Sei sulla pagina 1di 300

American Cancer Society

Atlas of
Clinical Oncology

Series Volumes
Blumgart, Jarnagin, Fong Hepatobiliary Cancer
Cameron Pancreatic Cancer
Carroll Prostate Cancer
Char Cancer of the Eye and Orbit
Clark, Duh, Jahan, Perrier Endocrine Tumors
Eifel, Levenback Cervical, Vulvar and Vaginal Cancer
Ginsberg Lung Cancer
Grossbard Malignant Lymphomas
Ozols Ovarian Cancer
Pollock Soft Tissue Sarcomas
Posner, Vokes, Weichselbaum Esophagus, Stomach and Small Bowel Cancer
Prados Brain Cancer
Raghavan Germ Cell Tumors
Rice, Taylor Endometrial and Uterine Cancer
Shah Head and Neck Cancer
Shipley Urothelial Cancer
Silverman Oral Cancer
Sober, Haluska Skin Cancer
Wiernik Adult Leukemias
Willett Colon and Rectal Cancer
Winchester, Winchester Breast Cancer
Yasko Bone Cancer
American Cancer Society

Atlas of
Clinical Oncology

Breast Cancer
American Cancer Society
Atlas of
Clinical Oncology

Editors

GLENN D. STEELE JR, MD


University of Chicago
THEODORE L. PHILLIPS, MD
University of California
BRUCE A. CHABNER, MD
Harvard Medical School

Managing Editor

TED S. GANSLER, MD, MBA


Director of Health Content, American Cancer Society
American Cancer Society
Atlas of
Clinical Oncology

Breast Cancer
David J. Winchester, MD, FACS
Associate Attending, Department of Surgery
Evanston Northwestern Healthcare
Evanston, Illinois
Assistant Professor of Surgery
Northwestern University Medical School
Chicago, Illinois

David P. Winchester, MD, FACS


Chairman, Department of Surgery
Evanston Northwestern Healthcare
Evanston, Illinois
Professor of Surgery
Northwestern University Medical School
Chicago, Illinois

2000
B.C. Decker Inc.
Hamilton • London
Contributors
JOEL R. BERNSTEIN, MD GEOFFREY C. FENNER, MD
Department of Radiology Evanston Northwestern Healthcare
Evanston Northwestern Healthcare Evanston, Illinois
Evanston, Illinois Breast Reconstruction
Screening and Diagnostic Imaging
RICHARD E. FINE, MD
WILLIAM D. BLOOMER, MD The Breast Center
Department of Radiology Marietta, Georgia
Evanston Northwestern Healthcare Image-Directed Breast Biopsy
Evanston, Illinois
Breast Cancer and Radiation Therapy ROBERT A. GOLDSCHMIDT, MD
Department of Pathology
DAVID R. BRENIN, MD Evanston Northwestern Healthcare
Columbia-Presbyterian Medical Center Evanston, Illinois
New York, New York Histopathology of Malignant Breast Disease
Unusual Breast Pathology
HANINA HIBSHOOSH, MD
WENDY R. BREWSTER, MD Columbia University, College of Physicians and
Department of Obstetrics and Gynecology Surgeons
University of California Columbia-Presbyterian Medical Center
Irvine, California New York, New York
Estrogen Replacement Therapy for Unusual Breast Pathology
Breast Cancer Survivors
GABRIEL N. HORTOBAGYI, MD
MASSIMO CRISTOFANILLI, MD Department of Breast Medical Oncology
Department of Breast Medical Oncology MD Anderson Cancer Center
MD Anderson Cancer Center Houston, Texas
Houston, Texas Breast Cancer Risk and Management:
Breast Cancer Risk and Management: Chemoprevention, Surgery, and Surveillance
Chemoprevention, Surgery, and Surveillance
JAN M. JESKE, MD
PHILIP J. DISAIA, MD Department of Radiology
Department of Obstetrics and Gynecology Evanston Northwestern Healthcare
University of California Evanston, Illinois
Irvine, California Screening and Diagnostic Imaging
Estrogen Replacement Therapy for Breast Cancer
Survivors JANARDAN D. KHANDEKAR, MD
Department of Medicine
WILLIAM L. DONEGAN, MD Evanston Northwestern Healthcare
Department of Surgery, Sinai Samaritan Evanston, Illinois
Medical Center Surveillance of the Breast Cancer Patient
Milwaukee, Wisconsin
Carcinoma of the Breast in Men
Contributors v

DAVID W. KINNE, MD LISA NEWMAN, MD, FACS


Columbia-Presbyterian Medical Center Department of Surgical Oncology
New York, New York University of Texas, MD Anderson Cancer Center
Unusual Breast Pathology Houston, Texas
Breast Cancer Risk and Management:
MICHAEL A. LACOMBE, MD Chemoprevention, Surgery, and Surveillance
Department of Radiology
Evanston Northwestern Healthcare PHILIP N. REDLICH, MD, PHD
Evanston, Illinois Department of Surgery
Breast Cancer and Radiation Therapy Medical College of Wisconsin
Milwaukee, Wisconsin
LAURIE H. LEE, PA-C Carcinoma of the Breast in Men
Division of General Surgery
Evanston Northwestern Healthcare STEPHEN F. SENER, MD
Evanston, Illinois Division of General Surgery
Surgical Management of Ductal Carcinoma In Situ Evanston Northwestern Healthcare
Evanston, Illinois
GERSHON Y. LOCKER, MD Surgical Management of Ductal Carcinoma In Situ
Division of Hematology
Evanston Northwestern Healthcare S. EVA SINGLETARY, MD, FACS
Evanston, Illinois MD Anderson Cancer Center
Adjuvant Systemic Therapy of Early Breast Cancer Houston, Texas
Locally Advanced Breast Cancer
HENRY T. LYNCH, MD
Creighton University MARGARET A. STULL, MD
Omaha, Nebraska Department of Radiology
Genetics, Natural History, and DNA-Based University of Illinois
Genetic Counseling in Hereditary Breast Cancer Chicago, Illinois
Screening and Diagnostic Imaging
JANE F. LYNCH, BSN
Creighton University ANN D. THOR, MD
Omaha, Nebraska Department of Pathology
Genetics, Natural History, and DNA-Based Evanston Northwestern Healthcare
Genetic Counseling in Hereditary Breast Cancer Evanston, Illinois
Prognostic and Predictive Markers in Breast
DOUGLAS E. MERKEL, MD Cancer
Division of Hematology-Oncology
Evanston Northwestern Healthcare DAVID J. WINCHESTER, MD, FACS
Evanston, Illinois Department of Surgery
Treatment of Metastatic Breast Cancer Evanston Northwestern Healthcare
Evanston, Illinois
DAN H. MOORE II, PHD Northwestern University Medical School
Geraldine Brush Cancer Research Institute Chicago, Illinois
Pacific Medical Center Evaluation and Surgical Management of Stage I
Department of Epidemiology and Biostatistics and II Breast Cancer
University of California
San Francisco, California DAVID P. WINCHESTER, MD, FACS
Prognostic and Predictive Markers in Breast Cancer Department of Surgery
Evanston Northwestern Healthcare
THOMAS A. MUSTOE, MD Evanston, Illinois
Division of Plastic Surgery, Northwestern University Northwestern University Medical School
Medical School Chicago, Illinois
Chicago, Illinois
Breast Reconstruction
B.C. Decker Inc.
4 Hughson Street South
P.O. Box 620, L.C.D. 1
Hamilton, Ontario L8N 3K7
Tel: 905-522-7017; 1-800-568-7281
Fax: 905-522-7839
E-mail: info@bcdecker.com
Website: http://www.bcdecker.com

© 2000 American Cancer Society


All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted,
in any form or by any means, electronic, mechanical, photocopying, recording, or otherwise, without prior
written permission from the publisher.
00 01 / PC / 9 8 7 6 5 4 3 2 1

ISBN 1–55009–112–3
Printed in Canada

Sales and Distribution


United States U.K., Europe, Scandinavia, Middle East South America
B.C. Decker Inc. Blackwell Science Ltd. Ernesto Reichmann, Distribuidora
P.O. Box 785 Osney Mead de Livros Ltda.
Lewiston, NY U.S.A. 14092-0785 Oxford OX2 0EL Rua Coronel Marques
Tel: 905-522-7017/1-800-568-7281 United Kingdom 335-Tatuape, 03440-000
Fax: 905-522-7839 Tel: 44-1865-206206 Sao Paulo-SP-Brazil
e-mail: info@bcdecker.com Fax: 44-1865-721205 Tel/Fax: 011-218-2122
website: http://www.bcdecker.com e-mail: info@blackwell-science.com

Canada Australia Foreign Rights


B.C. Decker Inc. Blackwell Science Asia Pty, Ltd. John Scott & Co.
4 Hughson Street South 54 University Street International Publishers’ Agency
P.O. Box 620, L.C.D. 1 Carlton, Victoria 3053 P.O. Box 878
Hamilton, Ontario L8N 3K7 Australia Kimberton, PA 19442
Tel: 905-522-7017/1-800-568-7281 Tel: 03 9347 0300 Tel: 610-827-1640
Fax: 905-522-7839 Fax: 03 9349 3016 Fax: 610-827-1671
e-mail: info@bcdecker.com e-mail: info@blacksci.asia.com.au
website: http://www.bcdecker.com

Japan South Korea


Igaku-Shoin Ltd. Seoul Medical Scientific Books Co.
Foreign Publications Department C.P.O. Box 9794
3-24-17 Hongo, Bunkyo-ku, Seoul 100-697
Tokyo 113-8719, Japan Seoul, Korea
Tel: 3 3817 5676 Tel: 82-2925-5800
Fax: 3 3815 6776 Fax: 82-2927-7283
e-mail: fd@igaku.shoin.co.jp

Notice: The authors and publisher have made every effort to ensure that the patient care recommended herein, including choice of drugs
and drug dosages, is in accord with the accepted standard and practice at the time of publication. However, since research and regulation
constantly change clinical standards, the reader is urged to check the product information sheet included in the package of each drug, which
includes recommended doses, warnings, and contraindications. This is particularly important with new or infrequently used drugs.
Contents
1 Genetics, Natural History, and DNA-Based Genetic Counseling
in Hereditary Breast Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
Henry T. Lynch, MD, Jane F. Lynch, BSN

2 Breast Cancer Risk and Management: Chemoprevention, Surgery, and Surveillance . . . 19


Massimo Cristofanilli, MD, Lisa Newman, MD, FACS, Gabriel N. Hortobagyi, MD

3 Screening and Diagnostic Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41


Jan M. Jeske, MD, Joel R. Bernstein, MD, Margaret A. Stull, MD

4 Image-Directed Breast Biopsy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 65


Richard E. Fine, MD

5 Histopathology of Malignant Breast Disease. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89


Robert A. Goldschmidt, MD

6 Unusual Breast Pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 99


David R. Brenin, MD, Hanina Hibshoosh, MD, David W. Kinne, MD

7 Prognostic and Predictive Markers in Breast Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . 113


Ann D. Thor, MD, Dan H. Moore II, PhD

8 Surgical Management of Ductal Carcinoma In Situ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 131


Stephen F. Sener, MD, Laurie H. Lee, PA-C

9 Evaluation and Surgical Management of Stage I and II Breast Cancer. . . . . . . . . . . . . . 139


David J. Winchester, MD, FACS

10 Locally Advanced Breast Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 153


S. Eva Singletary, MD, FACS

11 Breast Reconstruction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 171


Geoffrey C. Fenner, MD, Thomas A. Mustoe, MD
viii Contents

12 Adjuvant Systemic Therapy of Early Breast Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . 201


Gershon Y. Locker, MD

13 Breast Cancer and Radiation Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 219


Michael A. LaCombe, MD, William D. Bloomer, MD

14 Carcinoma of the Breast in Men . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 239


Philip N. Redlich, MD, PhD, William L. Donegan, MD

15 Estrogen Replacement Therapy for Breast Cancer Survivors . . . . . . . . . . . . . . . . . . . . . 253


Wendy R. Brewster, MD, Philip J. DiSaia, MD

16 Surveillance of the Breast Cancer Patient . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 263


Janardan D. Khandekar, MD

17 Treatment of Metastatic Breast Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 271


Douglas E. Merkel, MD

Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 287
Preface
The organization of this book reflects a logical, beyond the regimented doctrines of an overwhelm-
stepwise evaluation and treatment of the patient ing preoccupation of cancer treatment, immediate
with breast cancer. It emphasizes the importance reconstruction with microvascular surgery and
of early detection, but highlights a move toward other techniques have provided an answer to some
risk identification and reduction. The understand- of the physical and psychological challenges of
ing of the breast cancer patient has evolved from breast cancer. Recognition of other competing
the radical mastectomy for all patients to a tai- causes of mortality in the breast cancer survivor has
lored approach employing aggressive applica- led to a more comprehensive consideration of hor-
tions of treatment modalities according to their mone replacement therapy to address quality of life
respective risk reductions. issues and to reduce the risk of cardiovascular
Despite shifting efforts to identify high-risk disease and osteoporosis. In the patient with
patients and address their risk with pre-emptive metastatic disease, the introduction of novel forms
strategies, there remains a worldwide educational of treatment, such as with Herceptin, has led to sig-
challenge to adopt early detection screening guide- nificant improvements in survival. In total, these
lines. Although there is continuing progress in innovations represent significant progress and pro-
implementing mortality reducing surveillance vide important directions for future interventions.
guidelines as reflected by the increased prevalence One of the biggest challenges to the clinician
of preinvasive breast cancer, the full spectrum of has been the recognition of improved methods of
disease remains a challenge to the medical commu- diagnosis and treatment and utilization of these
nity. The high prevalence of breast cancer continues improvements despite ingrained practices. This
to drive improvements in all areas of detection, pattern is well documented by the great variation
diagnostic evaluation, disease characterization, observed nationally in the implementation of
multimodality therapy, quality of life issues, and, breast preserving surgery and the utilization of
finally, in the treatment of patients whose disease adjuvant treatment.
has extended beyond our capabilities to detect or It is the goal of this book to identify signifi-
contain local or regional cancer. cant improvements in each area of breast cancer
One of the most important innovations in the diagnosis and treatment, and to help accelerate
understanding of breast cancer has been the identi- the dispersion of this knowledge to an ever-broad-
fication of genetic mutations that have allowed the ening spectrum of physicians and scientists who
opportunity to intervene with proven surgical or are dedicated to preventing and treating one of the
chemopreventive strategies for high-risk patients. most common afflictions of women.
Diagnostic imaging technology continues to pro- We wish to thank our distinguished authors for
vide increased resolution and precision, resulting their timely and expert contributions to this effort.
in an enhanced ability to preserve tissue. The sur- We also wish to thank the American Cancer Soci-
gical treatment of breast cancer is in the process of ety, particularly Ted Gansler, for a helpful review
taking another significant step forward, with the of this book.
development of sentinel lymph node biopsy. The
definition of prognostic factors has helped to guide DJW
important adjuvant therapy decisions. Moving DPW
To our parents, for their example and knowledge,
and to Marilyn, Doris, Eric, Laura, Elena, and
Colin, who have allowed us to spend their time
working on this project.

David J. Winchester
David P. Winchester
1
Genetics, Natural History, and
DNA-Based Genetic Counseling
in Hereditary Breast Cancer
HENRY T. LYNCH, MD
JANE F. LYNCH, BSN

The extraordinary advances in molecular genet- genetic and phenotypic heterogeneity, natural
ics during the past decade have established history, genetic counseling issues, and cancer
beyond doubt that there is a Mendelian inherited control implications.
basis for a subset of virtually all forms of can-
cer.1 Specifically, more than 30 hereditary cancer GENETICS
syndromes have been shown to harbor germ-line
mutations. These culprit molecular genetic fac- The combination of carcinoma of the breast
tors include oncogenes such as the RET proto- and ovary in families, now known as the
oncogene for the multiple endocrine neoplasia HBOC syndrome, was first reported in the
type 2 syndromes, the mismatch repair genes early 1970s.4–6 The molecular genetic discov-
(hMSH2, hMLH1) in hereditary nonpolyposis eries that confirmed beyond any doubt the
colorectal cancer (HNPCC) of the Lynch I and hereditary basis for HBC and HBOC have
Lynch II syndrome variants, and tumor suppres- progressed at an explosive rate, particularly
sor genes. Examples of the latter include APC, over the past decade. This avalanche of knowl-
which predisposes to familial adenomatous edge was heralded by the gene linkage study
polyposis (FAP), and BRCA1 and BRCA2 muta- of Hall and colleagues,7 which identified a
tions in hereditary breast cancer, the subject of locus on chromosome 17q for families with
this chapter. site-specific breast cancer. Subsequently,
In the United States in 1999, it was pro- Narod and colleagues8 reported that this same
jected that 176,300 new cases of carcinoma of locus was responsible for the HBOC syndrome.
the breast would be diagnosed, and 43,700 The culprit gene, now known as BRCA1, was
would die from this disease.2 The current then cloned.9 More recently, a second breast
authors estimate that approximately 10 percent cancer susceptibility locus on chromosome
(17,600) of these newly diagnosed patients will 13q, known as BRCA2, was identified by link-
manifest a hereditary breast cancer (HBC) dis- age analysis10 and subsequently cloned.11
order, the most common of which will be the Approximately 45 percent of all hereditary
hereditary breast-ovarian cancer (HBOC) syn- breast cancer-prone families, including those
drome.3 It is the purpose of this chapter to characterized as HBOC, owe this condition to
update what is known about HBC, including its mutations of the BRCA1 gene;12 a slightly lower

1
2 BREAST CANCER

percentage is due to BRCA2 mutations. Initial and/or BRCA2 mutations, family history
studies estimated that carriers of the BRCA1 remains a factor in predicting breast cancer risk.
germ-line mutation harbor a lifetime risk for In families with breast and ovarian cancers, the
breast cancer of about 85 percent12,13 and a risk aggregation of these two cancers appears to be
for ovarian cancer that ranges between 40 and explained by BRCA1/BRCA2 mutation-carrier
66 percent.12,13 Carriers of the BRCA2 muta- probability.” This study clearly enunciates the
tion, on the other hand, have a lifetime risk of need for obtaining a well-orchestrated cancer
breast cancer of about 85 percent, but their risk family history for the assessment of breast/ovar-
for ovarian cancer is somewhat lower (10 to ian cancer risk.
20 %).12,13 Male BRCA2 mutation carriers have Previously, the best prediction of a patient’s
an approximate 7 percent lifetime risk for lifetime breast cancer risk was 50 percent. This
breast cancer. Other cancers occurring in BRCA2 estimate was based upon the patient’s position
mutation carriers include carcinoma of the pan- in the pedigree, namely, having one or more
creas, head and neck, and intraocular malignant first-degree relatives with a syndrome cancer
melanoma. Males who are harbingers of germ- in the direct genetic lineage of an HBC or
line mutations in BRCA1/BRCA2 will have a HBOC family. The identification of the muta-
two- to three-fold increased lifetime risk for tions for BRCA19 and BRCA211 now enables
prostate cancer. physicians and genetic counselors to predict a
These initial breast and ovarian cancer gene patient’s lifetime risk for carcinoma of the
penetrance risks were based upon publications breast and ovary, in context with the penetrance
of highly extended pedigrees selected because of these genes.
of their profound familial cancer aggregations;
they are thereby biased in the direction of can- Cancer Family History and
cer excess. In contrast, recent observations of Mutation Search
breast and ovarian cancer occurrence in the
Ashkenazi Jewish founder mutations (185delAG The search for a germ-line mutation should be
and 5382insC mutations on the BRCA1 gene, performed only on families with substantial
and 6174delT on the BRCA2 gene) indicate that evidence of a hereditary cancer syndrome.
the lifetime risk for breast cancer is only 56 Therefore, to establish a hereditary breast can-
percent, and that of ovarian cancer 16 percent.14 cer syndrome diagnosis, a detailed collection of
Claus and colleagues15 examined the family a patient’s cancer family history, with as much
history of carcinoma of the breast and ovary in a pathologic corroboration as possible, is manda-
data set involving 4,730 patients with breast can- tory. The family history may potentially consti-
cer and 4,688 controls who were enrolled in the tute the most cost-beneficial component of a
Cancer and Steroid Hormone Study. Attention patient’s medical workup; its collection and
was given to the association between family his- evaluation in the typical clinical setting, how-
tory of carcinoma of the breast and/or ovary and ever, remains notoriously neglected.16,17 This
breast cancer risk when controlling for the car- problem was well documented by David and
rier status of BRCA1 and BRCA2 mutations. The Steiner-Grossman17 through a survey of 76
question examined pertained to whether the fam- acute care, non-psychiatric hospitals in New
ily history of carcinoma of the breast remained a York City. Only four of the 64 reporting hospi-
predictive risk factor once the carrier status for tals indicated that family history information
BRCA1 and/or BRCA2 was given consideration. was reported in their medical records. Such
Findings disclosed that among those women serious omissions must be resolved in order to
“…with a moderate family history of breast can- enhance cancer control. Otherwise, the oppor-
cer, that is, predicted noncarriers of BRCA1 tunity to search for germ-line cancer prone
Genetics, Natural History, and DNA-Based Genetic Counseling in Hereditary Breast Cancer 3

mutations may be lost. This is a pity since, females, or the few cancers that are occurring
when a cancer-causing mutation is identified, may be in the paternal lineage. The overall
this information can, in concert with genetic effect is that it becomes exceedingly difficult to
counseling, be used effectively to benefit the predict whether such a small family should be a
patient and family members. candidate for searching for a mutation in
BRCA1 or BRCA2.
ASSESSMENT OF BREAST
CANCER-PRONE FAMILIES HETEROGENEITY AND HEREDITARY
BREAST CANCER
Building the case for hereditary cancer is fre-
quently based upon the cardinal clinical fea- Virtually all forms of hereditary cancer show
tures of hereditary cancer (namely, early age of significant genetic and phenotypic heterogene-
cancer onset, pattern of multiple primary can- ity. For example, breast cancer occurs in signif-
cers [such as breast and ovarian cancer], verti- icant excess in disorders associated with extra-
cal transmission of cancer, and increased num- breast cancer sites, such as Li-Fraumeni
ber of cancer occurrences) (Table 1–1). It is syndrome (Figure 1–1), Bloom’s syndrome,
virtually axiomatic that the larger the breast Cowden’s disease, ataxia-telangiectasia, the
cancer-prone family, the greater the number of breast-gastrointestinal tract cancer syndrome,
expected carcinomas of the breast or ovary. One extraordinarily early-onset breast cancer (Fig-
can be more confident of a likely hereditary eti- ure 1–2), and the HBOC syndrome (Figure
ology for breast cancer if there is evidence for 1–3). Undoubtedly, other tumor combinations
earlier age of onset of cancer, especially when and/or hereditary syndromes which will qualify
there is familial clustering of these cancers, as hereditary breast cancer are yet to be identi-
particularly ovarian cancer, among primary and fied. Space does not allow a discussion of each
secondary relatives (see Table 1–1). In such a of these breast cancer-associated disorders (for
setting, there is an increased probability that a more detail see Lynch and colleagues18).
germ-line mutation (BRCA1, BRCA2) will be Clearly, it is no longer appropriate to char-
found. On the other hand, when dealing with acterize hereditary breast cancer as a generic
families that are small, there may be a limited term. Rather, one must be more precise and
number of patients with cancer, a deficit of denote the particular breast cancer-associated

Table 1–1. ESTIMATED PROBABILITY OF BRCA1 MUTATION BASED ON FAMILY HISTORY

Family History Probability of BRCA1 Mutation (%)

Single affected person


Breast cancer at < 30 years of age 12
Breast cancer at < 40 years of age 6
Breast cancer at 40–49 years of age 3
Ovarian cancer at < 50 years of age 7
Sister pairs
Both with breast cancer at < 40 years of age 37
Both with breast cancer at 40–49 years of age 20
Breast cancer at < 50 years of age, ovarian cancer at < 50 years of age 46
Both with ovarian cancer at < 50 years of age 61
Families
Breast cancer only, three or more cases at < 50 years of age 40
Two or more breast cancers and one or more ovarian cancers 82
Two or more breast cancers and two or more ovarian cancers 91

Reprinted with permission of author (Barbara Weber, MD) and publisher from Weber B. Breast cancer susceptibility genes: current challenges
and future promises. Ann Intern Med 1996;124:1088–90.
4
BREAST CANCER

Cancer Sites
Individual number Ad Adrenal cortical
Unaffected Br Breast
Current age Bt Brain tumor
Cancer by pathology, Co Colon
age at diagnosis CSU Cancer site unknown
Current age Leu Leukemia
Cancer by family history, Lu Lung
age of death Lym Lymphoma
Lyx Larynx
Multiple primary cancers by pathology Nb Neuroblastoma
Pan Pancreas
Cancer by death certificate or Pro Prostate
medical records Sar Sarcoma
Number of unaffected progeny
(Both sexes)
Proband

Twins

Figure 1–1. Updated pedigree of a family with sarcoma, breast cancer and brain tumors, lung and laryngeal cancer, and adrenocortical carcinoma (SBLA syndrome, also known as Li-
Fraumeni syndrome). Reprinted with permission from Lynch et al. Genetic and pathologic findings in a kindred with hereditary sarcoma, breast cancer, brain tumors, leukemia, lung,
larygeal, and adrenal cortical carcinoma. Cancer 1978; 41:2055–64.
Genetics, Natural History, and DNA-Based Genetic Counseling in Hereditary Breast Cancer 5

syndrome relating to a particular patient/family. cancer burden will be hereditary. Interestingly,


Such syndrome identification is important not Ramus and colleagues22 reported a patient from a
only for molecular genetic assessment but, Hungarian family who manifested both breast
moreover, for targeted surveillance and man- and ovarian cancer and was found to have trun-
agement purposes. cating mutations in both the BRCA1 and BRCA2
genes. This patient “... carried the 185delAG
GENOTYPE-PHENOTYPE mutation in BRCA1 as well as the 6174delT
DIFFERENCES mutation in BRCA2. Both of these mutations are
common in Ashkenazi Jewish breast cancer
More than 200 different BRCA1 germ-line patients.”23–25 Recently, Liede and colleagues26
mutations have been identified in HBOC fami- identified an Ashkenazi Jewish kindred with
lies. Certain types of these mutations may give three mutations, namely BRCA1 185delAG,
rise to differing patterns of cancer occurrence. BRCA1 5382insC, and BRCA2 6174delT. Each
For example, Gayther and colleagues19 suggest founder mutation has been shown to have a fre-
that the position of the BRCA1 mutation has a quency of approximately 1 percent in the Ashke-
significant influence on the ratio of breast to nazi population.27–29
ovarian cancer in HBOC kindreds. Specifically,
they reported that mutations in the 3' third of the
gene are associated with a lower proportion of
ovarian cancer. However, these findings must be
reviewed cautiously. Serova and colleagues20
were unable to confirm these findings. How-
ever, the findings of the latter did suggest that
the risk of ovarian cancer is greater in families
with mutations associated with reduced RNA
levels. In the case of BRCA2, Gayther and col-
leagues21 found that “... truncating mutations in
families with the highest risk of ovarian cancer
relative to breast cancer are clustered in a region
of approximately 3.3 kb in exon 11 (p = .0004).”
Further research in this area may establish links
between specific mutations and specific cancer
risk that will be extremely useful for genetic
counseling. Until confirmation of these geno-
type-phenotype findings is more firmly estab-
Cancer Sites
lished, however, the current authors prefer to Individual number
Unaffected Br Breast
St Stomach
withhold this information in the genetic coun- Current age
Cancer by pathology,
seling setting. age at diagnosis
Current age
Most of the hereditary cases will harbor a Multiple primary cancers by pathology
age at death
BRCA1 or BRCA2 germ-line mutation. However, Cancer by death certificate or
medical records
one should expect (albeit rarely) to encounter Number of unaffected progeny
(Both sexes)
families where both BRCA1 and BRCA2 muta- Proband
tions are segregating, considering the high preva- Figure 1–2. Pedigree of a family showing extremely early age
lence of carcinoma of the breast and ovary in the of onset of hereditary breast cancer. Reprinted with permis-
general population, coupled with the fact that sion from Lynch et al. Extremely early onset hereditary breast
cancer (HBC): surveillance/management implications. Nebr
approximately 5 to 10 percent of the total breast Med J 1988; 73:97–100.
6
BREAST CANCER

Cancer Sites
Individual number Br Breast
Unaffected Co Colon
Current age CSU Cancer site unknown
Lu Lung
Cancer by pathology, Mmel Malignant melanoma
age at diagnosis Omen Omentum
Current age Ov Ovarian
Cancer by family history, Pro Prostate
age of death Psu Primary site unknown
Multiple primary cancers by St Stomach
medical records or death certificate
Twins
Multiple primary cancers by pathology Number of unaffected progeny
(Both sexes)
Cancer by death certificate or
medical records Proband

Figure 1–3. Updated pedigree of a large hereditary breast-ovarian cancer family. Adapted with permission from Lynch et al. Management of familial breast cancer, parts I and II.
Arch Surg 1978;113:1060–71.
Genetics, Natural History, and DNA-Based Genetic Counseling in Hereditary Breast Cancer 7

PATHOLOGY OF HEREDITARY lar neoplasia are signatures of the BRCA2 HBC


BREAST/OVARIAN CANCER IN phenotype.32 In contrast, BRCA1 HBC cases
CONCERT WITH BRCA1 OR manifest a deficit of TLG carcinomas and lob-
BRCA2 MUTATIONS ular neoplasia.32 Armes and colleagues37 con-
firm an excess of TLG carcinoma (in their
Pathology studies have shown differences cases, pleomorphic lobular carcinomas) in
between BRCA1- and BRCA2-related breast BRCA2 HBC in a population-based study of
cancers when compared with sporadic controls. BRCA2 cases that were not specifically
Specifically, Marcus and colleagues30–32 have recruited from large HBOC families.
shown that BRCA1 HBC has a highly distinctive The pathophenotype of BRCA2 HBC may
pathology phenotype, consisting of an increased be more heterogeneous than BRCA1 HBC
number of aneuploid cancers, more medullary when the amount of high-grade carcinoma in
carcinomas, and high proliferation rates as mea- the syndrome is considered. There have been
sured by DNA flow cytometry and mitotic reports of BRCA2 families with predominantly
grade, and lesser ductal carcinoma in situ high-grade carcinomas.38,39 However, we have
(DCIS) than in nonfamilial cases. In alluding to not seen high-grade predominance in the four
high S-phase fraction in HBC and attributing it BRCA2 families we have studied nor as the
to the BRCA1-linked subset, it was suggested average phenotype of the “other” HBC group in
that the mutation resulted in enhanced cellular which most Creighton BRCA2 families would
proliferation.33 This prediction was borne out by reside.32 Similarly, the Breast Cancer Linkage
the demonstration of the antiproliferative effect Consortium has not observed unusually high
of BRCA1 mRNA protein in vitro and in grades in its BRCA2 family series.40 The higher
vivo34–36 after the gene was cloned. The current grades reported from Iceland39 and, to a lesser
authors have proposed a model for the BRCA1 extent, from the Linkage Consortium,40,41 may
pathophenotype that considers the tumors to be well be associated with a site on the BRCA2
in an advanced state of genetic evolution.30 gene, in these cases the 999del5 mutation.37
In contrast, “other” HBCs (cases from HBC Lakhani and colleagues41 confirmed many
families with no BRCA1 mutations, no 17q of the original observations of Marcus and col-
linkage, and a paucity of ovarian cancer affect- leagues.32,33,42 Specifically, they showed that
eds, or with BRCA2 mutations or 13q linkage) “Cancers associated with BRCA1 mutations
appear to lack the systematic high grade, aneu- exhibited higher mitotic counts (p = .001), a
ploidy, and high proliferation of BRCA1 HBCs, greater proportion of the tumor with a continu-
and they are not deficient in in situ carci- ous pushing margin (p < .0001), and more lym-
noma.30,31 This “other” group also has more phocytic infiltration (p = .002) than sporadic
invasive lobular, tubular, tubulolobular, and (ie, control) cancers. Cancers associated with
cribiform special type carcinomas, which we BRCA2 mutations exhibited a higher score for
have designated as “tubular-lobular group” tubule formation (fewer tubules) (p = .0002), a
(TLG). Indeed, the excess of TLG and “no spe- higher proportion of the tumor perimeter with a
cial type” (NST) invasive carcinomas with TLG continuous pushing margin (p < .0001), and a
“features” (10 to 50% tumor composition) par- lower mitotic count (p = .003) than control can-
allels a trend for more lobular neoplasia (lobu- cers.” These authors concluded that this
lar carcinoma in situ and atypical lobular histopathology information may improve the
hyperplasia) in “other” HBC. These features classification of breast cancers in those show-
are present in the subset of mutation-confirmed ing a positive family history for this disease.
BRCA2 HBC cases in the “other” HBC group,32 Specifically, employing multifactorial analysis
which suggests that TLG carcinomas and lobu- results from their previous estimates, they
8 BREAST CANCER

found that 7.5 percent of individuals with second (nonbreast) cancers in young women
breast cancer in Britain who had been diag- with a family history (FH) suggestive of inher-
nosed at between the ages of 20 and 29 years ited breast cancer susceptibility compared with
harbor a BRCA1 mutation.43 Further, “Assum- young women without an FH.” As expected, the
ing that the odds ratios from our analysis are patients with a positive FH showed an
independent of age, only about 2 percent of increased risk of contralateral breast cancer.
case subjects in this age group in whom the This matter of contralateral breast cancer must
mitotic count is below 5 per 10 hpf, without be given careful consideration when counseling
continuous pushing margins, and in whom women with positive family histories who are
there is no lymphocytic infiltrate would be considering the option of breast conserving
expected to carry a BRCA1 mutation. By con- surgery and radiation therapy versus modified
trast, about 45 percent of case subjects in the radical mastectomy. Given these findings,
20- to 29-year-old group with 20 to 39 mitoses Chabner and colleagues45 conclude that “...
per 10 hpf, continuous pushing margins occu- young women can be offered conservative
pying more than 75 percent of the tumor surgery and radiation therapy as a reasonable
perimeter, and a prominent lymphocytic infil- option at breast cancer diagnosis.” However,
trate would be expected to be BRCA1 carriers. the investigators appropriately call attention to
The corresponding proportions based on the limitations in their study: a relatively short
mitotic count would be 4 and 16 percent.” follow-up time, small size of their cohort, and
the absence of specific genetic findings on
RADIATION EFFECTS AND BRCA1 their patients, including an absence of BRCA1
AND BRCA2 MUTATIONS or BRCA2 mutation findings.

Questions have been raised relevant to potential OVARIAN CARCINOMA


carcinogenic risk of radiation exposure for
women who harbor the BRCA1 or BRCA2 Any discussion of the genetics of carcinoma of
mutations. Scully and colleagues44 raised the the breast must include ovarian cancer. This dis-
possibility that there may be an interaction ease will affect approximately 1 percent of
between BRCA1 and BRCA2 gene products women in the United States during their lifetime,
with respect to proteins involved in the repair of where it accounts for about 14,500 deaths annu-
radiation-induced DNA errors. However, this ally2 with a five-year survival rate of < 30 per-
issue remains controversial due to lack of con- cent. The biological mechanism of transforming
firmation of this risk in the past by other inves- benign cells to carcinoma remains elusive,
tigators.45 Nevertheless, recent evidence has although it likely involves a multistep process
indicated that both BRCA1 and BRCA2 are requiring an accumulation of genetic lesions
associated with defective repair of radiation- involving different gene classes. As mentioned,
induced DNA damage.44,46 the etiologic association of ovarian cancer with
In a study by Chabner and colleagues45 of breast cancer was first reported in the early
201 patients, 29 of whom had positive family 1970s in a series of breast cancer-prone pedi-
histories of breast cancer (a mother or sister grees;4–6,8 both BRCA1 and BRCA2 mutations
previously diagnosed before the age of 50 were subsequently found to predispose to ovar-
years, or ovarian cancer at any age) and who ian cancer. In BRCA1, the lifetime risk for ovar-
had undergone breast-conserving surgery and ian cancer is in the range of approximately 50
radiation therapy for early-stage breast cancer, percent, while in BRCA2 the lifetime ovarian
there was no evidence associated with a higher cancer risk is about 20 percent. Ovarian carci-
rate of “...local recurrence, distant failure, or noma is also an integral lesion in Lynch syn-
Genetics, Natural History, and DNA-Based Genetic Counseling in Hereditary Breast Cancer 9

drome II.47 Lynch and colleagues48 have pro- gral part of the process of genetic predisposition
vided an extensive review of the genetics of testing, on either a clinical or research basis. Pre-
ovarian cancer. disposition testing should be performed on
patients for whom there is a strong family his-
GENETIC COUNSELING tory of cancer that is consonant with a likely
hereditary etiology, where the results can be ade-
We believe that genetic counseling should be quately interpreted, and where there is a poten-
mandatory for patients who are at high risk for tial to aid in the medical management of the
breast cancer and are contemplating DNA test- patient and/or family members.
ing in the search for specific germ-line muta- The American Society of Clinical Oncology
tions. Counseling should take place prior to col- also recognizes the need to strengthen regulatory
lection of DNA and at the time of disclosure of authority over laboratories that provide cancer
results. The ideal individual for initial gene test- predisposition tests that will ultimately be used
ing in a family where a hereditary form of breast in making informed clinical decisions. In the
cancer is considered likely would be one who interest of protecting patients and their families,
has had a syndrome cancer, particularly if diag- ASCO endorses the adoption of legislation to
nosed at an early age, and who is in the direct prohibit discrimination by insurance companies
line of descent of syndrome cancer expression. or employers based on an individual’s suscepti-
The clinician’s task during the genetic counsel- bility to cancer. Finally, ASCO and the American
ing process is to help the patient answer crucial Cancer Society prudently endorses the need for
questions which may arise during the genetic all individuals at hereditary risk for cancer to
testing process. Importantly, patients need to have, in concert with medical care, appropriate
decide whether to be tested for the presence of a genetic counseling which should be covered by
germ-line mutation in BRCA1 or BRCA2, once public and private third-party payers.
the facts are understood. They should be aware Figure 1–4 is an algorithm depicting the
of the potential for fear, anxiety, apprehension, process used by the Creighton cancer genetic
intrafamily strife, as well as insurance/ employ- research team to ascertain, test, and counsel
ment discrimination. Finally, they need to know HBC/HBOC-prone families. Detailed informa-
the best type of medical management for them, tion about the natural history of HBC/HBOC
based upon the test result. was provided and the pros and cons of DNA
Because there are a limited number of certi- testing were discussed with more than 2,000
fied genetic counselors who have sufficient members of 29 large families with BRCA1
knowledge of oncology to effectively counsel mutations and 8 families with BRCA2 mutations
cancer-prone families, the American Society of (Table 1–2).50 The current authors found that
Clinical Oncology (ASCO) has recommended the perceived risk for cancer was associated
that, whenever possible, physicians should per- with the individual’s position in the pedigree.
form genetic counseling.49 In its published posi- There was a significant tendency to overesti-
tion49 on genetic testing for cancer susceptibility, mate risk rather than underestimate it (p < .001)
ASCO recognizes that the clinical oncologist’s by a chi-square test from Fleiss and colleagues51
role should be to document the family history of (Table 1–3).
cancer, provide counseling with respect to a Fifty-seven family members who had pro-
patient’s inordinate lifetime cancer risk, and pro- vided blood samples several years ago declined
vide options for prevention and early detection the opportunity to receive the results of their
to those families for whom genetic testing may DNA testing. Thirty of the 57 responded to an
aid in the genetic counseling process. Informed anonymous questionnaire by giving one or
consent by the patient is considered to be an inte- more reasons for declining. Their responses
10 BREAST CANCER

varied, but fear of insurance discrimination was Of those choosing to learn their mutation
cited by 37 percent of this group, and fear of a status, the majority identified their children as
positive result was cited by 20 percent. a primary reason for being tested (Table 1–4).

Figure 1–4. Algorithm for education and assessment of BRCA1 and BRCA2 families. Reprinted with
permission from Lynch et al. Cancer. In press.
Genetics, Natural History, and DNA-Based Genetic Counseling in Hereditary Breast Cancer 11

Table 1–2. DEMOGRAPHIC CHARACTERISTICS OF STUDY SUBJECTS IN 29 BRCA1 FAMILIES AND 8 BRCA2 FAMILIES

BRCA1 BRCA2

Total number counseled and given gene status 339 85

Mutation positive 137 (40) 42 (49)


Mutation negative 198 (58) 43 (51)
Ambiguous 4 ( 1) 0

Male 89 (26) 20 (24)


Female 250 (74) 65 (76)

Age Group
< 25 11 ( 4) 4 ( 6)
25-50 89 (36) 26 (40)
> 50 150 (60) 35 (54)
Cancer status
Breast and/or ovarian cancer affected 59 (24) 16 (25)

Reproduced with permission of author and publisher from Lynch et al. An update on DNA-based BRCA1/BRCA2 genetic counseling in hereditary
breast cancer. Cancer Genet Cytogenet 1999;109:91–8.

Although most of these individuals freely chose transvaginal ovarian ultrasound as often as rec-
to be tested, occasionally they reported pressure ommended (Table 1–6). Most said they would
within the family either for or against their consider increasing surveillance and/or having
being tested. prophylactic oophorectomy if the test showed
Overall, 167 of 403 queried family members them to be mutation carriers. Note that all data
(41%) cited health management (especially on post-test management reflect what patients
surveillance for early detection and prophylac- say they will consider in the decision-making
tic surgery) as a reason for seeking genetic test- process, not necessarily what they plan to do.
ing, making it the second most common reason The emotional responses to disclosure of
reported. In the subgroup where mutation sta- germ-line mutation results cannot always be
tus information would have the greatest impact anticipated. Our data show that the majority of
on recommendations for surveillance (women patients who are negative express relief. How-
under the age of 40), 72 of 118 queried, or 61 ever, some may experience disbelief or survivor
percent, cited this reason (other subgroup data guilt. Those who are told they do have the
are not shown). germ-line mutation express a variety of reac-
Patients’ self reports indicated that currently
the majority were having breast cancer screening
tests at the recommended frequency (Table 1–5). Table 1–3. ESTIMATE OF PERCEIVED RISK OF
HARBORING A BRCA1/BRCA2 MUTATION
Nevertheless, the majority said that they would
consider increasing surveillance if the mutation Perceived Risk of
Mutation Carriage (n (%))
test showed they were mutation carriers. Only a
slim majority (13 of 25 overall) would consider Pedigree Risk* < 50% 50% > 50%
decreasing surveillance if the test showed they < 25% 27 (38.0%) 27 (38.0%) 17 (23.9%)
were not mutation carriers. Sixty-two percent 50% 44 (17.7%) 74 (29.8%) 130 (52.4%)
Affected/obligate 3 (3.8%) 5 (6.3%) 71 (89.9%)
(59 of 95) of all women over the age of 25 years gene carrier
said they would consider prophylactic mastec-
*These individuals tended to overestimate their risk more often than
tomy if the test showed they were mutation car- to underestimate it (p < .001) (chi square test51).
riers. Patient self reports indicated that a minor- Reproduced with permission of author and publisher from Lynch et al.
An update on DNA-based BRCA1/BRCA2 genetic counseling in
ity of these women are having CA-125 and hereditary breast cancer. Cancer Genet Cytogenet 1999;109:91–8.
12 BREAST CANCER

Table 1–4. REASONS FOR SEEKING RISK levels of stress, depression rates in decliners
ASSESSMENT IN COUNSELED MEMBERS OF 29
BRCA1 FAMILIES AND 8 BRCA2 FAMILIES increased from 26 percent at baseline to 47 per-
cent at one-month follow-up; depression rates
Counseled BRCA1 Counseled BRCA2
Individuals Individuals
in noncarriers decreased and in carriers showed
(n = 319) (n = 84) no change (odds ratio [OR] for decliners versus
noncarriers = 8.0; 95% confidence interval [CI]
Top three reasons
for seeking risk 1.9 to 3.5; p = .0004). These significant differ-
assessment ences in depression rates were still evident at
Children 170 (53) 47 (56)
Surveillance/ 125 (39) 42 (50) the six-month follow-up evaluation (p = .04).”
prophylaxis It was concluded that in BRCA1/BRCA2-linked
Curiosity 94 (29) 24 (29)
families, individuals showing high levels of
Reproduced with permission of author and publisher from Lynch et al. cancer-related stress who ultimately declined
An update on DNA-based BRCA1/BRCA2 genetic counseling in
hereditary breast cancer. Cancer Genet Cytogenet 1999;109:91-8.
genetic testing appeared to be at increased risk
for depression. It was reasoned that they could
derive benefit through education and counsel-
tions, including acceptance because the results ing even though they might ultimately decline
are not a surprise to them, relief of anxiety with to be tested; these are the individuals who
the removal of uncertainty about their genetic require monitoring for the potential occurrence
risk status, a positive attitude in terms of pre- of adverse psychological effects.
vention, feelings of sadness, or even anger. The An important genetic counseling question is,
genetic counselor must be responsive to all of Do patients who received information about their
these emotions. genetic risk status, including the presence of a
At a baseline interview in a study by Ler- BRCA1/BRCA2 germ-line mutation, heed sur-
man and colleagues,52 breast-ovarian cancer- veillance and management recommendations?
related stress symptoms were predictive of the These recommendations include the need for
onset of depressive symptoms in family mem- increased frequency of mammography, breast
bers who were invited but declined testing. self-examination, and physician breast examina-
“Among persons who reported high baseline tion. Recommendations for ovarian screening

Table 1–5. SURVEILLANCE PRACTICES AND ATTITUDES TOWARD


PROPHYLACTIC MASTECTOMIES PRIOR TO GENETIC TEST RESULT DISCLOSURE*

BRCA1 BRCA2

Bilateral mastectomies prior to the counseling session


Breast cancer-affected 39/250 (16) 8/65 (12)
Breast cancer-free (Prophylactic) 23/250 ( 9) 3/65 ( 5)
Current surveillance practices‡
Mammography Ever 145/153 (95) 42/46 (91)
As recommended 92/118 (78) 25/36 (69)
Physician BE Ever 105/107 (98) 39/40 (97)
As recommended 84/90 (93) 37/38 (97)
Self BE Ever 102/116 (88) 38/42 (90)
As recommended 54/88 (61) 27/37 (73)
If carrier, will consider prophylactic mastectomy† 51/77 (66) 8/18 (44)
If carrier, will consider increasing surveillance‡ 62/65 (95) 24/25 (96)
If noncarrier, will consider decreasing surveillance‡ 9/20 (45) 4/5 (80)

*In counseled female members of 29 BRCA1 families and counseled female members in 8 BRCA2 families (number/number queried† (percent)).

The number queried varies from item to item since not all questions were asked and/or responded to within the genetic counseling setting.

Excluding women aged < 25 or with bilateral mastectomies.
BE= breast examination.
Reproduced with permission of author and publisher from Lynch et al. Cancer Genet Cytogenet 1999;109:91–8.
Genetics, Natural History, and DNA-Based Genetic Counseling in Hereditary Breast Cancer 13

Table 1–6. SURVEILLANCE PRACTICES AND ATTITUDES TOWARD


PROPHYLACTIC OOPHORECTOMIES PRIOR TO GENETIC TEST RESULT DISCLOSURE*

BRCA1 BRCA2

Bilateral oophorectomies prior to the counseling session


Ovarian cancer-affected 7/250 ( 3) 0/65 ( 0)
Ovarian cancer-free 83/250 (33) 5/65 ( 8)
Current surveillance practices‡
CA-125 Ever 26/65 (40) 4/19 (21)
As recommended 10/49 (20) 2/17 (12)
Ultrasound Ever 32/71 (45) 11/25 (44)
As recommended 11/51 (22) 2/16 (12)
Pelvic Ever 86/96 (90) 50/51 (98)
As recommended 64/79 (81) 45/46 (98)
If carrier, will consider prophylactic oophorectomy‡ 72/81 (89) 10/20 (50)
If carrier, will consider increasing surveillance‡ 50/53 (94) 25/26 (96)
If noncarrier, will consider decreasing surveillance‡ 8/15 (53) 5/6 (83)

*In counseled female members of 29 BRCA1 families and counseled female members in 8 BRCA2 families (number/number queried† (percent)).

The number queried varies from item to item since not all questions were asked and/or responded to within the genetic counseling setting.

Excluding women aged < 25 or with bilateral oophorectomies.
Reproduced with permission of author and publisher from Lynch et al. An update on DNA-based BRCA1/BRCA2 genetic counseling in hereditary
breast cancer. Cancer Genet Cytogenet 1999;109:91–8.

included transvaginal ovarian ultrasound, tomy on a retrospective cohort study of all


Doppler color bloodflow imagery, and CA-125. women with a breast cancer-positive family his-
However, the patients were thoroughly educated tory who underwent bilateral prophylactic mas-
about the limitations of ovarian cancer screening. tectomy at the Mayo Clinic between 1960 and
The option of prophylactic mastectomy and/or 1993. These women were divided into high-risk
prophylactic oophorectomy was also discussed versus moderate-risk groups based on their fam-
during genetic counseling sessions.50 ily history. Those at high risk showed pedigrees
Preliminary data show that psychological consistent with a single-gene, autosomal domi-
assessment 6 months following BRCA1/BRCA2 nant predisposition to carcinoma of the breast,
testing among unaffected individuals (both whereas those at moderate risk showed positive
male and female) from HBOC families did not family histories that did not meet these high-risk
reflect adverse psychological effects.53 How- criteria. To predict the number of breast cancers
ever, with respect to screening, we did find that expected in these two groups had prophylactic
rates of adherence to mammography recom- mastectomies not been performed, the
mendations among mutation carriers was not researchers used a nested-sister control study
increased. It was also noted that carriers of for the high-risk group and the Gail Model for
deleterious genes who said they would consider the moderate-risk group. Their findings were
prophylactic surgery nevertheless showed low based upon 693 women with a family history of
rates of actually adopting such options. How- breast cancer (214 high-risk and 425 moderate-
ever, these observations are based upon short- risk) who had bilateral prophylactic mastec-
term experience; longer-term data will be tomies. Their median follow-up was 14.4 years,
required to determine how often women may while the median age at prophylactic mastec-
opt for prophylactic surgery.53 tomy was 43 (mean 42.4) years. The Gail Model
prediction for breast cancer occurrence
PROPHYLACTIC MASTECTOMY expected in the moderate-risk group was 37.4.
However, only four breast cancers occurred fol-
In a landmark study, Hartmann and colleagues54 lowing prophylactic mastectomy in this group
pursued the efficacy of prophylactic mastec- (89.5% reduction, p < .001).
14 BREAST CANCER

Breast cancer occurrences in the 214 high- tomy more limited gains for young women
risk probands were compared to their sisters with BRCA1 or BRCA2 mutations.”
who had not undergone prophylactic mastec-
tomy. These 214 probands had a total of 403 CONSERVATIVE VERSUS
sisters. Of keen interest was the finding that CONVENTIONAL SURGERY IN
there have been 156 breast cancers in the sis- HEREDITARY BREAST CANCER
ters; 115 occurred before the sister proband’s
mastectomy, 38 after the sister proband’s pro- Should a patient with HBOC, particularly one
phylactic mastectomy, and 3 with date who is harboring a BRCA1 or BRCA2 germ-
unknown. In comparison, 3 of the 214 probands line mutation, be managed differently from a
had developed breast cancer. This represents a patient with the more common sporadic form
> 90 percent reduction in the incidence of of this disease? We have taken the position
breast cancer with current follow-up. Breast that, because of the early age of breast cancer
cancer mortality was also reduced significantly onset and excess lifetime risk for bilaterality,
in both the high and moderate-risk groups. coupled with the potential deficiency of repair
The investigators concluded that prophy- of radiation-induced DNA damage,44 the
lactic mastectomy resulted in a significant patient should be given the option of total mas-
reduction in the incidence of and mortality tectomy as opposed to conservative (“lumpec-
from breast cancer among these women with tomy”) management, and seriously consider
positive family histories of breast cancer. contralateral prophylactic mastectomy, assum-
This information will be useful for genetic ing that the ipsilateral breast cancer is likely to
counseling.54 have adequate control.
Schrag and colleagues55 discuss the deci-
sion analysis involved in prophylactic mastec- POTENTIAL FOR TARGETED
tomy and oophorectomy and life expectancy BRCA1/BRCA2 MUTATION THERAPY
outcome among patients with BRCA1 and
BRCA2 germ-line mutations. They found that, In addition to identifying cancer risk status
on average, a 30-year-old woman harboring through mutations such as BRCA1 and
such a mutation would gain from 2.9 to 5.3 BRCA2, this knowledge has the potential to
years of life expectancy from prophylactic provide individualized highly-targeted molec-
mastectomy and from 0.3 to 1.7 years from ular genetic therapies based upon mutation
prophylactic oophorectomy. These findings discoveries.56 Specifically, once the functions
were dependent upon their cumulative risk of of cancer susceptibility genes have been iden-
cancer. Gains in life expectancy also would tified, knowledge as to how such gene-deter-
decline with age at the time of prophylactic mined biochemical functions can be employed
surgery. They would be minimal for a 60-year- for targeted radiation and chemotherapy
old woman. Importantly, in women aged 30, an should emerge.
oophorectomy may be delayed for 10 years Abbott and colleagues57 examined the pro-
with minimal loss of life expectancy. This tein product of the BRCA2 gene in terms of its
would allow women to complete their families. having an important role in mediating repair of
These investigators concluded that “On the double-strand breaks in DNA. They identified a
basis of a range of estimates of the incidence human pancreatic carcinoma cell line which
of cancer, prognosis, and efficacy of prophy- lacked one copy of the BRCA2 gene and con-
lactic surgery, our model suggests that prophy- tained a mutation (617delT) in the remaining
lactic mastectomy provides substantial gains in copy.58 They performed in vitro and in vivo
life expectancy and prophylactic oophorec- experiments in this cell line as well as with other
Genetics, Natural History, and DNA-Based Genetic Counseling in Hereditary Breast Cancer 15

carefully matched cell lines. They then exam- taught how to perform this procedure and
ined double-strand break repair with attention demonstrates proficiency in doing so on return
given to sensitivity to drugs and radiation effect medical visits. When patients reach the age of
that induce double-strand breaks. Their findings 20 years, we begin semiannual breast examina-
disclose that “... BRCA2-defective cells are tion by the physician and at age 25 initiate
unable to repair the double-strand DNA breaks annual mammography.
induced by ionizing radiation. These cells were With respect to ovarian cancer, we discuss
also markedly sensitive to mitoxantrone, transvaginal ovarian ultrasound, Doppler color
amsacrine, and etoposide... (two-sided p = .002) bloodflow imaging, and CA-125, with their lim-
and to ionizing radiation (two-sided p = .001). itations, and initiate this at age 30 and perform
Introduction of antisense BRCA2 deoxyribonu- it annually. The option of prophylactic bilateral
cleotides into cells possessing normal BRCA2 oophorectomy is also discussed. If the patient is
function led to increased sensitivity to mitox- interested in this option and has completed her
antrone (two-sided p = .008). Tumors formed by child bearing, the oophorectomy can be per-
injection of BRCA2-defective cells into nude formed between the ages of 35 and 45 years.
mice were highly sensitive (> 90% tumor size
reduction, two-sided p = .002) to both ionizing CONCLUSION
radiation and mitoxantrone when compared
with tumors exhibiting normal BRCA2 func- It is necessary to keep an open mind about the
tion.” Abbott and colleagues concluded that pros and cons of DNA testing and genetic coun-
these BRCA2-defective cancer cells were highly seling and its translation into medical practice by
sensitive to agents that contribute to double- the basic scientist, medical and molecular
strand breaks in DNA. geneticist, physician, genetic counselor, and
ethicist. For example, how does one interpret
SURVEILLANCE AND MANAGEMENT some of the ethical positions of today suggesting
FOR HEREDITARY BREAST CANCER that genetic testing be limited to a research set-
ting or even curtailed until specific benefit of
When the diagnosis of a hereditary breast can- such DNA testing can be more fully established?
cer-prone syndrome has been established, the Prodigious advances in science and technol-
surveillance and management strategies are then ogy (ie, better surveillance, more effective surgi-
melded to the natural history of the particular cal management including data supporting pro-
HBC syndrome. We recommend that patients phylactic surgery, and improved molecular
receive intensive education regarding the natural genetic technology with lower cost for germ-line
history, genetic risk, and availability of DNA mutation discovery) may resolve some of the
testing such as BRCA1, BRCA2, or p53, depend- concerns about molecular genetic testing that
ing upon the hereditary breast cancer syndrome cause certain physicians, basic scientists, and
of concern. We initiate such education between ethicists to believe it should be limited. How
the ages of 15 and 18 years but do not perform does one educate these colleagues about newly
any DNA testing until the patient is > 18 years emerging benefits of molecular genetic testing
of age and has given informed consent. which could prove to be lifesaving?
When patients are 18 years old, the authors Molecular genetic advances are occurring at
provide instruction in breast self examination such a rapid pace that it is exceedingly difficult
(BSE) with physician assessment of their per- to keep physicians fully informed of this
formance. Although the effectiveness of BSE progress. For example, it is predicted that the
has been controversial, the authors are con- entire human genome will be identified by the
vinced that it can be effective if the woman is year 2003.
16 BREAST CANCER

The lay press strives to keep the public fully 9. Miki Y, Swensen J, Shattuck-Eidens D, et al. A
informed about the impact new gene discover- strong candidate for the breast and ovarian can-
ies may have on patients and their close rela- cer susceptibility gene BRCA1. Science 1994;
266:66–71.
tives. Unfortunately, certain members of the 10. Wooster R, Neuhausen SL, Mangion J, et al. Local-
media have overinterpreted the benefit of ization of a breast cancer susceptibility gene,
germ-line testing and have not fully dealt with BRCA2, to chromosome 13q12–13. Science
some of its drawbacks. In turn, some molecular 1994;265:2088–90.
genetic laboratories have made testing appear 11. Wooster R, Bignell G, Lancaster J, et al. Identifi-
to be the panacea for cancer control. Some may cation of the breast cancer susceptibility gene
BRCA2. Nature 1995;378:789–92.
offer molecular testing without sufficient evi-
12. Easton DF, Bishop DT, Ford D, Crockford GP, The
dence that the family of concern merits testing. Breast Cancer Linkage Consortium. Genetic
Genetic counseling may not be provided to linkage analysis in familial breast and ovarian
those being tested. In spite of these misgivings, cancer: results from 214 families. Am J Hum
we believe that hereditary breast cancer Genet 1993;52:678–701.
patients, when properly counseled and DNA 13. Easton DF, Ford D, Bishop DT, The Breast Cancer
tested, will benefit immensely during this excit- Linkage Consortium. Breast and ovarian can-
cer incidence in BRCA1 mutation carriers. Am
ing era of molecular genetics.
J Hum Genet 1995;56:265–71.
Patients must be encouraged to meticulously 14. Struewing JP, Hartge P, Wacholder S, et al. The
examine their family histories of cancer. Should risk of cancer associated with specific muta-
they be found to harbor a germ-line cancer pre- tions of BRCA1 and BRCA2 among Ashkenazi
disposing mutation, this knowledge may be used Jews. N Engl J Med 1997;336:1401–8.
to encourage screening and detect cancer at an 15. Claus EB, Schildkraut J, Iversen ES Jr, Berry D,
Parmigiani G. Effect of BRCA1 and BRCA2 on
early stage so that a cure might be possible and/or
the association between breast cancer risk and
cancer prevented through the option of prophy- family history. J Natl Cancer Inst 1998;90:
lactic surgery. 1824–9.
16. Lynch HT, Follett KL, Lynch PM, et al. Family
REFERENCES history in an oncology clinic. Implications for
1. Vogelstein B, Kinzler KW. The genetic basis of cancer genetics. JAMA. 1979;242:1268–72.
human cancer. New York: McGraw-Hill; 1998. 17. David KL, Steiner-Grossman P. The potential use
2. Landis SH, Murray T, Bolden S, Wingo PA. Cancer of tumor registry data in the recognition and
statistics, 1999. CA Cancer J Clin 1999;49:8–31. prevention of hereditary and familial cancer.
3. Lynch HT, Lemon SJ, Durham C, et al. A descrip- NY State J Med 1991;91:150–2.
tive study of BRCA1 testing and reactions to dis- 18. Lynch HT, Lemon SJ, Marcus JN, et al. Breast
closure of test results. Cancer 1997;79:2219–28. cancer genetics: heterogeneity, molecular
4. Lynch HT, Krush AJ. Carcinoma of the breast and genetics, syndrome diagnosis, and genetic
ovary in three families. Surg Gynecol Obstet counseling. In: Bland KI, Copeland EMI, edi-
1971;133:644–8. tors. The breast: comprehensive management
5. Lynch HT, Krush AJ, Lemon HM, et al. Tumor of benign and malignant diseases. 2 ed.
variation in families with breast cancer. JAMA Philadelphia (PA): W. B. Saunders Company;
1972;222:1631–5. 1998. p. 370–94.
6. Lynch HT, Guirgis HA, Albert S, et al. Familial 19. Gayther SA, Warren W, Mazoyer S, et al.
association of carcinoma of the breast and Germline mutations of the BRCA1 gene in
ovary. Surg Gynecol Obstet 1974;138:717–24. breast and ovarian cancer families provide evi-
7. Hall JM, Lee MK, Newman B, et al. Linkage of dence for a genotype-phenotype correlation.
early-onset breast cancer to chromosome Nat Genet 1995;11: 428–33.
17q21. Science 1990;250:1684–9. 20. Serova O, Montagna M, Torchard D, et al. A high
8. Narod SA, Feunteun J, Lynch HT, et al. Familial incidence of BRCA1 mutations in 20 breast-
breast-ovarian cancer locus on chromosome ovarian cancer families. Am J Hum Genet
17q12–q23. Lancet 1991;388:82–3. 1996;58:42–51.
Genetics, Natural History, and DNA-Based Genetic Counseling in Hereditary Breast Cancer 17

21. Gayther SA, Mangion J, Russell P, et al. Variation 35. Rao VN, Shao N, Ahmad M, Reddy ESP. Anti-
of risks of breast and ovarian cancer associated sense RNA to the putative tumor suppressor
with different germline mutations of the gene BRCA1 transforms mouse fibroblasts.
BRCA2 gene. Nat Genet 1997;15:103–5. Oncogene 1996;12:523–8.
22. Ramus SJ, Friedman LS, Gayther SA, Ponder 36. Holt JT, Thompson ME, Szabo CI, et al. Growth
BAJ. A breast/ovarian cancer patient with retardation and tumour inhibition by BRCA1.
germline mutations in both BRCA1 and Nat Genet 1996;12:298–302.
BRCA2. Nat Genet 1997;15:14–15. 37. Armes JE, Egan AJM, Southey MC, et al. The his-
23. Tonin P, Moslehi R, Green R, et al. Linkage analy- tologic phenotypes of breast carcinoma occur-
sis of 26 Canadian breast and breast-ovarian ring before age 40 years in women with and
cancer families. Hum Genet 1995;95:545–50. without BRCA1 or BRCA2 germline mutations:
24. Neuhausen S, Gilewski T, Norton L, et al. Recur- a population-based study. Cancer 1998;83:
rent BRCA2 617delT mutations in Ashkenazi 2335–45.
Jewish women affected by breast cancer. Nat 38. Collins N, McManus R, Wooster R, et al. Consistent
Genet 1996;13:126–8. loss of the wild type allele in breast cancers from
25. Couch FJ, Farid LM, DeShano ML, et al. BRCA2 a family linked to the BRCA2 gene on chromo-
germline mutations in male breast cancer cases some 12q12–13. Oncogene 1995;10:1673–5.
and breast cancer families. Nat Genet 1996;13: 39. Sigurdsson H, Agnarsson BA, Jonasson JG, et al.
123–5. Worse survival among breast cancer patients in
26. Liede A, Metcalfe K, Offit K, et al. A family with families carrying the BRCA2 susceptibility
three germline mutations in BRCA1 and gene. [abstract] Breast Cancer Res Treat 1996;
BRCA2. Clin Genet 1998;54:215–8.
37:33.
27. Struewing JP, Abeliovich D, Peretz T, et al. The car-
40. Breast Cancer Linkage Consortium. Pathology of
rier frequency of the BRCA1 185delAG mutation
familial breast cancer: differences between
is approximately 1 percent in Ashkenazi Jewish
breast cancers in carriers of BRCA1 or BRCA2
individuals. Nat Genet 1995;11:198–200.
mutations and sporadic cases. Lancet 1997;
28. Roa BB, Boyd AA, Volcik K, Richards CS.
349:1505–10.
Ashkenazi Jewish population frequencies for
41. Lakhani SR, Jacquemier J, Sloane JP, et al. Multi-
common mutations in BRCA1 and BRCA2. Nat
factorial analysis of differences between spo-
Genet 1996;14:185–7.
radic breast cancers and cancers involving
29. Oddoux C, Struewing JP, Clayton CM, et al. The
BRCA1 and BRCA2 mutations. J Natl Cancer
carrier frequency of the BRCA2 6174delT muta-
tion among Ashkenazi Jewish individuals is Inst 1998;90:1138–45.
approximately 1%. Nat Genet 1996;14:188–90. 42. Eng C, Li FP, Abramson DH. Mortality from sec-
30. Marcus JN, Watson P, Page DL, et al. Hereditary ond tumors among long-term survivors of
breast cancer: pathobiology, prognosis, and retinoblastoma. J Natl Cancer Inst 1993;85:
BRCA1 and BRCA2 gene linkage. Cancer 1996; 1121–6.
77:697–709. 43. Ford D, Easton DF, Peto J. Estimates of the gene
31. Marcus JN, Page DL, Watson P, et al. BRCA1 and frequency of BRCA1 and its contribution to
BRCA2 hereditary breast carcinoma pheno- breast and ovarian cancer incidence. Am J
types. Cancer 1997;80 Suppl:543–56. Hum Genet 1995;57:1457–62.
32. Marcus JN, Watson P, Page DL, et al. BRCA2 44. Scully R, Chen J, Plug A, et al. Association of
hereditary breast cancer phenotype. Breast BRCA1 with Rad51 in mitotic and meiotic
Cancer Res Treat 1997;44:275–7. cells. Cell 1997;88:265–75.
33. Marcus JN, Watson P, Page DL, Lynch HT. The 45. Chabner E, Nixon A, Gelman R, et al. Family his-
pathology and heredity of breast cancer in tory and treatment outcome in young women
younger women. J Natl Cancer Inst Monogr after breast-conserving surgery and radiation
1994;16:23–34. therapy for early-stage breast cancer. J Clin
34. Thompson ME, Jensen RA, Obermiler PS, et al. Oncol 1998;16:2045–51.
Decreased expression of BRCA1 accelerates 46. Lim DS, Hasty P. A mutation in mouse Rad51
growth and is often present during sporadic results in early embryonic lethal that is sup-
breast cancer progression. Nat Genet 1995;9: pressed by a mutation in p53. Mol Cell Biol
444–50. 1996;16:7133–43.
18 BREAST CANCER

47. Watson P, Lynch HT. Extracolonic cancer in testing in families with hereditary breast-
hereditary nonpolyposis colorectal cancer. ovarian cancer: a prospective study of patient
Cancer 1993;71:677–85. decision-making and outcomes. JAMA 1996;
48. Lynch HT, Casey MJ, Lynch J, et al. Genetics and 275:1885–92.
ovarian carcinoma. Semin Oncol 1998;25: 54. Hartmann LC, Schaid DJ, Woods JE, et al. Effi-
265–81. cacy of bilateral prophylactic mastectomy in
49. American Society of Clinical Oncology. State- women with a family history of breast cancer.
ment of the American Society of Clinical N Engl J Med 1999;340:77–84.
Oncology: genetic testing for cancer suscepti- 55. Schrag D, Kuntz KM, Garber JE, Weeks JC. Deci-
bility. J Clin Oncol 1996;14:1730–6. sion analysis—effects of prophylactic mastec-
50. Lynch HT, Watson P, Tinley S, et al. An update on tomy and oophorectomy on life expectancy
DNA-based BRCA1/BRCA2 genetic counsel- among women with BRCA1 or BRCA2 muta-
ing in hereditary breast cancer. Cancer Genet tions. N Engl J Med 1997;336:1465–71.
Cytogenet 1999;109:91–8. 56. Livingston DM. Genetics is coming to oncology.
51. Fleiss JL. Statistical methods for rates and pro- JAMA 1997;277:1476–7.
portions. 2nd ed. New York: John Wiley & 57. Abbott DW, Freeman ML, Holt JT. Double-strand
Sons; 1981. break repair deficiency and radiation sensitiv-
52. Lerman C, Hughes C, Lemon SJ, et al. What you ity in BRCA2 mutant cancer cells. J Natl Can-
don’t know can hurt you: adverse psychologic cer Inst 1998;90:978–85.
effects in members of BRCA1–linked and 58. Goggins M, Schutte M, Lu J, et al. Germline
BRCA2-linked families who decline genetic BRCA2 gene mutations in patients with appar-
testing. J Clin Oncol 1998;16:1650–4. ently sporadic pancreatic carcinomas. Cancer
53. Lerman C, Narod S, Schulman K, et al. BRCA1 Res 1996; 56:5360–4.
2
Breast Cancer Risk and Management:
Chemoprevention, Surgery,
and Surveillance
MASSIMO CRISTOFANILLI, MD
LISA NEWMAN, MD, FACS
GABRIEL N. HORTOBAGYI, MD

In the past decade, the systematic use of mam- The development of effective chemopreven-
mography as part of diagnostic screening pro- tion strategies requires a systematic approach
grams and the extensive use of stereotactic focusing on multiple investigational aspects of
fine-needle biopsy techniques have greatly the problem including (1) a clear description of
improved our ability to detect pre-invasive as the specific risk factors that may be used in
well as microinvasive breast carcinomas. The selecting cohorts of women at increased risk and
consequent earlier detection of breast lesions is as end points for chemoprevention studies; (2)
considered the most important factor explain- the identification and preclinical evaluation of
ing the recent decline in overall mortality from cancer chemoprevention agents and subsequent
breast cancer observed in the United States.1 development of definitive, large-scale clinical tri-
This progress has translated into many efforts als; and (3) the identification and characteriza-
to gain insight into the biologic mechanisms tion of specific molecular biomarkers that may
responsible for cancer development and pro- be quantitatively assessed and used as surrogate
gression and to identify potential areas of inter- end-point biomarkers (SEBs) in future trials.
vention for prevention studies.2
About 20 percent of women diagnosed with BREAST CANCER RISK ASSESSMENT
proliferative breast disease display atypical
hyperplasia, a condition associated with increas- Chemoprevention is traditionally defined as the
ed risk of breast cancer and probably a precursor inhibition or reversal of carcinogenesis, before
to the disease.3 The recognition of the importance overt malignancy, by intervention with chemi-
of these benign breast lesions as risk factors and cal agents. Most breast cancer chemopreventive
the identification of genetic alterations (BRCA1, studies are conducted within cohorts of women
BRCA2, p53) associated with genetic predisposi- considered at high risk of the disease.7 In the
tion to breast cancer have spurred investigation context of chemoprevention investigations, the
in the areas of prophylactic surgery and chemo- most important cancer risk factors are consid-
prevention in the management of women at high ered to be those that can be measured quantita-
risk of breast cancer and directed attention to tively in the subject at risk. These factors are
specific interventions and to the design of com- called risk biomarkers, and they can be used to
prehensive surveillance programs.4–6 identify cohorts for chemoprevention trials.8

19
20 BREAST CANCER

Generally, the risk biomarkers are grouped models, which consider a variety of risk factors
in the following categories: (1) genetic predis- and project a cumulative risk for the develop-
position, (2) carcinogen exposure, (3) carcino- ment of disease over a finite period of time.
gen effect/exposure, (4) previous cancers, and Among the various models proposed, the model
(5) intermediate biomarkers. In some cases, developed by Gail and colleagues is probably the
risk biomarkers that are measurable and may most widely accepted.12 Proposed in the original
undergo selective modulation by chemopreven- form in 1989 and subsequently modified, this
tive agents can be used as SEBs in clinical model was designed to calculate the absolute
chemoprevention studies.9 Today, there is not a breast cancer risk in women on the basis of the
single ideal risk biomarker or SEB for breast data from the Breast Cancer Detection Demon-
cancer, and the selection of high-risk subjects stration Project (BCDDP) that recruited 280,000
for breast cancer chemoprevention studies is women in 28 centers in the United States in the
generally done on the basis of the presence of mid-1970s. It represented an attempt to define
proliferative atypical breast disease or epidemi- the contribution of accumulated breast cancer
ologic and genetic factors known to increase a risk from a number of risk factors combined in a
woman’s risk of developing breast cancer.10,11 multivariate logistic regression model. The vari-
The quantification of the risk of developing ables identified included age of the patient, num-
cancer is usually estimated from epidemiologic ber of first-degree relatives with breast cancer,

Table 2–1. RISK FACTORS CONSIDERED FOR THE CALCULATION OF ABSOLUTE BREAST CANCER RISK

Risk Factor Associated Relative Risk Expansion Variables

Age at menarche (y)


³ 14 1.000
12 to 13 1.099 A
< 12 1.207
No. of breast biopsies
Age < 50 y
0 (0) 1.000
1 (1) 1.698
³ 2 (2) 2.882
Age ³ 50 y B
0 (0) 1.000
1 (1) 1.273
³ 2 (2) 1.620
Age at first-term live birth (y) No. of first-degree relatives
with breast cancer
< 20 0 (0) 1.000
1 (1) 2.607
³ 2 (2) 6.798
20 to 24 0 (0) 1.244
1 (1) 2.681
³ 2 (2) 5.775
C
25 to 29 or nulliparous 0 (0) 1.548
1 (1) 2.756
³ 2 (2) 4.907
³ 30 0 (0) 1.927
1 (1) 2.834
³ 2 (2) 4.169
Absolute risk = A ´ B ´ C ´ 1.82 *
A ´ B ´ C ´ 0.93†
*Presence of atypical hyperplasia in biopsies.

No atypical hyperplasia.
Adapted from Gail MH, Brinton LA, Byar DP, et al. Projecting individualized possibilities of developing breast cancer for white females who are
being examined annually. J Natl Cancer Inst 1989;81:1879–86.
Breast Cancer Risk and Management: Chemoprevention, Surgery, and Surveillance 21

nulliparity or age at first live birth, number of The BRCA1 gene, a breast cancer susceptibil-
breast biopsies, presence or absence of atypical ity gene localized to chromosome 17q, was
hyperplasia, and age at menarche (Table 2–1). described for the first time in 1990. It is a tumor-
The individualized absolute risk can be esti- suppressor gene postulated to be important in
mated by combining the relative risk based on regulating the growth of breast epithelial cells
the woman’s age and individual risk factors and and in the process of deoxyribonucleic acid
then combining this information with an esti- (DNA) damage repair.17 The breast cancer sus-
mate of the baseline hazard rate for a woman ceptibility gene BRCA1 accounts for 45 percent
with no risk factors.12,13 In particular, women of hereditary cases of breast cancer and 80 to 90
with a strong family history of breast cancer (2 percent of hereditary cases of combined breast
first-degree relatives affected, or 1 first-degree and ovarian cancers. The BRCA1 mutations are
and 2 second-degree relatives affected) or of found in approximately 7 to 12 percent of women
breast and ovarian cancers (3 affected relatives with breast cancer of early onset. Mutations have
with breast cancer and 1 relative with ovarian been described as spreading evenly across the
cancer) have a 25 percent or greater lifetime risk entire gene. The most commonly described muta-
of developing breast cancer. A computer pro- tion is a specific alteration causing a deletion of
gram called RISK has been subsequently devel- adenine and guanine (185delAG). This mutation
oped and allows physicians to calculate a is present in 1 percent of the Ashkenazi Jewish
woman’s absolute risk to develop breast cancer population and contributes to a risk of breast can-
with 95 percent confidence interval bounds.14 cer as high as 21 percent among Jewish women.
Women who carry a BRCA1 mutation were ini-
Familial and Genetic Factors tially estimated to have an 87 percent lifetime risk
of breast cancer and a 44 percent lifetime risk of
Identification of cohorts at genetic risk for can- ovarian cancer.18 Subsequent studies suggest
cer is an appealing concept because it may offer risks of 56 percent and 16 percent, respectively.
the opportunity to explore the steps in breast More recent data from population-based
carcinogenesis, from the inheritance of a pre- studies suggest that BRCA1 mutations account
disposing mutation through the development of for only 10 to 20 percent of inherited breast
preinvasive lesions or overt malignancy. How- cancer, and BRCA2 mutations are responsible
ever, germline genetic changes are rare and for half this fraction. The discrepancy between
reported in only a small proportion of women estimates from early studies is probably related
who will eventually develop breast cancer.15 to the fact that initial data were derived from
Breast cancer attributed to a family history linkage analysis that probably tends to overesti-
of the disease has been reported to account for 6 mate the true incidence of BRCA1 and BRCA2
to 19 percent of all cases of breast cancer. mutations in hereditary breast cancer.19–21
Hereditary breast cancers, which constitute a The breast cancer susceptibility gene
proportion of these cases, are characterized by BRCA2, localized to chromosome 13q, was
early onset, a high incidence of bilateral disease, described in 1994. Linkage studies suggest that
association with other malignancies, and auto- 35 percent of high-risk families may have
somal dominant inheritance. Genetic-linkage BRCA2 mutations.22 Male breast cancer, a rare
studies of families with multiple members with condition that represents less than 1 percent of
breast cancer have allowed major improvements all cancer and 0.1 percent of cancer-related
in our understanding of the genetic alterations deaths in men, has been found to be associated
associated with hereditary predisposition. Such with mutations in the BRCA2 gene.23
studies led to the discovery of germline muta- Familial clustering of breast cancer has also
tions in the BRCA1 and BRCA2 genes.16 been described in families diagnosed with Cow-
22 BREAST CANCER

den syndrome and Li-Fraumeni syndrome, and cer. A latency period of at least 10 years is usu-
more recently, ataxia-telangiectasia.24,25 Cow- ally required in the majority of the patients; a
den syndrome involves multiple hamartomatous shorter latency period (5 to 10 years) has been
lesions, especially of the skin, and mucous reported occasionally.31
membranes, and carcinoma of the breast and These observational studies suggest that the
thyroid. Li-Fraumeni syndrome, associated with carcinogenic process is related to two factors:
a high incidence of p53 mutations, consists of a the dose of radiation delivered and the person’s
familial aggregation of breast carcinomas, soft age at the time of exposure. In general, young
tissue sarcomas, brain tumors, osteosarcomas, women (less than 30 years of age) represent a
leukemias, and adrenocortical carcinomas. cohort with a higher relative risk compared
Hereditary breast cancer syndromes are with the risk for other age groups. The cluster-
clinically relevant because they raise the possi- ing of breast carcinoma in women irradiated for
bility of effective identification, through Hodgkin’s disease at an early age is probably
genetic testing, of patients at high risk and related to the increased sensitivity of the
optimal quantification of the risk. In view of incompletely differentiated breast epithelium to
the high risk of developing an invasive breast the carcinogenic action of radiation.33
cancer during their lifetime and in considera- The high risk of breast carcinoma in young
tion of the imperfect nature of early detection, women with Hodgkin’s disease treated with
patients with hereditary breast cancer syn- irradiation mandates regular follow-up with
dromes constitute an ideal target for chemo- breast examination and yearly mammography
prevention studies. starting within 8 years of completion of the
radiation treatment.37
Environmental Radiation Exposure The hypothetical risk of breast cancer
derived from prolonged screening needs to be
Epidemiologic observations suggest that expo- mentioned. Using a risk estimate provided by
sure of breast tissue to ionizing radiation is asso- the Biological Effects of Ionizing Radiation
ciated with an increased risk of breast cancer. In (BEIR) V Report of the National Academy of
particular, an increased risk of breast carcinoma Sciences and a mean breast glandular dose of
has been clearly documented in young women 4 mGy from bilateral mammography, with two
that have survived atomic bombs, in patients views per breast, one can estimate that annual
who have undergone repeated fluoroscopies (eg, mammography of 100,000 women for 10 con-
in patients with tuberculosis), and in patients secutive years beginning at age 40 years will
treated with radiation for postpartum mastitis, result in, at most, 8 breast cancer deaths during
thymic enlargement, and Hodgkin’s disease.26–37 their lifetime. On the other hand, researchers
Treatment-associated second neoplasms have have shown a 24 percent mortality reduction
emerged as a major complication in patients from biennial screening of women in this age
cured of Hodgkin’s disease. Sporadic cases of group; this will result in one life lost. An
breast carcinoma after mantle irradiation for assumed mortality reduction of 36 percent from
Hodgkin’s disease were reported beginning in annual screening would result in 36.5 lives
1978, by which time clinical data on a large saved per life lost and 91.3 years of life saved
cohort of women treated and cured for their dis- per year of life lost. Thus, the theoretic radia-
ease in the 1960s had become available.25 tion risk from screening mammography is
Subsequent reports directed attention to the extremely small compared with the established
carcinogenic risk associated with radiation benefit from this life-saving procedure and
exposure and the latency time between expo- should not unduly distract women under age 40
sure and clinical manifestations of breast can- years who are considering screening.38
Breast Cancer Risk and Management: Chemoprevention, Surgery, and Surveillance 23

Intermediate Biomarkers prevention studies, the main advantage being


the possibility of a more objective assessment
A key element in the control and prevention of of the effectiveness of interventions through
invasive breast cancer is the recognition of early, repeated core biopsies.
preclinical changes with or without associated In recent years, the availability of sensitive
characteristic molecular abnormalities that may assays of DNA damage and genetic instability
identify a woman as being at high risk for devel- have prompted novel investigations and may help
opment of the disease.39 These alterations, called define subsets of women who are at high risk of
intermediate biomarkers, may be used as SEBs developing breast cancer. Among the various mol-
and provide a more cost-effective and rapid ecular markers investigated, p53 overexpression,
means of testing chemopreventive interventions. dysplasia, and aneuploidy have been found to be
To date, the most specific intermediate biomark- associated with increased risk of invasive breast
ers for invasive breast cancer development are carcinoma.43,44 Rohan and colleagues conducted a
precancerous and preinvasive lesions, such as case-control study within a cohort of 4,888 Cana-
histologic changes of atypical hyperplasia, lobu- dian women in the National Breast Screening
lar carcinoma in situ (LCIS), and ductal carci- Study (NBSS) who were diagnosed with benign
noma in situ (DCIS). Observations about the breast disease. Case subjects were the women in
value of molecular alterations, for example, whom breast cancer subsequently developed. The
c-erbB-2 overexpression and p53 accumulation, c-erbB-2 protein overexpression and p53 accu-
as markers of disease progression and increased mulation were determined by immunohisto-
risk in patients with benign breast conditions are chemical techniques. Accumulation of p53 was
contradictory. 40 associated with an increased risk of breast cancer
Intermediate biomarkers are essentially pre- (adjusted odds ratio 2.5), while c-erbB-2 protein
cancerous lesions identified as being directly overexpression was not.44 Even with multiple
on the control pathway to cancer. Their pres- methodologic problems, this investigation pro-
ence puts carriers at high risk for invasive dis- vides an interesting model for the clinical use of
ease. A hypothetical model of intraepithelial biomarkers in benign breast disease.
breast neoplasms postulates progression from The design of prospective trials that includes
focal aberrant ductal or lobular proliferation the evaluation of p53 status along with other
(hyperplasia) to cellular pleomorphism, disor- markers studied in randomly obtained fine-
ganized growth, and abnormal growth (dyspla- needle aspirates may provide a unique opportu-
sia), and, finally, focally invasive cancer.41 nity to identify specific, measurable, intermedi-
The histologic abnormalities, described by ate biomarkers that may be used in short-term
Page and colleagues3 and collectively known as trials to verify the efficacy of chemopreventive
proliferative breast disease (PBD), are associated agents.
with increased risk of breast cancer; the lesions
and associated risks of cancer development vary
ESTROGENS AND
from moderate hyperplasia (two-fold risk), to
BREAST CANCER RISK
atypical ductal hyperplasia (four-fold risk), to
carcinoma in situ (CIS) (11-fold risk).3,4,42
The presence of these lesions in the con- Endogenous Hormones
tralateral breast of patients with a history of The controversy surrounding exogenous hor-
breast cancer has been associated with up to a mone use and breast cancer risk is predicated
0.8 percent chance per year of developing a on the concept that breast carcinogenesis is a
new primary tumor. In this context, these hormone-dependent process. The ability to
lesions appear to be the ideal target for chemo- control breast cancer with hormonal manipula-
24 BREAST CANCER

tion has been recognized since 1986, when menopausal woman, androstenedione, synthe-
Beatson reported on oophorectomy as a suc- sized in the adrenal gland, is the principal estro-
cessful treatment for this disease.45 Since that gen precursor following the decline of ovarian
time several other epidemiologic, experimental function. Increased conversion of androstened-
and clinical lines of evidence have developed ione to estrone by fat cells that results in elevated
that also support this concept.46 levels of this predominant postmenopausal
It is well established that menstrual factors estrogen is reputed to be the underlying explana-
resulting in exposure of the breast to increased tion for the increased risk of breast cancer seen
numbers of ovulatory estrogen cycles over a in obese postmenopausal women.47–52 In con-
lifetime, such as early menarche (< 13 years), trast, in premenopausal obese women, derange-
late menopause (> 50 years), and nulliparity ment of the estrogen-progesterone balance and
can increase the risk of breast cancer.46,47 Con- subsequent menstrual disturbances result in a
versely, bilateral oophorectomy at a young age decreased risk of breast cancer.50,53
and interruptions of the menstrual cycle in the Male breast cancer is also likely to be related
form of multiple pregnancies may confer a pro- to factors resulting in abnormalities of estrogen
tective effect.48 metabolism, such as liver disease or genetic
The impact of pregnancy on the risk of defects such as Klinefelter’s syndrome.53–55
breast cancer is strongest in the case of the
first pregnancy occurring at a young age Exogenous Hormones
(before 20 years). The rate of proliferation of
the ductal epithelium is normally high after
Oral Contraceptives
puberty. The hormonal influences associated
with pregnancy induce a process of terminal Oral contraceptives (OCs) have been marketed
ductal and lobular stem cell differentiation, extensively over the past 30 to 40 years; world-
theoretically rendering the breast more resis- wide users are estimated to be over 200 million
tant to carcinogenesis.47,48 Henderson and col- women, and in the United States, it is projected
leagues hypothesized that completion of a that approximately 80 percent of all women
full-term pregnancy is crucial for this protec- will have used OCs by the age of 40 years.48
tive effect because the rapid increase in free Studies evaluating a possible association
estradiol during the first trimester of preg- between OCs and breast cancer risk have been
nancy is “equivalent to several ovulatory hampered by changes in the composition of
cycles over a relatively short period of time.” OCs over time and by individual patient varia-
They hypothesized that failure to over-ride this tion in the duration of use.56–64
estrogenic surge with the subsequent hormonal Early evidence that OCs could significantly
changes of advanced pregnancy (as occurs increase the risk of breast cancer was reported
with first-trimester abortions) can result in by Pike and colleagues in 1983.56 In their case-
increased risk of breast cancer.49 control study of 314 breast cancer patients <
It is also well established that estrogen and 37 years of age and 314 matched controls, the
progesterone exert proliferative effects on use of OCs with a relatively high progesterone
human breast tissue49,50 and that estrogen can content for more than 6 years and starting use
promote mammary tumorigenesis in animal of OCs before age 25 years were associated
models as well as in in vitro tissue cultures.47–51 with a relative risk of 4.9 for breast cancer
Postmenopausal obesity has been associated development. Since that time, many studies
with increased breast cancer risk, and this rela- have been conducted in the United States
tionship appears to be mediated by age-related attempting to quantify the level of risk of
variations in estrogen metabolism. In the post- breast cancer conferred by the use of OCs. The
Breast Cancer Risk and Management: Chemoprevention, Surgery, and Surveillance 25

results of the largest studies are summarized in Table 2–2. SUMMARY OF RANDOMIZED STUDIES
EVALUATING THE RISK ASSOCIATED WITH THE USE
Table 2–2. Most studies56–61,63 have been case- OF ORAL CONTRACEPTIVES (OCs)
control studies, in which the rates of use of
Number of Age Years Relative
OCs among groups of breast cancer patients Study Patients (y) Use Risk
were compared with the rates of use of OCs
Pike56 314P < 37 ³6 4.9
among matched groups of noncancer patients.
314C
The results are relatively inconsistent, with CASH57 689P < 55 >4 1.1
some studies demonstrating an increased risk 1,077C
Stadel58 2,088P < 45 >4 1.1
of breast cancer associated with use of OCs, 2,065C
whereas others demonstrate a protective effect Miller59 521P < 45 3–4 0.8
521C >7 1.4
associated with the use of OCs. It should be
Jick60 127P < 43 >5 0.7
noted that in most studies, the relative risk esti- 174C ³ 10 1.4
mate is close to unity, indicating that any effect Romieu61 >118K < 65 <1 1.2
Cohort 3 0.9
of OCs is modest in magnitude. However, the
high incidence of breast cancer in the United P = patients; C = controls; CASH = Cancer and Steriod Hormone
Study, Centers for Disease Control.
States suggests that even small increases in rel-
ative risk could translate into more cases of
breast cancer. was associated with the use of OCs. This relative
One of the most widely quoted case-control risk was not affected appreciably by the duration
studies on OCs and breast cancer is the Cancer of use, history of fibrocystic breast disease, or
and Steroid Hormone (CASH) Study by the family history of breast cancer. These results
Centers for Disease Control.57 This project, first were validated in an updated review of the
reported in 1983, evaluated 689 patients diag- Nurses’ Health Study reported in 1997, with
nosed with breast cancer between the ages of 20 over 1.6 million person-years of follow-up.62
and 54 years who were identified through the In 1996, a meta-analysis of 54 epidemiologic
Surveillance, Epidemiology, and End Results studies of OCs and breast cancer was published
(SEER) program. These patients were matched in Lancet.65 This review compiled data on more
with 1,077 controls, and on initial analysis, the than 53,000 breast cancer patients and more
risk of breast cancer was lower for women that than 100,000 controls from 25 countries. Cur-
had been users of OCs than for women who rent users of OCs had a small but statistically
never used OCs (relative risk, 0.9). Results of the significant increase in the risk of breast cancer
CASH study were re-evaluated and reported in (relative risk, 1.24; p < .00001), and this risk did
1991 by Wingo and colleagues.64 In this report, not persist after 10 years following discontinua-
a particular focus was placed on the issue of tion of the OCs. The tumors detected in users of
OCs having variable, age-related associations OCs were also found to be of earlier stage than
with breast cancer risk. This analysis revealed a those that were detected in women that did not
trend toward increased risk for users of OCs use OCs. Two theoretical mechanisms could
younger than 35 years (relative risk, 1.4), and a explain these findings. One explanation is related
slight decrease in risk for users of OCs from 45 to the concept of estrogen acting as a promoter
to 54 years of age (relative risk, 0.9). rather than as a cause of the neoplastic process.
The Nurses’ Health Study61 provides data Under this circumstance, it would be expected
regarding the relative risk of breast cancer that more tumors would be detected during and
among users of OCs followed up in a prospective following use of OCs because the estrogen con-
fashion. In a cohort of more than 100,000 nurses tent would merely be expediting the clinical
with more than 1 million person-years of follow- appearance of a pre-existing but previously
up, no significant increase in breast cancer risk occult tumor. The second explanation is that
26 BREAST CANCER

women who are users of OCs are necessarily myocardial infarction and sudden death.66,72–76
receiving follow-up care, which presumably Estrogen replacement therapy also reduces rates
includes surveillance for breast cancer. of bone resorption by as much as 60 percent,
thereby lowering rates of osteoporosis and
Hormone Replacement Therapy osteoporotic fractures.66,75,76 Unfortunately,
ERT exerts a dose-dependent and duration-of-
The controversy surrounding hormone replace- use–dependent proliferative effect on the uter-
ment therapy (HRT) and breast cancer is com- ine lining, and several studies have demon-
plicated not only by the prevalence of breast strated an increased rate of endometrial cancer
cancer but also by the fact that we live in an associated with ERT.66,74,77–79 Results from the
aging society. The health risks associated with Postmenopausal Estrogen/Progestin Interven-
the postmenopausal estrogen-deficient state, tions (PEPI) trial, however, demonstrated that
namely, cardiovascular disease and osteoporo- the addition of cyclic micronized progesterone
sis, are being faced by increasing numbers of to ERT negated the risk of endometrial hyper-
women still in the prime years of their life. plasia (as measured by baseline and annual
Approximately one quarter of the American endometrial aspiration biopsy), without
population is currently over the age of 55 years, adversely affecting the favorable impact of ERT
and cardiovascular disease is the leading cause on the cholesterol profile.71
of death among postmenopausal women. Car- The effect of ERT on the risk of breast can-
diovascular disease accounts for nearly three cer remains an unresolved question. To date,
times as many deaths as cancer among women no prospective, randomized study has been
over the age of 65 years.66 Osteoporosis afflicts completed, and many of the patterns and
approximately 25 million Americans, and it is inconsistencies demonstrated in the available
estimated that half of all women will experi- data are similar to those seen in the published
ence an osteoporotic fracture by the age of 75 series of OC use and breast cancer. The results
years. In particular, hip fractures are a major of several studies of HRT and breast cancer
problem; they are associated with a 34 percent conducted in the United States and abroad are
mortality rate within 6 months, and the corre- given in Table 2–3. The relative risk esti-
sponding health-care costs are several billion mates80–85 vary considerably. A protective
dollars annually.67 effect was seen in the study by Gambrell and
Menopause also causes several other colleagues,81 in which a relative risk of 0.3 was
symptoms that have a significant adverse
impact on the quality of life. Vasomotor symp-
toms are experienced by 80 percent of Table 2–3. HORMONE REPLACEMENT THERAPY:
BREAST CANCER RISK IN THE GENERAL POPULATION
menopausal women; urinary incontinence,
vaginal dryness, sleep disturbances, depres- Number Mean Number
of of Years Relative
sion, anxiety, and memory losses are being Study Year Women Follow-Up Risk
reported increasingly and have all been related
Hoover80 1976 1,891 12 1.3
to changes in estrogen.66,68–70
Estrogen replacement therapy (ERT) in post- Gambrell81 1983 5,563 7 0.4
menopausal women has been proved to reverse
Hunt82 1987 4,544 6 1.59
several risk factors for cardiovascular disease,
such as low high-density lipoprotein (HDL) Bergkvist83 1989 23,244 5.7 1.1
cholesterol levels,71 and ERT has been associ- Mills84 1989 20,341 6 1.7
ated with a 40 to 60 percent reduction in com-
plications of cardiovascular disease, such as Colditz85 1995 23,965 16 1.321
Breast Cancer Risk and Management: Chemoprevention, Surgery, and Surveillance 27

associated with HRT (combined estrogen and for cardiac disease, there was a lesser benefit
progestin) among more than 5,500 women fol- from HRT (relative risk of death, 0.89). After
lowed up at the Wilford Hall United States Air 10 years of HRT, however, mortality rates
Force Medical Center. In this study, the relative began to rise, predominantly from an increas-
risk estimate was calculated by comparing the ing rate of breast cancer–related deaths.
observed breast cancer incidence with the inci- In conclusion, the use of ERT and OCs
dence expected on the basis of the SEER pro- appears to be associated with an increased risk
gram. Mills and colleagues84 followed up a of breast cancer, particularly in younger
cohort of more than 20,000 Seventh Day women.87 In the presence of atherosclerotic
Adventist women and found a relative risk of disease and osteoporosis, the use of ERT
1.7 associated with any history of HRT use; seems to reduce the incidence of severe com-
this relative risk increased to 2.5 for current plications (eg, myocardial infarction and bone
HRT users, and to 2.8 for HRT users with a fractures) and appears to be associated with
history of benign breast disease. In 1995, improved survival. Recommendation for rou-
Colditz and colleagues85 reported on findings tine use should be restricted to a select group
from the Nurses’ Health Study. A significant of women, and the decision on treatment
increase in the relative risk of breast cancer should be based on a detailed evaluation of the
associated with current HRT use (relative risk, risk/benefit ratio.
1.32) was demonstrated.
Updated results of the Nurses’ Health Study BREAST CANCER
were published in 1997.86 At the time of the CHEMOPREVENTIVE AGENTS
updated report, more than 120,000 registered
nurses had been followed up with biannual The importance of estrogens as breast cancer
questionnaires since 1976, and more than 3,600 promoters has been sustained by direct and
deaths had been documented. Several impor- indirect observations since the 1960s. An
tant findings were reported. Use of HRT was important source of indirect evidence is pro-
associated with significant decreases in mortal- vided by the relevant clinical observations
ity compared with nonuse (relative risk of derived from clinical trials of the adjuvant use
death, 0.63). This benefit of HRT was strongest of tamoxifen. Several large randomized studies
for women who had pre-existing risk factors for have demonstrated that adjuvant therapy with
atherosclerotic heart disease (eg, current the nonsteroidal antiestrogen tamoxifen citrate
tobacco use, parental history of premature is associated with reduction in the risk of devel-
myocardial infarction, diabetes, or hyperten- oping a second contralateral primary breast
sion). For women considered to be at low risk cancer by 30 to 50 percent.88–90

Table 2–4. SUMMARY OF ONGOING RANDOMIZED CHEMOPREVENTIVE TRIALS

Number
Study Agent of Patients Patient Population

BCPT-P1 (USA)94 TAM vs. placebo 13,388 High-risk, pre-/postmenopausal


Royal Marsden (UK)93 TAM vs. placebo 2,012 High-risk pre-/postmenopausal
National Tumor Institute (ITALY)96 TAM vs. placebo 5,408 Post-hysterectomy
No risk of breast cancer, pre-/postmenopausal
MD Anderson Cancer Center (USA)115 TAM +/-4-HPR Ongoing Ductal carcinoma in situ
(pre-operative)
National Tumor Institute (ITALY) 94112 4-HPR 2,972 History of breast cancer
STAR (USA)94 TAM vs. Raloxifene Ongoing High-risk, pre-/postmenopausal

BCPT = Breast Cancer Prevention Trial; NSABP = National Surgical Adjuvant Breast and Bowel Project; TAM = tamoxifen; 4-HPR = fenretinide
(N-[4-hydroxyphenyl] retinamide, 4-HPR); STAR = Study of Tamoxifen and Raloxifene.
28 BREAST CANCER

These observations confirm the central ticular mention.90 This trial demonstrated a sig-
role for estrogens in the promotion of breast nificant decrease in the incidence of contralateral
cancer and suggest an opportunity for devel- breast cancer but also a six-fold increase in the
oping a preventive strategy based on using incidence of endometrial cancer and an unex-
selective antiestrogens to modulate estrogenic pected excess of gastrointestinal malignancies.
activity. Of particular interest is the overview analy-
Preclinical data and clinical observations sis of the major randomized trials of adjuvant
derived from clinical chemoprevention trials tamoxifen among nearly 30,000 women with
conducted in other malignancies indicated a early breast cancer.95 A recent update of this
potential activity of retinoids as chemopreven- analysis demonstrated a reduction in the inci-
tive agents. The major ongoing chemopreven- dence of contralateral breast cancer incidence of
tion trials are listed on Table 2–4. 47 percent with 5 years of treatment. The pro-
portional reduction in contralateral breast cancer
Tamoxifen appeared to be unrelated to the estrogen receptor
(ER) status of the original tumor. The treatment
Tamoxifen is a nonsteroidal triphenylethylene appeared to be associated with a significant
derivative, which is generally classified as an increase in the incidence of endometrial cancer
antiestrogen with partial estrogen-agonist activ- and a slight, not significant increase in colorec-
ity in some tissues. In fact, results from chemo- tal cancer (larger with only 1 year of tamoxifen).
preventive and adjuvant trials suggest that treat- In 1986, a pilot study was started in the
ment with tamoxifen is associated with an United Kingdom to test the feasibility and tox-
increase in bone mineral density and decreased icity associated with long-term tamoxifen treat-
serum cholesterol, particularly in postmeno- ment in women at high risk of breast cancer.93
pausal women.91,92 Between October 1986 and June 1993, a total
More importantly, the use of adjuvant of 2,012 women were accrued and randomly
tamoxifen following primary surgery for estro- assigned to tamoxifen (20 mg/day) or placebo
gen-sensitive early breast cancer has been asso- for up to 8 years. A total of 265 women were on
ciated with prolonged disease-free survival and HRT at entry and 131 were randomized to
reduction in the risk of death by 20 to 30 per- tamoxifen treatment. With a median follow-up
cent. One of the major arguments in favor of of 36 months and with a compliance of 77 per-
the use of tamoxifen as a chemopreventive cent of the women assigned to the treatment
agent is derived from the observation of a sig- arm, no obvious effect on bone mineral density
nificant reduction in the incidence of primary was observed and only marginal effects on clot-
tumors in the contralateral breast in women ting factors. Tamoxifen was associated with a
treated with adjuvant tamoxifen.88,89,93,94 significant reduction in the serum cholesterol
The National Surgical Adjuvant Breast and level. More importantly, there was an increased
Bowel Project (NSABP) and other European incidence of uterine fibromata and benign ovar-
studies showed a reduction between 40 and 50 ian cysts; however, no increase in endometrial
percent in the incidence of primary tumors in the cancer incidence was reported.
contralateral breast among women taking tamox- On the basis of these encouraging data, in
ifen as adjuvant therapy.88,91,92 Among the Euro- 1992, a large, multicenter, randomized, double-
pean trials, the Stockholm trial, designed to eval- blind trial funded by the National Cancer Insti-
uate the efficacy and toxicity of adjuvant tute (NCI) was begun to test whether tamoxifen
tamoxifen (40 mg daily) for 2 years in post- could prevent breast cancer in high-risk
menopausal patients (23 percent older than 50 women.94 The population at risk was defined,
years) with unilateral breast cancer, deserves par- taking into account the following risk factors,
Breast Cancer Risk and Management: Chemoprevention, Surgery, and Surveillance 29

as indicated by the Gail model: age, age at slight increase in the incidence of deep vein
menarche and at first live birth, number of thrombosis and pulmonary embolism in the
first-degree relatives affected, and, finally, the tamoxifen group was also reported; all these
number of previous breast biopsies and the events were more frequently observed in
presence or absence of atypical hyperplasia. women older than 50 years of age. Because of
The Breast Cancer Prevention Trial (BCPT- these reported complications, the decision
P1) enrolled 13,388 women older than 35 years about whether to use tamoxifen as a chemopre-
between April 1992 and September 1997. The ventive agent must be carefully weighed for
research, coordinated by the NSABP, involved every patient on the basis of an accurate evalu-
more than 300 centers across the United States ation of the patient’s age group, personal breast
and Canada. The study was closed and prelimi- cancer risks, and comorbid conditions.
nary results released 14 months earlier than Similar trials of tamoxifen for chemopreven-
planned.94 In the median average follow-up time tion have been conducted in Italy and the United
of 54.6 months, 89 cases of invasive breast can- Kingdom.96 The Italian study has completed
cer occurred in the group of women assigned to accrual of 5,408 women who have had hysterec-
tamoxifen treatment (6,681 women) compared tomy and have no factors associated with
with 175 cases in the group assigned to placebo increased risk of breast cancer. Preliminary
(6,707 women), corresponding to a 49 percent analysis at a median follow-up of 46 months did
risk reduction. There was also a 50 percent not show any difference in breast cancer inci-
reduction in the incidence of noninvasive breast dence in the tamoxifen group compared with
cancer. Tamoxifen reduced the occurrence of the placebo-control group.96 Differences in the
ER-positive tumors by 69 percent, but no effi- study populations, age distributions, history of
cacy was seen in the prevention of ER-negative HRT, and family history may account for the
tumors. The risk reduction was not age depen- inability of studies to confirm the effectiveness
dent; the risk reduction in women aged 49 years of tamoxifen for chemoprevention.
or younger was 44 percent, and it was 55 percent
in women older than 60 years. The NCI and the Selective Estrogen Receptor Modulators
Endpoint Review, Safety Monitoring, and Advi-
sory Committee agreed that the participants and In the past decade, the reports of significant
their physicians should be told which treatment side effects associated with the prolonged use
has been assigned because of the clear evidence of tamoxifen have stimulated research directed
that tamoxifen reduced breast cancer risk. toward the development of other selective
The Breast Cancer Prevention Trial was also estrogen receptor modulators (SERMs). Among
designed to evaluate the possible benefit of the various products investigated, raloxifene
tamoxifen in reducing cardiac events and osteo- has demonstrated antitumor activity and a
porosis-related complications. There was no favorable toxicity profile and is being further
difference between the tamoxifen group and the investigated.97–99
placebo group in the number of heart attacks, Preclinical data have shown that raloxifene,
whereas there was a 19 percent reduction in the an antiestrogen with no estrogen-agonist effect
incidence of fractures of the hip, wrist, and on the uterus, inhibits mammary carcinogenesis
spine (111 cases in the tamoxifen group versus in a rat model of breast cancer in a manner sim-
137 cases in the placebo group). Treatment ilar to tamoxifen when raloxifene is used in
with tamoxifen was associated with an combination with 9-cis retinoic acid.97 Clinical
increased incidence of endometrial cancer (33 trials have been started in an attempt to establish
cases versus 14 cases in the placebo group), in the role of raloxifene in preventing osteoporosis
particular in women aged 50 years or older. A in postmenopausal women, and preliminary
30 BREAST CANCER

results from two randomized clinical trials have sensitive to the antiproliferative effects of
recently become available. The Multiple Out- retinoids. Enhanced sensitivity has been shown
comes of Raloxifene Evaluation (MORE) trial by estrogen receptor–positive cell lines, whereas
was specifically designed to evaluate the possi- estrogen receptor–negative cell lines have shown
bility of reducing the risk of fractures in post- minimal sensitivity to these compounds.110,111
menopausal women receiving raloxifene; a The mechanisms of the antiproliferative effect
markedly reduced risk of newly diagnosed breast are still being investigated. At least, in some
cancer was demonstrated with raloxifene com- cell lines, apoptosis, instead of differentiation,
pared with placebo (0.21% versus 0.82%).100 seems to be the prevalent mechanism of growth
Jordan and colleagues recently reported the inhibition.111
results of a multicenter, double-blind random- A synthetic retinoid, fenretinide (N-[4-
ized trial conducted in about 12,000 women. hydroxyphenyl] retinamide, 4-HPR), has demon-
Treatment with raloxifene was associated with a strated the capacity to inhibit the growth of
58 percent reduction in the risk of developing breast cancer cell lines and chemically induced
primary breast cancer.101 These results have mammary tumors in rats, without the toxicity
stimulated the design of a second major breast associated with other retinoids. This compound
cancer prevention trial, the Study of Tamoxifen was the first retinoid to be tested in clinical tri-
and Raloxifene (STAR) that will compare the als and has been proved to be well tolerated at
toxicity, risks, and benefits of raloxifene with a daily dose of 200 mg with a 3-day monthly
those of tamoxifen. Women enrolled in the drug holiday.112,113
study will be randomly assigned to receive An Italian prospective randomized trial
either 20 mg of tamoxifen or 60 mg of ralox- designed to evaluate the role of fenretinide in
ifene for 5 years, with follow-up planned for an reducing the incidence of contralateral breast
additional 2 years. cancer began in March 1987. In July 1993,
A number of other SERMs have emerged accrual of 2,972 women, age 30 to 70 years,
(eg, toremifene, trioxifene, droloxifene, TAT-59) with history of T1-2, N0 breast cancer was
for clinical use.102–105 The majority of these completed.112 Preliminary data suggested that
drugs are presently in phase I to II clinical tri- fenretinide can reduce the incidence of ovarian
als and have already demonstrated their clinical carcinomas.113 Though safer than other
activity in the management of breast cancer. retinoids in experimental models, fenretinide
They represent possible candidates for future produced visual (dark adaptation) and ophthal-
chemoprevention studies. mologic complaints (ocular dryness, lacrima-
tion, conjunctivitis, photophobia) in 20 percent
Retinoids and 8 percent of women, respectively, at 5
years.114 Such effects are thought to be caused
Retinoids are a family of natural and synthetic by the reduction of plasma retinal levels, which
compounds structurally related to vitamin A. occurs after administration of the retinoid.
They are a group of molecules capable of influ- In animal models, 9-cis-retinoic acid in
encing many biologic functions, such as prolif- combination with antiestrogens (tamoxifen or
eration, differentiation, and induction of apop- raloxifene) resulted in effective chemopreven-
tosis.106–109 Retinoids function via two types of tion of a rat model of breast cancer induced by
nuclear receptors, the retinoid alpha-receptors the carcinogen nitrosomethylurea.97,111
(RARs) and the retinoid X receptors (RXRs), The University of Texas M. D. Anderson
each of which is encoded by three genes.93 Cancer Center is presently investigating the role
Preclinical data have demonstrated that of tamoxifen and fenretinide in reducing the risk
carcinogen-induced mammary carcinomas are of invasive breast cancer in patients diagnosed
Breast Cancer Risk and Management: Chemoprevention, Surgery, and Surveillance 31

with DCIS.115 This phase II trial is accruing Heterocyclic amines, a group of mutagenic
women presenting with small breast lesions and compounds identified in cooked foods, seem to
mammographic calcifications suspicious for be related to the increased risk of breast cancer
malignancy. After histologic confirmation associated with high intake of well-done meat.
through core biopsies, participants are randomly Recently, a case-control study among 41,836
assigned to receive tamoxifen, fenretinide, or a women demonstrated that women who con-
combination of both. The treatment is planned sumed well-done meats, including hamburger,
for 3 weeks before definitive surgery. An impor- beef steak, and bacon, had higher adjusted odds
tant objective of this trial is to perform a detailed ratios for breast cancer (up to 4.62), if they con-
quantitative assessment of biomarkers to be used sumed all three different meats well done.121
as surrogate end points. The proposed SEBs These data have provided the rationale for
include estrogen and progesterone receptors, diet interventions, consisting of a low-fat diet
nuclear retinoid receptors, transforming growth and fish-oil supplements, that have been found
factor (TGF)-b, HER-2/neu, proliferation (Ki-67 to be able to produce increases in total omega-
immunostaining), angiogenesis (factor VIII) 3 PUFAs in adipose tissue and in the ratio of
markers, and chromosomal aberrations. This omega-3/omega-6 PUFAs in patients with
trial is expected to provide insight into the bio- breast cancer.120 The Canadian Diet and Breast
logic mechanisms of antiestrogens and retinoids, Cancer Prevention Study Group has conducted
or the combination of both, in reducing the pro- a multicenter randomized trial involving
gression of DCIS to invasive cancer. women with breast densities detected on mam-
mography and showed that after 2 years of a
Dietary Interventions low-fat diet, with less than 15 percent of calo-
ries from fat, there was a significant reduction
Epidemiologic observations of large interna- in the number of radiographic abnormalities.122
tional differences in the incidence of breast Adjuvant dietary recommendations of 15 per-
cancer have provided a basis for formulating cent of calories from fat for women with post-
hypotheses on a possible relation between diet menopausal breast cancer are currently being
and the development of cancer. The age- evaluated in the Women’s Intervention Nutri-
adjusted incidence of breast cancer varies from tion Study and in the Women’s Healthy Eating
22 per 100,000 in Japan to 68 per 100,000 in and Living Study.123
the Netherlands.116 The ratio of breast cancer The role of alcohol consumption and smok-
mortality between the United States and Japan ing are also being extensively investigated as
is 3:1 for premenopausal women and 8:1 for possible risk factors for breast cancer. While
postmenopausal women.117 These important the majority of the studies has documented that
differences may possibly be related to fat intake high alcohol intake is associated with a signifi-
and total calories in the diet. Clinical data col- cant increased incidence of breast cancer, no
lected from case-control studies have demon- definitive pathogenetic role for active or pas-
strated a positive correlation between diets high sive smoking has been demonstrated.124,125
in fat and meat and breast cancer.118–122 Experi- The use of natural products contained in
mental studies have shown that omega-6 essential oils and soy-based products, for
polyunsaturated fatty acids (PUFAs) contained example, the monoterpenes limonene and
in high-fat diets promote both mammary perillyl alcohol and the isoflavonoid genis-
tumorigenesis and cell proliferation in chemi- tein, all showed preclinical evidence of tumor
cally induced mammary tumors, whereas regression.126–129 The effects of limonene and
omega-3 PUFAs, contained in fish oil, can limonene-related monoterpenes, perillyl alco-
inhibit these effects.119,120 hol and perillic acid, on cell growth, cell cycle
32 BREAST CANCER

progression, and expression of cyclin D1 has tion analyses (approximately 700 per breast) of
been investigated in T-4D, MCF-7, MDA-MB- the margins identified 3 cases of breast tissue
231 breast cancer cell lines. The results extending into the pectoralis fascia, 1 case of
revealed that limonene-related monoterpenes pectoralis muscle involvement, 2 cases of infe-
caused a dose-dependent inhibition of cell pro- rior skin flap involvement, and 1 case of axil-
liferation. Of the three monoterpenes tested, lary tissue involvement. It would be expected
perillyl alcohol was the most potent and that prophylactic subcutaneous mastectomy
limonene was the least potent inhibitor of cell (defined as removal of all gross breast tissue,
growth. Growth inhibition induced by perillyl usually via an inframammary incision, but
alcohol and perillic acid was associated with a sparing the nipple-areolar complex) would
fall in the proportion of cells in the S phase, result in additional breast tissue left on the skin
accumulation of cells in the G1 phase, and a flaps of the nipple-areolar complex.
decrease in cyclin D1 mRNA levels.128 The The clinical significance of retained breast
potential preventive role of genistein, a compo- tissue in the setting of prophylactic mastec-
nent of soy, has been evaluated in rats. Pharma- tomy for humans is not yet defined. In the
cologic doses of genistein given to immature rodent model of mammary tumors and prophy-
rats enhance mammary gland differentiation, lactic mastectomy, it is clear that the extent of
resulting in a significantly less proliferate breast tissue removed does not clearly corre-
gland that is not as suspectible to mammary late with the extent of protection against breast
cancer.129 These components are presently cancer. Wong and colleagues131 performed
being tested in several clinical chemopreven- varying degrees of partial versus total mastec-
tive studies. tomy versus sham surgery in a series of rats,
either before or after administration of the car-
SURGICAL PROPHYLAXIS cinogen DMBA; at 8 months of age, there were
AND MODULATION OF RISK no significant differences in the number of car-
cinogen-induced tumors between any of the
Prophylactic Mastectomy groups. Using a mouse model with a high inci-
dence of spontaneous mammary tumor devel-
It seems intuitive that mastectomy would be an opment (and therefore theoretically more sim-
effective means of preventing breast cancer, ilar to the human experience of spontaneous
especially in this era of immediate reconstruc- breast cancer incidence) Nelson and col-
tion techniques that produce cosmetically leagues132 similarly found no difference in the
acceptable results. However, animal models as number of tumors that developed in mice that
well as clinical data from trials in humans have underwent either sham surgery, 50 percent
confirmed that this is not always the case. Stud- mastectomy, or total mastectomy.
ies have demonstrated that even total mastec- Data on prophylactic bilateral mastectomy in
tomy (defined as removal of the entire breast, humans are limited. The often-cited studies by
including the nipple-areolar complex, but spar- Pennisi and Capozzi133 and Woods and
ing the axillary contents) is frequently incom- Meland134 in the plastic surgery literature each
plete; microscopic amounts of breast tissue reported on at least 1,500 women who under-
may be left in the skin flaps, attached to the went subcutaneous mastectomies, and in both
pectoralis fascia and extending into the axilla. studies, the subsequent incidence of breast car-
Temple and colleagues130 evaluated 10 prophy- cinoma was less than 1 percent. However, both
lactic mastectomy specimens from 5 patients these studies have been criticized for their lim-
considered to be at high risk for developing ited applicability to truly high-risk women,
breast cancer. In this study, random frozen sec- since many of the prophylactic procedures were
Breast Cancer Risk and Management: Chemoprevention, Surgery, and Surveillance 33

performed in women who would be considered risk associated with prophylactic mastectomy.
at the present time to be at only low or interme- Another patient population that might be
diate risk for breast cancer. Detailed follow-up considered to be suitable for prophylactic mas-
information is also lacking in these large series. tectomy is women with a history of unilateral
It remains to be determined if a prophylac- breast cancer, in whom the risk of subsequent
tic mastectomy is clearly indicated in women at contralateral breast cancer is approximately 0.5
high risk of developing breast cancer. Women to 0.7 percent per year.143 However, it has been
with BRCA1 mutations, who may have a cumu- argued that the risk of death from the primary
lative breast cancer risk of 40 to 85 percent, cancer is still greater than the risk of develop-
would be the obvious candidates. ing a second primary cancer for the majority of
Schrag and colleagues135 developed a statis- breast cancer patients, making the survival ben-
tical decision model to calculate the benefit efit of prophylactic surgery questionable.144 On
that BRCA1 or BRCA2 mutation carriers might the other hand, the rationale has also been
derive from prophylactic mastectomy. Using a offered that optimal immediate reconstruction
risk-reduction estimate of 85 percent associated cosmesis can be attained with bilateral trans-
with prophylactic mastectomy, this study deter- verse rectus abdominis myocutaneous (TRAM)
mined that a 30-year-old BRCA1 mutation car- flaps. To address this issue Kroll and col-
rier would gain 2.9 to 5.3 years of life leagues145 studied 88 patients with unilateral
expectancy following preventive surgery. breast cancer who had undergone bilateral mas-
Lynch and colleagues136 reported on the results tectomies with immediate breast reconstruc-
of a series of women who had undergone exten- tion. Previously unsuspected invasive breast
sive genetic counseling and subsequently tested cancer was found in 3.4 percent of the con-
positive for BRCA1 gene mutations. Only 35 tralateral mastectomy specimens.
percent of these patients said they would con- The Society of Surgical Oncology has delin-
sider undergoing prophylactic mastectomy. eated categories of patients for whom prophy-
This finding underscores the complexity of lactic mastectomy may reasonably be consid-
identifying high-risk women who will benefit ered on the basis of clinical features (and not
psychologically as well as clinically from pre- including genetic testing results).144 For women
ventive surgery. with no history of breast cancer, the indications
In addition, there are many case reports doc- include atypical hyperplasia, family history of
umenting the failure of prophylactic total mas- premenopausal bilateral breast cancer, and
tectomy to protect against breast cancer.137–141 dense, nodular breasts associated with atypical
However, very intriguing data were recently hyperplasia. For women with a known unilateral
reported by Hartmann and colleagues from the breast cancer, the indications for considering
Mayo Clinic.142 A retrospective analysis was contralateral prophylactic mastectomy include
performed on 639 women with moderate or diffuse microcalcifications, LCIS, a large, diffi-
high risk for breast cancer (by family history) cult-to-evaluate breast, history of LCIS, and
that had undergone bilateral prophylactic sub- family history of early-onset breast cancer.
cutaneous mastectomy between 1960 and 1993.
The breast cancer incidence in these women Prophylactic Oophorectomy
was compared with the number of expected and/or Hysterectomy
cases based on the Gail model and with the
number of cases that occurred among female Several studies have documented lower breast
siblings who had not undergone prophylactic cancer incidence among women who underwent
surgery; these estimations revealed an approxi- oophorectomy at a young age. The effect of hys-
mately 90 percent reduction in breast cancer terectomy on breast cancer risk is less clear, but
34 BREAST CANCER

it has been postulated that hysterectomy may gression. In the future, prospective clinical trials
have some secondary effects by affecting ovar- of chemoprevention strategies should be limited
ian blood flow and ovulation. Schairer and col- to high-risk populations identified on the basis
leagues evaluated 15,844 women undergoing of a combination of epidemiologic, histopatho-
surgery in the Uppsala health care region of logic, and genetic data and should make use of
Sweden and found a 50 percent reduction in SEBs to evaluate the efficacy of drug interven-
breast cancer risk in those women who under- tions. This approach will contribute greatly to
went bilateral oophorectomy prior to age 50 reducing the patient population under study,
years, compared with the risk of the background eventually reducing the costs related to these
population.146 Hysterectomy alone had no con- investigations and helping to clarify the biology
sistent association with change in breast cancer of each drug’s mechanism of action.
risk. In a case-control series from Italy, women The initial results of the BCPT-P1 have
who underwent premenopausal oophorectomy demonstrated, for the first time, the possibility
with hysterectomy or hysterectomy alone had of reducing the risk of breast cancer in a high-
reduced relative risk of developing breast cancer risk group of women, with a marginal toxicity.
(0.8 and 0.7, respectively).147 However, given The ongoing STAR preventive trial is designed
the importance of the ovarian function in main- to determine if raloxifene has a chemopreven-
taining cardiovascular and bone health, there are tive efficacy comparable with tamoxifen with
presently no indications for recommending less associated toxicity. In the meantime, a
these procedures as prophylaxis against breast longer follow-up of the BCPT-P1 trial will clar-
cancer in any subset of patients. ify if the treatment with tamoxifen represents a
true preventive intervention or a treatment for
CONCLUSIONS preclinical conditions with consequent delay-
ing of the onset of invasive breast cancer.
The recently reported encouraging results with The potential role of dietary intervention in
the use of tamoxifen and the ongoing clinical tri- modifying the risk of breast cancer is probably
als introducing SERMs (raloxifene), retinoids, presently underestimated; the ongoing clinical
and other approaches suggest an increasing trials may contribute essential information to
awareness in physicians of the field of chemo- their potential clinical applicability.
prevention and its potentially enormous socio- In conclusion, more attention should be
economic implications. Breast cancer chemo- directed to the biologic relationships among
prevention is a field in constant evolution and hormone modulation, diet, and the risk of
has the potential to significantly affect the lives breast cancer to develop an “ideal lifestyle
of thousands of women by reducing their risk of model” to propose for the high-risk groups. In
breast cancer. this context, the role of prophylactic surgery,
In the last decade, the increasing information with the psychologic consequences related to
available on the differential contribution of the change in body image, even if associated
genetic, dietary, and environmental factors has with improved outcome in high-risk women,
greatly improved our ability to determine the will come to be considered as a secondary,
absolute breast cancer risk for every woman and rather than a primary, option for breast cancer
properly select high-risk groups for interven- risk management.
tional studies. Interestingly, the concomitant
evaluation of histopathologic factors and mul-
REFERENCES
tiple biomarkers has offered the opportunity to
increase our understanding of the important bio- 1. Chu KC, Tarone RE, Kessler LG, et al. Recent
logic modifications associated with tumor pro- trends in U.S. breast cancer incidence, survival,
Breast Cancer Risk and Management: Chemoprevention, Surgery, and Surveillance 35

and mortality rates. J Natl Cancer Inst 1996; 17. Ford D, Easton DF, Petro J. Estimates of the gene
88:1571–9. frequency of BRCA1 and its contribution to
2. Vogel VG. Subjects and recruitment strategies for breast and ovarian cancer incidence. Am J
a short-term phase II chemoprevention trial of Hum Genet 1995;57:1457–62.
breast cancer using surrogate endpoint bio- 18. Struewing JP, Hartge P, Wacholder S, et al. The
markers. J Cell Biochem 1993;17G:257–8. risk of cancer associated with specific muta-
3. Page DL, Dupont WD, Rogers LW. Atypical tions of BRCA1 and BRCA2 among Ashke-
hyperplastic lesions of the female breast. A nazi Jews. N Engl J Med 1997;336:1401–8.
long-term follow-up study. Cancer 1985;55: 19. Shattuck-Eidens D, Oliphant A, McClure M, et al.
2698–708. BRCA1 sequence analysis in women at high
4. Dupont WD, Page DL. Relative risk of breast can- risk for susceptibility mutations. JAMA 1997;
cer varies with time since diagnosis of atypical 278:1242–50.
hyperplasia. Hum Pathol 1989;20:723–5. 20. Couch FJ, DeShano M, Blackwood MA, et al.
5. Hill AD, Doyle JM, McDermott EW. Hereditary BRCA1 mutations in women attending clinics
breast cancer. Br J Surg 1997;84:1334–9. that evaluate the risk of breast cancer. N Engl J
6. Tseng SL, Yu IC, Yue CT, et al. Allelic loss at Med 1997;336:1409–15.
BRCA1, BRCA2, and adjacent loci in relation 21. Iglehart JD, Miron A, Rimer BK, et al. Overesti-
to TP53 abnormality in breast cancer. Genes mation of hereditary breast cancer risk. Ann
Chromosomes Cancer 1997;20:377–82. Surg 1998;228(3):375–84.
7. Sporn MB, Newton DL. Chemoprevention of can- 22. Krainer M, Silva-Arrieta S, FitzGerald MG, et al.
cer and retinoids. Fed Proc 1979;38:2528–34. Differential contributions of BRCA1 and
8. Kelloff GJ, Boone CW, Steele VE. Progress in BRCA2 to early onset breast cancer. N Engl J
cancer chemoprevention: perspectives on agent Med 1997;336:1416–21.
selection and short-term clinical intervention 23. Prechtel D, Werenskiold AK, Prechtel K, et al.
trials. Cancer Res 1994;54:2015S–24S. Frequent loss of heterozygosity at chromo-
9. Kelloff GJ, Boone CW, Crowell JA, et al. Surro- some 13q12-13 with BRCA2 markers in spo-
gate endpoint biomarkers for phase II cancer radic male breast cancer. Diagn Mol Pathol
chemoprevention trials. J Cell Biochem Suppl 1998;7(1):57–62.
1994;19:1–9. 24. Malkin D, Li FP, Strong LC, et al. Germ line p53
10. Grizzle WE, Myers RB, Arnold MM, Srivastava S. mutations in a familial syndrome of breast can-
Evaluation of biomarkers in breast and prostate cer, sarcomas, and other neoplasms. Science
cancer. J Cell Biochem Suppl 1994;19:259–66. 1990;250:1233–8.
11. Fabian CJ, Kamel S, Zalles C, Kimler BF. Identi- 25. Swift M, Morrell D, Massey RB, Chase DL. Inci-
fication of a chemoprevention cohort from a dence of cancer in 161 families affected by
population of women at high risk for breast ataxia-telangiectasia. N Engl J Med 1991;325:
cancer. J Cell Biochem 1996;25S:112–22. 1831–6.
12. Gail MH, Brinton LA, Byar DP, et al. Projecting 26. Tokunaga M, Land CE, Yammoto T, et al. Inci-
individualized probabilities of developing dence of female breast cancer among atomic
breast cancer for white females who are being bomb survivors, Hiroshima and Nagasaki,
examined annually. J Natl Cancer Inst 1989; 1950–1980. Radiat Res 1987;112:243–72.
81:1879–86. 27. Mackenzie I. Breast cancer following multiple
13. Benichou J, Gail MH, Mulvihill JJ. Graphs to esti- fluoroscopies. Br J Cancer 1965;19:1–8.
mate an individualized risk of breast cancer. 28. Miller AB, Howe GR, Sherman GJ, et al. Mortal-
J Clin Oncol 1996;14(1):103–10. ity from breast cancer after irradiation during
14. Smith J. New computer program assesses a fluoroscopic examinations in patients being
woman’s risk for developing breast cancer. treated for tuberculosis. N Engl J Med 1989;
J Natl Cancer Inst 1998;90(18):1332. 321:1285–9.
15. Lynch HT, Krush AJ. The cancer family syndrome 29. Shore RE, Hildreth N, Woodard E, et al. Breast
and cancer control. Surg Gynecol Obstet 1971; cancer among women given x-ray therapy for
132:247–50. acute postpartum mastitis. J Natl Cancer Inst
16. Marcus JN, Watson P, Page DL, et al. Hereditary 1986;77:689–96.
breast cancer: pathobiology, prognosis, and 30. Hildreth NG, Shore RE, Dvoretsky PM. The risk
BRCA1 and BRCA2 gene linkage. Cancer of breast cancer after irradiation of the thymus
1996;77:697–709. in infancy. N Engl J Med 1989;321:1281–4.
36 BREAST CANCER

31. Goss PE, Sierra S. Current perspectives on radia- 44. Rohan TE, Hartwick W, Miller AB, Kandel RA.
tion-inducted breast cancer. J Clin Oncol 1998; Immunohistochemical detection of c-erbB-2
16(1):338–47. and p53 in benign breast disease and breast
32. van Leeuwen FE, Klockman WJ, Hagenbeek A, et cancer risk. J Natl Cancer Inst 1998;90:126–9.
al. Second cancer risk following Hodgkin’s dis- 45. Beatson GT. On the treatment of inoperable cases
ease: a 20-year follow-up study. J Clin Oncol of carcinoma of the mamma: suggestions for a
1994;12:312–25. new method of treatment with illustrative
33. Kaldor JM, Day NE, Brand P, et al. Second malig- cases. Lancet 1986;2:104–7.
nancies following testicular cancer, ovarian 46. Kelsey JL, Gammon MD. The epidemiology of
cancer and Hodgkin’s disease: an international breast cancer. CA Cancer J Clin 1991;41:147–65.
collaborative study among cancer registries. Int 47. Hulka BS, Liu ET, Lininger RA. Steroid hor-
J Cancer 1987;39:571–85. mones and risk of breast cancer. Cancer 1994;
34. Tucker MA, Coleman CN, Cox RS, et al. Risk of 74:1111–24.
second cancers after treatment for Hodgkin’s 48. Russo J, Tay LK, Russo IH. Differentiation of the
disease. N Engl J Med 1988;318:76–81. mammary gland and susceptibility to carcino-
35. Hancock SL, Tucker MA, Hoppe RT. Breast can- genesis. Breast Cancer Res Treat 1982;2:5–73.
cer after treatment of Hodgkin’s disease. J Natl 49. Henderson BE, Ross R, Bernstein L. Estrogens as
Cancer Inst 1993;85:25–31. a cause of human cancer: the Richard and
36. Sankila R, Garwicz S, Olsen JH, et al. Risk of Hilda Rosenthal Foundation Award Lecture.
subsequent malignant neoplasm among 1641 Cancer Res 1988;48:246–53.
Hodgkin’s disease patients diagnosed in child- 50. Pike MC, Spicer DV, Dahmoush L, Press MF.
hood and adolescence: a population-based Estrogens, progestogens, normal breast cell
cohort study in the five Nordic countries. J proliferation, and breast cancer risk. Epidemiol
Clin Oncol 1996;14:1442–6. Rev 1993;15:17–35.
37. Aisenberg AC, Finkelstein DM, Doppke KP, et al. 51. Spicer DV, Pike MC. Breast cancer prevention
High risk of breast carcinoma after irradiation through modulation of endogenous hormones.
of young women with Hodgkin’s disease. Can- Breast Cancer Res Treat 1993;28:179–93.
cer 1997;79(6):1203–10. 52. Dickson RB, Thompson EW, Lippman ME. Reg-
38. Feig SA, Hendrick RE. Radiation risk from ulation of proliferation, invasion, and growth
screening mammography of women aged 40- factor synthesis in breast cancer by steroids. J
49 years [Monogr]. J Natl Cancer Inst 1997; Steroid Biochem Mol Biol 1990;37:305–16.
(22):119–24. 53. Cauley JA, Gutal JP, Kuller LH, et al. The epidemi-
39. Marshall CJ, Schumann GB, Ward JH, et al. Cyto- ology of serum sex hormones in postmenopausal
logic identification of clinically occult prolifer- women. Am J Epidemiol 1989;129:1120–31.
ative breast disease in women with a family 54. Moore M. Male breast cancer. In: Harris JR, Lipp-
history of breast cancer. Ann Pathol 1991;95: man ME, Morrow M, Hellman S, editors. Dis-
157–65. eases of the breast. Philadelphia: Lippincott-
40. McKittrick R, Fabian C, Kamel S, et al. Dysplasia Raven; 1996. p. 859.
and other biomarker abnormalities as potential 55. Committee on the Relationship Between Oral
surrogate endpoint biomarkers in breast Contraceptives and Breast Cancer. Institute of
chemoprevention trials [abstract]. Proc Am Medicine, Division of Health Promotion and
Soc Clin Oncol 1995;14:A348. Disease Prevention. Oral contraceptives and
41. Alpers CE, Wellings SR. The prevalence of carci- breast cancer. Washington, D.C.: National
noma in situ in normal and cancer-associated Academy Press; 1991.
breast. Hum Pathol 1985;16:796–807. 56. Pike MC, Henderson BE, Krailo MD, et al. Breast
42. Frykberg ER, Bland KI. In situ breast carcinoma. cancer in young women and use of oral contra-
Adv Surg 1993;26:29–72. ceptives: possible modifying effect of formula-
43. Eriksson ET, Schimmelpenning H, Aspenblad U, tion and age at use. Lancet 1983;2:926–9.
et al. Immunohistochemical expression of the 57. The Centers for Disease Control Cancer and
mutant p53 protein and nuclear DNA content Steroid Hormone Study. Long-term oral con-
during the transition from benign to malig- traceptive use and the risk of breast cancer.
nant breast disease. Hum Pathol 1994;25: JAMA 1983;249:1591–5.
1228–33. 58. Stadel BV, Rubin GL, Webster LA, et al. Oral con-
Breast Cancer Risk and Management: Chemoprevention, Surgery, and Surveillance 37

traceptives and breast cancer in young women. 72. Stampfer MJ, Colditz GA. Estrogen replacement
Lancet 1985;2:970–3. therapy and coronary heart disease: a quantita-
59. Miller DR, Rosenberg L, Kaufman DW, et al. tive assessment of the epidemiologic evidence.
Breast cancer risk in relation to early contra- Prev Med 1991;20:47–63.
ceptive use. Obstet Gynecol 1986;68:863–8. 73. Wenger NK, Speroff L, Packard B. Cardiovascu-
60. Jick SS, Walker AM, Stergachis A, Jick H. Oral lar health and disease in women. N Engl J Med
contraceptives and breast cancer. Br J Cancer 1993;329:247–56.
1989;59:618–21. 74. Grady D, Rubin SM, Petitti DB, et al. Hormone
61. Romieu I, Willett WC, Colditz GA, et al. Prospec- therapy to prevent disease and prolong life in
tive study of oral contraceptive use and risk of postmenopausal women. Ann Intern Med
breast cancer in women. J Natl Cancer Inst 1992;117:1016–37.
1989;81:1313–21. 75. Lindsay R. Estrogen therapy in the prevention and
62. Hankinson SE, Colditz GA, Manson JE, et al. A management of osteoporosis. Am J Obstet
prospective study of oral contraceptive use and Gynecol 1987;156:1347–51.
risk of breast cancer (Nurses’ Health Study, 76. Weiss NS, Ufe CL, Ballard JH, et al. Decreased
United States). Cancer Causes Control 1997;8: risk of fractures of the hip and lower forearm
65–72. with postmenopausal use of estrogen. N Engl J
63. Weinstein AL, Mahoney MC, Nasca PC, et al. Med 1980;303:1195–8.
Breast cancer risk and oral contraceptive use: 77. Mack TM, Pike MC, Henderson BE, Pfeffer RI.
results from a large case-control study. Epi- Estrogens and endometrial cancer in a retirement
demiology 1991;2:353–8. community. N Engl J Med 1976;294:1261–2.
64. Wingo PA, Lee NC, Ory HW, et al. Age-specific 78. Boring JE, Bain CJ, Ehrmann RL. Conjugated
differences in the relationship between oral estrogen use and risk of endometrial cancer.
contraceptive use and breast cancer. Obstet Am J Epidemiol 1986;124:434–41.
Gynecol 1991;78:161–70. 79. Antunes CM, Stolley PD, Rosenshein NB, et al.
65. Collaborative Group on Hormonal Factors in Endometrial cancer and estrogen use: report of
Breast Cancer. Breast cancer and hormonal a large case-control study. N Engl J Med 1979;
contraceptives. Collaborative reanalysis of 300:9–13.
individual data on 53,297 women with breast 80. Hoover R, Gray LA, Cole P, MacMahon B.
cancer and 100,239 women without breast can- Menopausal estrogens and breast cancer. N
cer from 54 epidemiological studies. Lancet Engl J Med 1976;295:401–5.
1996;347:1713–27. 81. Gambrell RD, Maier RC, Sanders BI. Decreased
66. Smith HO, Kammerer-Doak DN, Barbo DM, incidence of breast cancer in postmenopausal
Sarto GE. Hormone replacement therapy in the estrogen-progestogen users. Obstet Gynecol
menopause: a pro opinion. CA Cancer J Clin 1983;62:435–43.
1996;46:343–63. 82. Hunt K, Vessey M, McPherson K, Coleman M.
67. Consensus Conference. Osteoporosis. JAMA Long-term surveillance of mortality and can-
1984;252:799–802. cer incidence in women receiving hormone
68. Bhatia NN, Bergman A, Karram MM. Effects of replacement therapy. Br J Obstet Gynecol
estrogen on urethral function in women with 1987;94:620–35.
urinary incontinence. Am J Obstet Gynecol 83. Bergkvist L, Adami H-O, Persson I, et al. The risk
1989;160:176–81. of breast cancer after estrogen and estrogen-
69. Campbell S, Whitehead M. Oestrogen therapy and progestin replacement. N Engl J Med 1989;
the menopausal syndrome. Clin Obstet Gynecol 321:293–7.
1977;4:31–47. 84. Mills PK, Beeson L, Phillips RL, Fraser GE.
70. Sherwin BB. Estrogen and cognitive function in Prospective study of exogenous hormone use
women. Proc Soc Exper Biol Med 1998;217:17. and breast cancer in Seventh-Day Adventists.
71. The Writing Group for the PEPI Trial. Effects of Cancer 1989;64:591–7.
estrogen or estrogen/progestin regimens on 85. Colditz GA, Hankinson SE, Hunter DJ, et al. The
heart disease risk factors in postmenopausal use of estrogens and progestins and the risk of
women: the Postmenopausal Estrogen/Prog- breast cancer in postmenopausal women. N
estin Interventions (PEPI) trial. JAMA 1995; Engl J Med 1995;332:1589–93.
273:199–208. 86. Grodstein F, Stampfer MJ, Colditz GA, et al. Post-
38 BREAST CANCER

menopausal hormone therapy and mortality. N 98. Gradishar WJ, Jordan VC. Clinical potential of new
Engl J Med 1997;336:1769–75. antiestrogens. J Clin Oncol 1997;15(2):840–52.
87. Colditz GA. Relationship between estrogen lev- 99. Boss SM, Huster WJ, Neild JA, et al. Effects of
els, use of hormone replacement therapy, and raloxifene hydrochloride on the endometrium
breast cancer. J Natl Cancer Inst 1998;90(11): of postmenopausal women. Am J Obstet
814–23. Gynecol 1997;177:1458–64.
88. Fornander T, Rutqvist LE, Cedermark B, et al. 100. Cummings SR, Norton L, Eckert S, et al. Ralox-
Adjuvant tamoxifen in early breast cancer: ifene reduces the risk of breast cancer and may
occurrence of new primary cancers. Lancet decrease the risk of endometrial cancer in post-
1989;1:117–20. menopausal women. Two-year findings from
89. Andersson M, Storm HH, Mouridsen HT. Incidence the multiple outcomes of raloxifene evaluation
of new primary cancers after adjuvant tamox- (MORE) trial [abstract 3]. Proc Am Soc Clin
ifen therapy and radiotherapy for early breast Oncol 1998;17:2a.
cancer. J Natl Cancer Inst 1991;83:1013–7. 101. Jordan VC, Glusman JE, Eckert S, et al. Incident
90. Rutqvist LE, Johansson H, Signomklao T, et al. primary breast cancers are reduced by ralox-
Adjuvant tamoxifen therapy for early stage ifene: integrated data from multicenter, double-
breast cancer and second primary malignan- blind, randomized trials in ~ 12,000 postmeno-
cies. J Natl Cancer Inst 1995;87:645–51. pausal women [abstract 466]. Proc Am Soc
91. Chang J, Powles TJ, Ashley SE, et al. The effect Clin Oncol 1998;17:122a.
of tamoxifen and hormone replacement ther- 102. Valavaara R. Phase II trials with toremifene in
apy on serum cholesterol, bone mineral density advanced breast cancer: a review. Breast Can-
and coagulation factors in healthy post- cer Res Treat 1990;16:S31–5.
menopausal women participating in a random- 103. Brunning PF. Droloxifene, a new anti-oestrogen
ized, controlled tamoxifen prevention study. in postmenopausal advanced breast cancer: pre-
Ann Oncol 1996;7(7):671–5. liminary results of a double blind dose finding
92. Powles TJ, Hickish T, Kanis JA, et al. Effect of phase II trial. Eur J Cancer 1992;28A:1404–7.
tamoxifen on bone mineral density measured 104. Sato M, Turner CH, Wang T, et al. LY353381.HC1:
a novel raloxifene analog with improved SERM
by dual-energy x-ray absorptiometry in healthy
potency and efficacy in vivo. J Pharmacol Exp
premenopausal and postmenopausal women. J
Ther 1998;287(1):1–7.
Clin Oncol 1996;14(1):78–84.
105. Toko T, Saito H, Fujioka A, et al. Antitumor
93. Powles TJ, Jones AL, Ashley SE, et al. The Royal
activity of miproxifene phosphate (TAT-59)
Marsden Hospital pilot tamoxifen chemopre-
against human mammary carcinoma. Gan to
vention trial. Breast Cancer Res Treat 1994;31:
Kagaku Ryoho 1998;25(6):829–38.
73–82.
106. Sporn MB, Roberts AB. Role of retinoids in dif-
94. Fisher B, Joseph P, Costantino D, et al., and other ferentiation and carcinogenesis. Cancer Res
NSABP investigators. Tamoxifen for preven- 1983;43:3034–40.
tion of breast cancer: report of the National 107. Lotan R. Retinoids and apoptosis: implications
Surgical Adjuvant Breast and Bowel Project for cancer chemoprevention and therapy. J Natl
P-1 study. J Natl Cancer Inst 1998;90:1371–88. Cancer Inst 1995;87:1655–7.
95. Early Breast Cancer Trialists’ Collaborative 108. Delia D, Aillo A, Formelli F, et al. Regulation of
Group. Tamoxifen for early breast cancer: an apoptosis induced by the retinoid N-(4-hydroxy-
overview of the randomized trials. Lancet phenyl) retinamide and effect of deregulated
1998;351:1451–67. bcl-2. Blood 1995;85:359–67.
96. Veronesi U, Maisonneuve P, Costa A, et al. Pre- 109. Mangelsdorf DJ, Umesono K, Evans RM. The
vention of breast cancer with tamoxifen: pre- retinoid receptors. In: Sporn MB, Roberts AB,
liminary findings from the Italian randomized Goodman DS, editors. The retinoids: biology,
trial among hysterectomised women. Italian chemistry, and medicine. 2nd ed. New York:
Tamoxifen Prevention Study. Lancet 1998; Raven Press; 1994. p. 319–49.
352:93–7. 110. Toma S, Isnardi L, Raffo P, et al. Effects of ALL-
97. Anzano MA, Peer CW, Smith JM, et al. Chemo- trans-retinoic acid and 13-cis-retinoic acid on
prevention of mammary carcinogenesis in the breast-cancer cell lines: growth inhibition and
rat: combined use of raloxifene and 9-cis- apoptosis induction. Int J Cancer 1997;70:
retinoic acid. J Natl Cancer Inst 1996;88:123–5. 619–27.
Breast Cancer Risk and Management: Chemoprevention, Surgery, and Surveillance 39

111. Bishoff ED, Gottardis MM, Moon TE, et al. therapy for early breast cancer. The Women’s
Beyond tamoxifen: the retinoid X receptor- Intervention Nutrition Study. J Clin Oncol
selective ligand LGD1069 (TARGRETIN) 1993;11:2072–80.
causes complete regression of mammary carci- 124. Willett WC, Stampfer MJ. Sobering data on alco-
noma. Cancer Res 1998;53:479–84. hol and breast cancer. Epidemiology 1997;
112. De Palo G, Camerini T, Marubini E, et al. Chemo- 8(3):225–7.
prevention trial of contralateral breast cancer 125. Mezzetti M, La Vecchia C, Decarli A, et al. Pop-
with fenretinimide. Rationale, design, method- ulation attributable risk for breast cancer: diet,
ology, organization, data management, statistics nutrition, and physical exercise. J Natl Cancer
and accrual. Tumori 1997;83:884–94. Inst 1998;90(5):389–94.
113. De Palo G, Veronesi U, Camerini T, et al. Can 126. Jirtle RL, Haag JD, Ariazi EA, Gould M.
fenretinide protect women against ovarian can- Increased mannose 6-phosphate/insulin-like
cer ? J Natl Cancer Inst 1995;87:146–7. growth factor II receptor and transforming
114. Cancer incidence in five continents. Age-stan- growth factor b-1 levels during monoterpene-
dardized incidence rates, four-digit rubrics, induced regression of mammary tumors. Can-
and age-standardized and cumulative incidence cer Res 1993;53:3849–52.
rates, three-digit rubrics. IARC Sci Publ 1992; 127. Messina MJ, Pensky V, Setchell KD, Barnes S. Soy
120:871–1011. intake and cancer risk: a review of the in vitro
115. Dhingra K. A phase II chemoprevention trial and in vivo data. Nutr Cancer 1994;21:113–31.
designed to identify surrogate endpoint bio- 128. Bardon S, Picard K, Martel P. Monoterpenes
markers in breast cancer. J Cell Biochem 1995; inhibit cell growth, cell cycle progression. Nutr
23Suppl:19–24. Cancer 1998;32(1):1–7.
116. Mariani L, Formelli F, De Palo G, et al. Chemo- 129. Lamartiniere CA, Zhang JX, Cotroneo MS.
prevention of breast cancer with fenretinide (4- Genistein studies in rats: potential for breast
HPR): study of long-term visual and ophthal- cancer prevention and reproductive and devel-
mologic tolerability. Tumori 1996;82:444–9. opmental toxicity. Am J Clin Nutr 1998;68(6
117. National Cancer Institute. Annual Cancer Statis- Suppl):1400S–5S.
tics Review including Cancer Trends: 1950- 130. Temple WJ, Lindsay RL, Magi E, Urbanski SJ.
1985. Bethesda, MD: U.S. Department of Technical considerations for prophylactic mas-
Health and Human Services; 1988. tectomy in patients at high risk for breast can-
118. Wynder EL, Cohen LA, Muscat JE, et al. Breast cer. Am J Surg 1991;161:413–5.
cancer: weighing the evidence for a promoting 131. Wong JH, Jackson CF, Swanson JS, et al. Analy-
role of dietary fat. J Natl Cancer Inst 1997; sis of the risk reduction of prophylactic partial
89:766–75. mastectomy in Sprague-Dawley rats with 7,12-
119. Rose DP, Connolly JM, Liu XH. Dietary fatty dimethylbenzanthracene-induced breast can-
acids and human breast cancer cell growth, cer. Surgery 1986;90:67–71.
invasion, and metastasis. Adv Exper Med Biol 132. Nelson H, Miller SH, Buck D, et al. Effectiveness
1994;364:83–91. of prophylactic mastectomy in the prevention
120. Bagga D, Capone S, Wang HJ, et al. Dietary mod- of breast tumors in C3H mice. Plast Reconstr
ulation of omega-3/omega-6 polyunsaturated Surg 1989;83:662–9.
fatty acid ratios in patients with breast cancer. 133. Pennisi VR, Capozzi A. Subcutaneous mastec-
J Natl Cancer Inst 1997;89:1123–31. tomy data: a final statistical analysis of 1500
121. Zheng W, Gustafson DR, Sinha R, et al. Well- patients. Plast Surg 1989;13:15–21.
done meat intake and the risk of breast cancer. 134. Woods JE, Meland NB. Conservative manage-
J Natl Cancer Inst 1998;90:1724–9. ment in full-thickness nipple-areolar necrosis
122. Boyd NF, Greenberg C, Lockwood G, et al. after subcutaneous mastectomy. Plast Reconstr
Effects at two years of a low-fat, high-carbohy- Surg 1989;84:258–64.
drate diet on radiologic features of the breast: 135. Schrag D, Kuntz KM, Garber JE, Weeks JC.
results from a randomized trial. J Natl Cancer Decision analysis: effects of prophylactic mas-
Inst 1997;89:488–96. tectomy and oophorectomy on life expectancy
123. Chlebowski RT, Blackburn GL, Buzzard JM, et among women with BRCA1 or BRCA2 muta-
al. Adherence to a dietary fat intake reduction tions. N Engl J Med 1997;336:1465–71.
program in postmenopausal women receiving 136. Lynch HT, Lemon SJ, Durham C, et al. A descrip-
40 BREAST CANCER

tive study of BRCA1 testing and reactions to dis- women with a family history of breast cancer.
closure of test results. Cancer 1997;79:2219–28. N Engl J Med 1999;340:77–84.
137. Willemsen HW, Kaas R, Peterse JH, Rutgers EJ. 143. Singletary SE, Taylor SH, Guinee VF. Occur-
Breast carcinoma in residual breast tissue after rence and prognosis of contralateral carcinoma
bilateral subcutaneous mastectomy. Eur J Surg of the breast. J Am Coll Surg 1994;178:390–6.
Oncol 1998;24:331–8. 144. Bilimoria MM, Morrow M. The woman at
138. Ziegler LD, Kroll SS. Primary breast cancer after increased risk for breast cancer: evaluation and
prophylactic mastectomy. Am J Clin Oncol management strategies. CA Cancer J Clin
1991;14:451–4. 1995;45:263–78.
139. Jameson MB, Roberts E, Nixon J, et al. Metastatic 145. Kroll SS, Miller MJ, Schusterman MA, et al.
breast cancer 42 years after bilateral subcuta- Rationale for elective contralateral mastectomy
neous mastectomies. Clin Oncol 1997;9:119–21. with immediate bilateral reconstruction. Ann
140. Goodnight JE, Quagliana JM, Morton DL. Failure Surg Oncol 1994;1:457–61.
of subcutaneous mastectomy to prevent devel- 146. Schairer C, Persson I, Falkeborn M, et al. Breast
opment of breast cancer. J Surg Oncol 1984; cancer risk associated with gynecologic
26:198–201. surgery and indications for such surgery.
141. Eldar S, Meguid MM, Beatty JD. Cancer of the Obstet Gynecol 1997;70:150–4.
breast after prophylactic subcutaneous mastec- 147. Parazzini F, Braga C, LaVecchia C, et al. Hys-
tomy. Am J Surg 1984;148:692–3. terectomy, oophorectomy in premenopause,
142. Hartmann LC, Schaid DJ, Woods JE, et al. Effi- and risk of breast cancer. Obstet Gynecol
cacy of bilateral prophylactic mastectomy in 1997;90:453–6.
3
Screening and
Diagnostic Imaging
JAN M. JESKE, MD
JOEL R. BERNSTEIN, MD
MARGARET A. STULL, MD

Mammography is currently the best available Diagnostic mammography is performed on


screening modality for early detection and patients presenting with signs or symptoms of
diagnosis of breast cancer. Periodic examina- potential breast pathology. The request for a diag-
tion of asymptomatic females with mammo- nostic mammogram should be considered a con-
graphy has been shown to reduce breast cancer sultation to provide evaluation of the patient’s
mortality.1 In accordance with the American symptoms or mammographic findings and make
Cancer Society recommendations, the available further management recommendations. Tai-
scientific data suggest a benefit from annual lored mammographic images, physical exami-
mammographic screening of all women begin- nation, and breast ultrasound are frequently
ning at the age of 40 years, combined with used to further investigate and explain a partic-
annual physical examination and monthly ular clinical or radiographic concern. Alterna-
breast self examination.2 For women between tive projections and magnification views often
20 and 39 years of age, the ACS recommends a supplement the standard mammogram. Spot
breast physical exam every three years and compression views of a particular site of con-
monthly breast self exam. Patients with a first- cern can improve visualization of an underlying
degree premenopausal relative diagnosed with lesion and allow for more accurate assessment
breast cancer may consider beginning annual of lesion margins. Magnification mammogra-
screening examinations 10 years prior to the phy is best suited for enhanced characterization
age at which the relative was diagnosed, in an and visualization of microcalcifications.
attempt to benefit from early detection.3 It is well known that mammography is
Screening mammography evaluates asymp- unable to detect every breast cancer. The false
tomatic women with the goal of discovering negative rate of mammography ranges from
unsuspected breast cancer at an early and four to thirty-four percent.4 The diagnosis of
potentially curable stage. The routine screening breast cancer therefore, is not excluded by a
mammogram is comprised of a craniocaudal negative mammogram. Patient management
(CC) and mediolateral oblique (MLO) image of should take into account the clinical assess-
each breast. Abnormalities detected on the ment, despite a negative mammogram.
screening exam are further evaluated with a When appropriate, the diagnostic exam
diagnostic mammogram. includes ultrasonography of a mammographic

41
42 BREAST CANCER

finding, palpable abnormality, or site of pain. ing to one of the following decision categories:
Breast sonography aids in the characterization
of mammographically detected masses and can 1. Category 0: incomplete. Needs additional
confirm equivocal radiographic findings. imaging evaluation. This category typically
Breast ultrasound is the initial imaging modal- arises after a screening mammography,
ity of choice for evaluating palpable masses in when the patient must be recalled for addi-
women < 30 years of age and in lactating and tional evaluation before a final assessment
pregnant women, as per the American College can be made.
of Radiology standards.5 2. Category 1: negative. The study is normal.
3. Category 2: benign finding. There is a
BREAST IMAGING REPORTING benign finding described and no evidence of
AND DATA SYSTEMS malignancy.
4. Category 3: probably benign finding—short
To improve the quality of mammography interval follow-up suggested. Lesions in this
reporting and early breast cancer detection, a category have imaging characteristics that
consortium of medical experts has developed are most likely benign. Follow-up is per-
the Breast Imaging Reporting Data System formed at a 6-month interval to establish sta-
(BIRADS).6 The American College of Radiol- bility of a lesion with low probability of can-
ogy (ACR), in collaboration with the National cer and detect those few cancers that initially
Cancer Institute, the Centers for Disease Con- present with benign morphology. Ninety-
trol and Prevention, the Food and Drug Admin- eight percent of these lesions subsequently
istration, the American Medical Association, the prove to be benign.7 This approach limits
American College of Surgeons, and the College unnecessary tissue sampling.
of American Pathologists, created BIRADS to 5. Category 4: suspicious abnormality—
standardize communication of mammographic biopsy should be considered. Lesions in this
results, reduce ambiguous breast imaging category have a 30 percent positive predic-
reports, and facilitate the collection and analysis tive value for being malignant; therefore,
of medical audit data at individual mammogra- biopsy is recommended.7
phy practices as well as at the national level. 6. Category 5: highly suggestive of malig-
There are four main sections in BIRADS: nancy—appropriate action should be taken.
breast imaging lexicon, reporting system, fol- These lesions have morphologic features
low-up and outcome monitoring, and ACR characteristic of cancer. Intervention is
National Mammography Database (NMD). The required. The positive predictive value for
lexicon provides standardized language for category 5 lesions is 97 percent.7
lesion characterization. It also provides descrip-
tive terms for masses, calcifications, architec- These assessment categories are consistent
tural distortion, and associated findings of skin with those mandated by the final regulations
and nipple retraction as well as trabecular under the Mammography Quality Standard Act
thickening and axillary adenopathy. (MQSA).8 These categories are not intended to
The reporting system uses a standardized replace clinical evaluation of the breast. Clini-
format for the mammographic report, noting cal assessment directs the ultimate course of
available comparison films, breast tissue com- action when the mammogram is “negative” and
position, a concise description of any signifi- there is concern about a clinically suspicious
cant findings, and a final assessment with abnormality.
appropriate recommendations. The mammo- The BIRADS section on follow-up and out-
graphic study is classified by BIRADS accord- come monitoring enables an individual radiolo-
Screening and Diagnostic Imaging 43

gist to assess his or her overall mammography shape, margins, and density. Round or oval
interpretation skills by performing a mammog- shaped masses are typically associated with a
raphy audit. There is a detailed description of benign etiology, most commonly a cyst or
the necessary core data to be collected and cal- fibroadenoma. Increasing lobulations, irregular
culated for a comprehensive medical audit. The shapes, and spiculations increase the probabil-
ACR BIRADS committee is encouraging mam- ity of malignancy.
mography practices to participate in the Assessment of the lesion margin adds
National Mammography Database. The pro- important distinguishing information. Cir-
gram will enable evaluation of mammographic cumscribed lesions with sharp, distinct mar-
screening in the diagnosis of clinically occult gins are almost always benign (Figure 3–1).
breast cancer at the national level. Poorly defined margins reflect the irregular
interface of the cancer cells invading the sur-
MAMMOGRAPHIC APPEARANCE rounding breast tissue. Due to superimposed
OF BREAST CANCER normal fibroglandular tissue, lesion margins
Breast cancer has numerous clinical and imaging can occasionally be obscured and difficult to
presentations. The classic mammographic appear- accurately assess. Spot compression views
ance of infiltrating breast cancer is an irregular and ultrasonography allow for further charac-
mass, often with ill-defined or spiculated mar- terization of lesion margins, particularly in
gins. In addition to a discrete mass, the invasive dense breast tissue.
tumor can also present as a subtle asymmetric
density or an architectural distortion. Clustered
pleomorphic calcifications are the common pre-
sentation of in situ carcinoma that may or may not
be associated with invasive disease.
Secondary signs of malignancy, often asso-
ciated with advanced stages of breast cancer,
are detectable clinically and radiographically as
areas of skin thickening or dimpling, nipple
retraction, and axillary adenopathy. Diffuse
skin thickening and breast edema manifested as
increased mammographic density are associ- A
ated with lymphangitic spread of cancer involv-
ing the dermal lymphatics with inflammatory
cancer. The underlying primary tumor is often
obscured by the diffuse breast edema. These
findings must be differentiated from mastitis.
Typically, the diagnosis of inflammatory cancer
is made clinically and by biopsy. Isolated nip-
ple and areolar thickening can occur in patients
with Paget’s disease of the nipple.

B
Mammographic Analysis of Masses
Figure 3–1. A, Mammographic image reveals the circum-
The mammographic mass is a space-occupying scribed and gently lobulated margins of this fibroadenoma. B,
Corresponding ultrasound shows the solid nature of this benign
lesion seen in two different projections. Mam- tumor as evidenced by the homogenous hypoechoic internal
mographic analysis of the mass is based on its architecture.
44 BREAST CANCER

ment (Figure 3–2). The complex cystic struc-


ture containing internal echoes or an intracystic
mass requires further evaluation so as to not
overlook pathology such as papillary carci-
noma or a necrotic neoplasm (see Figure 3–8).
Thorough sonographic evaluation of all lesion
margins and internal architecture is necessary
to detect subtle signs of malignancy. Subtle
margin irregularity and internal heterogeneity
may be the only findings to suggest a malignant
process. Sonographic features associated with
Figure 3–2. Sonographic depiction of a simple cyst. Note the
breast malignancy include marked hypo-
lack of internal echoes and increased echogenicity deep to echogenicity, irregular margins, and shadow-
the fluid (posterior acoustic enhancement). ing.9 Malignant lesion margins visualized with
ultrasound are often poorly defined, with an
Most breast cancers are mammographically angulated, microlobulated, or branching pattern.
dense (more radio-opaque) relative to an equal
volume of normal fibroglandular tissue. The Imaging Specific Types of
presence of radiolucent fat within a lesion is Infiltrating Breast Cancer
characteristic of a benign etiology. Fat contain-
ing lesions include hamartoma (fibroadeno- Approximately 85 percent of breast carcinomas
lipoma), lipoma, galactocele, fat necrosis, and arise from ductal structures, with the remaining
lymph nodes. 15 percent arising from lobular structures.
Infiltrating ductal carcinoma accounts for the
Sonographic Evaluation of Masses largest group of breast cancers, representing 65
to 80 percent of cases.10 The classic mammo-
Sonograpraphy is an excellent method for dis- graphic presentation of infiltrating ductal carci-
tinguishing a simple cyst from a solid, circum- noma is a high-density mass with spiculated
scribed tumor. On sonography, the simple cyst margins (Figure 3–3A). Sonographically, this
has smooth, rounded, or oval margins, contains lesion is typically seen as a shadowing, hypo-
no internal echoes, and has a sharply defined echoic mass with irregular margins (Figure
posterior wall with posterior acoustic enhance- 3–3B). The presentation of infiltrating ductal car-

A B
Figure 3–3. A, Classic mammographic appearance of infiltrating ductal carcinoma demonstrating irregular, spiculated margins.
B, Sonographic visualization of the same infiltrating ductal carcinoma illustrating a hypoechogenic mass with irregular margins.
Screening and Diagnostic Imaging 45

A B
Figure 3–4. A, Mammographic spot view of a 0.8 cm infiltrating ductal carcinoma with microlobulated and partially ill-defined
margins. B, Sonographic image of the same infiltrating ductal carcinoma demonstrating the marked irregularity of the tumor mar-
gins (arrows), despite the small size.

cinoma, however, can mimic a benign lesion with infiltrating lobular carcinoma appears sono-
partially circumscribed margins (Figure 3–4). graphically indistinguishable from infiltrating
Infiltrating lobular carcinoma is the second ductal carcinoma (Figure 3–5B).3
most common type of invasive breast cancer, Medullary, colloid, and papillary carcino-
representing approximately 15 percent of mas often present as partially circumscribed
cases.10 It has a higher rate of multicentricity mammographic masses. Medullary carci-
and bilaterality than infiltrating ductal carci- noma, accounting for approximately six per-
noma.11 Infiltrating lobular carcinoma is known cent of breast carcinomas, typically presents
for its insidious nature, delaying clinical and in patients < 50 years of age and can be mis-
mammographic diagnosis. The subtle nature of taken for a fibroadenoma (Figure 3–6). The
infiltrating lobular carcinoma is thought to be slow growing colloid carcinoma, also known
due to its pattern of single-file cellular infiltra- as mucinous carcinoma, comprises only two
tion and lack of associated desmoplastic reac- percent of all breast cancers and is more com-
tion.12 This tumor often presents as an evolving mon in older females (Figure 3–7). Papillary
asymmetric density, or, less often, a spiculated carcinoma represents less than one percent of
mass on mammography (Figure 3–5A).13,14 all breast cancers and is associated with spon-
Despite its elusive appearance on mammography, taneous serosanguinous nipple discharge. An

A B

Figure 3–5. A, The arrow identifies a vague, developing asymmetric density on this spot compression view. B, Despite the sub-
tle mammographic appearance, the ultrasound image clearly visualizes this infiltrating lobular carcinoma revealing the markedly
jagged margins.
46 BREAST CANCER

intracystic mass or intraductal lesion depicted raphy, indistinguishable from infiltrating ductal
by sonography raises concern for a papillary carcinoma (Figure 3–9). Tubular carcinoma can
neoplasm (Figure 3–8). be confused or associated with a radial scar
Tubular carcinoma accounts for less than (sclerosing papillomatosis), a benign entity
two percent of breast cancers. Due to its very (Figure 3–10).
slow growth, this tumor is typically small at the Phyllodes tumor, once termed “cystosarcoma
time of detection. Tubular carcinoma often pre- phyllodes,” comprises less than one percent of
sents as a small spiculated mass on mammog- breast tumors. Approximately ten percent of

B
B
Figure 3–7. A, This partially circumscribed mammographic
Figure 3–6. A, Mammographic image of a 6 cm circum- nodule represents a colloid (mucinous) carcinoma. B, The het-
scribed medullary carcinoma. B, Ultrasound reveals the het- erogeneous hypoechoic and lobulated appearance of this col-
erogeneous hypoechoic nature of this medullary carcinoma. loid carcinoma is easily visualized with ultrasound.
Screening and Diagnostic Imaging 47

A B
Figure 3–8. A, Papillary carcinoma with the typical appearance of a partially circumscribed mass. B, Sonographic image of the
same papillary neoplasm reveals the intracystic mass (arrow).

phyllodes tumors are malignant. This tumor can graphic detail of this finding as well as greater
present as a rapidly growing palpable mass. awareness of its importance. Current mammo-
Breast imaging usually shows a large rounded or graphic techniques can detect calcification in as
lobulated circumscribed mass. Cystic spaces can many as 50 percent of all breast cancers.16
be seen by sonography (Figure 3–11). Screening studies have shown that 90 percent of
Metastasis to the breast, although uncom- all cases of nonpalpable ductal carcinoma in situ
mon, can occur from a variety of primary malig- (DCIS)17 and 70 percent of minimal carcino-
nancies, including melanoma, lymphoma, lung mas18 were detected on the basis of microcalci-
cancer, and contralateral breast carcinoma. fications. Many women, however, have some
Mammographically, the metastatic lesions tend form of calcification in their breasts, the great
to be round and lack spiculations (Figure 3–12). majority of which are benign. Thus, we are chal-
lenged to both detect and analyze calcifications
BREAST CALCIFICATIONS seen on mammograms to accurately diagnose
breast cancer without incurring consequences of
The presence of suspicious microcalcifications a false positive or false negative study.
on a mammogram can make possible the early
diagnosis of clinically occult breast cancer. Since Detection
the description of calcifications on radiographs
of breast cancer by Leborgne in 195115 there have Nowhere in medical imaging are fine detail
been substantial improvements in the mammo- images as vital as they are in the detection and

A B
Figure 3–9. A and B, Mammographic and sonographic images demonstrating the irregular shape and spiculated margins of this
0.5 cm tubular carcinoma (arrow).
48 BREAST CANCER

A B
Figure 3–10. A and B, The imaging appearance of this benign radial scar (arrow) mimics the tubular carcinoma in Figure 3–9.

evaluation of breast microcalcifications, which of low suspicion calcifications. While some cal-
may be as small as 0.1 mm. It is necessary to cifications have classically benign or malignant
optimize the technique in all stages of produc- features that allow the mammographer to easily
tion of the mammographic image to yield high characterize them for appropriate action, there is
contrast, high resolution, and motion-free a sizable intermediate or indeterminate group
films. Viewing conditions, including the rou- that requires thorough analysis to assess the like-
tine use of a hand magnifying lens to system- lihood of malignancy. The characteristics most
atically search each film, are also important. useful in evaluating calcifications include size,
Magnification radiography in the study of number, form, distribution, and location.
breast calcifications, although at the cost of a
higher radiation dose (2´), provides greater Size
resolution than that achieved with a hand lens.
Calcifications associated with malignancy are
Analysis often as small as 0.1 to 0.3 mm in diameter and
usually < 0.5 mm. However, larger granular
Due to the frequency of calcifications on mam- forms, up to 2 mm, and longer fine linear forms
mograms, careful analysis is needed to recognize of calcification, may occasionally be seen. The
clearly benign calcifications and allow follow-up individual calcifications in cancer often vary in

A B
Figure 3–11. A, This large phyllodes tumor encompasses much of the mammographically visualized breast tissue. B, Sonogra-
phy confirms the solid nature of this mass, heterogeneous and hypoechoic.
Screening and Diagnostic Imaging 49

A B
Figure 3–12. A, Bilateral craniocaudal (CC) images demonstrating numerous round masses in this patient with multiple myeloma
metastasis. B, Sonographic image of the largest metastatic lesion in the right breast, depicting the heterogeneous hypoechoic nature.

size. In a group of mixed-size calcifications, cinoma, but are not the most common presenta-
the degree of suspicion should relate to the tion. Irregular granular forms are more fre-
smallest forms. quently seen, often differing in size and varying
from jagged “fractured crystal” shapes to round
Number punctate dots similar to lobular calcifications.
It is this overlap of benign and malignant
The presence of a focal group of five heteroge- shapes of granular calcifications that results in
neous microcalcifications in a volume of 1 cm3 the large indeterminate group of microcalcifi-
of tissue has been accepted as suspicious.19 cations requiring biopsy. Magnification studies
Biopsy of fewer calcifications may be per-
are invaluable in characterizing these calcifica-
formed if new, pleomorphic, or fine linear or
tions, allowing elimination of some typically
branching calcifications have developed since a
lobular forms from consideration for biopsy.
prior mammogram. Generally, the greater the
number of suspicious calcifications grouped in
a small area, the higher the chance of malig- Distribution
nancy.20,21 Magnification studies are used to Bilateral diffusely scattered calcifications are
more accurately determine the number of calci-
almost always benign and are often associated
fications present. The pathologist invariably
with adenosis. However, the calcifications of
finds more calcifications than are visible on the
adenosis or sclerosing adenosis may be focal
mammogram.
and indistinguishable from malignancy.
Malignant calcifications are usually found as
Shape
a focal cluster involving a small area of one
Small round to oval dense punctate calcifica- breast but can be more extensive, presenting
tions located in cystically dilated acini are con- as one or several clusters in the distribution of
sidered benign lobular calcifications. Malig- the ductal system of one lobe, or virtually an
nant calcifications are typically ductal in entire breast. While some benign masses con-
origin, forming in ductal cellular secretions or tain coarse calcifications, the presence of fine
necrotic cellular debris. Fine linear and branch- or pleomorphic calcifications associated with
ing ductal calcifications or pleomorphic calci- a mass increases the likelihood of malignancy
fications grouped to form a cast of the duct are and may suggest a related extensive intraduc-
most typical of malignancy, often comedo car- tal component.
50 BREAST CANCER

A B
Figure 3–13. Skin calcification. A, Lucent centered sebaceous gland calcification (arrow) appears intramammary on standard
projection. B, Tangential view confirms location of calcium in the skin (arrow).

Location Skin Calcification and Pseudocalcification

Calcifications must be proven to be within the Skin calcification is commonly related to seba-
breast to accurately evaluate their significance. ceous glands and appears as lucent, centered
Mimics of breast parenchymal calcification rings in a peripheral location (Figure 3–13A).
include skin calcium, artifacts, and pseudocal- Skin calcification may be punctate or irregular,
cifications. and may appear to lie within the breast
parenchyma on standard views. Therefore, a
Benign Calcifications tangential view skin localization study (Figure
3–13B) may be necessary to prove a cutaneous
Analysis of breast calcifications by an experi- location. Calcium in warts, moles, scars, and
enced mammographer will allow accurate diag- dermal lesions as well as pseudocalcifications
nosis of characteristically benign calcifications. due to tattoos, talc, deodorant, or film artifacts
The various types are described below. can be misleading (Figure 3–14).

Vascular Calcifications

Calcification caused by atherosclerosis in arter-


ial walls is usually easy to recognize due to the
typical continuous linear tubular pattern (Figure
3–15A). Early changes of short segment calcifi-
cations appearing as discontinuous deposits in
one wall may have a granular or fine linear
appearance that can arouse suspicion (Figure
3–15B). Magnification views in alternate pro-
jections will usually allow a correct diagnosis.

Calcium in Cysts

Figure 3–14. Pseudocalcification. Talc powder in moles


Thin, curvilinear calcifications defining the
beneath breast mimics parenchymal calcification. margin of a mass are seen with cyst wall calci-
Screening and Diagnostic Imaging 51

A B
Figure 3–15. Vascular calcification. A, Typical tubular arterial calcification. B, Irregular linear calcification, due to incomplete arte-
rial wall involvement, can appear suspicious.

fications. Intracystic calcium particles sus- ducts diffusely and often bilaterally. Large,
pended in fluid, known as “milk of calcium,” tubular periductal calcifications can appear in
may appear in multiple tiny cysts or a single plasma-cell mastitis (Figure 3–18). Increased
larger cyst. This diagnosis is best proven with a density of the subareolar parenchyma may be
90-degree lateral film showing a meniscus or found. These large, rod-like secretory calcifica-
teacup shape of layered calcium in a cyst (Fig- tions are usually easily differentiated from
ure 3–16). These calcifications may be difficult malignant calcifications by their large size and
to see when viewed en face in the CC view. greater length.

Fibroadenoma Fat Necrosis

Calcifications appear in fibroadenomas as a Calcifications due to fat necrosis are often seen
result of involution which may be due to myx- as fine-rim calcifications surrounding a lucent
oid degeneration, hyalinization, or infarction. center, varying in size from a few millimeters to
Early calcifications may occur in the periphery
of the mass and progress to large, geographic
areas of calcium the appearance of which has
been compared to popcorn (Figure 3–17).
Eventually, the soft-tissue component may be
completely replaced by a dense conglomerate
of calcifications. However, when this classic
pattern is not followed, an involuted fibroade-
noma may appear as fine pleomorphic calcifi-
cations without a visible mass and biopsy may
be required for diagnosis.

Secretory Calcification

Inspissated ductal secretions in normal or


dilated ducts may calcify to form solid, coarse, Figure 3–16. Milk of calcium. Layered sedimented calcium in
microcysts appears curvilinear or teacup-shaped on horizon-
and linear ductal casts involving one or more tal beam lateral film.
52 BREAST CANCER

A B C
Figure 3–17. Fibroadenoma. A, Few early coarse peripheral calcifications. B, Classic popcorn calcification. C, Large dense cal-
cification nearly replaces mass.

several centimeters. Small ring forms, usually these situations, careful analysis of the calcifi-
< 5 mm in diameter, are often idiopathic (Figure cations with magnification views may allow
3–19A). Dystrophic calcifications deposited periodic follow-up, but biopsy will often be
after trauma, hemorrhage, surgical biopsy, and necessary to exclude carcinoma.
radiation may appear as larger and less regular
calcifications surrounding an oil cyst (Figure Malignant and
3–19B). It is important to note that this type of Indeterminate Calcifications
calcium may appear several years after a
lumpectomy and breast radiation. In its early Microcalcifications are present in as many as
stages, it can be difficult to differentiate from 50 percent of all breast cancers and in an even
recurrent malignant calcifications. higher percentage of stage 0 and stage 1 breast
cancers.16–18 The presence of clustered micro-
Lobular Calcifications: calcifications may be the only indication of
Adenosis/Sclerosing Adenosis early preinvasive malignancy. Mammographic

Lobular calcifications form in the acini in asso-


ciation with such entities as adenosis, scleros-
ing adenosis, atypical lobular hyperplasia, and
cystic hyperplasia. Characteristically, these cal-
cifications are small, dense, and round (Figure
3–20A). If a lobule is distorted by surrounding
sclerosis, the individual forms may be more
irregular. The distribution of calcifications in
adenosis and sclerosing adenosis is often bilat-
eral, diffuse, and inhomogeneous due to vari-
able involvement of individual lobules. These
characteristic findings indicate a benign
process. Alternatively, the calcifications can be
more focal, presenting as unilateral loosely
grouped calcifications, regional calcifications,
Figure 3–18. Secretory calcification. Large, solid rod-like and
or a solitary small cluster (Figure 3–20B). In tubular calcifications appear in a ductal orientation.
Screening and Diagnostic Imaging 53

A B
Figure 3–19. Fat necrosis. A, Dense round lucent-centered calcifications caused by idiopathic fat necrosis. B, Postoperative oil
cysts with thin eggshell (white arrow) and course rim calcification (black arrow).

detection of microcalcifications in patients fine, pleomorphic, linear, and branching calcifi-


with DCIS accounts for this entity rising from cations (Figure 3–21) or multiple irregular gran-
a small percentage of lesions found at biopsy to ules forming castings arranged in a ductal distri-
the current rate of 20 to 40 percent for biopsies bution (Figure 3–22). The extent of involvement
for clinically occult lesions.22 Stomper and col- may vary from < 1 cm to an entire lobule or even
leagues,23 in a group of 100 patients with a whole breast (Figure 3–23). Holland observed
DCIS, reported that 84 percent of cases pre- a significant discrepancy between the estimated
sented with microcalcifications, either alone mammographic and actual histopathologic
(72%) or as calcifications associated with a extent of DCIS.24 This discrepancy is most pro-
soft-tissue density (12%). nounced for low grade DCIS. Mammography
Classic malignant calcifications are typically underestimates the histopathologic extent by 16
associated with comedo carcinoma but are also percent for high grade DCIS and 47 percent for
present in other histologic subtypes of DCIS. low grade DCIS.24
Characteristic malignant calcifications occur as Clustered irregular granular calcifications,

A B
Figure 3–20. Lobular calcification. A, Punctate, round, scattered calcifications (were bilateral) due to adenosis. B, Small group
of round clustered calcifications (arrow), likely acinar, in a dilated lobule.
54 BREAST CANCER

calcifications (Figure 3–24), that may have the


most similarity to benign forms of calcium and
thus require the greatest scrutiny. Invasive
breast cancer associated with DCIS involving

Figure 3–21. Linear and branching calcifications typical of


comedo type ductal carcinoma in situ.

not clearly ductal in distribution, or mixed


forms of granular and casting calcifications, are
A
a more common presentation of DCIS (65%)
compared to the classic pure casting and linear
forms (35%).23 These granular calcifications are
seen more frequently in low-grade, noncomedo
carcinoma, although there is enough overlap
that one cannot reliably subtype DCIS based on
the mammographic morphology. It is also this
group, because of the variability of the granular

Figure 3–23. Distribution of microcalcifications in ductal car-


Figure 3–22. Granular calcifications forming ductal casts cinoma in situ. A, focal (arrow); B, segmental; C, diffuse (whole
(arrows) in comedo type ductal carcinoma in situ. breast).
Screening and Diagnostic Imaging 55

25 percent or more of the area of the tumor is situ component and defining the margins of the
classified as an extensive intraductal component resection. Magnification mammography is
(EIC) (Figure 3–25). Magnification mammog- needed to accurately determine the number,
raphy is helpful in defining the extent of the in shape, and distribution of calcifications. How-
ever, even after careful study, clustered granular
calcifications are often considered indetermi-
nate and biopsy is performed, resulting in a
yield for malignancy of 20 to 33 per-
cent.20,21,25,26

GALACTOGRAPHY

Galactography or ductography may be used to


evaluate patients presenting with spontaneous
isolated bloody or clear nipple discharge.
Numerous studies document a 10 to 15 percent
incidence of carcinoma in women with sponta-
neous unilateral discharge from a single
duct.27–29 The incidence of carcinoma in
A patients with bloody versus serous discharge is
similar.27 Other types of discharge, including
green, yellow, or milky discharges, have not
been associated with carcinoma.12
Galactography is not indicated in pregnant or
lactating women or when the discharge occurs
from multiple bilateral ducts. Galactography

C
Figure 3–24. Variability of calcifications in non-comedo duc-
tal carcinoma in situ. A, round, punctate granules; B, pleomor-
phic granules (arrow) which vary in size; V, “fractured crystal,” Figure 3–25. Extensive intraductal component. Calcifications
highly irregular granules (arrow). within a tumor mass and extention into surrounding tissue.
56 BREAST CANCER

postradiation developments and that of recurrent


carcinoma is critical for patient care.

Postexcision Mammography

Magnification mammography is useful after


surgery to ensure complete excision of the
malignant lesion. If the targeted lesion and all
tumor-related calcifications are not clearly
included on the specimen radiograph, or if
there is discordance between the pathology
results and the preoperative diagnosis, magnifi-
Figure 3–26. Galactographic image demonstrating the
abrupt duct termination due to an intraductal filling defect
cations mammography can reveal the retained
(arrow), in this case, a benign intraductal papilloma. primary lesion or residual malignant calcifica-
tions. Postoperative magnification mammogra-
phy is useful prior to radiation to ensure com-
should not be performed on a patient with active
plete excision of the calcium-containing tumor.
mastitis because it may worsen the inflammation.
Unfortunately, mammography cannot defini-
Evaluation of women with bloody or serous
tively predict the histologic extent of tumors.
nipple discharge should begin with mammogra-
Mammography performed within the first
phy. If the mammogram is unrevealing, ductog-
few weeks after tumorectomy is often limited
raphy can be performed by painlessly cannulat-
by the patient’s discomfort, breast edema, post-
ing the discharging duct and gently injecting
surgical architectural distortion, and the pres-
radiographic contrast material. Postinjection
ence of postoperative fluid collections (ie, sero-
mammographic images reveal intraductal fill-
mas and hematomas) (Figure 3–27A). Post-
ing defects or abrupt duct termination when
excision changes frequently result in increased
pathology is present (Figure 3–26). The benign
density that can obscure subtle residual malig-
intraductal papilloma is the most common
nant calcifications (Figure 3–27B).
cause of spontaneous serosanguinous nipple
Although these alterations regress and stabi-
discharge. Benign duct ectasia may also cause
lize with time, they are accentuated and pro-
nipple discharge.
longed by subsequent radiation therapy.30,31
Prior to surgical excision of the ductal
lesion, preoperative galactography can be per-
formed with a mixture of iodinated contrast Postradiation Mammography
material and methylene blue dye to enable and Long-Term Follow-up
intraoperative localization of the involved duct.
Mammography is important for long-term mon-
It has been suggested that this technique can
itoring to evaluate for recurrent disease or new
allow a more accurate and limited resection.27
lesions in either breast. This should commence
with a post-treatment baseline mammogram
EVALUATION OF THE being performed within 3 to 9 months following
CONSERVATIVELY TREATED BREAST tumor excision and completion of radiation
therapy. Standard views may be supplemented
Mammography is an essential tool for monitor-
with special projections to fully define post-
ing conservatively treated breast cancer patients.
therapy changes. Magnification mammography
Recognizing the distinctions between mammo-
is particularly important when evaluating the
graphic appearance of the expected postsurgical,
breast for retained or recurrent malignant
Screening and Diagnostic Imaging 57

microcalcifications. Subsequent annual or more Recurrent Breast Cancer


frequent mammograms should be obtained as
indicated by clinical or radiographic evaluation. The mammographic indications of tumor recur-
Comparison of the post-treatment mammo- rence at the surgical site frequently develop
grams to preceding studies is necessary to accu- between 2 to 3 years following conservative
rately assess radiographic changes following breast surgery.33 The development of increased
completion of therapy. Among the most com-
mon post-therapy changes are breast edema,
skin thickening, postoperative fluid collections,
scarring, fat necrosis, and calcifications.
The mammographic findings of skin thick-
ening, irregular breast parenchyma, and breast
edema following surgery and radiation are most
prominent on the post-treatment baseline study
and typically diminish over 2 to 3 years follow-
ing conservative therapy.30,32 Once mammo-
graphic stability of the breast has been estab-
lished, any increase in the architectural
distortion or enlargement of the dense scar at
the surgical site suggests the presence of recur-
rent tumor. Interrupted lymphatic drainage after
extensive axillary node dissection may produce
chronic breast edema. Recurrent edema with
erythema may be a manifestation of infection
(mastitis), inflammatory breast carcinoma, or
recurrent breast cancer with lymphatic involve- A
ment. Postoperative fluid collections, such as
hematomas or seromas at the lumpectomy site,
present mammographically as high-density oval
masses that may have ill-defined or spiculated
margins (see Figure 3–27). These diminish in
size as the fluid is resorbed over a period of 6 to
18 months.30,31 If the fluid collection is enlarg-
ing or an abscess is suspected, ultrasonography
can be used to evaluate the process further and
to guide diagnostic needle aspiration.
Coarse, benign, dystrophic calcifications
can develop several years after radiation and
surgery. These calcifications frequently repre-
sent fat necrosis or occasionally calcified
suture material in the surgical bed. Sometimes B
the developing benign calcifications have an
Figure 3–27. Postoperative fluid collection obscuring resid-
indeterminate appearance necessitating tissue ual malignant microcalcifications. A, The large high density
sampling. Occasionally fat necrosis can present oval mass with ill-defined margins represents the postsurgi-
as an irregular mass-like lesion that may simu- cal fluid collection. B, Magnification view 2 months later
shows postsurgical scar with resorption of the seroma and
late tumor recurrence. adjacent subtle residual malignant calcifications ( arrows).
58 BREAST CANCER

A B C
Figure 3–28. Recurrent infiltrating ductal carcinoma. Sequential views of the right breast show
A, the spiculated primary tumor (arrow) in the lateral breast; B, post-lumpectomy changes; and
C, recurrent invasive disease (arrow) in the surgical bed.

skin thickening, progressive architectural distor- EVALUATION OF


tion, enlargement of the surgical scar, a new soft- THE AUGMENTED BREAST
tissue mass, or new pleomorphic microcalcifica-
tions raise the suspicion of recurrent disease The presence of breast implants causes technical
(Figures 3–28 and 3–29). Calcifications associ- problems that impair the ability to detect breast
ated with recurrent DCIS frequently resemble cancer by mammography. The radiopaque
the mammographic appearance of the original implant blocks x-ray transmission, which limits
tumor.34 Contrast-enhanced MR imaging may the imaging of breast tissue. Implants compress
help distinguish scar tissue from recurrent dis- breast tissue against the skin which can obscure
ease.35 Computed tomography (CT) scanning a significant amount of anterior breast tissue on
can be used to assess extensive local breast dis- conventional mammographic images. Implants
ease (Figure 3–30), revealing direct chest-wall also diminish the compressibility of the breast,
invasion or tumor recurrence in the chest wall. particularly if there is capsular contracture.
Computed tomography is also useful in assess- Improved visualization of the anterior breast
ing regional and distant metastatic disease. tissue is provided by implant displacement

A B

Figure 3–29. Recurrent DCIS and invasive ductal carcinoma developing 3 years after breast conservation therapy. A, Image on
12/96 shows architectural distortion at the lumpectomy site (arrow). B, Spot magnification view demonstrates faint microcalcifica-
tions (curved arrow) adjacent to enlargement of the surgical scar representing DCIS associated with invasive tumor (straight arrow).
Screening and Diagnostic Imaging 59

(Figure 3–32). Lesions are usually more con-


spicuous on the implant displacement views
(see Figure 3–31). Patients with cosmetic aug-
mentation or reconstruction with implants may
develop parenchymal scarring that should not
be confused with a malignant process. Dys-
trophic calcifications and calcifications associ-
ated with the fibrous capsule surrounding the
implant can occur but are usually clearly benign
in appearance.
Ultrasonography of the augmented breast
assists in the evaluation of palpable and mam-
mographically detected masses (Figure 3–33).
Ultrasound can identify a palpable implant valve
and distinguish a focal implant herniation or
Figure 3–30. Recurrent infiltrating ductal carcinoma after left contained implant rupture from a breast
mastectomy and radiation therapy. CT scan shows dermal
involvement (short white arrows), left chest wall and axillary parenchymal abnormality, eliminating the need
recurrence with direct invasion into the mediastinum and sub-
pleural space (long white arrow). Confluent pathologic medi-
astinal adenopathy (black arrows) is present. Tumor surrounds
the left axillary clips.

views (Figure 3–31). This modified positioning


technique supplements the standard views in
women with cosmetic augmentation. Unfortu-
nately, breast tissue near the chest wall is not
completely imaged with either standard or
modified views.
Breast cancer has the same mammographic
features in women with or without implants

Figure 3–31. Multicentric DCIS in a woman with subpectoral


implants. Implant displacement and standard MLO view (pho-
tographed back to back for comparison) show multiple groups Figure 3–32. Multifocal invasive disease with EIC adjacent to
of pleomorphic microcalcifications (arrows). The malignant implant. Mammographic image reveals three irregular masses
calcifications are more conspicuous on the implant displace- (arrows) with bridging spicules and pleomorphic malignant
ment image. calcifications in the right axillary tail and inferior axilla.
60 BREAST CANCER

for tissue sampling.30,36 If a suspicious or inde- MAGNETIC RESONANCE


terminate parenchymal abnormality is con- IMAGING OF THE BREAST
firmed, ultrasound guided fine-needle aspiration
or core biopsy limits the risk of implant rupture. High–spatial–resolution MR imaging of the
Magnetic resonance imaging is widely used breast is evolving as an important adjunctive
to evaluate prosthetic implant integrity and can diagnostic tool for the detection, characteriza-
aid in differentiating an implant complication tion, staging, and monitoring of breast cancer.
from an intramammary lesion.37,38 Dynamic, Contrast-enhanced MR imaging may allow
contrast-enhanced MR imaging shows promise more accurate preoperative evaluation of pri-
for improved detection and monitoring of mary malignant breast lesions that may be
breast cancer in certain women with cosmetic underestimated or not seen on mammography or
breast augmentation or breast reconstruction by ultrasonography. Magnetic resonance imag-
using prosthetic implants.36 ing has revealed unsuspected multifocal, multi-
centric, diffuse, and bilateral disease in patients
with a solitary mammographic lesion.39–41
The sensitivity of contrast-enhanced MR
imaging approaches 100 percent in the detec-
tion of invasive breast cancer when compared to
mammography and physical examination.35,39,41
The specificity of breast MR imaging ranges
from 37 to 97 percent.39,41,42 This wide range is
attributed to the overlap in contrast enhance-
ment of benign and malignant lesions. Higher
specificity for breast MR imaging can be
achieved using a dynamic contrast-enhanced
technique with three-dimensional imaging. On
dynamic contrast-enhanced studies, malignant
lesions typically exhibit rapid enhancement,
whereas benign lesions show slower or no
A
enhancement. False-positive enhancing lesions
include fibroadenomas, sclerosing adenosis,
radial scars, mastitis, atypical hyperplasia, lobu-
lar neoplasia and normal breast tissue during
various phases of the menstrual cycle.35–37
Magnetic resonance imaging is also capa-
ble of demonstrating mammographically and
clinically occult in situ carcinoma.39–46 High-
resolution contrast-enhanced dynamic MR
imaging may enable more accurate evaluation
of tumors in dense fibroglandular tissue and
B assist in screening of high-risk patients.35,39–41 It
may also be useful in differentiating recurrent
Figure 3–33. Stromal fibrosis presenting as an indeterminate
mass. A, Implant displacement view shows the oval lesion carcinoma from scarring and in evaluating the
(arrow) with partially defined margins located superficial to the response to neoadjuvant chemotherapy.39
implant. B, Ultrasound demonstrates the oval hypoechoic mass
directly subjacent to the dermis. The anterior aspect of the
Limitations of MR imaging include the high
intact hypoechoic implant is seen deep to the breast tissue. cost of the examination, limited availability of
Screening and Diagnostic Imaging 61

dedicated imaging equipment, and significant clinical studies are needed to determine the
overlap in the enhancement patterns of benign efficacy of the CAD methods.
and malignant lesions. Additional large multi-
institutional studies will help define the clinical CONCLUSION
usefulness and cost effectiveness of MR imag-
ing in the assessment of breast cancer. Increased public awareness of the importance
of breast cancer screening and continued tech-
nical advances in breast imaging will enhance
NUCLEAR MEDICINE TECHNIQUES
patient care by allowing early detection, stag-
FOR BREAST IMAGING
ing, and monitoring of the disease. Recognition
Scintigraphic imaging of the breast frequently of the diverse imaging presentations of breast
uses the single-gamma radiotracer technetium- cancer is crucial for early diagnosis and proper
99m (Tc-99m) sestamibi. Studies have indicated management.
95 to 97 percent sensitivity for breast tumors > 1
cm.47 The sensitivity, however, is poor for small, REFERENCES
nonpalpable or medially located lesions, ranging 1. Breast cancer screening for women ages 40–49.
from 26 percent for lesions < 0.5 cm to 56 per- NIH Consens Statement. 1997;Jan 21–23;
cent for lesions between 0.5 cm and 1 cm.47 The 15(1):1–35.
specificity for Tc-99m sestamibi imaging ranges 2. Feig SA, D’Orsi CJ, Hendrick RE, et al. American
from 73 to 90 percent.47,48 College of Radiology Guidelines for Breast
Cancer Screening. Am J Roentgenol 1998;171:
Breast cancer imaging has also been per- 29–33.
formed using positron emission tomography 3. Kopans DB. Breast imaging. Philadelphia: Lip-
(PET) imaging of a structural analog of glu- pincott-Raven Publishers; 1998.
cose, fluorodeoxyglucose (FDG). The sensitiv- 4. Huynh PT, Jarolimek, AM, Daye S. The false-
ity and specificity for PET scanning of the negative mammogram. Radiographics 1998;
18:1137–54.
breast range from 70 to 90 percent and 85 to 95
5. American College of Radiology (ACR). 1998
percent, respectively.47,48 As with scintigraphic Standards. Reston (VA): American College of
imaging, lesions < 1 cm are not reliably Radiology; 1998. p. 317.
detected with PET imaging. Improvements in 6. American College of Radiology (ACR). Breast
the ability to detect small lesions will be neces- imaging reporting and data system (BIRAD-
sary before clinical utility of scintigraphy and STM). 3rd ed. Reston (VA): American College
of Radiology;1998.
PET can be proven.
7. Orel SG, Kay N, Reynolds C, et al. BI-RADS cat-
egorization as a predictor of malignancy. Radi-
COMPUTER-AIDED DIAGNOSIS ology 1999;211:845–50.
8. Federal Register, Part II, Department of Health
Computer-aided detection and diagnosis and Human Services, 1997, October 28.
(CAD) is an evolving technology that functions 9. Stavros AT, Thickman D, Rapp CL, et al. Solid
breast nodules: use of sonography to distin-
as a “second reader” of the mammographic guish between benign and malignant lesions.
films.49 Current CAD requires digitizing the Radiology 1995;196:123–34.
mammography films to allow computer analy- 10. Adler DD. Imaging evaluation of spiculated
sis. The computer program identifies areas of masses. In: Friedrich M, Sickles EA, editors.
the breast that match certain prescribed tissue Radiological diagnosis of breast diseases.
Berlin: Springer-Verlag; 1997.
patterns, densities, or calcifications for further
11. Fisher ER. What is early breast cancer? In: Zander
review by the interpreting radiologist. Limita- J, Baltzer J, editors. Early breast cancer; histo-
tions of CAD include the increased film han- pathology, diagnosis, treatment. New York:
dling time and high start-up costs. Prospective Springer;1985.
62 BREAST CANCER

12. Bassett LW, Jackson VP, Johan R, et al. Diagnosis 27. Van Zee KJ, Perez GO, Minnard E, et al. Preoper-
of diseases of the breast. Philadelphia: W. B. ative galactography increases the diagnostic
Saunders Company; 1997. yield of major duct excision for nipple dis-
13. Mendelson EB, Harris KM, Doshi N, et al. Infiltrat- charge. Cancer 1998;82:1874–80.
ing lobular carcinoma: mammographic patterns 28. Tabar L, Dean PB, Pentek Z. Galactography: the
with pathologic correlation. Am J Roentgenol diagnostic procedure of choice for nipple dis-
1985;153:265–71. charge. Radiology 1983;149:31–8.
14. Newstead GM, Baute PB, Toth HK. Invasive lob- 29. Leis HP. Management of nipple discharge. World
ular and ductal carcinoma: mammographic J Surg 1989;13:736–42.
findings and stage at diagnosis. Radiology 30. Mendelson EB. Evaluation of the postoperative
1992;184:623–7. breast. Radiol Clin North Am 1992;30:107–38.
15. Leborgne R. Diagnosis of tumor of the breast by 31. Kopans DB. The altered breast: pregnancy, lactation,
simple roentgenography: calcification in carci- biopsy, mastectomy, radiation, and implants. In:
noma. Am J Roentgenol 1951;65:1–11. Kopans DB. Breast imaging. 2nd ed. Philadel-
16. Miller RR, Davis R, Stacey AJ. The detection and phia: Lippincott-Raven; 1997. p. 445–96.
significance of calcification in the breast. A 32. Brenner RJ, Pfaff JM. Mammographic features
radiological and pathological study. Br J after conservative therapy for malignant breast
Radiol 1976;49:12–26. disease: serial findings standardized by regres-
17. Feig SA, Shaber GS, Patchefsky A. Analysis of sion analysis. Am J Roentgenol 1996;167:
clinically occult and mammographically occult 171–8.
breast tumors. Am J Roentgenol 1977;128: 33. Mendelson EB, Tobin CE. Imaging the breast
403–8. after surgery and radiation therapy. In: Syl-
18. Moskowitz M. The predictive value of certain labus: a categorical course in breast imaging.
mammographic signs in screening for breast Radiological Society of North America; 1995.
cancer. Cancer 1983;51:1007–11. p. 175–84.
19. Sickles EA. Mammographic features of early breast 34. Liberman L, Van Zee KJ, Dershaw DD, et al.
cancer. Am J Roentgenol 1984;143:461–4. Mammographic features of local recurrence in
20. Egan RL, McSweeney MD, Sewell CW. Intra- women who have undergone breast-conserving
mammary calcification without an associated therapy for ductal carcinoma in situ. Am J
mass in benign and malignant disease. Radiol- Roentgenol 1997;168:489–93.
ogy 1980;137:1–7. 35. Viehweg P, Paprosch I, Strassinopoulou M, et al.
21. Muir BB, Lamb J, Anderson TS, Kirkpatrick AE. Contrast-enhanced magnetic resonance imag-
Microcalcification and its relationship to can- ing of the breast: interpretation guidelines. Top
cer of the breast: experience in a screening Magn Reson Imaging 1998;9:17–43.
clinic. Clin Radiol 1983;34:193–200. 36. Brenner RJ. Tumor detection in the augmented
22. Rebner M, Rajic V. Noninvasive breast cancer. breast. In: Gorczyca DP, Brenner RJ, editors. The
Radiology 1994;190:623–31. augmented breast: radiologic and clinical per-
23. Stomper PC, Connolly JL, Meyer JE, Harris JR. spectives. New York: Thieme; 1997. p. 154–69.
Clinically occult ductal carcinoma in situ 37. Gorczyca DP. Magnetic resonance imaging of the
detected with mammography: analysis of 100 failing implant. In: Gorczyca DP, Brenner RJ,
cases with radiographic-pathologic correlation. editors. The augmented breast: radiologic and
Radiology 1989;172:235–41. clinical perspectives. New York: Thieme; 1997.
24. Holland R, Hednriks JH, Verbeek AL, et al. p. 121–43.
Extent, distribution and mammographic/histo- 38. Reynolds HE. Evaluation of the augmented breast.
logical correlations of breast ductal carcinoma Radiol Clin North Am 1995;33:1131–45.
in situ. Lancet 1990;335:519–22. 39. Orel SG, Schnall MD, Powel, et al. Staging of sus-
25. Meyer JE, Kopans DB, Stomper PC, Lindfors KK. pected breast cancer: effect of MR imaging and
Occult breast abnormalities: percutaneous pre- MR-guided biopsy. Radiology 1995;196:
operative needle localization. Radiology 1984; 115–22.
150:335–7. 40. Mumtaz H, Hall-Craggs M, Davidson T, et al.
26. Feig SA. Mammographic evaluation of calcifica- Staging of symptomatic primary breast cancer
tions. In: Kopans DB, Mendelson EB, editors. with MR imaging. Am J Roentgenol 1997;169:
Syllabus: a categorical course in breast imag- 417–24.
ing. Radiol Soc North Am; 1995. 41. Nunes LW, Schnall MD, Orel SG, et al. Breast MR
Screening and Diagnostic Imaging 63

imaging: interpretation model. Radiology Menstrual cycle and age: influence on parenchy-
1997;202:833–41. mal contrast medium enhancement in MR imag-
42. Hulka CA, Smith BL, Sgroi DC, et al. Benign and ing of the breast. Radiology 1997;203:145–9.
malignant breast lesions: differentiation with 46. Soderstrom CE, Harms SE, Copit DS, et al.
echo-planar MR imaging. Radiology 1995; Three-dimensional RODEO breast MR imag-
197:33–8. ing of lesions containing ductal carcinoma in
43. Orel SG, Reynolds C, Schnall MD, et al. Breast situ. Radiology 1996;201:427–32.
carcinoma: MR imaging before re-excisional 47. Williams MB, Pisano ED, Schnall MD, et al.
biopsy. Radiology 1997;205:429–36. Future directions in imaging of breast diseases.
44. Stompre PC, Herman S, Klippenstein DL, et al. Radiology 1998;206:297–300.
Suspect breast lesions: findings at dynamic 48. Wahl RL. Nuclear medicine techniques in breast
gadolinium-enhanced MR imaging correlated imaging. Sem Ultrasound CT MRI 1996;17:
with mammographic and pathologic features. 494–505.
Radiology 1995;197:387–95. 49. Feig SA, Yaffe MJ. Digital mammography. Radi-
45. Muller-Schimpfle M, Ohmenhauser K, Stoll P, et al. ographics 1998;18:893–901.
4
Image-Directed Breast Biopsy
RICHARD E. FINE, MD

The recent increase in the detection of nonpal- NEEDLE LOCALIZATION


pable breast abnormalities requiring further BREAST BIOPSY
evaluation is thought to be the direct result of
more favorable participation in mammography The gold standard with which image-guided
screening. Appropriate diagnostic work-up percutaneous breast biopsy is compared is the
will lead to a relative increase in lesions that needle or wire localization open surgical breast
are of sufficient risk to warrant a biopsy. In biopsy. However, this traditional management
fact, it has been estimated that approximately of a suspicious nonpalpable breast abnormality
1.2 million breast biopsies are performed per is not without its own error rate. The inability to
year in the United States. Unfortunately, an successfully remove the appropriate lesion
average positive predictive value for mammog- ranges from 0.5 to 17 percent.10–15 Some of the
raphy of 20 percent (range 15 to 35%) will reasons given for unsuccessful biopsies include
yield a significant number of biopsies per- (1) poor radiologic placement of the localiza-
formed for benign disease.1–4 If 5 women are tion wire, (2) preoperative and intraoperative
identified on the mammogram to have a lesion dislodgment of the wire, (3) surgical inaccu-
requiring biopsy, only 1 of these 5 women will racy and inadequacy in excising the appropriate
be found to have breast cancer. Therefore, if tissue, (4) failure to obtain a specimen radio-
traditional methods for histologic confirmation graph, and (5) the pathologist missing the focus
are used, all 5 women would proceed to the of disease when searching through a larger tis-
operating room for an open surgical biopsy sue sample provided by the surgeon.
after first having had wire localization in the The needle localization and open surgical
radiology suite. Image-guided percutaneous breast biopsy is typically more invasive.
breast biopsy, an effective alternative, has Although a surgeon may discount the impor-
recently gained favor. Image-guided percuta- tance of a scar on the breast, women frequently
neous breast biopsy would provide a secondary have a great concern over even a one- to two-
level of screening for these five women in a less inch scar, especially on the superior aspect of the
invasive, cost-effective manner as well as a his- breast. The possibility of altered breast shape
tologic diagnosis without sacrificing accu- associated with tissue removal is also important.
racy.5–9 The patient with breast cancer may then This fear is thought to be responsible for women
proceed to definitive surgical management and failing to participate in recommended screening.
the other four women with a benign diagnosis In addition to cutaneous scarring, parenchy-
may be placed in an appropriate follow-up pro- mal scarring may also complicate future mam-
tocol. It is with this concept in mind that we mographic follow-up.6 Kopans has suggested
review the state-of-the-art in image-guided per- that significant parenchymal scarring is rarely
cutaneous breast biopsy. associated with a properly performed needle

65
66 BREAST CANCER

localization breast biopsy.12 However, surgeons comfort while improving the technical ease and
are frequently faced with mammographic success of removing the lesion with a needle
reports indicating architectural distortion at the localization and open surgical breast biopsy.10
site of a prior biopsy that might mimic the While planning the incision site, cosmesis
changes associated with a malignancy. should be taken into consideration without
Despite the potential advantages of image- ignoring cancer surgery principles.18 If a lesion
guided percutaneous breast biopsy, there are has a relatively low probability of malignancy
still reasons why a standard needle localization and is within a reasonable distance from the
open surgical biopsy may be chosen for histo- nipple-areolar complex, a circumareolar inci-
logic diagnosis. Some patients desire complete sion should be considered. Regardless of the
surgical removal of a breast abnormality and wire insertion site, localization mammography
will not be satisfied with a “sampling proce- should be used to estimate the location of the
dure.” Certain facilities and insurance plans do lesion within the breast.21 The breast incision
not provide access to facilities where image- does not necessarily require inclusion of the
guided procedures (ie, stereotactic) are per- guide wire insertion site. In addition to using
formed. There are also certain lesion character- localization mammography, familiarity with
istics, patient characteristics, and potential the localization wire lengths and inherent
pathologic entities that may render image- markings may aid in a more accurate estimation
guided breast biopsy difficult or inappropriate. of lesion location (Figures 4–1A to 4–1C).19–21
The essentials for a properly performed wire Once the lesion location is determined, the
localization breast biopsy include accurate local- incision is planned to avoid tunneling of a more
ization, a comfortable, confident patient, and suspicious lesion through benign breast tis-
appropriate surgical planning and tech- sue.22 The incision is carried straight down
nique.10,12–15 The radiologist most often localizes through the subcutaneous layer without the
the lesion with orthogonal mammography.12,16 development of any flaps until the body of the
Increasingly, stereotaxis and ultrasonography are wire is encountered and can be brought into the
used to identify the location of a nonpalpable confines of the biopsy cavity. This is designed
lesion.17,18 Whichever technique is used, it is to maintain more support during closure and to
important to have the wire within 1 cm of the avoid potential indentation.18,19 On the basis of
lesion for the localization to be considered accu- the relationship of the lesion to the tip of the
rate and to limit the potential for error.19 wire, the excision is performed.
The majority of these procedures are per- If the needle localization biopsy is being per-
formed under local anesthesia with or without formed as a follow-up to an abnormal lesion on
intravenous sedation.12,14,20 Appropriate instilla- image-guided percutaneous breast biopsy or a
tion of local anesthesia for both image-guided presumed malignancy, it is then performed as a
percutaneous biopsies and open surgical proce- needle localization lumpectomy.23 Margin
dures is important for gaining the patient’s con- assessment becomes crucial to the success of the
fidence in the earliest aspect of the procedure. procedure. A technique of assisting the patholo-
This will ensure a calmer patient through the gist with margin assessment involves intraopera-
remainder of the procedure. A skin wheal is tive inking of the margins by the surgeon using
raised using a small (27- or 30-gauge) needle the Davidson™ multicolor inking system. The
with 1 percent Xylocaine ™ (lidocaine). Deeper anterior, 12, 3, 6, and 9 o’clock positions as well
local anesthetic should be placed (25-gauge nee- as the deep margins may be marked with corre-
dle) as a block around the potential dissection sponding colors (red, green, blue, black, yellow,
site. Despite the added cost, some have found and orange), using a cotton-tipped applicator
sedation or general anesthesia to lessen the dis- (Figure 4–2). The specimen is then dipped into 3
Image-Directed Breast Biopsy 67

Figure 4–1. A, Based on the wire length, angle of insertion,


and the localization mammograms, the position of the wire tip
and lesion are determined and the incision planned. B, The
incision is carried down through subcutaneous tissue, the
wire is brought into the confines of the cavity and then a
2-0 silk suture is used for traction, and dissection is carried
out around the course and tip of the wire. C, The specimen
radiograph maybe magnified to determine the completeness
A of excision of microcalcifications.

B C

percent acetic acid (vinegar) to set the colors. If there is focal margin involvement of the main
Subsequently, the specimen is sent to the radiol- specimen, the resection is felt to be adequate, if
ogy department for a specimen radiograph and the additional margins are clear.
then on to the pathology department. The speci-
men is sent dry so as to allow further stabiliza-
tion of the ink prior to pathology sectioning.
When the probability of malignancy is high
or already confirmed, a technique to ensure
adequate margins may be instituted, especially
when the procedure is for a potential multifocal
process, such as ductal carcinoma in situ
(DCIS). Once the main biopsy/lumpectomy
specimen is removed, random additional mar-
gins may be taken from the walls of the biopsy
cavity (12, 3, 6, 9 o’clock, and deep positions).
The thickness of the additional margins is
approximately 5 to 10 mm. The multicolor ink-
ing system is used to mark the side of the spec-
Figure 4–2. The Davidson™ multicolor inking system is used
imen representing the new margin of resection. to identify margins of resection.
68 BREAST CANCER

After hemostasis is obtained with electro- Stereotactic Breast Biopsy: Equipment


cautery, the wound is closed. Closure consists
only of reapproximating the subcuticular and Percutaneous breast biopsy for nonpalpable dis-
dermal layers. There is neither draining nor ease requires imaging. The two main imaging
reconstruction of the deep aspect of the biopsy modalities are stereotaxis and ultrasonography.
cavity. If postoperative radiation therapy is a These modalities are complementary to one
probability for the patient, small hemoclips are another and therefore knowledge of both is
placed in the walls and base of the cavity. This required to provide the physician with the full
will assist the radiation oncologist in planning range of options. Stereotactic breast biopsy is
therapy (especially the boost therapy required performed using specialized mammography
for close margins or used as a standard treat- equipment. The equipment obtains stereo mam-
ment in many centers). A clear, waterproof, mogram images of a lesion within the breast
Tegaderm™ dressing is applied, which allows and then relies on computerized triangulation
the patient to shower or bathe in the immediate of the targeted lesion to calculate the three-
postoperative period. dimensional position of this lesion.5,24 There are
The patient returns to the office during the two main types of stereotactic equipment, the
following week for wound assessment, to dis- upright add-on and the dedicated prone.25–27
cuss the pathology, and to plan future follow- Add-on stereotactic equipment uses standard
up. A baseline mammogram of the biopsied upright mammography with an attachable plat-
breast is obtained after 6 months to look for any form to perform targeting and biopsy (Figure
parenchymal scarring and to evaluate for 4–3). Add-on stereotactic units provide the
appropriate and adequate biopsy. If the preop- advantage of maximum use of the equipment,
erative mammogram contained suspicious cal- which has dual capabilities: screening or diag-
cifications at the site of the biopsy, a baseline nostic mammography and stereotactic breast
mammogram should be obtained prior to the biopsy. This can provide considerable cost sav-
initiation of radiotherapy. ings by avoiding not only dedicated equipment
but also dedicated space within a breast diag-
nostic facility. Despite these potential advan-
IMAGE-GUIDED BREAST BIOPSY

The physician will have several concerns about


instituting an image-guided breast biopsy pro-
gram. Will patients accept sampling rather than
excision? What will be the false-negative rate?
Will we maintain the proper indications, and
who should perform the image-guided breast
biopsy—radiologists or surgeons?
While the physicians deal with these concerns,
the patient is exposed to media headlines such as
“The Needle Replaces the Knife.” It does not take
a very sophisticated patient-consumer to under-
stand that a needle is less invasive than a knife.
In addition to the media deluge, patients are also
exposed to corporate-driven advertisement of
new breast biopsy devices. Acceptance of a new
technology, in the face of physician reluctance,
Figure 4–3. The StereoLoc™ is the upright add-on stereotactic
is now being influenced by outside sources. guidance system used with the Lorad™ mammography unit.
Image-Directed Breast Biopsy 69

tages, add-on stereotactic breast biopsy units outweighed by several advantages. The prone
traditionally have been less popular than dedi- position allows gravity to aid the technologist in
cated prone stereotactic tables. Because of the reaching more posterior lesions. A greatly
upright patient position and patient visualiza- enhanced work space underneath the table and
tion of the procedure, there is the potential for out of the patient’s view allows for the addition
an increase in syncopal episodes.5,28 In addition, of more advanced breast biopsy devices.
the upright units provide minimal work space There are two dedicated prone systems avail-
and limited access to the breast. Dedicated prone able, the Mammotest/Mammovision™ (Fischer
stereotactic tables are more costly and require Imaging, Denver, Colorado) and the Lorad
dedicated space for performing stereotactic Stereoguide™ (Lorad, a division of Trex Medical,
breast biopsy. These disadvantages appear to be Danbury, Connecticut) (Figures 4–4A and 4–4B).

Figure 4–4. A, Fischer Mammotest/Mammovision™. B, Lorad Stereoguide™.


70 BREAST CANCER

The Stereoguide™ table has, until recently, also biopsy devices (Mammotome™, MIBB™) (Fig-
been distributed as the ABBI™ table by the U.S. ure 4–5). Breast biopsy acquisition devices
Surgical Corporation. The Fischer Mammotest/ (ABBI™, Site-Select™) have larger cannula
Mammovision™ is a unidirectional patient- type tools and are designed for an image-
positioning table, with the aperture for the guided excision of tissue as a substitute for tra-
patient’s breast located at one end of the table. ditional surgical biopsy.
Access around the breast is approximately 180º Fine-needle aspiration using stereotactic
to 210º with rotation of the C-arm and mam- guidance was the first minimally invasive
mography tube head. Special software features biopsy technique used for nonpalpable lesions
increase the angle of access to the breast up to and is preferred by some to this day. In 1989,
240º with “target on scout” and close to 360º Lancet published a classic article from the
with the “lateral arm.” The Lorad Stereoguide™ Karolinski Institute, which evaluated the
is a bidirectional patient-positioning table with stereotactic fine needle biopsy of 2,594 mam-
the aperture for the breast in the center of the mographically detected, nonpalpable lesions
table and a foot extension at either end. With from 1983 to 1987.29 Of the 2,005 (77.3%)
the facility to rotate the patient 180º and the cases judged to be benign, only 1 turned out to
addition of the 180º rotation of the C-arm, there be malignant 14 months later. Of the 576 cases
is a true 360º access to the breast. (21.9%) selected for needle localization fol-
lowed by open breast biopsy on the basis of
Specimen Acquisition Devices cytologic and/or mammographic interpretation,
cancer was identified in 429 (75.7%). Dowlat-
The stereotactic biopsy equipment is amenable shahi and colleagues published 528 cases of
to the addition of a number of different devices stereotactic FNA, in corroboration with the
that are used in specimen acquisition. These University of Kiel from the Federal Republic of
include fine-needle aspiration (FNA), auto- Germany.17 In their report stereotactic guidance
mated Tru-cut needle core, and vacuum-assisted with 23-gauge FNA had a sensitivity of 95 per-

Figure 4–5. Prone stereotactic tables can perform fine-needle aspiration, needle core biopsy, vacuum-
assisted biopsy, and wire localization procedures.
Image-Directed Breast Biopsy 71

cent and accuracy of 92 percent. Furthermore, sistently larger tissue sample. The standard use
this article confirmed the accuracy of stereo- of the 14-gauge needle eliminated the issue of
tactic localization by imaging the tip of the nee- insufficient sampling. Several different gauge
dle within 2-mm of the center of the lesion in needles for automated Tru-cut™ biopsy have
96 percent of the cases. been evaluated. The lower rate of insufficient
Acceptance of FNA as a standard technique sampling and increased sensitivity, without
for the performance of stereotactic biopsy had increased complications, have led to the mini-
limited success, especially in the United States. mum 14-gauge size becoming the standard.31,32
Fine-needle aspiration has long been recog- Another principle identified to increase the
nized to have several potential pitfalls. This accuracy is the routine use of pre- and postfire
includes insufficient sampling as high as 38 stereotactic imaging. Prefire stereo images
percent, a low ranging sensitivity ranging assess the appropriate alignment of the needle
between 68 and 93 percent, and specificity to the lesion, and postfire stereo images docu-
varying between 88 and 100 percent.30 The ment the penetration of the needle through the
broad range of sensitivity and specificity is lesion. Tru-cut™-type core-needle biopsy with
dependent on a number of factors, including the stereotactic localization was demonstrated by
type of lesion to be sampled, the individual per- Parker and others to be a cost-effective proce-
forming the aspiration, and the individual inter- dure, which was less invasive and reduced
preting the cytologic specimen. Finally, cytol- patient anxiety. It has a lower false-negative
ogy rarely provides a specific benign diagnosis. rate when compared with FNA.5–8 Furthermore,
the need for cytologic expertise is avoided,
Automated Tru-cut™ Type Biopsy which is important in the community setting
where expert cytopathologist interpretation
In the late 1980s, Parker (Radiology Imaging may not be available.
Associates, Englewood, Colorado) and others Initial experience with stereotactic breast
began working with the automated Tru-cut biopsy was based on film screen technology
Biopsy™ instrument (Bard Urologic, Coving- until 1993, when the charged coupled device
ton, Georgia) designed for biopsy of the (CCD) camera replaced the film cassette
prostate. He combined this technology on the image receptor. This facilitated digital recre-
prone Fischer™ stereotactic table for perform- ation of the breast lesion on a computer moni-
ing large-core stereotactic breast biopsy. In tor. Several advantages for digital image acqui-
1991, Parker published a case series of 102 sition were immediately recognized. As the
patients, where every patient had a stereotactic- software improved, image acquisition time
guided, large-core needle biopsy, followed by decreased. This significantly reduced proce-
traditional surgical excision of the lesion.5 dure time, which, in turn, increased patient
There was agreement of histologic results in 98 comfort. A more comfortable patient was less
cases (96%). One cancer missed with a core likely to move and as a result the breast and
was determined to be a very difficult lesion to target lesion remained stationary for improved
localize because of its posterior position. This targeting accuracy. A critical difference
article set the stage for the standardization of between digital image acquisition and film
stereotactic biopsy techniques, which is still screen image acquisition involves postprocess-
being followed today. In addition to the use of ing of the image. Once a film is developed, the
dedicated prone stereotactic equipment, he also characteristics of the lesion on the film cannot
advocated the use of “long throw” biopsy nee- be changed. When a digital image is acquired,
dle devices. The longer excursion of the inner the lesion can be enhanced, magnified, and
and outer sheaths of the needle provided a con- even inverted to appear black on a white back-
72 BREAST CANCER

ground. These postprocessing features are The first digital image to be taken is the
especially helpful when evaluating small clus- zero-degree scout image. No matter what
ters of microcalcifications. An additional approach to the breast is taken, the scout image
bonus for patients included lower radiation is perpendicular to the plane of the skin. Once
with each procedure, related to a narrow field the appropriate lesion is identified, it is posi-
of view. Digital imaging for stereotactic biopsy tioned in the middle third of the scout image,
is not full field (ie, the entire breast is not from left to the right. A set of stereo images are
imaged). The area of the breast imaged is lim- obtained by rotating the mammography tube
ited to a square area of 5 ´ 5 cm. The true head to a +15° and –15° to yield an arc of sepa-
learning curve in performing stereotactic ration between the two stereo images of 30°
breast biopsy revolves around this narrow field (Figure 4–6). When targeting for an automated
of view. The lesion for which the biopsy is to Tru-cut instrument, a central target is chosen on
be performed requires prior identification on a the lesion in image number one, and a corre-
high-quality, film-screen diagnostic mammo- sponding target is chosen on the lesion in image
gram. The physician must recognize and trans- number two. Additional targets or offsets are
fer the appearance of the lesion to a digital chosen around the center of the lesion, for
image. The digital image is at least four times example, at 12, 3, 6, and 9 o’clock positions
magnified, is dependent on monitor resolution, (Figure 4–7). With the appropriate targets
and the lesion is not seen in association with entered into the system, the software determines
the full anatomic image of the breast. the horizontal, parallax shift of the lesion from
stereo image number one to stereo image num-
Performing Stereotactic Breast Biopsy ber two. The vertical coordinate is unchanged. A
trigonometric formula then calculates the hori-
After review of the mammogram, the general zontal, vertical, and depth coordinates of the
approach to the breast is chosen. The shortest lesion’s center within the breast.
skin-to-lesion distance and the ability to visual- With the three-dimensional coordinates of
ize the lesion are both factors in choosing the the lesion calculated, the puncture device or
approach.33 Also important is assuring enough stage, which houses the biopsy instrumenta-
tissue beyond the lesion to account for the tion, is motor driven to the calculated horizon-
excursion (“throw”) of the biopsy needle dur- tal and vertical positions. The biopsy device is
ing sampling. A lesion in the lower inner aspect advanced toward the skin, and the site for
of the breast may be approached from the medi- insertion is identified. A local anesthetic is
olateral position. A lesion in the most inferior injected to raise an appropriate skin wheal.
six o’clock position of the breast may be Additional local anesthetic may be judiciously
approached from the mediolateral or laterome- injected into the deeper aspects of the breast.
dial position on the Fischer Mammotest™ table. This is done in a radial manner to avoid plac-
The “lateral arm” may be attached to access the ing too much local anesthetic in any one area
lesion through the noncompressed portion of for fear of moving the targeted lesion, or plac-
the breast. The six o’clock lesion may also be ing too much local anesthetic in front of the
approached from the caudal cranial position (ie, lesion for fear of limiting its visualization. An
from below) on the Lorad Stereoguide™ table. 11-blade scalpel is used to make a vertical-ori-
The mammography technologist is responsible ented skin incision of 2 to 4 mm in length. The
for positioning the patient for the desired needle is advanced into the breast to a position
approach. Other responsibilities of the technol- several millimeters short of the center of the
ogist include calibration and maintenance of calculated depth of the lesion. On the basis of
the equipment and quality assurance. the mechanics of the biopsy instruments in use
Image-Directed Breast Biopsy 73

Figure 4–6. Viewing the stereotactic tables from above. The diagram illustrates the movement of the tube head for acquiring
stereo images and the resultant z-value determination.

(dead space at needle tip, sample notch size, tionship to the lesion to be biopsied. There
excursion or throw of instrument), a precalcu- must be symmetry of alignment between the
lated pull-back is determined by each biopsy two stereo images. If the alignment is satisfac-
instrument manufacturer. The depth chosen is tory, the automated Tru-cut instrument is fired
the calculated depth minus this predetermined and the first sample from the center of the
pull-back depth. By using the pull-back depth, lesion is obtained. Prior to removing the needle
the sampling notch will be positioned for from the breast, it is important to document
more adequate, fuller sampling of the lesion symmetrical penetration of the needle through
(Figure 4–8). the center of the lesion by taking stereo postfire
Prefire stereo digital images are acquired to images of the lesion (Figures 4–9A, 4–9B). The
assess the alignment of the needle tip in rela- needle is withdrawn from the breast, the speci-

Figure 4–7. Digital stereo images in reverse video with central targets and multiple offsets on the
Fischer™ system.
74 BREAST CANCER

men is placed on moistened gauze, and the ital images are acquired. The purpose of these
biopsy instrument is recocked and reinserted images is to document the removal of the
into the breast through the same skin incision to microcalcifications and to verify the presence
acquire the next target or offset sample. The of residual calcifications. A specimen radi-
same process is repeated until all samples are ograph of the tissue documents the inclusion of
obtained. The average number of needle core microcalcifications within the core sam-
biopsy samples for a nodular density is five or ples.36,37 Frequently, only a few calcifications
six (Figure 4–10). When biopsies of microcal- are evident on specimen radiography. Increas-
cifications are performed, a greater sampling is ingly, the literature has begun to support con-
required. Even in open biopsy surgical litera- cern over insufficient sampling of core biopsy
ture, pathologic assessment has identified atyp- for microcalcifications. Israel and Fine
ical hyperplasia and DCIS at a “distance” from reported a series of 500 consecutive core-nee-
the targeted microcalcifications.34 Lieberman dle biopsies with a sensitivity of 97.8 percent
from Memorial Sloan Kettering Hospital dis- and a false-negative rate of 1.5 percent. How-
cussed the issue of how many core biopsy spec- ever, upgrading of diagnosis on open surgical
imens are needed.35 In her report, biopsies of excision was evident in 33 percent of the cases,
145 lesions were performed; 92 were nodular where atypical ductal hyperplasia was identi-
densities and 53 were microcalcifications. Five fied on core-needle biopsy, and DCIS was iden-
cores yielded diagnosis in 99 percent of the tified on excision.38 The presentation for the
biopsies for nodular densities. Five cores upgrading of diagnosis was microcalcifica-
yielded a diagnosis in only 87 percent of the tions, where the average core sampling was
patients presenting with microcalcifications; between 9 and 12 samples. Not surprisingly,
six or more cores yielded a diagnosis in 92 per- atypical ductal hyperplasia diagnosed at stereo-
cent of the cases. When the targeted samples tactic core-needle biopsy has been called an
have been obtained, a set of postprocedure dig- indication for open surgical biopsy.39,40 Consis-

Figure 4–8. Positioning the needle at the lesion center will result in failure to sample the front half
of the lesion. With a 5-mm pull-back to the front of the lesion, the entire length of the lesion will be
sampled.
Image-Directed Breast Biopsy 75

A B
Figure 4–9. A, Prefire alignment; the relation of the tip of the needle to the lesion must be symmetrical. B, Postfire alignment;
the relation of the tip of the needle to the lesion must be symmetrical.

tent with the reporting of others, Liberman gauge.43 The Mammotome™ system is ideally
reported a series of 25 cases of atypical ductal suited for taking biopsy specimens of microcal-
hyperplasia identified on stereotactic core-nee- cifications under stereotactic guidance. Once
dle biopsy, with significant upgrading (52%) to the calcifications have been imaged stereotacti-
carcinoma on open surgical excision.41 cally, they are targeted on the computer monitor.
By changing the position of the sampling notch,
Vacuum-Assisted Biopsy Devices multiple tissue samples can be obtained using
only a single target probe insertion. The target
A solution to decrease the upgrading of diagno- may be centered in the cluster of microcalcifi-
sis is to increase tissue sampling. Tissue sam- cations or preferentially placed at the cluster’s
pling can be increased by increasing the number edge so that the overlying probe will not
of samples taken, increasing the size of the tis- obscure a small cluster of microcalcifications
sue sample taken, and/or the manner in which when digital images are obtained. When the
the tissue samples are obtained. A vacuum- three-dimensional coordinates are calculated by
assisted biopsy satisfies these requirements.25,42 the stereotactic system software, the driver and
Not only is the sample size larger, but the sam- probe are positioned at the corresponding hori-
ples may be taken in a circumferencial, contigu- zontal and vertical coordinates. The driver and
ous manner to allow reconstruction of the histo- probe are advanced toward the breast, where
logic architecture of the area of the biopsy. The local anesthetic is injected. The probe is inserted
Mammotome™ consists of a driver, which may through a vertical skin incision to the appropriate
be housed on either of the dedicated prone
stereotactic systems. Inside the driver, there is a
biopsy probe that consists of three parts. The
probe body has a sampling notch with several
holes connected to a vacuum. The second part
of the probe is a rotating cutter lumen that fits
inside the probe body and is rotated by a circu-
lar gear mechanism in the driver. The third and
last component of the probe is the knockout pin,
which fits inside the cutter lumen and is
attached to the rear vacuum to assist in retriev- Figure 4–10. 14-gauge needle core biopsy
ing tissue samples from the probe body. The sample with histologic confirmation of
fibroadenoma.
probe is manufactured in two sizes: 14- and 11-
76 BREAST CANCER

Figure 4–11. Local anesthetic injection, skin incision with an


11-blade scalpel followed by insertion of vacuum-assisted
probe into the skin, in preparation for biopsy.

depth (Figure 4–11). Prefire stereo images assess the lesion is confirmed with postfire (align-
the alignment of the probe with the microcalcifi- ment) stereo images (Figure 4–12). A vacuum
cations. The driver is designed with a spring- pulls breast tissue into the sampling notch, and
loaded mechanism to permit automated advance- the cutter is advanced across the sampling
ment of the probe through the breast tissue. The notch, cutting the tissue free from inside the
sampling notch may be positioned within the breast. The tissue sample is removed from the
breast by the automated forward movement of specimen retrieval chamber. The entire process
the probe, or it can be manually aligned with the is then repeated. The number of biopsy samples
lesion by taking the driver to the appropriate taken is based on the size of the lesion and the
depth with the probe already in its full excur- volume of tissue desired. On average, approxi-
sion. The alignment of the sampling notch with mately 12 to 16 tissue samples are obtained for
a small cluster of microcalcifications, fre-
quently resulting in removal of the entire mam-
mographic evidence of the lesion. The vacuum
assistance provides several advantages. It elimi-
nates the need for pinpoint accuracy required
with automated Tru-cut biopsy instruments and
facilitates removal of multiple tissue samples
without removal of the biopsy probe.42 In addi-
tion, the notch may be positioned for specific
directional sampling, on the basis of the align-
Figure 4–12. The vacuum assisted probe may be used for ment of the probe notch with the lesion. The
directional sampling. In this example, the majority of samples
will be taken between 3:00 and 9:00 through the 6:00 position. larger tissue samples provide a greater chance
Image-Directed Breast Biopsy 77

of lesion removal and a greater percentage of is essential that the physician be able to recog-
positive specimen radiographs (Figure 4–13). nize and correct for targeting errors (Figures
Burbank and Jackman have demonstrated that 4–14A to 4–14C). Certain patient characteris-
the improved accuracy with the directional vac- tics will interfere with the success of a stereo-
uum-assisted biopsy device decreases the tactic breast biopsy. Patients with neurologic or
upgrading of diagnosis seen with core-needle musculoskeletal conditions may not tolerate
biopsy.44,45 Burbank showed that the 14-gauge positioning on the stereotactic table. Patients
device provided no upgrading of atypical ductal that are coughing because of an acute or
hyperplasia to carcinoma at open biopsy. Jack- chronic respiratory condition will increase
man compared 14-gauge vacuum-assisted breast motion and lesion movement, which may
device to 14-gauge core-needle biopsy and interfere with accurate targeting. Patients with
illustrated a reduction of upgrading of atypical a high level of anxiety, especially those suffer-
ductal hyperplasia from 48 to 18 percent. Post- ing from claustrophobia or agoraphobia, may
procedure imaging yields a well-defined air- require sedation. As any biopsy has the poten-
contrast cavity in the majority of cases. The 11- tial for bleeding complications, those patients
gauge core probe also allows a marker clip to be with a history of bleeding abnormalities or who
placed in the wall of the biopsy cavity to assist are taking anticoagulants will require correc-
in the localization of the area in the future when tion prior to biopsy. The small or ptotic breast
all evidence of the lesion has been eliminated.46 creates one of the most common difficulties in
This marker clip and/or the residual cavity may stereotactic breast biopsy. A breast that flattens
be localized if the diagnosis requires further to a marginal thickness in compression may
surgical management. The complication rate for lead to “stroke margin problems.” The stroke
the device is less than 1 percent, which is com- margin is defined as the distance in millimeters
parable to core-needle biopsy.7,42 Ecchymosis of from the postfired biopsy needle/probe to the
the skin at the insertion site is common as well back of the breast or the rear image receptor.
as intraparenchymal hemorrhage localized to The stroke margin must be greater than zero on
the biopsy cavity, but clinically significant the Lorad Stereoguide™ system or greater than
hematomas that interfere or complicate subse- a positive 4 mm on the Fischer Mammotest™
quent surgery or follow-up are rare. table (Figure 4–15). When a breast is very thin,
or the lesion is more posteriorly positioned, a
DIFFICULTIES IN STEREOTACTIC negative stroke margin may be encountered.
BREAST BIOPSIES This situation will result in the biopsy needle or
probe striking the rear image receptor and
It is important for the physician to anticipate piercing the back of the patient’s breast skin.
that some patients and some lesions will be dif-
ficult for obtaining biopsy specimens. Certain
lesion characteristics, such as low-density nod-
ules, faint or nonclustered microcalcifications,
or vague asymmetric densities, may be difficult
to visualize with digital imaging despite post-
processing features. The position of certain
lesions, such as those that are very superficial,
those against the chest wall, or those in the axil-
lary tail of the breast, may require innovative
positioning by the experienced technologist. Figure 4–13. Postprocedure images after six samples with
the vacuum-assisted device illustrated the air-contrast cavity
However, some lesions may be inaccessible. It and the majority of calcifications removed.
78 BREAST CANCER

C
Figure 4–14. A, Correct pre- and postfire needle/probe alignment with the lesion
result in favorable tissue sampling. B, An “X” targeting error will result in the needle/
probe being off to one side of the lesion. A “Y” targeting error will result in the needle/
probe being above or below the lesion. C, A “Z” axis targeting error will illustrate the
needle/probe being too far in front or too far past the lesion. All three targeting errors
become less important with the vacuum-assisted device.
Image-Directed Breast Biopsy 79

Figure 4–15. With an adequate stroke margin, the back of the breast and image receptor are
protected. Ignoring a negative stroke margin will result in patient discomfort and damage to the
image receptor.

Several mechanisms for correcting stroke mar- ment, manual insertion of the Mammotome™
gins are available. The most commonly probe, use of the lateral arm, and implementa-
employed is pulling back the prefire position of tion of a double-paddle technique.
the needle/probe several millimeters until the
calculated stroke margin is adequate (Figure BREAST ULTRASONOGRAPHY
4–16). Other methods for dealing with stroke
margin difficulties include, but are not limited Breast ultrasonography is an effective diagnostic
to, taking a different approach to the breast tool when used in the proper clinical setting.
lesion, using a shorter “throw” biopsy instru- Appropriate indications for breast ultrasonog-

Figure 4–16. The most common method for correcting for an inadequate stroke
margin involves reducing the depth of the needle/probe by “x” mm.
80 BREAST CANCER

sent as a “thickening” or a mammographic asym-


metry. When a focal abnormality is identified, an
ultrasound-guided percutaneous biopsy will pro-
vide an efficient and cost-effective diagnosis.
Breast ultrasonography can easily be inte-
grated into the surgeon’s practice as a direct
extension of his clinical breast evaluation. The
patient in the supine position with the ipsilateral
arm raised, much like the clinical breast exami-
Figure 4–17. The patient is positioned supine, with the ipsi- nation, is ready for ultrasound examination
lateral arm raised for ultrasound scanning. The position may
(Figure 4–17). The 7.5- to 10-MHz linear array
be altered by rolling the patient and/or propping the patient
with a pillow to allow scanning through the thinner portions transducer is used for a targeted diagnostic
of the breast. ultrasound examination of a clinical abnormal-
ity. Whole breast ultrasound is usually done for
raphy have been somewhat controversial. A two reasons:
lower comfort level with hands-on breast ultra-
1. In the patient with a suspected breast cancer
sonography and a heavy reliance on ultrasonog-
to look for other areas of involvement;
raphers has, until recently, minimized the impor-
2. High-risk patient with a clinically difficult
tance of breast ultrasonography. The availability
breast and normal or non-specific mammo-
of computer-enhanced imaging and high-fre-
gram to look for an occult lesion.
quency transducers have allowed the indications
for breast ultrasonography to move well beyond The scanning techniques commonly used
distinguishing cystic from solid lesions.47,48 include the radial scan, transverse sweeping
Diagnostic breast ultrasonography may be scan, and tangential scan of the nipple. The
used to evaluate palpable abnormalities, espe- axilla may also be scanned, especially in associ-
cially in a radiographically dense breast. Mam- ation with a known malignancy. The normal
mographically indeterminate lesions will not sonographic anatomy of the female breast that
only be evaluated for cystic versus solid nature, can be routinely imaged includes skin, subcuta-
but the sonographic characteristics may assist neous fat, breast parenchyma, Cooper’s liga-
in distinguishing a lesion’s benign or malignant ment, retromammary fat, pectoral muscle and
nature. Diagnostic breast ultrasonography will fascia, ribs, and pleura.49 Diagnosis of ultra-
assist the clinician in evaluating the patient that sound abnormalities is dependent on familiarity
is pregnant or lactating. Postoperative follow- with the normal sonographic breast anatomy and
up for both benign and malignant disease may the recognition of patterns of different breast
be enhanced with ultrasonography, including types (Figure 4–18).
monitoring and management of seromas and
hematomas. The chest wall and the conserved Indications for Intervention
breast can be assessed for recurrent disease.
The axilla may be scanned for preoperative A symptomatic or enlarging cyst, whether pal-
staging. Associated with every diagnostic indi- pable or nonpalpable, is a common indication
cation is the ability to use ultrasonography to for intervention. Typically, a symptomatic, pal-
guide interventional procedures. pable cyst is aspirated by inserting a needle
The goals of diagnostic breast ultrasonog- under palpation guidance and withdrawing
raphy are to reduce the number of benign biop- fluid until the lesion is no longer palpable.
sies by recognizing simple cysts and areas of Ultrasound guidance for aspiration may be just
fibroglandular tissue, which may clinically pre- as appropriate for the palpable lesion as it is
Image-Directed Breast Biopsy 81

obtained in a cost-effective, efficient manner in


the office setting.
Ultrasound-guided aspiration and/or biopsy
will assist in the management of postoperative
complications, such as seromas or hematomas.
With the increasing use of breast conservation
therapy, the architectural distortion identified at
the lumpectomy site may be ideally suited for
further evaluation with image-guided breast
biopsy. Ultrasound-guided FNA of clinical

Figure 4–18. Normal breast anatomy.

necessary for the nonpalpable lesion.50,51 Ultra-


sound guidance may assist in getting the needle
into a thick-walled cyst, documenting any cyst
wall irregularities and confirming total cyst
collapse (Figures 4–19A, 4–19B).
Cysts not meeting the criteria for a simple
cyst require aspiration. A lesion with a mixed
internal echo pattern and posterior enhancement
suggests the presence of fluid versus a solid
lesion, and an aspiration may be attempted prior
A
to a core-needle biopsy.52 The aspiration of thick,
paste-like material, frequently found with mam-
mary duct ectasia, might require a local anes-
thetic and the use of a larger-gauge (18 g) needle.
The presence of a nonpalpable, solid lesion
is an indication for an ultrasound-guided core-
needle biopsy to obtain a histologic diagnosis.53
Once again, ultrasound guidance is advanta-
geous for guiding a core-needle biopsy for the
palpable solid lesion as well as the nonpalpable
lesion. Ultrasound visualization and documen-
tation of the needle within the biopsy lesion
enhances the accuracy. For the solid lesion with
smooth margins, a homogeneous internal echo
pattern and ellipsoid shape, such as a fibroade-
noma, core-needle biopsy will achieve a spe- B
cific benign diagnosis. For the more suspicious Figure 4–19. A, This anechoic, smooth-walled lesion with
lesion with jagged edges, nonhomogeneous and reverberation artifact is being prepared for aspiration, using
an attachable needle guide. The lesion is aligned with the
irregular shadowing considered to be suspi- puncture lines on the ultrasound monitor. B, Postcyst aspira-
cious for carcinoma, the diagnosis may be tion documenting complete resolution.
82 BREAST CANCER

adenopathy may assist in staging or evaluation thick slice of this lesion in its location within the
of recurrent disease. With image-guided aspira- breast. This ultrasound plane is parallel to the
tion and drainage, frequently a part of the sur- long axis of the ultrasound transducer. There-
gical management of intracavitary abscesses, it fore, the intervening instrument must be guided
only makes sense that a more conservative along the long axis of the transducer. When the
approach should be considered for breast tip of the needle is seen advancing toward the
abscess. Ultrasound guidance is advantageous lesion and penetrates that lesion, we know that
for the aspiration of pus or the insertion of a the needle is in the same 1.5-mm plane as the
catheter for drainage and also to monitor the slice of the lesion being visualized and therefore
resolving abscess. within the lesion (Figure 4–20).54–56
To aspirate an enlarging or symptomatic
Technique for Ultrasound Guidance cyst, one simply wipes the skin with an alcohol
preparation and inserts a small needle (25-, 22-,
Whether guiding a 25-gauge needle for the aspi- 20-gauge); feedback is immediate when fluid is
ration of a simple cyst, a needle for the insertion seen in the syringe. When using a larger-gauge
of a local anesthetic, a wire for localization, or a needle for aspiration of a complex cyst, a local
14-gauge needle for core-needle biopsy, the anesthetic may be injected with a small-gauge
principles for ultrasound-guided biopsy remain needle (30-, 27-, 25-gauge) and aspiration per-
the same. It must be remembered that an ultra- formed with an 18-gauge needle. Cytologic
sound image visualized on the ultrasound mon- evaluation of the aspirated fluid is reserved for
itor represents a “slice” of the breast of approx- bloody fluid or lack of cyst resolution.55
imately 1.5 mm in thickness. When scanning Ultrasound core-needle biopsy for histo-
brings the lesion requiring intervention into logic diagnosis requires more planning. When
focus, we once again are only seeing a 1.5-mm the lesion requiring a core-needle biopsy is
identified, the skin is marked at the edge of the
transducer for the proposed insertion site of the
needle. The optimal insertion site and approach
to the lesion is the shortest skin-to-lesion dis-
tance. For best cosmesis, care should be taken
to avoid placing the scar of the insertion site
near the inner portion of the breast. At this
point, the breast is prepared with an appropriate
antiseptic solution. The transducer is likewise
prepared, or a sterile sleeve may be placed over
the transducer. Sterile ultrasound gel is avail-
able in individual packets. The local anesthetic
is then injected under direct ultrasound visual-
ization. The skin is anesthetized as well as the
track leading to the lesion. In addition, the local
anesthetic is applied above, below, and to the
opposite side of the lesion. The use of ultra-
sound visualization limits obscuring the lesion
with the administration of too much local anes-
Figure 4–20. The ultrasound plane is approximately 1 to thesia in any one area. A small skin incision is
1.5 mm thick. The biopsy needle must remain within this thin made at the transducer edge using an 11-blade
ultrasound plane at the same time that the lesion is main-
tained within that same plane to confirm accurate biopsy. scalpel. The core biopsy needle is then inserted
Image-Directed Breast Biopsy 83

Figure 4–21. A, With the ultrasound transducer and breast


stabilized with the nondominant hand, the biopsy instrument
is advanced forward through a small skin incision maintain-
ing orientation of the needle in the long axis of the trans-
ducer. B, Documenting alignment and penetration under real-
A time ultrasound guidance.

into the breast, and the lesion is approached mented as it approaches the lesion. When the
with ultrasound guidance. The needle may be position of the needle is confirmed at the front
guided freehand or directed by an attachable of the lesion, the needle/gun combination is
needle guide that is available from certain ultra- fired.53,57,58 There are several needle/gun com-
sound manufactures.49,53,54 The attachable nee- binations available, some of which are dispos-
dle guides assist the neophyte in maintaining able and others that use disposable needles
the proper needle alignment within the narrow within a more permanent housing (gun). The
ultrasound plane to ensure accurate lesion sam- mechanism of tissue acquisition is similar with
pling. Visualization is maximized by keeping the automated movement of an inner sheath
the needle at a more shallow angle and there- that contains a sampling notch followed imme-
fore parallel to the sole of the transducer. The diately by an outer sheath that cuts the tissue
shallow angle of the needle will also minimize free. This action is accomplished under direct
the chance of penetrating the chest wall and ultrasound visualization (Figure 4–21). The
subsequently the pleura. The freehand method needle is then withdrawn, and the specimen is
of biopsy allows greater flexibility in the needle transferred onto a moistened telfa pad. Addi-
insertion site and angle of approach of the nee- tional tissue samples are taken in the same
dle to the breast lesion. With either method, the manner through the existing incision until the
needle is observed and the information docu- lesion is adequately sampled. Approximately
84 BREAST CANCER

three to five tissue samples are obtained, SUMMARY


depending on the quality of the samples and the
confirmation of the needle penetrating the When patients are referred with mammographic
lesion. When adequate sampling is achieved, abnormalities requiring further evaluation, the
manual compression is applied. The incision mammographic lesion is evaluated to determine
may be approximated with a Steri-strip™, and a if the work-up is complete. When the abnormal-
Tegaderm™ dressing is applied (Figure 4–22). ity is determined to require a biopsy, options are
The accuracy of ultrasound core-needle presented to the patient. The options presented
biopsy has been widely documented.53,54,56,59 should include traditional, open surgical biopsy
Staren reviewed his series of more than 1,000 and percutaneous, image-guided breast biopsy.
consecutive diagnostic ultrasound scans. Of The option of image-guided breast biopsy must
these, 210 patients underwent ultrasound- include a discussion of monitoring and follow-
guided core biopsy of nonpalpable, mammo- up. If this type of breast biopsy is acceptable,
graphically detected lesions using a 14-gauge then the physician must choose the most appro-
needle. Symptomatic cysts underwent ultra- priate method of imaging to guide the biopsy.
sound-guided FNA. Lesions characterized as Microcalcifications, which cannot be visu-
fibroadenoma, indeterminate, or suspicious alized with the current ultrasound technology,
underwent ultrasound-guided FNA and/or core- will require stereotactic guidance. Certain
needle biopsy. There were no false positives nodular densities, architectural distortions, and
and the false-negative rate was 3.6 percent. asymmetric densities without ultrasound find-
Small lesions were noted with ultrasound char- ings will also be amenable to stereotactic
acteristics that warranted biopsy on diagnostic biopsy.45,53,54 When both mammography and
grounds alone. Staren concluded that ultra- ultrasonography visualize a lesion, such as a
sound-guided aspiration and/or biopsy could solid nodular density, ultrasonography is the
accurately diagnose nonpalpable, mammo- preferable method for image guidance.52,53,55
graphically detected breast masses. The real-time nature of ultrasound imaging
provides increased accuracy and is more cost
effective. In addition, ultrasound-guided biop-
sies are more comfortable for the patient. The
patient may lie supine, and local anesthetic may
be injected under direct visualization. This con-
trasts with stereotactic breast biopsy, where the
patient lies prone on a stereotactic table with
her breast in compression for the entire proce-
dure and her neck hyperextended. Also, the
ability to guide the injection under direct visu-
alization resolves concerns associated with the
liberal use of local anesthesia and the potential
for obscuring or moving a lesion undergoing
stereotactic core-needle biopsy.
The vast majority of nonpalpable lesions
recommended for biopsy are evaluated with
percutaneous image-guided breast biopsy. If a
benign diagnosis is obtained, no further work-
Figure 4–22. The skin incision is re-approximated using a
Steri-strip™ after manual compression has been applied for
up is recommended, and the patient is placed in
hemostasis. A dressing may then be applied. a follow-up protocol. A specific benign diagno-
Image-Directed Breast Biopsy 85

sis (fibroadenoma) requires only a return to 3. Wilhelm NC, DeParedes ES, Pope RT. The chang-
routine screening. With microcalcifications or ing mammogram: a primary indication for nee-
nodular densities with a less specific benign dle localization biopsy. Arch Surg 1986;121:
1311.
diagnosis, short-term mammography in 4 to 6 4. Miller RS, Adelman RW, Espinosa MH, et al. The
months is recommended.31 early detection of nonpalpable breast carci-
The obvious indication to proceed with noma with needle localization. Experience
open surgical excision is an image-guided with 500 patients in a community hospital. Am
biopsy for malignancy and atypical hyperpla- Surg 1992;58:193–8.
sia. Medical judgment or lack of pathologic and 5. Parker SH, Lovin JD, Jobe WE, et al. Nonpalpable
breast lesions: stereotactic automated large-
radiologic diagnostic concordance would also
core biopsies. Radiology 1991;180:403–7.
be sufficient cause for further intervention. The 6. Elvecrog EL, Lechner MC, Nelson MT. Nonpal-
issue of pathologic concordance with a suspi- pable breast lesions: correlation of stereotaxic
cious mammographic lesion has fueled a large-core needle biopsy and surgical biopsy
debate over the indication to perform an image- results. Radiology 1993;188:453–5.
guided breast biopsy on a highly suspicious 7. Parker SH, Burbank F, Jackman RJ, et al. Percuta-
lesion. Histologic confirmation assists in neous large-core breast biopsy: a multi-institu-
tional study. Radiology 1994;193:359–64.
patient planning and allows wider excision for
8. Dershaw DD, Morris EA, Liberman L, et al. Non-
clear margins at the first surgical setting. diagnostic stereotaxic core breast biopsy:
Image-guided breast biopsy of a suspicious results of rebiopsy. Radiology 1996;198:323–5.
lesion may bypass open biopsy altogether for 9. Liberman LL, Fahs MC, Dershaw DD, et al.
those patients that require a mastectomy and Impact of stereotaxic core breast biopsy on
are not candidates for breast conservation. His- cost of diagnosis. Radiology 1995;195:633–7.
tologic confirmation of an obvious cancer with 10. Tinnemans JGM, Wobbes T, Hendricks JHCL, et
al. Localization and excision of nonpalpable
image-guided technology leaves a tumor in situ breast lesions. A surgical evaluation of three
to aid in the successful performance of the sen- methods. Arch Surg 1987;122:802–6.
tinel lymph node biopsy procedure.60 Concerns 11. Norton LW, Zeligman BE, Pearlman MD. Accu-
over potential added cost with image-guided racy and cost of needle localization breast
biopsy are exaggerated. biopsy. Arch Surg 1988;123:947–50.
As technology advances and the approach to 12. Kopans DB, Meyer JE, Lindfors KK, McCarthy
KA. Spring-hookwire breast lesion localizer:
breast cancer treatment evolves, additional
use with rigid compression mammographic
indications for image-guided biopsy of suspi- systems. Radiology 1985;157:537–8.
cious lesions will emerge. For many, image- 13. Bigelow R, Smith R, Goodman PA, Wilson GS.
guided percutaneous breast biopsy has perma- Needle localization of non-palpable breast
nently altered the management of nonpalpable lesions. Arch Surg 1985;120:565–9.
breast disease. Image-guided percutaneous 14. Landercasper J, Gunderson SB, Gunderson AL, et
breast biopsy will provide the stage for achiev- al. Needle localization and biopsy of nonpal-
pable lesions of the breast. Surg Gynecol
ing nonoperative histologic diagnosis and the
Obstet 1987;164:477–81.
potential for future therapeutic modalities. 15. Homer MJ, Smith TJ, Marchant DJ. Outpatient
needle localization and biopsy for nonpalpable
REFERENCES breast lesions. JAMA 1984;252:2452–4.
16. Feig SA. Localization of clinically occult breast
1. Kopans DS. The positive predictive value of mam- lesions. Radiol Clin North Am 1983;21:155–
mography. AJR Am J Roentgenol 1992;158: 71.
521–6. 17. Dowlatshahi K, Gent HJ, Schmidt R, et al. Non-
2. Sailors DM, Crabtree JD, Land RL, et al. Needle palpable breast tumors: diagnosis with stereo-
localization for non-palpable breast lesions. taxic localization and fine-needle aspiration.
Am Surg 1994;60:186–9. Radiology 1989;170:427–33.
86 BREAST CANCER

18. Schwartz GF, Goldberg BB, Riften MD, D’Orazio diagnosed by stereotactic core needle biopsy.
SE. Ultrasonography: an alternative to x-ray Ann Surg Oncol 1996;3(5):482–8.
guided needle localization of nonpalpable 35. Lieberman LL, Dershaw DD, Rosen PR, et al.
breast masses. Surgery 1988;104:870–3. Stereotactic 14-gauge breast biopsy: how many
19. Kopans DB. Breast Imaging. Philadelphia: core biopsy specimens are needed? Radiology
Lippincott Williams & Wilkins; 1998. 1994;192:793–5.
20. Wilhelm MC, Wanebo HJ. Technique and guide- 36. Lieberman LL, Evans WP, Dershaw DD, et al.
lines for needle localization biopsy of nonpal- Radiography of microcalcifications in stereo-
pable lesions of the breast. Surg Gynecol taxic mammary core biopsy specimens. Radi-
Obstet 1988;176:439–41. ology 1994;190:223–5.
21. Swann CA, Kopans DB, McCarthy KA, et al. 37. Meyer JE, Lester SC, Grenna TH, White FV.
Practical solutions to problems of triangulation Occult breast calcifications sampled with large-
and preoperative localization of breast lesions. core biopsy: confirmation with radiography of
Radiology 1987;163:577–9. the specimen. Radiology 1993;188:581–2.
22. Leeming R, Madden M, Levy L. An improved 38. Israel PZ, Fine RE. Stereotactic needle core
technique for needle localization biopsies of the biopsy for occult breast lesions: a minimally
breast. Surg Gynecol Obstet 1993;177:85–7. invasive alternative. Am Surg 1995;61:87–91.
23. Fisher B. Reappraisal of breast biopsy prompted 39. Jackman RJ, Nowels KWW, Shepard MJ, et al.
by the use of lumpectomy. JAMA 1985;253: Stereotaxic large-core needle biopsy of 450
3585–8. non-palpable breast lesions with surgical cor-
24. Fine RE, Boyd BA. Stereotactic breast biopsy: a relation in lesions with cancer or atypical
practical approach. Am Surg 1996;62:96–102. hyperplasia. Radiology 1994;193:91–5.
25. Lovin JD, Parker SH, Leuthke JM, Hopper KD. 40. Liberman L, Dershaw DD, Rosen PP, et al. Stereo-
taxic core biopsy of breast carcinoma: accu-
Stereotactic percutaneous breast core biopsy,
racy of predicting invasion. Radiology 1995;
technical adaptation, and initial experience.
194:379–81.
Breast Dis 1990;176:741–7.
41. Liberman L, Cohen MA, Dershaw DD, et al.
26. Parker SH, Lovin JD, Jobe WE, et al. Stereotactic
Atypical ductal hyperplasia diagnosed at
breast biopsy with a biopsy gun. Radiology
stereotaxic core biopsy of breast lesions: an
1990;176:741–7.
indication for surgical biopsy. AJR Am J
27. Caines JS, McPhee MD, Konak GP, Wright BA.
Roentgenol 1995;164:1111–3.
Stereotactic needle core biopsy of breast
42. Burbank F, Parker SH, Fogerty TJ. Stereotactic
lesions using a regular mammographic table breast biopsy: improved tissue harvesting with
with an adapable stereotaxic device. AJR Am J the mammotome. Am Surg 1996;62:738–44.
Roentgenol 1994;163:317–21. 43. Berg WA, Kerbs TL, Campassi C, et al. Evalua-
28. Parker SH, Burbank F. State of the art: a practical tion of 14 and 11-gauge directional vacuum-
approach to minimally invasive breast biopsy. assisted biopsy probes and 14-gauge biopsy
Radiology 1996;200:11–20. guns in a breast parenchymal model. Radiol-
29. Azavedo E, Svane G, Auer G. Stereotactic fine- ogy 1997;205:203–8.
needle biopsy in 2594 mammographically 44. Burbank F. Stereotactic breast biopsy of atypical
detected non-palpable breast lesions. Lancet ductal hyperplasia and ductal carcinoma in in
1989;171:373–6. situ lesions: improved accuracy with direc-
30. Schmidt RA. Stereotactic breast biopsy. Cancer J tional vacuum-assisted biopsy. Radiology
Clin 1994;44:172–91. 1997;202:843–7.
31. Parker SH. When is a core really a core? Radiol- 45. Jackman RJ, Burbank SH, Parker SH, et al. Atyp-
ogy 1992;185:641–2. ical ductal hyperplasia diagnosed at stereotac-
32. Dowlatshahi K, Yaremko ML, Kluskens LF, Jokich tic breast biopsy: improved reliability with 14-
PM. Nonpalpable breast lesions: findings of gauge, directional vacuum-assisted biopsy.
stereotaxtic needle-core biopsy and fine-needle Radiology 1997;204:485–8.
aspiration cytology. Radiology 1991;181:745–50. 46. Burbank F, Forcier N. Tissue marking clip for
33. Soo MS. Imaging-guided core biopsies in the stereotactic breast biopsy: initial placement
breast. South Med J 1998;91:994–1000. accuracy, long-term stability and usefulness as
34. Tocino I, Gaargia B, Carter D. Surgical biopsy a guide for wire localization. Radiology 1997;
findings in patients with atypical hyperplasia 205:407–15.
Image-Directed Breast Biopsy 87

47. Dempsy PJ. Breast sonography: historical per- ultrasound. Baltimore: Williams & Wilkins;
spectives, clinical application and image inter- 1990. p. 71.
pretation. Ultrasound Q 1988;6:69. 55. Staren ED. Surgical office-based ultrasound of the
48. McSweeney MB, Murphy CH. Whole breast sono- breast. Am Surg 1995;61:619–26.
graphy. Radiol Clin North Am 1985;23:157–67. 56. Fornage BD, Coan JD, David CZ. Ultrasound-
49. Leucht W. Teaching atlas of breast ultrasound. guided needle biopsy of the breast and other
New York: Thieme; 1992. interventional procedures. Radiol Clin North
50. Kopans DB, Meyer JE, Lindfors KK, et al. Breast Am 1992;30:167–85.
sonography to guide cyst aspiration and wire 57. Staren ED. Ultrasound-guided biopsy of non
localization of occult solid lesions. AJR Am J palpable breast masses by surgeons. Ann Surg
Roentgenol 1984;143:489–92. Oncol 1996;3:476–82.
51. Meyer JE, Christian RL, Frenna TH, et al. Image- 58. Parker SH. Needle selection. In: Parker SH, Jobe
guided aspiration of solitary occult breast WE, editors. Percutaneous breast biopsy. New
“cysts.” Arch Surg 1992;127:433–35. York: Raven; 1993. p. 7.
52. Jackson VP. The role of ultrasound in breast imag- 59. Parker SH, Jobe WE, Dennis MA, et al. Ultra-
ing. Radiology 1990;177:305–11. sound-guided automated large-core breast
53. Parker SH, Stavros AS. Interventional breast biopsy. Radiology 1993;187:507–11.
ultrasound. In: Parker SH, Jobe WE, editors. 60. Pijpers R, Meijer S, Hoekstra OS, et al. Impact
Percutaneous breast biopsy. New York: Raven; of lymphoscintigraphy on sentinel node iden-
1993. p. 129. tification with technitium-99m colloidal albu-
54. Fornage BD. Interventional ultrasound of the min in breast cancer. J Nucl Med 1997;38:
breast. In: McGahan JP, editor. Interventional 366–8.
5
Histopathology of Malignant
Breast Disease
ROBERT A. GOLDSCHMIDT, MD

The basic classification of malignant breast dis- years from the bulky mass with a high risk of
eases has remained relatively unchanged since invasion to minute foci of questionable clinical
the most recent WHO revision in 1982.1 These significance, numerous studies have been under-
conditions can be broadly divided into epithelial taken to identify prognostic factors and optimize
and nonepithelial lesions, with separation of the therapy for the individual patient.
former into in situ and invasive tumors (Table The most important change in our concept
5–1). Although recent studies have shed new of DCIS was from the monolithic view of a sin-
light onto our understanding of the basic biol- gle disease highly likely to invade if left
ogy and natural history of breast cancer, this tra- untreated to the realization that DCIS repre-
ditional classification still retains its relevance sents a nonobligate precursor with variable risk
for clinicians involved in the diagnosis and of progression, depending on a combination of
treatment of malignant breast disease. factors. These factors include histologic pattern
(and by extension histologic grade), lesion size,
DUCTAL CARCINOMA IN SITU margin status, and ancillary studies such as pro-
liferation markers and c-erbB-b2.
Since, by definition, ductal carcinoma in situ
(DCIS) is an atypical proliferation of cells con-
Classification and Grading
fined by an intact basement membrane to the
of Ductal Carcinoma In Situ
ductolobular system of the breast, it cannot
cause serious morbidity unless it becomes inva- Although the traditional classification of DCIS
sive. Thus, the major goal of any pathologic based on architectural pattern is now recog-
evaluation of a patient with DCIS should be to
determine the level of risk of subsequent inva-
Table 5–1. MALIGNANT DISEASES OF THE BREAST:
sion so that optimal treatment can be offered EPITHELIAL TUMORS
and possible over- or undertreatment avoided.
In Situ Lesions Invasive Lesions
In the premammographic era, pure DCIS was
most often seen as a mass lesion of high-grade, Lobular carcinoma in situ (LCIS) Invasive lobular
comedo type and usually treated, appropriately, Ductal carcinoma in situ (DCIS) Ductal
comedo no special type (NST)
by mastectomy. In the current clinical setting, micropapillary tubular
however, the vast majority of DCIS present as a cribriform mucinous
solid medullary
mammographic abnormality or may be entirely papillary invasive
incidental to the lesion seen on radiography. As cribriform
the pattern of the disease has shifted over the papillary

89
90 BREAST CANCER

nized to be limited in terms of prognostic value, criteria for microinvasion are not well estab-
it remains in use by many pathologists and mer- lished, nor is its clinical significance, but at
its review for that reason. In addition, before present it is probably best to limit such a diag-
prognosis can be assessed, the pathologist must nosis to those cases where single tumor cells
establish a diagnosis by applying criteria to sep- are clearly evident outside an affected duct.
arate DCIS from other atypical or nonatypical Micropapillary DCIS, sometimes referred
proliferations. These criteria are almost purely to as “clinging carcinoma,” may vary in appear-
architectural and include the solid, cribriform, ance from a relatively flat proliferation with
micropapillary, papillary, and intraductal come- short projections to a pattern of long, slender
docarcinoma variants. epithelial fronds lacking fibrovascular cores
Comedo DCIS is the only type likely to pre- (Figure 5–2). Peripheral spaces formed by so-
sent as a palpable mass and accounts for the called “Roman bridges” are common and lead
majority of cases of DCIS diagnosed before the some to group micropapillary and cribriform
advent of mammography. As will be seen, it is lesions together. Key features of micropapillary
also the most likely to be high grade and as carcinoma distinguishing it from proliferations
such, the most likely to be associated with con- that appear similar but are benign are
current or subsequent invasion. The histologic monomorphism and lack of polarity. Myoep-
features of comedo DCIS are solid growth with ithelial cells should be absent from their usual
central necrosis, often with calcification (Fig- peripheral location, and the atypical cells
ure 5–1). Marked nuclear atypia is often seen, should be homogeneous in appearance.
but a recent consensus conference on the clas- Micropapillary carcinoma is usually composed
sification of DCIS2 allows use of the comedo of cells with low-grade nuclei, although cases
designation in the absence of high nuclear with high nuclear grade are not rare. Centrally
grade. Marked fibrosis and elastosis of the sur- located necrotic debris and microcalcification
rounding stroma are frequently present, as is an may be present, especially in cases with high-
associated periductal lymphocytic infiltrate. grade nuclei. There is some evidence that
Not infrequently, a question of possible micropapillary DCIS may be more likely to
microinvasion arises as small nests of tumor involve multiple quadrants than other forms of
cells are trapped in the fibroinflammatory reac- DCIS.3 Further studies will be necessary to
tion encircling the affected ducts. Diagnostic establish the clinical significance of this finding.

Figure 5–2. Micropapillary DCIS. Left, dilated ducts with pap-


illary tufting of the epithelium (original magnification x100).
Figure 5–1. DCIS, comedo type, with central necrosis. This Right, the micropapillary structures show stratification and loss
would qualify as a high-grade lesion based on nuclear mor- of polarity (original magnification x200). This is a low-grade
phology (original magnification x400). lesion based on nuclear grade and lack of necrosis.
Histopathology of Malignant Breast Disease 91

Cribriform DCIS describes a lesion charac-


terized by the formation of secondary microlu-
mens. These lumens tend to be round and uni-
formly distributed, although some variability is
acceptable. The classic term used to describe
the monomorphous nature of the cells outlining
the spaces is “rigid bridges,” referring to the
lack of stretched or elongated cells separating
glandular spaces (Figure 5–3). Nuclear mor-
phology is typically low grade, although high-
grade variants exist. Likewise, necrosis is com-
monly encountered in cribriform DCIS and Figure 5–3. Cribriform DCIS with central necrosis (x400).
This lesion is intermediate grade based on necrosis and
may be so prominent as to mimic comedo low-grade nuclear features.
DCIS. Fortunately, as will be seen in the dis-
cussion on grading, this distinction is obviated on the presence or absence of comedo-type
in current classification systems. necrosis. Using these parameters, tumors can
Solid DCIS, as the name implies, consists of be divided into three groups. Group 1 (low-
a solid proliferation of neoplastic cells filling a grade) includes those tumors with either low-
ductal structure (Figure 5–4). Nuclear grade or intermediate-grade nuclei and no necrosis.
may vary widely, and spotty necrosis may be Group 2 (intermediate-grade) describes those
encountered. As in the other forms of DCIS, the tumors with low- or intermediate-grade nuclei
monomorphous nature of the cell population is and comedo necrosis, whereas Group 3 (high-
the hallmark of the process. grade) encompasses all tumors with high-grade
Papillary DCIS is the least common of the nuclei regardless of necrosis.
well-described variants and exhibits prominent Tumor grade has emerged as a significant
papillary features with fibrovascular cores but prognostic factor for risk of recurrence, although
no myoepithelial cell layer (Figure 5–5). As in other pathologic features may also be important.
other variants, a monomorphous cytologic The Van Nuys grading scheme is part of a prog-
appearance is essential to the diagnosis. nostic index for DCIS that includes tumor grade,
As previously alluded to, DCIS grade has a tumor size, and margin width. Evaluation of the
largely supplanted pattern as the most impor- latter two factors in DCIS can be problematic,
tant guide to clinical behavior and treatment.
There have been a variety of different systems
proposed, but all include some assessment of
nuclear features combined with other factors,
typically necrosis or cell polarity, with separa-
tion of cases into either two or three grades.
Two recent studies4,5 have compared a number
of different grading systems for their interob-
server reproducibility. Although none of the
systems demonstrated a high degree of agree-
ment among reviewing pathologists, both stud-
ies found the Van Nuys system of Silverstein
and Lagios6 to be the most reproducible. This
scheme relies on the distinction of three nuclear Figure 5–4. Solid DCIS. There is a monotonous proliferation
of cells filling the duct (x200). Lack of necrosis and non high-
grades based on size, texture and nucleoli, and grade nuclear features qualify this as a low-grade lesion.
92 BREAST CANCER

method of margin examination will rely on the


experience and preference of the pathologist
and surgeon, at least until better clinical studies
are available. Since most “recurrences” of
DCIS probably represent persistence following
incomplete removal, the issue of margins is not
of trivial importance. Routine specimen mam-
mography is often helpful in guiding the patho-
logic sampling by identifying areas of suspi-
cious calcification near resection margins.
A recent study published by Silverstein and
Lagios7 uses a retrospective analysis of DCIS
Figure 5–5. Papillary DCIS. Left, an intraductal proliferation
of papillary structures with fibrovascular cores (original mag- patients to demonstrate that margin width is the
nification x100). Right, high power shows nuclear atypia and only significant risk factor for local recurrence8.
stratification (original magnification x200).
In their series, there was no difference in recur-
rence rate between patients who received post-
however. Since DCIS only rarely forms a grossly operative radiation and those treated by lumpec-
visible mass, measurement of lesion size is typi- tomy only, so long as there was an uninvolved
cally done from microscopic slides. If, as is often margin width of 10mm or more in all directions.
the case, tumor is present on more than one In both the radiated and non-radiated groups,
slide, the pathologist must be able to reconstruct the risk of recurrence after 8-year follow-up was
the specimen to estimate size. This requires that 3–4%. Tumor size, nuclear grade, and presence
the sections be submitted in an orderly fashion to of comedonecrosis did not alter relative risk in
permit reconstruction. Even so, it is sometimes these cases. For margin widths of 1-10mm,the
difficult to know how to report lesion size when recurrence rates were higher (12–20%), but
small foci of DCIS are scattered throughout a there was still no statistically significant benefit
lumpectomy specimen, and such data are not to radiation therapy in this group. Only when
readily available in existing clinical studies. final margin width was < 1mm did postopera-
If tumor size assessment is occasionally a tive radiation show a significant reduction in
problem, margin width determination is even recurrence rates.
more of one. The most common approach Given the above considerations, the pathol-
involves the application of colored ink(s) to the ogy report in cases of DCIS must include a
surface of a specimen that has been oriented by large amount of data. Many institutions have
the surgeon. The specimen is then submitted for found that the use of a template form ensures
microscopic examination in serial transverse all vital data is present. Such a form would typ-
section and the shortest distance between tumor ically include features such as nuclear grade,
and ink reported as the margin width. Since this pattern, presence of necrosis, distance to mar-
method can only examine a tiny fraction of the gin, lesion size, presence of calcifications, and
actual surface area of the specimen, it is a crude any other parameters deemed to be important.
measurement at best. Some workers recom- This type of systematic reporting scheme has
mend an alternate method in which sections are the added advantage of making any retrospec-
shaved tangentially from the surface of the tive clinical studies much easier to perform.
specimen to permit wider sampling of the mar-
gins. Yet another technique advocated by some LOBULAR CARCINOMA IN SITU
surgeons is the separate removal of shaved mar-
gins from the biopsy cavity after the specimen Lobular carcinoma in situ (LCIS) is, by defini-
has been resected. Ultimately, selection of a tion, a microscopic process and as such is
Histopathology of Malignant Breast Disease 93

almost always an incidental finding seen in LCIS and ductal carcinoma in situ (DCIS)
association with some other gross or mammo- involving lobular units, since the therapeutic
graphic abnormality. In its most classic form, implications of these lesions may be quite differ-
described by Foote and Stewart in 1941,8 it is ent. In most cases of in situ duct carcinoma
readily identifiable by the general pathologist. involving the terminal duct/lobular unit in a sec-
The lobular acini are filled and distended by a ondary fashion, there is at least some retention
poorly cohesive proliferation of cells with of features suggestive of ductal origin. The pres-
round, rather bland nuclei, scant cytoplasm, and ence of large pleomorphic nuclei and/or sec-
inconspicuous nucleoli (Figure 5–6). Problems ondary lumens in such a proliferation would
in interpretation of these lesions can arise, how- favor DCIS over LCIS. Likewise, as previously
ever, from variations in qualitative and quanti- mentioned, identification of intracytoplasmic
tative aspects of the neoplastic process. For lumens with mucin droplets strongly suggests
example, the cytologic features of the neoplas- LCIS. Occasionally, a case will defy classifica-
tic cells may demonstrate more variability, with tion, even after exhaustive examination of the
pleomorphic nuclei and abundant cytoplasm. In specimen. If there are foci of coexistent invasive
such cases, it is often difficult to determine carcinoma or unequivocal DCIS, the distinction
whether one is dealing with lobular carcinoma becomes one of academic interest only, as treat-
in situ or so-called “lobular cancerization” by an ment would be dictated by those lesions. With a
in situ duct carcinoma. Since there is no reli- biopsy containing only an in situ lesion of inde-
able marker to distinguish the origin of such a terminate etiology, however, communication
lesion as either lobular or ductal, the distinction between the pathologist, surgeon, oncologist,
must ultimately be made by the pathologist’s and patient is essential for optimal care.
assessment of the light microscopic features.
The situation may be further complicated by INVASIVE BREAST CARCINOMA
the frequent association of typical ductal carci-
noma in situ and LCIS in the same biopsy. The Invasive (infiltrating) breast carcinoma can be
identification, when present, of intracytoplas- broadly subdivided into ductal and lobular cat-
mic lumens and associated mucin globules in egories, with a number of recognized variants
the atypical cells is helpful. The finding of such of each. Although current evidence suggests
cells in a proliferative lesion involving the ter- that the majority of invasive cancers arise from
minal duct/lobular unit is compelling evidence cells of the terminal duct lobular unit, their
for lobular carcinoma in situ. wide variation in appearance and clinical pre-
Another common feature, seen in up to 75
percent of patients with LCIS, is a pattern of so-
called pagetoid spread of atypical cells along
small ductules and occasional larger ducts. Typ-
ically, this manifests as a proliferation of atypical
cells lining the duct, with an overlying layer of
intact ductal epithelium. In the atrophic breast of
the postmenopausal patient, the latter pattern
may be the only evidence of LCIS. Some experts
disagree as to whether this appearance repre-
sents true ductal involvement rather than an
“unfolding” of the lobule yielding a pseudoduc-
tular appearance. Recognition of the process,
however, is more important than the terminol- Figure 5–6. Lobular carcinoma in situ. The acini are filled
and distended by a monotonous proliferation of small cells
ogy. The important distinction is between true with bland nuclear features (original magnification x100).
94 BREAST CANCER

sentation continues to make subtyping a useful often swirling around native ductal structures in
exercise until some better method of predicting a so-called “targetoid” fashion (Figure 5–7).
behavior becomes available. There are a number of variants of infiltrat-
ing lobular carcinoma that have been described,
Infiltrating Lobular Carcinoma and many tumors show mixtures of two or more
types. Most of these variants consist of cells
Infiltrating lobular carcinoma (ILC) is gener- with the same cytologic features as the classical
ally considered to account for up to 16 percent9 type but different patterns of growth such as
of invasive breast cancers, depending on the alveolar, solid, or tubulolobular. While it is
study population and the rigidity of diagnostic unclear whether identification of these variants
criteria. It may present as a scirrhous mass has clinical significance, the pleomorphic type
grossly and mammographically indistinguish- does merit separate distinction. This variant
able from infiltrating ductal carcinoma, consists of cells which infiltrate in the same
although it is often more insidious, with only manner as classical ILC but have high-grade
vague gross findings and occasionally negative nuclei. Several studies have suggested a more
mammographic appearance. In contrast to inva- aggressive behavior for these tumors.12,13
sive ductal carcinoma, invasive lobular cancer
presents as a less distinct tumor that is less Infiltrating Ductal Carcinoma
apparent on physical examination and mam-
mography. As a result, the microscopic extent Invasive breast cancers that do not exhibit the
of disease is often much greater than grossly features described above for lobular variants
appreciated, and clear lumpectomy margins are are considered to be ductal in origin. While this
somewhat more difficult to achieve.10 The clas- distinction is somewhat arbitrary, it is firmly
sical microscopic description, generally cred- embedded in the literature of breast cancer and
ited to Foote and Stewart,11 is of a diffusely serves as a useful tool for the recognition and
infiltrative tumor composed of cells with small, subclassification of malignant breast disease.
round nuclei with minimal pleomorphism or This large group of tumors accounts for the
mitotic activity. Intracytoplasmic lumina yield- majority (85 to 95%) of invasive breast cancer
ing a signet-ring appearance are often present cases and can be broadly divided into those of
but are not pathognomonic since they may be “no special type” or “not otherwise specified”
seen in ductal carcinomas also. Linear files (NST, NOS) and “special type” tumors of dis-
(“Indian files”) of infiltrating tumor cells are tinctive appearance and behavior.
the most characteristic pattern of invasion, Various studies place the percentage of NST
breast carcinomas at 50 to 75 percent of all
invasive breast cancers.13 The tumors within
this large group vary widely in appearance and
often contain minor components of special type
histology. An appreciation of the cytologic fea-
tures of the tumor cells and of the architectural
pattern of the invasive process is useful when
studying breast cancer. As will be discussed
later, these factors also form the basis for grad-
ing NST tumors. In regard to cytology, the
nuclei of NST neoplasms can vary from small
and rather bland in appearance to those exhibit-
ing marked enlargement and pleomorphism.
Figure 5–7. Infiltrating lobular carcinoma. The tumor cells
infiltrate in typical linear files (original magnification x200). Mitotic activity can likewise range from mini-
Histopathology of Malignant Breast Disease 95

mal to brisk and generally follows nuclear


grade. Architectural patterns are typically
described on the basis of degree of gland for-
mation, which may be quite prominent or com-
pletely absent. In addition, both cytologic and
architectural features may vary widely within a
single tumor. Other features, such as extensive
necrosis or widespread DCIS, may also have
prognostic importance and should be noted.
Ductal carcinomas of special type include a
group of tumors distinguished from NST
tumors by their unique histologic appearance
Figure 5–8. Tubular carcinoma. Left, the tumor consists of a
and often, less aggressive behavior. Within this haphazard arrangement of small tubular structures (original
group are the tubular, mucinous, medullary, magnification x100). Right, the tumor glands are lined by
cells with low-grade nuclei (original magnification x400).
invasive cribriform, papillary, and metaplastic
variants. Tubular carcinoma has become the
most commonly diagnosed special type tumor breast cancer and has a distinctive gross and
since the advent of mammography, due to its microscopic appearance. Such tumors tend to be
small size and lack of clinical symptoms. The soft, well-defined, rounded masses with a glis-
majority of tubular carcinomas are < 1.0 cm in tening mucoid surface. The tumor cells show
diameter, accounting for 7 to 21 percent of minimal pleomorphism and form tight clusters
mammographically detected lesions in various which float in pools of extracellular mucin (Fig-
studies.14,15 Precisely defining tubular carci- ure 5–9). As with tubular carcinoma, 90 percent
noma is elusive, particularly regarding the mucinous morphology is the generally accepted
extent of tubule formation required to make the minimum for designation as mucinous carci-
diagnosis. The basic requirement is the pres- noma, with its associated favorable prognosis.
ence of tubular structures lined by a single layer Medullary carcinoma is the most controver-
of epithelial cells of low-nuclear grade. The sial of the special-type tumors, both in terms of
tubule lumens are rounded and/or angulated, histologic criteria for diagnosis and subsequent
and mitosis is rare. The tumor stroma is quite behavior. Poor intraobserver reproducibility, and
characteristic, consisting of a cellular desmo- disagreement over the diagnostic requirements
plastic reaction, often with a central fibrous
scar from which the tubules radiate (Figure
5–8). Low-grade DCIS is a frequent accompa-
niment of tubular carcinoma. As previously
alluded to, tumors that are not purely tubular
are somewhat controversial. Most authors will
accept 90 percent tubular architecture as a min-
imum criterion for the diagnosis of tubular car-
cinoma, and many use 75 percent as a cut-off.
Lesser degrees of tubular differentiation are gen-
erally reported as “tubular features” in an NST
tumor. The distinction is not trivial, as several
studies have shown prognostic differences
related to varying degrees of tubule formation.16 Figure 5–9. Mucinous carcinoma. Left, clusters of tumor
Mucinous (colloid) carcinoma of the breast cells float in a pool of extracellular mucin (original magnifica-
tion x100). Right, in this case, the cells exhibit minimal
accounts for one to three percent of invasive nuclear atypia (original magnification x200).
96 BREAST CANCER

for medullary carcinoma, likely account for the cribriform designation comes from its resem-
reported frequency range of 2 to 10 percent.17 In blance to cribriform DCIS, from which it may
our own practice, medullary carcinoma is a dis- occasionally be difficult to distinguish. The his-
tinctly unusual variant. Disclaimers aside, the tologic appearance is of infiltrating sheets and
classic description of medullary is of a soft, nests of cells of low-nuclear grade with the typ-
fleshy, circumscribed tumor with a homoge- ical punched-out spaces seen in its in situ
neous appearance. Microscopically, the tumor namesake (Figure 5–11). The 90 percent rule
cells have large, often bizarre nuclei, abundant for designation as a special-type tumor gener-
mitoses, and grow in syncytial sheets. Equally ally applies for combinations of invasive cribri-
important for the diagnosis is the presence of a form carcinoma and NST tumors, while tumors
significant lymphoplasmacytic infiltrate that composed entirely of tubular and invasive crib-
usually occupies narrow bands of fibrovascular riform structures in any proportion still qualify
stroma within the tumor and may also surround as special type.
the periphery of the tumor. Lastly, microscopic
circumscription is essential. The tumor must Grading of Invasive Breast Carcinoma
have a smooth, pushing margin without infiltra-
tion of surrounding breast tissue or fat by tumor Grading schemes for invasive breast carcinoma
cells (Figure 5–10). Foci of DCIS surrounding originated with the work of Greenhough19 60
the tumor, however, are not uncommon and years ago and have not changed substantially
should not preclude a diagnosis of medullary since that time. It is a testament to the power of
carcinoma. It is important to adhere to strict histological grading of breast cancer that virtu-
diagnostic criteria because the less aggressive ally all of the numerous methods and variations
behavior ascribed to medullary carcinoma proposed over the years correlate to some
belies its high-grade appearance. Attempts to degree with clinical behavior. While no single
distinguish an intermediate variant18 (atypical system has achieved universal acceptance, all
medullary carcinoma) exhibiting some, but not use some combination of nuclear features,
all, of the classic features and having an inter- mitotic activity, and gland formation. One of
mediate prognosis remain controversial. the more commonly used methods, and that
Invasive cribriform carcinoma is a relatively adopted by the WHO, is the Scarff-Bloom-
infrequent variant in pure form, although com- Richardson system,20 which assigns a score of
bination with tubular carcinoma is not rare. The one to three points for each of the three above-

Figure 5–10. Medullary carcinoma. Left, the tumor is sharply


demarcated from the surrounding fat (original magnification
x100). Right, the tumor cells are highly pleomorphic with Figure 5–11. Invasive cribriform carcinoma. Nests of tumor
numerous mitoses and a dense lymphoid infiltrate (original with a cribriform configuration infiltrate the stroma (original
magnification x400). magnification x200).
Histopathology of Malignant Breast Disease 97

Figure 5–12. Invasive ductal carcinoma, no special type. Figure 5–13. Invasive ductal carcinoma, no special type.
Left, the tumor makes some glandular structures (original Left, the prominent gland formation scores 1 point for archi-
magnification x200). Right, the nuclei are moderately tecture (original magnification x100). Right, high nuclear
enlarged with inconspicuous nuclei and no mitoses (original grade and moderate mitotic activity score 3 and 2 points
magnification x400). In the Scarff-Bloom-Richardson system, respectively (original magnification x400). The total score of
this tumor would score 1 point for architecture, 2 points for 6 points makes this a grade II tumor.
nuclei, and 1 point for mitosis, making it grade I.

mentioned parameters. Based on the point total, dant problems. Frozen section is entirely appro-
tumors are assigned to grade I (three to five priate, however, when confirmation of invasive
points), grade II (six to seven points), or grade tumor is necessary prior to concurrent axillary
III (eight to nine points) (Figures 5–12 to dissection or when the surgeon requires intraop-
5–14). Further refinement of this method by erative margin assessment. The popularity of
Elston and Ellis21 defined specific criteria for sentinel node techniques using frozen section to
nuclear grade, architectural pattern, and mitotic determine whether to proceed with axillary dis-
rate, and has resulted in a system showing both section is also increasing.
strong correlation with outcome and reasonably In general, all lumpectomy and re-excision
good interobserver reproducibility. specimens must be assessed, at a minimum, for
tumor size, tumor type, and margin width. This
SPECIMEN HANDLING requires input from the surgeon as to specimen
AND REPORTING orientation, and some method for identifying

As the surgical approach to breast cancer has


changed, so has the pathologist’s approach to
specimen handling. Many lumpectomy speci-
mens arrive in the pathology lab with a diagno-
sis already established by fine-needle or stereo-
tactic core biopsy. If the specimen is oriented by
the surgeon using sutures or some other method,
the margins can then be optimally assessed on
permanent section. If the lesion was nonpalpa-
ble and removed using stereotactic localization,
radiography of the specimen is essential to con-
firm the adequacy of excision. Hormone recep- Figure 5–14. Invasive ductal carcinoma, no special type.
tor assays and other ancillary studies no longer Left, this tumor shows no gland formation (original magnifi-
cation x100). Right, the nuclei are high grade and show
require fresh tumor tissue, thus obviating the numerous mitoses (original magnification x400). The total
need to perform a frozen section, with its atten- score of 9 points makes this tumor grade III.
98 BREAST CANCER

margins on the histologic sections. Typically, Screening Pathology. Consistency achieved by


this involves the application of colored inks to 23 European pathologists in categorizing duc-
tal carcinoma in situ of the breast using five
the surfaces of the specimen prior to sectioning.
classifications. Hum Pathol 1998;29:1056–62.
Adequate fixation prior to processing is of 6. Silverstein MJ, Lagios MD, Craig PH, et al. A
utmost importance in achieving optimal results prognostic index for ductal carcinoma in situ of
for histologic examination. Since most breast the breast. Cancer 1996;77:2267–74.
specimens are fairly fatty, overnight formalin 7. Silverstein MJ, Lagios MD, Groshen S, Waisman
JR, Lewinsky BS, et al. The influence of mar-
fixation is often necessary. The attendant delay gin width on local control of ductal carcinoma
in reporting is more than offset by the quality of in situ of the breast. NEJM 1999;340:1455–61.
the information that can be gleaned from high 7. Foote F, Stewart F. Lobular carcinoma in situ: a
quality histologic sections. rare form of mammary carcinoma. Am J Pathol
Because of the large amount of pathologic 1941;17:491–6.
8. Elston CW, Ellis IO. Systemic pathology, Vol.13.
data entering into breast cancer prognosis and The breast. Edinburgh: Churchill Livingstone;
treatment strategy, many pathologists find it 1998.
helpful to utilize some sort of standardized 9. Yeatman TJ, Cantor AB, Smith TJ, et al. Tumor
reporting format as part of the pathology biology of infiltrating lobular carcinoma. Ann
report. As described above for DCIS, we use a Surg 1995;222:549–61.
10. Foote FW, Stewart F. A histologic classification of
breast cancer worksheet to describe relevant carcinoma in the breast. Surgery 1946;19:74–9.
features of invasive tumors. Features such as 11. Eusibi V, Magalhaes F, Azzopardi JG. Pleo-
tumor size, grade, subtype, and margin status morphic lobular carcinoma of the breast: an
are described, and this data is included as part aggressive tumor showing apocrine differentia-
tion. Hum Pathol 1992;23:655–62.
of the final report. The design of such a format
12. Weidner N, Semple JP. Pleomorphic variant of
should include input from clinicians to ensure invasive lobular carcinoma of the breast. Hum
they receive all information required for opti- Pathol 1992;23:1167–71.
mal patient management as well as any data 13. Rosen PP. Breast pathology. Philadelphia: Lippin-
that may be helpful in future retrospective stud- cott-Raven; 1997.
14. Cooper HS, Patchefsky AS, Krall RA. Tubular car-
ies. Standardized forms have been developed
cinoma of the breast. Cancer 1978;42:2334–42.
by the Association of Directors of Surgical 15. Parl FF, Richardson LD. The histologic and bio-
Pathology (ADSP) to serve this purpose. logic spectrum of tubular carcinoma of the
breast. Hum Pathol 1983;14:694–8.
REFERENCES 16. Ridolfi R, Rosen P, Port A, et al. Medullary carci-
noma of the breast. A clinicopathologic study
1. Azzopardi JG, Chepick OF, Hartmann WH. The with 10 year follow-up. Cancer 1977;40:1365–
World Health Organization histological typing 85.
of breast tumors. 2nd ed. Am J Clin Pathol 17. Wargotz ES, Silverberg SG. Medullary carcinoma
1982;78:806–16. of the breast. A clinicopathologic study with
2. The Consensus Conference Committee. Consensus appraisal of current diagnostic criteria. Hum
conference on the classification of ductal car- Pathol 1988;19:1340–46.
cinoma in situ. Cancer 1997;80:1798–1802. 18. Greenhough RB. Varying degrees of malignancy
3. Bellamy COC, McDonald C, Salter DM, et al. in cancer of the breast. J Cancer Res 1925;9:
Noninvasive ductal carcinoma of the breast: 452–63.
the relevance of histologic categorization. Hum 19. Bloom HJG, Richardson WW. Histological grad-
Pathol 1993;24:16–23. ing and prognosis in breast cancer. A study of
4. Douglas-Jones AG, Gupta SK, Attanoos RL, et al. 1409 cases of which 359 have been followed
A critical appraisal of six modern classifica- for 15 years. Br J Cancer 1957;11:359–77.
tions of ductal carcinoma in situ of the breast 20. Elston CW, Ellis IO. Pathological prognostic fac-
(DCIS): correlation with grade of associated tors in breast cancer. I. The value of histologi-
invasive tumor. Histopathology 1996;29:397– cal grade in breast cancer: experience from a
409. large study with long-term follow-up. Histo-
5. European Commission Working Group on Breast pathol 1991;19:403–10.
6
Unusual Breast Pathology
DAVID R. BRENIN, MD
HANINA HIBSHOOSH, MD
DAVID W. KINNE, MD

This chapter reviews clinical and pathologic fea- and a slightly higher proportion in men.1,2,3 It
tures of uncommon breast malignancies. The occurs in both an invasive and noninvasive
majority of the data used in the course of writing form. The World Health Organization (WHO)
the chapter was obtained from small studies of defined papillary carcinoma as follows: “A rare
specific tumor subtypes, or has been gleaned carcinoma whose invasive pattern is predomi-
from larger studies that included several types of nantly in the form of papillary structures. The
more common breast cancers. Unfortunately, same architecture is usually displayed in the
there is often insufficient information available metastases. Frequently, foci of intraductal pap-
to draw absolute conclusions regarding therapy illary growth are recognizable.”4 Further, the
and prognosis. WHO classification states that “papillary carci-
Much of the data cited was collected prior to noma arising, and limited to a mammary cyst,
the widespread use of breast conservation. For is [to be] referred to as noninvasive intracystic
this reason, the vast majority of patients studied carcinoma.”4 Invasive carcinoma, however, may
were treated using mastectomy. The reliance on be associated with an intracystic carcinoma.5
mastectomy has resulted in a lack of information Papillary carcinoma occurs most frequently
regarding the natural history and radiosensitivity in the central portion of the breast, and is asso-
of many of the tumors presented. Therefore, the ciated with a malignant nipple discharge in 22
risk of local recurrence for patients with rare to 34 percent of patients.1,6 The mean age of
breast malignancies opting for breast conserva- diagnosis for papillary carcinoma, 63 to 67
tion is unclear. There is, however, no reason to years, is older compared to the more common
suspect a significant difference in the risk of local types of breast cancer.1,2,7 The tumors tend to
recurrence in this group of patients compared to grow slowly, not infrequently being present for
patients with more common types of breast can- more than 1 year prior to patients seeking treat-
cer. Except where specifically indicated, the clin- ment. On physical exam, papillary carcinomas
ically appropriate use of breast conservation are well-circumscribed, and often lobulated.
should be considered in the informed treatment There may be a bloody nipple discharge. The
of patients with rare breast malignancies. average clinical size is 2 to 3 cm.3 Clinically
enlarged axillary lymph nodes are not uncom-
PAPILLARY CARCINOMA mon in patients with larger tumors containing
areas of hemorrhagic necrosis. Mammographi-
Papillary carcinoma accounts for 1 to 2 percent cally, papillary carcinomas typically have sharp
of newly diagnosed breast cancers in women, margins and are rounded or lobulated. Breast

99
100 BREAST CANCER

ultrasound may reveal a solid component in a with whole breast irradiation in patients with
cyst that otherwise appears benign. noninvasive papillary carcinoma. The use of
The appearance of the gross tumor varies breast conservation, however, appears reason-
with the proportion of the cystic component. able for these patients, as there is no reason to
Some fibrosis may be present. The cut surface of suspect a significant difference in the risk of
the tumor is typically described as tan or gray, local recurrence compared to patients with more
and areas of focal hemorrhage and necrosis are common types of noninvasive breast cancer.
not uncommon.3 Larger tumors may form a large The low rate of axillary metastasis observed
cyst containing partially clotted blood and tumor makes elimination of axillary dissection appro-
fragments. Microscopically, the tumors form a priate in patients with noninvasive papillary car-
predominately frond-like pattern (Figures 6–1 cinoma and a clinically negative axilla.
and 6–2). Cystic areas may be present but are not Even less data exists to aid in treatment
a prerequisite for diagnosis. Distinguishing selection for patients with invasive papillary
between benign and malignant papillary tumors carcinoma. Fisher and colleagues reported on
can be challenging. Kraus and Neubecker,8 35 patients with invasive papillary cancer.2 Of
Lefkowitz and colleagues,7 and Rosen3 have the 22 patients who underwent axillary dissec-
attempted to delineate guidelines for diagnosis. tion, 32 percent were found to have axillary
Various immunohistochemical markers have metastases. Of patients with axillary metastases,
been evaluated but have proved to be of little only two (nine percent) had four or more lymph
help. Analysis of DNA content, however, has nodes involved. Life-table plots calculated by
demonstrated significant differences between Fisher and colleagues showed a favorable prog-
papillary carcinoma and benign lesions.9,10 nosis comparable to patients with tubular can-
Papillary carcinoma has a favorable progno- cers. At 5-year follow-up, only one patient had
sis. Noninvasive papillary carcinoma is a variant died of papillary carcinoma. Recurrences, when
of ductal carcinoma in situ (DCIS), and is asso- they do occur, are typically “late,” coming > 5
ciated with a less than one percent rate of axil- years after the initial diagnosis.3 The majority of
lary metastasis.3,7 To date, there is no significant reports concerning the treatment of invasive
body of data addressing the use of radiation papillary carcinoma have addressed patients
therapy in the treatment of this lesion. As is the whose primary therapy consisted of mastectomy
case with DCIS, there are no prospective trials with or without axillary dissection. When clini-
comparing mastectomy to breast preservation cally appropriate, the use of breast conservation,

Figure 6–1. Papillary carcinoma demonstrating frond-like Figure 6–2. Intracystic papillary carcinoma. Note solid cyst
pattern (original magnification x400). wall on periphery (original magnification x400).
Unusual Breast Pathology 101

whole breast irradiation, and axillary dissection histology of carcinoma at metastatic sites may
or sentinel node biopsy is a reasonable option. not be predicted by the extent and subtype seen
in the breast.
METAPLASTIC The number of reported patients with meta-
MAMMARY CARCINOMA plastic mammary carcinomas is insufficient to
draw accurate conclusions concerning therapy
Metaplastic mammary carcinoma refers to a and prognosis. The majority of the data has been
classic breast carcinoma containing a variable obtained from small studies of specific tumor
component exhibiting a nonglandular growth subtypes. Most patients underwent mastectomy.
pattern. These tumors constitute fewer than one Rosen and Ernsberger reported that in four of
percent of breast cancers.11,12 The metaplastic seven patients treated with excisional biopsy
changes typically manifest as squamous cells, alone, disease locally recurred between 1 and 3.5
spindle cells, and/or as areas of heterologous years after diagnosis.16 There is no information
mesenchymal growth showing cartilaginous or concerning the responsiveness of metaplastic
osseous differentiation. The histologic diversity carcinoma to radiation or chemotherapy.15 When
observed in metaplastic mammary carcinoma compared to the more common histologies,
has led to various subdesignations including metaplastic mammary carcinoma has a low rate
spindle-cell carcinoma, carcinoma with osseous of axillary lymph node involvement.17–19 Distant
metaplasia, carcinoma with pseudosarcomatous failure, however, is common, with an overall 5-
metaplasia, squamous cell carcinoma with pseu- year survival rate reported to be 44 percent.13
dosarcomatous stroma, and carcinosarcoma. The Given the low rate of axillary metastasis and
histogenesis of these carcinomas is assumed to a lack of prognostic information gained by
be of ductal origin. Results derived from ultra- axillary staging in these patients, elimination of
structural and immunohistochemical studies axillary lymphadenectomy or the use of sentinel
suggest that metaplastic mammary carcinomas lymph node biopsy alone may be appropriate.
originate from undifferentiated multipotential
cells.13 Tavassoli suggested that myoepithelial APOCRINE CARCINOMA
cells are the cell of origin.11 Although the num-
ber of reported cases is small, all of the subtypes Apocrine carcinoma of the breast reportedly
appear to have a similar prognosis14 and will be accounts for 0.4 percent of new mammary
presented as a single group in this chapter. malignancies.20,21 This tumor derives its name
Metaplastic mammary carcinoma typically
presents as a mass. Skin changes and fixation
to underlying tissues have been reported.15 The
gross appearance of the tumor varies with sub-
type, but most are described as hard with well-
circumscribed borders. Cystic degeneration
may occur when there is an extensive squamous
metaplastic component.14 Histologically, meta-
plastic carcinoma is divided broadly into
tumors showing squamous and/or heterologous
(cartilaginous or osseous) (Figure 6–3) or pseu-
dosarcomatous differentiation. The former
appears to be more frequent; however, mixed
and transition forms are common. The extent
Figure 6–3. Metaplastic carcinoma with cartilagenous differ-
and degree of differentiation varies widely. The entiation (original magnification x400).
102 BREAST CANCER

from the apocrine glands normally present in ADENOID CYSTIC CARCINOMA


skin. Apocrine carcinomas of the breast, how-
ever, do not originate from apocrine glands of Adenoid cystic carcinoma of the breast, also
the skin. Apocrine carcinomas appear to arise known as cylindroma, is a rare neoplasm
from the apocrine metaplasia commonly found accounting for < 0.1 percent of mammary car-
in excised breast tissue.3 The histologic similar- cinomas.29,15 First described in the breast by
ity of apocrine metaplasia commonly found in Geschickter30 in 1945 and again by Foote and
excised breast tissue, rare carcinomas with Stewart5 in 1946, its characteristic histopatho-
apocrine differentiation, and apocrine glands of logic appearance is identical to like-named
the skin is due to their common embryological tumors arising from the salivary glands. Ade-
derivation from the epidermis. noid cystic carcinomas typically present in the
Apocrine carcinoma presents in a fashion sixth or seventh decade of life. The characteris-
similar to other, more common breast cancers. tic presentation is that of a 2 to 3 cm movable
The reported age range of affected patients is tumor which may be tender or painful.31 The
from 19 to 86 yrs.22–25 Most patients with infil- lesions tend to be centrally located in the
trating apocrine carcinoma of the breast present breast32 and may exhibit skin changes when
with a palpable mass.24,25 Abati and colleagues superficial. These tumors have a gray to pale
found that approximately one-third of both the yellow cut surface with well-defined margins.
intraductal and invasive lesions were detected Larger lesions have been found to undergo cys-
mammographically.24 Infiltrating apocrine car- tic degeneration.31,33 Adenoid cystic carcino-
cinomas are hard on palpation. Grossly, the mas of the breast have marked histological het-
lesions are typically gray to white with infiltrat- erogeneity, making diagnosis by needle biopsy
ing borders.3 Some tumors are cystic or have a problematic. Examination of many microscopic
medullary appearance.3 Microscopically, the fields may be required before the classic cylin-
cytoplasm is markedly eosinophilic and may be dromatous and/or cribriform growth pattern is
granular or homogeneous. The cellular architec- identified. Ro and colleagues divided adenoid
ture of both intraductal and invasive apocrine cystic carcinomas of the breast into three histo-
carcinomas is similar to that seen with more logic grades based on the proportion of solid
common mammary carcinomas. The distinction growth to the overall tumor size.34 Tumors with
between atypical apocrine hyperplasia and apoc- no solid component were classified as grade I,
rine intraductal carcinoma can be difficult.26–28 those with < 30 percent solid component were
The prognosis for patients with apocrine grade II, and tumors consisting of > 30 percent
carcinoma of the breast is generally considered solid component were grade III. Ro noted that
to be analogous to patients with similarly tumors with a solid component were more
staged ductal carcinomas.23–25 Abati and col- likely to be larger, recur, or metastasize.
leagues identified a 15 percent local recurrence Adenoid cystic carcinoma of the breast has
rate in 20 patients with intraductal apocrine an excellent prognosis. The rate of axillary
carcinomas treated by biopsy alone, but no metastasis is low, less than one percent.31,34 Dis-
recurrences in two patients treated with tant metastasis is rare.31 When systemic recur-
lumpectomy and irradiation.24 The majority of rence does occur, it is typically pulmonary.
reported patients with invasive apocrine carci- Metastases to bone,34 liver,34 brain,35 and kid-
noma have been treated with mastectomy and ney36 have also been reported. Distant metas-
some form of axillary dissection. The radiosen- tases typically occurr in patients who had nega-
sitivity of these lesions has yet to be deter- tive axillary dissections.31 There is no
mined, but the use of breast conserving therapy prospective data to support one therapeutic
may be appropriate. modality over another in the treatment of this
Unusual Breast Pathology 103

disease. Reported data on prognosis has been squamous debris. Microscopically, squamous
gathered mostly from patients treated with mod- cell carcinomas of the breast resemble similar
ified radical or radical mastectomy. The use of tumors arising in other sites. Keratin pearls and
breast conservation, however, appears reason- keratohyaline granules may be present.3
able for these patients, as there is no reason to As squamous cell carcinoma is a very rare
suspect a significant difference in the risk of lesion, information on prognosis and treatment
local recurrence compared to patients with more is limited. The majority of patients reported in
common types of breast cancer.37 The low rate the literature have been treated by mastectomy
of axillary metastasis observed, combined with with axillary dissection. Radiosensitivity of
a lack of prognostic information gained from this tumor has not been defined.
axillary staging, makes elimination of axillary
dissection appropriate in patients with adenoid SECRETORY CARCINOMA
cystic carcinoma and a clinically negative axilla.
Secretory carcinoma is a rare tumor affecting
SQUAMOUS CELL CARCINOMA both adults and children. In 1966, McDivitt and
Stewart described a series of seven young
Squamous cell carcinoma of the breast is an patients with this tumor, referring to it as juvenile
extremely rare form of metaplastic carcinoma carcinoma.41 It soon became apparent, however,
consisting of a lesion entirely, or nearly entirely, that most patients found to have this tumor were
composed of keratinizing squamous cell carci- not juveniles. Tavassoli and Norris, reporting on
noma. Typically, lesions composed of > 90 per- a series of 19 patients, found the median age at
cent keratinizing squamous carcinoma have the time of diagnosis to be 25 years, with six
been placed in this group. One must be careful patients being > 30 years of age.42 As it became
to exclude a metastatic squamous cell carci- obvious that the majority of patients with
noma or skin carcinoma involving the breast “juvenile carcinoma of the breast” were adults,
prior to accepting squamous cell carcinoma as a it was redesignated “secretory carcinoma.”
primary breast tumor. The usual precursor of Secretory carcinoma has been described in
this cancer is thought to be squamous meta- patients from the first to the eighth decade of
plasia, which occurs in a wide variety of set- life. Typically, these lesions are palpable and
tings including fibroadenoma, cystic lesions, present as painless, well-circumscribed masses.
phyllodes tumors, gynecomastia, mammary Grossly, secretory carcinomas are white to gray
duct hyperplasia, papillomatosis, subareolar or tan to yellow in color and may be lobulated.3
abscesses, and areas of inflammation. 3 In some The margins are usually well-circumscribed
cases squamous cell carcinoma may represent a and rarely infiltrative. Microscopically, the
variant of metaplastic carcinoma in which the cells are filled with secretory material which is
adenocarcinomatous component has been over- pale pink or amphophilic when stained with
grown by the squamous component.15 hematoxylin and eosin (Figure 6–4).3,41,42
The mean age at diagnosis of patients with Secretory carcinoma is considered a low-
squamous cell carcinoma of the breast is simi- grade carcinoma with an excellent prognosis.
lar to that seen with more common breast Axillary metastasis has been identified in
cancers.38,39 The lesions are usually palpable, approximately 20 percent of cases, but very few
and fixation to the chest wall as well as skin patients have been reported to have distant
involvement have been observed. Calcifica- metastasis.42–44 There is, however, a risk of late
tions may be seen on mammography.40 Grossly, local recurrence.43,45 Wide local excision is pre-
the tumors frequently undergo cystic degenera- ferred in children, with an attempt to preserve
tion producing a cavity filled with necrotic the breast bud. In adults, breast conservation is
104 BREAST CANCER

tumors are palpable.3 Grossly, there are no fea-


tures specifically associated with endocrine dif-
ferentiation. Microscopically, most carcinomas
of the breast with endocrine differentiation con-
tain argyrophilic cytoplasmic granules. Rarely,
choriocarcinomatous differentiation may occur,
resulting in tumors that are microscopically
similar to syncytiotrophoblast and cytotro-
phoblast and are strongly reactive for HCG.3
There is general agreement that patients
with carcinomas of the breast with endocrine
Figure 6–4. Secretory carcinoma (original magnification x400). differentiation have a similar prognosis to those
with like-staged more common mammary
appropriate. Axillary lymphadenectomy should cancers.3,46 Treatment selection should be based
be performed selectively based on physical on conventional clinical and pathologic criteria.
examination or with the identification of
metastases on sentinel node biopsy. Radiosen- PHYLLODES TUMOR
sitivity of this tumor has not been defined, and
the majority of reported patients treated using First characterized by Muller50 in 1838, phyl-
breast conservation have not received postoper- lodes tumors are fibroepithelial neoplasms
ative radiation therapy.45 accounting for approximately 0.3 to 0.5 percent
of breast tumors in women.51,52 This tumor’s
CARCINOMA OF THE BREAST WITH other name, cystosarcoma phyllodes, is used
ENDOCRINE DIFFERENTIATION less often and considered by some as misleading
for a lesion that is more often benign than
Rarely, tumors of the breast may undergo malignant. These tumors may be locally aggres-
endocrine metaplasia and have the ability to pro- sive but have minimal capability for metastasis.
duce ectopic hormones such as human chorionic Phyllodes tumors have been the subject of many
gonadotropin (HCG), calcitonin, adrenocorti- reports, but their optimal management has yet to
cotropin, and epinephrine. Endocrine differenti- be clearly defined.
ation may arise in the setting of ductal carcinoma Patients typically present with firm, discrete,
in situ, small-cell undifferentiated carcinoma, mobile masses often clinically indistinguishable
mucinous carcinoma, lobular carcinoma, and from fibroadenomas. Palpable axillary lymph
infiltrating ductal carcinoma.3,46 Rarely, the nodes may be present in as many as 20 percent
microscopic architecture of a breast cancer with of patients but they are infrequently involved by
endocrine differentiation may mimic the histo- tumor.3,53 The median age at presentation in the
logic structure of nonmammary tissue that con- majority of published series is the fourth or fifth
tains the ectopic hormone being produced. decade, with a range of 10 to 86 years.3,53,54 It is
The clinical presentation of patients with important to note, however, that the mean age of
carcinomas of the breast with endocrine differ- presentation for patients with a fibroadenoma,
entiation is similar to patients with more com- approximately 30 years, is significantly lower
mon mammary neoplasms. Systemic symptoms than that for phyllodes tumors.55 The occurrence
attributable to the ectopic hormone produced of phyllodes tumors in patients < 30 years is
are absent in nearly all cases. However, rare rare.3 Bernstein and colleagues identified race-
reports of systemic manifestations ascribed to specific differences in the incidence and mean
ectopic hormones do exist.47–49 Most of these age of diagnosis of patients in Los Angeles
Unusual Breast Pathology 105

County with phyllodes tumors.56 The average


annual age-adjusted incidence rate for all racial-
ethnic groups combined was 2.1 per 1 million
women in the population. Latina whites had the
highest incidence rate (2.8 per 1 million popula-
tion) followed by non-Latina whites, Asians, and
African Americans, respectively.56 Bernstein and
colleagues found that the mean age of diagnosis
for non-Latina whites was 53.7 years, for Latina
whites 45.8 years, African Americans 48.7 years,
and Asians 32.9 years. Clinically, tumors that
Figure 6–5. Cut surface of phyllodes tumor.
exhibit rapid growth, are > 4 cm, or previously
stable tumors that suddenly increase in size,
should arouse suspicion. Mammographically, hypercellular stromal overgrowth. Cellular
these lesions are smooth and lobulated. With pleomorphism is common, with typically > 5
locally invasive disease, margin irregularity may mitoses per 10 HPF.3 Although the proportion
be present. Ultrasound typically reveals a solid of patients with clinically enlarged axillary
mass with no posterior shadowing. Cysts may be lymph nodes approaches 20 percent,3,54 the rate
present within the lesion. of pathologically confirmed axillary metastasis
Grossly, phyllodes tumors are well circum- is well under 5 percent.53,58,59
scribed and firm. On sectioning, the tumors are The likelihood that a phyllodes tumor will
gray to tan and bulging (Figure 6–5). Focal cys- metastasize and/or locally recur depends on its
tic necrosis may be present. Microscopically, histologic classification. Histologically, benign
phyllodes tumors can be difficult to differenti- lesions have a local recurrence rate of six to ten
ate from benign cellular fibroadenomas. Typi- percent.54,60,61 and minimal risk of systemic
cally, an increased cellularity of the stromal metastasis.3,62 Low-grade malignant phyllodes
component is present. Long epithelial-lined tumors locally recur in 25 percent to 32 percent
clefts (intracanalicular pattern) are a prominent of cases3,57 and have a reported incidence of
feature and may help differentiate these tumors distant metastasis of under 5 percent.3 Tumors
from fibroadenomas (Figure 6–6). Mixoid with a malignant histologic classification have
changes may be present. Some degree of a high rate of local recurrence and a 25 percent
epithelial hyperplasia is common, with as many risk of systemic metastasis.3,57,62
as 10 percent of tumors containing squamous
metaplasia.57 Histologically, these tumors are
divided into three groups: benign, low-grade
malignant (borderline), and high-grade malig-
nant (Table 6–1). Benign phyllodes tumors are
characterized by having < 1 mitosis per 10
high-power fields (HPF). The stromal expan-
sion is uniform throughout the lesion, and the
cellularity is modest in extent with mild cellu-
lar atypia. Low-grade malignant tumors typi-
cally have microscopically invasive borders,
moderate heterogeneously distributed stromal
expansion, and < 5 mitoses per 10 HPF. High-
grade malignant phyllodes tumors have marked Figure 6–6. Phyllodes tumor (original magnification x100).
106 BREAST CANCER

Table 6–1. DIFFERENTIATION OF BENIGN, the lesion and established the following crite-
BORDERLINE, AND MALIGNANT PHYLLODES TUMORS
ria: (1) a close association between breast tissue
Benign Borderline Malignant and the infiltrating lymphoma, (2) no history of
Borders Pushing Mostly pushing Infiltrating extramammary lymphoma, and (3) the breast
Atypia Slight Moderate Marked must be the primary clinical site.
Mitoses/10HPF <1 1–4 ³5 Patients with primary breast lymphoma typ-
Stromal overgrowth Rare Occasional Frequent ically present with a palpable, sometimes ten-
HPF = high-power field
der, breast mass.74–76 Rapid growth of the tumor
is common.75,76 Diffuse infiltration, skin
changes, and clinically palpable axillary nodes
Primary therapy of phyllodes tumors is have been described.75–77 Lymphoma “B” type
aimed at reducing the risk of local recurrence. symptoms are rare.3,74–76,78 Bilateral involve-
These tumors must be excised to clear surgical ment has been reported in up to 25 percent of
margins. The magnitude of the negative mar- patients.74,78,79 The majority of patients present
gin must be dictated by the histologic features in their sixth decade of life, but a bimodal age
of the tumor and the size of the breast. Exci- distribution with peaks in the mid-30s and mid-
sion with negative margins up to 2 cm has 60s has been reported.3,74–76,78–80 Mammog-
been suggested by some authors.3,15,53,54,62 raphy and ultrasound of patients with primary
Mastectomy may be indicated if the lesion breast lymphoma demonstrates a solitary mass
cannot be completely excised in a cosmeti- in the majority of cases. The imaging charac-
cally acceptable wide local excision. Axillary teristics of this lesion are nonpathognomonic.81
dissection is not required. Clinically suspi- Grossly, these tumors have a gray-white cut
cious nodes are invariably hyperplastic and surface, are well circumscribed, fleshy, and
should be individually biopsied. Locally may be nodular.3 The majority of primary
recurrent phyllodes tumor does not mandate breast lymphomas are classified as, mixed, or
mastectomy. Complete excision to wide nega- large cell with diffuse architecture and a B-cell
tive margins is acceptable.15 phenotype.3,75,76,78,79 T-cell tumors are rare.74,78
The role of radiotherapy in the treatment of In some cases, the linear arrangement of lym-
patients with phyllodes tumor remains unclear. phoma cells in the stroma may mimic invasive
There are multiple reports of insensitivity to lobular carcinoma. Immunostains for epithelial
radiation when used for palliation.63–65 Two and lymphoid markers may be required to dif-
investigators, however, describe the use of post- ferentiate between the two.3
operative whole breast irradiation following Patients with primary breast lymphoma
breast-preserving surgery for phyllodes must be staged in a manner similar to other
tumor.66,67 Local recurrence rates were not lymphoma patients. Local excision followed
reported in these studies. by radiation therapy provides excellent local
control.70,75,82,83 Negative margins are not
PRIMARY BREAST LYMPHOMA required. Most patients who fail therapy will
recur at distant sites or in the other breast.3 As
Primary lymphoma of the breast is a rare dis- primary therapy appears to have little impact on
ease, accounting for < 1.0 percent of all breast survival, radical surgery is rarely required in
malignancies.68–70 The origin of this tumor the treatment of patients with primary breast
remains unclear. Several investigators have lymphoma. Patients with stage I disease and
suggested that mucosa-associated lymphoid tis- those with histologically low-grade tumors
sue (MALT) may play a role in its develop- have the most favorable prognosis.69 Systemic
ment.71–73 In 1972, Wiseman and Liao defined therapy should be considered in all cases.3
Unusual Breast Pathology 107

BREAST SARCOMA sarcomas of the breast are similar to their more


common counterparts occurring in other areas
First described in 1828 by Chelius,84 breast sar- of the body (Figure 6–7). In the breast, however,
coma accounts for less than one percent of all metaplastic carcinoma must be excluded prior
breast malignancies,85,86 with an annual inci- to establishing a diagnosis of mammary sar-
dence in the United States of approximately coma.3 Diagnosis of this lesion requires exten-
17.5 new cases per 1 million women.87 The rar- sive sampling to rule out the presence of in situ
ity of this lesion has resulted in reports on or invasive carcinoma. Immunohistochemical
breast sarcoma typically addressing a heteroge- studies for epithelial markers may be useful in
neous group of tumors, including malignant difficult cases. Axillary lymph node involve-
phyllodes tumors. Mammary sarcomas should ment is exceedingly uncommon.3 Gutman and
be limited to tumors arising from interlobular colleagues identified axillary nodal metastases
mesenchymal elements comprising the sup- only in the context of disseminated disease.89
porting stroma.3 These tumors include liposar- Breast sarcomas should be treated similarly
coma, leiomyosarcoma, osteogenic sarcoma, to sarcomas occurring elsewhere in the body.
chondrosarcoma, malignant fibrous histiocy- Surgery is the mainstay of treatment. Wide local
toma, fibrosarcoma, rhabdomyosarcoma, pri- excision with histologically negative margins is
mary angiosarcoma, and hemangiopericytoma. required.15,89,93 Mastectomy may be necessary
Also included in this category, but discussed to ensure complete excision of larger tumors.
separately in this review, are postradiotherapy Neoadjuvant chemoradiation should be consid-
angiosarcomas. Phyllodes tumors, which arise ered in patients with large tumors. No staging or
from intralobular and periductal stroma, should therapeutic role for axillary lymphadenectomy
be excluded. has been demonstrated.89 Axillary lymph node
Typically, breast sarcomas present as painless, dissection need be performed only if required
mobile, well-circumscribed breast masses.3,15,88,89 for complete excision of the tumor.
There is commonly a history of rapid growth in Gutman and colleagues, reporting on 60
a pre-existing mass.15,89 Skin involvement and cases, found a median disease-free survival of
nipple changes have been reported but are infre- 17.7 months and median overall survival of 67
quent.88,90 Enlarged axillary lymph nodes may be months (median follow-up of 120 months).89
palpable, but pathologically confirmed axillary Local failure was reported in 19 patients. Those
metastases are rare. The mean age at the time of suffering local failure typically did so within
presentation in three recent reports ranged from the first 24 months. Patients with sarcomas < 5
44 to 55 years (range 16 to 87 years).89–91 Mam-
mographically, these lesions typically appear as
well-circumscribed, dense masses. Rarely, the
presence of osseous trabeculae within an
osteogenic sarcoma may be noted.92
Grossly, the majority of breast sarcomas are
well circumscribed. The tumors usually grow as
expansile masses, compressing surrounding tis-
sue as they enlarge. The margin of a liposar-
coma may be multinodular and infiltrative. The
cut surface is typically yellow, gray or white in
color. There may be a whorled texture as well as
areas of necrosis. Gelatinous areas are fre-
quently noted in liposarcomas. Histologically,
Figure 6–7. High grade sarcoma (original magnification x100).
108 BREAST CANCER

cm in size were found to have a significantly cent when a median latency period of 74
better prognosis.89 Pollard and colleagues iden- months is considered.97
tified an overall 5-year mortality of 64 percent The median interval between breast-preserv-
with a local recurrence rate of 44 percent in the ing therapy and the occurrence of angiosarcoma
25 patients in their series.90 The role of adjuvant of the breast has been reported to be between 6
radiotherapy has yet to be defined.89 and 11 years (range 2 to 44).95,97,98,100 Patients
typically present with skin changes reminiscent
POSTRADIOTHERAPY of a hematoma. These changes may be present
ANGIOSARCOMA over a broad area within the radiation field. An
underlying breast mass may be present. Red-
The occurrence of sarcoma following radiother- dish-purple skin patches as well as vesicles have
apy has been well described.94,95 The wide- also been reported.97 Findings on mammog-
spread acceptance of breast preservation in the raphy, ultrasound, and MRI are nonspecific.103
treatment of breast cancer may have an unfore- Grossly, the tumors may be friable, firm, or
seen secondary result: an increase in the number spongy. Areas of cystic hemorrhagic necrosis
of patients at risk for developing post irradiation are common in high-grade lesions.3 Histologi-
sarcoma. Postradiotherapy sarcoma was defined cally, the postirradiated angiosarcoma of the
by Cahan and colleagues in 1948 as sarcoma chest wall primarily occurs in the skin, with
developing in a previously irradiated field after occasional extension to underlying subcuta-
a latency period of several years.96 Angiosar- neous tissue or breast. It shows a wide spectrum
coma, osteosarcoma, malignant fibrous histio- of degree of differentiation but is commonly
cytoma, and fibrosarcoma have all been high grade. Well-formed inter-anastomosing
reported in the irradiated breast. Angiosarcoma vascular channels corresponding to low-grade
has been the topic of many recent reports.97–100 angiosarcoma frequently merge with high-grade
The rarity of these tumors makes the true inci- solid or spindle-cell-containing regions (Figure
dence of postradiotherapy angiosarcoma of the 6–8). Distinction from postirradiation benign
breast difficult to determine. The estimated risk vascular changes including vascular or lym-
of patients treated with whole breast irradiation phatic ectasia and the so called “atypical vascu-
ranges from 0.06 to 0.4 percent.97,100–102 Strobbe lar lesions” in addition to hemangiomas is
and colleagues, reporting on 21 patients with mandatory.104 Axillary lymph node involvement
postradiotherapy angiosarcomas of the breast is uncommon. Gutman and colleagues, report-
collected from the Netherlands cancer registry, ing on 17 patients with angiosarcoma, identified
cite a potential incidence as high as 1.59 per- axillary nodal metastases only in the context of
disseminated disease.89
Surgical treatment of postradiotherapy angio-
sarcoma should consist of complete resection
with wide negative margins. Salvage mastec-
tomy with en-bloc resection of involved skin
and adjacent structures is often required.97,105
Reconstruction of the resulting defect may
require musculocutaneus flaps and/or skin
grafts. Rarely, patients with small lesions may
be adequately treated with a partial mastec-
tomy.105 Axillary dissection need be performed
only if required for complete excision of the
Figure 6–8. Angiosarcoma (original magnification x100). tumor.105,106
Unusual Breast Pathology 109

The prognosis for primary angiosarcoma of 12. Rottino A, Wilson K. Osseous, cartilaginous and
the breast appears to depend on the grade of the mixed tumors of the human breast: a review of
the literature. Arch Surg 1945;50:184–93.
tumor.105 The prognosis for postradiotherapy
13. Kaufman MW, Marti JR, Gallager HS, Hoehn JL.
angiosarcomas, however, remains unclear. The Carcinoma of the breast with pseudosarcoma-
number of reported patients with postradiother- tous metaplasia. Cancer 1984;53:1908–17.
apy angiosarcomas is insufficient to draw accu- 14. Oberman HA. Metaplastic carcinoma of the breast:
rate conclusions regarding prognosis. High- a clinicopathologic study of 29 patients. Am J
grade lesions are aggressive and tend to recur Surg Pathol 1987;11:918–29.
15. Rosen P. Invasive mammary carcinoma. In: Harris
locally.107 Low-grade tumors appear to have a
JR, Hellman S, Harris JR, et al., editors. Dis-
more favorable prognosis.3,98,108 ease of the breast. Philadelphis: Lippincott–
Raven Publishers, 1996.
REFERENCES 16. Rosen PP, Ernsberger D. Low-grade adenosqua-
mous carcinoma a variant of metaplastic mam-
1. Haagensen CD. In: Diseases of the breast. 2nd ed. mary carcinoma. Am J Surg Pathol 1987;11:
Philadelphia: W. B. Saunders; 1971. 351–8.
2. Fisher ER, Palekar AS, Redmond C, et al. Patho- 17. Bauer TW, Rostock RA, Eggleston JC, Baral E.
logic findings from the National Surgical Spindle cell carcinoma of the breast. Hum
Adjuvant Breast Project (Protocol No. 4). VI. Pathol 1984;15:148–52.
Invasive papillary cancer. Am J Clin Pathol 18. Gersell DJ, Katzenstein AA. Spindle cell carcinoma
1980;73:313–20. of the breast. Hum Pathol 1981;12:550–60.
3. Rosen PP. In: Rosen’s breast pathology. Philadel- 19. Huvos AG, Lucas JC, Foote FW Jr. Metaplastic
phia: Lippincott-Raven Publishers; 1997. breast carcinoma. NY State J Med 1973;73:
4. World Health Organization. Histological typing of 1078–81.
breast tumours. 2nd ed. International histolog- 20. Mossler JA, Barton TK, Brinkhous AD, et al.
ical classification of tumours #2. Genova: Apocrine differentiation in human mammary
World Health Organization; 1981. carcinoma. Cancer 1980;46:2463–71.
5. Foote FW Jr, Stewart FW. A histologic classifica- 21. Azzopardi JG. Problems in breast pathology. Lon-
tion of carcinoma of the breast. Surgery don; W. B. Saunders; 1979.
1946;19:74–99. 22. Eusebi V, Betts C, Haagensen DE, et al. Apocrine
6. Carter D. Intraductal papillary tumors of the differentiation in lobular carcinoma of the
breast. A study of 76 cases. Cancer 1977;39: breast: a morphologic, immunologic and ultra-
1689–92. structural study. Hum Pathol 1984;15:134–40.
7. Lefkowitz M, Lefkowitz W, Wargotz ES. Intra- 23. Lee BJ, Pack GT, Scharnagel I. Sweat gland can-
ductal (intracystic) papillary carcinoma of the cer of the breast. Surg Gynecol Obstet 1933;
breast and its variants: a clinicopathological 54:975–96.
study of 77 cases. Hum Pathol 1994;25:802–9. 24. Abati AD, Kimmel M, Rosen PP. Apocrine mam-
8. Kraus FT, Neubecker RD. The differential diagno- mary carcinoma a clinicopathologic study of
sis of papillary tumors of the breast. Cancer 72 cases. AJCP 1990;94:371–7.
1962;15:444–5. 25. d’Amore ESG, Terrier-Lacombe MJ, Travagli JP,
9. Tsuda H, Uei Y, Fukutami T, Hirohashi S. Differ- et al. Invasive apocrine carcinoma of the
ent incidence of loss of heterozygosity on breast: a long term follow-up study of 34 cases.
chromosome 16q between intraductal papil- Breast Cancer Res Treat 1988;12:37–44.
loma and intracystic papillary carcinoma of the 26. O’Malley FP, Page DL, Nelson EH, Dupont WD.
breast. Jpn J Cancer Res 1994;85:992–6. Ductal carcinoma in situ of the breast with
10. Tsuda H, Takarabe T, Susumu N, et al. Detection of apocrine cytology: definition of a borderline
numerical and structural alterations and fusions category. Hum Pathol 1994;25:164–8.
of chromosomes 16 and 1 in low-grade papil- 27. Tavossoli FA, Norris HJ. Intraductal apocrine car-
lary breast carcinoma by fluorescence in situ cinoma: a clinicopathologic study of 37 cases.
hybridization. Am J Pathol 1997;151:1027–34. Mod Pathol 1994;7:813–8.
11. Tavassoli FA. Classification of metaplastic carci- 28. Carter D, Rosen PP. Atypical apocrine metaplasia
noma of the breast. Pathol Annu 1992;27: in sclerosing lesions of the breast: a study of 51
89–119. patients. Mod Pathol 1991;4:1–5.
110 BREAST CANCER

29. Cavanzo FJ, Taylor HB. Adenoid cystic carcinoma with endocrine differentiation: a review. Vir-
of the breast. Cancer 1969;24:740–5. chows Arch 1994;425:449–57.
30. Geschickter CF. In: Diseases of the breast. 47. Coombes RC, Easty GC, Detre SI, et al. Secretion
Philadelphia: J. B. Lippincott; 1945. of immunoreactive calcitonin by human breast
31. Peters GN, Wolff M. Adenoid cystic carcinoma of carcinomas. Br Med J 1975;4:197–9.
the breast. Report of 11 new cases: review of 48. Woodard BH, Eisenbarth G, Wallace NR, et al.
the literature and discussion of biological Adrenocorticotropin production by a mam-
behavior. Cancer 1982;52:680–6. mary carcinoma. Cancer 1981;47:1823–7.
32. Qizilbash AH, Patterson MC, Oliveira KF. Ade- 49. Kaneko H, Hojo H, Ishikowa S, et al. Norepi-
noid cystic carcinoma of the breast. Arch nephrine-producing tumors of bilateral breasts.
Pathol Lab Med 1977:101;302–6. A case report. Cancer 1978;41:2002–7.
33. Rosen PP. Adenoid cystic carcinoma of the breast: 50. Muller J. Uber den fein eran Bau and die Forman
a morphologically heterogeneous neoplasm. der Krakhaften Geschwilste. Berlin: G. Reiver;
Pathol Annu 1989;24:237–54. 1838.
34. Ro JY, Silva EG, Callager HS. Adenoid cystic car- 51. Oberman HA. Cystosarcoma phyllodes. A clini-
cinoma of the breast. Hum Pathol 1987;18: copathologic study of hypercellular periductal
1276–81. stromal neoplasms of the breast. Cancer
35. Orenstein JM, Dardick I, van Nostrand AW. Ultra- 1965;18:697–710.
structural similarities of adenoid cystic carci- 52. Rowell MD, Perry RR, Hsiu JG, Barranco SC.
noma and pleomorphic adenoma. Histopathol- Phyllodes tumors. Am J Surg 1993;165:376–9.
ogy 1985;9:623–38. 53. Reinfuss M, Mitus J, Krzysztof D, et al. The treat-
36. Herzberg AJ, Bossen EH, Walther PJ. Adenoid ment and prognosis of patients with phyllodes
cystic carcinoma of the breast metastatic to the tumor of the breast an analysis of 170 cases.
kidney. A clinically symptomatic lesion requir- Cancer 1996;77:910–16.
ing surgical management. Cancer 1991;68: 54. Rajan PB, Cranor ML, Rosen PP. Cystosarcoma
1015–20. phyllodes in adolescent girls and young
37. Leeming R, Jenkins M, Mendelsohn G. Adenoid women: a study of 45 patients. Am J Surg
cystic carcinoma of the breast. Arch Surg Pathol 1998;22:64–9.
1992;127:233–5. 55. Foster ME, Garrahan N, Williams S. Fibroade-
38. Rostock RA, Bauer TW, Eggleston JC. Primary noma of the breast: a clinical and pathological
squamous carcinoma of the breast: a review. study. J R Coll Surg Edin 1988;33:16–19.
Breast 1984;10:27–31. 56. Bernstein L, Deapen D, Ross RK. The descriptive
39. Shousha S, James AH, Ferandez MD, Bull TB. epidemiology of malignant cystosarcoma phyl-
Squamous cell carcinoma of the breast. Arch lodes tumors of the breast. Cancer 1993;71:
Pathol Lab Med 1984;108:893–6. 3020–4.
40. Tashjian J, Kuni CC, Bohn LE. Primary squamous 57. Grimes MM. Cystosarcoma phyllodes of the breast:
cell carcinoma of the breast; mammographic histologic features, flow cytometry analysis and
findings. J Can Assoc Radiol 1989;40:228–9. clinical correlations. Mod Pathol 1992;5:232–9.
41. McDivitt, RW, Stewart FW. Breast carcinoma in 58. Palmer ML, De Risi DC, Pelikan A, et al. Treat-
children. JAMA 1966;195:388–90. ment options and recurrence potential for cys-
42. Tavassoli FA, Norris HJ. Secretory carcinoma of tosarcoma phyllodes. Surg Gynecol Obstet
the breast. Cancer 1980;45:2404–13. 1990;170:193–6.
43. Akhtar M, Robinson C, Ali MA, Godwin JT. 59. Norris HJ, Taylor HB. Relationship of histologic
Secretory carcinoma of the breast in adults. features to behavior of cystosarcoma phyl-
Light and electron microscopic study of three lodes: analysis of ninety-four case. Cancer
cases with review of the literature. Cancer 1967;20:2090–9.
1983;51:2245–54. 60. Ciatto S, Bonardi R, Cataliotti L, Cardona G and
44. Rosen PP, Cranor ML. Secretory carcinoma of the members of the Coordinating Center and Writ-
breast. Arch Pathol Lab Med 1991;115:141–4. ing Committee of FONCAM. Phyllodes tumor
45. Krausz T, Jenkins D, Grontoft O, et al. Secretory of the breast: a multicenter series of 59 cases.
carcinoma of the breast in adults: emphasis on Eur J Surg Oncol 1992;18:545–9.
late recurrence and metastasis. Histopathology 61. Zurrida S, Bartoli C, Galimberti V, et al. Which
1989;14:25–36. therapy for unexpected phyllode tumour of the
46. Maluf HM, Koerner FC. Carcinomas of the breast breast? Eur J Cancer 1992;28:654–7.
Unusual Breast Pathology 111

62. Hines JR, Marad TM, Beal JM. Prognostic indica- 79. Wiseman C, Liao KT. Primary lymphoma of the
tors in cystosarcoma phyllodes. Am J Surg breast. Cancer 1972;29:1705–12.
1987;153:276–80. 80. Adair FE, Hermann JB. Primary lymphosarcoma
63. Sheen-Chen S, Chou F, Chen W. Cystosarcoma of the breast. Surgery 1944;16:836–53.
phyllodes of the breast: a review of clinical, 81. Liberman L, Giess CS, Dershaw DD, et al. Non-
pathological and therapeutic options in 18 Hodgkin lymphoma of the breast: imaging
cases. Int Surg 1991;76:101–4. characteristics and correlation with histopatho-
64. Al-Jurf A, Hawk Wa, Crile G. Cystosarcoma phyl- logic findings. Radiology 1994;192:157–60.
lodes. Surg Gynecol Obstet 1978;146:358–64 82. DeBlasio D, McCormick B, Straus D, et al. Defin-
65. Blichert-Toft M, Hansen JPH, Hansen OH, itive irradiation for localized non-Hodgkin’s
Schiodt T. Clinical course of cystosarcoma lymphoma of the breast. Int J Radiat Oncol
phyllodes related to histologic appearance. Biol Phys 1989;17:843–6.
Surg Gynecol Obstet 1975;140:929–31. 83. Smith MR, Brustein S, Straus DJ. Localized non-
66. Cohn-Cedermark G, Rutqvist L, Rosendahl I, Sil- Hodgkin’s lymphoma of the breast. Cancer
fversward C. Prognostic factors in cystosar- 1987;59:351–4.
coma phyllodes—a clinicopathologic study of 84. Chelius MJ. Teleangiektasie. Heidelberger Klin
77 patients. Cancer 1991;68:2017–22. Ann 1828;499–517.
67. Hopkins ML, McGowan TS, Rawlings G, et al. 85. Oberman HA. Sarcomas of the breast. Cancer
Phyllodes tumor of the breast: a report of 14 1965;10:1233–43.
cases. J Surg Oncol 1994;56:108–12. 86. Callery CD, Rosen PP, Kinne DW. Sarcomas of
68. Fischer MG, Chideckel NG. “Primary” lym- the breast. A study of 32 patients with reap-
phoma of the breast. Breast 1984;10:7–9. praisal of classification and therapy. Ann Surg
69. Giardini R, Piccolo C, Rilke F. Primary non- 1985;201:527–32.
Hodgkin’s lymphomas of the female breast. 87. May DS, Stroup NE. The incidence of sarcomas
Cancer 1992;69:725–35. of the breast among women in the U.S. 1973–
1986. Plast Reconstruct Surg 1991;87:193–4.
70. Anania G, Baccarani U, Risaliti A, et al. Primary
88. Moore MP, Kinne DW. Breast sarcoma. Surg Clin
non-Hodgkin’s T-cell lymphoma of the breast.
North Am 1996;76:383–92.
Eur J Surg 1997;163:633–5.
89. Gutman H, Pollock RE, Ross MI, et al. Sarcoma
71. Mattia AR, Ferry JA, Harris NL. Breast lymphoma.
of the breast: implications for extent of therapy
A B-cell spectrum including the low grade B-
the M.D. Anderson experience. Surgery 1994;
cell lymphoma of mucosa associated lymphoid
116:505–9.
tissue. Am J Surg Pathol 1993;17:574–87.
90. Pollard SG, Marks PV, Temple LN, Thompson
72. Isaacson PG. Lymphomas of mucosa-associated
HH. Breast sarcoma: a clinicopathologic
lymphoid tissue (MALT). Histopathology
review of 25 cases. Cancer 1990;66:941–4.
1990;16:617–19. 91. Terrier MJ, Terrier-Lacombe H, Mourisees S, et
73. Lamovec J, Jancar J. Primary malignant lymphoma al. Primary breast sarcoma: a review of 33
of the breast. Lymphoma of the mucosa-associ- cases with immunohistochemistry and prog-
ated lymphoid tissue. Cancer 1987;60:3033–41. nostic factors. Breast Cancer Res Treat 1989;
74. Arber DA, Simpson JF, Weiss LM, Rappaport H. 13:39–48.
Non-Hodgkin’s lymphoma involving the 92. Elson BC, Ikeda DM, Andersson I, Wattsgard C.
breast. Am J Surg Pathol 1994;18:288–95. Fibrosarcoma of the breast: mammographic
75. El-Ghazaway IMH, Singletary SE. Surgical man- findings in five cases. AJR 1992;158:993–5.
agement of primary lymphoma of the breast. 93. Mies C. Mammary sarcoma and lymphoma. In:
Ann Surg 1991;214:724–6. Bland KI, Copeland EM, editors. The breast.
76. Misra A, Kapur BML, Rath GK. Primary breast Philadelphia: W. B. Saunders; 1998.
lymphoma. J Surg Oncol 1991;47:265–70. 94. Laskin WB, Silverman TA, Enzinger FM. Postra-
77. Hugh JC, Jackson FI, Hanson J, Poppema S. Pri- diation soft tissue sarcomas: an analysis of 53
mary breast lymphoma. An immunohistologic cases. Cancer 1988;62:2330–40.
study of 20 new cases. Cancer 1990;66:2602–17. 95. Brady MS, Gaynor JJ, Brennan MF. Radiation-
78. Jeon HJ, Akagi T, Hoshida Y, et al. Primary non- associated sarcoma of bone and soft tissue.
Hodgkin malignant lymphoma of the breast. Arch Surg 1992;127:1379–85.
An immunohistochemical study of seven 96. Cahan WG, Woodard HW, Higinbotham NL, et al.
patients with breast lymphoma in Japan. Can- Sarcoma arising in irradiated bone: report of
cer 1992;70:2451–9. 11 cases. Cancer 1948;1:3–29.
112 BREAST CANCER

97. Strobbe LJA, Peterse HL, van Tinteren H, et al. surgery and irradiation for early mammary
Angiosarcoma of the breast after conservation cancer. Radiother Oncol 1994;30:271–3.
therapy for invasive cancer, the incidence and 103. Schnarkowski P, Kessler M, Arnholdt H, Helm-
outcome. An unforeseen sequela. Breast Can- berger T. Angiosarcoma of the breast: mammo-
cer Res Treat 1998;47:101–9. graphic, sonographic, and pathological find-
98. Bolin DJ, Lukas G. Low-grade dermal angiosar- ings. Euro J Radiol 1997;24:54–6.
coma of the breast following radiotherapy. Am 104. Fineberg S, Rosen PP. Cutaneous angiosarcoma
Surg 1996;62:668–71. and atypical vascular lesions of the skin and
99. Del Mastro L, Garrone O, Guenzi M, et al. breast after radiation therapy for breast carci-
Angiosarcoma of the residual breast after con- noma. Am J Clin Path 1994;102:757–63.
servative surgery and radiotherapy for primary 105. Rosen PP, Kimel M, Ernsberger D. Mammary
carcinoma. Ann Oncol 1994;5:163–5. angiosarcoma. The prognostic significance of
100. Slotman BJ, van Hattum AH, Meyer S, et al. tumor differentiation. Cancer 1988;62:2145–51.
Angiosarcoma of the breast following conserv- 106. Jardines L. Other cancers in the breast. In: Harris
ing treatment for breast cancer. Eur J Cancer JR, Hellman S, Harris JR, editors. Diseases of
1994;30:416–7. the breast. Philadelphia: Lippincott-Raven
101. Wijnmaalen A, van Ooijen B, van Geel BN, et al. Publishers; 1996.
Angiosarcoma of the breast following lumpec- 107. Naka N, Ohsawa M, Tomita Y, et al. Prognostic
tomy, axillary lymph node dissection, and factors in angiosarcoma: a multivariate analy-
radiotherapy for primary breast cancer: three sis of 55 cases. J Surg Oncol 1996;61:170–6.
case reports and a review of the literature. Int J 108. Donnell RM, Rosen PP, Lieberman PH, et al.
Radiat Oncol Biol Phys 1993;26:135–9. Angiosarcoma and other vascular tumors of the
102. Zuccali R, Merson M, Placucci M, et al. Soft tis- breast. Pathologic analysis as a guide to prog-
sue sarcoma of the breast after conservative nosis. Am J Surg Pathol 1981;5:629–42.
7
Prognostic and Predictive Markers
in Breast Cancer
ANN D. THOR, MD
DAN H. MOORE II, PHD

OVERVIEW malignant processes.8 Needle biopsies (fine-


needle aspiration [FNA] and core-needle
Breast cancer is a highly prevalent and morbid biopsy) have contributed to this process,
disease, afflicting approximately 1 in 9 women although their role in screening, diagnosis, and
in the United States. The death rate from breast subclassification is variable and still in evolu-
cancer in the United States has recently tion.9–13 Marker studies represent another phase
declined for most age groups, although it in this evolution.
remains a major killer with 45,000 deaths annu- Randomized clinical trials (RCTs) have pro-
ally.1 Despite the overall decline, the incidence vided important data that allow comparison of
of ductal carcinoma in situ (DCIS) and stage I chemotherapeutic strategies for adjuvant, neoad-
disease has risen significantly. In parallel, ther- juvant, palliative, or preventive treatments. Tis-
apeutic opportunities (both traditional and sues acquired from patients enrolled on these
alternative) for breast cancer patients have randomized trials have become an important
rapidly expanded. Surgical, radiologic, and resource for correlative studies, allowing analy-
medical oncologic modalities are increasingly sis of tumor markers for prognosis, prediction of
diverse, including both therapeutic and preven- therapeutic benefit, or molecular epidemiologic
tive strategies. In this era of diversity, individu- studies. Knowing which option to choose, in
alization of treatment strategies to maximize what order, and in which combination is the
response and minimize morbidity and mortality challenge that drives prognostic and predictive
has become the goal.2,3 breast cancer marker studies.
As documented in the literature of the This explosion in breast cancer care options
1930s, breast masses identified at surgery and has occurred nearly simultaneously with the
presumed to be cancer could be immediately molecular revolution that occurred in the later
removed by mastectomy on the basis of physi- decades of the 20th century. The identification
cal findings.4 The evolution of biopsy followed and characterization of deoxyribonucleic acid
by histopathology as the diagnostic test preced- (DNA), the introduction of monoclonal anti-
ing definitive surgery began in the second quar- bodies, and molecular biologic methods have
ter of the 20th century5–7 and became firmly given scientists and physicians new tools to
established within decades. Histologic criteria study old problems. As a result, our under-
became further defined with experience, result- standing of breast carcinogenesis and cancer
ing in subclassification of preneoplastic and biology has been greatly modified. We now

113
114 BREAST CANCER

appreciate the immense genetic heterogeneity regularly perform careful histopathologic


of the disease, both within and between indi- analyses as recommended by the Association
vidual patients. Numerous genetic, transcrip- of Directors of Anatomic and Surgical Pathol-
tional, and protein alterations may someday be ogy.23 The clinical application of markers, such
used to diagnose and subclassify breast can- as proliferative rate and oncogenes, has been
cers, augmenting and perhaps replacing more controversial. The American Society of
histopathology as the gold standard (Figure Clinical Oncology (ASCO) and other specialty
7–1). Until the value of these new markers is groups have been reviewing these issues and
determined by careful study, however, use of are expected to publish updated recommenda-
classic markers in breast cancer remains critical tions. Markers for therapeutic prediction,
to the practice of breast cancer care. It is likely including erbB-2 (HER-2/neu), represent the
that, over the next decade, emphasis will be new frontier.
placed on predictive markers and quantitation
of molecular targets to guide novel therapeutics Identification of New Markers
(see Figure 7–1).
Potential new breast cancer markers are gener-
Clinical Use of Usual Breast Cancer ally evaluated using at least one of three types of
Markers in the 1990s studies: (1) early exploratory studies, which gen-
erally seek associations between markers and
Breast cancer markers can be broadly subdi- disease characteristics; (2) studies to determine
vided into (1) clinical or histologic “markers” whether factors provide improved means of
or characteristics (such as tumor size, nodal identifying patients at high or low risk for dis-
metastases), which are useful to define or sub- ease progression or death (using various statisti-
divide the disease; and (2) markers that are cal methods, see below); and (3) studies to
identifiable by specialized testing of the cancer, determine if markers predict benefit from a
sera, nipple aspirate, or other biologic sample. given therapeutic regimen.24 In general, analy-
Both types of markers have clinical rele- ses seek to determine if a marker is specific for
vance14–22 and utility (Table 7–1). the disease or tissue type, whether it relates to
Many physicians currently use breast can- other disease characteristics of interest, or if it
cer markers only for prognosis. Pathologists has prognostic (the ability to portend outcome
independent of therapy) or predictive (the ability
to portend outcome dependent on therapy) value.
Cancer–associated markers (1960s-) Widely accepted methodologic principles
have been published to guide the design, conduct,
Breast cancer–associated markers (1970s-) and analysis of clinical trials.16,20,21 However, few
guidelines have been published for clinical test-
Prognostic markers associated with breast
ing of prognostic or predictive markers. General
cancer (1980s-) guidelines, which are not marker specific, have
been reported and are summarized in Table 7–2.
Predictive markers associated with
For specific breast cancer markers, relevance and
breast cancer (1990s-) applicability may change with time. Recommen-
dations published by governing bodies or profes-
? Molecular targets for therapeutic
sional organizations (such as National Cancer
intervention (2000-) Institute [NCI], Food and Drug Administration
[FDA], American Cancer Society [ACS], ASCO,
Figure 7–1. Evolution of cancer markers. College of American Pathologists [CAP],
Prognostic and Predictive Markers in Breast Cancer 115

American College of Surgeons) are usually Table 7–1. UTILITY OF BREAST CANCER MARKERS

updated every few years. Referral to their rec- • Risk assessment


ommendations on a regular basis is advisable. • Early detection
To exemplify the evolving nature of the • Cancer subclassification
• Differential diagnosis
field, the NCI convened a consensus conference
• Prognosis
in 1985. Only nodal status and tumor size were • Therapeutic prediction
recognized. In 1990, five factors were recom- • Disease monitoring
mended by a similar group of experts: nodal
metastases, tumor size, histologic grade, histo-
logic subtype, and steroid receptor status.3 sentinel node processing (step sections,
Additional markers, including proliferation rate, immunohistochemistry), or the clinical rele-
ploidy, and oncogenes (such as p53 mutation/ vance of micro- or single-cell metastases. Res-
overexpression and erbB-2 amplification/over- olution of these issues will better define the
expression), were recognized as promising role of the pathologist in this procedure. It will
markers. At that time, predictive markers (asso- also determine whether microscopic cellular
ciated with a treatment response) were not rec- metastases is itself a marker of prognosis.
ognized except for steroid receptors.
Tumor Size
Lymph Node Metastases
Tumor size is an independent prognostic
Nodal metastases are a well-recognized risk fac- marker that is particularly important in node-
tor for poor outcome in breast cancer patients negative breast cancer patients.17 Most actuar-
(discussed in detail elsewhere).23,25,26 Nodal pos- ial survival data have emphasized 1 cm and
itivity and the number of nodes with metastases larger tumors, broken into subgroups, for
are associated with an increased risk of disease prognostication. Given the increased inci-
recurrence or progression.27 Nodal metastases dence of tumors ² 1 cm, subclassification of
have often been divided into subgroups for prog- these smaller tumors by size is important as
nostic purposes or randomized trial entry. While well.27–33 Subsetting this group into ² 0.5 cm
subclassification can be afforded using this tech- versus 0.5 to 1 cm has been used by some. Fur-
nique, the biologic value of nodal metastases ther subdivisions of this group are likely as
should be considered as a continuum that corre- more data are available. For these small, node-
lates with outcomes data on survival and recur- negative tumors (T1a), proliferation rate
rence.27 Lymph node metastases are often the appears to be a very important prognostic
strongest independent variable (marker) of out- marker as well.34,35
comes in breast cancer patients.
There are several important issues related to Table 7–2. GENERAL GUIDELINES FOR CLINICAL
lymph node metastases that have not yet been MARKER UTILIZATION
clarified. These include the biologic signifi- • Markers should exhibit significant and independent
cance (and definition) of microscopic nodal predictive value, validated by clinical testing (ie, they should
metastases, nodal metastases detected by only not be implemented solely on the basis of retrospective
data analysis).3
immunohistochemical assay (and the need to • Assays used should be feasible, reproducible, widely
perform such assays), and issues related to lim- available, and subject to quality control.3
ited axillary dissections (sentinel node proce- • Marker analysis should provide data that are readily inter-
pretable by the clinician, with therapeutic implications.3
dures). There is no consensus regarding the use • The measurement of a factor should not consume tumor
of frozen sections or cytology techniques on specimen needed for other tests, particularly careful
sentinel lymph nodes, the optimal protocol for cytologic and/or histologic analysis.20
116 BREAST CANCER

Tumor Grade Determination of steroid receptors on cyto-


logic or surgical preparations of primary breast
Several schemes have been proposed for grading
tumors is standard and almost universally per-
breast carcinomas. These generally include
formed. Such tests should also be obtained on
architectural cellular arrangement, nuclear fea-
presurgical breast cancer samples (cytology or
tures, and other items such as mitotic rate. The
core biopsy) if neoadjuvant chemo- or radiother-
Surveillance, Epidemiology, and End Results
apy will be given. Receptor determination may
(SEER) data from thousands of patients have
also be performed on tumor metastases if the
shown that breast tumor grading, irrespective of
steroid receptor status was not determined on the
the scheme used, has prognostic signifi-
primary tumor or if there is reason to suspect
cance.36,37 The 1990 Consensus Conference rec-
biologic cancer progression (the development of
ommendation—that nuclear grade be evalu-
an estrogen receptor–negative phenotype).
ated—has now been largely superseded by
Immunochemical assays for estrogen and
increasing consensus that a single classification
progesterone receptors (ER, PgR) are most
scheme should be adopted. The Elston scheme,
often used.17 Pathologists evaluate the receptor
which includes nuclear and architectural features
status of the invasive component only. Gener-
and mitotic count, is increasingly used.38,39 Con-
ally, immunopositivity of benign breast epithe-
sensus on a preferred grading scheme is likely
lium adjacent to the cancer is sought as an
to be forthcoming.40–44 Nuclear grading is also
internal positive control for the assay. Many
possible on cytology preparations from touch
pathologists have developed their own defini-
imprint or fine-needle aspiration. Athough not
tion of positivity, which is used in reporting.
equivalent to the combined architectural/cyto-
This may include evaluation of the percentage
logic systems used in surgical pathology, it
of cells staining as well as consideration of
may provide important data on nuclear
stain intensity. An effort should be made to
grade.9–11 Clearly, patients with low-grade
have the local institutional scoring system
(better-differentiated) breast cancers have a
defined and the methodology clearly stated in
better prognosis than those with high-grade
the assay report. Methods and scoring differ-
(poorly differentiated) carcinoma. The reader
ences have been estimated to contribute to dis-
is referred to Chapters 5 and 6 for a detailed
agreements between laboratories in up to 30
discussion on breast pathology.
percent of specimens. Data on ER from archival
cases often used different methodologies for
Steroid Receptor Analysis ER or PgR, and cut-off levels for positivity
Steroid receptors have been routinely deter- were generally determined by the local labora-
mined on surgically resected primary breast tory. Immunohistochemical methods now in
cancers since the late 1970s.45 Receptor- use for ER and PgR can be applied to archival
containing tumors have a better short-term fixed-embedded tissue specimens that are
prognosis, although the magnitude of this dif- decades old, should concern arise about old
ference is relatively small (8 to 10 percent dif- hormone receptor data.
ference in recurrence rate for node-negative The estrogen receptor is a good example of a
patients at 5 years).17,46 Long-term relapse and marker that is both prognostic and predictive.
survival rates between receptor-positive and Patients with tumors that are ER positive are
receptor-negative tumor patients, however, tend more likely to have a better outcome indepen-
to merge.17,44,47 Despite this, steroid receptor dent of treatment. These same patients are also
assays are often used as a marker of probable more likely to respond to tamoxifen therapy (a
sensitivity to tamoxifen or other agents that positive predictive factor).17 Adding to the com-
bind the estrogen receptor. plexity of the issue, recent data suggest that
Prognostic and Predictive Markers in Breast Cancer 117

optimal ER assay cut points may be different to reviewed elsewhere.18 Mitotic counts are now
optimize either prognostic or predictive estima- a composite of a commonly used grading sys-
tions.48 In summary, ER and PgR have both pos- tem.39,40 Separate reporting of the mitotic score,
itive prognostic and predictive values associated in addition to the Elston grade, is supported by
with a more favorable patient outcome. a recent multivariate analysis of outcomes,
which included both statistical models.34 When
Cancer Subtyping other systems of mitotic quantitation are
reported, the mitotic rate should be compared
Tumor subtyping has been recognized to have with the mean or median for other similar
independent prognostic significance in breast breast cancers at the same institution, using
cancer.22 Ten to 30 percent of invasive ductal the same scoring methodology. In general,
carcinomas are of a special type, many of invasive lobular carcinomas have a signifi-
which can be recognized on cytology prepara- cantly lower mitotic rate than infiltrating duc-
tions. Three of these were recognized by the tal carcinomas.34 Scoring of in situ carcinomas
1990 National Institutes of Health (NIH) Con- has not yet been associated with prognostic or
sensus Conference as having a favorable prog- predictive value. Although mitotic counting
nosis—the tubular, colloid, and papillary vari- was first reported nearly a century ago, it
ants.3 Each of these three patterns have remains an important prognostic marker of
cytologic correlates and are often classifiable breast cancer biology.34,49
on FNA (refer to Chapters 5 and 6). With the Other techniques that estimate the percent-
rapid expansion of molecular technologies, age of cells in the S phase include flow cytom-
including the promising array-based formats etry and thymidine (or thymidine analogue)
(which may be used simultaneously to measure uptake and immunohistochemical detection of
hundreds to thousands of genes or proteins proliferation associated antigens, such as
from a breast cancer), subtyping based on gene Ki-67 or proliferating cell nuclear antigen
expression will soon be possible. (PCNA)/cyclin.18,49 Of these, flow cytometry
or the immunohistochemical detection of pro-
Proliferation Rate liferation associated antigens (such as Ki-67)
are the most commonly used. When flow
Cellular proliferation is an important biologic cytometry is performed on a small sample,
characteristic of cancer but has been less macrodissection to increase the tumor/benign
widely accepted as an independent prognostic ratio is advisable, as a false diploid reading
marker. Part of the reticence to adopt it as a may result.
routinely reported marker may be due to the
wide variety of tests for quantitation. The pro- Summary of Commonly Reported
liferation rate is based on the principle of cell Breast Cancer Markers
replication (cell cycle states G1, S, M, G2).
Cells can also be in a resting phase, known as Clearly, no single agent or combination treat-
G0. The mitotic rate, which is generally scored ment is appropriate for all patients.44 Improve-
from routine hematoxylin and eosin–stained ments in outcome, with accurate forecasting of
slides of primary tumor, has been quantitated who will derive the greatest benefit, are impor-
using several systems, including mitotic fig- tant. “Therapeutic modeling” using markers has
ures per 10 high-powered fields, mitotic fig- been evaluated in detail by a multidisciplinary
ures per 1,000 cells, or as a percentage of panel sponsored by the ASCO. Three steps were
invasive cancer cells. The strengths and weak- suggested for the clinical integration of prognos-
nesses of these visual counts have been tic data: (1) analysis of a given patient’s risk of
118 BREAST CANCER

recurrence and survival based on historic out- actions, rapid progression of prognostic mark-
come and multiple prognostic factors; (2) identi- ers from the research arena to translational
fication of various treatment options and their (clinical) applications may be ill advised.
potential therapeutic benefit and risk; and (3) an While centralized banking has met resis-
overall assessment of the expected benefit, risks, tance from some local institutions and patholo-
cost, and other personal factors that might influ- gists, nationally applied standards and safe-
ence treatment decisions and outcome.19 Deter- guards may eventually make these a safer place
mination of prognostic factors in breast cancers, than local archives for long-term storage of
with subsequent use of those data to make ther- slides or blocks. Given the evolving technolo-
apeutic decisions and predict outcome, is com- gies, banks of tumor DNA may someday be
plicated but feasible. The goal of using markers commonplace as well. The emergence of pre-
should be to “contribute to a decision in practice dictive factors and a new format for marker val-
that results in a more favorable clinical outcome idation (the RCT) has greatly affected the labo-
for the patient.”19 Similar guidelines should be ratory development and analysis of new
applicable to predictive factor analysis. markers. Marker/chemotherapy interactions
may also explain, to some extent, discordant
Evolving Markers marker data on distinct patient subsets, obtained
Based on Cancer Biology using retrospective archival tumor tissues.50,51

In the early days of marker development, it was Historic Overview: Identification of


assumed that we could subset patients for coun- Novel Cancer Markers
seling and treatment on the basis of marker and
clinical data that predicted outcome. Unfortu- The first widely studied cancer-associated
nately, clinical and tumor biology heterogeneity marker was carcinoembryonic antigen (CEA).
among breast cancer patients made an exact It was detectable in tumors as well as body flu-
prediction of outcome for an individual patient ids and generated great excitement as a marker
more difficult than anticipated. While prognos- for the diagnosis or monitoring of cancer. It is
tic studies can provide an estimate of risk, expressed by adenocarcinomas (including
translation to a single patient is inherently more breast cancers); however, benign epithelium
complex. Recently reported treatment/marker and inflammatory states produce CEA as well.
interactions have made prognostic marker stud- The challenges in CEA research were making
ies even more difficult. The use of archival reagents and assays that were specific for
tumor banks, comprising heterogeneously CEA, quantitating the protein in human tissues
treated patients is no longer acceptable for ver- and body fluids, determining the associations
ification studies of prognostic markers. with clinical and disease parameters, and bet-
Increasingly, such studies are performed on ter definition of CEA biology and structure.
patient samples derived from cooperative We know now that CEA is a member of a large
group-based randomized trials. This design family of proteins with homology to other
allows testing for treatment/marker interac- cross-reacting antigens.52–55 Many of the early
tions.50,51 Marker/therapy interactions may be studies used nonspecific reagents and generated
confounding. While the relationship may be conflicting results. The CEA has shown lim-
similar (a poor prognosis and marker of poor ited usefulness in breast cancer immunodiag-
response to outcome), it is not always so. A nostics and as a marker of disease progression.
marker may be associated with a negative prog- Because it is not part of a critical molecular
nostic value and a positive predictive value, or pathway, targeting of CEA for therapeutic
vice versa. Given the complexity of such inter- intent has not been useful.
Prognostic and Predictive Markers in Breast Cancer 119

In the early 1980s, scientists used breast Promising Prognostic


cancer cells, cell lines, or derived cellular prod- and Predictive Markers
ucts (eg, membrane extracts) to generate mono-
clonal antibodies against breast cancer associ- A number of biology-associated markers with
ated antigens. Numerous reagents were reported prognostic or predictive value have
discovered, such as the human milk fat globule been reported (Table 7–3). While many of
(HMFG) membranes.56,57 Expression patterns these are of biologic interest, relatively few
of these antigens were variable. None have will likely have independent prognostic value.
demonstrated independent value as prognostic With or without prognostic value, genes or
or predictive tumor markers, although some of their encoded products may be useful as thera-
these reagents have been used to monitor dis- peutic targets.
ease progression.
The development of molecular methods, Growth Factors and Receptors
with subsequent studies of critical cell surface
receptors, oncogenes, and tumor suppressor Breast epithelial cells are, by necessity, respon-
genes, has significantly altered the emphasis of sive to a wide range of growth factors and their
breast cancer marker studies away from anti- receptors including hormones and their cognate
gens identified by chance. In the early 1980s, receptors, ER and PgR, prolactin, insulin, and a
gene sequence data were first used to generate variety of other factors. Estrogen and ER pro-
monoclonal antibody probes against peptides, mote cancer development through an indirect
such as the mutant ras gene.58 This allowed process that includes the promotion of cell
visualization and quantitation of ras gene alter- growth and the activation of estrogen-respon-
ations in situ in human colon and breast adeno- sive genes. Some growth factors/receptors are
carcinomas.57–60 Rapid expansion of these tech- considered oncogenes as well because in the
nologies to other important breast cancer aberrant state, they may cause cancer. Members
markers, including the clinical acceptance of of the type I growth factor receptor family (epi-
immunohistochemical assays to analyze estro- dermal growth factor receptor [EGFR], erbB-2
gen and progesterone receptors, firmly estab- [HER-2/neu], erbB-3, erbB-4) have prognostic,
lished this approach. predictive, and therapeutic target value. For the
Some have suggested a “growing backlash purpose of this chapter, only the highlights of
of negative sentiment concerning breast cancer the voluminous literature will be cited.
prognostic factors in the oncology community
today.”61 However, with a shift in emphasis erbB-2
from prognosis to prediction and an increased
Over a decade has passed since the HER-2/neu
use of targeted molecular therapeutics, the
(erbB-2) gene was first identified in chemically
field of markers in breast cancer care has
solidified. The ability of scientists to insert
(transfect) genes into cells and the develop- Table 7–3. IMPORTANT MARKER GROUPS
ment of knock-in and knock-out transgenic IN BREAST CANCER

mice now allows hypothesis testing to deter- • Oncogenes


mine the specific biologic role(s) of specific • Tumor suppressor genes
genes or signal transduction pathways. These • Programmed cell death associated
• Angiogenesis associated
tools have revolutionized our understanding of • Growth factors and their receptors
cancer biology and, in the process, have identi- • Adhesion molecules
fied entirely new targets (markers) for molecu- • Proteases/Protease inhibitors
lar therapeutics. • Metastasis associated
120 BREAST CANCER

induced glial tumors in rats.62,63 The human decrease the sensitivity or specificity of the test
equivalent of the proto-oncogenic neu, known (no matter what procedure is used), resulting in
as erbB-2 is located on chromosome 17.64 The false negatives or false positives. For this reason,
erbB-2 gene encodes a transmembrane protein, calibration of erbB-2 assays, using controls with
p185, with structural homology to EGFR. This various levels of gene amplification, are neces-
structural homology is one of the features link- sary and can be purchased commercially. These
ing these two genes as members of the type 1 should be fixed embedded pellets of cell lines,
receptor tyrosine kinase (RTK) gene superfam- with and without gene amplification, rather than
ily, which also contains the less well studied human tumors that have been positive before.
members erbB-3 and erbB-4.65,66 Although “Kits” that support erbB-2 testing from
encoded by individual genes, these members reagents to recommended scoring systems have
are highly homologous. Each possesses an just been released. It is possible that these sys-
extracellular ligand-binding domain, and li- tems will be superior to the currently used “in-
gands that bind to all but erbB-2 have been house” technologies, although there is little
identified. These four members can form data yet to support that conclusion.
homo- and heterodimers, with 10 possible While the pathologist may use a special
dimers. The biologic differences in a prognostic scoring system (such as the 0, 1+, 2+, or 3+
or predictive sense are not yet known for these system for the Dako HercepTest®), providing
different configurations. an estimate of the percentage of erbB-2–posi-
The erbB-2 overexpression/amplification is a tive invasive cancer cells will allow greater
complex process,67–69 which occurs in approxi- comparison with other laboratories. While
mately one-third of invasive and up to two-thirds reagent issues are beyond the scope of this
of in situ carcinomas.70–77 The erbB-2 alterations chapter, the methodology for scoring deserves
have been associated with a poor prognosis in brief mention. In general, (1) membranous
breast cancer patients,17,50,78–89 although it is usu- reactivity only should be considered positive;
ally less predictive of outcome than lymph node (2) the invasive component of a tumor only (not
metastases. A resurgence of interest in erbB-2 as in situ disease) should be scored; (3) erbB-2
a breast cancer marker has recently occurred staining should not be observed in adjacent
because erbB-2 alterations may predict chemore- stroma or inflammatory cells, nor should benign
sponsiveness81,83,85 and the FDA has recently epithelium show strong membranous reactivity;
approved the drug Herceptin® (Trastuzumab, (4) reporting should include an approximate
Genentech, Inc.), which targets erbB-2. estimate of the percentage of immunopositive
Cancers without erbB-2 alterations have two invasive cancer cells; (5) positive and negative
copies of the gene (unless deletions have controls should be included in each assay; and
occurred) and encode low levels of protein. All (6) the method and primary reagent used by the
normal cells and the majority of breast cancer laboratory should be reported with the assay
cells bear two copies of the erbB-2 gene and pro- result. While some recommend a reporting/
duce low levels of the encoded protein p185. scoring of staining intensity, few have com-
Assays to evaluate erbB-2 generally measure pared that data with outcome. There is little
either gene copy number or protein expression. data that intensity, by itself, has prognostic or
Abnormal erbB-2 can be defined as protein predictive value.81 Cells with concentric mem-
expression at levels above normal cells or gene branous staining only are recommended for
copy number > 2. Assays, therefore, need to be scoring by some (Dako HercepTest®). This has
precise and have the discriminatory power to not been proven to be superior to focal mem-
separate abnormal from normal. Variance in brane staining for prognostic or predictive pur-
assay procedures or reagents may increase or poses. However, in general, intensity and con-
Prognostic and Predictive Markers in Breast Cancer 121

centric staining are associated with higher lev- CAF have now been reported by others as
els of gene amplification. Discrete cut points well.85,86 Data from older cooperative group tri-
used for data analysis in some studies are sub- als suggest a relative resistance of erbB-
optimal; the biologic relevance of erbB-2 likely 2–altered breast cancers to methotrexate-based
represents a continuum. regimens.50,87,88 Taxol resistance has also been
Two commercial fluorescence in situ associated with erbB-2 overexpression/ amplifi-
hybridization (FISH) assays for erbB-2 have cation, although this issue remains controver-
recently been approved by the FDA for progno- sial.80 Some reports have also suggested resis-
sis (not qualification of patients for Herceptin). tance of patients with ER+ tumor to tamoxifen
The majority of studies have shown compara- if erbB-2 and/or EGFR are overexpressed,89–93
bility between immunohistochemical data and although the interaction has not been demon-
FISH,77 although some have reported that FISH strated by all.94–96
methods are superior.84 The FISH methodology
is generally similar to immunohistochemistry, erbB-2 as a Therapeutic Target. In 1998 the FDA
although reagents are more expensive, requir- announced approval of Herceptin® (Trastuzumab,
ing special microscopic equipment and greater Genentech, Inc.) for the treatment of metastatic
pathologist time for scoring. Differences breast cancer. This approval occurred in five
between kits include probe labeling (direct via months, a nearly unparalleled “fast-track.” Her-
indirect) and the use of a centromeric probe for ceptin is thought to offer a less toxic approach
chromosome 17 in addition to the erbB-2 for treating breast cancer, as it directly targets
probe. Intratumor heterogeneity of erbB-2 gene erbB-2 associated growth promotion.97 Her-
copy number and chromosome 17 centromeric ceptin is a genetically engineered (humanized)
copy number are common.79 Scoring systems monoclonal antibody which binds erbB-2.
that reflect this heterogeneity have not been Early studies of breast cancer patients with
widely applied and, therefore, the biologic rele- advanced disease have shown that as a single
vance is unknown. agent, or in combination with other chemo-
therapy, Herceptin significantly improved out-
erbB-2 as a Predictive Marker in Breast Cancers. come for some patients.98,99 It is somewhat
The erbB-2 data from nearly 1,000 stage II unclear how patients should be selected for
breast cancers derived from a randomized three- treatment with this agent. Most believe that
arm trial (Cancer and Leukemia Group B, breast cancers without erbB-2 alterations will
CALGB trial 8541) of cytoxan, adriamycin, and not be responsive to Herceptin, although there
5-fluorouracil (CAF) have suggested interac- has not been a clinical trial to test this hypothe-
tions between erbB-2 and chemotherapy (dose sis. In completed trials, patients with the greater
of CAF81,83,85). This conclusion is supported by number of cells with concentric erbB-2
both molecular and immunohistochemical immunostaining had a greater response rate.
erbB-2 data. Patients whose tumors had ampli-
fied or overexpressed erbB-2, treated with dose Epidermal Growth Factor Receptor
intensive CAF, had a significantly better sur-
vival than patients without erbB-2 abnormali- The EGFR gene is amplified with overexpres-
ties assigned to the same treatment arm.81,83 In sion or is overexpressed in many breast can-
this study, stage II breast cancer patients whose cers. This receptor allows breast cancer cells to
tumors had alterations of both erbB-2 and p53 bind a variety of autocrine or paracrine growth
treated with dose-intensive CAF had the most factors (including epidermal growth factor
favorable outcome (90% 10-year survival).81 [EGF], transforming growth factor-alpha
Interactions between erbB-2 and response to [TGF-a]).100–103 The EGFR is upregulated by
122 BREAST CANCER

estrogens via direct binding to the promotor The p53 gene is relatively large, and muta-
region of the gene.104–106 It is also constitutively tions have been reported in both introns and
activated by amplified erbB-2.107 Binding of exons. In breast cancers, mutations appear to
ligands such as EGF to EGFR triggers rapid cluster in exons 5 to 9. Given the molecular com-
tyrosine phosphorylation of the erbB-2 pro- plexity of this large gene, studies of mutations
tein108 as well as other downstream sub- based on genetic sequencing have been limited.
strates.109,110 Newer technologies are being developed for
The EGFR is overexpressed by over one- sequencing in high-throughput formats. Given
third of infiltrating ductal carcinomas, is uni- the size of the gene and its many functions, the
versally expressed by medullary carcinomas, location and type of genetic abnormality may be
and is generally not detected in lobular or col- important to determine its clinical value.
loid carcinomas.111 Overexpression of EGFR Patients with germline p53 mutations
has been reported in male breast cancers, (LiFraumeni syndrome) have an increased inci-
although infrequently.112 Overexpression of dence of breast cancers.132 Recent evidence
EGFR has been associated with increased suggests a relationship between BRCA-1 and
metastatic potential and a worse prognosis in p53 in hereditary breast cancer such that p53
both node-positive and node-negative breast acts as a cancer cofactor in these patients.133
cancer patients.113–118 The EGFR may interact Most p53 abnormalities identified in breast
with erbB-2 to confer relative resistance to cancers, however, occur as spontaneous,
tamoxifen in ER-positive patients,89–93,115,116 somatic events. The p53 abnormalities have
although, as stated above, this issue is some- been reported in invasive and in situ carcino-
what controversial. Co-overexpression of mas as well as in rare precursor lesions.
erbB-2 and EGFR is seen in approximately
one-fifth of breast cancers. In summary, EGFR STATISTICAL ISSUES
should be considered prognostic and possibly IN CANCER MARKER STUDIES
predictive on the basis of the data that are cur-
rently available. There are many statistical tools for studying the
relationships between patient characteristics
p53 (such as age at diagnosis and genetic makeup),
tumor parameters (eg, tumor size and grade),
Nearly one-third of breast cancers have muta- adjuvant therapy (primarily radiation and
tions of the tumor suppressor gene p53. This has chemotherapy), markers, and length of sur-
been associated with histologic and clinical vival. This subsection will highlight key issues
aggressiveness.86,119–123 Mutations often result in that are important in marker analyses.
overexpression of the encoded protein as a result
of a prolonged half-life and protein accumula- Randomized Clinical Trials
tion. Fortunately, this effect allows immunohis-
tochemical detection of p53 as a surrogate for An RCT is the most rigorous way to evaluate
mutational analyses.81,120,124–128 This should be treatment efficacy, compare different treat-
considered a screening method, as some muta- ments, or test the predictive value of a given
tions are clearly not detected. The p53 gene as a marker. The FDA requires proof of efficacy
breast cancer marker appears more prognostic in from one or more RCTs for approval of any
node-negative as compared with node-positive new treatment for cancer. The key to an RCT is
breast cancer patients. In addition to prognostic blinded randomization of patients into treat-
value, p53 data may help identify patients likely ment arms. Randomization minimizes the like-
to respond to chemo- or radiotherapy.129–131 lihood that differences in outcome between two
Prognostic and Predictive Markers in Breast Cancer 123

treatment groups are due to factors other than (K-M) survival curves. A description of how to
the difference in treatment. Formal statistical calculate points for drawing this curve can be
methods for ensuring that patients are assigned found in many medical-statistical books.134
at random to a treatment group must be applied. Comprehensive statistical software can calcu-
The importance of this step cannot be overem- late these curves with precision. The K-M
phasized. Promising results from small studies curve is nonparametric, that is, there are no
without randomization are often discovered to parameters to be estimated to determine its
be due to baseline differences between those shape. It is highly flexible and can be used to fit
who received the new treatment and those who any set of survival data consisting of time and a
did not, rather than the differences in treatment. censoring indicator. The basic idea underlying
Failures in randomization can result in statisti- the K-M curve is that it starts with 100 percent
cal nightmares in the interpretation of marker survival at time 0 (usually the left side of the
data. The study by Thor34 is a case in point. curve along the Y axis). A decrement is made at
An RCT usually involves patients from mul- each event, and the size of the decrement is
tiple treatment centers. Once such trials have equal to the number of patients who experience
been reviewed and approved, patients are that event at that time, divided by the number of
assigned at random to treatment groups during patients who were still alive and being followed
the enrollment period. Enrollments cease when immediately prior to the event time. A little
trials are closed. Patients “on protocol” are then known but useful feature of the curve is that the
followed up for a predetermined length of time, number of patients at the end of the study can
and their outcome is recorded. The usual out- be estimated by examining the horizontal
comes of interest are disease recurrence (other- decrement at the last event. The number
wise known as disease-free survival [DFS]), remaining is equal to the reciprocal of this
disease-specific death (DSS), and death from decrement. For example, if the survival curve
other causes. Recurrence or death is often falls by 0.1 at the time of the last event, there
referred to as “events” in statistical jargon. were, with high probability, 10 patients fol-
Depending on the eligibility criteria, many lowed up for this length of time.134
patients will not have had a recurrence and/or
will still be alive at the end of the study so that Comparisons of Survival
the survival time for those patients is not
known. These patients are generally removed The survival experience of any number of
from analyses of outcomes, a process called groups can be compared visually by plotting K-
“censoring.” The number of patients who are M survival curves for each group. There are
censored or have events determines the statisti- many statistical tests for comparing survival
cal power of a given study. curves; the most widely used is the log-rank
test. It summarizes the survival experience of
Kaplan-Meier Survival Curves two (or more) groups by forming a 2 ´ 2 con-
tingency table each time an event occurs. This
Survival experience is usually quantitated as leads to a series of contingency tables and dif-
the length of time patients are followed up and ferent weighting schemes for combining the
whether or not outcomes of interest, or events, results. The log-rank test weights the chi-square
occurred. When an event has occurred, the statistics from these tables equally. Harrington
length of time is recorded as length of follow- and Fleming proposed a weighting scheme
up to the event time. Time after the event is dis- under greater control by the user. This allows
regarded. The simplest way to summarize data comparisons of survival curves at specific time
of this type is to plot them as Kaplan-Meier points, such as early or late. These might be
124 BREAST CANCER

appropriate to determine if the effect of a new though the shape of the survival function is not
treatment or marker is most pronounced early specified, the relative hazards associated with
on. Similarly, later events may be analyzed by different factors require estimation of parame-
changing the Harrington-Fleming parameter.135 ters. In using this model to assess the impor-
tance of factors on survival, it is important to
Proportional Hazard Statistical Models test the proportional hazards assumption. For
example, when the number of positive nodes is
The K-M survival curves can be used to study used, the model assumes that the relative hazard
the effect of a continuous variable (eg, age), but of having 1 positive node compared with none
it is necessary to define one or more cut points is the same as that of having 20 nodes com-
to subdivide patients into strata. A K-M curve pared with 19. Some software programs have
can be calculated for each stratum, and the the capability of testing these assumptions sta-
strata can be compared using the log-rank or tistically. However, it is important for the user
other tests. However, results of such compar- to understand the meaning of the assumptions
isons are usually highly dependent on the cut behind the proportional hazards model and to
points; therefore, a researcher who finds con- make adjustments when possible to make the
flicting results may not know how to report his assumptions more relevant. For example, the
findings. Cut points are also commonly used to logarithm of the number of positive nodes (with
maximize the p values, a practice which should 1 added before taking the logarithm to avoid
be discouraged. taking the log of 0) is often a more realistic way
The most widely used statistical tool for to model the effects of positive nodes than
studying the effects of continuous variables on using the actual number. A similar transforma-
survival or recurrence is the proportional haz- tion may be appropriate for tumor size. This
ards model. This method was formulated by kind of variable transformation is often applied
Cox;136 hence, it is also known as the Cox model. in marker analyses, although it is usually
It is based on an assumption that the hazard described in detail only in figure legends or the
(defined as the instantaneous risk of experienc- statistical methods section.
ing an event at any given time) for a patient with
a risk factor, say age, at level x is proportional to Univariate and Multivariate Analyses
the hazard for another patient at level x' and that
the ratio of these hazards is constant over the fol- When establishing the usefulness of a breast
low-up time. A simple equation describes the cancer marker, it is important to perform both
ratio of these hazards for any values of x and x': univariate and multivariate analyses. Univariate
analysis determines whether the factor predicts
Ratio of hazards = exp[ _ (x – x')] the end point. It does not consider the influence
of other factors and, therefore, can be mislead-
where _ is a parameter to be estimated from the ing. In evaluating novel markers, it is often
data. This model is readily extended to many unknown whether the marker is a cause or a
variables. result of the cancerous process. Thus, associa-
Comprehensive statistical software pro- tions between the presence or absence of a
grams include routines for estimating the para- marker with a better or worse survival does little
meters of the Cox proportional hazards model to advance our understanding of the underlying
and for testing their statistical significance. biology or improve predictability of outcome.
This method is widely used for studying the Probability values (p-values) are poor indicators
relation between multiple factors and survival. of relative statistical ranking of the importance
It is said to be semiparametric because even of multiple factors. For example, a factor that is
Prognostic and Predictive Markers in Breast Cancer 125

“significant” at p = .001 may or may not be a In deciding how many patients should be
better predictor of outcome than one with a p = included in a study, it is important to realize
.02. Each factor may interact with others in ways that statistical power is dictated by the number
that are not assessed by the size of the p value. of events (ie, recurrences or deaths), not the
The only robust way to evaluate the performance number of subjects at the start of follow-up.
of different subsets of factors is to perform ran- When multiple factors are under study, a rule of
domization tests of their performance.137 thumb is that 10 events are required for every
A much better understanding of the rela- factor studied. In node-negative disease where
tionship of the marker can be acquired when it 5-year survival is often above 90 percent, this
is tested in the “presence” of other well-estab- means that 100 patients per factor under study
lished factors (ie, multivariate analyses). For will be required. Prognostic marker studies
example, a factor that is highly correlated with should include both univariate and multivariate
the number of positive nodes may provide no analyses of outcomes. Prognostic marker stud-
additional independent prognostic/predictive ies generally exclude consideration of treat-
information when the latter factor is added to ment/marker interactions. If such interactions
the predictive equation. The only way to find exist, they will go unrecognized when the study
this out is to perform a multivariate analysis, population is treated heterogeneously. To iden-
where all factors are considered together in a tify such interactions, study of tumors derived
statistical model for predicting outcome. The from patients entered in RCTs is necessary.
Cox multivariate model described above is a
widely used and useful tool for determining the REFERENCES
statistical utility of a new factor in the presence
of established factors. When this analysis is 1. Parker SL, Tong T, Bolden S, Wingo PA. Cancer
statistics, 1996. CA Cancer J Clin 1996;46(1):
performed, it is important to include the most 5–27.
powerful usual markers in the model. Failure to 2. Ernster VL, Barclay J, Kerlikowske K, et al. Inci-
do so may cause a new marker to appear impor- dence of and treatment for ductal carcinoma in
tant, when in reality it lacks independent value. situ of the breast. JAMA 1996;275(12):913–8.
3. NIH Consensus Conference. Treatment of early
stage breast cancer. JAMA 1991;265:391–5.
SUMMARY 4. Tod MC, Dawson EK. The diagnosis and treat-
ment of doubtful mammary tumours. Lancet
It is important for clinicians and translational 1934;II:1041–5.
scientists to work closely with statisticians for 5. Muir R. The pathogenesis of Paget’s disease of the
both marker study design and analysis. Statisti- nipple and associated lesions. Br J Surg 1934;
22:728–37.
cians who regularly participate in breast cancer
6. Foote FW Jr, Stewart F. Lobular carcinoma in situ.
marker or outcome studies often have the great- A rare form of mammary cancer. Am J Pathol
est insights; they know which variables should 1941;7:491–6.
be considered in the analysis and what patient 7. Ewing J. Epithelial and other tumours of the
populations are best suited for hypothesis test- breast. Neoplastic disease. Philadelphia: W. B.
Saunders; 1940.
ing. Survival analysis requires “expert knowl-
8. Page DL, Anderson TJ. How should we categorize
edge” as many variables can affect study out- breast cancer? Breast 1993;2:217–9.
comes.138 One of the most common errors that is 9. Kreuzer G, Boquoi E. Aspiration biopsy cytology,
made in breast marker studies is the use of mammography and clinical exploration: a
diverse patients with short-term follow-up. Well- modern set up in diagnosis of tumors of the
breast. Acta Cytol 1976;20:319–23.
established factors associated with survival, such
10. Kline TS, Joshi LP, Neal HS. Fine-needle aspiration
as tumor stage and patient age, must be consid- of the breast: diagnoses and pitfalls. A review of
ered in trial design and statistical analyses. 3545 cases. Cancer 1979;44:1458–64.
126 BREAST CANCER

11. Dabbs DJ. Role of nuclear grading of breast car- tumor size, lymph node status and survival in
cinomas in fine needle aspiration specimens. 24,740 breast cancer cases. Cancer 1989;63:
Acta Cytol 1993;37(3):361–6. 181–7.
12. Abiati A, Consensus Committee. The uniform 26. Cazin JL, Gosselin P, Boniface B, et al. Compara-
approach to breast fine needle aspiration biopsy: tive values of several tumour markers: example
a synopsis. Acta Cytol 1996;40:1120–6. of untreated breast carcinoma. Br J Cancer
13. Bassett L, Winchester DP, Caplan RB, et al. 1990;62(6):1031–3.
Stereotatic core-needle biopsy of the breast: a 27. Harris JR, Morrow M, Norton L. Malignant
report of the Joint Task Force of the American tumors of the breast. In: DeVita VT, Hellman
College of Radiology, American College of S, Rosenberg SA, editors. Cancer: principles
Surgeons, and College of American Patholo- and practice of oncology. 5th ed. New York:
gists. CA Cancer J Clin 1997;47(3):171–90. Lippincott-Raven Publishers; 1997.
14. Aziz K. Tumour markers: current status and future 28. Mansour EG, Ravdin PM, Dressler L. Prognostic
applications. Scand J Clin Lab Invest 1995; factors in early breast carcinoma. Cancer
Suppl 221:153–5. 1994;74:381–400.
15. Miller WR, Ellis IO, Sainsbury JR, Dixon JM. 29. Hermanek P, Sovin LH, Fleming ID. What do we
ABC of breast diseases. Prognostic factors. need beyond TNM? Cancer 1996;77(5):815–7.
BMJ 1994;309:1573–6. 30. Goldhirsch A, Wood WC, Senn HJ, et al. Interna-
16. Levine MN, Browman GP, Gent M, et al. When is tional Consensus Panel on the Treatment of
a prognostic factor useful? A guide for the per- Primary Breast Cancer. Eur J Cancer 1995;
plexed. J Clin Oncol 1991;9:348–56. 31A:1754–9.
17. Clark GM. Prognostic and predictive factors. In: 31. Bast RC, Desch CE, Hayes DF, et al. Update of
Harris JR, Hellman S, Lippman M, Morrow M, recommendations for the use of tumor markers
editors. Diseases of the breast. Philadelphia: J. in breast and colorectal cancer. J Clin Oncol
B. Lippincott-Raven; 1996. 1997;16:793–5.
18. Thor AD, Edgerton SM. Cellular markers of pro- 32. Nemoto T, Vana J, Bedwani RN, et al. Management
liferation and oncogenes. In: Colvin RB, Bhan and survival of female breast cancer: results of a
AD, McCluskey RT, editors. Diagnostic national survey by the American College of
immunopathology. 2nd ed. New York: Raven Surgeons. Cancer 1980;45(12):2917–24.
Press, Ltd; 1995. 33. Koscielny S, Tubiana M, Le MG, et al. Breast can-
19. Hayes DF, Bast RC, Desch CE, et al. Tumor cer: relationship between the size of the pri-
marker utility grading system: a framework to mary tumour and the probability of metastatic
evaluate clinical utility of tumor markers. J dissemination. Br J Cancer 1984;49(6):709–15.
Natl Cancer Inst 1996;88(20):1456–66. 34. Thor AD, Liu S, Moore DH, Edgerton SM. Com-
20. Gasparini G, Pozza F, Harris AL. Evaluating the parison of mitotic index, in vitro bromo-
potential usefulness of new prognostic and pre- deoxyuridine labeling, and MIB-1 assays to
dictive indicators in node-negative breast can- quantitate proliferation in breast cancers. J Clin
cer patients. J Natl Cancer Inst 1993;85(15): Oncol 1999;17(2):470–7.
1206–19. 35. Rosen PP, Groshen S, Saigo PE, et al. A long-term
21. McGuire WL. Breast cancer prognostic factors: follow up study of survival in stage I
evaluation guidelines. J Natl Cancer Inst 1991; (T1NoMo) and stage II (T,N, M0) breast carci-
83(3):154–5. noma. J Clin Oncol 1989;7:355–66.
22. Page DL. Prognosis and breast cancer. Recogni- 36. Henson DE. The histologic grading of neoplasms.
tion of lethal and favorable prognostic sub- Arch Pathol Lab Med 1988;112:1091–6.
types. Am J Surg Pathol 1991;15(4):334–49. 37. Henson DE, Ries L, Freedman LS, Carriaga M.
23. Connolly JL, Fechner RE, Kempson RL, et al. Relationship among outcome, stage of disease,
Recommendations for the reporting of breast and histologic grade for 22,616 cases of breast
carcinoma. Association of Directors of cancer. The basis for a prognostic index. Can-
Anatomic and Surgical Pathology. Hum Pathol cer 1991;68:2142–9.
1996;27(3):220–4. 38. Elston CW, Ellis IO. Method for grading breast
24. Simon R, Altman DG. Statistical aspects of prog- cancer. J Clin Pathol 1993;46:189–90.
nostic factor studies in oncology. Br J Cancer 39. Elston CW, Ellis IO. Pathological prognostic fac-
1994;69(6):979–85. tors in breast cancer: I. The value of histologi-
25. Carter CL, Allen C, Henson DE. Relation of cal grade in breast cancer: experience from a
Prognostic and Predictive Markers in Breast Cancer 127

large study with long-term follow-up. Histo- anti-CEA sera, used for immunohistology. The
pathology 1991;19:403–10. need for tissue controls and absorptions. Am J
40. Robinson IA, McKee G, Kissin MW. Typing and Clin Pathol 1983;79:25–31.
grading breast carcinoma on fine-needle aspi- 53. Nap M, Keuning H, Burtin P, et al. CEA and NCA
ration: is this clinically useful information? in benign and malignant breast tumors. Am J
Diagn Cytopathol 1995;13(3):260–5. Clin Pathol 1984;82:526–34.
41. Cajulis RS, Hessel RG, Hwang S, et al. Simpli- 54. Thompson J, Zimmermann W. The carcinoembry-
fied nuclear grading of fine-needle aspirates of onic antigen gene family: structure, expression
breast carcinoma: concordance with corre- and evolution [review]. Tumor Biol 1988;9:
sponding histologic nuclear grading and flow 63–83.
cytometric data. Diagn Cytopathol 1994;11(2): 55. Thor A, Muraro R, Gorstein F, et al. Adjunct to the
124–30. diagnostic distinction between adenocarcino-
42. Howell LP, Gandour-Edwards R, O’Sullivan D. mas of the ovary and the colon utilizing a
Application of the Scarff-Bloom-Richardson monoclonal antibody (COL 4) with restricted
tumor grading system to fine-needle aspirates carcinoembryonic antigen reactivity. Cancer
of the breast. Am J Clin Pathol 1994;101(3): Res 1987;47:505–12.
262–5. 56. Foster CS, Edwards PAW, Dinsdale EA, Neville
43. Dabbs DJ, Silverman JF. Prognostic factors from AM. Monoclonal antibodies to the human mam-
the fine-needle aspirate: breast carcinoma mary gland. Virchows Arch 1982;394:279–93.
nuclear grade. Diagn Cytopathol 1994;10(3): 57. Burchell J, Gendler S, Taylor-Papadimitriou J, et
203–8. al. Development and characterization of breast
44. Hirshaut Y, Pressman P. Breast cancer: the com- cancer reactive monoclonal antibodies directed
plete guide. New York: Bantam; 1996. to the core protein of the human milk mucin.
45. Jordan VC. Tamoxifen: a guide for clinicians and Cancer Res 1987;47:5476–82.
patients. Huntington NY: PRR Publishers; 1996. 58. Thor A, Horan Hand P, Wunderlich D, et al.
46. Clark GM, McGuire WL. Steroid receptors and Monoclonal antibodies define differential ras
gene expression in malignant and benign
other prognostic factors in primary breast can-
colonic diseases. Nature 1984;311:562–5.
cer. Semin Oncol 1988;15:20.
59. Horan Hand P, Thor A, Wunderlich D, et al.
47. Early Breast Cancer Trialists’ Collaborative
Monoclonal antibodies of predefined speci-
Group. I. Systemic treatment of early breast
ficity detect activated ras gene expression in
cancer by hormonal, cytotoxic, or immune ther-
human mammary and colon carcinomas. Proc
apy: 133 randomized trials involving 31,000
Natl Acad Sci U S A 1984;81:5227–31.
recurrences and 24,000 deaths among 75,000
60. Thor A, Ohuchi N, Horan Hand P, et al. ras gene
women. Lancet 1992;339:1–15, 71–85.
alterations and enhanced levels of ras p21
48. Elias JM, Masood S. Estrogen receptor assay: are
expression in a spectrum of benign and malig-
we all doing it the same way? A survey. J His- nant human mammary tissues. Lab Invest
totechnol 1995;18:95–6. 1986;55(6):603–15.
49. Thor AD, Yandell DW. Molecular pathology of the 61. Osborne CK. Prognostic factors for breast cancer:
breast. In: Harris, JR, Hellman S, Lippman M, have they met their promise? J Clin Oncol
Morrow M, editors. Diseases of the breast. 1992;10(5):679–82.
Philadelphia: J. B. Lippincott-Raven; 1996. 62. Bargmann CI, Hung MC, Weinberg RA. Multiple
50. Ravdin PM, Chamness GC. The c-erbB-2 proto- independent activations of the neu oncogene by
oncogene as a prognostic and predictive a point mutation altering the transmembrane
marker in breast cancer: a paradigm for the domain of p185. Cell 1986;45(5):649–57.
development of other macromolecular mark- 63. Schecter AL, Stern DF, Vaidyanathan L, et al. The
ers—a review. Gene 1995;159(1):19–27. neu oncogene: an erbB related gene encoding a
51. Berry DA, Thor A, Cirrincione C, et al. Scientific 185,000 Mr tumour antigen. Nature 1984;312:
inference and predictions: multiplicities and 513–6.
convincing stories: a case study in breast can- 64. Coussens L, Yang-Feng TL, Liao YC, et al. Tyro-
cer therapy. In: Bernardo JM, Berger JO, sine kinase receptor with extensive homology
Dawid AP, et al., editors. Bayesian statistics. to EGF receptor shares chromosomal location
Vol 5. Oxford University Press; 1996. with neu oncogene. Science 1985;230:1132–9.
52. Nap M, Klaski A, Hoor R, Fleuren G-J. Cross- 65. Kraus MH, Issuing T, Miki N, et al. Isolation and
reactivity with normal antigens in commercial characterization of erbB-3, a third member of
128 BREAST CANCER

the erbB/epidermal growth factor receptor 79. Szollosi J, Balazs M, Feuerstein BG, et al. erbB-2
family: evidence for overexpression in a set of (HER2/neu) gene copy number, p185HER-2
human mammary tumors. Proc Natl Acad Sci overexpression and intratumor heterogeneity in
U S A 1989;86(23):9193–7. human breast cancer. Cancer Res 1995;55(22):
66. Plowman GD, Culouscou J-M, Whitney GS, et al. 5400–7.
Ligand-specific activation of HER4/p180 80. Press MF, Bernstein L, Thomas PA, et al. HER-
erbB-4, a fourth member of the epidermal 2/neu gene amplification characterized by flu-
growth factor receptor family. Proc Natl Acad orescence in situ hybridization: poor prognosis
Sci U S A 1993;90:1746–50. in node-negative breast carcinomas. J Clin
67. Nandi S, Guzman RC, Yang J. Hormones and Oncol 1997;15(8):2894–904.
mammary carcinogenesis in mice, rats, and 81. Thor AD, Berry DA, Budman DR, et al. erbB-2,
humans: a unifying hypothesis. Proc Natl Acad p53 and efficacy of adjuvant therapy in lymph
Sci U S A 1995;92(9):3650–7. node-positive breast cancer. J Natl Cancer Inst
68. Ethier SP. Growth factor synthesis and human 1998;90:1346–60.
breast cancer progression. J Natl Cancer Inst 82. Thor AD, Budman DR, Berry DA, et al. Selecting
1995;87(13):964–73. patients for higher dose adjuvant CAF: c-erbB-2,
69. Stern DF. Biology of ErbB2/HER2/Neu. [Sub- p53, dose and dose intensity in stage II, node
mitted] positive breast cancer [abstract]. Proc ASCO
70. King CR, Kraus MH, Aaronson SA. Amplification 1997;16:128a.
of a novel v-erb-related gene in a human mam- 83. Muss HB, Thor AD, Berry DA, et al. C-erbB-2
mary carcinoma. Science 1985;229:974–6. expression and response to adjuvant therapy in
71. Kraus MH, Popescu NC, Amsbaugh SC, King women with node-positive early breast cancer.
CR. Overexpression of the EGF receptor- N Engl J Med 1994;330:1260–6.
related proto-oncogene erbB-2 in human mam- 84. Pauletti G, Godolphin W, Press MF, Slamon DJ.
mary tumor cell lines by different molecular
Detection and quantitation of HER-2/neu gene
mechanisms. EMBO J 1987;6:605–10.
amplification in human breast cancer archival
72. Slamon DJ, Clark GM, Wong S, et al. Human
material using fluorescence in situ hybridiza-
breast cancer: correlation of relapse and sur-
tion. Oncogene 1996;13(1):63–72.
vival with amplification of the HER-2/neu
85. Paik S, Bryant J, Park C, et al. erbB-2 and
oncogene. Science 1987;235:177–82.
response to doxorubicin in patients with axil-
73. Slamon DJ, Godolphin W, Jones LA, et al. Studies of
lary lymph node-positive, hormone receptor-
the HER-2/neu proto-oncogene in human breast
negative breast cancer. J Natl Cancer Inst
and ovarian cancer. Science 1989;244:707–12.
74. Thor AD, Schwartz LH, Koerner FC, et al. Analy- 1998;90(18):1361–70.
sis of c-erbB-2 expression in breast carcinomas 86. Clark GM. Should selection of adjuvant
with clinical follow-up. Cancer Res 1989;49: chemotherapy for patients with breast cancer
7147–52. be based on erbB-2 status? J Natl Cancer Inst
75. Paik S, Hazan R, Fisher ER, et al. Pathologic find- 1998;90(18):1320–1.
ings from the National Surgical Adjuvant Breast 87. Gusterson BA, Gelber RD, Goldhirsch A, et al.
and Bowel Project: prognostic significance of Prognostic importance of c-erbB-2 expression in
erbB-2 protein overexpression in primary breast breast cancer. J Clin Oncol 1992;10(7):1049–56.
cancer. J Clin Oncol 1990;8:103–12. 88. Allred DC, Clark GM, Tandon AK, et al. HER-
76. Liu E, Thor A, He M, et al. The HER2 (c-erbB-2) 2/neu in node-negative breast cancer: prognos-
oncogene is frequently amplified in in situ carci- tic significance of overexpression influenced
nomas of the breast. Oncogene 1992;7:1027–32. by the presence of in situ carcinoma. J Clin
77. Kallioniemi OP, Kallioniemi A, Kurisu W, et al. Oncol 1992;10(4):599–605.
erbB-2 amplification in breast cancer analyzed 89. Leitzel K, Teramoto Y, Konrad K, et al. Elevated
by fluorescence in situ hybridization. Proc Natl serum c-erbB-2 antigen levels and decreased
Acad Sci U S A 1992;89(12):5321–5. response to hormone therapy of breast cancer.
78. Dittadi R, Catozzi L, Gion M, et al. Comparison J Clin Oncology 1995;13(5):1129–35.
between Western blotting, immunohistochemi- 90. Borg A, Baldetorp B, Ferno M, et al. erbB-2
cal and ELISA assay for p158neu quantitation amplification is associated with tamoxifen
in breast cancer specimens. Anticancer Res resistance in steroid-receptor positive breast
1993;13(5C):1821–4. cancer. Cancer Lett 1994;81:137–44.
Prognostic and Predictive Markers in Breast Cancer 129

91. Benz CC, Scott GK, Sarup JC, et al. Estrogen- dermal growth factor receptor sites in 216
dependent, tamoxifen-resistant tumorigenic human breast cancer biopsies. Breast Cancer
growth of MCF-7 cells transfected with HER2/ Res Treat 1992;20(3);177–83.
neu. Breast Cancer Res Treat 1993;24(2):85–95. 103. Ro J, North SM, Gallick GE, et al. Amplified and
92. Wright C, Nicholson S, Angus B, et al. Relation- overexpressed epidermal growth factor recep-
ship between c-erbB-2 protein product expres- tor gene in uncultured primary human breast
sion and response to endocrine therapy in carcinoma. Cancer Res 1988;48:161–4.
advanced breast cancer. Br J Cancer 1992; 104. Chrysogelos SA, Yarden RI, Lauber AH, Murphy
65(1):118–21. JM. Mechanisms of EGF receptor regulation in
93. Carlomagno C, Perrone F, Gallo C, et al. c-erbB- breast cancer cells. Breast Cancer Res Treat
2 overexpression decreases the benefit of adju- 1994;31:227–36.
vant tamoxifen in early-stage breast cancer 105. Ishii S, Xu Y-H, Stratton RH, et al. Characteriza-
without axillary lymph node metastases. J Clin tion and sequence of the promoter region of the
Oncol 1996;14(10):2702–8. human epidermal growth factor receptor gene.
94. Archer SG, Eliopoulos A, Spandidos D, et al. Proc Natl Acad Sci U S A 1985;82:4920–4.
Expression of ras p21, p53 and c-erbB-2 in 106. Koenders PG, Beex LV, Geurts-Moespot A, et al.
advanced breast cancer and response to first Epidermal growth factor receptor-negative
line hormonal therapy. Br J Cancer 1995; tumors are predominantly confined to the sub-
72(5):1259–66. group of estradiol receptor-positive human pri-
95. Loaiciga K, Beckmann MW, Niederacher D, et mary breast cancers. Cancer Res 1991;51(17):
al. p185/HER2 and pS2 protein overexpression 4544–8.
in breast cancer specimens: improvement for 107. Worthylake R, Opresko LK, Wiley HS. ErbB-2
prediction of response to endocrine therapy? amplification inhibits down-regulation and
Oncol Rep 1994;1:625–9. induces constitutive activation of both erbB-2
96. Elledge RM, Green S, Ciocca D, et al. HER-2 and epidermal growth factor receptors. J Biol
expression and response to tamoxifen in estro- Chem 1999;274:8865–74.
gen receptor-positive breast cancer: a South- 108. King CR, Borrello I, Bellot F, et al. EGF binding
west Oncology Group study. Clin Cancer Res to its receptor triggers a rapid tyrosine phos-
1998;4:7–12. phorylation of the erbB-2 protein in the mam-
97. Diamond A, The Advisory Board Company. Her- mary tumor cell line SK-BR-3. EMBO J 1988;
ceptin® offers new treatment option for 7(6):1647–51.
advanced breast cancer patients. The Oncology 109. Buday L, Downward J. Epidermal growth factor
Roundtable, Oncology Watch 1998;Issue #7. regulates p21ras through the formation of a
98. Park JW, Hong K, Carter P, et al. Development of complex of receptor, Grb2 adapter protein, and
anti-p185HER2 immunoliposomes for cancer Sos nucleotide exchange factor. Cell 1993;
therapy. Proc Natl Acad Sci U S A 1995;92: 73(3):611–20.
1327–31. 110. Sadowski HB, Shuai K, Darnell JE Jr, Gilman
99. Pegram M, Lipton A, Pietras R, et al. Phase II MZ. A common nuclear signal transduction
study of intravenous recombinant humanized pathway activated by growth factor and
anti-p185 HER-2 monoclonal antibody (rhuMAb cytokine receptors. Science 1993;261:1739–44.
HER-2) plus cisplatin in patients with HER-2/ 111. Skoog I, Macias A, Azavedo E, et al. Receptors
neu overexpressing metastatic breast cancer. for EGF and oestradiol and thymidine kinase
Proc Am Soc Clin Oncol 1995;14:A124. activity in different histological subgroups of
100. Davidson NE, Gelmann EP, Lippman ME, Dick- human mammary carcinomas. Br J Cancer
son RB. Epidermal growth factor receptor gene 1986;54(2):271–6.
expression in estrogen receptor-positive and 112. Winchester DJ, Goldschmidt RA, Kahn SH, et al.
negative human breast cancer cell lines. Mol Flow cytometric and molecular prognostic
Endocrinol 1987;1(3):216–23. markers in 91 male breast carcinoma patients
101. Ennis BW, Lippman ME, Dickson RB. The EGF [meeting abstract]. 46th Annual Cancer Sympo-
receptor system as a target for antitumor ther- sium in Conjunction with Society of Head and
apy. Cancer Invest 1991;9(5):553–62. Neck Surgeons; 1993 March 18–21; Los Ange-
102. Falette N, Lefebvre MF, Meggouh F, et al. Mea- les, CA: Society of Surgical Oncology; 1993.
surement of occupied and non-occupied epi- 113. Toi M, Nakamura T, Mukaida H, et al. Relation-
130 BREAST CANCER

ship between epidermal growth factor receptor human breast cancer. Oncogene 1991;6(12):
status and various prognostic factors in human 2277–84.
breast cancer. Cancer 1990;65:1980–4. 125. Nigro JM, Baker SJ, Preisinger AC, et al. Muta-
114. Sainsbury JRC, Farndon JR, Harris AL, Sherbet tions in the p53 gene occur in diverse human
GV. Epidermal growth factor receptors on tumour types. Nature 1989;342:705–8.
human breast cancers. Br J Surg 1985;72:186–8. 126. Davidoff AM, Humphrey PA, Iglehart JD, Marks
115. Nicholson S, Wright C, Sainsbury JR, et al. Epi- JR. Genetic basis for p53 overexpression in
dermal growth factor receptor (EGFr) as a human breast cancer. Proc Natl Acad Sci
marker for poor prognosis in node-negative U S A 1991;88(11):5006–10.
breast cancer patients: neu and tamoxifen failure. 127. Prosser J, Thompson AM, Cranson G, Evans HJ.
J Steroid Biochem Mol Biol 1990;37(6):811–4. Evidence that p53 behaves as a tumour sup-
116. Nicholson S, Richard J. Sainsbury C, et al. Epi- pressor gene in sporadic breast tumours.
dermal growth factor receptor (EGFr); results Oncogene 1990;5(10):1573–9.
of a 6 year follow-up study in operable breast 128. Runnebaum IB, Nagarajan M, Bowman M, et al.
cancer with emphasis on the node negative Mutations in p53 as potential molecular mark-
subgroup. Br J Cancer 1991;63:146–50. ers for human breast cancer. Proc Natl Acad
117. Sainsbury JR, Farndon JR, Sherbet GV, Harris Sci U S A 1991;88(23):10657–61.
AL. Epidermal growth factor receptors and 129. Bergh J, Norberg T, Sjogren S, et al. Complete
oestrogen receptors in human breast cancer. sequencing of the p53 gene provides prognostic
Lancet 1985;1(8425):364–6. information in breast cancer patients, particu-
118. Klijn JG, Berns PM, Schmitz PI, Foekens JA. The larly in relation to adjuvant systemic therapy and
clinical significance of epidermal growth factor radiotherapy. Nature Med 1995;1(10):1029–34.
receptor (EGF-R) in human breast cancer: a 130. Hawkins DS, Demers GW, Galloway DA. Inacti-
review on 5232 patients. Endocr Rev 1992; vation of p53 enhances sensitivity to multiple
13(1):3–17. chemotherapeutic agents. Cancer Res 1996;56
119. Stark A, Hulka BS, Conway JS, et al. Her-2/neu (4):892–8.
amplification and the risk of subsequent inva- 131. Levine AJ. p53, the cellular gatekeeper for
sive breast cancer. [In press] growth and division. Cell 1997;88(3):323–31.
120. Thor AD, Yandell DW. Prognostic significance of 132. Kleihues P, Schauble B, zur Hausen A, et al.
p53 overexpression in node-negative breast Tumors associated with p53 germline muta-
carcinoma: preliminary studies support cau- tions: a synopsis of 91 families. Am J Pathol
tious optimism [editorial]. J Natl Cancer Inst 1997;150(1):1–13.
1993;85(3):176–7. 133. Sobol H, Stoppa-Lyonnet D, Bressac-De Paillerets
121. Thor AD, Moore DH II, Edgerton SM, et al. B, et al. BRCA1-p53 relationship in hereditary
Accumulation of p53 tumor suppressor gene breast cancer. Int J Oncol 1997;10:349–53.
protein: an independent marker of prognosis in 134. Marubini E, Valsecchi MG. Analysing survival
breast cancers. J Natl Cancer Inst 1992;84: data from clinical trials and observational stud-
845–55. ies. New York: Wiley & Sons; 1995.
122. Yandell DW, Thor AD. p53 analysis in diagnostic 135. Harrington DP, Fleming TR. A class of rank test
pathology: biologic implications and possible procedures for censored survival data. Bio-
clinical applications. Diagn Mol Pathol 1993; metrika 1982;69:53–56.
2(1):1–3. 136. Cox DR. Regression models and life tables (with
123. Thor AD. Prognostic and predictive markers in discussion). J R Statist Soc 1972;34(B):187–220.
breast cancer: issues related to molecular 137. Edgington ES. Randomization tests. New York:
determinants of outcome. Breast J 1998;4(5): Marcel Dekker; 1995.
379–82. 138. Berry DA. When is a confirmatory randomized
124. Horak EK, Smith K, Bromley L, et al. Mutant clinical trial needed? [editorial]. J Natl Cancer
p53, EGF receptor and c-erbB-2 expression in Inst 1996;88:1606–7.
8
Surgical Management of
Ductal Carcinoma In Situ
STEPHEN F. SENER, MD
LAURIE H. LEE, PA-C

Although Broders first defined the pathologic ologists of the natural evolution of calcifica-
entity of ductal carcinoma in situ (DCIS) in tions related to DCIS, and technologic
1932, in situ disease remained a clinical curios- advances in mammography equipment.
ity until the mid-1970s because of the unusual Since 1976, the Cancer Incidence and End
association of a palpable mass with noninfil- Results (CIER) Committee of the American
trating cancer.1 With the widespread acceptance Cancer Society, Illinois Division, has published
of screening mammography for breast cancer incidence data on approximately 85 percent of
detection came a significant increase in the patients treated for cancer at Illinois hospitals. A
number of patients with nonpalpable DCIS. retrospective report from the CIER Committee
Reports over the last two decades have demon- was based on 10,974 breast cancer patients
strated equivalent survival results for the treat- diagnosed from 1970 to 1975.3 Only 2 percent
ment of DCIS with mastectomy versus breast- of patients during this time period had in situ
conservation therapy. Yet, the limitations of disease, reflecting the stage distribution com-
early studies led to ambivalence about the effi- monly seen in the era before the availability of
cacy of breast conservation. Illustrating this mammography.4,5 The number of patients with
sentiment was the fact that mastectomy was DCIS, as a percentage of the total number of
more commonly used than lumpectomy for female breast cancer patients, has steadily
patients with DCIS from 1985 through 1991, as increased to 12.9 percent in 1995, corroborating
reported by the American College of Surgeons data from the NCDB.2,6 The combined statewide
using the National Cancer Data Base (NCDB).2 registry and mammography survey data from
This chapter focuses on clinical research 1985 to 1994 revealed that 2.2 patients with
that has attempted to predict risk factors for DCIS were identified per 1,000 mammograms.
ipsilateral recurrence after treatment of DCIS Data from Evanston Northwestern Healthcare
with breast conservation therapy. compiled from 1994 to 1996 demonstrated that
the number of patients with DCIS gradually
INCIDENCE increased until age 70 years and then remained
constant thereafter (Figure 8–1).7
The increased incidence of DCIS over the last Despite the fact that most investigators
two decades has resulted from a significant regard DCIS as a preinvasive phase of malig-
increase in the number of screening mammo- nant transformation, in some women, it is pre-
grams per year, heightened awareness by radi- sent but never becomes clinically relevant. For

131
132 BREAST CANCER

example, the incidence of occult multicentric cancers as biologically aggressive.14 The risk of
DCIS in mastectomy specimens is higher than disease-related mortality, although low, was
the ipsilateral breast tumor recurrence rate after finite, most likely due to difficulty in identify-
lumpectomy, with or without radiation.8 As fur- ing areas of invasion within the excised breast.
ther evidence for the heterogeneous natural his- Clearly distinct from these subsets are the cur-
tory of DCIS, seven autopsy series of women rent, more common patients with screening
without a history of breast cancer were collec- mammograms who present with localized, non-
tively evaluated, and it was demonstrated that palpable areas of calcification representing
the median prevalence of DCIS was 8.9 percent DCIS, in whom the incidence of multicentricity
(range 0 to 14.7 percent), depending on the level or invasion is low.
of scrutiny of the pathologic examination.9 The difficulty in identifying areas of inva-
sion continues to exist today and leads to a low
TREATMENT OF PRIMARY but continuing risk of mortality from DCIS,
DUCTAL CARCINOMA IN SITU especially for patients with multicentric or geo-
graphically large, comedo DCIS.15 Treatment
Total Mastectomy recommendations must take into account the
risk of noninvasive and invasive recurrence as
Reports on the treatment of DCIS by mastec- well as that of disease-related mortality. Cur-
tomy serve as historical benchmarks for com- rently, mastectomy is generally reserved for
parison with breast conservation therapy, con- patients in whom lumpectomy results in either
sisting primarily of patients presenting prior to positive pathologic margins or unacceptable
the wide acceptance of mammographic screen- cosmesis. A small percentage of patients who
ing. Patients in these series frequently had nip- might otherwise be candidates for breast con-
ple discharge, Paget’s disease of the nipple, or servation therapy will elect mastectomy due to
palpable DCIS, and pathologic review fre- a lack of interest in cosmesis, inaccessibility of
quently demonstrated evidence of invasion. radiation treatment facilities, or a history of
Locoregional recurrence and disease-related connective tissue disease.
mortality rates were about 1 percent.10–13 Chest
wall recurrences were invasive and defined the
Lumpectomy with Radiation Therapy

The National Surgical Adjuvant Breast Project


(NSABP) Protocol B-17 was initiated in 1985
to test whether radiation after lumpectomy for
localized DCIS prevented recurrence of cancer
in the surgically treated breast.16–18 The updated
report (1997) presented findings on 814 eligi-
ble patients through 8 years of follow-up. Of
403 patients treated by lumpectomy alone, 104
(26.8%) had an ipsilateral breast recurrence.
Fifty-one (13.4%) recurrences were noninva-
sive and 53 (13.4%) were invasive. Of the 411
patients treated by lumpectomy and radiation,
47 (12.1%) had an ipsilateral breast recurrence.
Thirty (8.2%) recurrences were noninvasive,
Figure 8–1. Numbers of noninvasive and invasive breast
cancers by age among 609 patients treated at Evanston
and 17 (3.9%) were invasive. Thus, the addition
Northwestern Healthcare, 1994 to 1996. of radiation to lumpectomy in the treatment of
Surgical Management of Ductal Carcinoma In Situ 133

localized DCIS significantly reduced the inci- recurrence was 5 years and to a noninvasive
dence of noninvasive and invasive ipsilateral recurrence was 4 years.24 A longer time interval
breast recurrence (p < .000005). Despite a bet- to an invasive than a noninvasive recurrence has
ter disease-free survival in those treated with also been reported by two other groups.20,22 In
lumpectomy and radiation (75% versus 62%, NSABP B-17, 58 percent of all recurrences
p = .00003), overall survival was equivalent were within 2 years of treatment.17 Fowble and
(95% versus 94%). colleagues concluded from their data that
Numerous retrospective studies have been increased attention to efforts that assure com-
done to determine variables associated with an plete excision of DCIS prior to radiation (exci-
increased risk of local recurrence after breast sion to negative margins > 2 mm, and negative
conservation therapy. The recurrence and sur- preradiotherapy mammography) reduced the
vival results of lumpectomy and radiation for risk of noninvasive local recurrence. And, as
mammographically detected DCIS are shown that type of recurrence was eliminated, late
in Table 8–1.12,16–24 Ipsilateral breast recurrence invasive recurrence became the predominant
rates ranged from 0 to 10 percent at 5 years and type of ipsilateral breast failure.20
8 to 23 percent at 10 years. The most compre-
hensive evaluations had the lowest recurrence Factors Associated with Ipsilateral
rates and included mammographic and patho- Breast Recurrence after Lumpectomy
logic correlation, with microscopic margin with Radiation Therapy
analysis, classification of architectural pattern,
determination of tumor size, and use of Treatment-Related Factors
postlumpectomy mammography to assess the
completeness of excision. Recent studies have indicated that microscopic
The patterns of recurrence after lumpectomy margin status is a predictor for ipsilateral breast
and radiation for mammographically detected recurrence.12,17,20,21,23,24 For example, in the
DCIS are shown in Table 8–2. Most ipsilateral series reported by Fowble and colleagues, the
breast recurrences occurred in the same vicinity 5-year actuarial breast recurrence rate was 0
as the primary tumor, and approximately 50 per- percent for patients with negative or unknown
cent were invasive cancers when detected. Solin margins and 8 percent for those with positive or
and colleagues reported that the median time close margins.20 Solin and colleagues reported
interval from diagnosis of DCIS to an invasive a recurrence rate of 29 percent for patients with

Table 8–1. RECURRENCE AND SURVIVAL RESULTS OF LUMPECTOMY AND RADIATION


FOR MAMMOGRAPHICALLY DETECTED DCIS

Ipsilateral Cause-Specific
Recurrence (%) Survival (%)

Authors (ref. #) Number of Patients 5 yr 10 yr 5 yr 10 yr Median Follow-Up (y)

NSABP B-1716–18 399* 10 12.1 — 75† 8.0 (mean)


Kuske, et al19 44 7 — — — 4.0
Fowble, et al20 110 1 15 100 100 5.3
Vicini, et al21 105 8.8 10.2 — 99 6.5
Hiramatsu, et al22 54 2 23 — 96 6.2
Sneige, et al23 31 0 8 — — 7.2
Silverstein, et al12 133‡ 7 19 — 97 7.8
Solin, et al24 110 7 14 100 96 9.3
*81 percent detected by mammography

8-year disease-free survival

89 percent detected by mammography
134 BREAST CANCER

Table 8–2. PATTERNS OF IPSILATERAL BREAST RECURRENCE AFTER LUMPECTOMY


AND RADIATION FOR MAMMOGRAPHICALLY DETECTED DCIS

Number of Local Invasive Median Time (y)


Authors Recurrences Recurrence*(%) Recurrence(%) to Recurrence

NSABP B-1718 47 — 36 —
Kuske, et al19 3 33 100 2.6
Fowble, et al20 3 33 100 8.8
Vicini, et al21 10 70 70 2.4
Hiramatsu, et al22 4 75 25 6.1
Sneige, et al23 1 — 0 8
Silverstein, et al12 16 100 50 4.9
Solin, et al24 15 73 40 5

* recurrence within lumpectomy site

positive or close margins and 7 percent for DCIS grew in a continuous pattern, implying
those with negative margins.24 The median time that margin assessment should be accurate.
interval to recurrence was 3.6 years for patients However, well-differentiated DCIS grew in a
with positive margins and 4.3 years for those discontinuous (multifocal) pattern in 70 percent
with negative margins. From the NSABP B-17 of patients, making margin analysis problematic.
trial, the breast recurrence rates, with a mean Of specimens with multifocal DCIS, about 65
follow-up of 43 months, were 10 percent for percent had gaps < 5 mm, 20 percent had gaps
patients with positive or unknown margins and 5 to 10 mm, and 10 percent had gaps >10 mm.
4 percent for those with negative margins.17 The influence of pathologic factors on ipsi-
The presence of malignant-appearing calcifi- lateral breast recurrence remains an area for
cations on preradiation mammography has been active investigation. It was initially suggested by
highly predictive of recurrence. Two series have Solin and colleagues that high-grade DCIS or
reported five such patients, all of whom had dis- comedo necrosis was associated with a higher
ease recurrence.23,25 A third series reported that rate of breast recurrence.28 However, this series
there were no recurrences in 37 patients with with a shorter follow-up underestimated the
negative preradiation mammography.20 number of recurrences in low-grade and non-
comedo DCIS, and recurrences with high-grade
Pathologic Factors or comedo DCIS were predominant. Longer fol-
low-up revealed late recurrences with low-grade
Confounding the pathologist’s ability to assess and noncomedo DCIS, a finding also reported
the adequacy of surgical margins is the growth by Silverstein and colleagues.15,29
pattern of DCIS within the duct system. Silver-
stein and colleagues demonstrated that even with Combination of Treatment-Related
negative margins and preradiation mammogra- and Pathologic Factors
phy to confirm excision of all calcifications, (Van Nuys Prognostic Index)
more than 50 percent of patients had residual
DCIS in re-excision or mastectomy specimens.15 In 1995, Silverstein and colleagues devised a
Holland demonstrated that mammography may scoring system, based on retrospective data,
underestimate the extent of DCIS by 2 cm in 15 that combined three independent predictors of
to 20 percent of patients.26 An additional study local recurrence after breast conservation treat-
by Faverly and colleagues demonstrated a differ- ment in patients with DCIS: tumor size, margin
ence in the growth patterns of well- and poorly width, and pathologic classification.30 Scores,
differentiated DCIS.27 Poorly differentiated ranging from 1 to 3, were assigned to each of
Surgical Management of Ductal Carcinoma In Situ 135

the variables defined by multivariate analyses Clinical Factors


(Table 8–3). The Van Nuys Prognostic Index
(VNPI) scoring system was validated using Solin and colleagues reported that the ipsilat-
data from 394 patients treated for DCIS with eral breast recurrence rate for women < 50
breast conservation: 209 by lumpectomy alone years of age was 25 percent compared with 2
and 185 by lumpectomy and radiation. Patients percent for those ³ 50 years, and the time inter-
were divided into three groups with different val to recurrence was 4.9 years for the younger
probabilities for ipsilateral breast recurrence, women compared with 8.7 years for the single
on the basis of VNPI scores (3 to 4, 5 to 7, 8 to recurrence in women ³ 50 years.24 Van Zee and
9). The 12-year local recurrence-free survival colleagues also reported an increased recur-
rates were 98 percent for those with VNPI = 3 rence risk for women < 40 years of age versus
and 4; 70 percent for those with VNPI = 5 to 7; those ³ 40 years.31 However, other investigators
and 28 percent for those with VNPI = 8 and 9. were unable to confirm the correlation of
The 12-year breast cancer-specific survival young age with increased risk of breast recur-
rates were 100 percent for those with VNPI = 3 rence after breast conservation treatment.20,22,23
and 4; 99 percent for those with VNPI = 5 to 7; An increased risk of breast recurrence was
and 95 percent for those with VNPI = 8 and 9. associated with a family history of breast cancer
In patients with VNPI scores of 3 and 4, by two groups of investigators.22,25 However,
there was no difference in disease-free survival this finding remains unconfirmed by others, so
in those treated with lumpectomy and radiation the impact of family history on ipsilateral breast
versus those treated by lumpectomy alone. In recurrence remains uncertain at this time.20
patients with intermediate VNPI scores (5 to 7),
there was a 13 percent lower local recurrence Lumpectomy Alone
rate in those treated with lumpectomy and radi-
ation versus those treated with lumpectomy Lagios and colleagues proposed that lumpec-
alone (p = .027). Even though there was a sig- tomy alone was appropriate treatment for
nificantly lower local recurrence rate in selected patients with mammographically
patients with VNPI = 8 and 9 treated by detected DCIS.14,32 Selection criteria included
lumpectomy and radiation versus lumpectomy tumor size of 25 mm or less, histologically
alone, close to 60 percent of those treated by negative margins of excision, and postopera-
lumpectomy and radiation had an ipsilateral tive mammography to confirm the absence of
breast recurrence with an 81-month median fol- calcifications remaining in the breast. Follow-
low-up. Although this prognostic scheme has a up of the original 79 patients reported in 1989
rational formula, it was defined using a retro- revealed a 15-year actuarial local recurrence
spectively identified cohort. Further validation rate of 19 percent.33 The local recurrence rates
in a prospective analysis will be important to were 33 percent for patients with high-grade
confirm its conclusions. DCIS, 10 percent for those with intermediate-

Table 8–3. THE VAN NUYS PROGNOSTIC INDEX (VNPI) SCORING SYSTEM

Score 1 2 3

Size (mm) 15 or less 16 to 40 41 or more


Margin width (mm) 10 or more 1 to 9 Less than 1
Pathologic class Non–high grade Non–high grade High grade
No necrosis Necrosis
Nuclear grade 1 to 2 Nuclear grade 1 to 2 Nuclear grade 3

VNPI = size score + margin score + pathologic class score.


136 BREAST CANCER

grade DCIS, and 6 percent for those with low- and 1 (3%) was stage IV. However, node infor-
grade DCIS. In addition, local recurrence rates mation was available for only 5 of the 35
were 68 percent for patients with margins < 1 patients with invasive recurrence because 30
mm, 20 percent for those with margins 1 to 9 patients initially underwent axillary dissection
mm, and 7 percent for those with margins of with the treatment of their primary tumor. Thus,
10 mm or more. Other authors have reported some of the remaining 30 patients were probably
similar results (14 to 27% breast recurrence understaged. At 8-year follow-up after treatment
rates) with follow-up times of 45 to 90 of the 35 patients for local invasive recurrence,
months.18,34-37 These data have led to the con- the probability of developing distant metastases
clusion that there may be subsets of patients was 27 percent and the breast cancer–specific
with DCIS for whom lumpectomy alone is ade- mortality rate was 14.4 percent. The 8-year
quate treatment. breast cancer mortality rate for 448 patients
The NSABP B-17 trial represents the only that had breast conservation treatment for
randomized comparison of lumpectomy with or DCIS was 2.1 percent. The results indicated
without radiation therapy. Although the data that, regardless of treatment choice, the overall
from this study strongly supported the use of mortality rates were low.
radiation therapy to decrease the risk of ipsilat- Re-excision of recurrent disease may be a
eral breast recurrence, DCIS consists of a broad consideration for patients treated initially with
spectrum of disease defined by grade and lumpectomy alone. After re-excision, radiation
extent. Failure to scrutinize these pathologic therapy should be included in the treatment pro-
variations has been a criticism of the study.38 gram. For most, a recurrence occurs in the con-
Although there may be subsets of patients that text of an irradiated breast, and a completion
are adequately treated by lumpectomy alone, a mastectomy is usually the treatment of choice.
successful outcome with this approach is
dependent on careful selection of good-risk RECOMMENDATIONS FOR FOLLOW-UP
patients and demonstration of clear surgical CARE OF PATIENTS WITH DCIS
margins after lumpectomy.
In 1992, a collaborative effort of the American
RESULTS OF TREATMENT FOR Colleges of Radiology and Surgeons, the Col-
IPSILATERAL BREAST TUMOR lege of American Pathologists, and the Society
RECURRENCE of Surgical Oncology led to a publication regard-

Survival results after treatment of an ipsilateral Table 8–4. RESULTS OF TREATMENT FOR
breast recurrence following conservative IPSILATERAL BREAST RECURRENCE FOLLOWING
surgery for DCIS are shown in Table 8–4. LUMPECTOMY AND RADIATION FOR DCIS

Patients with noninvasive recurrence were Number Developing Metastases /


treated by complete total mastectomy and none Total Number
developed distant metastases thereafter. In 707 Noninvasive Invasive
patients treated by Silverstein and colleagues Authors Recurrence Recurrence

(which included 259 patients initially treated by NSABP B-1716 0/20 1/8
mastectomy), the 8-year local recurrence rate Kuske, et al19 — 0/3
Fowble, et al20 — 1/3
was 12.5 percent.39 There were 74 recurrences: Vicini, et al21 0/3 1/7
39 were noninvasive, and 35 were invasive. At Hiramatsu, et al22 0/3 1/1
Sneige, et al23 0/1 —
the time of local invasive recurrence, 18 of 35 Silverstein, et al39 0/39 5/35
(51%) were stage I, 4 (11%) were stage IIA, 8 Solin, et al24 0/9 1/6
(23%) were stage IIB, 4 (11%) were stage IIIB, Total 0/75 10/61 (16%)
Surgical Management of Ductal Carcinoma In Situ 137

ing standards of care for invasive and noninva- Cancer in Illinois: incidence reports. Chicago:
sive breast cancer treated by breast conservation American Cancer Society, Illinois Division,
Inc.; 1983–1995.
therapy.40 A task force of the same four national
7. Sener SF, Winchester DJ, Winchester DP, et al.
organizations published a subsequent separate Spectrum of mammographically detected breast
standard of care for patients with DCIS.41 cancers. Am Surg 1999;65:731–6
The goals of routine follow-up include the 8. Schwartz GF, Patchefsky AS, Feig SA, et al. Mul-
identification of treatment sequelae and early ticentricity of non-palpable breast cancer. Can-
detection of recurrent or new breast cancers. cer 1980;45:2913–6.
9. Welch HG, Black WC. Using autopsy series to
Regular clinical examinations and breast imag-
estimate the disease “reservoir” for ductal car-
ing are the cornerstones of effective follow-up cinoma in situ of the breast: how much more
care. Routine tests for metastatic disease are not breast cancer can we find? Ann Intern Med
indicated for asymptomatic patients after treat- 1997;127:1023–8.
ment for DCIS. Clinical examinations should be 10. Ashikari R, Hajdu SI, Robbins GF. Intraductal
done every 6 months for at least 5 years and per- carcinoma of the breast (1960–1969). Cancer
1971;28:1182–7.
haps through 8 years, when the risk of ipsilateral 11. Kinne D, Petrek JA, Osborne MP, et al. Breast car-
breast recurrence after breast-conservation ther- cinoma in situ. Arch Surg 1989;124:33–6.
apy approaches that of contralateral breast can- 12. Silverstein MJ, Barth A, Poller DN, et al. Ten-year
cer. A preradiation therapy ipsilateral mammo- results comparing mastectomy to excision and
gram (with magnification views, as necessary) radiation therapy for ductal carcinoma of the
should be done to ensure that there are no resid- breast. Eur J Cancer 1995;37:1425–7.
13. Rosner D, Bedwani RN, Vana J, et al. Noninvasive
ual suspicious calcifications after lumpectomy. breast carcinoma: results of a national survey
A baseline mammogram of the treated breast by the American College of Surgeons. Ann
should be done during the first year after breast- Surg 1980;192:139–47.
conservation therapy, and thereafter annually or 14. Lagios MD, Westdahl PR, Margolin FR, Rose
more frequently, if warranted by clinical or radi- MR. Duct carcinoma in situ: relationship of
extent of noninvasive disease to the frequency
ographic findings. Mammography of the con-
of occult invasion, multicentricity, lymph node
tralateral breast should be done at least annually, metastases, and short-term failures. Cancer
depending on clinical or radiographic findings. 1982;50:1309–14.
15. Silverstein MJ, Waisman JR, Gamagami P, et al.
REFERENCES Intraductal carcinoma of the breast (208
cases): clinical factors influencing treatment
1. Broders AC. Carcinoma in situ contrasted with choice. Cancer 1990;66:102–7.
benign penetrating epithelium. JAMA 1932;99: 16. Fisher B, Constantino J, Redmond C, et al.
1670–4. Lumpectomy compared with lumpectomy and
2. Winchester DJ, Menck HR, Winchester DP. radiation therapy for the treatment of intraductal
National treatment trends for ductal carcinoma breast cancer. N Engl J Med 1993;328:1581–6.
in situ of the breast. Arch Surg 1997;132:660–5. 17. Fisher ER, Constantino J, Fisher B, et al. Patho-
3. Cancer Incidence and End Results Committee. logic findings from the National Surgical
Cunningham MP, Chairman. Breast cancer: a Adjuvant Breast Project (NSABP) Protocol
report by 53 Illinois hospitals on cases diag- B-17. Intraductal carcinoma (ductal carcinoma
nosed 1970–1975. Chicago: American Cancer in situ). Cancer 1995;75:1310–9.
Society, Illinois Division, Inc.; 1982. 18. Fisher B, Dignam J, Wolmark N, et al. Lumpec-
4. Hughes KS, Lee AK, Rolfs A. Controversies in tomy and radiation therapy for the treatment of
the treatment of ductal carcinoma in situ. Surg intraductal breast cancer: findings from NSABP
Clin North Am 1996;76(2):243–65. B-17. J Clin Oncol 1998;16(2):441–52.
5. Rosen PP, Braun DW, Kinne D. The clinical sig- 19. Kuske RR, Bean JM, Garcia DM, et al. Breast
nificance of pre-invasive breast carcinoma. conservation therapy for intraductal carcinoma
Cancer 1980;46:919–25. of the breast. Int J Radiat Oncol Biol Phys
6. Cancer Incidence and End Results Committee. 1993;26:391–6.
138 BREAST CANCER

20. Fowble B, Hanlon AL, Fein DA, et al. Results of 30. Silverstein MJ. The Van Nuys Prognostic Index.
conservative surgery and radiation for mam- In: Silverstein MJ, editor. Ductal carcinoma in
mographically detected ductal carcinoma in situ of the breast. Baltimore: Williams and
situ (DCIS). Int J Radiat Oncol Biol Phys Wilkins; 1997. p. 491–501.
1997;38(5):949–57. 31. Van Zee P, Liberman L, McCormick B, et al. Long
21. Vicini FA, Lacerna MD, Goldstein NS, et al. Duc- term follow-up of DCIS treated with breast
tal carcinoma in situ detected in the mammo- conservation: effect of age and radiation
graphic era: an analysis of clinical, pathologic, [Abstr]. Soc Surg Oncol 1996;82:26.
and treatment-related factors affecting out- 32. Lagios MD, Margolin FR, Westdahl PR, Rose
come with breast-conserving therapy. Int J MR. Mammographically detected ductal carci-
Radiat Oncol Biol Phys 1997;39(3):627–35. noma in situ. Frequency of local recurrence
22. Hiramatsu H, Bornstein BA, Recht A, et al. Local following tylectomy and prognostic effect of
recurrence after conservative surgery and radi- nuclear grade and local recurrence. Cancer
ation therapy for ductal carcinoma in situ. Pos- 1989;63:618–24.
sible importance of family history. Cancer J 33. Lagios M. Lagios experience. In: Silverstein MJ,
Sci Am 1995;1:55–61. editor. Ductal carcinoma in situ of the breast.
23. Sneige N, McNeese MD, Atkinson EN, et al. Duc- Baltimore: William and Wilkins; 1997. p. 361–5.
tal carcinoma in situ treated with lumpectomy 34. Shreer I. Conservation therapy of DCIS without
and irradiation: histopathologic analysis of 49 radiation. Breast Dis 1996;9:27–36.
specimens with emphasis on risk factors and 35. Arnesson LG, Olsen K. Linkoping experience. In:
long term results. Hum Pathol 1995;26:642–9. Silverstein MJ, editor. Ductal carcinoma in situ
24. Solin LJ, McCormick B, Recht A, et al. Mammo- of the breast. Baltimore: Williams and Wilkins;
graphically detected, clinically occult ductal 1997. p. 373–8.
carcinoma in situ (intraductal carcinoma) 36. Schwartz GF. Treatment of subclinical ductal car-
treated with breast conserving surgery and cinoma in situ by excision and local surveil-
definitive breast irradiation. Cancer J Sci Am lance. In: Silverstein MJ, editor. Ductal carci-
1996;2:158–65. noma in situ of the breast. Baltimore: Williams
25. McCormick B, Rosen PP, Kinne D, et al. Ductal and Wilkins; 1997. p. 353–60.
carcinoma in situ of the breast: an analysis of 37. Silverstein MJ. Van Nuys experience by treatment.
local control after conservative surgery and In: Silverstein MJ, editor. Ductal carcinoma in
radiotherapy. Int J Radiat Oncol Biol Phys situ of the breast. Baltimore: Williams and
1991;21:289–92. Wilkins; 1997. p.443–8.
26. Holland R, Hendriks JHCL, Verbeek ALM, et al. 38. Page D, Lagios M. Pathologic analysis of the
Extent, distribution, and mammographic/histo- National Surgical Adjuvant Breast Project
logical correlations of breast ductal carcinoma (NSABP) B-17 Trial. Unanswered questions
in situ. Lancet 1990;335:519–22. remaining unanswered considering current
27. Faverly DRG, Burgers L, Bult P, Holland R. Three concepts of ductal carcinoma in-situ [Editor-
dimensional imaging of mammary ductal car- ial]. Cancer 1995;75:1219–22.
cinoma in situ: clinical implication. Semin 39. Silverstein MJ, Lagios MD, Martino S, et al. Out-
Diagn Pathol 1994;11:193–8. come after invasive local recurrence in patients
28. Solin LJ, Yeh IT, Kurtz J, et al. Ductal carcinoma in with ductal carcinoma in situ of the breast. J
situ (intraductal carcinoma) of the breast treated Clin Oncol 1998;16:1367–73.
with breast-conserving surgery and definitive 40. Winchester DP, Cox JD. Standards for breast-con-
irradiation. Cancer 1993;71:2532–42. servation treatment. CA Cancer J Clin 1992;
29. Solin LJ, Kurtz J, Fourquet A, et al. Fifteen-year 42:134-62.
results of breast conserving surgery and defin- 41. Winchester DP, Strom EA. Standards for diagno-
itive breast irradiation for the treatment of duc- sis and management of ductal carcinoma in
tal carcinoma in situ of the breast. J Clin Oncol situ of the breast. CA Cancer J Clin 1998;48
1996;14:754–63. (2):108–28.
9
Evaluation and Surgical
Management of Stage I and II
Breast Cancer
DAVID J. WINCHESTER, MD, FACS

Implementation of screening mammography cytologic diagnosis, a thorough breast exami-


and increased breast cancer awareness account nation and an explanation of options should be
for the vast majority of breast cancers present- conducted by the surgeon. If a stereotactic core
ing at an earlier stage. This combined with biopsy is performed prior to surgical evalua-
extensive data supporting the choice of breast tion, it is possible that a subtle physical finding
preservation has lead to a dramatic change in may be overlooked, leading to a more compli-
the treatment for early stage breast cancer. Hal- cated image-directed biopsy instead of an in-
sted firmly established radical mastectomy as office needle biopsy. If complicated by a post-
the sole surgical procedure for breast cancer. procedural hematoma, the therapeutic surgical
Although this provided improved locoregional procedures may also require localization.
control of disease, the results were disfiguring Although large postprocedural hematomas are
(Figure 9–1, right breast). Clinical trials have unusual, when they do occur, the physical
led to the evolution of therapy that has lessened examination may have very little role in formu-
physical deformity (Figure 9–1, Figure 9–2) lating a treatment plan.
and improved survival.

DIAGNOSTIC EVALUATION

Although the surgeon is sometimes the first


physician that encounters the patient with breast
cancer, this diagnosis may be initially suspected
by primary care physicians or other specialists.
Although few of these physicians will be
directly involved in the diagnostic procedures,
all should be familiar with the key issues rele-
vant to the initial evaluation of women with sus-
pected breast cancer.
A suspicious finding or an interval change
on a mammogram may require additional imag- Figure 9–1. Metachronous bilateral breast cancers treated
with radical mastectomy (left) and modified radical mastec-
ing studies. Prior to obtaining a histologic or tomy (right).

139
140 BREAST CANCER

A B
Figure 9–2. A and B, Excellent cosmetic outcome from breast preserving therapy (left breast).

The same concerns also apply to the radio- invasive from in situ tumors, although there may
graphic evaluation. Prior to embarking on a tis- be reliable signs in certain tumor types.1 Hor-
sue diagnosis, it is important to obtain a mam- mone receptor assays and immunohistochemi-
mogram and additional views or a sonogram, if cal stains for prognostic markers such as HER-
indicated. The usefulness of these studies is 2/neu and p53 can be determined from both
adversely affected by a breast hematoma. It is core samples and cytology preparations. The
also imperative that the contralateral breast be false-negative and false-positive rates of core
thoroughly evaluated by physical examination, biopsy and FNA cytology are comparable.2,3
mammography, and, if indicated, sonography.
After the physical examination and radio- THERAPY
graphic studies, a cytologic or histologic diagno-
sis is required prior to any therapeutic proce- The treatment of breast cancer continues to be
dures. If the lesion can be appreciated on physical refined to an individualized approach that
examination, an office-based aspiration or core strives to preserve the breast, chest wall mus-
biopsy is simple, cost effective, and expeditious cles, and lymphatics, when possible. To achieve
and simplifies the subsequent treatment in terms improved survival, multimodality therapy has
of localization of the malignancy. The diagnosis also been increasingly used, but according to
can be reliably obtained with either a cytologic need and efficacy. To define the optimal local,
aspiration or a histologic core biopsy. regional, and systemic therapies of breast can-
Establishing a diagnosis with core biopsy cer, the patient needs to be staged according to
provides histologic confirmation of malignancy the TNM (tumor, nodes, metastases) staging
and has the ability to distinguish between inva- system defined by the American Joint Commit-
sive and in situ carcinoma. This approach tee on Cancer4 (Tables 9–1 and 9–2). For most
requires a local anesthetic and has a greater patients with early stage breast cancer, surgical
potential for formation of a hematoma. Com- intervention serves as the first phase of treat-
pared with core biopsy, fine-needle aspiration ment. This step also moves beyond clinical stag-
(FNA) cytology is a simpler, less invasive tech- ing to pathologic staging to provide important
nique but requires expertise in the preparation prognostic information to direct adjuvant ther-
and interpretation of cytologic preparations. It apy decisions. In addition to the stage of the
has very limited capabilities in distinguishing tumor, the presence of ductal carcinoma in situ
Evaluation and Surgical Management of Stage I and II Breast Cancer 141

Table 9–1. TUMOR-NODE-METASTASIS (TNM) along with that, the shortest recovery as com-
CLASSIFICATION SYSTEM FOR BREAST CANCER
pared with a mastectomy with reconstruction.
Primary Tumor (T) Multiple prospective randomized studies have
TX Primary tumor cannot be assessed
T0 No evidence of primary tumor
confirmed the efficacy of BPT.6–11 No study
Tis Carcinoma in situ or Paget’s disease of the has identified a survival disadvantage of this
nipple, with no associated tumor approach. A nonrandomized comparison12 as
T1 Tumor 2 cm in greatest dimension
T1a 0.5 cm well as a meta-analysis of randomized trials13
T1b > 0.5 cm and 1.0 cm have shown equivalent survival rates between
T1c > 1.0 cm and 2.0 cm
T2 Tumor > 2 cm, and 5 cm in greatest dimension
these two approaches.
T3 Tumor > 5 cm in greatest dimension
T4 Tumor of any size with direct extension to chest
Patient Selection
wall or skin
T4a Extension to chest wall
T4b Edema, ulceration, or satellite nodules Despite the extensive data to support the use of
T4c Both T4a and T4b BPT, there are still patients who are not candi-
T4d Inflammatory carcinoma
dates for this approach. As surgery is usually the
Regional Lymph Nodes (N)
NX Regional lymph nodes cannot be assessed first treatment modality, the choice of therapy is
N0 No regional lymph node metastasis guided by the surgeon’s evaluation. A more
N1 Metastasis to ipsilateral axillary lymph node(s)
N2 Metastasis to ipsilateral axillary node(s) fixed to effective evaluation process ideally includes the
one another preoperative evaluation from other treating
N3 Metastasis to ipsilateral internal mammary lymph
physicians to provide a cohesive and compre-
node(s)
Distant Metastasis (M) hensive treatment plan before therapy begins.
MX Presence of distant metastasis cannot be Most randomized clinical trials included
assessed
M0 No distant metastasis patients with T1 and T2 tumors.6–11 Thus,
M1 Distant metastasis (includes metastases to supra tumors up to 5 cm can be safely managed with
clavicular lymph node[s])
this approach. A good cosmetic result may be
difficult to achieve with large T2 tumors, and
(DCIS) also has implications for the local man- neoadjuvant chemotherapy may improve the
agement of breast cancer. Small invasive breast ability to preserve breast tissue without com-
cancers accompanied by extensive DCIS may promising survival.14,15 Saving the breast is
require total mastectomy. important for psychological and cosmetic rea-
sons. This requires sound surgical judgment and
meticulous techniques. A large breast can more
Breast Preservation Therapy

Adherence to screening mammography guide- Table 9–2. BREAST CARCINOMA STAGES


lines has made most patients candidates for STAGE T N M
breast preservation therapy (BPT). Acceptance
0 Tis N0 M0
of this treatment approach has been gradual and I T1 N0 M0
dependent upon regional preferences and avail- IIA T0 N1 M0
ability of radiation therapy facilities.5 When T1 N1 M0
T2 N0 M0
considering treatment options for early breast IIB T2 N1 M0
cancer, good cosmesis is an important goal. T3 N0 M0
IIIA T0 N2 M0
Improvements in surgery and radiotherapy have T1 N2 M0
minimized the incidence of poor results seen T2 N2 M0
initially (Figures 9–4 and 9–5). For most T3 N1 or N2 M0
IIIB T4 Any N M0
patients, the best cosmetic result can be Any T N3 M0
achieved with breast preservation therapy and, IV Any T Any N M1
142 BREAST CANCER

patients who are interested in cosmesis and


committed to radiation therapy are good candi-
dates for BPT. Centrally located lesions includ-
ing Paget’s disease (Figure 9–6) were at one
time considered a relative contraindication to
BPT. Adequate treatment of tumors in this loca-
tion may necessitate resection of a portion or all
of the nipple-areolar complex (Figure 9–7).
This treatment has the distinct advantage of
maintaining the breast mound and a sensate
breast. Nipple reconstruction may be per-
formed, if desired.
Physical findings and mammographic
Figure 9–4. Poor cosmetic outcome resulting from poor
dimensions may not correlate with the histo-
incision placement, incision size, and hematoma formation.
logic findings after a lumpectomy is per-
formed.17 Thus, the choice of lumpectomy is
readily accommodate a larger lumpectomy than ultimately contingent upon the ability to
a smaller breast. Resection of more than a quad- achieve histologically clear surgical margins.
rant of the breast begins to have a significant Close or involved surgical margins are impor-
impact on the cosmetic result and leads to con- tant predictors of local failure and should
sideration of alternatives such as mastectomy prompt consideration of either re-excision or
with reconstruction.16 With the development of completion mastectomy, depending on the
microvascular techniques, reconstructive options extent of the margin involvement.18–20
and results have made total mastectomy a good Placement of the incision is important to
option for many patients. create a good cosmetic result and to allow for
In the preoperative evaluation of the patient, additional surgery in the case of microscopi-
the size of the primary tumor is an important cally involved margins. Optimal cosmesis usu-
discriminator in the selection of the surgical ally places incisions within skin folds or in a
therapy. Given an acceptable ratio between the curvilinear fashion around the nipple (Figure
size of the tumor and the size of the breast, 9–8). Incisions should be placed directly over

Figure 9–5. Poor cosmetic outcome resulting from radiation


injury. Figure 9–6. Paget’s disease.
Evaluation and Surgical Management of Stage I and II Breast Cancer 143

the primary to avoid tunneling and limit the


deformity and extent of dissection in the breast.
With the exception of superficial lesions, resec-
tion of skin or subcutaneous tissue is not
required. A small ellipse of skin may be helpful
for specimen orientation.
Aside from the size of the tumor, multicen-
tric tumors or extensive intraductal cancer are
also important in identifying poor candidates
for BPT. Clearing the surgical margin for in situ
disease is equally important for local control.
Mammographically occult in situ disease may
Figure 9–7. Central lumpectomy.
therefore have the potential to alter the surgical
therapy for even small invasive tumors. Syn-
chronous tumors located in different quadrants can extend into the breast parenchyma. Cyto-
must be approached with the same margin cri- keratin stains may facilitate the identification
teria as those for solitary lesions, leading to a of lobular cells but have uncertain prognostic
compromised cosmetic result. In addition, mul- information in axillary staging.23
ticentric disease also suggests that other areas Randomized clinical trials addressing breast
in the breast may contain unrecognized foci of preservation therapy have not separated or
cancer. Two or more ipsilateral tumors should excluded lobular carcinomas.6–11 Several non-
lead to strong consideration of a mastectomy randomized studies have found no difference in
with reconstruction. the local disease-free survival rates between
The histologic subtype of the breast primary breast preservation patients with lobular carci-
may have an impact on the local management. noma and those with ductal carcinoma24–26
In addition to DCIS, which has the propensity whereas others have noted a difference.17,27–29
to extend great distances in the breast without Analysis of the National Cancer Data Base did
any mammographic or physical findings, inva- not identify any significant differences in size,
sive lobular carcinoma may also have a perva- stage, or survival according to histology.30 It
sive presentation. In the case of in situ ductal would appear that there are not any specific his-
carcinoma, preoperative magnification views tologic categories that should exclude consider-
may help identify extensive pleomorphic calci- ation of BPT. The same principles of a careful
fications around a stellate mass. The surgical preoperative assessment and microscopic eval-
procedure should attempt to remove all suspi- uation of lumpectomy margins should lead to
cious calcifications. successful BPT for all histologic variants.
Invasive lobular carcinomas have a more
indolent presentation with a less defined mass
with indistinct borders. Mammographic find-
ings are subtle21,22 and more likely to underesti-
mate tumor dimensions, compared with other
invasive cancers.17 These characteristics account
for the greater likelihood of requiring re-
excision after lumpectomy.17 The histologic
evaluation of the lumpectomy and regional
lymph nodes is more difficult because of the
frequency of single malignant lobular cells that Figure 9–8. Incision placement for lumpectomy.
144 BREAST CANCER

Risk Factors for Local Recurrence lumpectomy candidates have also shown that
microscopic foci of cancer are identified beyond
Several important variables exist for local 2 cm of the primary in 41 percent of patients.35
recurrence after lumpectomy. The only variable Although an adequate margin is important, a
to predict local recurrence from analysis of more extensive resection needs to be balanced
National Surgical Adjuvant Breast and Bowel with the cosmetic result of the operation. In most
Project (NSABP) B-06 was age under 45 instances, resection of the pectoralis fascia with
years.31 Other variables analyzed included vas- the lumpectomy specimen will avoid concerns
cular and lymphatic invasion, tumor grade, and about posterior extension. Without muscle
size.31 Treatment-related variables include the involvement, inclusion of the pectoralis fascia
extent of resection, margin involvement, and with the lumpectomy specimen should assist in
the implementation of radiotherapy. Thus, the good local control, even with a close margin.
risk of local recurrence can be dramatically The handling of the surgical specimen
affected by treatment decisions. becomes critical when close or involved micro-
Defining the optimal margin distance is scopic margins are identified on the lumpec-
based on a subjective and individualized assess- tomy specimen. Inability to accurately define
ment of each patient, balancing the cosmetic specimen orientation risks having to remove too
outcome and pathologic characteristics. When large a specimen or removing the wrong area of
comparing a more extensive quadrantectomy persistent involvement. The surgeon and the
with lumpectomy, the risk of a local recurrence is pathologist should work closely together at the
reduced significantly in the former.32 Despite a time of surgery. Specimens should either be
significant reduction in local recurrence with a inked on six sides by the surgeon or have appro-
more extensive resection,32 radiation therapy has priate markers to allow the pathologist to do so.
been shown to add to local control in these Specimens should be submitted fresh for patho-
patients.33 Margin involvement is a strong predic- logic examination so that any questions about
tor of local recurrence, and identification of an the orientation can be addressed immediately.
involved margin should prompt consideration of Achieving good hemostatis is important for
re-excision or completion mastectomy.19,20 obvious reasons. Large lumpectomy cavities can
Despite the usefulness of a microscopic margin be defined for the radiotherapist by placing
assessment, clear surgical margins under the radiopaque surgical clips at the borders of the
most stringent conditions still do not ensure local specimen. This may help facilitate the delivery of
control rates that are equivalent to those achieved a radiation therapy boost to the lumpectomy site.
with the addition of radiotherapy. In the Uppsala
Swedish trial, only patients with tumors < 20 Total Mastectomy
mm were included. Each patient underwent a
sector resection consisting of removal of a por- Total mastectomy remains an excellent choice for
tion of the skin and pectoralis fascia. Each mar- many patients with breast cancer. A clear advan-
gin was assessed twice. Any microscopic margin tage of mastectomy is the avoidance of radiation
involvement or lymph node involvement was an therapy for patients without large tumors or mul-
exclusion criterion. Patients were randomized to tiple involved lymph nodes. This has more appeal
observation or radiotherapy after sector resec- for patients that are not motivated to achieve
tion. Despite these favorable conditions and good cosmesis. Older, less mobile patients may
careful analysis of margins, local recurrence was find this preferable to the alternative of lumpec-
significantly more common in the observation tomy and radiation therapy.
arm of the study.34 Serial sectioning studies of Total mastectomy is indicated for multicen-
mastectomy specimens of patients that would be tric disease or tumors with extensive coexistent
Evaluation and Surgical Management of Stage I and II Breast Cancer 145

DCIS, where achieving a clear surgical margin peutic operation for a diagnosis that led to the
becomes difficult with a segmental mastec- genetic evaluation. Counseling these patients
tomy. It is also indicated for individuals who can be very difficult as they have to cope with
are not radiation therapy candidates, including both a diagnosis of cancer and an emotional
those with active scleroderma, history of prior decision as to whether or not they wish to
radiotherapy, ataxia telangectasia, and early undergo a bilateral mastectomy. For those
pregnancy and for those who opt for it. Excel- patients who might have greater difficulty in
lent cosmetic results can be achieved with a reaching a comfortable decision regarding a
variety of reconstructive options, which can bilateral operation, a safe approach is to pro-
occur either simultaneously or as a delayed pro- ceed with a lumpectomy and axillary staging
cedure. If a patient is contemplating recon- procedure in conjunction with genetic counsel-
struction, a skin-sparing mastectomy should be ing, with or without genetic testing. With this
performed. This operation involves the removal approach, the more important delivery of sys-
of the nipple-areolar complex and breast tissue temic therapy is not delayed. During
but differs from a standard incision in preserv- chemotherapy, the time-consuming process of
ing as much of the skin over the breast as pos- genetic testing can be performed, if indicated.
sible (Figure 9–9). If the patient is found not to carry a genetic
Most patients with early-stage breast cancer mutation, radiation therapy serves as the last
can undergo immediate reconstruction. This has step of the treatment plan. For those patients
the advantages of limiting the surgical interven- with an identified mutation, a completion mas-
tions to a single-stage procedure and providing tectomy and contralateral prophylactic mastec-
the patient with the psychological benefit of an tomy with reconstruction can be performed.
immediately reconstructed breast. Immediate Although a prophylactic mastectomy does not
reconstruction also best preserves the elasticity guarantee prevention of future breast cancer
of the elevated flaps and helps maintain the nat- events, recent data suggest that it is an effective
ural contour of the breast, including the infra- means of reducing risk.37 As an alternative,
mammary fold, which may be affected with a tamoxifen can be used for both adjuvant treat-
delayed reconstruction. Considerations for ment and chemoprevention. Although data are
delayed reconstruction include urgency to relatively early, a clear reduction in high-risk
address adjuvant systemic treatment, a patient patients was identified in a randomized study
who remains undecided regarding reconstruc- of tamoxifen users.38 If elected, compartmen-
tion options, or a patient who is likely to receive talizing treatment and prophylactic issues helps
chest wall radiation therapy. Although radiation to ease the sudden burden of complex decisions
therapy can be successfully delivered after auto- that a patient will face at the time of diagnosis.
genous reconstruction with good cosmetic
results, the incidence of capsular contraction
after radiation therapy is prohibitive in those
patients undergoing implant reconstruction.36
For patients with a strong familial history of
breast cancer, a decision may be made to com-
bine a treatment operation with a prophylactic
procedure. The identification of breast cancer
susceptibility genes has fostered this concept,
but in practical terms, it is very difficult to
assess risk and screen for a genetic mutation in Standard Skin-sparing
a timely fashion before embarking on a thera- Figure 9–9. Incision placement for mastectomy.
146 BREAST CANCER

Radical Mastectomy important reasons: staging of the breast cancer


and providing regional control. Lymph node
In the context of early breast cancer, there is vir- involvement represents the most important
tually no need to resect the pectoralis muscles variable, aside from metastatic disease, to pre-
and axillary tissue. Occasionally, tumors located dict outcome.47 This information is important
posteriorly along the chest wall may focally in defining the prognosis and in tailoring the
invade the pectoralis muscle. It should be pointed adjuvant therapy of patients with breast cancer.
out that invasion of the pectoralis muscle does Staging of the axilla represents a critical vari-
not constitute chest wall invasion and is staged able in defining the prognosis of patients pre-
according to the size of the primary tumor.4 senting with early breast cancer. In the context
Small breast cancers that present with muscle of early detection and screening mammog-
involvement are usually located peripherally or raphy, nodal involvement is at times the only
posteriorly, and extension, in part, reflects prognosticator that leads to the clear recom-
proximity to the muscle. This scenario can be mendation of chemotherapy.
safely managed with resection of a portion of Aside from providing important prognostic
the muscle as part of either a lumpectomy or a information, axillary dissection represents the
total mastectomy. With either surgical approach, most effective means of controlling regional
radiation therapy should be considered. disease.48 What constitutes an adequate axillary
dissection? This has been well established with
Management of the Axilla multiple studies analyzing the inclusion of
metastatic disease based on the arbitrary divi-
Just as treatment of the primary tumor of the sion of level I, II, and III axillary lymph
breast has evolved from a single, radical opera- nodes.49–51 On average, there is a one percent
tion for all scenarios to a more directed chance of metastatic disease in level III lymph
approach consisting of lumpectomy, the stan- nodes that would not be detected in levels I and
dard axillary dissection is quickly being II.49–51 Mathiesen demonstrated that the poten-
replaced by sentinel lymphadenectomy. Intro- tial for identifying micrometastases increased
duced by Morton and colleagues in 1992 for the till 10 lymph nodes were removed from the
treatment of melanoma,39 this technique was axilla.52 Unless extensive axillary involvement
quickly applied to breast cancer.40,41 Like lym- is recognized at the time of surgery, a level I/II
phatic mapping for other disease sites, the sen- node dissection should encompass axillary dis-
tinel lymph node is identified through the con- ease in 99 percent of patients.
stant anatomic relationship between a tumor In the presence of axillary disease, an axil-
and draining lymphatics. Conceptually, each lary dissection is an excellent operation for
specific area in the breast drains to a sentinel regional control and prognostic information.
lymph node which may be located anywhere However, for the great majority of patients with
within the axilla or internal mammary chain early breast cancer, an axillary dissection does
(Figure 9–10). Larger tumors may have more not confer any therapeutic benefit. The greatest
than one draining lymphatic (Figure 9–11). The concern, particularly for younger, active
sentinel lymph node biopsy continues to be patients is the risk of developing lymphedema.
refined and defined for patients with early This risk is directly related to the extent of the
breast cancer; in several studies, it has been axillary dissection and is further increased with
demonstrated to yield reliable correlation to an the addition of radiation therapy.53 This risk
axillary dissection.40–46 remains indefinitely for the life of the patient.
The axillary dissection has always been rec- Other potential side effects include paresthe-
ognized as an excellent procedure for two sias, loss of mobility, and cosmetic deformity.
Evaluation and Surgical Management of Stage I and II Breast Cancer 147

sensitive enough to exclude the presence of axil-


lary disease.57 It is unlikely that any imaging
technology will compare with the sensitivity of a
microscopic examination, which has the poten-
tial to identify single metastatic cells. The short-
coming of a standard axillary dissection is that
pathologists are incapable of reviewing every cell
of every lymph node. The ability to detect micro-
metastases is directly related to the intensity with
which a lymph node is analyzed. Serial section-
ing studies have identified a higher incidence of
true nodal positivity and mortality in those with
unrecognized micrometastases.23,58 Outside of
Figure 9–10. Breast lymphatic drainage patterns. investigational studies, serial sectioning of axil-
lary dissection specimens is impractical.
Avoiding the side effects of an axillary dis- Sentinel lymphadenectomy has several con-
section in a substantial number of patients who ceptual advantages over standard axillary dissec-
do not achieve any therapeutic benefit has been tion. Most significant to the patient is that the
a major impetus in identifying an alternative risk of long term complications is virtually elim-
means of staging the axilla. Other methods that inated by avoiding an extensive axillary dissec-
have been evaluated to replace axillary dissec- tion. Recovery is much shorter, and for most,
tion have been imaging studies including ultra- BPT, under these circumstances, can be accom-
sonography, computed tomography, and scintig- plished as an outpatient procedure. Compared to
raphy. All these techniques have had the same a standard axillary incision, the sentinel lymph
major limitation of an unacceptably high false- node can be removed through a smaller incision
negative rate.54–56 Positron emission tomog- with transcutaneous localization of the node
raphic (PET) scanning has emerged as a more with a hand held gamma probe (Figure 9–12). In
sensitive test that relies on the metabolic differ- addition to the reduction in morbidity, sentinel
ences of tumors rather than on anatomic changes. lymphadenectomy provides the pathologist with
To date, this test shows promise but is still not the opportunity to perform a much more com-
prehensive analysis of the specimen, given the
more limited material to analyze. Although diffi-

Figure 9–11. Lymphatic drainage pattern of an upper breast Figure 9–12. Transcutaneous localization of axillary sentinel
tumor. lymph node.
148 BREAST CANCER

cult to prove, this may, in fact, provide a more nique to most accurately identify the sentinel
sensitive means of staging the axilla, provided node. Variables to be defined include the loca-
the sentinel node is correctly identified. tion of the injection, the radiopharmaceutical
The immediate question addressed by pre- compound and optimal size, the time interval
liminary studies has been the ability of sentinel between the injection and the surgical proce-
lymphadenectomy to detect micrometastases as dure, the combination of radiocolloid with iso-
efficiently as an axillary dissection can. As a sulfan blue, size limitations of the tumor, effect
result, comparative studies have included both of excisional biopsy on the accuracy of sentinel
operations in the same patient. Although the end node localization, and the microscopic and sub-
points for these studies have all been the same, microscopic evaluation of nodal tissue. The
the technique has varied widely. Conceptually, a long-term regional recurrence risk after a sen-
visual tracer, a radiolabeled protein, or a combi- tinel lymph node biopsy has yet to be addressed.
nation of both are injected around the breast Until these questions have been answered, sen-
tumor. Initially, this process was performed with tinel lymphadenectomy should be performed as
isosulfan blue, a vital blue dye used for lym- a protocol.
phatic mapping for melanoma. This approach
created difficulties in defining the breast lym- Adjuvant Therapy
phatics because of the three-dimensional nature The surgical treatment for early breast cancer
of breast cancers and the potential for missing serves as a very important therapeutic step but
multiple sentinel nodes located within the three- also provides important prognostic information
dimensional axillary nodal basin. Because of the to define subsequent adjuvant therapy deci-
brevity of time in which isosulfan blue migrates sions. Adjuvant chemotherapy has evolved from
through the sentinel node, the timely identifica- a narrowly defined node-positive indication to
tion of multiple sentinel nodes is difficult. more encompassing indications based on identi-
The introduction of radiocolloid for this tech- fied benefits. Nonetheless, the efficacy of com-
nique greatly enhanced this procedure by provid- bination chemotherapy is well correlated with
ing a second means of localizing a sentinel node nodal involvement, and surgical staging, partic-
using a hand-held gamma probe. This has simpli- ularly for early breast cancer, is important in
fied the procedure by obviating the lymphatic defining optimal adjuvant therapy decisions.
mapping required to identify the blue lymph
node. Additionally, technitium-labeled sulfur col-
loid binds to lymphatic tissue and provides a
much greater window of opportunity to localize
sentinel lymph nodes. Lymphoscintigraphy done
prior to the surgical procedure can assist in con-
firming the migration and location of radiocol-
loid (Figure 9–13). Another conceptual benefit of
the use of radiocolloid is that it makes it possible
to localize internal mammary nodes. For tumors
located medially in the breast, these may be the
only sentinel nodes for the tumor.
Although data are quickly emerging to sup-
port sentinel lymphadenectomy in the staging of
breast cancer, it has not yet become uniformly
accepted as the standard of care. A question yet
Figure 9–13. Lymphscintigram of breast pri-
to be answered is the optimization of the tech- mary and axillary sentinel lymph nodes.
Evaluation and Surgical Management of Stage I and II Breast Cancer 149

RECURRENCE limited and tailored approach. Diagnostic eval-


uation also continues to improve in defining the
An ipsilateral breast recurrence after BPT may be extent of the disease and in providing accurate
difficult to distinguish from a second primary staging information to guide resection and
tumor. Information important to help make this adjuvant therapy. With these strategies, the
distinction includes the location of the recurrence treatment of breast cancer has become more
relative to the initial primary tumor, the histo- precise and effective.
logic features, and the disease-free interval. Prox-
imity to a previous lumpectomy site increases the REFERENCES
likelihood of the tumor being a recurrence. In the
1. Shin HJ, Sneige N. Is a diagnosis of infiltrating
absence of any previous history of in situ carci-
versus in situ ductal carcinoma of the breast
noma, an in situ component is suggestive of a possible in fine-needle aspiration specimens?
new primary lesion. Local recurrences are most Cancer 1998;84:186–91.
likely to occur within several years of initial 2. Shabot M, Goldberg I, Schick P, et al. Aspiration
treatment. A long disease-free interval is more cytology is superior to Tru-Cut needle biopsy
suggestive of a second primary tumor. The dis- in establishing the diagnosis of clinically sus-
picious breast masses. Ann Surg 1982;196:
tinction between a new primary tumor and recur-
122–26.
rent breast cancer is more important in under- 3. Ballo SM, Sneige N. Can core needle biopsy
standing the biology of the disease and the replace fine-needle aspiration cytology in the
efficacy of the treatment selected. In practical diagnosis of palpable breast carcinoma? A
terms, an ipsilateral event is managed in a simi- comparative study of 124 women. Cancer 1996;
lar fashion with either scenario. Without the 78:773–7.
4. American Joint Committee on Cancer. Breast. In:
ability to deliver additional radiotherapy in most
Fleming ID, Cooper JS, Henson DE, et al. edi-
patients, re-excision is not a safe option, and tor. AJCC Cancer Staging Manual. 5th Edition.
most patients are treated with a completion mas- Philadelphia: J. B. Lippincott; 1997. pp. 171–80.
tectomy. An ipsilateral event should also lead to 5. Winchester DJ, Menck HR, Winchester DP.
a metastatic evaluation; recurrences are com- National treatment trends for ductal carcinoma
monly the harbinger of metastatic disease.59 in situ of the breast. Arch Surg 1997;132(6):
660–5.
In patients previously treated with mastec-
6. Sarrazin D, Le M, Rouesse J, et al. Conservative
tomy, a chest wall recurrence can usually be treatment versus mastectomy in breast cancer
managed with a local excision. The goal of exci- tumors with macroscopic diameter of 20 mil-
sion should be a margin-free resection. In some limeters or less. Cancer 1984;53(5):1209–13.
situations, this may necessitate resection of 7. Fisher B, Anderson S, Redmond CK, et al.
muscle or a portion of the chest wall. In patients Reanalysis and results after 12 years of follow-
up in a randomized clinical trial comparing
who have undergone previous radiation treat- total mastectomy with lumpectomy with or
ments, a more extensive resection may represent without irradiation in the treatment of breast
the only therapeutic option to achieve local con- cancer. N Engl J Med 1995;333(22):1456–61.
trol. Closure of the chest wall defect may be 8. Veronesi U, Banfi A, Del Vecchio M, et al. Com-
facilitated by a myocutaneous flap closure. Hor- parison of Halsted mastectomy with quadran-
tectomy, axillary dissection, and radiotherapy
monal and cytotoxic chemotherapy must also be
in early breast cancer: long term results. Eur J
considered at the time of a local recurrence. Can Clin Oncol 1986;22(9):1085–9.
9. Van Dongen J, Bartelink H, Fentimen I, et al. Ran-
CONCLUSION domized clinical trial to assess the value of
breast-conserving therapy in stage I and II
Local and regional surgical treatment of early breast cancer, EORTC 10801 trial. J Natl Can-
breast cancer continues to evolve toward a more cer Inst Monogr 1992;11:15–8.
150 BREAST CANCER

10. Blichert-Toft M, Rose C, Anderson JA, et al. Dan- graphic features of 455 invasive lobular carci-
ish randomized trial comparing breast conser- nomas. Radiology 1992;185(3):705–8.
vation therapy whit mastectomy: six years of 23. Clare SE, Sener SF, Wilkens W, et al. Prognostic
life table analysis. J Natl Cancer Inst Monogr significance of occult lymph node metastases
1992;11:19–25. in node-negative breast cancer. Ann Surg
11. Lichter A, Lippman M, Danforth D, et al. Mastec- Oncol 1997;4(6):447–51.
tomy versus breast conserving therapy in the 24. Weiss MC, Fowble BL, Solin LJ, et al. Outcome
treatment of Stage I and II carcinoma of the of conservative therapy for invasive breast can-
breast: a randomized trial at the National Can- cer by histologic subtype. Int J Radiat Oncol,
cer Institute. J Clin Oncol 1992;10:976–83. Biol, Phys 1992;23(5):941–7.
12. Winchester DJ, Menck HR, Winchester DP. The 25. Poen JC, Tran L, Juillard G, et al. Conservation
National Cancer Data Base report on the therapy for invasive lobular carcinoma of the
results of a large nonrandomized comparison breast. Cancer 1992;69(11):2789–95.
of breast preservation and modified radical 26. White JR, Gustafson GS, Wimbish K, et al. Con-
mastectomy. Cancer 1997; 80:162–7. servative surgery and radiation therapy for
13. Early Breast Cancer Trialist’s Collaborative Group. infiltrating lobular carcinoma of the breast.
Effects of radiotherapy and surgery in early breast The role of preoperative mammograms in
cancer. N Engl J Med 1995;333:1444–55. guiding treatment. Cancer 1994;74(2):640–7.
14. Fisher B, Bryant J, Wolmark N, et al. Effect of 27. du Toit RS, Locker AP, Ellis IO, et al. An evalua-
preoperative chemotherapy on the outcome of tion of differences in prognosis, recurrence
women with operable breast cancer. J Clin patterns and receptor status between invasive
Oncol 1998;16:2672–85. lobular and invasive carcinomas of the breast.
15. Fisher B, Brown A, Mamounas E, et al. Effect of Eur J Surg Oncol 1991;17(3):251–7.
preoperative chemotherapy on local-regional 28. Silverstein MJ, Lewinsky BS, Waisman JR, et al.
disease in women with operable breast cancer: Infiltrating lobular carcinoma. Is it different
findings from NSABP B-18. J Clin Oncol from infiltrating duct carcinoma? Cancer
1997;15:2483–93. 1994;73(6):1673–77.
16. Olivotto IA, Rose MA, Osteen RT, et al. Late cos- 29. Mate TP, Carter D, Fischer DB, et al. A clinical
metic outcome after conservative surgery and and histopathologic analysis of the results of
radiotherapy: analysis of causes of cosmetic conservation surgery and radiation therapy in
failure. Int J Radiat Oncol Biol Phys 1989;1: stage I and II breast carcinoma. Cancer 1986;
747–53. 58(9):1995–2002.
17. Yeatman TJ, Cantor AB, Smith TJ, et al. Tumor 30. Winchester DJ, Chang HR, Graves TA, et al. A
biology of infiltrating lobular carcinoma. comparative analysis of lobular carcinoma of
Implications for management. Annals of Surg the breast: presentation, treatment, and out-
1995;222(4):549–59. come. J Am Coll Surg 1998;186(4):416–22.
18. Gage I, Schnitt SJ, Nixon AJ, et al. Pathologic 31. Fisher ER, Sass R, Fisher B, et al. Pathologic find-
margin involvement and the risk of recurrence ings from the National Surgical Adjuvant
in patients treated with breast conserving Breast Project (protocol 6). II. Relation of local
surgery. Cancer 1996;78:1921–8. breast recurrence to multicentricity. Cancer
19. Smitt MC, Nowels KW, Zdeblick MJ, et al. The 1986;57(9):1717–24.
importance of the lumpectomy surgical margin 32. Veronesi U, Volterrani F, Luini A, et al. Quadran-
status in long term results of breast conserva- tectomy versus lumpectomy for small size
tion. Cancer 1995;76:259–67. breast cancer. Eur J Cancer 1990;26:671–3.
20. Anscher M, Jones P, Prosnitz L, et al. Local fail- 33. Veronesi U, Luini A, Del Vecchio M, et al. Radio-
ure and margin status in early-stage breast car- therapy after breast preserving surgery in
cinoma treated with conservative surgery and women with localized cancer of the breast. N
radiation therapy. Ann Surg 1993;218:22–8. Engl J Med 1993;328:1587–91.
21. Krecke KN, Gisvold JJ. Invasive lobular carcinoma 34. Liljegren G, Holmberg L, Adami HO, et al. Sector
of the breast: mammographic findings and resection with or without postoperative radio-
extent of disease at diagnosis in 184 patients. therapy for stage 1 breast cancer. Five year
AJR Am J Roentgenol 1993;161(5):957–60. results of a clinical trial. J Natl Cancer Inst
22. Le Gal M, Ollivier L, Asselain B, et al. Mammo- 1994;86:717–22.
Evaluation and Surgical Management of Stage I and II Breast Cancer 151

35. Holland R, Veling S, Mravunac M, et al. Histologic egory and operable breast cancer prognosis.
multifocality of Tis, T1–2 breast carcinomas: Tumori 1989;75:18–22.
implication for clinical trials of breast conserv- 48. Fisher B, Redmond C, Fisher ER, et al. Ten-year
ing treatment. Cancer 1985;56:979–90. results of a randomized clinical trial comparing
36. Schuster RH, Kuske Rb, Young VL, Fineberg B. radical mastectomy and total mastectomy with
Breast reconstruction in women treated with or without radiation. N Engl J Med 1985;312:
radiation therapy for breast cancer: cosmesis, 674–81.
complications, and tumor control. Plast Recon- 49. Rosen P, Martin M, Kinne D, et al. Discontinuous
str Surg 1992;90:445–52. or “skip”: metastases in breast carcinoma:
37. Hartmann LC, Schaid DJ, Woods JE, et al. Effi- analysis of 1228 axillary dissections. Ann Surg
cacy of bilateral prophylactic mastectomy in 1983;197:276.
women with a family history of breast cancer. 50. Veronesi U, Rilke F, Luini A, et al. Distribution of
N Engl J Med 1999;340:77–84. axillary node metastases by level of invasion.
38. Fisher B, Joseph P, Constantino D, et al. Tamox- Cancer 1987;59:682–7.
ifen for prevention of breast cancer: report of 51. Boova R, Bonanni R, Rosato F. Patterns of axil-
the National Surgical Adjuvant Breast and lary nodal involvement in breast cancer. Ann
Bowel Project P-1 study. J Natl Cancer Inst Surg 1982;196:642–4.
1998;90:1371–88. 52. Mathiesen O, Carl J, Bonderup O, Panduro J. Axil-
39. Morton DL, Wen DR, Wong JH, et al. Technical lary sampling and the risk of erroneous staging
details of intraoperative lymphatic mapping for of breast cancer. An analysis of 960 consecutive
early stage melanoma. Arch Surg 1992;127: patients. Acta Oncol 1990;29:721–5.
392–9. 53. Larson D, Weinstein M, Goldberg I, et al. Edema of
40. Krag DN, Weaver DL, Alex JC, Fairbank JT. Sur- the arm as a function of the extent of axillary
gical resection and radiolocalization of the sen- surgery in patients with Stage 1 and 2 carcinoma
tinel lymph node in breast cancer using a of the breast treated with primary radiotherapy.
gamma probe. Surg Oncol 1993;2:335–9. Int J Radiat Oncol Biol Phys 1986;12:1575–82.
41. Giuliano AE, Kirgan DM, Guenther JM, Morton 54. Tate JJ, Lewis V, Archer T, et al. Ultrasound detec-
DL. Lymphatic mapping and sentinel lym- tion of axillary lymph node metastases in breast
phadenectomy for breast cancer. Ann Surg cancer. Eur J Surg Oncol 1989;15:139–41.
1994;3:391–8. 55. March DE, Wechsler RJ, Kurtz AB, et al. Ct-
42. Albertini JJ, Lyman GH, Cox C, et al. Lymphatic pathologic correlation of axillary lymph nodes
mapping and sentinel node biopsy in the in breast cancer. J Comput Assist Tomogr
patient with breast cancer. JAMA, 1996;276: 1991;15:440–4.
1818–22. 56. Kao Ch, Wang SJ, Yeh SH. Technetium-99m MIBI
43. Veronesi U, Paganelli, Galimberti V, et al. Sen- uptake in breast carcinoma and axillary lymph
tinel-node biopsy to avoid axillary dissection node metastases. Clin Nucl Med 1994;19:
in breast cancer with clinically negative lymph- 898–900.
nodes. Lancet 1997;349:1864–7. 57. Avril N, Dose J, Janicke F, et al. Assessment of
44. Krag D, Weaver D, Ashikaga T, et al. The sentinel axillary lymph node involvement in breast can-
node in breast cancer: a multicenter validation cer patients with positron emission tomog-
study. N Engl J Med 1998;339:941–6. raphy using radiolabeled 2–(fluorine-18)-flu-
45. Guiliano AE, Jones RC, Brennan M, Statman R. oro-2–deoxy-D-glucose. J Natl Cancer Inst
Sentinel lymphadenctomy in breast cancer. J 1996;88:1204–9.
Clin Oncol 1997;15:(6):2345–50. 58. International (Ludwig) Breast Cancer Study
46. Winchester DJ, Sener SF, Winchester DP, et al. Group. Prognostic importance of occult axil-
Sentinel lymphadenectomy for breast cancer: lary lymph node metastases from breast can-
Experience with 180 consecutive patients: effi- cers. Lancet 1990;335:1565–8.
cacy of filtered technetium 99m sulphur col- 59. Bedwinek J, Fineberg B, Lee J, et al. Analysis of
loid with overnight migration time. J Am Coll failures following local treatment of isolated
Surg 1999;188:597–603. local-regional recurrence of breast cancer. Int J
47. Ciatto S, Cecchini S, Iossa A, Grazzini G. “T” cat- Radiat Oncol Biol Phys 1981;7:581–5.
10
Locally Advanced Breast Cancer
S. EVA SINGLETARY, MD, FACS

Integration of systemic chemotherapy and/or selected patients with LABC, and recent inno-
hormonal therapy with surgery and irradiation vations in systemic therapy.
is considered the standard of care in the treat-
ment of locally advanced breast cancer DEFINITION OF LOCALLY ADVANCED
(LABC). Because the greatest risk for patients BREAST CANCER
with LABC is the development of distant
metastases and subsequent death, the goals of Locally advanced breast cancer generally refers
surgery are to provide maximal locoregional to large primary tumors (> 5 cm) associated
control with minimal disfigurement and to per- with skin or chest-wall involvement or with
mit accurate staging to determine prognosis. fixed (matted) axillary lymph nodes (T3/T4;
Breast conservation surgery is sometimes pos- N2/N3).1 In the most recent TNM staging sys-
sible after tumor downstaging with induction tem,1 tumors associated with disease in the ipsi-
chemotherapy, but close cooperation between lateral supraclavicular nodal basin have been
the medical and surgical oncologists and the eliminated from the LABC category because
radiation therapist is required to determine the the supraclavicular basin lies outside the pri-
feasibility of this option. Similarly, the surgeon mary lymphatic drainage pathways of the axilla
must be familiar with the natural history of and internal mammary nodes; tumors associ-
LABC to assess the advisability of major resec- ated with supraclavicular disease have been
tions of either persistent advanced primary reclassified as stage IV disease. However, as
disease or locoregional recurrences. If life patients with distant metastases confined to
expectancy is very short, as is the case with supraclavicular nodes have a better prognosis
patients who have bulky visceral disease or than patients with metastases at other distant
metastases nonrespondent to multiple chemo- sites and can be rendered disease free with
therapy regimens, the true benefit of a complex locoregional therapy,2 metastases limited to the
but technically feasible operation should be ipsilateral sub- or supraclavicular fossa will be
evaluated carefully. However, in selected included in the definition of LABC offered
patients with advanced disease, surgery may here. Large primary tumors (> 5 cm) with no
achieve quality palliation of local symptoms of evidence of nodal involvement (T3;N0) have a
pain, hemorrhage, and malodorous ulceration. more favorable prognosis than LABC, with a 5-
This chapter defines LABC and addresses year survival rate of 70 to 80 percent; thus, in
the role of surgery after tumor downstaging the most recent TNM staging system, T3N0
with induction chemotherapy, the use of mas- lesions have been reclassified as stage IIB dis-
tectomy for inflammatory breast cancer, the ease. However, as most series have classified
feasibility of immediate reconstruction in T3N0 lesions as LABC for the purposes of

153
154 BREAST CANCER

treatment, these tumors will also be included in complete response (no evidence of tumor by
the present definition of LABC. physical or radiographic examination). Seventy-
one percent had a partial response (³ 50 percent
ROLE OF SURGERY AFTER tumor shrinkage). Only 10 percent had a minor
INDUCTION CHEMOTHERAPY or no significant response to induction chemo-
therapy. Tumor progression occurred in 2 per-
Since the mid-1970s, patients with LABC cent of patients. This trial demonstrated that the
treated at The University of Texas M. D. Ander- majority of patients will have significant tumor
son Cancer Center have received three to four shrinkage with induction chemotherapy and
cycles of doxorubicin-based combination that the likelihood of tumor progression is low.
chemotherapy prior to local therapy; local ther- As the virulence of a tumor is associated with
apy is followed by the completion of systemic chemoresistance, tumors that progress during
therapy and irradiation. Between 1974 and aggressive chemotherapy are unlikely to be
1996, patients with LABC were treated in four controlled with surgery, and a crossover
trials addressing four major concerns about the chemotherapy regimen should be considered.
use of induction chemotherapy: (1) whether This study also confirmed that induction
tumor progression will occur during induction chemotherapy is well tolerated and that surgical
chemotherapy, rendering the tumor unre- procedures after induction chemotherapy can
sectable even with radical surgery; (2) whether be completed without an increased rate of
operative morbidity is increased after induction infection or delayed wound healing.4
chemotherapy; (3) whether the histologic stag- The above trial refuted the concept that histo-
ing information obtained from the surgical logic staging information obtained after induc-
specimen after induction chemotherapy main- tion chemotherapy would not have predictive
tains its prognostic correlations with survival; power. The histologically confirmed response in
(4) and whether breast conservation therapy the mastectomy specimen after induction
with or without an axillary node dissection is chemotherapy was an excellent prognostic fac-
feasible and safe in patients with LABC. tor for survival and was more accurate than
In the first clinical trial at M. D. Anderson clinical assessment of response.5,6 The number
Cancer Center (1974 to 1985), induction combi- of positive axillary nodes after induction
nation chemotherapy was administered to 174 chemotherapy also remained prognostic for
evaluable patients (191 registered) with nonin- survival: actuarial 5-year survival rates were 70
flammatory stage III breast cancer.3 After three percent for patients with negative lymph nodes,
cycles of 5-fluorouracil, doxorubicin, and 62 percent for patients with one to three posi-
cyclophosphamide (FAC), patients with an tive lymph nodes, 47 percent for patients with
excellent tumor response underwent irradiation four to ten positive lymph nodes, and 21 per-
of the chest wall and regional lymph nodes. cent for patients with more than ten positive
Patients with a substantial volume of residual lymph nodes.6 The 5-year disease-free survival
tumor underwent mastectomy and irradiation. rates were 72, 46, 35, and 6 percent, respec-
After completion of locoregional therapy, FAC tively. When the subsets of patients with four or
was reinitiated and continued until a dose of 450 more positive lymph nodes were combined, the
mg/m2 of doxorubicin was reached. Then treat- overall survival rate at 5 years was 38 percent,
ment with cyclophosphamide, methotrexate, and the disease-free survival rate dropped to
and 5-fluorouracil (CMF) was instituted and only 20 percent. As patients with four or more
continued for a total treatment period of 2 years. positive lymph nodes after induction chemo-
After the three cycles of induction chemo- therapy have a survival rate similar to that
therapy with FAC, 17 percent of patients had a obtained in historical trials of mastectomy and
Locally Advanced Breast Cancer 155

postoperative irradiation without systemic ther- ond-line therapy. Significantly, there was exten-
apy,2,7 these patients should be considered for sive downstaging in a large proportion of
innovative clinical trials. patients in the study: 17 mastectomy specimens
The second M. D. Anderson clinical trial had no evidence of residual tumor, and 54 mas-
(1985 to 1989) was designed to determine tectomy specimens had < 1 cm3 of residual
whether the extent of residual disease in the tumor. This finding led us to consider the pos-
mastectomy specimen after induction chemo- sibility of performing breast conservation
therapy can be used as a guide in planning post- surgery for locally advanced disease.
operative adjuvant therapy. Three cycles of vin- In a retrospective review of the mastectomy
cristine, doxorubicin, cyclophosphamide, and specimens in which the tumor shrank by ³ 50
prednisone (VACP) were administered at 21- percent with induction chemotherapy, the fac-
day intervals, then a modified radical mastec- tors most commonly associated with multiple-
tomy was performed. Patients with histologi- quadrant involvement that would exclude
cally confirmed complete remission and those breast conservation surgery were demonstrated
with < 1 cm3 of residual tumor received five to be persistent skin edema, residual tumor size
additional cycles of VACP; those with no > 4 cm, extensive intramammary lymphatic
response to induction chemotherapy were invasion, and known mammographic evidence
crossed over to receive five cycles of methotrex- of multicentric disease.8
ate, 5-fluorouracil, and vinblastine (MFVb). The objective in the third M. D. Anderson
Patients with partial response and ³ 1 cm3 or clinical trial (1989 to 1992) was to determine
more of residual tumor were randomly assigned prospectively what fraction of patients with
to receive five additional cycles of either VACP LABC may be candidates for breast conserva-
or MFVb. All patients received radiation to the tion surgery after induction chemotherapy.9 Of
chest wall and regional lymph nodes. Eight 203 evaluable patients with LABC who com-
patients whose tumors remained inoperable pleted four cycles of induction chemotherapy
after induction chemotherapy underwent irradi- with (FAC), 51 (25 %) elected and underwent
ation before mastectomy and MFVb. Irradiation breast conservation surgery (Figure 10–1). The
had a minimal effect on wound healing provided breast preservation rate for patients with ulcer-
wound tension and thin skin flaps were avoided. ative lesions or dermal lymphatic involvement
If mastectomy resulted in a large defect, flap (stage IIIB) was only 6 percent. With a median
coverage consisting of healthy autogenous tis- follow-up of > 60 months, only 5 (ten %) of the
sue was preferred to the use of skin grafts. 51 patients who underwent breast conservation
Of 193 evaluable patients in this second trial surgery had relapses in the breast.
(200 registered), 161 had a partial or greater In the fourth M. D. Anderson clinical trial
clinical response to the three cycles of induc- (1992 to 1996), the objective was to determine
tion chemotherapy. Among the patients with a if high-dose chemotherapy would increase the
partial response, no statistically significant dif- extent of tumor downstaging with induction
ference (p = .64) was detected in the 4-year sur- chemotherapy and allow more patients the
vival rates for the MFVb and VACP groups (75 option of breast conservation surgery. One hun-
and 58%, respectively).8 Of the 32 patients in dred and seventy patients with LABC were ran-
this study whose tumors showed a minor or no domly assigned to receive either four cycles of
response to the induction chemotherapy, only standard FAC (1000 mg/m2 5-fluorouracil, 50
16 remain alive and only 8 are disease-free at mg/m2 doxorubicin, and 500 mg/m2 cyclophos-
the time of writing. The lack of impact on sur- phamide) at 21-day intervals or dose-intensive
vival of the crossover regimen in this study was FAC (1200, 60, and 1000 mg/m2 of the three
probably due to the absence of an effective sec- drugs, respectively) at 18-day intervals with
156 BREAST CANCER

A B
Figure 10–1. A, Patient with locally advanced breast cancer who desired breast conservation therapy. B, After four cycles of
induction chemotherapy, a segmental mastectomy and axillary node dissection were performed. The patient then completed
chemotherapy followed by irradiation of the breast and regional nodal basins.

prophylactic subcutaneous administration of treated with FAC alone (p = .963). Although a


recombinant human granulocyte colony-stimu- higher percentage of patients underwent breast
lating factor (G-CSF). After surgery, patients conservation therapy in the group that received
with < 1 cm3 of residual tumor received four FAC plus G-CSF (42 v 29 %), this difference
additional cycles of FAC or dose-intensive did not achieve statistical significance.
FAC. Patients with a clinical partial response To determine if there may be an alternative
but with > 1 cm3 of residual tumor and those to axillary node dissection after tumor down-
with four or more positive lymph nodes in the staging, we analyzed 147 consecutive patients
surgical specimen were treated postoperatively in the FAC versus high-dose FAC study who
with four more cycles of FAC or dose-intensive had both physical and ultrasound examinations
FAC followed by four cycles of methotrexate of the axilla at diagnosis and prior to surgery10
and vinblastine. Patients with no change or pro- (Figure 10–2). Of the 133 patients with pal-
gression of disease received six cycles of pable axillary disease on initial examination, 43
methotrexate and vinblastine. In all patients, patients (32%) were downstaged to a negative
locoregional radiotherapy was instituted within axilla as assessed by physical and ultrasound
6 weeks of completion of chemotherapy. examination following induction chemother-
One hundred and sixty-six patients were apy. There was a pathologic complete axillary
evaluable for response. Patients who received lymph node response found in 30 patients
FAC plus G-CSF were more likely to have a (23%). Of the 72 patients with axillary metas-
complete or partial clinical response compared tases that were cytologically proven by fine-
with patients who received standard FAC (84 v needle aspiration on initial evaluation, 15
66%). However, the two regimens produced (21%) were confirmed to have histologically
similar results in terms of histologic downstag- negative axillary lymph nodes following induc-
ing of the primary tumor. There was a complete tion chemotherapy. Of the 28 patients in whom
histologic response (no tumor present) seen in the axilla became clinically negative but the
25 percent of patients treated with FAC plus G- findings on axillary ultrasound remained posi-
CSF and in 16 percent of patients treated with tive after induction chemotherapy, 21 (75%)
FAC alone (p = .155). There was a near-com- were found to have macroscopic axillary nodal
plete histologic response (< 1 cm3 of tumor pre- disease upon dissection. When both the physi-
sent) seen in 25 percent of patients treated with cal and ultrasound examination were negative
FAC plus G-CSF and in 24 percent of patients following induction chemotherapy, 53 percent
Locally Advanced Breast Cancer 157

of patients (29 of 55) were found to still have fossa. Preliminary analysis based on 78 evalu-
axillary nodal metastases. However, 96 percent able patients (104 patients registered at the time
(25 of 26) had only 2 to 5 mm foci of disease. of the analysis) who had completed the induc-
On the basis of our analysis of the FAC ver- tion chemotherapy and surgery showed that
sus high-dose FAC study, we are currently con- paclitaxel and FAC have a similar ability to
ducting a clinical trial to assess whether patients downstage both the primary tumor and the axil-
with a negative axilla by physical and ultra- lary nodal disease.11 No or minimal residual dis-
sound examination can be safely treated without ease was found in the breast in 41 percent of the
axillary node dissection. In this clinical trial, 41 patients on the FAC treatment arm, com-
patients with T2-3, N0-1 breast cancer are ini- pared to 32 percent of the 37 patients who
tially randomized to receive four cycles of either received induction chemotherapy with pacli-
paclitaxel or standard FAC preoperatively.11 taxel (p = .44). Sixty-nine patients underwent an
After the completion of induction chemother- axillary node dissection. Negative or < 4 posi-
apy, patients who have become candidates for tive lymph nodes were found in 68 percent of
breast conservation surgery and who have clini- the 41 patients who received FAC chemother-
cally negative axilla are further randomly apy, compared to 77 percent of the 37 patients
assigned to either irradiation of the axilla or a treated with paclitaxel (p = .75). In the eight
standard level I and II axillary lymph node dis- patients who did not undergo an axillary node
section. After completion of four cycles of post- dissection, no axillary recurrences had been
operative FAC, irradiation is delivered to the detected in a 24-month follow-up period.
breast and, in patients with a nondissected How safe is conservative surgery for LABC
axilla, the lower axilla and the supraclavicular in terms of long-term local control? In review

A B

Figure 10–2. A, Normal fat-replaced axillary lymph node. The sonogram shows a large node completely replaced by echogenic
fat with a thin hypoechoic rim outlining the periphery of the node. B, Hypoechoic metastatic foci in an axillary lymph node
confirmed by ultrasound-guided fine needle aspiration.
158 BREAST CANCER

of our database of all patients treated at M. D. This survival rate is similar to the overall sur-
Anderson with breast conservation therapy vival rate of 89 percent for the entire group of
(patients with early-stage breast cancer and 93 patients (median follow-up of 73 months).
those with LABC), 949 patients were found to The local recurrence rate in our selected series
have been treated with breast conservation of breast conservation therapy for LABC was
surgery at our institution between 1982 and similar to the local failure rate observed for
1994.12 Of this group, 93 patients received breast conservation therapy in our patients with
induction chemotherapy prior to surgery on or early-stage breast cancer and was also consis-
off protocol. The initial stage distribution of tent with the experience of other investigators
these 93 patients was as follows: stage IIA, 22.6 (Table 10–1).13,15–20 The results of our study also
percent; stage IIB, 24.7 percent; stage IIIA, indicate that most patients with local recurrence
32.3 percent; stage IIIB, 16.1 percent; and stage can be treated without an adverse effect on over-
IV (supraclavicular lymph node metastases all survival.
only), 4.3 percent. In most patients (88%), The role of axillary node dissection after
induction chemotherapy consisted of FAC or induction chemotherapy in patients with LABC
high-dose FAC for three to five cycles. After has become controversial. There are four main
segmental mastectomy and axillary node dis- arguments against the routine use of axillary
section, patients underwent four to eight cycles node dissection for LABC. First, induction
of chemotherapy followed by radiotherapy. chemotherapy in patients with LABC and oper-
Breast irradiation consisted of 50 Gy of exter- able breast cancer has been shown to down-
nal-beam radiation to the intact breast and a 10 stage positive axillary lymph nodes to negative
to 15 Gy boost to the segmental mastectomy nodes in 23 to 44 percent of patients.6,10,14,20,21
site, which had been marked intraoperatively Second, in most treatment protocols, patients
with clips. Of the 93 patients, 86 completed with LABC routinely receive additional post-
postoperative therapy. Two patients refused operative chemotherapy and radiotherapy
radiotherapy after chemotherapy, four patients regardless of the findings at axillary node dis-
refused chemotherapy but did receive radio- section. Third, some LABC series have
therapy, and one patient refused all postopera- reported that axillary node dissection alone,
tive therapy. axillary irradiation alone, or a combination of
Overall, nine patients had a local recurrence, surgery and irradiation produce equivalent axil-
for a local failure rate of 9.7 percent. In six lary control rates after induction chemother-
patients, local recurrence was the first site of apy.22–24 The fourth argument is somewhat more
relapse; recurrence was in the breast paren- complex. There has been a survival advantage
chyma in three patients, in the skin of the breast suggested for high-dose chemotherapy over
in two patients, and in the breast parenchyma standard anthracycline-based chemotherapy in
and an axillary node in one patient. The median patients with multiple positive axillary nodes
time to local recurrence in these six patients was after induction chemotherapy.25,26 However,
55 months. Three patients had local recurrence high-dose chemotherapy off-protocol cannot be
after the development of distant metastases. In recommended in the absence of prospective
the nine patients with local recurrence, the sur- randomized data demonstrating such a survival
gical margin of the segmental mastectomy spec- benefit. One of the lessons learned from the
imens was negative in all patients but close in high-dose chemotherapy protocols for metasta-
three patients. The six patients with local recur- tic breast cancer was that patients with previ-
rence only or local recurrence prior to distant ously demonstrated resistance to chemotherapy
metastases had an overall survival rate of 83 usually do not benefit from this procedure.27 In
percent at a median follow-up of 88 months. addition, proponents of axillary node dissection
Locally Advanced Breast Cancer 159

in patients receiving induction chemotherapy Table 10–1. RATES OF BREAST CONSERVATION


THERAPY AND SUBSEQUENT LOCAL
assert that the number of positive nodes RECURRENCE AFTER INDUCTION CHEMOTHERAPY
detected after tumor downstaging may affect
Percent Percent Local
whether patients should be crossed over to a dif- Author Patients Stage BCT Recurrence
ferent chemotherapeutic agent or be given high-
Bonadonna14 157 II/III 81 1
dose chemotherapy. For example, phase II trials
Calais15 158 II/III 49 8
have demonstrated high activity of taxane-based Veronesi16 226 II/III 90 6
chemotherapy (paclitaxel and docetaxel) in Schwartz17 160 II/III 34 2
Touboul*18 97 II/IV 62 16
anthracycline-resistant breast cancer.28,29 How- Merajver*19 89 III 28 14
ever, if the trend in therapy is toward a sequen- Fisher20 747 I/II 68 8
Peoples12 93 II/III/IV N/A 10
tial or combined approach using anthracycline
and taxane-based regimens prior to local ther- *Local therapy consisted of primary radiation therapy.
BCT = breast conservation therapy
apy, with no further systemic intervention Adapted from Hunt KK, Buzdar AU. Breast conservation after tumor
planned, then the histologic assessment of the downstaging with induction chemotherapy. In: Singletary SE, editor.
Breast cancer—M. D. Anderson Solid Tumor Oncology Series.
axilla becomes only a prognostic tool. New York: Springer-Verlag; 1999. p. 196–207.
Whether chemotherapy can substitute for
surgery for local control of occult axillary
metastases is still unknown. Data concerning patients who will benefit from a cross-over
locoregional recurrences of the chest wall fol- chemotherapy regimen if tumor response is
lowing mastectomy show that optimal local con- inadequate to the initial drugs.
trol is provided by using both systemic therapy
and irradiation rather than chemotherapy INFLAMMATORY BREAST CANCER
alone.30 The use of axillary irradiation in patients
with clinically node-negative stage I or stage II Because today’s combination chemotherapy
breast cancer reduced the rate of axillary recur- regimens can often render inflammatory breast
rence by 1 and 3 percent, respectively.31–33 The cancer (IBC) resectable, mastectomy now has a
Early Breast Cancer Trialists’ Collaborative role in the treatment of this disease. In our
Group overview analysis of randomized trials review of 178 women treated for IBC in dox-
comparing axillary surgical clearance versus orubicin-based multimodality therapy proto-
radiotherapy found no difference in mortality cols between 1974 and 1993, the addition of
between groups regardless of the type of axillary mastectomy led to significant improvement in
treatment.34 Sentinel lymph node biopsy in locoregional disease control.36 Locoregional
patients with LABC has not been sufficiently relapse rates were 16.3 percent (16 of 98
studied yet and will prove accurate only if patients) for patients who underwent chemo-
metastatic deposits within each axillary lymph therapy, mastectomy, and radiotherapy, and
node respond identically to chemotherapy.35 35.7 percent (15 of 42 patients) for patients
Based upon the clinical trials conducted who underwent only chemotherapy and radio-
thus far, we have learned that induction therapy (p = .016). However, when patients
chemotherapy can be given safely without were stratified on the basis of tumor response
increasing the morbidity of local treatment. The to induction chemotherapy, only patients with a
histologic findings after induction chemother- partial response to chemotherapy demonstrated
apy remain important in defining prognosis. significant improvement in local control with
Very few patients will have progression of their the addition of mastectomy. As only 12 percent
disease and some will become candidates for of patients (21 of 178) had a complete clinical
breast conservation therapy. Most importantly, response, demonstration of a statistically sig-
neoadjuvant chemotherapy may identify nificant improvement in local control with the
160 BREAST CANCER

use of mastectomy was not feasible in this patients who had no significant response to
review. The amount of residual disease found induction chemotherapy.
on histologic examination of the mastectomy These retrospective data suggest that optimal
specimen was highly predictive of long-term local control for most patients with IBC is
local control: no patient with < 1 cm3 residual obtained with the addition of mastectomy to
disease (n = 38) had a locoregional recurrence. chemotherapy and radiotherapy. Other benefits
The effect of the addition of mastectomy on of mastectomy are that it allows accurate assess-
disease-specific and disease-free survival was ment of the amount of residual disease after
also dependent on the tumor response to induc- induction chemotherapy and that lower doses of
tion chemotherapy (Figure 10–3). Patients who radiation can be used for subclinical disease.37
had a complete or partial clinical response to However, mastectomy should be used only selec-
induction chemotherapy and were treated with tively in patients who have no significant
mastectomy in addition to chemotherapy and response to induction chemotherapy, as these
irradiation had significantly improved 5-year patients are at high risk for both local and distant
disease-specific survival compared with patients failure regardless of surgical intervention.
who had a similar response to induction Although accelerated fractionation radiotherapy
chemotherapy but did not undergo mastectomy has been proposed to exploit the biologic char-
(62.0 v 43.0 %; p = .018). No improvement in acteristics of IBC, an improvement in local con-
survival (disease-specific or disease-free) with trol rates with this technique has not been con-
the addition of mastectomy was detected in firmed.38,39 However, this accelerated schedule

A B

C D
Figure 10–3. Kaplan-Meier actuarial survival curves for disease-specific (A) and disease-free (B) survival in patients with a
complete or partial response to induction chemotherapy and for disease-specific (C) and disease-free (D) survival in patients
with no significant response to induction chemotherapy, stratified by type of treatment received. C = chemotherapy; M = mas-
tectomy; RT = radiotherapy. Reprinted with permission from Fleming RYD, Armar L, Buzdar AU, et al. Effectiveness of mastec-
tomy by response to induction chemotherapy for control in inflammatory breast carcinoma. Ann Surg Oncol 1997;4:452–61.
Locally Advanced Breast Cancer 161

expedites delivery of the radiotherapy, which Forty-three patients were entered in this IBC
may be especially useful if the goal of therapy is protocol between 1994 and 1998. There was a
palliation. Interstitial irradiation has been stud- clinical complete response observed in seven
ied as a possible substitute for surgery in patients percent and a clinical partial response observed
who experience significant tumor reduction with in 65 percent. This overall rate of response to
chemotherapy, but early results in terms of local induction chemotherapy of 72 percent is iden-
control do not appear promising.40 tical to the rate of response in the 178 patients
Attempts to improve tumor downstaging in with IBC treated on FAC induction chemother-
IBC with high-dose chemotherapy have shown apy protocols between 1974 and 1993. How-
promise in terms of clinical response rates, but ever, 2-year disease-free survival and overall
the effect on long-term survival is still unclear. survival were 61 and 78 percent, respectively,
In a review of five trials of either single-agent in the current protocol (median follow-up,
or combination chemotherapy followed by 20 months), compared to 52 and 71 percent,
autologous bone marrow transplantation respectively, in our previous studies (median
(ABMT) for IBC and other stage III breast can- follow-up, 86 months). In our next protocol for
cers, Antman and colleagues41 reported that 44 IBC, patients will receive four cycles of induc-
(79%) of 56 patients had a clinical complete tion chemotherapy with FAC followed by two
response after induction chemotherapy but six-week courses of paclitaxel (175 mg/m2 as a
before ABMT, and that 89 percent had a com- 3-hour infusion weekly), with a two-week break
plete response after ABMT. Disease-free status between the two courses. Patients with minimal
was maintained in 54 percent of patients, with tumor response after the FAC-paclitaxel induc-
follow-up ranging from 1 to 37 months. tion chemotherapy will be considered as candi-
The use of a different, crossover chemother- dates for high-dose chemotherapy with autolo-
apy regimen prior to mastectomy in patients gous peripheral blood progenitor cell support.
with IBC with less than a partial response to Surgical therapy will be planned at the comple-
induction chemotherapy may also be effective, tion of all systemic therapy. In patients who
as nonanthracyline-resistant drugs such as have a complete clinical response as docu-
paclitaxel and docetaxel are now available. The mented by physical examination, radiological
current trial at M. D. Anderson involves the use imaging, and core needle biopsies, locoregional
of paclitaxel if less than a partial response is irradiation concomitant with weekly paclitaxel
obtained with four cycles of induction FAC. If a will be offered as an alternative to surgery.
complete response or partial response is
achieved with paclitaxel, the patient undergoes RECONSTRUCTIVE SURGERY
mastectomy followed by four cycles of pacli-
taxel and irradiation. In patients who have a The goal of reconstructive surgery for patients
complete or partial response with the initial four with LABC can be to (1) repair defects, or (2)
cycles of FAC, mastectomy is performed and an repair defects and re-create a breast mound. In
additional four cycles of FAC are given, fol- patients with LABC who need or elect to have
lowed by four cycles of paclitaxel and irradia- standard mastectomy and who desire breast
tion. In patients with no significant response to reconstruction to improve the cosmetic out-
either FAC or paclitaxel induction chemo- come, reconstruction is often delayed until
therapy, the radiation oncologist and surgeon completion of both adjuvant chemotherapy and
plan whether to treat the breast with preopera- irradiation. As most locoregional recurrences
tive irradiation and then perform mastectomy or are in the skin or subcutaneous tissue of the
to proceed with definitive irradiation as the only chest wall,42 a flat postmastectomy chest wall
local modality, with the intent of palliation. often makes irradiation technically easier than
162 BREAST CANCER

does a reconstructed breast mound, especially if result, except for the anticipated skin tanning
inclusion of the internal mammary nodal basin and slight fibrosis of the reconstructed breast
is necessary. However, in selected patients with mound. In a series from M. D. Anderson48 of 19
excellent response to induction chemotherapy patients who received radiotherapy after recon-
or when palliative debulking surgical proce- struction with an autogenous tissue flap, either
dures are needed, the use of an autogenous flap for known local recurrence (n = 4) or as adju-
to create a breast mound or provide skin cover- vant therapy for high risk of recurrence (n = 15),
age of the operative defect before radiotherapy the cosmetic result was dependent on the initial
is instituted if feasible. outcome of the reconstruction.
Implant-based reconstruction in an irradi- The two tissue flaps used most frequently
ated field has been associated with a high com- for reconstruction after breast surgery are the
plication rate as well as patient discomfort and latissimus dorsi and rectus abdominis myocuta-
dissatisfaction because of loss of skin elasticity neous flaps. The advantages of the latissimus
and fibrosis of underlying tissues after irradia- dorsi flap include its reliable blood supply and
tion.43,44 The M. D. Anderson series of 298 the relative rarity of donor site morbidity. This
patients who received submuscular implants flap is also relatively thin, so it matches the
revealed that the rates of capsular contracture thickness of the native chest wall skin fairly
(Baker III or greater), pain, implant exposure, closely and is excellent for providing coverage
and implant removal were significantly higher of soft-tissue defects (Figure 10–4). The chief
(p = .028) in 13 patients with implants within disadvantage of the latissimus dorsi flap is its
an irradiated field than in 230 patients with limited size; an implant is usually required if
implants who received no radiotherapy.45 The the patient desires a reconstructed breast
effects of irradiation were slightly less detri- mound. The amount of available surplus skin
mental in patients with implants placed beneath varies from patient to patient, but in general the
autogenous-tissue flaps: the complication rate latissimus dorsi flap is never > 10 cm wide or
was 40 percent in 19 patients with implants 20 cm long.
placed in an irradiated area and 8 percent in 36 Rectus abdominis myocutaneous flaps can
patients with implants who had not undergone be quite large and are most useful for defects
radiotherapy. too large to repair with a latissimus dorsi flap.
The use of a myocutaneous flap for breast The chief disadvantage is that they tend to be
reconstruction, either before or after irradiation, bulky and thus do not closely match the thick-
does not interfere with the resumption of ness of the native chest wall skin. The thickness
chemotherapy or the ability to detect locore- of this flap can be an advantage, however, if the
gional recurrence.46 Irradiation of the recon- defect is located directly over the central area of
structed breast-mound flap does not impair the the chest wall; in this case, the excess flap bulk
flap’s blood supply. In the M. D. Anderson series can be used to reconstruct a breast mound.
of 61 patients who required complex chest wall The two main types of rectus abdominis myo-
resections,47 prior irradiation that included the cutaneous flaps are the transverse rectus abdo-
internal mammary artery, which provides blood minis myocutaneous (TRAM) flap and the verti-
to the rectus abdominis flap, or the thoracodor- cal rectus abdominis myocutaneous (VRAM) flap.
sal artery, which provides blood to the latis- The TRAM flap has a greater arc of rotation and
simus dorsi flap, did not compromise the viabil- a more symmetrical and easily concealed donor
ity of these flaps for wound coverage. Provided site than does the VRAM flap (Figure 10–5). The
that the flap has an adequate vascularization VRAM flap leaves a more noticeable donor scar
without evidence of significant fat necrosis, the but is technically easier to construct and has a
irradiation itself does not alter the cosmetic more reliable blood supply (Figure 10–6). The
Locally Advanced Breast Cancer 163

A B

Figure 10–4. A, B, Patient with locally advanced cancer of the


left breast who refused chemotherapy and radiation therapy.
C, Operative defect after modified radical mastectomy.
D, Closure of operative defect with a latissimus dorsi myo-
cutaneous flap. E, Patient with excellent range of motion 10
days after surgery. (Reprinted with permission from Single-
tary SE. Breast surgery. In: Roh MS, Ames FC, editors. Atlas
of advanced oncologic surgery. New York: Gower Medical
Publishing, 1993. p. 14.1–14.9.)

E
164 BREAST CANCER

TRAM flap is used most often when cosmetic ribs have been removed, the use of mesh does
considerations are important. improve chest wall mechanics and reduces the
For major chest wall resections, the rectus duration of ventilator dependence. Marlex, a
abdominis flap is capable of covering a wide nonabsorbable durable mesh, can be used for
area from the clavicle to the costal margin and flat surfaces of the chest wall. If the defect is
from the sternum to the midaxillary line. large, a “sandwich” of Marlex mesh and methyl
Because this flap is bulky, it provides sufficient methacrylate can be formed to restore a more
chest wall stability even when up to five ribs or normal contour.49 If the mesh is covered by
the entire sternum is resected, without the need well-vascularized tissue, the risk of infection
for prosthetic mesh. However, if three or more and extrusion is usually low.

Figure 10–5. A, Operative defect following full-thickness


chest-wall excision. B, Myocutaneous TRAM flap harvested
from the lower abdominal wall and transferred to the chest.
C, Postoperative result with the chest wall reconstruction
shaped into a facsimile of a breast. (Reprinted with permis-
sion from Singletary SE, Hortobagyi GN, Kroll SS. Surgical
and medical management of local-regional treatment failures
in advanced primary breast cancer. Surg Oncol Clin N Am
1995;4(4):671–84.)
C
Locally Advanced Breast Cancer 165

Figure 10–6. A, Operative defect of the chest wall after radi-


cal excision. B, Myocutaneous VRAM flap harvested from the
lateral abdominal wall. C, Postoperative result. (Reprinted
with permission from Singletary SE, Hortobagyi GN, Kroll
SS. Surgical and medical management of local-regional
treatment failures in advanced primary breast cancer. Surg
Oncol Clin N Am 1995;4(4):671–84.)

C
166 BREAST CANCER

STRATEGIES FOR results on positron emission tomography, which


IMPROVING SYSTEMIC THERAPY reflect the metabolic alterations in the breast
cancer following chemotherapy, may hold
Advances in the treatment of LABC are largely promise as a predictor of response.55,56 Studies
dependent on improvements in systemic chemo- are also under way to evaluate the role of mag-
therapy. Two major strategies to improve sys- netic resonance imaging in accurately measur-
temic therapy include improved selection and ing true tumor response to chemotherapy.57
individualization of chemotherapy regimens and However, the optimal timing of these modalities
the development of novel targeted therapies. If in relation to the cycle of chemotherapy has not
the chemosensitivity of a specific breast cancer yet been determined.
could be predicted before or soon after the initia- An exciting new area is the identification of
tion of chemotherapy, an optimal treatment regi- several specific targets for novel therapeutic
men could be designed for that tumor. One pos- approaches based on an understanding of the
sible way to predict chemosensitivity is to molecular genetic and biochemical features of
measure levels of cellular proteins associated the tumor. These therapeutic approaches may
with drug resistance, including MDR1 (mul- include monoclonal antibodies either alone or
tidrug-resistance protein, or P-glycoprotein), conjugated to a cytotoxic substance; vaccines; or
MRP (multidrug resistance-associated protein), gene therapy to either suppress an oncogene or
glutathione S-transferase, and dihydrofolate replace the product of an inactivated tumor sup-
reductase.50 Although MDR1 is often not pressor gene.58 These strategies may be com-
expressed in early-stage breast cancer, it is bined with current chemotherapy regimens to
detectable at a high frequency in patients with produce a synergistic effect. Antibodies against
LABC 51 and appears to correlate with a poor the HER-2/neu and epidermal growth factor
response to chemotherapy. However, the other receptor oncogene products have been demon-
proteins associated with drug resistance are strated to have a synergistic effect when com-
either not detectable at a sufficient frequency or bined with cisplatin, doxorubicin, and cyclo-
have not yet been shown to be reliable enough to phosphamide.59,60 Novel therapies may also be
predict in vivo drug resistance. The earlier in used to protect normal cells against the effects of
vitro chemosensitivity assays were problematic chemotherapy drugs and thus lessen side effects.
because the patient’s tumor cells had to be grown For example, gene therapy may allow the human
in culture for a prolonged period and the assays multidrug-resistance gene to be transfected into
could reliably predict only chemoresistance, not human marrow progenitors to instill a preferen-
chemosensitivity. Newer chemosensitivity assays tial resistance to a chemotherapy drug such as
that preserve the cellular spatial relationships paclitaxel.61 Although the BRCA1 gene is rarely
of the tumor and do not require a prolonged mutated in sporadic breast cancer, levels of
culture period are currently being assessed.52 BRCA1 mRNA and its protein are decreased in
Another approach to predicting chemosensi- both hereditary and sporadic disease. Results of
tivity is to measure the effects of the chemother- a pilot trial with an ovarian cancer nude mouse
apy on the intact tumor in vivo. This can be done model indicate that delivery of a nonmutated
by studying sequential needle aspirates to deter- BRCA1 gene into the tumor via a retroviral vec-
mine changes in flow cytometric DNA profiles tor can suppress tumor growth.62 Encouraging
and nuclear morphometric features that measure observations have also been reported for an E1B
alterations in DNA content and cell cycle char- gene-attenuated adenovirus, ONYX-015, that
acteristics during chemotherapy; changes in targets the p53 gene of tumors but not of normal
these features have been shown to correlate with cells.63 The tumor-specific cytolysis produced by
subsequent tumor regression.53,54 Alternatively, this adenovirus appears to augment the efficacy
Locally Advanced Breast Cancer 167

of concomitant chemotherapy. Other therapeutic 9. Booser D, Frye D, Singletary S, et al. Response to


possibilities include inhibitors of angiogene- induction chemotherapy for breast cancer: a
prospective multimodality treatment program.
sis64–66 and matrix metalloproteinases67; retinoids
[abstract] Proc Am Soc Clin Oncol 1992;11:82.
to induce differentiation68; and vaccines directed 10. Kuerer HM, Newman LA, Fornage BD, et al. Role
against tumor antigens such as muc-1.69 of axillary lymph node dissection after tumor
Translational research that brings new treat- downstaging with induction chemotherapy for
ment concepts from the laboratory to the clini- locally advanced breast cancer. Ann Surg
cal arena is essential for continued progress in Oncol 1998;5(8):673–80.
11. Buzdar AU, Hortobagyi GN, Asmar L, et al.
the management of LABC. Clinicians must be
Prospective randomized trial of paclitaxel
prepared to consider the feasibility of molecu- alone versus 5-fluorouracil/doxorubicin/
lar control of the underlying process of mam- cyclophosphamide as induction therapy in
mary carcinogenesis as part of their treatment patients with operable breast cancer. Semin
armamentarium for both early-stage breast can- Oncol 1997;24:1–4.
cer and LABC. 12. Peoples GE, Regan Q, Heaton KM, et al. Breast
conservation therapy for large primary and
locally advanced breast cancers after induction
REFERENCES chemotherapy. Ann Surg Oncol. In press.
1. Anonymous. Breast. In: Fleming ID, Cooper JS, 13. Hunt KK, Buzdar AU. Breast conservation after
Henson DE, et al, editors. AJCC cancer staging tumor downstaging with induction chemother-
manual. 5th ed. Philadelphia: Lippincott- apy. In: Singletary SE, editor. Breast Cancer-
Raven; 1997. p.171–80. M. D. Anderson Solid Tumor Oncology Series.
2. Strom EA, McNeese MD, Fletcher GH, et al. New York: Springer-Verlag; 1999. p.196–207.
Results of mastectomy and postoperative irra- 14. Bonadonna G, Veronesi U, Brambilla C, et al. Pri-
diation in the management of locoregionally mary chemotherapy to avoid mastectomy in
advanced carcinoma of the breast. Int J Radiat tumors with diameters of three centimeters or
Oncol Biol Phys 1991;21:319–23. more. J Natl Cancer Inst 1990;82:1539–45.
3. Hortobagyi GN, Ames FC, Buzdar AU, et al. Man- 15. Calais G, Berger C, Descamps P, et al. Conserva-
agement of stage III primary breast cancer with tive treatment feasibility with induction chemo-
primary chemotherapy, surgery, and radiation therapy, surgery, and radiotherapy for patients
therapy. Cancer 1988;62:2507–16. with breast carcinoma larger than 3 cm. Cancer
4. Broadwater JR, Edwards MJ, Kuglen C. Mastec- 1994;74:1283–8.
tomy following preoperative chemotherapy. 16. Veronesi U, Bonadonna G, Zurrida S, et al. Con-
Ann Surg 1991;213:126–9. servation surgery after primary chemotherapy
5. Feldman LD, Hortobagyi GN, Buzdar AU, et al. in large carcinomas of the breast. Ann Surg
Pathological assessment of response to induc- 1995;222:612–8.
tion chemotherapy in breast cancer. Cancer 17. Schwartz GF. Breast conservation following
Res 1986;46:2578–81. induction chemotherapy for locally advanced
6. McCready DR, Hortobagyi GN, Kau SW, et al. breast cancer: a personal experience. The
The prognostic significance of lymph node Breast Journal 1996;2:78–82.
metastases after preoperative chemotherapy for 18. Touboul E, Buffat L, Lefranc J, et al. Possibility of
locally advanced breast cancer. Arch Surg conservative local treatment after combined
1989;124:21–5. chemotherapy and preoperative irradiation for
7. Hortobagyi GN, Singletary SE, Buzdar AU, et al. locally advanced noninflammatory breast can-
Primary chemotherapy for breast cancer: M. D. cer. Int J Radiat Oncol Biol Phys 1996;34:
Anderson experience. In: Banzet P, editor. Pro- 1019–28.
ceedings of the 3rd International Congress on 19. Merajver SD, Weber BL, Cody R, et al. Breast
Neoadjuvant Chemotherapy. New York: conservation and prolonged chemotherapy for
Springer-Verlag; 1991. p. 145–8. locally advanced breast cancer: the University
8. Singletary SE, McNeese MD, Hortobagyi GN. of Michigan experience. J Clin Oncol 1997;15:
Feasibility of breast conservation surgery after 2873–81.
induction chemotherapy for locally advanced 20. Fisher B, Bryant J, Wolmark N, et al. Effect of
carcinoma. Cancer 1992;69:2849–52. preoperative chemotherapy on the outcome of
168 BREAST CANCER

women with operable breast cancer. J Clin 32. Delouche G, Bachelot F, Premont M, Kurts JM.
Oncol 1998;16:2672–85. Conservation treatment of early breast cancer:
21. Schwartz GF, Birchansky CA, Komarnicky LT, et long-term results and complications. Int J
al. Induction chemotherapy followed by breast Radiat Oncol Biol Phys 1987;13:29–34.
conservation for locally advanced carcinoma 33. Wazer DE, Erban JK, Robert NJ, et al. Breast con-
of the breast. Cancer 1994;73:362–9. servation in elderly women for clinically nega-
22. Scholl SM, Fourquet A, Asselain B, et al. Neoad- tive axillary lymph nodes without axillary dis-
juvant versus adjuvant chemotherapy in pre- section. Cancer 1994;74:878–83.
menopausal patients with tumors considered 34. Early Breast Cancer Trialists’ Collaborative
too large for breast conserving surgery: pre- Group. Effects of radiotherapy and surgery in
liminary results of a randomised trial: S6. Eur early breast cancer. N Engl J Med 1995;333:
J Cancer 1994;5:645–52. 1444–55.
23. Delena M, Varini M, Zucali R, et al. Multimodal 35. Singletary SE. Management of the axilla in early
treatment for locally advanced breast cancer. stage breast cancer. In: Perry MC, editor.
Cancer Clinical Trials 1981;4:229–36. American Society of Clinical Oncology Edu-
24. Perloff M, Lesnick GJ, Korzun A, et al. Combina- cational Book. Alexandria (VA): American
tion chemotherapy with mastectomy or radio- Society of Clinical Oncology; 1998. p.132–41.
therapy for stage III breast carcinoma: Cancer 36. Fleming RYD, Asmar L, Buzdar AU, et al. Effec-
and Leukemia Group B study. J Clin Oncol tiveness of mastectomy by response to induction
1988;6:261–9. chemotherapy for control in inflammatory breast
25. Rahman ZU, Frye DK, Buzdar AU, et al. Impact of carcinoma. Ann Surg Oncol 1997;4:452–61.
selection process on response rate and long- 37. Singletary SE, Ames FC, Buzdar AU. Manage-
term survival of potential high-dose chemo- ment of inflammatory breast cancer. World J
therapy candidates treated with standard-dose Surg 1994;18:87–92.
doxorubicin-containing chemotherapy in 38. Barker JL, Montague ED, Peters LJ. Clinical
patients with metastatic breast cancer. J Clin experience with irradiation of inflammatory
Oncol 1997;15:3171–7. carcinoma of the breast with and without elec-
26. Hortobagyi GN, Buzdar AU, Champlin R, et al. tive chemotherapy. Cancer 1981;45:625–29.
Lack of efficacy of adjuvant high-dose (HD) 39. Thoms WW, McNeese MD, Fletcher GH, et al.
tandem combination chemotherapy (CT) for Multimodal treatment for inflammatory breast
high-risk primary breast cancer (HRPBC)—a cancer. Int J Radiat Oncol Biol Phys 1989;17:
randomized trial. [abstract] Proc Am Soc Clin 739–45.
Oncol 1998;17: 123a. 40. Brun B, Ottmezguine Y, Feuilhade F, et al. Treat-
27. Dunphy FR, Spitzer G, Buzdar AU, et al. Treat- ment of inflammatory breast cancer with com-
ment of estrogen receptor negative or hormon- bination chemotherapy and mastectomy versus
ally refractory breast cancer with double high- breast conservation. Cancer 1988;61:1096–
dose chemotherapy intensification and bone 103.
marrow support. J Clin Oncol 1990;8:1207–16. 41. Antman K, Bearman SI, Davidson N, et al. Dose
28. Holmes FA, Walters RS, Theriault RL, et al. Phase intensive therapy in breast cancer: current sta-
II trial of Taxol, an active drug in the treatment tus. In: Gale RP, Champlin RE, editors. New
of metastatic breast cancer. J Natl Cancer Inst strategies in bone marrow transplantation. New
1991;83:1797–1805. York: Alan R Liss; 1990. p. 423–6.
29. Valero V, Holmes FA, Walters RS, et al. Phase II 42. Newman LA, Kuerer HM, Hunt KK, et al. Pre-
trial of docetaxel, a new highly effective anti- sentation, treatment and outcome of local
neoplastic agent in the management of patients recurrence after skin-sparing mastectomy and
with anthracycline-resistant breast cancer. J immediate breast reconstruction. Ann Surg
Clin Oncol 1995;13:2886–94. Oncol 1998;5(7):620–6.
30. Buzdar AU, McNeese MD, Hortobagyi GN, et al. 43. Halpern J, McNeese MD, Kroll SS, Ellerbrock N.
Is chemotherapy effective in reducing the local Irradiation of prosthetically augmented breasts:
failure rate in patients with operable breast a retrospective study on toxicity and cosmetic
cancer? Cancer 1990;65:394–9. results. Int J Radiat Oncol Biol Phys 1990;18:
31. Osborne MP, Ormiston N, Harmer OL, et al. 189–91.
Breast conservation in the treatment of early 44. Forman DL, Chiu J, Restifo RJ, et al. Breast
breast cancer: a 20-year follow-up. Cancer reconstruction in previously irradiated patients
1984;53:349–55. using tissue expanders and implants: a poten-
Locally Advanced Breast Cancer 169

tially unfavorable result. Ann Plast Surg 1998; apy. Ann NY Acad Sci 1993;698:423–8.
40:360–4. 57. Orel SG, Schnall MD, Powell CM, et al. Staging
45. Evans GRD, Schusterman MA, Kroll SS, et al. of suspected breast cancer: effect of MR imag-
Reconstruction and the radiated breast: is there ing and MR-guided biopsy. Radiology 1995;
a role for implants? Plast Reconstr Surg 1995; 196:115–22.
96:1111–8. 58. Dhingra K, Hittelman WN, Hortobagyi GN.
46. Schusterman MA, Kroll SS, Miller MJ, et al. The Genetic changes in breast cancer—conse-
free transverse rectus abdominis musculocuta- quences for therapy? Gene 1995;159:59–63.
neous flap for breast reconstruction: one cen- 59. Baselga J, Norton L, Masui H, et al. Antitumor
ter’s experience with 211 consecutive cases. effects of doxorubicin in combination with
Ann Plast Surg 1994;32:234–42. anti-epidermal growth factor receptor mono-
47. McKenna RJ, Moutain CF, McMurtrey MJ, et al. clonal antibody. J Natl Cancer Inst 1993;85:
Current techniques for chest wall reconstruc- 1327–33.
tion: expanded possibilities for treatment. Ann 60. Hancock MC, Langton BC, Chan T, et al. A
Thorac Surg 1988;46:508–12. monoclonal antibody against the c-erbB-2
48. Hunt KK, Baldwin BJ, Strom EA, et al. Feasibil- protein enhances the cytotoxicity of cis-
ity of postmastectomy radiation therapy after diammine dichloroplatinum against human
TRAM flap breast reconstruction. Ann Surg breast and ovarian tumor cell lines. Cancer Res
Oncol 1997;4:377–84. 1991;51:4575–80.
49. Kroll SS, Walsh G, Ryan B, King RC. Risks and 61. Ward M, Richardson C, Pioli P, et al. Transfer and
benefits of using Marlex mesh in chest wall expression of the human multiple drug resis-
reconstruction. Ann Plast Surg 1993;31:303–6. tance gene in human CD34+ cells. Blood 1994;
50. Verrele P, Meissonnier F, Fonck Y, et al. Clinical 84:1408–14.
relevance of immunohistochemical detection of 62. Holt JT, Thompson ME, Szabo C, et al. Growth
multidrug resistance. P-glycoprotein in breast retardation and tumor inhibition by BRCA1.
carcinoma. J Natl Cancer Inst 1991;83:111–6. Nat Genet 1996;12:298–302.
51. Ro J, Sahin A, Ro JY, et al. Immunohistochemical 63. Heise C, Sampson-Johannes A, Williams A, et al.
analysis of P-glycoprotein expression corre- ONYX-015, an E1B gene-attenuated adeno-
lated with chemotherapy resistance in locally virus, causes tumor-specific cytolysis and anti-
advanced breast cancer. Hum Pathol 1990;21: tumoral efficacy that can be augmented by
787–91. standard chemotherapeutic agents. Nature
52. Blackman KE, Fingert HJ, Fuller AF, Meitner PA. 1997;3:639–45.
The fluorescent cytoprint assay in gynecologi- 64. Thorpe PE, Burrows FJ. Antibody-directed target-
cal malignancies and breast cancer. Methodol- ing of the vasculature of solid tumors. Breast
ogy and results. Contrib Gynecol Obstet 1994; Cancer Res Treat 1995;36:237–51
19:53–63. 65. O’Reilly MS, Holmgren L, Chen C, Folkman J.
53. Brifford M, Spyratos F, Hacene K, et al. Evaluation Angiostatin induces and sustains dormancy of
of breast carcinoma chemosensitivity by flow human primary tumors in mice. Nat Med 1996;
cytometric DNA analysis and computer assisted 2:689–92.
image analysis. Cytometry 1991;13:250–8. 66. Folkman J. Clinical implications of angiogenesis
54. O’Reilly SM, Camplejohn RS, Rubens RD. DNA research. N Engl J Med 1995;333:1757–63.
flow cytometry and response to preoperative 67. Sledge GW Jr, Qulali M, Goulet R, et al. Effect of
chemotherapy for primary breast cancer. Eur J matrix metalloproteinase inhibitor batimastat on
Cancer 1992;28:681–3. breast cancer regrowth and metastasis in athymic
55. Jansson T, Westlin JE, Ahlstrom H, et al. Positron mice. J Natl Cancer Inst 1995;87:1546–50.
emission tomography studies in patients with 68. Lotan R. Retinoids in cancer chemoprevention.
locally advanced and/or metastatic breast can- FASEB J 1996;10:1031–9.
cer: a method for early therapy evaluation. J 69. Gilewski T, Adluri R, Zhang S, et al. Preliminary
Clin Oncol 1995;13:1470–7. results: vaccination of breast cancer patients
56. Nieweg OE, Wong W-H, Singletary SE, et al. lacking identifiable disease with muc-1-key-
Positron emission tomography of glucose hole limpet hemocyanin (klh) conjugate and
metabolism in breast cancer: potential for tumor qs21. [abstract] Proc Am Soc Clin Oncol 1996;
detection, staging, and evaluation of chemother- 15:555.
11
Breast Reconstruction
GEOFFREY C. FENNER, MD
THOMAS A. MUSTOE, MD

In contrast to the 1960s when silicone implants nosed at an earlier age and stage, have excellent
were the mainstay of breast reconstruction, prognoses, and represent an increasing percent-
patients in the 1990s may choose from an age of patients seeking consultation and alter-
impressive spectrum of reconstructive options. natives for breast restoration.
Techniques, instruments, and materials have The female breast is intimately associated
evolved that provide all patients, regardless of with a woman’s selfesteem, sexuality, and inter-
age, stage, previous treatment, or laterality, personal relations. The response to the impact
choices that may optimally represent their and presumed implications of breast cancer
desires and expectations. Breast cancer aware- varies widely among women. Breast cancer
ness has increased the sophistication of represents a therapeutic myriad with emotional
patients, however, it remains the plastic sur- and physical implications, both for the present
geon’s responsibility to educate patients and and future. Although breast reconstruction may
coordinate expectations and outcome. be viewed as a positive alternative to breast
Increased detection of breast cancer has par- loss, it represents only one facet newly diag-
alleled improved techniques and availability of nosed cancer patients must face. Each patient
screening mammography, an increased female upholds an individual, often rigid, esthetic stan-
population, and the impact of changes in the dard, emotional drive, and physiology which
age of childbearing, menarche, and menopause. guides them towards a specific reconstructive
Today, ductal carcinoma in situ (DCIS) repre- technique. It remains the plastic surgeon’s
sents 15 to 20 percent of all breast cancer responsibility to inform, educate, and perform
cases;1 it is treated by either localized resection with this in mind.
or total mastectomy. Genetic testing and better The first breast reconstruction was per-
elucidation of risk factors has identified addi- formed by Czerny, in 1895, when he success-
tional patients as potential candidates for pro- fully transplanted a lipoma from a patient’s flank
phylactic mastectomy. As many as 15 percent to a submammary position.3 Multiple develop-
of patients undergoing breast conservation, and ments over the past 100 years have improved
who require a proportionately large lumpec- reconstructive options as well as ultimate out-
tomy, attain poor esthetic outcome and may be comes for women faced with mastectomy.
better served in the longterm, by preoperative
consideration of completion mastectomy and IMMEDIATE RECONSTRUCTION
autologous reconstruction.2 Included in this
group are patients with small breasts, propor- Immediate reconstruction provides significant
tionately large lesions, and centrally located advantages for the newly diagnosed breast can-
lesions. These women, often having been diag- cer patient (Table 11–1). Greater understanding

171
172 BREAST CANCER

Table 11–1. ADVANTAGES OF greater freedom in choosing clothing.6 Patients


IMMEDIATE RECONSTRUCTION
undergoing mastectomy and immediate recon-
Diminished psychologic trauma struction demonstrate a similar psychosocial
Facilitates coverage of radical defects
Eases recipient pedicle dissection outcome to that of breast conservation patients,
Superior esthetic results having had lumpectomy with or without radia-
Incorporates skin sparing mastectomy
tion.7 Body image may be adversely affected
Minimizes anemhesia and improves cost
due to greater breast and donor site scarring
compared to patients having undergone breast
of tumor biology and technical advances in conservation. Overall, psychologic morbidity is
reconstructive surgery have led to greater similar, and clearly favorable compared to that
acceptance of immediate postmastectomy of patients having had delayed reconstruction.8
reconstruction. The first large series reported in The opportunity to attain optimal esthetic
1982, conveyed excellent outcome, less expense results is enhanced with immediate reconstruc-
than delayed reconstruction, and no apparent tion. The newly raised mastectomy skin flaps
effect on the natural course of the malignancy.4 tend to preserve the shape of the natural breast,
Initial options for immediate reconstruction providing a structural template that determines
through the mid-1990s included various the shape of the underlying volume, whether an
expanders and implants, yet now include vari- implant or flap reconstruction. Skin flap fibro-
ous flaps and even free-flap reconstruction. Sat- sis associated with delayed reconstruction rep-
isfactory outcome is dependent upon patient resents inherent tissue loss and requires either
selection as well as communication between the greater tissue expansion or greater skin replace-
ablative and reconstructive surgeons. ment at the time of autologous reconstruction.
The advantages of immediate reconstruc- Fibrosis of the mastectomy skin flaps are an
tion include diminished psychosocial trauma, impediment in achieving a natural breast shape.
superior esthetic results, decreased surgical Skin-sparing mastectomy in immediate recon-
morbidity, and lower cost than delayed recon- struction further increases the ability to attain a
struction. Historically, delayed reconstructions symmetric result, limits scarring to the periare-
were more often performed due to heightened olar region, and minimizes the need for con-
fear of recurrence, concerns that immediate tralateral procedures such as reductions and
reconstruction would mask subsequent detec- mastopexies.9–13
tion of a recurrence, and the possibility that Administration of adjuvant therapy is not
immediate reconstruction would be compro- delayed in patients undergoing immediate
mised by and hinder the initiation of adjuvant breast reconstruction, nor is the rate of compli-
therapy. It was also felt that patients would be cations higher after immediate reconstruc-
more appreciative of reconstruction if required tion.14–15 The usual 3 to 4 week interval prior to
to live for a time with the postmastectomy the initiation of adjuvant chemotherapy is
defect. These ideas have since been rendered ample time for uncomplicated, postreconstruc-
obsolete by the need to consider the emotional tive wound healing and patient recovery. Only
impact of mastectomy and by the technical and 1 to 2 percent of patients have their chemother-
therapeutic advances of the past 15 years. apy delayed beyond 3 to 4 weeks due to com-
Patients undergoing immediate reconstruc- plications from immediate reconstruction, such
tion tend to incorporate the new breast into as delayed healing.16
their body image, thereby maintaining greater Although neoadjuvant and adjuvant chemo-
selfesteem, personal sexuality, and confidence therapy have no relative impact upon immedi-
in interpersonal relationships.5 They tend to ate reconstruction, adjuvant radiation is known
have less “cancer anxiety,” less recall, and to unequivocally detract from the esthetic result
Breast Reconstruction 173

and increase the local complication rate. This is compensated for through surgical and design
influenced by many factors, including recon- modifications. It is interesting that the rate of fat
structive technique and the type and dose of necrosis and volume loss in TRAM flaps, postra-
radiation. Historically, radiation exaggerated diation, was higher in pedicled (33%) than in free
the extent of fibrous capsular contracture pre- TRAMs (6%) reconstructions.26
sent, to some extent, in all expander/implant Immediate postmastectomy reconstruction
reconstructions.17–19 Poor outcome paralleled for locally advanced disease has been reported
the need for substantial expansion and the use as encouraging. Sultan reported on 22 patients
of large, smooth, silicone implants. The rate of with stage IIB or III disease who had under-
poor cosmetic results in early series ranged gone neoadjuvant chemotherapy and comple-
from 18 to 40 percent, with a failure rate up to tion of chemotherapy 3 weeks subsequent to
40 percent.20 Evans and colleagues reported a surgery. Perioperative morbidity was 14 per-
43 percent complication rate among radiated cent. Delay in resumption of chemotherapy
implant reconstruction patients, compared to a occurred in no instances, and patients
12 percent rate in nonradiated patients.18 expressed appreciation for having been offered
Schuster reported a 55 percent complication this option.27 Styblo reported on 21 patients
rate and unacceptable cosmesis in 24 percent of with stage III disease who had undergone
postreconstructive patients requiring adjuvant immediate TRAM reconstruction. There were
radiation.17 Of patients who had undergone a no delays in reinstitution of adjuvant treatment
composite autogeneous/implant reconstruction, and no increase in local relapse.28 It has been
40 percent of the radiated and 8.3 percent of the shown that breast reconstruction may facilitate
nonradiated patients had major complica- resection, without an increase in local compli-
tions.18 Dickson reported an overall complica- cations or relapse.
tion rate of 70 percent for patients having Immediate reconstruction also has eco-
immediate prosthetic reconstruction with radia- nomic advantages. Ablation and reconstruction
tion, and rates of 30 percent for skin necrosis are combined in one procedure, thereby limit-
and 67 percent for capsule contracture.21 ing anesthetic risk and the time committed to
Although the general consensus is to avoid postoperative recovery. Patients welcome the
prosthetic reconstruction in patients, an antici- opportunity for a single procedure with less
pated need for adjuvant radiation, the regiment impact on occupational and domestic responsi-
is most often recommended postoperatively. bilities. Avoidance of a staged second surgery
Use of textured saline prosthesis as well as and hospitalization in delayed reconstruction
improved radiation techniques have demon- have obvious cost advantages.
strated improved overall tolerance and dimin-
ished complications in some early reports, but SKIN-SPARING MASTECTOMY
there is no consensus.22–24
In contrast, tolerance of autologous tissue to Toth and Lappert first described skin-sparing
radiation is generally good. Zimmerman reported mastectomy (SSM) in 1991.9 The technique is
the effect of postoperative radiation on immedi- indicated for patients with early stage (I and II)
ate free transverse rectus abdominis myocuta- breast cancer, patients managed with prophylac-
neous (TRAM) reconstruction. He reported no tic mastectomy, and in attempts to facilitate a
total or partial losses. Cosmesis, as rated by highly esthetic outcome through maximal skin
patients, was excellent in 60 percent of cases, preservation (Figure 11–1). Incisions are planned
good in 30 percent, and fair in 10 percent.25 that will remove the breast, nipple-areolar com-
Although some variable degree of cutaneous plex, adjacent biopsy scars, and the skin over
fibrous contracture may occur, this can usually be more superficial tumors. Kroll and colleagues in
174 BREAST CANCER

rate was 4.8 percent in the former group and 9.5


percent in the latter; native skin flap necrosis
occurred in 10.7 percent of the SSM patients
and in 11.2 percent of the non-SSM patients.13
Because local recurrence after SSM is low and
the likelihood of local control and survival are
high, SSM with immediate reconstruction is an
acceptable treatment for breast cancer.

BREAST IMPLANTS

The number of women with breast implants


ranges between 1.5 and 2 million. The modern
Figure 11–1. Skin-sparing mastectomy.
silicone implant has been available since 1963
and has undergone a multitude of subsequent
1991 reported only one local recurrence in 100 mechanical and material improvements. All
cases with a follow-up of 23 months.10 implants consist of a silicone elastomer shell
Local recurrence is dependent upon tumor that may be single or double lumen, with a
size and locoregional nodal involvement. smooth or textured surface. Contents of single
Despite variation in mastectomy technique, chamber implants consist of either silicone gel,
including SSM, the rate of local recurrence has which is factory sealed and nonadjustable, or
remained stable. Skin-sparing mastectomy is saline, which may be adjusted intra- and/or
more challenging for the oncologic surgeon, perioperatively. Dual chamber implants were
more time consuming, and requires delicate devised to provide the benefits and camouflage
handling of the skin flaps to avoid ischemic of silicone texture (outer lumen), along with
complications. These efforts to preserve the postoperative saline adjustability (inner
skin envelope and inframammary fold are lumen). Various natural oils, triglycerides, and
greatly appreciated by the patient and result in water soluble hydrogels are currently under
greater symmetry, often diminishing the need investigation but are not currently available in
for a contralateral procedure. Subsequent areo- the United States.
lar tatooing may completely camouflage the Silicone is ubiquitous in our environment.
central incisions. Individual exposure occurs through contact
Newman reported a 6.2 percent local recur- with needles, syringes, medications (insulin,
rence rate in 372 patients who underwent SSM simethicone), lipstick, creams, cosmetics, and
for TI/II lesions. Ninety-six percent of these implantable devices, such as pacemakers, joint
recurrences presented as palpable skin flap replacements, defibrillators, shunts, stents, and
masses.11 Hidalgo reported on 28 patients who implants.29 Extensive research undertaken
underwent immediate reconstruction (92% since the FDA-directed silicone breast implant
receiving TRAM flaps) after SSM, with a mean moratorium in 1992 has confirmed that
follow-up of 27 months. Complications at the implantable medical grade silicone is among
reconstructive site were limited to cellulitis and the least bioreactive, most inert substances
marginal periareolar skin loss. Esthetic results available for implantation.30–32 Studies have
were judged as excellent in 75 percent of failed to show linkage between connective tis-
patients.12 Carlson compared 327 patients given sue disease and silicone gel implants. The sili-
SSM to 188 non-SSM patients. After a mean cone elastomer shell and gel of breast implants,
follow-up of 41 months, the local recurrence however, like all implanted devices, will trigger
Breast Reconstruction 175

a foreign body inflammatory cell response, Table 11–2. BAKER’S CLASSIFICATION OF


CAPSULE CONTRACTURE
with giant cell formation and eventual scarring.
The extent and impact of this fibrotic capsular Class I Augmented breast feels as soft as an
unoperated-upon breast
response upon the fluid, and physical character- Class II Minimal; less soft, the implant can be palpated
istics of breast implants is dependent upon cap- but is not visible
sular density, implant–tissue incorporation, the Class III Moderate; more firm, the implant can be easily
palpated and is visible
presence of myofibroblasts, and/or the presence Class IV Severe; the breast is hard, tender, painful, cold,
of intracapsular silicone or sepsis. and distorted
Capsular contracture represents the most Reproduced with permission from Little G, Baker JL. Results of
common complication of breast implants. It closed compression capsulotomy for treatment of contracted breast
implant capsules. Plast Reconstr Surg 1980;65:30.
consists of progressive fibrous constriction
around breast implants and is unpredictable and
variable. It is graded according to a scale devel- the fibrous capsule is surgically released and or
oped by Baker (Table 11–2) and ranges from excised (capsulectomy) (Figure 11–3).
visually imperceptible (class I), to stone hard Silicone gel is composed of an amorphous
and painful (class IV). It may occur immedi- matrix consisting of silicone oils of various
ately or years after implantation. There is a sizes and weights. Smaller caliber oils are
greater incidence associated with smooth sili- known to diffuse through the elastomer shell
cone implants and with subglandular placement (silicone gel “bleed”) and become incorporated
in cosmetic augmentation. Some theories sug- into the fibrous capsule. Microscopic amounts
gest local contamination with Staphylococcus may percolate through lymphatic channels fol-
epidermidis as one inciting cause. The powder lowing macrophage ingestion and migrate to
from gloves and inflammation from even lim- the regional lymph nodes. As with exposure to
ited hematomas may play a role in some cases. other medial grade silicones, there is no evi-
Capsular contracture may cause implant dence to suggest that the minute quantities
deformation, migration, and rupture (Figure transgressing the elastomer shell have any
11–2) and may, on occasion, become calcified metabolic or long-term impact.
and detour from effective mammography. Indi- Silicone gel implant rupture occurs in up to
vidual perception is dependent upon severity 63 percent of patients after 12 years, docu-
and on the esthetic standard of the patient. Cap- mented during surgery in patients having their
sular contracture is not in itself a health risk. implants removed.33–36 Among asymptomatic
Twenty to 50 percent of reconstruction patients patients, the incidence of implant rupture is
who develop contractures require operative unknown but is believed to be significant.
intervention.
Contractures, historically, were released
through aggressive manual compression. The
goal was to “pop” the surrounding constricting
capsule, leading to a softer breast. This tech-
nique of closed capsulotomy resulted in occa-
sional implant rupture, extracapsular silicone
extravasation, and surgeon injury (game-
keeper’s thumb). In addition to long-term fail-
ure, the technique could potentiate liability risk
if future rupture was detected. Contractures
today are more commonly corrected through a
limited, outpatient, open capsulotomy, whereby Figure 11–2. Left breast class III capsule contracture.
176 BREAST CANCER

Silicone and saline implants are radio-


opaque on mammography and have led to con-
cerns regarding potential delay in breast cancer
detection.39–40 Implant characteristics, which
may affect the sensitivity of standard mammo-
graphy, include implant size, the proportion of
overlying breast tissue, implant placement
(subglandular versus submuscular) and the
presence and immobility of capsular contrac-
ture.41–42 As recommended by the American
Cancer Society and the American Society of
Plastic and Reconstructive Surgeons, women
with breast implants should maintain the same
schedule of mammography as all other women.
Figure 11–3. Excised implant and enveloping capsule con-
tracture. They should secure a certified facility that has
sufficient experience with breast implants and
confirm the availability of displacement mam-
Gradual attenuation of the elastomer shell, with mography (Eklund) and ultrasound. Patients
imperceptable rupture, is well documented. with postmastectomy implant reconstruction
Abrupt or premature rupture may be prompted are typically followed by physical examination
by capsular contracture, implant shell infolding only. One major epidemiologic study has con-
leading to accelerated stress fractures, and firmed that the stage at breast cancer detection
trauma. Once a gel implant shell ruptures, from in women with implants is identical or better
longevity or trauma, the contents are usually than it is in the general population;43 a second
contained within the surrounding fibrous cap- major study from the National Cancer Institute
sule. This is likely to remain undetected and has will be addressing this question as well. In
demonstrated no systemic effects. addition, there is no evidence that silicone is
Post-traumatic change in the form of herni- carcinogenic in humans. In fact, in two large
ation, deflation, malposition, or deformation studies women with implants exhibit 10 to 30
may manifest extracapsular extravasation. percent less breast cancer than would be statis-
When this occurs, free gel may infiltrate breast tically expected when matched with the general
parenchyma and tissue planes, and/or elicit a population; the results, however, did not show
granulomatous foreign body reaction. This statistical significance.44–47 This issue needs
may lead to regional silicone migration, sili- further study with larger numbers of patients.
cone mastitis, and formation of irregular nod- The most recent large study, sponsored by the
ules that may, on physical examination and NCI (in press), shows an incidence no different
mammography, simulate a malignancy.37 Sus- than for the matched control group.
pected implant rupture warrants evaluation. In 1992, a series of poorly documented case
Magnetic resonance imaging has a greater sen- reports and the subsequent intense media
sitivity than do either mammography or ultra- scrutiny, combined with a temporary suspen-
sound and is the test of choice for detecting sion of silicone gel implant usage by the FDA,
implant rupture.38 Early removal of the free sil- led to lawsuits and an eventual multibillion dol-
icone and implant, with or without implant lar settlement with the major implant manufac-
replacement, will help to avoid these sequellae turers. Only one implant company (Mentor)
and minimize subsequent confusion in mam- was allowed to provide gel implants for recon-
mographic screening. struction patients, with specific and rigid crite-
Breast Reconstruction 177

ria on a highly monitored, investigational basis. biopsy incisions, and a resultant, variable, ipsi-
There were a plethora of syndromes, autoim- lateral skin deficiency. Immediate reconstruc-
mune diseases, and symptoms associated with tion requires an initial assessment of skin flap
silicone breast implants, and intense litigation vascularity, trauma, and tension. Only the
followed. Many of these proposed associations, healthiest skin flaps should signify proceeding
such as rheumatoid arthritis, were, in fact, with immediate implant reconstruction. Ques-
shown in subsequent, large retrospective stud- tionable vascularity, or marginal necrosis, war-
ies to occur in a lesser percentage of augmented rants reappraisal and the choice of an alterna-
patients than in the general population. Sclero- tive option, such as an immediate expander or
derma-like syndromes were not shown to be autologous flap reconstruction, or delayed
associated with breast implants. The American reconstruction. Compromised flaps, and/or
College of Rheumatology issued on October 22, insertion of a large implant under tension, risks
1995, the following statement, based on accumu- dehiscence and implant exposure. Avoidance of
lated data: “Studies provide compelling evidence tight compressive dressings and constricting
that silicone implants expose patients to no bras, prompt drainage of hematomas or sero-
demonstrable additional risk for connective tis- mas, and early revision of marginal necrosis
sue or rheumatologic disease.” None of the pos- will minimize complications.
tulated syndromes have withstood the scrutiny of Implant position is determined by the
prospective epidemiologic testing.48–50 Results dimension and esthetics of the contralateral
from a large National Cancer Institute study breast. The position of the inframammary fold,
are still pending. breast base width, volume, and overlying skin
The FDA has recently submitted its require- redundancy, or ptosis, are critical in attaining
ments for submission of a “premarket approval” optimal symmetry with the native breast. The
which will, once again, enable marketing of gel IMF may be lowered up to 2 cm when attempt-
implants. The protocol requires patient moni- ing to simulate limited contralateral ptosis.
toring during an 18-month follow-up, and sub- Alternatively, a concurrent, or delayed con-
mission of a limited questionnaire. tralateral reduction or mastopexy may maxi-
mize esthetic outcome and symmetry.
PRIMARY IMPLANT RECONSTRUCTION Delayed implant reconstruction is a safer
and more popular option. The well-healed skin
One-stage primary implant reconstruction, the flaps are elevated in the subpectoral plane and
workhorse of breast reconstruction in the may be stretched, thinned, and scored to pro-
1980s, has become less frequently used due to vide improved projection without regard to vas-
improved outcome with expander or autologous cular compromise. The final outcome may be
reconstruction. Certain patients with A- to B- similarly improved by a symmetry procedure.
sized breasts, having limited to no ptosis, suffi- One-stage implant reconstruction is an
cient soft-tissue coverage, and who desire an option ideally suited to the rare patient with
expeditious and simplistic approach to breast small to moderate sized nonptotic breast who
restoration, may remain candidates for either possesses sufficient soft tissue coverage and
immediate or delayed single-staged implant who desires the simplest reconstructive option.
reconstruction. Even in this group, however, a Despite an initial desire to avoid a secondary
more natural shape can be achieved by an procedure, many patients require future implant
expander with a removable valve, that may also adjustments or symmetry procedures. This tech-
serve as a permanent implant. nique has been largely supplanted by adjustable
Inherent to mastectomy are resection of the and permanent expanders/implants and the pop-
nipple-areolar complex, inclusion of adjacent ular, time-tested, two-stage expander technique.
178 BREAST CANCER

ADJUSTABLE Poor candidates for implant reconstruction


IMPLANT RECONSTRUCTION are those with large, pendulous breasts. These
women, often obese, represent a challenge with
Adjustable implants or permanent expanders/ any technique and are unlikely to achieve satis-
implants represent an option intermediate to the factory symmetry without a contralateral reduc-
single-stage implant reconstruction and the tion or mastopexy. Prior radiation treatment is a
more conventional two-stage technique (Figure strong relative contraindication. The fibrotic
11–4). Postoperatively, adjustable implants and relatively ischemic nature of radiated skin
enable precision in symmetry and the ability to flaps resists expansion and tolerates an under-
attain a softer, often larger reconstruction with lying implant poorly, with a tendency towards
greater ptosis. The technique offers protection cutaneous erosion and exposure. These patients
against tension-related wound complications are better served with either an autologous or
and is generally considered preferable to pri- composite reconstruction.
mary implant reconstruction. It offers an excel- There are two types of adjustable prostheses
lent alternative to patients with limited skin currently available. One is a round and anatomic,
deficits, A- to B-sized contralateral breasts, textured or smooth, postoperatively adjustable,
and/or those patients who require only limited saline implant. The implants are successively
expansion. In addition, a second-stage implant expanded, with saline, by percutaneous injec-
exchange is often avoided. tion through a remotely positioned subcuta-
Indications for use of an adjustable implant neous injection port. Alternatively, Becker
include patients with an immediate or delayed expander/prostheses are composed of a dual
soft tissue deficit, with mild to moderate ptosis, chamber system. The inner lumen, like the
or who require salvage after failed primary Mentor implant, is filled and expanded with
implant reconstruction. Patients with asymmet- saline through a self-sealing, removable injec-
ric deformities from hypoplasia, trauma, burns, tion port. The outer lumen is factory sealed
or congenital deformities (including pectus with silicone gel. It provides patients with the
excavatum and Poland’s syndrome) are also tangible advantages and surface camouflage of
ideal candidates. Adjustability is beneficial in silicone and the postoperative adjustability of a
augmentation candidates with inherent paren- saline implant.
chymal asymmetries or tuberous breasts, or in Once optimal size and shape have been
patients with an unpredictable or poorly com- attained, as confirmed by patient and surgeon,
municated esthetic standard. and sufficient time for capsule maturation has
been allowed (4 to 6 months), the ports may be
removed under local anesthesia, usually through
a short segment of the lateral mastectomy inci-
sion. Vigorous retraction of the connecting tube
engages a self-sealing valve and prevents leak-
age of intrinsic saline. Removal of a small vol-
ume of saline prior to port removal may optimize
implant softness and simulate ptosis.
Preoperative considerations include accu-
rate assessment of size and base diameter.
These determinations may be aided by the use
of templates, sizers, and by the weight of the
mastectomy specimen. Preoperative markings
Figure 11–4. Bilateral reconstruction with adjustable expander/
prosthesis. should detail breast margins in the immediate
Breast Reconstruction 179

reconstruction setting or, in delayed reconstruc- cent skin through temporary overexpansion.
tion, mirror the contralateral breast. Expanders, presently available for immediate
Total muscular coverage, in the immediate breast reconstruction, enable focused expansion
reconstruction setting, will reduce the inci- and simulation of a realistic inframammary fold,
dence of implant exposure, infection, and cuta- without the physiologic, donor site, and rehabil-
neous complications. Sufficient coverage and itative demands of autologous reconstruction.
implant camouflage is provided by a submus- Second-stage exchange with either saline or sil-
cular pocket composed of the pectoralis, serra- icone implants is a simple, outpatient proce-
tus, and rectus muscles. The deflated implant is dure. Tissue expanders remain the most popular
placed, precisely, within the muscular pocket method of immediate breast reconstruction.
and cleared of redundant folds. The injection The advent of textured surfaces has some-
port is connected, drawn through the lateral ser- what lessened the incidence of capsule contrac-
ratus fibers, and fixed to the lateral chest wall. ture, has limited the incidence of perioperative
Patency of the filling system should be con- migration, and has resulted in more predictable,
firmed. Skin flap viability must be assessed successful results. The introduction of anatomic
prior to closure and all questionable skin expanders and implants has enabled preferen-
resected. The implants may be expanded to tial expansion of the lower pole, to better simu-
obliterate dead space but should not further late natural ptosis. Over longer follow-up, how-
stress the muscular or cutaneous closure. ever, there are still significant limitations in the
Expansion is usually initiated 7 to 14 days ability to consistently achieve a natural shape
postoperatively, following confirmation of skin and soft breast.
flap viability. The frequency and extent of each Although all patients who undergo a mastec-
expansion is dependent upon wound healing, tomy may be considered candidates for expander
skin sufficiency, and the patient’s tolerance and reconstruction, preferred patients are those with
comfort level. Typically, saline is injected to the smaller, minimally ptotic breasts. Some of these
point of tolerable skin tension, without blanch- patients may exhibit sufficiently vigorous skin
ing, on a weekly basis. Maintenance of the flaps to accommodate a primary implant recon-
implant at maximum volume for a minimum of struction. Those with limited deficits, and par-
3 months allows for capsule maturation. The ticularly those who increasingly undergo skin-
implant may then be adjusted, within a narrow sparing mastectomies, are candidates for the use
range, prior to port removal, to optimize con- of newer adjustable implants. The two-staged
sistency and ptosis. approach is a reliable, predictable reconstruction
and has the ability to incorporate maximal adja-
IMMEDIATE TWO-STAGE BREAST cent skin, achieve greater volumes and ptosis,
RECONSTRUCTION and enable patient-directed modifications. Final
refinements, including fold adjustments and
Tissue expansion in breast reconstruction was capsulotomy, are facilitated at the time of
pioneered through the efforts of Chadomer implant exchange, optimizing the esthetic result.
Radovan and initially reported in 1976.51 The Conversely, patients with large or pendulous
postmastectomy defect lacks both skin for cov- breasts require greater, more prolonged expan-
erage and the underlying breast mound. To be sion and often a contralateral symmetry proce-
reconstructed, the skin envelope must have a dure to achieve an acceptable result.
adequate laxity to allow the breast mound to Prior or anticipated chest-wall radiation
project sufficiently, achieve symmetric ptosis, after breast conservation or mastectomy remains
and remain soft in consistency. These goals a strong relative contraindication to immediate
often require the recruitment of substantial adja- expander reconstruction. Cutaneous radiation
180 BREAST CANCER

fibrosis resists effective expansion, limits ulti- adjuvant chemotherapy is imposed, pending
mate projection, and increases the risk of cap- wound stability at initiation. When imple-
sule contracture, skin flap necrosis, implant mented, completion of a chemotherapeutic reg-
exposure, and infection. Patients with compro- imen and granulocyte recovery is usually
mised wound healing ability, such as those with required prior to the second stage.
scleroderma and lupus, may also benefit from If adjuvant radiation is deemed necessary
alternative methods. subsequent to expander placement, full, prera-
Textured, anatomic expanders with inte- diation expansion, with preferably a 15 to 20
grated ports are preferable. Smooth-surfaced percent overcompensation, helps resist the
expanders have been shown to result in an fibrotic contracture associated with radiation.
unacceptable rate of capsule contracture, com- All patients undergoing postreconstruction
promising effective expansion.52 Anatomic radiation, however, risk radiation-induced
expanders enable preferential expansion of the implant complications. Radiation-induced cap-
lower pole, a more realistic shape, and greater sule contracture affects the quality of the
projection. Use of an integrated port affords expansion, often leads to local chest wall dis-
greater patient comfort, in that the overlying comfort, and potentiates the risk of expander
mastectomy skin flaps are usually anesthetic. extrusion or exposure. Patients should be mon-
The integrated ports, when compared to remote itored throughout their course and the expander
ports, also incur a lower rate of malfunction. incrementally deflated if skin flap compromise
Simulating contralateral base width and the is noted. Treatment options for patients with
height of maximal projection are the key ele- radiation-induced complications include expand-
ments in attaining optimal cosmetic outcome. er removal and delayed reconstruction, salvage
The goal is to accomplish the major surgical by autologous replacement (TRAM or latis-
steps at the first procedure, which requires simus), and, occasionally, delayed capsulotomy.
careful analysis of the contralateral breast. Sim- Patients with large, smooth implants seem to
ulation of ptosis can be accomplished through show the worst response.
overexpansion of the skin envelope and subse- Complications of expander reconstruction
quent deflation or secondary replacement with parallel those of primary and adjustable implant
an implant of lower vertical profile. Moderate reconstruction53–57 (Table 11–3). Advantages and
ptosis can be simulated through the use of over- their ranges are illustrated in Table 11–1. Cap-
expansion and anatomic expanders, and by sular contracture remains the single most trou-
lowering of the inframammary fold. Moderate blesome complication and is reported in 10 to
to severe ptosis can not be accurately matched 25 percent of patients.58–59 Progressive contrac-
and usually necessitates either a contralateral ture may lead to asymmetry, deformation, and
reduction or mastopexy or composite recon- pain, and require intervention, such as capsulo-
struction using the latissimus dorsi myocuta- tomy, in 20 percent of cases.
neous flap. Contralateral procedures are usually The advantages of prosthetic breast recon-
more precise when based upon the quality and struction include the ability to attain a reason-
extent of expansion achieved and are therefore ably good esthetic result without a complex,
preferentially performed during the second prolonged operation and hospital stay. Compro-
stage. The end point of the expansion process mising a donor site, with its potential compli-
occurs when adequate projection is achieved in cating morbidity, is also avoided. Prosthetic
relation to the contralateral breast, rather than reconstruction remains appealing for bilateral
the ultimate volume being attained. cases in which symmetry is less of a problem
Patients and their reconstructions are not and where bilateral autologous reconstruction
adversely affected if concurrent expansion would impose substantial demands on the
Breast Reconstruction 181

patient and surgeon. Similarly, in patients with Table 11–3. IMPLANT COMPLICATIONS

smaller breasts, in older patients, and in those No XRT (%) XRT (%)
less motivated, expander or implant reconstruc-
Implant loss/extrusion 3.4–18 4–10
tion remains a desirable option. Deflation 3–4
Infection 1.2–8 10
Capsule contracture 2.9–31 20
ADVANTAGES OF AUTOLOGOUS Skin necrosis 10–24 3–7
RECONSTRUCTION Satisfaction 80–98 49–55

XRT = external radiation beam therapy


Breast reconstruction with a silicone- or saline-
based implant is technically the simplest option
available to mastectomy patients. Recent apparent when lying supine, when the recon-
advances enhancing the potential esthetic out- structed breast remains fixed and projecting
come include permanent expander prosthesis while the native breast falls naturally to the
and postoperatively adjustable implants. Most side. This represents the most common adverse
competent surgeons can insert a prosthesis, postoperative development, occurring in 20 to
postmastectomy, in a wide range of patients. 40 percent of all mastectomy patients and requir-
Consequently, prosthetic reconstruction is the ing operative intervention in up to 20 percent of
most common mode of breast reconstruction cases.58–59 Implant-based reconstruction may,
available today. therefore, be a less strategic option for younger
Prosthetic reconstruction is safe and expedi- patients. Kroll reported on 325 postmastectomy
tious, with a limited recovery period. It is suited patients who had undergone either expander
to the patient desiring a simple approach toward or autologous reconstruction. Complications
breast restoration. Candidates include those occurred in 23 percent of expander patients,
who wish to avoid an external prosthesis, those compared to 9 and 3 percent in latissimus and
with limited expectations, those with smaller TRAM flap reconstructions, respectively.60
breasts and limited-to-no ptosis, those with Implants are devices and are susceptible to
existing medical risk factors and anxious device failure. It has been well demonstrated
patients who have difficulty comprehending that the silicone elastomer shell of both sili-
more technical procedures, and those desiring cone and saline implants fatigue over time.
an expeditious initiation of adjuvant treatment. This may manifest itself as either a silicone or
Expander/implant reconstruction may also saline bleed and/or leak. An intracapsular sili-
pacify younger patients who wish to ultimately cone implant rupture is likely to remain unde-
convert to autologous reconstruction following tected until some adverse event occurs. Most
anticipated pregnancies. commonly, this may involve an increased ten-
Implant-based breast reconstruction, how- dency toward progressive capsular contracture.
ever, has many disadvantages. The implant, Blunt trauma resulting from a car, bicycle, or
which is clad only by a thin layer of skin and rollerblade incident, or even an overzealous
muscle, is often poorly camouflaged and leads mammogram, may convert a contained rup-
to a round, “mechanical,” unnaturally aptotic ture into an extracapsular rupture. Patients
and asymmetric replacement. Peri-implant cap- typically notice a change in the shape and/or
sule contractures may impose further distor- volume of the implant. This scenario warrants
tion, migration, asymmetry, and discomfort. either mammographic, ultrasonic, or MRI
Capsular fibrosis limits the fluidity of both imaging to rule out rupture.38 Conversely, rup-
saline and silicone implants. It is noticeable ture of saline implants leads to implant defla-
upon palpation and in its inability to react nat- tion and a flat breast. In either case, implant
urally to positional changes. This is especially replacement is warranted.
182 BREAST CANCER

In contrast, autologous tissue has the CONVENTIONAL TRANSVERSE


warmth, consistency, feel, and reactive mobility RECTUS ABDOMINIS
of one’s own tissues. It is a malleable, con- MYOCUTANEOUS FLAP
formable, permanent medium that does not
elicit a foreign body fibrotic response and is The transverse rectus abdominis myocutaneous
more tolerant of adjuvant therapy, trauma, and flap, one of the most ingenious techniques in
infection (Table 11–4). In contrast to the greater plastic surgery, has established itself over time as
contracture and rupture rates of implants, autol- the flap of choice for autogenous breast recon-
ogous tissue softens and ages commensurate struction. It presents the reconstructive surgeon
with adjacent structures and is therefore an with the opportunity to a create a breast of
ideal option for younger patients. An autolo- unsurpassed esthetic beauty, is unparalleled in its
gous flap may be contoured to match a con- ability to simulate the opposite breast, and sec-
tralateral breast of almost any size and shape. ondarily improves the contour of the lower
Although the initial overall cost of the flap abdomen. Attaining consistently good results
reconstruction is greater, the long-term costs of requires careful planning and technical profi-
autologous reconstruction have been shown to ciency. The lower abdomen consistently provides
be less than those of prosthetic reconstruction exceptional and sufficient tissue for unilateral
due to subsequent secondary capsulotomies, and, in the majority of patients, bilateral breast
revisions, and implant exchanges required with reconstruction. The procedure is versatile and
the latter procedure. reliable when performed within its recognized
Autologous reconstruction is inherently vascular and volumetric constraints. Hartrampf’s
more complex from both a technical and an landmark introduction of the TRAM flap in
artistic standpoint. The functional and esthetic 1982, still the gold standard for autologous
outcome of the initial procedure, which lasts breast reconstruction, provided the foundation
from 4 to 5 hours, largely depends upon the sur- for the modern era of breast reconstruction.61
geon’s experience and/or microsurgical exper- The conventional, unipedicled TRAM flap,
tise. Although the initial procedure requires a as originally described, consists of a transverse
longer hospitalization (3 to 4 days) and postop- ellipse of skin and fat based on one rectus
erative recovery, the result is permanent and abdominis muscle and its intrinsic musculocu-
rarely requires a secondary adjunctive proce- taneous perforators from the superior deep epi-
dure. The TRAM flap is overwhelmingly the gastric pedicle. The pedicle branches as it trans-
flap of choice when available. Alternatives gresses through the substance of the ipsilateral
include the latissimus dorsi, Ruben’s or peri- rectus through a network of “choke” vessels,
iliac, lateral thigh, and gluteal flaps. which reconstitute in the midabdomen.61–65
This inflow communicates with the periumbili-
cal, myocutaneous perforators that supply the
Table 11–4. ADVANTAGES OF suprafacial and subcutaneous plexuses. Bost-
AUTOLOGOUS RECONSTRUCTION
wick has determined that blood flow in the con-
Soft ventional TRAM is based upon pedicle caliber,
Warm number of perforators, integrity of the
Pliable
Permanent
suprafascial plexus across the midline, and
Enables wide resection venous outflow.64–65 Perfusion has been graded
No foreign body response and is depicted as a sequence of zones, with
Natural consistency and appearance zone VI, the most distal tissue, representing
Tolerates adjuvant therapy well
Decreases need for symmetry procedure
strictly random perfusion (Figure 11–5). Flow
More economic in the longterm in the conventional TRAM is, therefore, sec-
Breast Reconstruction 183

ondary and unpredictable beyond the midline. plementary perfusion through a microvascular
Patient selection is critical and is limited, anastomosis, if intrinsic vascular insufficiency
among experienced surgeons, to those patients is noted. The flap is “supercharged” through an
who have tissue requirements met by the ipsi- anastomosis, most commonly to an axillary
lateral “hemi-TRAM.” recipient pedicle.
In the uncomplicated case, the flap extends Fascial donor site closure is achieved with
from the umbilicus to a point superior to the either interrupted figure-of-eight sutures or a
pubis. The incisions are beveled, after isolation running, heavy, braided, synthetic. The patient
of the umbilicus, to incorporate additional peri- is then flexed to 45° to facilitate abdominal clo-
umbilical perforators and subcutaneous fat. The sure and ascertain breast symmetry. Typically,
flap is elevated at the suprafascial level toward two drains are placed, both at the breast and
the medial and lateral row of ipsilateral muscu- abdominal sites. Postoperative flap monitoring
locutaneous perforators. The fascia is incised, is institution-specific and may encompass tem-
immediately adjacent to the perforators, facili- perature probes, ultrasound or laser doppler,
tating subsequence closure, and the underlying and clinical surveillance.
rectus is mobilized. Optimal perioperative conditions are para-
Most commonly, a full width muscle harvest mount to early and late flap success. Patient
with a thin strip of fascia is performed. The rec- core temperature, intravascular fluid status,
tus muscle is elevated beneath the superior anxiety and pain level, and position may all
abdominal skin flap to the costal margin. The have an impact on final outcome. If the start
superior epigastric pedicle is easily identified, time is late in the day or there is minimal urine
enabling transection of the lateral rectus fibers
as well as of the intercostal nerves. This facili-
tates muscle atrophy and, thereby, minimizes
the central xiphoid bulge, common initially
after this procedure. The flap is transposed
through a subcutaneous tunnel, which under-
mines the medial IMF, and is inset into the
breast defect. Zones IV and II may be discarded
prior to transposition to facilitate passage.
In an effort to preserve abdominal wall
integrity, an alternative “split-muscle” harvest
has been advocated.66–68 Pedicle (muscle)
width is based upon the laterality of the medial
and lateral row of perforators. It is usually pos-
sible to preserve a substantial (one-third) width
of the lateral rectus and often a slip of infraum-
bilical medial rectus. Although the muscle is,
in most cases, denervated, it is thought to
uphold the muscular interface of the semilunar
line and adds fibrous stability in the perioper-
ative period.
It is common practice to include a segment
of skeletonized inferior epigastric pedicle in the
event additional perfusion is necessary to sus-
tain the flap.69–70 This “lifeboat” enables sup- Figure 11–5. TRAM flap zones.
184 BREAST CANCER

upon foley placement, an initial fluid bolus may lack of the abdominoplasty effect inherent in
be required. An intraoperative hourly urine out- conventional TRAM.
put of 50 cc should be maintained to ensure Although largely supplanted by microsurgi-
adequate flap perfusion. In addition, mainte- cal advances, preoperative surgical delay of a
nance of normothermia may minimize vascular conventional TRAM is another technique for
vasoconstriction, spasm, and shivering, all of augmenting reliable flap dimensions.79–82 In
which may have an adverse impact on immedi- 1995, Codner demonstrated improved inflow
ate postoperative flap perfusion. The use of and diminished congestion after surgical
heating blankets and fluid warmers is routine. delay.82 Zones II and III proved more vigorous
Use of the superiorly based unipedicle and reliable, especially in the high-risk patient,
TRAM flap requires strict adherence to patient and lessened the need for a bipedicled approach.
selection criteria. Obtaining consistent results In 1997, Restifo documented greater pedicle
demands an assessment of potential risk fac- caliber (1.3 versus 1.8 mm) and flow rates (7.5
tors. It has been clearly demonstrated that versus 18.2 mL/min) compared to controls after
patients who smoke, are obese, have significant surgical delay.83 Staged interruption of the infe-
abdominal scarring, or have had previous radi- rior epigastric pedicle, on a physiologic basis, is
ation have an increased risk of complications, highly effective in augmenting vascularity and
including fat necrosis, partial flap failure, and is beneficial in the high-risk patient.
donor site complications.71–75 These risk factors Scars within the confines of the harvested
should not eliminate patients from the proce- flap, such as appendectomy, hernia, and mid-
dure so much as indicate modification to line scars, represent an ischemic boundary and
enhance blood supply to the transferred tissue. will diminish the volume tissue available, due
For instance, nicotine from cigarette smoking to variable vascular disruption. Regional inci-
has been recognized as a potent vasoconstrictor sions, such as paramedian, cholecystectomy, or
of the microcirculation. Patients who smoke or extended Pfannenstiel’s incisions may directly
are unable to abstain 4 to 6 weeks prior to disrupt the pedicle and potentiate donor site
surgery are at extremely high risk for partial complications. Shaw and colleagues assessed
flap and donor site necrosis.76 These patients, complications among TRAM patients with pre-
likely to fail conventional reconstruction, often existing scars.75 In 43 percent of the patients,
succeed under the preface of a delay, bipedicle, abdominal wall weakness, partial flap loss, fat
or free technique. necrosis, and donor site morbidity developed.
Alternatively, the midabdominal TRAM Paramedian scars precluded use of the free
was devised in response to a 20 to 60 percent of TRAM in three of three patients. Cholecystec-
partial flap loss or fat necrosis and a high rate tomy scars and multiple scars showed the high-
of hernia and abdominal wall weakness in high- est propensity toward skin-related complica-
risk patients.77–78 This flap is based on the per- tions. Conservatism, and often an alternative
forator-rich periumbilical region, and extends flap design, are warranted in patients with pre-
inferiorly to a tangent parallel to the anterior existing scars.
superior iliac spine (ASIS). Because muscle Operative assessment of the contralateral
integrity is preserved below the arcuate lines, a breast helps in formulating a reconstructive
lower incidence of hernia may be anticipated. strategy and optimizing symmetry. The location
Slavin’s review of 236 midabdominal flaps and breadth of the IMF is a critical landmark
showed a 2 percent rate of partial flap necrosis and serves as the basis for building a symmet-
and a 1 percent incidence of fat necrosis.77 The ric breast. Attention to the condition and vol-
primary disadvantage is an occasionally dis- ume of retained skin (mastectomy skin flap),
pleasing high- or midabdominal scar and the the size, shape, base width, and ptosis of the
Breast Reconstruction 185

contralateral breast and how it relates to the and fat necrosis, and are inherent to the pro-
IMF is necessary if symmetry is to be opti- cedure’s secondary blood supply and volume
mized. Patients with pre-existing macromastia constraints.57,66,73–76,84–86 These complications
may elect to undergo concurrent or delayed may impose prolonged wound healing and con-
contralateral breast reduction, both to alleviate siderable delay in the therapeutic sequence.
objective symptoms (shoulder pain and groov- Although the free TRAM procedure requires
ing, intertrigo, lower back pain) and improve greater technical proficiency and a slightly
the ultimate esthetic outcome. Patients with longer operating time, the flap has unparalleled
substantial glandular ptosis may elect to vascular reliability and versatility, and is the
undergo mastopexy for similar reasons. It is the flap of choice in high-risk patients. These
current authors’ preference to perform these include obese patients, smokers, and those
contralateral procedures as a second stage. patients with prohibitive scars or who have had
Improved accuracy of a symmetry procedure prior radiation treatment.73–76
may be attained after resolution of flap edema, Suprafascial elevation is identical to that
muscle atrophy, and skin retraction. Staging employed in the pedicled TRAM procedure.
also enables concurrent refinements (SAL, Widely dispersed perforators may be omitted
IMF revision) on the recently restored breast. due to the dominant inflow, thereby limiting the
The goal of reconstructive surgeons using fascial harvest. The lateral edge of the rectus
the TRAM flap for breast reconstruction is to muscle is elevated to discern the path of the
increase the total efficiency, reduce operative epigastric pedicle. This determines whether a
morbidity, and to be able to offer patients an medial and/or lateral muscular strip may be
absolute minimum complication rate and hos- preserved. The pedicle is ligated at the external
pital stay. Experience has demonstrated the iliac origin. Axillary recipient vessels are nor-
vascular and volumetric constraints of the mally reliable, even if prior radiation therapy
pedicled TRAM flap and led to technical has been implemented. Preference, in decreas-
refinements that are dependent upon individ- ing order, include the thoracodorsal, the cir-
ual patient risk factors. cumflex scapular, lateral thoracic, and internal
mammary vessels. This last option requires a
FREE TRANSVERSE RECTUS peristernal costochondrectomy at the third rib
ABDOMINIS MYOCUTANEOUS FLAP interface for adequate exposure. An interrupted
or running arterial microvascular anastomosis
The free TRAM flap, based on the dominant is typically performed with 9.0 nylon suture.
inferior epigastric blood supply and requiring a Venous anastomosis may be similarly per-
microvascular anastomosis, represents a reli- formed, or one may use an anastomotic cou-
able, versatile, highly esthetic option for both pling device (3M) for added speed. The occlud-
immediate and delayed reconstruction. The ing clamps are removed, and the quality of flap
rationale for employing this option stems from perfusion is confirmed both clinically and with
its benefits in immediate reconstruction, where the use of an intraoperative doppler.
the ultimate goal of the reconstructive surgeon Advantages of the free TRAM flap (Figures
is to provide a highly desirable restoration, 11–6A, 11–6B) include a more limited abdomi-
minimal complications, and an expeditious nal harvest site, which corresponds to a more
recovery that will remain invisible to the adju- expeditious, often less uncomfortable postoper-
vant therapeutic sequence (see Figure 11–5). ative recovery. The volume of rectus muscle
Complications associated with conventional harvested may be limited to a small cuff of
TRAM reconstruction, occurring in up to 25 fibers surrounding the perforators transgressing
percent of reported cases, are partial flap loss through the rectus muscles to supply the
186 BREAST CANCER

suprafascial and subcutaneous plexuses. Thus, a of high-risk patients (63 versus 28%), the free
medial and/or lateral strip of rectus muscle may TRAM group had fever complications (9 ver-
be preserved, which benefits young, active sus 28%) than the conventional TRAM group.
patients or those desiring future pregnancies. The advantages of the free TRAM procedure
Use of the free TRAM enables preservation of are outlined in Table 11–5.
an inframammary fold, does not compromise The main disadvantage of TRAM flap recon-
the marginal perfusion of the freshly elevated struction is the potential for weakening the
mastectomy skin flaps, optimizing the esthetic abdominal wall. Questions remain as to the best
outcome. Freedom upon insetting, due to the technique of abdominal closure and the impact
absence of the conventional muscular “leash,” of free versus pedicled flap reconstruction on
facilitates a quick, easy, and highly cosmetic the abdominal wall. Despite all the advantages
and symmetric reconstruction (Figures 11–7A, of the TRAM, it is a major surgical procedure
11–7B). The improved blood supply may expe- and carries the risk of abdominal weakness,
dite wound healing and initiation of adjuvant bulging, and hernia formation. True hernias
therapy, which may also be better tolerated than resulting from the procedure are extremely rare
in the conventional TRAM flap procedure. (< 3% of cases). Abdominal wall bulges, indi-
The only absolute contraindications to free cating a separation and attenuation of the inter-
TRAM reconstruction include prohibitive scar- nal and external oblique muscles, occur more
ring, violation of the inferior epigastric blood frequently (3 to 12% of cases).
supply from previous abdominoplasty, suction Several studies have obtained objective mea-
lipectomy, extended Pfannenstiel’s incision, or sures of abdominal muscle strength. Trunk mus-
previous TRAM procedure. Pre-existing med- cle strength as measured by an isoknetic dyna-
ical conditions may limit the patient’s ability to mometer demonstrated postoperative recovery
tolerate 4 to 6 hours combined anesthesia time. of 92, 96, and 98 percent at 3, 6, and 12 months,
This should be addressed preoperatively. respectively, for unilateral free TRAM flap
In a series of 211 free TRAM flaps, Schus- patients. Although the ability of one-half of the
terman reported a flap thrombosis rate of 3.3 group to perform situps was not affected, the
percent and a flap loss rate of 1.4 percent.87 other half demonstrated mild impairment.88
One study compared outcome among conven- Kind and colleagues compared the recovery
tional and free TRAM reconstruction. It was after pedicled and free TRAM reconstruction.
demonstrated that despite a higher percentage Flexion torque as measured by dynamometer

A B

Figure 11–6. A, Free TRAM reconstruction; B, 9 months postoperative.


Breast Reconstruction 187

A B
Figure 11–7. A and B, Right free TRAM reconstruction prior to nipple/areolar reconstruction.

was 58 and 89 percent at 6 weeks and 6 months, lined below, appear to have a positive impact.
respectively, for conventional TRAMs. In con- Approximation of the medial and lateral
trast, the free TRAM showed 89 and 93 percent remnants of the tendinous inscriptions appears
of preoperative flexion recovery at 6 weeks and to “restore the ribs” of abdominal support and
6 months respectively. The investigators con- relieve tension on the anterior rectus sheath.92
cluded that the pedicled TRAM caused a signif- Primary repair of the fascial defect should
icantly greater insult to the abdominal wall in the include approximation of the underlying mus-
early postoperative period but that the two tech- cular support. Kroll advocates approximation
niques equilibrated to over 90 percent of preop- of the anterior remnant of the internal oblique
erative levels at 12 months. It was also deter- to the linea alba and a reinforcing two-layer
mined that muscle splitting techniques appeared closure. Fascial sheath closure is strengthened
to offer no functional advantage.89 with sutures through the semilunar line.93
The ability of patients to perform situps after Abdominal wall complications are probably
various modes of TRAM reconstruction has best avoided through recognizing excessive
been studied. Percentages of patients able to tension during closure or undue attenuation of
perform a situp were 63.0, 57.1, 46, and 27 per- weakened fascia. The threshold for mesh rein-
cent for the single free TRAM, conventional forcement should correlate positively with
TRAM, bilateral free flap, and conventional these findings. Mesh enables reinforcement
flap, respectively.90 Fitoussi reported that 47 without excess tension and may facilitate post-
percent of single pedicle TRAM and 0 percent operative mobility, diminish pain, and expedite
of bipedicle TRAM patients could perform recovery. The infection rate after using mesh is
situps postoperatively and concluded that reported to be < 2 percent and is usually a result
although the hernia rate did not vary between of other miscalculations that result in exposure
the two groups, functional sequellae were statis- through marginal dehiscence or necrosis.
tically significant.91
Hernias occur in 5 to 6 percent of TRAM Table 11–5. ADVANTAGES OF FREE TRAM
patients, regardless of whether the procedure
Primary and dominant blood supply
was conventional or free, uni- or bipedicled. Greater available volume
The incidence appears more closely associated Less muscle harvest/abdominal dissection
More comfortable recovery
with the technique and detail of abdominal clo- More reliable in high-risk patients
sure rather than with the number or extent of Greater freedom in insetting
muscles harvested. Various modifications, out- Good tolerance to adjuvant therapy
188 BREAST CANCER

The other complication that has led to con- much the same manner as the free technique.
siderable investigation and comparison between The use of mesh has markedly reduced the inci-
TRAM techniques is the incidence of fat necro- dence of abdominal hernia formation and
sis. Kroll and colleagues reported in 1998 on the bulging. Although these patients have objective
incidence of fat necrosis among patients who loss of abdominal function, subjective interfer-
had had conventional versus free TRAM recon- ence with daily activity is rare. There are
struction. Of the 49 free TRAM patients, 8.2 reports of an increased incidence of long-term
percent exhibited clinical fat necrosis, with one lower back pain.
patient showing mammographic evidence. Of Use of the bipedicled TRAM for unilateral
the 67 pedicled TRAM patients, 27 percent reconstruction has invoked substantial contro-
demonstrated fat necrosis on examination and versy in the plastic surgery literature. Antago-
nine patients on mammogram.94 nists claim the morbidity from bilateral muscle
harvest, including abdominal wall weakness
BIPEDICLED TRANSVERSE RECTUS and the propensity toward future back pain,
ABDOMINIS MYOCUTANEOUS FLAP “can no longer be defended” in the current
realm of reliable microsurgical capability and
A unipedicled conventional TRAM will reli- surgical delay.97 Conversely, proponents claim
ably perfuse all of zone I, 20 percent of zone II, that the split muscle technique and addition of
and 80 percent of zone III.95 An alternative mesh reinforcement limit functional morbidity
technique or flap choice is warranted if tissue and that the resultant abdominal wall integrity
requirements exceed these specifications. Indi- is dependent upon the closure technique used.98
cations for a bipedicled TRAM include those They adhere to its use as a reliable alternative in
patients who insist on autogenous reconstruc- high-risk patients.
tion, who require additional volume, and for
whom microsurgical reconstruction is not pos- LATISSIMUS DORSI
sible due to an absence of reasonable recipient
axillary vessels. The indications parallel those The latissimus dorsi myocutaneous flap was
for surgical vascular delay. originally described by Tansini in 1906 and
The lower abdominal pannus is isolated on used to cover radical mastectomy defects.99 It
the medial and lateral row of perforators, bilater- has since demonstrated remarkable versatility
ally. Once the upper abdominal apron is elevated, and is useful in providing purely autogenous,
each superior epigastric pedicle is isolated with composite implant, and partial mastectomy
the assistance of doppler mapping, and a split reconstruction (Figure 11–8). The straightfor-
bipedicle muscle harvest is performed. The flap ward anatomy, easy elevation, relative lack of
is transposed and inset in much the same way as donor morbidity, and ability to provide an addi-
for an unipedicled TRAM flap. tional “curtain” of conforming tissue have
Multiple reports have investigated the long- made it a reasonable adjunct to breast recon-
and short-term impact of bilateral rectus har- struction, most commonly in healthy patients
vest. Hartrampf reported that 64 percent of considering expander reconstruction.
patients could not perform a single situp after The indications for latissimus reconstruc-
bipedicled reconstruction, compared to 17 per- tion vary widely and depend upon the prefer-
cent in the unipedicled group. Petit reported a ences and capabilities of the surgeon. Several
20 percent incidence of subsequent severe back subsets exist, all governed by the assumption
pain in bipedicled patients.96 that a TRAM flap has been ruled out for either
The bipedicled flap has reduced the inci- medical, anatomic, or personal reasons. The
dence of partial flap loss and fat necrosis in first set includes those patients who are other-
Breast Reconstruction 189

wise appropriate candidates for expander tissue. These patients typically have redundant
reconstruction but for whom less than optimal flank skin and additional subcutaneous bulk
coverage is predicted. This may include that may be incorporated into the flap to pro-
patients who have had a prior radical mastec- vide necessary volume and ptosis. Conversely,
tomy and lack a pectoralis, those who have thin patients with marked breast hypoplasia may
mastectomy skin flaps, or those who require a also attain sufficient volume, contour, and sym-
large skin resection due to inclusive resection metry from a purely autogenous latissimus
of a remote biopsy site or to prior radiation. myocutaneous flap.
The second set includes those patients The flap has an extremely reliable blood
amenable to expander reconstruction who have supply and is versatile even in smokers and dia-
sufficient coverage and high esthetic expecta- betics. Partial flap necrosis has been reported in
tions. The challenge in unilateral postmastec- up to 7 percent of patients.100 The most com-
tomy expander reconstruction is to provide a mon nuisance is the persistence of seromas,
breast form which simulates the contralateral which often requires prolonged drainage or
side. Prosthetic reconstruction provides a round, aspiration. Implant-related complications
firm, relatively immobile breast form which is include implant slippage and capsule contrac-
ideally suited for patients with small to interme- ture. Use of textured, saline expanders and
diate sized breast and limited to no ptosis. implants has reduced these complications.
Patients who are moderate or large in size and The latissimus dorsi flap represents a popu-
develop some degree of ptosis with age and lar, extremely reliable option for the mastectomy
childbirth will demonstrate variable asymmetry patient. The results are outstanding, when used
with unilateral prosthetic reconstruction. These in conjunction with the newer textured, anatomic
non-TRAM candidates may elect to undergo saline expanders and implants, typically better
either contralateral mastopexy or composite latis- than those achieved with expanders alone.
simus-expander reconstruction for improved
symmetry. The flap provides supplemental mus- RUBENS FLAP
cle and fat, which helps camouflage the under-
lying prosthesis and replaces the resected skin, Peter Paul Rubens was known for his portraits
leading to a more natural ptotic breast form. of females with particular fullness in the
The third category includes those patients suprailiac region. The skin and subcutaneous
who prefer an autogenous restoration but lack tissue in this region may be sustained by the
flap alternatives due to medical or surgical rea- deep circumflex iliac artery, as originally
sons. Most patients have a breast volume in
excess of their available flank tissue and require
supplemental volume in the form of an implant.
The resultant satisfaction in esthetic outcome
and greater projection and natural ptosis allay
most patient’s preoperative reluctance toward a
supplemental implant.
The fourth and fifth sets involve autogenous
latissimus reconstruction without supplemental
prosthesis and apply to two patient extremes
where the available flank tissue volume simu-
lates breast volume. Solely autogenous latis-
simus reconstruction is routinely possible in Figure 11–8. Latissimus dorsi myocutaneous flap recon-
heavier patients having substantial upper flank struction.
190 BREAST CANCER

described by Taylor.101 Hartrampf coined this the muscle layers are cut immediately adjacent
peri-iliac fat pad the “Rubens flap.”102 to the pedicle. The inferior skin flap is elevated
The TRAM is the flap of choice in autolo- above the tensor of fascia lata to the iliac crest.
gous breast reconstruction. One of the benefits Subperiosteal dissection will ensure the
of the TRAM flap is the performance of a con- integrity of both the deep circumflex iliac
current abdominoplasty, with resection of often artery (DCIA) pedicle and perforators. The lat-
large volumes of infraumbilical tissue. It is with eral femoral cutaneous nerve runs inferiorly,
some bewilderment that patients complain of a within 1 cm of the anterior superior iliac spine,
greater lower abdominal circumference, not a and may lie either above or below the DCIA.
reduction, and have greater difficulty wearing This nerve should be preserved.
their previously well-fitted clothing. Closing the Donor site closure is initiated by approxi-
anterior TRAM donor site leads to accentuation mation of the transversalis fascia to the ilio-
of the peri-iliac tissue and can cause an actual psoas fascia. The remaining flank muscles are
increase in the peri-iliac circumference. This secured to the iliac crest through drill holes and
redundancy represents the tissue available for heavy suture or wire.
free tissue transfer after a previous TRAM flap. Deep circumflex iliac artery pedicle length
The predominant indication for use of the facilitates anastomosis to the preferred thora-
Rubens flap is therefore a prior TRAM harvest codorsal vessels in the majority of cases. This
or abdominoplasty. Other indications for use of flap tends to be less robust than the TRAM and
the Rubens flap include thin patients and pro- may exhibit a weak doppler signal, at best. The
hibitive anterior abdominal scars. flap provides excellent projection (Figure
Flap dissection requires a precise knowl- 11–9B) and is an ideal option for bilateral
edge and familiarity with the intrinsic support reconstruction, which may be performed simul-
of the abdominal wall. The primary disadvan- taneously, concurrent with mastectomy.
tage of the flap is the occurrence of an occa-
sional flank hernia. Compulsive closure of the SUPERIOR GLUTEAL FLAP
donor site is paramount to the success of this
procedure and requires a dedicated surgeon to The superior gluteal flap was the first free
do so. Other potential morbidity includes long- flap described for breast reconstruction.103
standing seromas that require prolonged Microsurgical expertise is essential for suc-
drainage and compression garments. Patients cess due to a tedious flap dissection, an inher-
with this problem also have a higher incidence ently short vascular pedicle, and because the
of prolonged discomfort and often require microanastomoses are most commonly per-
monitored physical therapy. formed to the delicate and variable internal
The flap is oriented parallel to the iliac crest, mammary vessels.104
with two-thirds of the skin paddle above and Candidates include patients who fail qualifi-
one-third below the crest (Figure 11–9A). An cation for implants due to prior chest-wall irra-
inguinal incision lateral to the femoral pulse diation or “implant anxiety” or who have
will expose the external oblique. Once this is abdominal scars precluding TRAM reconstruc-
incised, the round ligament is identified and tion. Such scars may have resulted from laparo-
retracted superiorly to expose the inguinal tomies, enterotomies, previous abdominoplas-
floor. An incision through the internal oblique, ties, liposuction, or TRAM harvests. This flap
transversalis, and transversalis fascia will may represent the only autogenous option in
expose the underlying deep circumflex iliac thin patients who lack sufficient abdominal or
vessels. This helps guide the remainder of the lateral thigh tissue for unilateral or bilateral
flap dissection. Once the skin and fat are cut, reconstruction.
Breast Reconstruction 191

Like the TRAM, the gluteal flap offers a per- that are based upon the lateral femoral circum-
manent, soft, warm, and natural reconstruction. flex vessels and make use of the lateral “riding
It has a more dense fat–septal network, provid- breeches” or “saddle bags.”105 The pedicle trans-
ing an intermediate size reconstruction with gresses through and requires the sacrifice of the
excellent projection. It may be the flap of choice modest tensor muscle. Preservation of adjacent
for patients who have had a previous TRAM and fascia lata helps to ensure lateral knee stability
require a staged contralateral mastectomy. It without functional compromise. More impos-
also offers an inconspicuous donor site. ing, and representing the primary disadvantage,
Flap dimensions typically extend from the are the often disfiguring lateral thigh scars,
lateral midsacrum to within 5 cm of the ASIS. which are long and remain poorly camouflaged.
The verticle height of the flap depends on the Preoperative design requires experience and
tissue needed but may vary from 10 to 15 cm. precision. An excessive subcutaneous harvest
Flap dissection necessitates identification of will result in objectionable lateral thigh contour
the fragile superior gluteal vessels deep to the deficits. These are difficult to correct but do
gluteus muscle. This pedicle emerges from the
greater sciatic foramen amidst numerous
branches and provides 1.5 to 2.0 cm of pedicle
length. The internal mammary vessels are
exposed and mobilized through a third perister-
nal rib resection. The external jugular vein,
rotated down from the mandibular angle, may
serve as a venous alternative if internal mam-
mary vein integrity is lacking.
Like its counterpart, the inferior gluteal
flap is indicated in rare patients who refuse
prosthetic reconstruction and who are not can-
didates for either TRAM, lateral thigh, or
latissimus flaps. Although the length of the
donor inferior gluteal vessels enable anasto-
mosis to the more forgiving thoracodorsal
pedicle and the donor site scar is the least con-
spicuous of any autogenous option, harvest
necessitates sacrifice of the gluteal motor
nerve, occasional sacrifice of the posterior
cutaneous nerve, and close dissection to the
A
sciatic nerve, all of which may lead to tran-
sient pain syndromes and weakness with
ambulation; prolonged rehabilitation may be
required. For these reasons, the gluteal flap is
generally the least favored flap in the breast
reconstruction algorithm.

B
LATERAL THIGH FLAPS
Figure 11–9. Ruben’s flap A, Bilateral flap design; B, Imme-
The lateral transverse thigh flap and tensor of diate postoperative projection demonstrated. Reproduced
with permission from William W. Shaw, MD, Division of Plastic
fascia lata flap are two reconstructive variants Surgery, UCLA.
192 BREAST CANCER

benefit from delayed suction lipectomy. Patients patient or the patient with marginal reserve who
may require prolonged drainage and garment desires to avoid an external prosthesis and could
compression to limit the tendency toward not tolerate a long, grueling procedure.
seroma formation. Advantages include a 7 to The TRAM flap, once again, is the flap of
8 cm vascular pedicle, excellent flap projection, choice, providing reliability and minimal mor-
and the ability to perform concurrent bilateral bidity in bilateral autologous reconstruction
simultaneous harvests and reconstruction. (Figure 11–10). Sufficient tissue is present for
bilateral reconstruction in 75 to 80 percent of
BILATERAL patients. The majority of patients when advised
BREAST RECONSTRUCTION of the ability to perform an immediate, single-
stage, highly esthetic and symmetric, perma-
Indications for contralateral prophylactic mas- nent, bilateral autogenous reconstruction, and
tectomy include a strong family history of simultaneously rid themselves of an often per-
breast cancer, positive genetic testing, lobular vasive lower abdominal pannus, are, most
carcinoma in situ (LCIS), cancer anxiety, and often, highly grateful and not overly concerned
equivocal or progressively difficult clinical about the possibility of having slightly smaller
and/or radiographic examinations. breasts if less than profound amounts of tissue
With improvements in breast cancer screen- are available. Advantages of bilateral TRAM
ing, a greater number of early breast cancers are reconstruction include the ability to perform a
being detected in young, premenopausal simultaneous harvest in the supine position.
patients, many of whom have some degree of Either bilateral conventional or free
familial cancer history. Patients with young TRAMS may be performed. The vascular relia-
families present with the intent to absolve breast bility of bilateral “hemi-TRAM” flaps is nor-
cancer risk for the benefit of their young ones mally adequate, because cross perfusion across
and represent a new indication for either pro- the midline is not necessary, depending on an
phylactic or bilateral mastectomy. Breast cancer absence of excessive scarring, obesity, prior
awareness has elevated the level of sophistica- radiation, and history of smoking. Use of the
tion of all patients. Prosthetic and autologous conventional flaps is usually faster and techni-
reconstruction is a known entity that continues cally simpler than free TRAM reconstruction
to become more reliable, safe, and esthetically but requires inherent sacrifice of both rectus
satisfying. As this awareness becomes more muscles, which may lead to objective and sub-
apparent and outcomes improve, it is not sur- jective abdominal wall weakness in the major-
prising that an increasing number of susceptible ity of more active patients. Extensive superior
women are at least questioning the option of abdominal dissection and tunneling is required
bilateral ablation and immediate reconstruction. and may prolong postoperative discomfort and
Esthetic outcome is often better in bilateral recovery. Transposition of bilateral flaps may
reconstruction than in unilateral reconstruction lead to an upper abdominal bulge and violate
due to the symmetry achieved. Macromastia some aspect of both inframammary folds, com-
and pseudoptosis are not compounding factors promising final cosmesis.
since skin redundancy may be addressed sym- The advantages of bilateral free TRAM
metrically. Bilateral implants and/or permanent reconstruction include limited muscle harvest,
expander implants, postmastectomy, usually limited upper abdominal dissection (which min-
provide exceptional results, in contrast to unilat- imizes discomfort and expedites recovery),
eral procedures, which exaggerate implant char- unparalleled esthetic outcome, and a greater tol-
acteristics. Postoperative adjustability ensures a erance to adjuvant radiation if deemed neces-
symmetric result. This is ideal for the older sary. Lateral extension of these free “hemi-flaps”
Breast Reconstruction 193

may be incorporated to boost tissue volume in


thin patients and is made possible by the excep-
tional blood supply. Assuming the presence of
suitable recipient vessels and sufficient experi-
ence of the reconstructive team, free TRAM
reconstruction for bilateral restoration is usually
preferred for the ultimate benefit of the patient.
The incidence of abdominal wall bulging and
hernia formation is similar for free and conven-
tional bilateral reconstruction and is dependent Figure 11–10. Bilateral free TRAM reconstruction.

upon the security and detail of the abdominal


closure. Some surgeons perform a mild bowel avoidance of drains. The resulting deformity
prep preoperatively to facilitate closure in bilat- after lumpectomy or quadrantectomy depends
eral cases. The potential to exclude medial, on initial breast size, tumor size and location,
diminutive, or outlying perforators in bilateral radiation dose, surgical technique, and adjuvant
free reconstruction facilitates fascial closure chemotherapy. The relative excision, in propor-
without the use of mesh. Although it is reported tion to breast size, is perhaps the most important
that mesh may be avoided in 60 to 80 percent of factor. Patients with large, pendulous breasts
patients having bilateral TRAM reconstruction, may easily accommodate a 4 cm lumpectomy.
the use of a more relaxed closure using mesh The same resection in a smaller-breasted woman
may facilitate postoperative comfort, recovery, may lead to an unacceptable cosmetic result.
and return of bowel motility. Prolene mesh is Radiation therapy exaggerates the tissue deficit
currently the authors’ preferred choice for rein- in the form of ischemic fibrous contracture.
forcement. Closure may be facilitated by a pre- The treated breast is subject to edema, retrac-
operative bowel prep, the appropriate use of tion, fibrosis, calcification, hyperpigmentation,
relaxing agents, avoidance of nitrous oxide depigmentation, telangiectasia formation, and
(which can lead to bowel dilitation) and the use atrophy. It is not until 24 to 36 months postradi-
of lateral external oblique relaxing incisions. The ation that radiation-induced changes stabilize.
incidence of true hernias is rare. Lower abdomi- Initial edema camouflages the initial deficit and
nal attenuation or abdominal wall bulging occurs is replaced with fibrosis and contracture that
in 4.4 to 20 percent of cases. tends to worsen with time. Deficits within the
lower pole tend to retract upward. Deficits along
RECONSTRUCTION OF THE the superomedial aspect of the breast are diffi-
PARTIAL MASTECTOMY DEFECT cult to camouflage due to the paucity of available
adjacent tissue and are, unfortunately, socially
Breast conserving surgery combined with adju- conspicuous. Centrally located lesions are more
vant radiation has been accepted as a regime forgiving unless resection involves some aspect
equivalent to modified radical mastectomy for of the nipple-areolar complex.
early stage (I and II) breast cancer. The tech- An assessment of the patient’s overall onco-
nique is popular due to its ability to eradicate logic risk for recurrence should be considered
breast cancer while preserving a maximal vol- prior to any attempt at partial mastectomy
ume of breast tissue. reconstruction. Breast cancer history, the nature
Skin incisions are designed directly over the of the inciting lesion, and the patient’s family
lesion, and skin and subcutaneous tissues are history should be reviewed prior to an addi-
preserved unless involved in the lesion. Closure tional procedure that may further affect subse-
involves subcuticular closure only and the quent screening examinations. In any event,
194 BREAST CANCER

stabilization of the breast appearance is a pre- the areolar defect, is de-epithelialized. Primary
requisite and occurs 1 to 3 years postradiation. skin closure is facilitated by undermining at the
Investigators have attempted to classify the parenchymal interface. The second technique
spectrum of partial mastectomy deficits and parallels conventional mastopexy and enables
relate them to specific treatment options. Clas- superior advancement of an inferior dermoglan-
sification is based upon the localized deficit of dular pedicle. It is performed through a Wise or
skin and glandular tissue, malposition and/or keyhole pattern incision.106,108–109
distortion of the areola, and the extent of Upper outer quadrant excisions are the most
fibrous contracture of the breast.106 Local flap frequent and, fortunately, the most forgiv-
transposition is recommended for mild defor- ing.106,108–109 The great majority of these exci-
mities, whereas myocutaneous flaps are reserved sions do not require reconstruction. Occasion-
for more extensive defects. ally, delayed augmentation, scar lengthening
Approximately 15 percent of patients treated via Z-plasty, and areolar transposition are indi-
with BCT are not content with the esthetic out- cated. If a discrepancy between the medial and
come.107 These patients often seek consultation lateral breast quadrant is recognized due to a
to improve selfesteem and body image. Careful substantial superolateral resection, immediate
assessment of the actual and apparent tissue centralization of the nipple-areolar complex
deficits are crucial in the selection of the appro- over the point of maximal projection is war-
priate reconstructive strategy. Contour deficits ranted. This involves simple areolar transposi-
signify substantial parenchymal loss, whereas tion after release of the dermal attachments.
radiation contracture represents extensive cuta- Wide excisions may require transfer of regional
neous deficits. Nipple-areolar distortion neces- or distant tissue. The latissimus dorsi myocuta-
sitates a substantial increase in cutaneous neous flap represents the ideal choice for these
replacement, as central areolar support requires defects (Figure 11–11).
dermal rather than subcutaneous support. Partial inferior defects may be corrected on
The majority of patients are poor candidates an immediate or delayed basis. The occasional
for implant reconstruction. Cutaneous fibrosis patient, lacking significant radiation change,
responds poorly to implant displacement, and may benefit from delayed insertion of a small
implant radio-opacity impairs an already com- round or custom (one-third) implant for volume
plex screening examination. Autologous tis- replacement. Most defects, however, benefit
sues, conversely, are reliable, versatile, and pro- from a procedure that parallels a standard supe-
vide all the components necessary for partial rior pedicle reduction mammoplasty.108–109 The
restoration. The inherent vascularity may actu- resection and reconstruction are facilitated
ally improve the quality of the relatively through a standard keyhole pattern. Medial and
ischemic and radiated recipient tissue.
Large central excisions involving the nipple-
areolar complex and primary closure take on a
flat, attenuated appearance, lacking projection.
These defects may be reconstructed in one of
two ways. It may be possible to mobilize a skin
glandular flap based on inferolateral perforators
from the underlying pectoral fascia, which is
then mobilized into the defect. The curvilinear
incision extends from the inferomedial aspect of
the previous areola to the central inframammary Figure 11–11. Reconstruction of the partial mastectomy
defect. Superolateral reconstruction with latissimus dorsi
fold. All but a central skin paddle, rotated into myocutaneous flap.
Breast Reconstruction 195

lateral parenchymal flaps are mobilized from site morbidity and a longer recovery. It would
the pectoralis fascia and inframammary fold appear less economic in restoration of limited
and mobilized into the inferior defect, whether tissue defects. It is indicated for the reconstruc-
it be lateral, central, or medial. tion of large inferior pole deficits in large-
Supra-areolar defects are socially conspicu- breasted women.
ous and necessitate local reconstruction due to Continued surveillance for recurrent cancer
the paucity of available adjacent tissue and the after partial reconstruction should proceed
tendency to develop a visible and depressed scar. unimpeded. Studies comparing pre- and post-
These defects are corrected by superior advance- operative mammograms after partial recon-
ment of the areolar complex, based on an infe- struction have confirmed the radiolucency of
rior pedicle, in a procedure similar to an inferior these flaps. The development of new microcal-
pedicle reduction mammoplasty.106,108–109 cifications, fat necrosis, and new lesions are
The latissimus dorsi myocutaneous flap rep- easily discernible. Some reports, interestingly,
resents the flap of choice for the majority of have noted improved mammographic visualiza-
partial mastectomy defects. Its regional loca- tion and resolution of breast density and fibro-
tion, malleability, ease of dissection, and lack sis as a result of improved local vascularity.
of donor site morbidity are ideally suited for Immediate reconstruction of partial mastec-
this indication. All breast conservation defects tomy defects is gaining popularity. The demand
should be reconstructed by overcorrecting the for these techniques has evolved due to a ten-
skin and soft tissue deficits. In general, twice dency toward more aggressive resection in BCT
the apparent tissue loss should be inset to com- and accumulated experience with unfavorable
pensate for normal wound contracture, contin- tumors. Petit and colleagues reported that imme-
ued retraction of the postradiation fibrosis, and diate reconstruction of the partial mastectomy
anticipated muscle atrophy inherent in raising defect was performed in 25 percent of cases.
muscle flaps. The muscle may be folded and They advocated close preoperative collaboration
contoured to accommodate the most irregular to optimize cosmetic results and enable
defects. Although small skin paddles may be “improved radicality” of the surgical breast con-
harvested to precisely accommodate the appar- servation.110 Thus, the potential for immediate
ent skin deficit, a typical 4 ´ 6 cm skin paddle partial mastectomy reconstruction facilitates a
facilitates flap harvest and replacement of com- more aggressive resection or marginal clearance
promised or contracted radiated skin. in BCT and may lessen the need and/or fre-
Although partial latissimus harvests are quency of re-excision. Also, it may lessen the
possible, the majority of partial mastectomy need for staged reconstruction following radia-
defects warrant total flap elevation. Preserva- tion-induced exaggeration of the defect.
tion of the thoracodorsal nerve will maintain
greater muscle bulk but lead to early postoper- NIPPLE-AREOLAR RECONSTRUCTION
ative contractions. Compulsive fixation at the
recipient site is necessary to avoid disruption. Nipple-areolar reconstruction is a critical stage
Transection or resection of the muscular inser- in breast reconstruction and may add remark-
tion will help avoid the typical bulge within the able realism to the new breast mound (Figure
anterior axilla. Finally, supporting the radiated 11–12). Areolar tattooing facilitates symmetry
native breast skin with a de-epithelialized por- in color, may camouflage minor discrepancies
tion of the transposed skin paddle will improve and scars, and lacks the morbidity associated
ultimate wound contour. with skin grafts. Nipple reconstruction is typi-
The TRAM flap represents a flap of sub- cally performed at a second stage, at the time of
stantial bulk, typically incurring greater donor port removal, or breast mound revision.
196 BREAST CANCER

has proven to be a reliable workhorse, with the


potential for a long projectile nipple if
needed.109 The donor site does require a skin
graft, most commonly harvested from the
groin, inner thigh, or axilla. Precise demarca-
tion of the central nipple complex is critical and
serves as a basis for dermal flap elevation. The
lateral dermal wings are elevated, preserving
the central nipple core and an inferior extension
of fat. These components are elevated, preserv-
ing the subcutaneous perforators, and then sur-
faced by the lateral wings. The circular de-
Figure 11–12. Completed nipple-areolar reconstruction. epithelialized harvest site is then covered with
a full thickness skin graft.
The Star flap,111 C-V flap,112 fishtail flap
Although single-stage reconstruction may be (McCraw), and double opposing tab flap
performed, attaining symmetry of nipple-areo- (Kroll) are additional flap options, most of
lar position is crucial to esthetic outcome and is which are modifications of the skate flap.
most accurately attained at a second stage, Although they provide less nipple projection
when dermal edema and skin elasticity have than does the skate flap, they avoid the need for
normalized. a skin graft. These are excellent alternatives for
The insensate, adynamic nipple remains sta- the majority of patients with small to moderate
tic in size, contour, and projection and will sized contralateral nipples.
likely be visualized through undergarments, Intradermal areolar tattoo has greatly simpli-
swimsuits, and clothing. The patient’s final fied the final phase of restoration and adds
assessment and perception may closely parallel abrupt and striking realism to the physical breast
the quality and symmetry of the newly con- form. It remains an artistic challenge among sur-
structed nipple. Consideration of a symmetry geons to simulate contralateral areolar pigment.
procedure should, therefore, be entertained This final phase enables the surgeon one ad-
prior to final nipple reconstruction and should ditional opportunity to optimize symmetry.
encompass whether the patient prefers support Nipple-areolar reconstruction may enhance the
(bra) and to what extent. Simulation in a bra or focus of the reconstructed breast and improve
sheer blouse preoperatively may help the overall patient incorporation of the reconstructed
patient’s understanding of these issues.
breast, both physically and psychologically.
Various techniques of nipple reconstruction
are available and provide a range of caliber
potential projection. Modification, reduction, REFERENCES
or composite grafting of the contralateral nip- 1. Ernster VL, Barclay J, Kerlikowske K, et al. Inci-
ple may be considered as an option in the dence of and treatment for ductal carcinoma in
patient with redundant nipples. Although this situ of the breast. JAMA 1996;275:913–8.
represents the most realistic reconstruction, it 2. Grisotti A. Conservative treatment of breast can-
necessitates a procedure on the remaining cer. In: Spear S, editor. The breast: principles
intact nipple and is sensitive perioperatively. and art. Philadelphia: Lippincott-Raven;
1998. p. 137.
Local flaps are the technique of choice for 3. Goldwyn RM. Vincenz Czerny and the begin-
nipple reconstruction, most of which are vari- nings of breast reconstruction. Plast Reconstr
ants of the original skate flap. The skate flap Surg 1978;61:673–81.
Breast Reconstruction 197

4. Georgiade G, Georgiade N, McKarty KS Jr, et al. consecutive cases. Plast Reconstr Surg 1993;
Rationale for immediate reconstruction of the 92:217–27.
breast following modified radical mastectomy. 17. Schuster RH, Kuske RB, Young VL, Fineberg B.
Ann Plast Surg 1982;8:20–28. Breast reconstruction in women treated with
5. Rosenqvist S, Sandelin K, Wickman M. Patients’ radiation therapy for breast cancer: cosmesis,
psychological and cosmetic experience after complications, and tumor control. Plast Recon-
immediate breast reconstruction. Eur J Surg str Surg 1992;90:445–52.
Oncol 1996;22:262–6. 18. Evans GR, Schusterman MA, Kroll SS, et al.
6. Wellisch DK, Schain WS, Noone RB, et al. Psy- Reconstruction and the radiated breast: is there
chosocial correlates of immediate versus a role for implants? Plast Reconstr Surg 1995;
delayed reconstruction of the breast. Plast 96:1111–5.
Reconstr Surg 1985;76:713–8. 19. Spear S, Majidian A. Immediate breast recon-
7. Shover LR, Yetman RJ, Tuason LJ, et al. Partial struction in 2 stages using textured integrated-
mastectomy and breast reconstruction. A com- valve tissue expanders and breast implants: a
parison of their effects on psychosocial adjust- retrospective review of 171 consecutive breast
ment, body image, and sexuality. Cancer reconstructions from 1989–1996. Plast Recon-
1995;75:54–64. str Surg 1998;101:53–63.
8. Noguchi M, Kitagawa H, Kinoshita K, et al. Psy- 20. Jackson WB, Goldson AL, Staud C. Post-opera-
chologic and self-assessments of breast con- tive irradiation following immediate breast
serving therapy compared with mastectomy reconstruction using a temporary tissue
and immediate breast reconstruction. J Surg expander. J Natl Med Assoc 1994;86:538–42.
Oncol 1993;54:260–6. 21. Dickson MG, Sharpe BT. The complications of tis-
9. Toth BA, Lappert P. Modified skin incisions for sue expansion in breast reconstruction: a review
mastectomy: the need for plastic surgery input of 75 cases. Br J Plast Surg 1987;40:629–35.
in pre-operative planning. Plast Reconstr Surg 22. Jacobson GM, Sause WT, Thompson JW, Plenk
1991;87:1048–53. HP. Breast irradiation following silicone gel
10. Kroll SS, Ames F, Singletary SE, et al. The onco- implants. Int J Radiat Oncol Biol Phys 1986;
logic risks of skin preservation at mastectomy 12:835–8.
when combined with immediate reconstruction 23. Chu FC, Kaufman TP, Dawson GA, et al. Radia-
of the breast. Surg Gynecol Obstet 1991; tion therapy of cancer in prosthetically aug-
172:17–20. mented or reconstructed breasts. Radiology
11. Newman LA, Keurer HM, Hunt KK, et al. Pre- 1992;185:429–33.
sentation, treatment, and outcome of local 24. Spear S. Prosthetic reconstruction in the radiated
recurrence after skin sparing mastectomy and breast. In: Spear S, editor. The breast: princi-
immediate breast reconstruction. Ann Surg ples and art. Philadelphia: Lippincott-Raven;
Oncol 1998;5:620–6. 1998. p. 399–406.
12. Hidalgo DA. Aesthetic refinement in breast 25. Zimmerman RP, Mark RJ, Kim AL, et al. Radiation
reconstruction: complete skin sparing mastec- tolerance of transverse rectus myocutaneous
tomy with autogenous tissue transfer. Plast free flaps used in immediate breast reconstruc-
Reconstr Surg 1998;102:63–70. tion. Am J Clin Oncol 1998;21:381–5.
13. Carlson GW, Bostwick J, Styblo TM, et al. Skin 26. Hunt KK, Baldwin BJ, Strom EA, et al. Feasibil-
sparing mastectomy: oncologic and reconstruc- ity of post-mastectomy radiation therapy after
tive considerations. Ann Surg 1997;225:570–75. TRAM flap breast reconstruction. Ann Surg
14. Yule GJ, Concannon MJ, Croll G, et al. Is there Oncol 1997;4:377–84.
liability with chemotherapy following immedi- 27. Sultan MR, Smith ML, Estabrook A, et al. Imme-
ate breast reconstruction? Plast Reconstr Surg diate breast reconstruction in patients with
1996;97:969–73. locally advanced disease. Ann Plast Surg
15. Grotting JC, Urist MM, Maddox WA, Vasconez LO. 1997;38:345–9.
Conventional TRAM flap versus free microsur- 28. Styblo TM, Lewis MM, Carlson GW, et al. Imme-
gical TRAM flap for immediate reconstruction. diate breast reconstruction for stage III breast
Plast Reconstr Surg 1989;84(6):1005–6. cancer using transverse rectus myocutaneous
16. Elliott LF, Eskenazi L, Beegle PH Jr, et al. Imme- flaps. Ann Surg Oncol 1998;3:375–80.
diate TRAM flap breast reconstruction: 128 29. Brody GS. Safety and effectiveness of breast
198 BREAST CANCER

implants. In: Spear S, editor. The breast: prin- up of the Los Angeles study. Plast Reconstr
ciples and art. Philadelphia: Lippincott-Raven; Surg 1997;99:1346–53.
1998. p. 336–46. 45. Deapon DM, Pike MC, Casagrande JT, Brody GS.
30. Peters W, Keystone E, Snow K, et al. Is there a rela- The relationship between breast cancer and
tionship between autoantibodies and silicone gel augmentation mammoplasty: an epidemiologic
implants? Ann Plast Surg 1994;32:1–5. study. Plast Reconstr Surg 1986;77:361–7.
31. Kossovsky N, Heggers JP, Robson MC. Experi- 46. Engel A, Lamm SH. Risk of sarcomas of the
mental demonstration of the immunogenicity breast among women with breast augmenta-
of silicone protein complexes. J Biomed Mater tion. Plast Reconstr Surg 1992;89:571–2.
Res 1987;21:1125–33. 47. Su CW, Dreyfuss DA, Krizek TJ, et al. Silicone
32. Heggers JP, Kossovsky N, Parsons RW, et al. Bio- implants and the inhibition of cancer. Plast
compatibility of silicone implants. Ann Plast Reconstr Surg 1995;96:513–8.
Surg 1983;11:38–45. 48. Brody GS, Conway DP, Deapon DM, et al. Con-
33. DeCamara DI, Sheridam SM, Kammer BA. Rup- sensus statement on the relationship of breast
ture and aging of silicone breast implants. Plast implants to connective tissue disorders. Plast
Reconstr Surg 1993;91:828–34. Reconstr Surg 1992;90:1102–5.
34. Greenwald WB, Randolph M, May JW. Mechani- 49. Gabriel SE, O’Faflon WM, Kurland LT, et al. Risk
cal analysis of explanted silicone breast of connective tissue diseases and other disor-
implants. Plast Reconstr Surg 1996;98:269–72. ders after breast implantation. N Engl J Med
35. Phillips JW, Decamara DL, Lockwood MD, et al. 1994;330:1697–702.
Strength of silicone breast implants. Plast 50. Giltay EJ, Moens HJB, Riley AH, et al. Silicone
Reconstr Surg 1996;97:1215–25. breast prostheses and rheumatic symptoms: a
36. Robinson OG, Bradley EL, Wilson DS. Analysis retrospective follow-up study. Ann Rheum Dis
of explanted silicone implants: a report of 300 1994;53:194–6.
patients. Ann Plast Surg 1995;34:1–6. 51. Radovan C. Breast reconstruction after mastec-
37. Ahn CY, Shaw WW. Regional silicone gel migra- tomy using the temporary expander. Plast
tion in patients with ruptured implants. Ann Reconstr Surg 1982;69:195–208.
Plast Surg 1994;33:201–8. 52. Colen SR. Immediate two stage breast reconstruc-
38. Ahn CY, DeBruhl ND, Gorczyca DP, et al. Com- tion utilizing a tissue expander and implant. In:
parative silicone breast implant evaluation Spear S, editor. The breast: principles and art.
using mammography, sonography, and mag- Philadelphia: Lippincott-Raven; 1998. p. 375–86.
netic resonance imaging: experience with 59 53. Forman DL, Chiu J, Restifo RJ, et al. Breast
implants. Plast Reconstr Surg 1994;94:620–7. reconstruction in previously irradiated patients
39. Leibman AL, Kruse BD. Imaging of breast cancer using tissue expanders and implants: a poten-
after augmentation mammoplasty. Ann Plast tially unfavorable result. Ann Plast Surg 1998;
Surg 1993;30:111–5. 40:360–3.
40. Silverstein MJ, Gamagami P, Handel N. Missed 54. Mandrekas AD, Zambacos GJ, Katsantoni PN.
breast cancer in an augmented woman using Immediate and delayed breast reconstruction
implant displacement mammography. Ann with permanent tissue expanders. Br J Plast
Plast Surg 1990;25:210–3. Surg 1995;48:572–8.
41. Carlson GW, Curley SA, Martin FE, et al. The 55. Francel TJ, Ryan JJ, Manson PM. Breast recon-
detection of breast cancer after augmentation struction utilizing implants: a local experience
mammoplasty. Plast Reconstr Surg 1993;91: and comparison of three techniques. Plast
837–40. Reconstr Surg 1993;92:786–94.
42. Gumico CA, Pin P, Young VL, et al. The effect of 56. Yeh KA, Lyle G, Wei JP, et al. Immediate breast
breast implants on the radiographic detection reconstruction in breast cancer: morbidity and
of microcalcifications and soft tissue masses. outcome. Am J Surg 64:1195–9.
Plast Reconstr Surg 1989;84:772–8. 57. Wickman M, Jurell G, Sandelin K. Technical
43. Birdsell DC, Jenkins H, Berkel H. Breast cancer aspects of immediate breast reconstruction: 2
diagnosis and survival in women with and year follow-up of 100 patients treated conserv-
without breast implants. Plast Reconstr Surg atively. Scand J Plast Reconstr Surg 1998;32:
1993;92:795–800. 265–73.
44. Deapon DM, Bernstein L, Brody GS. Are breast 58. Caffee HH. Textured silicone and capsule contrac-
implants anti-carcinogenic? A 14 years follow- ture. Ann Plast Surg 1990;24:197–9.
Breast Reconstruction 199

59. Pakium AI, Young CS. Submuscular breast recon- experience with 147 cases. Mayo Clin Proc
struction: a one stage method of tissue expan- 1994;69:635–40.
sion. Ann Plast Surg 1987;19:312–7. 74. Berrino P, Campora E, Leone S, et al. The trans-
60. Kroll SS, Baldwin B. A comparison of outcome verse rectus myocutaneous flap for breast
using three different methods of breast recon- reconstruction in obese patients. Ann Plast
struction. Plast Reconstr Surg 1992;90:455–62. Surg 1991;27:221–31.
61. Hartrampf CR Jr, Scheflan M, Black PW. Breast 75. Takeishi M, Shaw WW, Ahn CY, et al. TRAM
reconstruction with a transverse abdominal flaps in patients with abdominal scars. Plast
island flap. Plast Reconstr Surg 1982;69:216–9. Reconstr Surg 1997;99:713–22.
62. Boyd JB, Taylor GI, Corlett R. The vascular territo- 76. Kroll SS, Gheradini G, Martin JE, et al. Fat necro-
ries of the superior and deep inferior epigastric sis in free and pedicled TRAM flaps. Plast
systems. Plast Reconstr Surg 1984;73:1–16. Reconstr Surg 1998;102:1502–7.
63. Moon HK, Taylor GI. The vascular anatomy of the 77. Slavin SA, Goldwyn RM. The midabdominal rec-
transverse rectus myocutaneous flaps based on tus abdominus myocutaneous flap: review of
the deep superior epigastric system. Plast 236 flaps. Plast Reconstr Surg 1988;81:189–97.
Reconstr Surg 1988;82:815–32. 78. Slavin SA, Hein KD. The mid-abdominal transverse
64. Bostwick J. Plastic and reconstructive breast rectus abdominus myocutaneous flap. In: Spear
surgery. St Louis: Quality Medical Publishing; S, editor. The breast: principles and art. Philadel-
1990. phia: Lippincott-Raven; 1998. p. 565–76.
65. Watterson PA, Bostwick J, Hester TR, et al. 79. Callegari PR, Taylor GI, Caddy CM, et al. An
TRAM flap anatomy correlated with a ten year anatomic review of the delay phenomenon. I.
clinical experience with 556 patients. Plast Experimental studies. Plast Reconstr Surg
Reconstr Surg 1995;95:1185–94. 1992;89:397–407.
66. Hartrampf CR, Bennett GK. Autogenous tissue 80. Morris SF, Taylor GI. The time sequence of the
delay phenomenon: when is a surgical delay
reconstruction in the mastectomy patient: a
effective? An experimental study. Plast Reconstr
critical review of 300 patients. Ann Surg
Surg 1995;95:526–33.
1987;205:508–19.
81. Taylor GI. The surgically delayed unipedicled
67. Hartrampf CR, Michelow BJ. Breast reconstruc-
TRAM flap for breast reconstruction. Ann
tion with living tissue. Norfolk (VA): Hampton
Plast Surg 1996;36:242–5.
Press; 1991.
82. Codner MA, Bostwick J, Nahai F, et al. TRAM flap
68. Little JW. Breast reconstruction by the unipedicle
vascular delay for high risk breast reconstruc-
TRAM operation: muscle splitting technique.
tion. Plast Reconstr Surg 1995;96:1615–22.
In: Spear S, editor. The breast: principles and
83. Restifo RJ, Ahmed SS, Isenburg JS, et al. Timing,
art. Philadelphia: Lippincott-Raven; 1998.
magnitude and utility of surgical delay in the
p. 521–34. TRAM flap. I. Animal studies. Plast Reconstr
69. Takayanagi S. Extended transverse rectus abdomi- Surg 1997;99:12–16.
nus myocutaneous flap. Plast Reconstr Surg 84. Serletti JM, Moran SL. Free versus the pedicled
1993;92:757–8. TRAM flap: a cost comparison and outcome
70. Yamamota Y, Nohira K, Sugihara T, et al. Superi- analysis. Plast Reconstr Surg 1997;100:1418–
ority of the microvascularly augmented flap: 24.
analysis of 50 transverse rectus abdominus 85. Schusterman MA, Kroll SS, Weldon ME. Imme-
myocutaneous flaps for breast reconstruction. diate breast reconstruction: why the free
Plast Reconstr Surg 1996;97:79–83. TRAM over the conventional TRAM flap.
71. Paige KT, Bostwick J, Bried JT, Jones G. A com- Plast Reconstr Surg 1992;90:255–61.
parison of morbidity from bilateral, unipedi- 86. Kroll SS, Netscher DT. Complications of the
cled and unilateral, unipedicled TRAM flap TRAM flap breast reconstruction in obese
breast reconstructions. Plast Reconstr Surg patients. Plast Reconstr Surg 1989;84:866.
1998;101:1819–27. 87. Schusterman MA, Kroll SS, Miller MJ, et al. The
72. Williams JK, Bostwick J III, Bried JY, et al. free transverse rectus myocutaneous flap for
TRAM flap breast reconstruction after radia- breast reconstruction: one center’s experience
tion treatment. Ann Surg 1995;221:756–64. with 211 consecutive cases. Ann Plast Surg
73. Jacobson WM, Meland NB, Woods JE. Autolo- 1994;32:234–41.
gous breast reconstruction with use of trans- 88. Suominen S, Asko-Seljavaara S, Kinnunen J, et al.
verse rectus myocutaneous flap: Mayo clinic Abdominal wall competence after free trans-
200 BREAST CANCER

verse rectus abdominus myocutaneous flap cutaneous flap breast reconstruction. In: Spear S,
harvest: a prospective study. Ann Plast Surg editor. The breast: principles and art. Philadel-
39:299–34. phia: Lippincott-Raven; 1998. p. 477–90.
89. Kind GM, Rademaker AW, Mustoe TA. Abdomi- 101. Taylor GI, Townsend P, Corlett R. Superiority of
nal wall recovery following TRAM flap: a the deep circumflex iliac vessels as the supply
functional outcome study. Plast Reconstr Surg for free groin flaps. Clinical work. Plast
1997;99:417–28. Reconstr Surg 1979;64:745–59.
90. Kroll SS, Schusterman MA, Reece GP, et al. 102. Elliott LF, Hartrampf CR Jr. The Rubens flap.
Abdominal wall strength, bulging, and hernia The deep circumflex iliac artery flap. Clin
after TRAM flap breast reconstruction. Plast Plast Surg 1998;25:283–91.
Reconstr Surg 1995;96:616–9. 103. Fugino T, Harashina T, Endomoto K. Primary
91. Fitoussi A, Le Taillandier M, Biffaud JC, et al. breast reconstruction after a standard radical
Functional evaluation of the abdominal wall mastectomy by a free flap transfer. Plast
after raising a rectus abdominus myocutaneous Reconstr Surg 1976;58:372–4.
flap. Ann Chir Plast Esthet 1997;42:138–46. 104. Shaw WW. Superior gluteal free flap breast recon-
92. Hartrampf CR Jr, Bried JT. General considera- struction. Clin Plast Surg 1998;25:267–74.
tions in TRAM flap surgery. In: Hartrampf CR, 105. Elliott LF, Beegle PH, Hartrampf CR Jr. The lat-
editor. Breast reconstruction with living tissue. eral transverse thigh free flap: an alternative
New York: Raven Press; 1991. p. 33–70. for autogenous-tissue breast reconstruction.
93. Kroll SS, Marchi M. Comparison of strategies for Plast Reconstr Surg 1990;85:169–78.
preventing abdominal wall weakness after 106. Slavin SA. Reconstruction of the breast conser-
TRAM flap breast reconstruction. Plast Reconstr vation patient. In: Spear S, editor. The breast:
Surg 1992;89:1045–51. principles and art. Philadelphia: Lippincott-
94. Kroll SS, Gherardini G, Martin JE, et al. Fat Raven; 1998 p. 221–38.
necrosis in free and pedicled TRAM flaps. 107. Beadle F, Silver B, Botnick L, et al. Cosmetic
Plast Reconstr Surg 1998;102:1502–7. results following primary radiation therapy for
95. Shestak KC. Bipedicle TRAM flap reconstruction. early breast cancer. Cancer 1984;54:2911–8.
In: Spear S, editor. The breast: principles and art. 108. Kroll SS, Singletary SE. Repair of partial mastec-
Philadelphia: Lippincott-Raven; 1998. p. 535–46. tomy defects. Clin Plast Surg 1998;25:303–10.
96. Petit JY, Rietjens M, Ferreira MA, et al. Abdom- 109. Grisotti A. Immediate reconstruction after partial
inal sequellae after pedicled TRAM flap breast mastectomy. Oper Tech Plast Reconstr Surg
reconstruction. Plast Reconstr Surg 1997;99: 1994;1:1–12.
723–9. 110. Petit JY, Rietjens M, Garusi C, et al. Integration
97. Jensen JA. Is double pedicle TRAM flap recon- of plastic surgery in the course of breast con-
struction of a single breast within the standard serving surgery for cancer to improve cosmetic
of care? Plast Reconstr Surg 1989;102:586–7. results and radicality of tumor excision. Recent
98. Spear SL, Hartrampf CR Jr. The double pedicle Results Cancer Res 1998;152:202–11.
TRAM flap and the standard of care. Plast 111. Little JW, Spear SL. The finishing touches in nip-
Reconstr Surg 1998;100:1592–3. ple–areola reconstruction. Perspect Plast Surg
99. Maxwell GP. Iginio Tansini and the origin of the 1988;2:1–17.
latissimus dorsi musculocutaneous flap. Plast 112. Bostwick J III. Creating a nipple. In: Berger K,
Reconstr Surg 1980;65:686–92. Bostwick J III, editors. A women’s decision. St.
100. Hammond DC, Fisher J. Latissimus dorsi musculo- Louis: Quality Medical Publishing; 1994.
12
Adjuvant Systemic Therapy
of Early Breast Cancer
GERSHON Y. LOCKER, MD

The modern era of breast cancer treatment cer were cured by surgery. Regrettably this fact
began over 100 years ago with the development remains so even today. It is only in the last 30
of surgical techniques that emphasized the need years, however, that the other principles of
for total resection of tumor. Nevertheless, adjuvant therapy were applied to the treatment
despite gross total excision, many patients with of early breast cancer leading to the improve-
seemingly localized disease suffered relapse or ment of outcome.
distant recurrence and died of their cancer. This
was presumably due to the growth of micro- IDENTIFICATION OF POPULATIONS
scopic tumor unappreciated at the time of ini- AT HIGH RISK OF RELAPSE
tial therapy. The need for additional, adjuvant
therapy after surgery led to numerous random- Although not every women with early stage,
ized controlled trials (RCTs) addressing the “localized” breast cancer is rendered cancer
problem. The role of adjuvant systemic therapy free by local treatment, many women are. It is
in increasing survival and decreasing mortality critical, if additional treatments are to be used,
has been established by these studies and con- that they be directed at women at highest risk of
firmed by overview meta-analyses. The basis recurrence. Indiscriminate administration of
for this success is the recognition of and adher- adjunctive therapies runs the risk of unnecessar-
ence to the principles of adjuvant therapy, ily exposing those already cured to toxicity, mor-
which are that (1) local treatments do not cure bidity and, with some aggressive approaches,
all patients with seemingly localized cancer; (2) even mortality from treatment. Choice of
populations at high risk of relapse can be iden- patients to be treated is critical in the develop-
tified; (3) patterns of relapse and failure are ment of successful adjuvant therapy. For
understood; (4) palliative therapy against overt women with early stage, operable breast cancer,
macroscopic tumor can potentially eradicate the single most important prognostic feature for
occult microscopic disease; (5) the value of recurrence and death is the presence or absence
adjuvant therapy has been validated in RCTs; of tumor metastases in the axillary lymph
(6) the choice of adjuvant therapy considers the nodes.1,2 Patients with no axillary nodal metas-
biology of the tumor and of the patient and; (7) tases have a 70 to 75 percent chance of long-
the benefits of treatment outweigh the toxicity term disease-free survival (DFS) when treated
and risks of therapy. with surgery alone. Patients with any number
Halstead in the late 19th century appreci- of nodal metastases have a 25 to 30 percent
ated that not all patients with early breast can- chance of long-term survival.3 The risk of

201
202 BREAST CANCER

recurrence and death increases with the number ers, however, may be of value in determining
of involved nodes. Women with more than 10 the type of adjuvant therapy to be used (hor-
nodes involved have an extremely poor progno- mone receptors, (c-erbB-2). Studies in the past
sis, less than a 20 percent cure rate as shown in and current practice use nodal status and tumor
most studies. From the beginning of the era of size as primary determinants of need for treat-
adjuvant therapy trials, patients with involved ment. The other prognostic factors are used
axillary nodes were identified as the highest- (often together) only when the need for, or the
risk group and were the subjects of adjuvant potential benefits of, systemic therapy after
approaches. As systemic adjuvant therapies for local treatment is unclear.
node-positive disease became accepted, atten-
tion turned to improving the prognosis of the 30 UNDERSTANDING PATTERNS OF
percent of node-negative cases destined to have RELAPSE AND FAILURE
recurrences and die. Multiple studies have
shown that in these women, the size of the pri- There are multiple effective treatments of breast
mary tumor is the most important predictor of cancer. Which one is most appropriate to
surgical outcome.3 Women with tumors smaller increase survival and cure rate after primary
than 1 cm in diameter and no involved nodes therapy of early disease is dependent on the
have better than a 90 percent chance of long- nature of the failure. Is it due to inadequate con-
term DFS. Adjuvant therapy is not a major pri- trol of the primary tumor, regional spread, or
ority in this group and is not routinely given. distant metastases? The Halstedian view of
On the other hand, women with tumors > 5 cm, breast cancer spread was that of a prolonged
despite having no nodal metastases, have a period of local/regional disease before systemic
prognosis comparable to patients with axillary dissemination. By implication, failure to cure
nodal spread. Clearly, women with large might be due to incomplete surgery. As dis-
tumors, whatever the axillary status, are candi- cussed in Chapter 11, multiple studies have
dates for additional therapy beyond surgery. failed to confirm the survival benefit of more
Other than nodal status and tumor size, the use aggressive versus less aggressive surgery. A
of prognostic features to determine who should recent meta-analysis of 3,400 women in ran-
or should not be considered for adjuvant ther- domized trials of more versus less extensive
apy is very controversial. While tumor grade surgery found no difference in 10-year survival
may be helpful,1 the variability of grading between the two approaches.5 This was true
between pathologists makes it problematic. whether or not the patients had axillary nodal
Hormone receptor status, proliferation markers involvement. Radiation therapy is another
(%S phase, thymidine labeling, Ki67), onco- approach to local and regional control. If the
gene expression/mutation (c-erbB-2, p53) may Halstedian paradigm were true, local regional
be more reproducible and have also been corre- irradiation by killing residual tumor should
lated with outcome of local therapy.4 Neverthe- increase the overall cure rate and survival.
less, to date, none has been universally Again, most studies have failed to confirm sur-
accepted as an independent predictor for the vival benefit with irradiation of the chest
need for adjuvant treatment. The American wall/draining nodes, despite significantly
Society of Clinical Oncology (ASCO) Tumor decreasing local recurrence5 (see also Chapter
Marker Expert Panel, using criteria of evi- 16). In studies of local modalities, the decrease
dence-based medicine, could not endorse any in local recurrence did not improve survival
of these tumor markers by themselves as ade- benefit, in part because of the development of
quate to determine the need for additional ther- distant recurrences. This refutation of the Hal-
apy beyond breast surgery.4 Some of the mark- stedian theory strongly argues that failure to
Adjuvant Systemic Therapy of Early Breast Cancer 203

cure localized breast cancer with local therapies approach.8 Oophorectomy in premenopausal
is due to the presence of unappreciated distant women and tamoxifen in women of all ages
micrometastases, that are destined to grow and cause significant regressions of clinically
kill the patient. To improve surgical results, addi- advanced estrogen and or progesterone recep-
tional therapy must be directed to the systemic tor–containing breast cancers with generally
nature of breast cancer in high-risk women. acceptable toxicity. They were obvious candi-
dates to be tried in the adjuvant setting after
PALLIATIVE THERAPY AGAINST local therapy of early disease to treat occult
OVERT MACROSCOPIC TUMOR AND micrometastases.
POTENTIAL ERADICATION OF OCCULT Unfortunately, not all breast cancers are
MICROSCOPIC DISEASE estrogen dependent and responsive to hormonal
manipulation. After the introduction of chemo-
A basic assumption underlying adjuvant sys- therapeutic drugs in the 1940s and 1950s, mul-
temic therapy of early breast cancer is that tiple agents were identified that were able to
those therapies that may be only palliative cause temporary shrinkage of tumor in women
against bulky macroscopic metastases might be with disseminated breast cancer. Melphelan
curative against microscopic disseminated (PAM), thiotepa (T), cyclophosphamide (C),
tumor that is assumed to be present in high-risk methotrexate (M), fluorouracil (F), or vinblas-
patients. To develop effective adjuvant systemic tine (V) have a 20 to 30 percent chance of caus-
therapy of early disease, one must first identify ing transient regression of metastatic breast can-
effective and safe therapies for advanced-stage cer when used alone.9 In the late 1960s Cooper
breast cancer. combined five drugs and reported higher
Systemic therapy for overt advanced breast regression rates.10 This regimen was the fore-
cancer began 100 years ago when Beatson runner of the cyclophosphamide methotrexate
observed shrinkage of locally extensive breast fluorouracil (CMF) combination regimen,
cancers after oophorectomy in premenopausal which was in the 1970s the standard chemo-
women.6 This phenomenon is based on the therapeutic treatment of advanced disease
trophic effect of estrogen on approximately half (Table 12–1).11 In the late 1970s, doxorubicin
of all breast cancers studied. Removal of the (Adriamycin[A]) was introduced and found to
ovaries leading to a drop in endogenous estro- be the most active single agent against advanced
gen levels in younger women can arrest cancer breast cancer. It was soon combined with other
growth and result in regression. Another drugs11 (ex-cyclophosphamide adriamycin fluor-
approach to depriving breast cancers of estro- ouracil [ex-CAF]) and, in the 1980s, became the
gen effects is to block estrogen binding to the new standard treatment of overt metastatic dis-
protein, estrogen receptor (ER), in the breast ease (see Table 12–1). Finally, another class of
cancer cell cytoplasm. The receptor-estrogen drugs, the taxanes (paclitaxel–Taxol and doc-
complex mediates much of the effect of the hor- etaxel–Taxotere) were introduced in the last
mone on the cell, and blocking the interaction, decade with activity comparable with that of
such as by removing estrogen, leads to regres- doxorubicin in the palliation of advanced mam-
sion of the hormone-dependent cancer.7 Tamox- mary cancer. All these drugs can be given safely,
ifen (Nolvadex) is the prototype competitive though they have significant toxicity. All were
inhibitor of estrogen binding at its receptor. The candidates for use in the adjuvant setting.
ability to measure estrogen and progesterone A recent trend in the treatment of metastatic
receptors and thus predict responsiveness to breast cancer is the use of high-dose chemother-
endocrine therapy made hormonal treatment of apy, either with cytokine support of the bone
overt, metastatic breast cancer a standard marrow or with stem cell or bone marrow stor-
204 BREAST CANCER

age and reinfusion (“bone marrow transplanta- because once a patient relapses, there are effec-
tion”). These therapies are based on the assump- tive palliative treatments to prolong life in
tion of a dose-response curve for drug-induced advanced disease. Perhaps the most convincing
cancer cell death. While still controversial in effect of adjuvant treatment, however, is on the
advanced breast cancer (and very toxic), such percentage of patients alive long after treatment.
approaches are also candidates for evaluation Multiple randomized trials have shown that sys-
against the poorest-risk, early-stage disease. temic treatment of early-stage breast cancer sig-
nificantly and reproducibly decreases the risk of
ADJUVANT THERAPY VALIDATED IN death years after local therapy. This benefit per-
RANDOMIZED CONTROLLED TRIALS sists with time, suggesting the likelihood of cure.

Effective therapies of advanced breast cancer CHEMOTHERAPY


were long known and anecdotally used after
surgery for early breast cancer, but their value as Single Agents
adjuncts to primary therapy was difficult to
assess. Unlike in advanced disease, where tumor As in the case of advanced disease, the earliest
shrinkage after systemic treatment can be deter- trials of adjuvant chemotherapy were of single
mined directly by observation or radiographi- agents. The National Surgical Adjuvant Breast
cally, when used to increase survival after treat- and Bowel Project (NSABP) conducted ran-
ment of early stage disease, there are no direct domized trials in the 1950s and 1960s looking at
determinants of effectiveness. Cure in an indi- a short course of thiotepa or fluorouracil after
vidual patient with early-stage disease may have surgery versus surgery alone. Overall, there was
been achieved as a result of the systemic therapy no survival benefit to the chemotherapy, but
or might have occurred even if it had not been there was a suggestion that thiotepa might have
given as a result of the local treatment. Recur- some benefit in premenopausal women.12 The
rence may be a sign of failure of adjunctive ther- rationale for these studies was a belief that
apy but might have been delayed because of it. manipulation of the tumor during surgery might
The only way to truly evaluate the usefulness of promote detachment and spread of its cells and
additional treatments after primary therapy of that chemotherapy might kill the scattered cells.
breast cancer is by large RCTs. Initially, those It was only with the realization that failure may
trials randomized women with high-risk, early- be due to distant metastases already present at
stage disease to surgery alone or to surgery plus the time of surgery that trials of protracted
an experimental adjuvant therapy. As adjuvant chemotherapy were undertaken. The risk of tox-
treatments were proved effective, the next gener- icity of these more prolonged approaches led to
ation of trials randomized women to primary restriction of the trials to women with node-pos-
therapy plus “standard” adjuvant systemic treat- itive disease. The critical single-agent study was
ment versus primary therapy plus “experimen- done by the NSABP in the early 1970s. It ran-
tal” adjuvant treatment. The end point of such domized 349 women with node-positive disease
trials were disease-free interval (DFI) and DFS to surgery alone or surgery plus 2 years of inter-
(the time to recurrence and percentage of mittent oral melphalan.13 At 10 years, there was
patients alive without recurrence of cancer at any significant improvement in DFS and a trend
time point) and overall survival (time to death toward improved overall survival in the
and percentage of patients alive at any time chemotherapy group. The benefits, however,
point). In general, the benefits of adjuvant sys- were confined to women under 50 years. At
temic chemotherapy or hormonal therapies are about the same time, randomized studies were
more pronounced on DFS than overall survival showing the superiority of combination
Adjuvant Systemic Therapy of Early Breast Cancer 205

Table 12–1. ADJUVANT SYSTEMIC THERAPY REGIMENS USED IN THE TREATMENT OF EARLY BREAST CANCER

Regimen Indication Frequency Drugs Dose

CMF Poor prognosis node – Every 28 days Cyclophosphamide 100 mg/M2 po qd ´ 14 d


or 1 to 3 + nodes Methotrexate 40 mg/M2 IV dl and d8
(erbB-2 –) ´ 6 months Fluorouracil 600 mg/M2 IV dl and d8
AC Poor prognosis node – Every 21 days Cyclophosphamide 600 mg/M2 IV dl
or 1 to 3 + nodes Doxorubicin 60 mg/M2 IV dl
(erbB-2 + or –) ´ 4 treatments
CAF 4 or more + nodes Every 28 days Cyclophosphamide 100 mg/M2 po qd ´ 14d
Doxorubicin 30 mg/M2 IV dl and d8
´ 6 months Fluorouracil 600 mg/M2 IV dl and d8
AC® Paclitaxel 4 or more + nodes AC given ´ 4 as above
Every 21 days Followed by
´ 4 treatments Paclitaxel 175 mg/M2 3 h IV infusion
Tamoxifen Receptor Daily for 5 years 20 mg po daily
(ER or PR)-containing tumors
• Postmenopausal node ±
• Premenopausal node –

chemotherapy over single-agent therapy in the CMF remains a standard adjuvant systemic ther-
treatment of metastatic breast cancer. Would the apy for early-stage breast cancer today.
same be true for adjuvant therapy? Almost immediately after the adoption of
adjuvant chemotherapy for node-positive pre-
Combination Cyclophosphamide menopausal women, several questions arose:
Methotrexate Fluorouracil Therapy was combination chemotherapy truly better
and Meta-analyses than single-agent therapy? Is there really only a
disease-free but not overall survival benefit to
In the early 1970s Bonadonna at the National adjuvant chemotherapy? What is the optimal
Cancer Institute in Milan evaluated CMF given 2 duration of therapy? Is it of any benefit in poor-
weeks on and 2 weeks off for 12 months (“clas- risk, node-negative disease? Was it truly of no
sic CMF”).14 Three hundred and eighty-six value in postmenopausal women? Each of these
women with node-positive disease were random- issues was evaluated in individual randomized
ized to surgery ± CMF. As with the PAM trial, trials. Nevertheless, it became increasingly dif-
patients on CMF had a statisfically significant ficult, even with relatively large studies, to
improvement in DFS and a trend toward definitively come up with answers, as results
improved overall survival. Again the benefit was were often conflicting.
confined to premenopausal women with no sig- In 1985, 1990,18 and 1995,19 a consortium of
nificant benefit for postmenopausal patients.14 breast cancer researchers, the Early Breast Can-
Treatment with CMF (and its variants) became cer Trialists’ Collaborative Group (EBCTCG)
the standard adjuvant therapy for premenopausal conducted meta-analyses, using primary data
women with node-positive disease (see Table from multiple randomized trials of adjuvant
12–1). Although there are several ways of admin- chemotherapy to address these issues. The 1990
istering CMF (eg, classic monthly: po C ´ 14 overview looked at 13 studies (enrolling ~3,400
days + IV MF day 1 and 8; all IV every 3 weeks), women) comparing single-agent versus combi-
several studies support the classic 28-day pro- nation chemotherapy.18 There was greater bene-
gram as being the most effective.15,16 On the fit with polychemotherapy, a relative 17 ± 5 per-
other hand, in a subsequent randomized study, cent decrease in yearly risk of death due to
Bonadonna found that 6 months of CMF to be as breast cancer compared with single-agent ther-
efficacious as 1 year of drugs.17 Six months of apy, reinforcing the widely held clinical impres-
206 BREAST CANCER

sion. The 1995 overview, consequently, looked adjuvant chemotherapy, particularly CMF,
only at combination chemotherapy regimens, It became standard for both node-positive and
reviewed prolonged chemotherapy versus no high-risk, node-negative breast cancers. In post-
chemotherapy in 47 trials encompassing 18,000 menopausal women, chemotherapy was reserved
women; longer versus shorter chemotherapy in for receptor-negative patients because of the
6,100 patients in 11 trials; CMF versus anthra- perceived lesser benefit and increased toxicity.
cycline (doxorubicin or epirubicin)–based com- It was also being used for high-risk operable
binations in 6,000 women in 11 trials.19 For all male breast cancer.20
studies, there was a statistically significant ben-
efit in polychemotherapy versus no chemother- Doxorubicin
apy in terms of recurrence (relative decrease of
24%, p < .00001) and also survival (relative The suggestion that doxorubicin-based chemo-
decrease in mortality of 15%, p < .00001). therapy was more effective than CMF in the
Chemotherapy was not just delaying recurrence. palliation of advanced breast cancer11 led to the
Comparisons of standard durations of adjuvant use of combinations containing the drug as
CMF-like regimens (6 months) with more pro- adjuvant therapy particularly in poor prognostic
longed durations found no significant survival groups, such as those with multiple positive
benefit to more prolonged therapy,19 confirming nodes. The 1995 overview analyzed 11 CMF
Bonadonna’s results.17 The benefits of versus anthracycline (doxorubicin or epiru-
chemotherapy in the 1995 overview was seen in bicin) polychemotherapy trials.19 At 5 years
all nodal groups.19 In trials of combination after surgery, anthracycline combinations
chemotherapy versus control, mortality was cut decreased recurrence by an absolute 3.2 percent
by the same relative amount in both node-posi- (p = .006) and mortality by an absolute 2.7 per-
tive and node-negative patients. Given the dif- cent (p = .02). Individual trials themselves have
ferences in the risk of death in the two groups, been contradictory, but there are some general
the absolute benefit, however, was different. In trends. The NSABP Protocol B15 showed that
women under age 50 years, the absolute in node-positive women, a short four-treatment
decrease in death at 10 years was 12.4 percent course of IV doxorubicin/cyclophosphamide
(p < .00001) in the node-positive group and 5.7 (AC) (see Table 12–1) was equivalent to the
percent (p < .02) in the node-negative group. effect of classic CMF for 6 months.21 Further-
Finally, despite the many seemingly negative tri- more, the AC regimen was effective and well
als, polychemotherapy was found to be of value tolerated in postmenopausal women. It is now
in postmenopausal women but only in the 50- to widely used as an alternative to CMF for poor
69-year-old age group.19 For node-positive prognosis node-negative and 1- to 3-node–pos-
women over 50 years, chemotherapy decreased itive adjuvant therapy.21 Its popularity is based
the rate of death from any cause by an absolute on the briefer duration of treatment (3 versus 6
2.3 percent (p = .002), far less than in younger months), less overall toxicity and lower risk of
women. For node-negative older women, how- permanent menopause in younger women.
ever, the absolute decrease in rate of death Studies comparing CMF with CAF (doxoru-
(6.4% p < .005) was comparable with that in bicin substituted for methotrexate) have been
younger women. The paradox of chemotherapy harder to interpret. They have suffered from
having a greater absolute benefit in post- using the inferior all-IV 3-weekly CMF in the
menopausal node-negative women than in node- control arm or using epirubicin as their anthra-
positive ones is unexplained. As a result of the cycline drug. The Southwest Oncology Group
multiple studies that comprised the EBCTCG (SWOG), however, did conduct a study of stan-
overviews, by 1990, in premenopausal women, dard CMF versus CAF (± tamoxifen) in node-
Adjuvant Systemic Therapy of Early Breast Cancer 207

negative women. Their preliminary report was The assumption that there is a dose-
of a small but statistically significant 2 percent response relationship to tumor cell kill and the
improvement in survival (p = .03) for the CAF development of cytokine and stem cell bone
groups compared with the CMF groups,22 sim- marrow support has lead to a series of studies
ilar to the 1995 meta-analysis results.19 looking at higher-or more-intense-dose adju-
Whether such a small benefit in a relatively vant chemotherapy regimens. These studies are
good prognosis group warrants the excess toxi- of three types. Some escalate drugs two to four
city is questionable. The difference, however, times the conventional dosage and use cytokine
would be clinically significant in the high-risk support (escalated conventional dose). Others
multinode-positive patient. If the final results escalate drug doses minimally but give drugs at
of this study continue to show the difference, it briefer intervals (dose-dense therapy). There
would support CAF ´ 6 as an alternative for are few studies that address this concept. The
adjuvant chemotherapy for women with four or third approach uses massive (5- to 10-fold)
more positive lymph nodes. dose escalation and requires stem cell support
(“bone marrow transplant”). Only for the esca-
Investigational Approaches lated conventional dose approach are final
results of randomized trials available. They are
Unfortunately, even with aggressive doxoru- very disappointing. The NSABP trials B-2226
bicin-based chemotherapy, the prognosis and B-2527 randomized node-positive patients
remains poor for women with many axillary to conventional AC ´ 4 or to regimens contain-
nodes involved.19 Another approach to improv- ing higher doses of cyclophosphamide (up to
ing the results was the use of multiple four-fold escalation with GCSF). There was no
non–cross-resistant agents as adjuvant therapy. disease-free or overall survival benefit to the
Studies looking at CMF variants alternating higher-dose schedules. The CALGB 9344 trial
with doxorubicin regimens have not consis- randomized conventional AC ´ 4 (± paclitaxel)
tently shown benefit.21,23,24 The introduction of versus AC ´ 4 (± paclitaxel) with the doxoru-
the taxanes, which are highly active and not bicin escalated by 25 or 50 percent. There was
cross-resistant with doxorubicin, offer greater no benefit to the higher-dose doxorubicin regi-
hope of improving outcomes. Several studies mens compared with those containing conven-
are evaluating doxorubicin based regimens ± tional AC.25 To date, there is no convincing evi-
paclitaxel or docetaxel. The Cancer and Acute dence to support escalated standard-dose
Leukemia Group (CALGB) reported the pre- adjuvant chemotherapy.
liminary results of CALGB 9344, which found While high-dose chemotherapy with stem
that the addition of four doses of paclitaxel after cell transplant is widely employed in some cen-
AC ´ 4 (see Table 12–1) in node-positive ters as adjuvant therapy for women with 10 or
women decreased recurrence rate by 22 percent more involved nodes, data to support it are
and death by 26 percent. The absolute decrease incomplete. Proponents of the approach cite
in mortality at 18 months was 2 percent vastly superior survival results in women
(p = .039).25 While it is too soon to state that AC undergoing the technique compared with his-
´ 4 ® paclitaxel ´ 4 is the standard therapy for torical controls receiving conventional chemo-
node-positive disease (or if it is better than CAF therapy. Critics believe the superiority com-
´ 6), the data suggest a role for the taxanes. It is pared with historical controls is due to patient
reasonable to consider AC® paclitaxel as an selection and cite a small randomized study
alternative to CAF in high-risk situations where that found no benefit in the approach.28 The
the toxicity of prolonged doxorubicin adminis- Eastern Cooperative Oncology Group (ECOG)
tration is a concern (Table 12–2). has completed a study in women with 10 or
208 BREAST CANCER

Table 12–2. GUIDELINES FOR THE CHOICE OF ADJUVANT SYSTEMIC THERAPY

Node-Tumor-Receptor Status Premenopausal Postmenopausal Over Age 70 Years

Axillary node negative: tumor < 1 cm No Rx*‡ No Rx*‡ No Rx*‡


Axillary node negative: tumor > 1 cm or Tamoxifen or CMF† or Tamoxifen or Tamoxifen
Poor prognosis receptor positive AC or tamoxifen + AC tamoxifen + AC*
Axillary node negative: tumor > 1 cm or CMF† or AC AC or CMF† Chemo Rx on
Poor prognosis receptor negative‡ individual basis*
Axillary node positive: 1 to 3 + nodes CMF† or AC ± Tam Tamoxifen ± AC* Tamoxifen
Receptor positive (or CMF†)
Axillary node positive: 1 to 3 + nodes CMF† or AC AC (or CMF†) Chemo Rx on
Receptor negative‡ individual basis
Axillary node positive: ³ 4 + nodes CAF or AC® paclitaxel Tamoxifen + CAF Tamoxifen (± chemo Rx
Receptor positive ± tamoxifen or AC® paclitaxel on individual basis)
Axillary node positive: ³ 4 + nodes CAF or AC® paclitaxel CAF or Chemo Rx on
Receptor negative‡ AC® paclitaxel individual basis

* If multiple prognostic markers adverse consider Rx



If c-erbB-2 overexpressed doxorubicin based chemotherapy should be used

Consider Tamoxifen as chemopreventive agent
Rx = therapy; CMF = cyclophosphamide, methotrexate, fluorouracil; AC = adriamycin, cyclophosphamide; CAF = cyclophosphamide, adriamycin,
fluorouracil.

more positive nodes of CAF ´ 6 versus CAF metastatic breast cancer in postmenopausal
´ 6 followed by high-dose chemotherapy with women.29 Estrogen receptor could be used to
stem cell reinfusion. The CALGB trial is also predict response of metastatic disease.8 Initial
conducting a study in 10 or more node-positive reports of randomized trials of adjuvant
patients. They are randomized to CAF followed chemotherapy in early breast cancer found min-
by low-dose consolidation chemotherapy or by imal benefit in postmenopausal women com-
the same drugs given in high doses with stem pared with younger women.13,14 Trials in the
cell support. For women with four to nine United Kingdom and Scandinavia began look-
nodes involved, SWOG is conducting a study of ing at adjuvant tamoxifen after surgery in node-
sequential high conventional dose positive postmenopausal breast cancer patients.
doxorubicin® paclitaxel® cyclophosphamide In 1983, Baum reported preliminary results of
therapy versus AC ´ 4 followed by high-dose one of the studies. Tamoxifen given after
chemotherapy with stem cells. The latter trial, surgery decreased recurrence and increased
unfortunately, suffers from not having a truly survival.30 Since then over 50 randomized trials
standard control arm. Hopefully, when the have been conducted looking at the role of
longterm results of these large studies are adjuvant tamoxifen in 37,000 women. As with
reported, we will know what role (if any) high- chemotherapy, the EBCTCG conducted meta-
dose chemotherapy with stem cell support has analysis overviews of the tamoxifen trials, most
in the adjuvant treatment of early breast cancer. recently in 1995.31 Tamoxifen was beneficial in
node-positive and node-negative disease, in
HORMONAL THERAPY postmenopausal and premenopausal patients.
Although any duration of therapy was benefi-
cial, longer durations were more beneficial
Tamoxifen
than briefer duration. Only for women with
Multiple events in the late 1970s led to the tumors not containing ER was there no bene-
development of tamoxifen as the most widely fit.31 For all women given tamoxifen, there was
prescribed drug for the adjuvant systemic ther- a 26 percent relative decrease in recurrence, a
apy of early breast cancer. Tamoxifen was 14 percent relative decrease in death, and, at 10
shown to be an effective and safe therapy of years after surgery, an absolute decrease of 3.7
Adjuvant Systemic Therapy of Early Breast Cancer 209

percent in death from any cause, compared sidered an alternative to chemotherapy in pre-
with women not getting the drug (p < .00001).31 menopausal women. Tamoxifen is also of value
For women with tumors containing hormone in receptor-positive male breast cancer.36
receptor and receiving 5 years of tamoxifen, the
absolute decrease in death at 10 years was 5.6 Oophorectomy and Gonadotropin
percent for node-negative tumors and 10.9 per- Releasing Hormone Analogue
cent for node-positive tumors.31 In the 1990s,
tamoxifen became standard adjuvant therapy The sporadic use of adjuvant oophorectomy after
for all postmenopausal women with node-posi- breast cancer surgery in younger women was
tive and poor-prognosis, node-negative breast continued by surgeons for many years in the
cancers containing hormone receptor. hope of preventing recurrence. Randomized tri-
Several controversies persist in the use of als looking at its value date back 50 years.37
adjuvant tamoxifen. One is the duration of ther- Unfortunately, these early trials suffer from the
apy; the other is its role in premenopausal lack of hormone responsiveness of most breast
women. While the meta-analysis and individual cancers in premenopausal women and they pre-
randomized trials favor 5 years of therapy,31 two date our ability to predict responsiveness with
studies of 5 years versus more than 5 years in hormone receptor measurements. The EBCTCG
predominantly node-negative women found no conducted overview meta-analyses of adjuvant
benefit to additional years of therapy.32,33 The oophorectomy in 1985, 1990, and 1995. The
ECOG, however, found that more than 5 years most recent overview encompassed 12 trials ran-
of tamoxifen added to adjuvant chemotherapy in domizing 2,100 women to surgical or radiation
women with receptor containing tumors was oophorectomy versus no castration.38 In women
more beneficial than only 5 years of the drug.34 under the age of 50 years, oophorectomy
In general, the standard approach is to give resulted in an 18 percent relative decrease in
node-negative women 5 years of tamoxifen; for recurrence, an 18 percent relative decrease in
node-positive women, 5 years is suggested, but death, and, at 15 years after surgery, an absolute
individualizing duration of therapy on the basis decrease of 6.3 percent in death from any cause,
of risk of recurrence and toxicity is widely done. compared with women not getting the procedure
The Oxford Group is conducting a trial (p < .001).38 The relative benefit was the same in
(ATLAS), which is directly addressing the opti- node-negative and node-positive patients.38
mal duration of adjuvant tamoxifen therapy. These results are very similar to the chemother-
In the United States, chemotherapy is the apy meta-analysis results. Furthermore, in a
predominant form of adjuvant systemic therapy Scottish trial, adjuvant CMF was compared with
used in premenopausal women. This is not sur- oophorectomy in premenopausal women with
prising since many younger women have breast node-positive disease.39 There was no difference
cancers that do not contain hormone receptor seen in the overall result. In women with recep-
and are unlikely to benefit from adjuvant tor-positive tumors, the trend was the superiority
tamoxifen. Yet NSABP trial B-14 found tamox- of castration; in receptor-negative disease CMF
ifen to be an effective therapy in premeno- appeared better.
pausal axillary node-negative women.35 The In the United States, adjuvant surgical cas-
meta-analysis looking at 5 years of tamoxifen tration is rarely done these days, with tamox-
found no significant difference in the benefit of ifen the preferred adjuvant hormonal approach
tamoxifen between younger and older in younger women (if any hormonal approach
patients.31 This emphasizes that in hormone is used). The introduction of gonadotropin-
receptor-positive breast cancer (particularly releasing hormone (GnRH) analog has the
node-negative disease), tamoxifen can be con- potential to change this practice. These drugs,
210 BREAST CANCER

when given by slow-release depot injection, adjuvant therapy. The ATAC trial is a multina-
continually stimulate the pituitary, eventually tional randomized double-blinded study in
depleting it of FSH and LH. This results in the postmenopausal women of adjuvant tamoxifen
cessation of ovarian function, achieving a bio- versus the new AI anastrozole (Arimidex) ver-
chemical oophorectomy.7 The GnRH analog, sus the combination for 5 years. Another trial
goserelin (Zoladex) and leuprolide (Lupron) looks at women that have received 5 years of
have antitumor activity comparable with tamoxifen and are then being randomized to no
oophorectomy and with tamoxifen against further therapy or treatment with the AI letro-
overt hormone-responsive metastatic disease zole (Femara). The results of the toremifene,
in premenopausal women.40 Furthermore, their arimidex, and letrozole trials and a European
action on the ovary is reversible. Currently, the trial of the AI formestane are not yet known.
European “ZEBRA” study is comparing adju- To date, other than tamoxifen and oophorec-
vant goserelin with chemotherapy in pre- tomy, there are no standard hormonal adjuvant
menopausal women. Despite its proven effi- therapies.
cacy as adjuvant therapy in younger women,
oophorectomy is unlikely to be widely COMBINED CHEMOHORMONAL
accepted in the United States. THERAPY

Other Hormonal Approaches The 1995 EBCTCG overviews looked at the


relative benefits of adjuvant combined chemo-
Several other hormonal therapies have activity hormonal therapy versus single-modality treat-
against metastatic breast cancer and have been ment.19,31,38 There was a suggestion that in
evaluated as adjuvant therapy in early disease. women aged 50 to 69 years, tamoxifen plus
Toremifene (Fareston) is a derivative of tamox- chemotherapy decreased the annual risk of
ifen with a similar mechanism of action and death by 10 percent compared with tamoxifen
activity against disseminated disease.7 It is alone.31 The issue was prospectively studied in
being evaluated in randomized trials against newer trials. The NSABP, SWOG, and the
tamoxifen as adjuvant therapy in older women. International Breast Cancer Study Group, each
Progestins lower endogenous estrogen levels in found benefit in their studies of combined ther-
postmenopausal women and cause tumor apy versus tamoxifen alone in postmenopausal
regression in many women with advanced dis- women;44–46 the National Cancer Institute
ease.7 Medroxyprogesterone has been studied (NCI) of Canada did not.47 While it is prema-
as adjuvant therapy in randomized trials, with ture to suggest that all postmenopausal women
negative results.41,42 Aromatase inhibitors (AI) with receptor-positive cancer should receive
inhibit the enzyme that catalyzes the conver- chemotherapy and tamoxifen; certainly, it is
sion of androgen to estrogen. They, too, lower appropriate in selected high-risk women under
serum (and intracellular) estrogen levels in the age of 70 years. The best way to combine
older women and are effective hormonal thera- the two, simultaneously or sequentially, still
pies of metastatic breast cancer.7 Aminog- remains unresolved.
lutethimide, one of the first-generation aro- For women under the age 50 years, the 1995
matase inhibitors, however, was no better than overview suggested that the addition of
placebo in an adjuvant trial after surgery in oophorectomy to adjuvant chemotherapy was
postmenopausal women.43 New classes of of borderline benefit, with a nonstatistically
more potent and selective aromatase inhibitors significant decrease of 10 percent in the annual
have been recently introduced for the treatment mortality.38 Furthermore, the ECOG recently
of advanced disease and are being evaluated as reported the preliminary results of a random-
Adjuvant Systemic Therapy of Early Breast Cancer 211

ized study in premenopausal women with hor- apy. The NSABP trial B-18 randomized women
mone-sensitive, node-positive breast cancer. to receive four cycles of preoperative AC or
Women were randomized to receive CAF ´ 6 or four cycles of postoperative therapy.49,50 In
CAF ´ 6 + 2 years of goserelin or CAF ´ 6 + women with tumors > 5 cm in diameter, there
goserelin + tamoxifen. The results are prelimi- was a near-doubling of the breast conservation
nary. It seems unlikely that adding goserelin to rate with neoadjuvant chemotherapy.50 Never-
adjuvant chemotherapy improves results in theless, there was no difference in overall dis-
premenopausal women in whom the chemo- tant recurrence and survival.49 A new NSABP
therapy already achieved a chemical castration trial (B-27) is asking the same questions but
but may be of value in younger women who are looking at AC ± docetaxel given in various
still menstruating. Unfortunately, the meta- neoadjuvant and/or adjuvant combinations. In
analyses did not address these issues.31 The patients with large hormone receptor-positive
addition of tamoxifen in the ECOG trial seemed breast cancers, neoadjuvant tamoxifen has
beneficial in older women in whom chemother- been shown to be as effective as chemotherapy
apy led to menopause. Although not conclu- in shrinking the tumor to facilitate breast con-
sively proven effective, chemotherapy plus servation.51 It should be considered in patients
tamoxifen is often given to premenopausal that are not candidates for chemotherapy
women with receptor-positive early breast can- because of age or infirmity. For now, neoadju-
cer, usually because the chemotherapy has ren- vant therapies remain an effective way to facil-
dered them menopausal or more recently for itate breast conservation but not to improve
chemopreventive reasons (vide infra). survival over that achieved with postoperative
adjuvant treatment.
NEOADJUVANT THERAPY
DUCTAL CARCINOMA IN SITU
Increasingly, lumpectomy plus radiation has
become the desired standard of local therapy Although the primary role of adjuvant systemic
for early breast cancer. Unfortunately, tumor or therapy is to treat occult distant disease, there
breast size in many women make lumpectomy may also be a local benefit, at least in ductal
cosmetically or technically not feasible. The carcinoma in situ trial (DCIS). The NSABP
gratifying results with initial chemotherapy in trial B-24 randomized 1,804 women with DCIS
the treatment of locally inoperable breast can- treated with lumpectomy plus radiation to no
cer (such as inflammatory carcinoma) led to further therapy or to 5 years of tamoxifen.52 At
trials of preoperative chemotherapy in the hope 5 years, tamoxifen decreased the risk of the
of shrinking operable but large tumors to the development of invasive breast cancer in the
point that breast conservation could be accom- treated breast by 47 percent (2.1% versus 3.4%
plished. Bonadonna’s group in Milan, in two in control, p = .04) and of all breast cancer
trials using several preoperative chemotherapy events (ipsilateral and contralateral) by 34 per-
regimens, was able to perform breast preserva- cent (8.8% versus 13% in control, p = .007).52
tion surgery (quadrentectomy) in 66 percent of These results apply only to women who had
women with tumors > 5 cm.48 No one preoper- lumpectomy/radiation for DCIS. They are not
ative chemotherapy regimen was clearly supe- relevant to women treated with mastectomy.
rior.48 Building on these findings, the NSABP While the absolute magnitude of the benefit
investigated whether preoperative chemother- was small, certainly tamoxifen should be con-
apy, besides shrinking the primary tumor, sidered in many women with DCIS treated with
might also be more effective as an adjuvant breast conservation, particularly those with
systemic therapy than postoperative chemother- high-risk pathology.
212 BREAST CANCER

THE CHOICE OF ADJUVANT THERAPY that c-erbB-2 overexpression may predict unre-
AND THE BIOLOGY OF THE TUMOR sponsiveness to adjuvant tamoxifen,55,56 recent
AND OF THE PATIENT studies are not supportive.57,58
Though the biology of the tumor is impor-
Despite the successes of adjuvant chemother- tant, so too is the biology of the patient in the
apy and hormonal therapy, many patients that choice of adjuvant systemic therapy. Age is the
receive such treatments have recurrences and most important factor. There is little data on
die. To optimize results, when choosing treat- the efficacy of chemotherapy in patients over
ment, tumor and patient biology must be taken the age of 70 years; of the 19,000 reviewed in
into account. This is most apparent in the use of the EBCTCG chemotherapy overview, there
hormonal therapy. Although there is a small but were only 600 women over 70 years. They
real response rate with tamoxifen in metastatic could draw no conclusion as to the value of
hormone receptor-negative breast cancer, the chemotherapy in that age group.19 Chemother-
1995 meta-analysis found no significant bene- apy should be reserved for women over 70
fit to adjuvant tamoxifen in women with recep- years with poor prognostic tumors containing
tor-poor tumors.31 It is, therefore, critical that no hormone receptor, with a reasonable life
hormone receptor be measured in any patient expectancy, and that are physiologically in
with newly diagnosed breast cancer.4 Although excellent health. It is important to emphasize,
there was some controversy in the past as to however, that being over 70 years should not a
whether hormone receptor status is also of priori exclude a woman from consideration of
value in predicting response to chemotherapy, it chemotherapy. In the younger postmenopausal
is now known that no such relationship exists.19 groups, chemotherapy is clearly beneficial
On the other hand, there is preliminary data alone or when added to tamoxifen. Neverthe-
that suggest the presence of an overexpressed less, it should not be universally given. The
c-erbB-2 oncogene in breast cancers may have EBCTCG trial analyzed the same randomized
predictive value in the choice of chemother- trials where they found a survival benefit to
apy.53,54 The NSABP trial found that melphalan chemotherapy plus tamoxifen in women 50 to
with fluorouracil + doxorubicin was more effec- 69 years but found no increase in “quality (of
tive adjuvant therapy than melphalan with fluo- life)–adjusted survival” compared with tamox-
rouracil alone. However, when the data were ifen alone.59 The implication is not that
reanalyzed, the benefit was only seen in patients chemotherapy should not be given to women
whose tumors overexpressed c-erbB-2.53 The age 50 to 69 years but rather that its use in
CALGB trial randomized women to three dif- addition to tamoxifen should be limited to
ferent dose-intense CAF regimens. The two high-risk, poor-prognosis patients, despite
higher-dose regimens (both within the range of calls to the contrary. The administration of
standard dosage) were superior to the low-dose adjuvant systemic therapy cannot be consid-
regimen; however, an analysis of the data by ered a standardized process. It must always be
c-erbB-2 status found the difference to be pre- individualized.
sent only in the subset of oncogene overexpres-
sor.54 These data suggest that if adjuvant BENEFITS VERSUS TOXICITY AND
chemotherapy is to be given in the presence of RISKS OF THERAPY
c-erbB-2 overexpression, it should contain dox-
orubicin at full dose. What is not clear is The acute toxicities of adjuvant systemic ther-
whether lack of c-erbB-2 overexpression pre- apy of early breast cancer are significant but
dicts the effectiveness of nondoxorubucin-con- generally well tolerated and are easily justified
taining regimens. While there are suggestions given the potential benefit. The toxicities of
Adjuvant Systemic Therapy of Early Breast Cancer 213

chemotherapy can be divided into those of the ertheless, the incidence of second malignancies
CMF-like regimens and those of the doxoru- has been low. The ECOG estimated the risk of
bicin regimens. All chemotherapies used as secondary leukemia or myelodysplasia after its
adjuvant treatment cause significant myelosup- CMF adjuvant regimens to be less than 0.2 per-
pression, with leukopenia generally clinically cent similar to that of the general population.62
more significant than anemia or thrombocy- Bonadonna could find no increased risk of
topenia. In the NSABP trials of classic CMF malignancy in long-term follow-up of his adju-
´ 6, the incidence of neutropenia less than vant CMF patients.63 The M.D. Anderson Hos-
2,000 was ~ 10 percent and severe infection pital, in reviewing its adjuvant doxorubicin pro-
about 1 percent.21 With AC ´ 4, it is 4 percent grams, found the risk of secondary leukemia or
severe neutropenia and 2 percent severe infec- myelodysplasia to be 0.2 to 0.5 percent.64 There
tion.21 With 6 months of CAF, the risk of is some suggestion that concomitant high-dose
leukopenia and infection is higher. Thrombocy- cyclophosphamide may increase the doxoru-
topenia is seen in less than 1 percent of patients bicin leukemia risk.65 Other than menopause in
in most regimens.21 Doxorubicin-containing younger women, long-term complications of
regimens are more emetogenic than CMF; how- adjuvant chemotherapy are infrequent.
ever, the incidence of severe vomiting is rapidly Tamoxifen is a selective estrogen receptor
dropping with the introduction of serotonin modular (SERM) and so may be antiestrogenic
antagonists. Alopecia is nearly universal with or estrogenic, depending on its interaction with
doxorubicin and is seen in about 40 percent of the individual tissue receptor. Its toxicity pro-
CMF patients.21 Diarrhea is rarely seen with file reflects this duality. The most common
either regimens; the use of serotonin antagonist acute tamoxifen side effects are menopausal
antiemetics is associated with constipation (and symptoms. In the NSABP trial B-14, hot
mild headache). Cystitis is seen in about 1 per- flashes were seen in about two-thirds of
cent of patients receiving cyclophosphamide- patients, about a third had weight gain, fluid
containing regimens and correlates with longer retention, and vaginal discharge, and a quarter
durations of therapy.21 Other rare side effects of experienced nausea, and weight loss.35 Irregu-
both regimens include mucositis, thromboem- lar menses were seen in a fourth of pre-
bolic events, and, for doxorubicin, extravasa- menopausal women.35 The only significant
tion skin ulceration. acute toxicities were rare thromboembolic
The most common chronic chemotherapy events: deep vein thrombosis in 0.8 percent and
toxicity is the cessation of menses and induc- pulmonary embolus in 0.4 percent. Mood
tion of menopause in premenopausal women. swings and depression are unusual. Very-high-
This is more common with 6 months of CMF dose tamoxifen may cause retinal changes, but
(and CAF) than with AC ´ 4. In one study, these are rarely seen with conventional doses.
amenorrhea was seen in 68 percent of women There are reports of cataracts in patients on the
on CMF and 34 percent of women on AC.60 drug.66 In a large review of ocular toxicity from
Symptomatic cardiomyopathy is a rare compli- the NSABP, there were no cases of vision-
cation seen with doxorubicin-containing regi- threatening eye toxicity with tamoxifen.66
mens. The risk is less than 1 percent with There is an increased risk of developing
cumulative doxorubicin doses less than 350 uterine cancer in women receiving tamoxifen
mg/M2.61 The cumulative dose with AC ´ 4 is (2/1,000/y of therapy versus 1/1,000/y in con-
240/m2; with CAF, it is 360/m2. The risk is trol).67 In the 1995 meta-analysis, 10 years after
increased with age, left chest wall irradiation, breast surgery, women on 5 years of the drug
and prior heart disease. Chemotherapy agents had a 1.1 percent risk of uterine malignancy
are carinogenic in experimental systems. Nev- compared with 0.3 percent for those who did
214 BREAST CANCER

not receive the drug.31 Furthermore, most receptor-containing tumor warranting adjuvant
reported cases were early stage and highly cur- therapy (node + or –), tamoxifen should be
able,68,69 although fatal cases of uterine cancer given for 5 years. If at particularly high risk,
have been reported.70 In contrast to the uterine such as with multiple positive nodes or an
cancer effects are thoses of tamoxifen on the extremely large tumor, the addition of
development of contralateral breast cancer. chemotherapy (AC ´ 4) should be considered
Multiple studies have found that tamoxifen if the patient is in good health and has a rea-
given to prevent recurrence of previous breast sonable life expectancy. For receptor-negative
cancer significantly decreases the risk of devel- postmenopausal women with node-positive or
oping new contralateral breast cancer.68 The high-risk, node-negative disease, chemother-
1995 overview found a 47 percent decrease in apy (CMF or AC ´ 4 or for multiple nodes
the risk of contralateral malignancy in women AC ´ 4® paclitaxel ´ 4 or CAF) should be
receiving 5 years of the drug.31 This effect of administered. The borderline efficacy of CMF
tamoxifen was confirmed by the Breast Cancer in postmenopausal women make doxorubicin-
Prevention Trial (NSABP P-1), which found a containing regimens preferable if cardiac func-
virtually identical decrease in the development tion permits. Chemotherapy should be used
of new breast cancers in high risk women with- cautiously in women over 70 years. In the
out a history of the disease.71 This effect would United States, for premenopausal women with
suggest that even in women with a history of positive nodes, adjuvant chemotherapy is stan-
receptor-negative breast cancer, tamoxifen dard. For patients with 1 to 3 positive nodes,
might be considered not to prevent recurrence AC or CMF is widely used. For women with
but to decrease the risk of new malignancy. four or more involved nodes, CAF ´ 6 is the
Other beneficial effects of adjuvant tamox- standard. Recent data suggest that the addition
ifen include an estrogenic-like decrease in bone of taxanes to doxorubicin regimens also has a
loss in postmenopausal women,72–74 decrease in role in this group (AC ´ 4® paclitaxel ´ 4) and
cholesterol,75 and, in some studies, decreased may supplant CAF.25 If the patient’s tumor is
cardiac mortality.76–78 Overall, the risk/benefit receptor positive, tamoxifen is frequently
ratio strongly favors tamoxifen’s use as adju- added to chemotherapy, but the data to support
vant therapy for hormone receptor-containing this are limited. Nevertheless, if the chemother-
early-stage breast cancer. apy renders a woman menopausal or there is
great concern about contralateral breast can-
SUMMARY cer, few would fault the addition of tamoxifen
to chemotherapy for node-positive, premeno-
The addition of adjuvant chemotherapy or hor- pausal, receptor-positive, breast cancer adju-
monal therapies to local treatment signifi- vant therapy. In Europe, such women might
cantly decreases recurrence and mortality in undergo oophorectomy as an alternative to
women with axillary node-positive or high- chemotherapy ± tamoxifen.39 For high-risk,
risk, node-negative, operable breast cancer. node-negative premenopausal women, there
The choice of therapy should be individualized are several alternatives. If the tumor is hor-
on the basis of the perceived risk of recurrence, mone receptor positive, tamoxifen, CMF, or
particularly as determined by nodal status and AC (or chemotherapy + tamoxifen) are accept-
tumor size, patient age and general health, and able. If receptor negative, chemotherapy with
the presence or absence of hormone (estrogen CMF or AC is used. Although the SWOG trial
or progesterone) receptor in the tumor. Some suggested the superiority of CAF in this group
general guidelines are reasonable (see Table of patients,22 the added toxicity is likely to limit
12–2). For all postmenopausal women with the use of this combination in node-negative
Adjuvant Systemic Therapy of Early Breast Cancer 215

women. The use of high-dose chemotherapy 8. Desombre ER, Carbone PP, Jensen EV, et al. Spe-
with stem cell support should be considered cial Report. Steroid receptors in breast cancer.
investigational, even for patients with 10 or N Engl J Med 1979;301:1011–2.
9. Hoogstraten B, Fabian C. A reappraisal of single
more involved nodes. Neoadjuvant therapies
drugs for advanced breast cancer. Cancer Clin
are appropriate in an attempt to achieve breast Trials 1979;2:101–98.
conservation but have not been proved a more 10. Cooper RG. Combination chemotherapy in hor-
effective adjuvant systemic therapy than post- mone resistant breast cancer. Proc Am Assoc
operative treatment.49 Delay of local treat- Cancer Res 1963;10:15.
ment (surgery and/or radiation) for the 11. Bull JM, Tormey DC, Li SH, et al. A randomized
administration of neoadjuvant49 or adjuvant comparative trial of adriamycin versus metho-
trexate in combination drug therapy. Cancer
therapy79 does not negatively impact out-
1978;41:1649–57.
come. For women with high-risk DCIS treated 12. Fisher B, Ravdin RG, Ausman RK, et al. Surgical
with lumpectomy plus radiation, tamoxifen adjuvant chemotherapy in cancer of the breast:
therapy should be considered on the basis results of a decade of cooperative investiga-
of the preliminary results of NSABP trial tions. Ann Surg 1968;168:337–56.
B-24. 52 Whenever possible, women with 13. Fisher B, Carbone P, Economou SG, et al. L-Pheny-
lalanine mustard (L-PAM) in the management
early-stage breast cancer should be encour-
of primary breast cancer. A report of early find-
aged to enroll in RCTs of adjuvant therapies. ings. N Engl J Med 1975;292:117–22.
Only with such studies will further progress 14. Bonadonna G, Brusamolino E, Valagussa P, et al.
be made in reducing mortality in women with Combination chemotherapy as an adjuvant
breast cancer. treatment in operable breast cancer. N Engl J
Med 1976;294:405–10.
15. Engelsman E, Rubens RD, Klign JGM. Compari-
REFERENCES son of the classical CMF with a three weekly
intravenous CMF schedule in postmenopausal
1. Contesso H, Mouriesse H, Friedman S, et al. The patients with advanced breast cancer: an
importance of histologic grade in long-term EORTC study. Proceedings of the 4th EORTC
prognosis of breast cancer: a study of 1,010 Breast cancer Working Conference 1987;1:1.
patients, uniformly treated at the Institut Gus- 16. Goldhirsch A, Colleoni M, Coates AS, et al.
tave-Roussy. J Clin Oncol 1987;5;1378–6. Adding adjuvant CMF chemotherapy to either
2. Fisher ER, Gregorio RM, Fisher B, et al. The radiotherapy or tamoxifen: are all CMF’s alike?
pathology of invasive breast cancer. A syllabus the International Breast Cancer Study Group
derived from findings of the National Adjuvant (IBCSG). Ann Oncol 1998;9:489–93.
Breast Project. Cancer 1975;36:1–85. 17. Tancini G, Bonadonna G, Valgussa P, et al. Adju-
3. Donegan WL. Tumor-related prognostic factors for vant CMF in breast cancer: comparative 5 year
breast cancer. CA Cancer J Clin 1997;47:28–51. results of 12 versus 6 cycles. J Clin Oncol
4. Tumor Marker Guideline Panel, American Society 1983;1:2–10.
Clinical Oncology. Clinical practice guidelines 18. Early Breast Cancer Trialists’ Collaborative
for the use of tumor markers in breast and col- Group. Systemic treatment of early breast can-
orectal cancer. J Clin Oncol 1996;14:2843–77. cer by hormonal, cytotoxic, or immune ther-
5. Early Breast Cancer Trialists’ Collaborative Group. apy: 133 randomized trials involving 31,000
Effects of radiotherapy and surgery in early recurrences and 24,000 deaths among 75,000
breast cancer. An overview of the randomized women. Lancet 1992;339:1–15, 71–85.
trials. N Engl J Med 1995;333:1444–55. 19. Early Breast Cancer Trialists’ Collaborative
6. Beatson GT. On the treatment of inoperable cases Group. Polychemotherapy for early breast can-
of carcinoma of the mamma: suggestion for a cer: an overview of the randomized trials.
new method of treatment with illustrative Lancet 1998;352:930–42.
cases. Lancet 1996;2:104–7. 20. Bagley CS, Wesley MN, Young RC, Lipmann ME.
7. Locker GY. Hormonal therapy of breast cancer. Adjuvant chemotherapy in males with cancer
Cancer Treat Rev 1998;24:221–40. of the breast. Am J Clin Oncol 1987;6:45–50.
216 BREAST CANCER

21. Fisher B, Brown AM, Dimitrov NV, et al. Two tamoxifen after mastectomy for early breast
months of doxorubicin-cyclophosphamide cancer [letter]. Lancet 1983;2(8347):450.
with and without interval reinduction therapy 31. Early Breast Cancer Trialists’ Collaborative
compared with 6 months of cyclophosphamide, Group. Tamoxifen for early breast cancer: an
methotrexate, and fluorouracil in positive-node overview of the randomized trials. Lancet
breast cancer patients with tamoxifen-nonre- 1998;351:1451–67.
sponsive tumors: results from the NSABP B-15. 32. Fisher B, Dignam J, Bryant J, et al. Five versus
J Clin Oncol 1990;8:1483–96. more than five years of tamoxifen therapy for
22. Hutchins L, Green S, Ravdin P, et al. CMF versus breast cancer patients with negative lymph
CAF with and without tamoxifen in high risk nodes and estrogen receptor-positive tumors. J
node-negative breast cancer patients and a nat- Natl Cancer Inst 1996;88:1529–42.
ural history follow-up study in low-risk node 33. Stewart HJ, Forrest AP, Everington D, et al. Ran-
negative patients: first results of Intergroup domized comparison of 5 years of adjuvant
Trial INT 102. Proc Am Soc Clin Oncol tamoxifen with continuous therapy for oper-
1998;17:1a. able breast cancer. The Scottish Cancer Trials
23. Moliterni A, Bonadonna G, Valagussa P, et al. Breast Group. Br J Cancer 1996;74:297–9.
Cyclophosphamide, methotrexate and fluo- 34. Tormey DC, Gray R, Falkson G. Postchemother-
rouracil with and without doxorubicin in the apy adjuvant tamoxifen therapy beyond five
adjuvant treatment of resectable breast cancer years in patients with lymph node-positive
with one to three positive axillary nodes. J Clin breast cancer. Eastern Cooperative Oncology
Oncol 1991;9:1124–30. Group. J Natl Cancer Inst 1996;88:1828–33.
24. Tormey DC, Gray R, Abeloff MD, et al. Adjuvant 35. Fisher B, Costantino J, Redmond C. A random-
therapy with a doxorubicin regimen and long ized clinical trial evaluating tamoxifen in the
term tamoxifen in premonopausal breast can- treatment of patients with node negative breast
cer patients: an ECOG trial. J Clin Oncol 1992 cancer who have estrogen receptor positive
1992;10:1848–56. tumors. N Engl J Med 1989;320:479–84.
25. Henderson IC, Berry D, Demetri G, et al. 36. Ribeiro G, Swindell R. Adjuvant tamoxifen for
Improved disease-free and overall survival male breast cancer. Br J Cancer 1992;65:252–4.
from the addition of sequential paclitaxel but 37. Cole MP. A clinical trial of an artificial meno-
not from the escalation of doxorubicin dose in pause in carcinoma of the breast. INSERM
the adjuvant chemotherapy of patients with 1975;55:143.
node-positive primary breast cancer. Proc Am 38. Early Breast Cancer Trialists’ Collaborative
Soc Clin Oncol 1998;17:101a. Group. Ovarian ablation in early breast cancer:
26. Fisher B, Anderson S, Wickerham DL, et al. an overview of randomized trials. Lancet
Increased intensification and total dose of 1996;348:1189–96.
cyclophosphamide in a doxorubucin-cyclo- 39. Scottish Cancer Trials Breast Group and ICRF
phosphamide regimen for the treatment of pri- Breast Unit. Adjuvant ovarian ablation versus
mary breast cancer: findings from National CMF chemotherapy in premenopausal women
Surgical Adjuvant Breast and Bowel Project with pathological stage II breast carcinoma: the
B-22. J Clin Oncol 1997;15:1858–69. Scottish trial. Lancet 1993;341:1293–8.
27. Wolmark N, Fisher B, Anderson S. The effect of 40. Davidson NE. Ovarian ablation as treatment for
increasing dose intensity and cumulative dose young women with breast cancer. J Natl Can-
of adjuvant cyclophosphamide in node positive cer Inst Monographs 1994;16:95–9.
breast cancer: results of NSABP B-25. Breast 41. Focan C, Beaudin M, Salamon E. Adjuvant high
Cancer Res Treat 1997;46:26. dose medroxyprogesterone for early breast
28. Rodenhuis S, Richel DJ, van der Wall E, et al. A cancer: 13 years update of a multicenter ran-
randomized trial of high-dose chemotherapy domized trial. Eur J Oncol 1998;(Suppl 1):34.
and haematopoetic progenitor-cell support in 42. Pannuti F, Martoni A, Cilenti G, et al. Adjuvant
operable breast cancer with extensive axillary therapy for operable breast cancer with
node involvement. Lancet 1998;352:515–21. medroxyprogesterone acetate alone in post-
29. Mouridsen H, Palshof T. Tamoxifen in advanced menopausal patients or in combination with
breast cancer. Cancer Treat Rev 1978;5:131–41. CMF in premenopausal patients. Eur J Cancer
30. Baum M. Brinkley DM, Dosset JA, et al. Improved Clin Oncol 1988;24:423–9.
survival among patients treated with adjuvant 43. Jones AL, Powles TJ, Law M, et al. Adjuvant
Adjuvant Systemic Therapy of Early Breast Cancer 217

aminoglutethimide for postmenopausal patients lary lymph node-positive, hormone receptor-


with primary breast cancer: analysis at 8 years. negative breast cancer. J Natl Cancer Inst 1998;
J Clin Oncol 1992;10:1547–52. 90:1361–70.
44. Fisher B, Dignam J, DeCillis A, et al. The worth of 54. Thor AD, Berry DA, Budman DR, et al. ErbB-2,
chemotherapy and tamoxifen (TAM) over TAM p53, and efficacy of adjuvant therapy in lymph
alone in node negative patients with estrogen- node-positive breast cancer. J Natl Cancer
receptor positive invasive breast cancer: first Institute 1998;90:1346–60.
results from NSABP B-20 [abstract]. Proc Ann 55. Borg A, Caldetorp B, Ferno M, et al. ErbB-2
Meet Am Soc Clin Oncol 1997;16:1a. amplification is associated with tamoxifen
45. Albain K, Green S, Osborne CK, et al. Tamoxifen resistance in steroid-receptor positive breast
(T) versus cyclophosphamide, adriamycin and 5- cancer. Cancer Lett 1994;81:137–44.
FU plus either concurrent or sequential T in post- 56. Carlomagno C, Perrone F, Gallo C, et al. C-erbB-
menopausal receptor (+), node (+) breast cancer: 2 overexpression decreases the benefit of adju-
a Southwest Oncology Group phase III inter- vant tamoxifen in early-stage breast cancer
group (SWOG-8814, INT-0100) [abstract]. Proc without axillary node metastases. J Clin Oncol
Ann Meet Am Soc Clin Oncol 1997;16:128a. 1996;14:2702–8.
46. International Breast Cancer Study Group. Effec- 57. Soubeyran I, Quenel N, Coindre JM, et al. PS2 pro-
tiveness of adjuvant chemotherapy in combina- tein: a marker improving prediction of response
tion with tamoxifen for node-positive post- to neoadjuvant tamoxifen in post-menopausal
menopausal breast cancer patients. J Clin Oncol breast cancer. Br J Cancer 1996;74:1120–5.
1997;15:1385–94. 58. Berns EM, Foekens JA, van Staveren IL, et al.
47. Pritchard KI, Paterson AH, Fine S, et al. Random- Oncogene amplication and prognosis in breast
ized trial of cyclophosphamide, methotrexate cancer: relationship with systemic treatment.
and fluorouracil chemotherapy added to tamox- Gene 1995;159:11–8.
ifen as adjuvant therapy in postmenopausal 59. Gelber RD, Cole BF, Goldhirsch A, et al. Adjuvant
women with node-positive estrogen and/or chemotherapy plus tamoxifen compared with
progesterone receptor-positive breast cancer: a tamoxifen alone for postmenopausal breast
report of the National Cancer Institute of cancer: meta-analysis of quality-adjusted sur-
Canada Clinical Trials Group. J Clin Oncol vival. Lancet 1996;347:1066–71.
1997;15:2302–11. 60. Bines J, Oleske DM, Cobleigh MA. Ovarian func-
48. Bonadonna G, Valagussa P. Primary chemother- tion in premenopausal women treated with
apy in operable breast cancer. Semin Oncol adjuvant chemotherapy for breast cancer. J
1996;23:464–74. Clin Oncol 1996;14:1718–29.
49. Fisher B, Bryant J, Wolmark N, et al. Effect of 61. Von Hoff D, Layard MW, Basa P, et al. Risk fac-
preoperative chemotherapy on the outcome of tors for doxorubicin-induced congestive heart
women with operable breast cancer. J Clin failure. Ann Int Med 1979;91:710–7.
Oncol 1998;16:2672–85. 62. Tallman MS, Gray R, Bennett JM, et al. Leuke-
50. Fisher B, Brown A, Mamounas E, et al. Effect of mogenic potential of adjuvant chemotherapy
preoperative chemotherapy on local-regional for early-stage breast cancer: the ECOG expe-
disease in women with operable breast cancer: rience. J Clin Oncol 1995;13:1557–63.
findings from NSABP B-18. J Clin Oncol 63. Valagussa P, Tancini G, Bonadonna G. Second
1997;15:2483–93. malignancies after CMF for resectable breast
51. Willsher PC, Robertson JFR, Chan SY, et al. cancer. J Clin Oncol 1987;5:1138–42.
Locally advanced breast cancer: early results of 64. Diamandidou E, Buzdar AU, Smith TL, et al.
a randomized trial of multimodal therapy ver- Treatment-related leukemia in breast cancer
sus initial hormone therapy. Eur J Cancer patients treated with fluorouracil-doxorubicin-
1997;33:45–9. cyclophosphamide combination adjuvant
52. Wolmark N, Dignam J, Fisher B. The addition of chemotherapy: the University of Texas M.D.
tamoxifen to lumpectomy and radiotherapy in Anderson Cancer Center experience. J Clin
the treatment of ductal carcinoma in situ Oncol 1996;14:2722–30.
(DCIS): preliminary results of NSABP protocol 65. DeCillis A, Anderson A, Bryant J, et al. Acute
B-24. Breast Cancer Res Treat 1998;50:227. myloid leukemia and myelodysplastic syn-
53. Paik S, Bryant J, Park C, et al. ErbB-2 and drome on NSABP B-25: an update. Proc Am
response to doxorubicin in patients with axil- Soc Clin Oncol 1997;16:130a.
218 BREAST CANCER

66. Gorin MB, Day R, Costantino JP, et al. Long-term low-risk breast cancer patients: a randomized
tamoxifen citrate use and potential ocular toxi- study. J Clin Oncol 1994;12:992–7.
city. Am J Ophthalmol 1998;125:493–501. 74. Powles TJ, Hickish T, Kanis JA, et al. Effect of
67. Jordan VC, Assikis VJ. Endometrial carcinoma tamoxifen on bone mineral density measured
and tamoxifen: clearing up a controversy. Clin by dual-energy x-ray absorptiometry in healthy
Cancer Res 1995;1:467–72. premenopausal and postmenopausal women. J
68. Rutqvist LE, Johansson H, Signomklao T, et al. Clin Oncol 1996;14:78–84.
Adjuvant tamoxifen therapy for early stage 75. Love RR, Wiebe DA, Feyzi JM, et al. Effects of
breast cancer and second primary malignan- tamoxifen on cardiovascular risk factors in
cies. J Natl Cancer Inst 1995;87:645–51. postmenopausal women after 5 years of treat-
69. Fisher B, Costantino JP, Redmond CK, et al. ment. J Natl Cancer Inst 1994;86:1534–9.
Endometrial cancer in tamoxifen-treated breast 76. McDonald CC, Alexander FE, Whyte BW, et al.
cancer patients: findings from the National Cardiac and vascular morbidity in women
Surgical Adjuvant Breast and Bowel Project receiving adjuvant tamoxifen for breast cancer
(NSABP) B-14. J Natl Cancer Inst 1994;86: in a randomized trial. The Scottish Cancer Tri-
527–37. als Breast Group. Br Med J 1995;311:977–80.
70. Magriples U, Naftolin F, Schwartz PE, Carcangiu 77. Costantino JP, Kuller LH, Ives DG, et al. Coro-
ML. High-grade endometrial carcinoma in nary heart disease mortality and adjuvant
tamoxifen-treated breast cancer patients. J Clin tamoxifen therapy. J Natl Cancer Inst 1997;
Oncol 1993;11:485–90. 89:776–82.
71. Fisher B, Costantino JP, Wickerham DL, et al. 78. Rutqvist LE, Mattsson A. Cardiac and throm-
Tamoxifen for the prevention of breast cancer: boembolic morbidity among post menopausal
report of the NSABP P-1 study. J Natl Cancer women with early-stage breast cancer in a ran-
Inst 1998;90:1371–88. domized trial of adjuvant tamoxifen. The
72. Love RR, Mazess RB, Barden HS, et al. Effects of Stockholm Breast Cancer Study Group. J Natl
tamoxifen on bone mineral density in post- Cancer Inst 1993;85:1398–406
menopausal women with breast cancer. N Engl 79. Recht A, Come SE, Henderson IC, et al. The
J Med 1992;326:852–6. sequencing of chemotherapy and radiation ther-
73. Kristen B, Ejlertsen B, Dalgaard P, et al. Tamox- apy after conservative surgery for early-stage
ifen and bone metabolism in postmenopausal breast cancer. N Engl J Med 1996;334:1356–61.
13
Breast Cancer and
Radiation Therapy
MICHAEL A. LACOMBE, MD
WILLIAM D. BLOOMER, MD

The role of radiation therapy in the treatment of dard treatment. On the other hand, there may be
breast cancer has been affected by shifting subsets in the population of breast cancer
trends in treatment. Historically, mastectomy patients whose risk of recurrence is low enough
was the surgical treatment of choice for all after local excision alone to preclude radiother-
stages of disease. In later times, radiation ther- apy after lumpectomy. Future investigations in
apy was found to significantly reduce the risk BCT will be needed to individualize treatment
of local recurrence. Pioneers in radiotherapy based on these evolving parameters.
such as Gilbert Fletcher published important Routine postmastectomy radiotherapy has
dose response data for the control of regional fallen out of favor in recent years, despite proven
adenopathy.1 As the treatment of breast cancer efficacy in reducing local recurrence. Although
has changed, the role of radiotherapy has local recurrences were reduced, positive impact
evolved for all stages of disease. on overall survival was difficult to document.
Breast conservation therapy for ductal carci- Thus, the potential for distant metastatic disease
noma in situ (intraductal) and early-stage inva- is thought to be the single most important prog-
sive cancer has been shown to provide equiva- nostic factor for survival. Chemotherapy has
lent survival to mastectomy in properly selected emerged as an effective treatment for metastatic
patients. The importance of cosmesis in breast and potentially metastatic disease. Adriamycin-
conserving treatment can not be underesti- based chemotherapy is probably the most effec-
mated. Issues of self-esteem and sensuality add tive regimen, despite well-documented cardiac
complexity to the treatment decision. Breast morbidity associated with adriamycin. Cardiac
conservation therapy (BCT) is clearly estab- morbidity due to the late effects of radiation
lished as a standard treatment for early-stage began to be appreciated at the same time these
breast cancer. In fact, a consensus panel of the effective chemotherapy regimens were devel-
National Institutes of Health determined BCT oped.3 Additionally, systemic and local toxicity
to be the preferred treatment for patients eligi- prohibits concomitant delivery of radiotherapy
ble for the procedure.2 and systemic therapy for breast cancer. Radio-
Prognostic factors have been evaluated to therapy following mastectomy has therefore
determine those patients at high risk for recur- been infrequently used.
rence after BCT. There are many new patho- Recent data have emerged showing a sur-
logic and cytologic markers that may be used to vival benefit when radiotherapy is added to
predict local or distant recurrence after stan- mastectomy and chemotherapy as treatment for

219
220 BREAST CANCER

selected patients. Older studies of radiotherapy and increased screening. More patients are pre-
have been criticized for poor technique as well senting with stage I disease, fewer with stage II
as employing low-energy equipment.4 How- disease, with the number presenting with more
ever, historic studies of patients treated with advanced disease remaining the same. Studies
modern equipment showed a survival advan- comparing screened populations with unscreen-
tage. Thus, it is important to define those ed patients have demonstrated a reduction in
patients with locally advanced breast cancer overall breast cancer mortality in the screened
whose risk of local recurrence is significant patients.9,10 The logical conclusion is that the
enough to warrant radiotherapy. acceptance of a screening modality leads to
Confounding any evaluation of the treat- detection of tumors at an earlier stage as well as
ment results of breast cancer is the long natural initially increasing the incidence of the disease.
history of the disease, the interval evolution of Earlier staged tumors are more curable, and
surgical, radiotherapeutic, and systemic treat- their detection decreases mortality related to
ment, and the long latency of late-radiation the disease. The incidence of the disease may
sequelae developing. The long natural history eventually decline as a generation of patients
of breast cancer requires years of follow-up to completes screening.
dissect results of comparative trials. The devel- Acceptance of the equivalency of BCT to
opment of more effective therapies obscures mastectomy is not widespread. Although it is
results as local and distant recurrences may not estimated that 70 to 80 percent of patients
develop for many years. During these long fol- should be eligible for BCT, the actual number
low-up periods, the standards of treatment of patients undergoing BCT is as low as 10 to
change. Current studies in breast cancer ther- 15 percent.11,12 There is a trend away from this
apy may be virtually obsolete before the data however. One study showed the number of
matures. Trials have been criticized for using patients treated with mastectomy for stage I
chemotherapy regimens that are no longer breast cancer to have decreased from 56 to 43
standard.5 The toxicity of radiotherapy is most percent from 1985 to 1995.8 The most common
often defined by late effects on normal tissue. medical contraindication to BCT is multicen-
These late effects may take years to develop tric disease involving more than one quadrant
and are related to technique. For example, the of the breast.13 Intraductal cancer presents with
risk of second malignancy may not be evalu- multicentricity in a higher percentage of
able for decades.6 patients than does invasive cancer. On the other
The epidemiology of breast cancer is chang- hand, 20 percent of patients eligible for BCT
ing. There was a large increase in incidence choose mastectomy.13
seen during the 1980s, and a constant increase How does this apply to the radiotherapeutic
of about 1 percent per year has been seen until treatment of breast cancer? As more early-
recently.7 This is undoubtedly because of the stage and noninvasive cancers are detected,
acceptance of screening mammography. In more patients will be eligible for breast con-
1979, 22 percent of women over the age of 40 servation therapy. There appears to be growing
years had had at least one mammographic acceptance of BCT as the standard treatment in
study; presently, 74 percent of women over the eligible patients. Optimizing individual treat-
age of forty have undergone at least one study. ment based on predictive indicators for local
The incidence of ductal carcinoma in situ recurrence may lead to subsets of patients who
(intraductal) breast cancer increased from 7.4 do not require radiotherapy after excision.
percent in 1985 to 14.3 percent in 1995,8 a Likewise, those at highest risk for local recur-
result attributable almost entirely to both rence may benefit from additional or alterna-
improvements in mammographic techniques tive treatment.
Breast Cancer and Radiation Therapy 221

EARLY-STAGE BREAST CANCER from patients with breast cancer treated with
conservative surgery and radiotherapy. It is
Mastectomy and Breast Conservation very safe to conclude that the data supports the
Therapy: Equivalent Survival use of breast conservation therapy for the treat-
ment of early-stage invasive breast cancer.
Several randomized clinical trials have con-
firmed equivalent survival data when lumpec- Risk Factors for Local Recurrence
tomy and breast radiotherapy are compared to
modified radical mastectomy for early-stage Among patients treated with breast conserva-
invasive breast cancer. Local recurrence rates tion therapy, several risk factors have been
after conservative surgery and radiotherapy identified that predict for local recurrence.
range from 3 to 19 percent (Table 13–1).14–20 It These have been classified as patient-related,
should be noted that the study from the NCI tumor-related, and treatment-related factors. If
had the highest rate of local recurrence but did a risk factor was associated with a prohibitively
not require negative pathologic resection mar- high recurrence rate, this would be a con-
gins, the significance of which is discussed traindication to BCT only if the same factor
below. This same study was the only trial to was not a risk factor for recurrence after mas-
show a significant advantage to mastectomy tectomy. For example, several series have
with respect to local recurrence. Despite a shown age of diagnosis to be a predictive indi-
broad range of entry criteria (acceptable tumor cator for local recurrence after BCT, with
size ranged from 2 to 5 cm), the data are younger patients being at higher risk.23–26 How-
remarkably consistent. The most important ever, one cannot conclude that young age is a
finding was the equivalent overall survival contraindication for BCT, because other studies
between treatment arms in all studies. Local have shown the same population of young
recurrence in the mastectomy-treated patients patients to be at higher risk for local recurrence
ranged from 4 to 14 percent. Equivalent sur- after mastectomy.27,28
vival between extensive surgery and limited
surgery plus radiotherapy was confirmed in a PATHOLOGIC VARIABLES
meta-analysis of randomized trials in breast
cancer. Also, a threefold reduction in local Table 13–2 lists several factors that are or at one
recurrence was demonstrated with the addition time were thought to be factors predictive for
of radiotherapy to local excision.21 local recurrence. Multicentricity in more than
There are large retrospective reviews corre- one quadrant of the breast is a contraindication
lating these results.22 These studies have also to BCT. Several authors have concluded that the
been useful in identifying prognostic features risk for local failure is significantly higher in

Table 13–1. BREAST CONSERVATION THERAPY COMPARED WITH MASTECTOMY: RANDOMIZED TRIALS

Local Recurrence (%) Survival (%)


Follow-up
Trial (years) Mastectomy BCT p Value Mastectomy BCT p Value

NSABP B-0614 12 8 10 ns 59 63 ns
NCI15,16 10 6 19 .01 75 77 ns
Milan17 18 4 7 ns 65 65 ns
EORTC18 14 14 17 ns 61 54 ns
Danish19 — 4 3 ns 82 79 ns
IGR20 15 14 9 ns 65 73 ns

NS = not significant.
222 BREAST CANCER

patients with documented disease in more than (larger size at presentation, higher hormonal
one quadrant of the breast.29–31 This likely indi- receptor positivity, decreased axillary involve-
cates the presence of residual tumor burden ment), there is no difference in local control in
throughout the breast after local excision that patients with infiltrating lobular histology
cannot be controlled with radiotherapy doses of treated with local excision and radiotherapy.38
45 to 50 Gy. Since mastectomy for multicentric Patients with centrally located tumors were
disease is not associated with an increased risk initially thought to be unsuitable for BCT due to
of local recurrence,32 it is the standard of care cosmetic concerns about tumors near the nipple-
for patients with defined multicentricity. areolar complex.30 There was also a concern that
Historically, it was thought that patients involvement of major breast ducts might be asso-
with an extensive intraductal component (EIC) ciated with more diffuse disease. However, recent
associated with their invasive tumor were at analysis of tumors within 2 cm of the nipple-
significant risk for recurrence.33,34 This condi- areolar complex show no increased risk of local
tion has been defined as intraductal carcinoma failure when treated with breast conservation
occupying ³ 25 percent of the area encom- therapy, provided negative resection margins are
passed by invasive tumor as well as the sur- achieved.39 Good to excellent cosmesis can be
rounding stroma of the resected specimen. achieved in this scenerio.40,41
However, when this risk factor was re-evaluated Patients with Paget’s disease of the nipple
in association with negative surgical resection were previously considered poor candidates for
margins, it was found that if negative margins BCT because of its significant association with
for both invasive and noninvasive components separate palpable infiltrating malignancies. For
could be obtained, no increase in local recur- patients with Paget’s disease and no palpable
rence was observed.35,36 malignancy, the concern was the possibility of
The presence of lobular carcinoma in situ microscopic invasive disease. This concern is
(LCIS) within the tumor specimen likewise has probably less valid today with newer mammo-
no impact on the probability of local recur- graphic technologies. Moreover, it has been
rence.37 However, these patients may be at shown that if patients presenting with Paget’s dis-
higher risk for development of a second subse- ease of the breast without an associated palpable
quent cancer in the contralateral breast. Infil- mass are treated with local excision and radio-
trating lobular histology behaves differently therapy, there is no increased risk of recurrence.42
than infiltrating ductal histology. Although the Tumor size and histologic grade have prog-
natural history of lobular carcinoma may differ nostic significance for survival, risk of axillary
involvement, and the development of distant
Table 13–2. RISK FACTORS FOR LOCAL RECURRENCE
metastatic disease. However, local control may
AFTER BREAST CONSERVATION THERAPY not be impacted by either the size or histologic
Current
grade of the primary lesion. Most retrospective
Risk Factor Literature Support studies find no significant difference in local
control for T1 or T2 tumors. Recent studies fail
Young age at diagnosis23–27 Y
Multicentricity29–32 Y to correlate higher grade tumors with increased
Extensive intraductal component35,36 N local recurrence.43
LCIS found in specimen37 N
Infiltrating lobular histology38 N
Subareolar location39–41 N PATIENT VARIABLES
Paget’s disease42 N
Pathologic grade43 N
Family history44,45 N Significant family history of breast cancer is not
Positive surgical margins47,51 Y a contraindication for BCT. There is no higher
LCIS = lobular carcinoma in situ. risk of local failure in patients with a history of
Breast Cancer and Radiation Therapy 223

breast cancer in a first-degree relative. In fact, Focally positive and close margins appear to
several studies have shown that patients with be associated with a risk of local recurrence
breast cancer who have a first-degree relative that is intermediate between diffusely positive
with the disease may have an increased overall and negative margins. In some studies, this
survival.44,45 This observation may correlate intermediate risk of recurrence is statistically
with evolving data showing a good prognosis significant, implying re-excision should be per-
with BRCA1 genetic overexpression.46 formed.50 However, other studies do not show
Young age at diagnosis is associated with an significance related to close or focally positive
increased local recurrence rate. The studies dif- margins.51 The presence of two or more positive
fer in the age at which risk of recurrence is sig- margins has been shown to be predictive of
nificantly higher, but all conclude young age to higher local failure compared to one.52 Of par-
be a risk factor. Because these same patients are ticular note, proper evaluation of resection mar-
at higher risk for local failure after mastectomy, gins is essential, as one-third of patients with
young age is not a contraindication to BCT.24–27 focally positive shaved margins are negative
In addition, there is no impact on overall sur- when inked margins are properly evaluated.
vival for younger patients treated with breast The amount of tumor at the margin of a sur-
preservation therapy. gical specimen correlates with the risk of dif-
fuse involvement in the breast.53 There is a
THERAPEUTIC VARIABLES strong correlation between the extent of
involvement of surgical margins and residual
Surgical Factors tumor burden in the subsequently re-excised
specimen. The risk of residual disease is a con-
The principal surgical factor correlated with tinuum based upon distance from the tumor,
increased local–regional recurrence after BCT with margin distance determined within the
is an inability to resect the lesion with clear context of cosmetic outcome.54 Standard doses
pathologic margins. Definition of a “clear mar- of radiotherapy are less likely to control a
gin” varies. While the absence of tumor at the breast with a significant microscopic tumor
inked margin of the resected specimen is burden. Although boost doses of radiotherapy
defined by some authors as a negative margin, to the surgical bed are believed to increase local
others have differentiated between negative and control, definitive evidence is meager. One
close margins by using 1 to 3 mm to define a study looked at the effect on local control with
negative margin. These distinctions notwith- both re-excision and boost doses of radiother-
standing, there appears to be a consistently sig- apy.55 There was a statistically significant
nificant increase in local recurrence with dif- increase in local control with re-excision for
fusely positive margins compared to “negative” close, indeterminate, or positive margins com-
margins. The recurrence rate for diffusely posi- pared to no re-excision. There was no differ-
tive margins approaches 30 percent, compared ence in local control for these same patients
to 2 to 10 percent for negative margins. Thus, when evaluated by total dose delivered to the
the presence of diffusely positive pathologic tumor site, indicating that re-excision for ques-
lumpectomy margins is an indication for re- tionable margin status was more important than
excision. If re-excision is not possible, mastec- boost irradiation. This supports the notion that
tomy should be performed. Surgical margins the best radiation in the world cannot compen-
classified as negative, close, focally positive, or sate for inadequate surgery. The presence of a
more than focally positive correlate with a 2, 3, focally positive margin appears to be signifi-
9, and 28 percent risk of local recurrence, cant and should also be re-excised. Although
respectively (Table 13–3).47–49 the significance of close resection margin prob-
224 BREAST CANCER

ably depends more on the underlying defini- patients with scleroderma exhibited prohibitive
tions, re-excision does not appear necessary. toxicity. Patients with other collagen vascular
Ideally, negative margins should be obtained to diseases did not experience prohibitive toxicity
optimize local control. and could be considered candidates for BCT.58
The anticipated cosmetic outcome after The potentially deleterious effects of radio-
lumpectomy is critical to the surgical selection therapy on the developing fetus prohibit patients
of patients for BCT. The pressure of a large in the first two trimesters of pregnancy from
tumor-to-breast ratio is likely to lead to a less being candidates for BCT. Estimated cumula-
satisfactory cosmetic outcome from the point tive fetal doses of 3 to 4 Gy are delivered with
of view of both patient and physician. Excision tangential radiotherapy.59 Because there is no
of a major proportion of breast tissue to obtain known threshold dose for mutagenesis, radio-
negative margins (usually greater than a quad- therapy is an absolute contraindication during
rant) is a relative contraindication to BCT. Cos- the first two trimesters. Long-term effects of
metic evaluation of the breast is very subjec- radiation during the third trimester are unclear.
tive, with many layers of complexity in terms of
self-esteem and sensuality. While studies com- Boost to Lumpectomy Site
paring lumpectomy to quadrantectomy have
shown better local control with quadrantectomy Two questions exist regarding the use of irradi-
followed by radiotherapy, in general lumpec- ation to the lumpectomy site in conjunction
tomy plus radiotherapy provides equivalent with whole breast irradiation. First, is a boost
local control, with better cosmesis from the necessary? Most centers treat the whole breast
point of view of both patient and physician.56,57 with fraction sizes of 180 cGy to a total dose of
45 to 50 Gy followed by a boost to the tumor
Radiation Therapy Factors site of an additional 10 to 15 Gy. Protocols
established by the National Surgical Adjuvant
Certain collagen vascular diseases, such as Breast Project (NSABP) routinely treat at
scleroderma and systemic lupus erythematosus, 200 cGy per day to a total dose of 50 Gy with
have been associated with an increase in acute no boost to the tumor site. The principal ratio-
skin and subcutaneous toxicity to standard nale for boost irradiation is that it can be deliv-
doses of radiotherapy and may be related to ered safely without major cosmetic detriment.
inadequate repair of sublethal radiation injury. In a large retrospective study, 17 percent of
However, a retrospective review of patients patients who did not receive a boost showed
with collagen vascular disease showed that only local failure, compared to 11 percent for those

Table 13–3. MARGINS OF EXCISION CORRELATED TO RATE OF LOCAL RECURRENCE

Margin Status (%)

Study Negative Positive Close Indeterminate Focally More than


involved Focally involved

Anscher et al47 10 2 10
Gage et al51 2 16 3 9 28
Heimann et al49 2 11
Ryoo* et al48 6 13 8
Smitt et al63 2 18†
Solin* et al62 10 8 14 13
Spivack et al50 3.7 18

*Boost radiation delivered based on margin status.



Combined data on close or positive margins.
Breast Cancer and Radiation Therapy 225

who did.60 On the other hand, another study local control. Three randomized trials evaluat-
randomizing patients to a boost after whole ing the addition of tamoxifen to BCT have
breast radiation versus no boost found shown local control and event-free survival to
increased local control in boosted patients.61 be significantly improved in tamoxifen-treated
However, patients treated with a boost had a patients. Tamoxifen reduces ipsilateral and con-
worse cosmetic outcome, with a statistically tralateral recurrences.66–69 There is currently a
significant increase in telangiectasia formation. randomized trial is underway to evaluate the
Unfortunately, the dose per fraction in the study omission of breast radiation in elderly women
was 250 cGy for both the whole breast and with estrogen-receptor positive tumors treated
boost portions of treatment—this represents a with excision and tamoxifen.
higher fractional dose than is routinely used in
the United States. Cytologic Factors and Local Recurrence
The second question is whether an increased
radiation boost can compensate for close or New cytologic and genetic factors are being
positive resection margins. The data on this identified and associated with breast cancer
show mixed results. Retrospective data are prognosis. The majority of these studies to date
often confounded by the routine use of an are small retrospective series and have not
increased boost dose to patients with close mar- influenced the choice of therapy. Case control
gins. On the one hand, there are studies show- studies looking at the overexpression of
ing that the increasing dose used to boost close insulin-like growth factor-I receptor (IGF-IR)
margins increased local control.62 On the other and HER-2/neu indicate that overexpression
hand, studies demonstrate that patients with may predict for an increased risk of local recur-
positive margins have higher local recurrence rence.70,71 In these studies, the tumor tissue of
rates with or without a boost.63 The issue may all patients who had experienced local recur-
be resolved by a current EORTC study ran- rence at a single institution were evaluated for
domizing patients with inadequate surgical overexpression of these newly described mark-
margins to boost doses of 10 and 25 Gy. ers. Case controls were drawn from those
patients treated who did not experience local
Systemic Therapy Factors recurrence. Overexpression of IGF-IR and
HER-2/neu was found significantly more fre-
Two conclusions can be gleaned from data on the quently in patients who experienced recurrence.
addition of chemotherapy to BCT. Chemotherapy Although HER-2/neu overexpression negatively
added to lumpectomy and radiotherapy in impacted disease-free survival in patients
patients at high risk for development of metasta- treated with tamoxifen and radiotherapy,72 this
tic disease reduces the risk of ipsilateral breast effect was not seen in patients treated with
recurrence to as low as 2.6 percent.64 On the chemotherapy.
other hand, chemotherapy cannot substitute for The impact on local control, however, is
radiotherapy in BCT. Local recurrence in patients unclear. Several studies show that patients with
treated with local excision and chemotherapy has the germ-line mutations BRCA1 or BRCA2 may
been shown to be significantly higher than that experience statistically significant improvement
for standard lumpectomy and radiotherapy.65 in survival.73 Overexpression of p53 has been
associated with poor response to tamoxifen, but
Tamoxifen and Local Control no data exist on its role with radiotherapy or
local control.74 The “tumor suppressor” gene
Tamoxifen benefits estrogen-receptor positive p53 is thought to confer radioresistance by a loss
patients both in terms of overall survival and of apoptosis in response to radiation. The possi-
226 BREAST CANCER

bility of radioresistance has been raised in a pre- prognostic features, since these patients may
liminary study of patients treated with BCT or benefit least from the addition of radiother-
mastectomy and postoperative radiotherapy.75 apy.82,83 The two favorable prognostic factors
Finally, the presence of angiogenesis as mea- identified were advancing age of the patient
sured by microvessel count has been shown to be and small tumor size. In patients with T1
prognostically significant for survival. Those tumors and advancing ages, the local recur-
patients with node-negative cancers and a low rence risk ranged from 4 to 16 percent. Of note,
mean vessel count (MVC) had excellent overall the Uppsala study compared patients > 50 years
survival, but the impact on local recurrence was old with T1 tumors to all others and found a
not indicated.76 However, laboratory studies recurrence rate of “only” 15.9 percent.84 How-
have shown a synergistic effect between angio- ever, such a local recurrence is probably unac-
genesis inhibitors and radiotherapy.77 ceptable since the addition of radiation likely
reduces the risk of recurrence to < 5 percent. Of
Lumpectomy Alone note, some studies were unable to identify sub-
sets of patients who did not benefit from radio-
The NSABP B-06 trial determined that the risk therapy.85 As indicated above, there is currently
of local recurrence after local excision alone a prospective trial for patients > 70 years old
was as high as 35 percent, compared to 10 per- with T1 tumors evaluating the omission of radi-
cent when combined with radiotherapy. An ation in patients receiving tamoxifen, based on
alternative view of this data is that 65 percent the subset analysis of the prior studies and
of patients will not experience local recurrence knowledge of the impact of tamoxifen on local
after lumpectomy alone. Therefore, the major- control. Outside of a trial setting, the omission
ity of patients treated with radiotherapy would of radiation therapy from breast conserving
not have local recurrence in the absence of radi- therapy is not presently indicated. Interestingly,
ation. Attempts have been made to identify sub- practice patterns in the treatment of patients
sets of patients with early stage breast cancer > 65 years old have been shown to differ from
who do not require radiotherapy. Six studies younger patients, with this population more
listed in Table 13–4 prospectively randomized likely to be treated with local excision and no
patients to excision versus excision plus radio- radiotherapy.86,87 The ongoing study may justify
therapy.78–81 Although all studies have shown a these practice patterns.
significant increase in local recurrence in the
patients who did not receive radiation, none DUCTAL CARCINOMA IN SITU
showed a statistically significant survival
advantage with radiotherapy. The increasing acceptance of screening mam-
The stress of tumor recurrence on the mography has led to a dramatic increase in the
patient as well as the low morbidity of tangen- incidence of ductal carcinoma in situ (DCIS),
tial breast radiation must be considered when or intraductal carcinoma. Historically, patho-
evaluating these results. Although no survival logic evaluation of intraductal carcinoma
advantage was documented, local recurrence showed focality in the majority of cases. How-
may be predictive of subsequent increased mor- ever, Holland showed intraductal foci of intra-
tality. Also, a significant number of patients ductal disease remote from the primary site in
who experience recurrence after local excision 40 percent of patients.88 Thus, the rationale for
alone choose mastectomy at the time of recur- radiotherapy to the remaining breast following
rence, regardless of survival data. local excision appears appropriate for the
Within these studies, subset analyses were majority of patients with intraductal cancer.
performed to identify patients with favorable There was a subset of patients on NSABP B-06
Breast Cancer and Radiation Therapy 227

Table 13–4. COMPARISON OF LUMPECTOMY VERSUS LUMPECTOMY PLUS RADIOTHERAPY: RANDOMIZED TRIALS

Local Recurrence (%) Survival (%)


Follow-up
Trial (years) No XRT XRT p Value No XRT XRT p Value

NSABP B-0614 12 35 10 + 58 62 ns
Scottish trial69 5 28 6 + 85 88 ns
Ontario78 8 35 11 + 90 91 ns
Milan79,80 5 18 2 + 92 92 ns
Uppsala81 5 18 2 + 90 91 ns

XRT = external beam radiation therapy.

identified as having in situ histology. The local an entry criteria, a minimum of 3 mm negative
recurrence rate in patients undergoing excision surgical margins are required.
without radiotherapy was 43 percent, versus 7 The impact of local recurrence for DCIS
percent in patients receiving radiation.89,90 does not appear to have an impact on sur-
The largest prospective study of DCIS is the vival.99,100 However, the potentially devastating
NSABP randomized study B-17 comparing emotional trauma the patient experiences with
local excision alone with local excision plus recurrence should be balanced against the very
radiotherapy.91,92 There was a significant low risk of radiation complications. Decreased
decrease in local recurrence in the radiation mortality from breast cancer is likely the result
arm but no difference in overall survival. The of earlier detection of earlier-stage disease, par-
addition of radiation reduced the noninvasive ticularly DCIS. The reported 75 percent reduc-
cancer recurrence from 10.4 to 7.5 percent. tion in subsequent development of invasive
Invasive cancer recurrences were reduced from breast cancer for patients with DCIS treated
10.5 to 2.9 percent. Importantly, of those with with postexcisional radiotherapy supports this
recurrence in the local excision-only arm, half conclusion.
the recurrences were invasive. Other retrospec-
tive series confirm these findings.93–95 Thus, RADIOTHERAPY FOR LOCALLY
the standard of care for patients with DCIS who ADVANCED BREAST CANCER
are eligible for breast sparing treatment is local
excision followed by radiotherapy. Early studies of local-regional radiotherapy fol-
Several centers have attempted to identify lowing mastectomy for locally advanced dis-
subgroups of patients with an extremely low ease showed reduced recurrences in the axilla,
risk for recurrence after local excision. Tumor supraclavicular fossa, and chest wall. However,
grade, particularly the presence of comedocar- these patients experienced an increase in non-
cinoma, is a significant predictor of local recur- breast cancer-related mortality that negated the
rence. The Van Nuys Prognostic Index (VNPI) survival advantage.101,3
has been proposed, using 155 patients treated Cardiac toxicity due to chest wall radiother-
with local excision alone.96,97 There is a score apy is purported to be the causative factor in the
given to each of three factors: tumor grade, size increase in nonbreast cancer-related mortality.
of disease, and extent of negative surgical mar- Concerns about cardiac morbidity from radio-
gins. The result of a low VNPI appears to be therapy in the face of potentially cardiotoxic
predictive for decreased local recurrence.98 adriamycin-based chemotherapy led to decreased
There is presently a single-arm nonrandomized use of postmastectomy radiotherapy. Technical
study underway evaluating low-risk patients for factors in historically quoted studies may
local recurrence after local excision alone. As explain the high incidence of radiation cardiac
228 BREAST CANCER

toxicity. The principle factor is the use of an en for locally advanced breast cancer. Studies have
face internal mammary and supraclavicular looked at recurrence patterns in patients treated
portal, often referred to as a “hockey stick” with mastectomy alone.108 Results have shown
port. Other considerations are related to that patients with four or more nodes positive,
dosimetry, such as the use of orthovoltage large primary tumors > 5 cm, pectoral fascia, or
energy radiotherapy. The energy delivered with skin involvement are at greatest risk of local
orthovoltage radiation is lower in energy, less recurrence after mastectomy, as high as 25 to
penetrating, therefore leading to greater inho- 30 percent. Chest wall radiotherapy in these
mogeneity of dose. Comparative dosimetric patients reduces local recurrence to < 10 per-
analysis has shown the dose to the heart to be cent. Recurrence patterns show the chest wall
significantly higher with orthovoltage than and supraclavicular fossa to be the most com-
with modern megavoltage radiotherapy.102,103 mon sites of recurrence, with axillary recur-
When patients treated with orthovoltage rence seen less frequently. Patients with tumors
radiotherapy were excluded from analysis, a sur- < 2 cm and negative axillary lymph nodes have
vival benefit of approximately 10 percent was local recurrence risk of < 10 percent and would
seen in the patients receiving chest wall radio- not appear to benefit from radiotherapy.109
therapy.104 Recently, two prospective random- Patients with tumors between two and five cen-
ized studies reported results showing survival timeters or one to three involved axillary lymph
advantage from the addition of local–regional nodes are at intermediate risk, the role of radio-
radiotherapy after mastectomy and chemother- therapy is controversial in these patients
apy for node-positive premenopausal breast can- because of modest potential benefit.110
cer patients. Both studies show an improvement Extracapsular extension (ECE) of tumor in
in survival of 8 to 10 percent.105,106 Both studies axillary lymph nodes is a potential risk factor
have shown a benefit to radiating all patients, for regional recurrence. It is likely to be associ-
regardless of the number of lymph nodes ated with other poor prognostic indicators, such
involved. Although patients with one to three as multiple positive axillary nodes. While
nodes positive and four or more nodes positive shown to be significant on univariate analysis,
both showed benefit from radiation in subgroup
analysis, routine regional lymph node radiother- Table 13–5. POSTOPERATIVE CHEST WALL
apy in all patients with positive nodes is not gen- RADIOTHERAPY FOLLOWING MASTECTOMY
erally accepted practice (Table 13–5). Treatment indicated108–110
There has been criticism of the studies for Tumor greater than 5 cm or
Positive surgical margins or
inadequate evaluation of the axilla. Many patients Four or more positive axillary lymph nodes
had small numbers of axillary lymph nodes
Treatment to be considered112
excised. The patient with only one positive node Premenopausal patients with one to three positive nodes
excised may have had other involved nodes in the Inclusion of internal mammary nodes controversial
undissected axilla. Thus, a proportion of the Treatment not indicated
patients thought to have one to three nodes posi- All patients with
Tumor less than five centimeters and
tive may have actually had four or more involved Negative margins and
nodes. Prior studies have shown this population No positive lymph nodes
Postmenopausal patients with
of patients to be at lower risk for local–regional One to three positive lymph nodes
recurrence. There was also criticism that the
Treatment not indicated based on recent literature113
local recurrence rates in these studies were inor- Patients with extracapsular extension of lymph node
dinately high compared to prior studies.107 involvement
Prior retrospective studies have evaluated Treatment guidelines differ
the role of radiotherapy following mastectomy Male patients with breast cancer
Breast Cancer and Radiation Therapy 229

ECE was not found to predict for regional fail- surgery had a higher local recurrence rate than
ure on multivariate analysis.111,112 those with partial response, local excision, and
The recently published trials showing a sur- subsequent radiotherapy.118 Residual disease,
vival benefit to radiotherapy after mastectomy although not clinically detectable, may still be
included the internal mammary nodes. The argu- extensive in these patients. In a prospective
ment for treatment is that persistent microscopic evaluation of mastectomy specimens in patients
involvement after chemotherapy is still poten- who had a complete clinical response to neoad-
tially curable with regional radiotherapy. Theoret- juvant chemotherapy and radiation therapy,
ically, untreated microscopically involved nodes residual disease ranging from 0.6 to 6.5 cm was
may lead to development of distant disease. Cur- confirmed in all specimens.119
rently, there is an EORTC study underway evalu- What is the role of postmastectomy radiother-
ating the efficacy of treating internal mammary apy in patients who can not undergo BCT after
nodes; efficacy and toxicity data, however, will chemotherapy? Clinical staging often under-
not be available for 15 or more years. estimates the extent of disease when compared to
Detecting involved internal mammary pathologic staging. Neoadjuvant therapy only
nodes is difficult. The region is not clinically confounds the issue. Should regional lymphatic
evaluable as are the axilla or supraclavicular irradiation be used in all patients with locally
fossa. Computed tomography scanning can advanced disease? There is currently an inter-
detect grossly enlarged nodes but not micro- group trial evaluating neoadjuvant therapy that
scopic disease in normal-sized nodes. Sentinel prohibits chest wall radiotherapy after mastec-
lymph node mapping, on the other hand, may tomy, regardless of pathologic findings. In light
resolve this issue.113 of the recent data showing a survival advantage
in selected patients treated with postmastectomy
MULTIMODALITY THERAPY radiotherapy, this approach is difficult to justify.
FOR LOCALLY ADVANCED The sequencing of chemotherapy and radio-
BREAST CANCER therapy in all stages has been studied in detail.
Current practice focuses on systemic therapy as
Neoadjuvant chemotherapy is the subject of the top therapeutic priority since it has the great-
intense clinical investigation for tumor down- est impact on survival. Concurrent chemother-
staging before surgery. Patients who otherwise apy and radiotherapy are prohibitively toxic.
would not be candidates for breast conservation Although there are data showing a detrimental
therapy may experience a significant reduction impact on local control from delaying radiother-
in tumor burden after neoadjuvant therapy apy more than 6 months after surgery,120 a ran-
allowing for local excision with negative mar- domized trial comparing upfront chemotherapy
gins. Response rates are reported to be approx- followed by radiation versus upfront radiother-
imately 65 to 75 percent.114–116 Studies have apy followed by chemotherapy concluded that
shown no detriment to overall survival when delaying chemotherapy increased distant metas-
compared to standard postoperative treatment. tases and adversely affected survival. Although
Complete clinical response may define a more delayed radiotherapy increases local failure, a
favorable subgroup than a partial response significant percentage of patients experiencing
although this has not been universally noted.117 local recurrence can be salvaged.121
What is the optimal therapy for women
experiencing a complete response? In one TECHNIQUES OF RADIOTHERAPY
study, those patients who had a complete clini-
cal response to chemotherapy who then Breast irradiation after local excision is gener-
received radiotherapy to the breast without ally administered with megavoltage photon
230 BREAST CANCER

energies of 4 to 10 MV to deliver 45 to 50.4 Gy tions to maximize homogeneity of dose


to the whole breast at 180 cGy per fraction. Tan- throughout the breast tissue.
gential portals are established from midline to
midaxilla using wedge filters and a half beam RADIOTHERAPY COMPLICATIONS
block or independent jaws to minimize pul-
monary radiation. Many centers deliver a boost Dosimetric analyses of historic treatment tech-
to the tumor bed using electron beam radiation niques show an average of 25 percent of the car-
for an additional 900 to 1500 cGy. Alternatively, diac volume received at least 50 percent of the
NSABP protocols prescribe 5000 cGy at 200 prescribed dose. With modern treatment ener-
cGy per fraction, with an optional boost. Superi- gies, 5.7 percent of the cardiac volume receives
orly, the tail of the breast is encompassed while this same dose.124 Serum troponin measure-
excluding the humoral head. Inferiorly, a reason- ments during radiotherapy, a measure of cardiac
able margin of 1.5 to 2.0 cm below the infra- injury, revealed no significant elevation in one
mammary fold is standard. The chest wall cur- study.125 Patients treated for left-sided tumors
vature necessitates treatment of a small volume who also had internal mammary nodes treated
of lung tissue. If the maximum width of lung tis- with modern techniques were evaluated after
sue treated is < 2 cm, the risk of pulmonary ten years for possible late effects to the heart.
injury is exceptionally low (Figure 13–1).122 Thallium stress tests revealed no statistical evi-
The boost treatment volume generally dence of increased abnormality when compared
encompasses the surgical bed with margins. to the general population.126 No increased rate
Some centers advocate the use of clips at the of myocardial infarction or cardiac-related
time of surgery to outline the surgical bed for deaths was seen in retrospective reviews of
boost treatment.123 Electron energies of 6 to patients treated with tangential radiation.127,128
15 MeV are used, depending on the depth Other reported complications from breast
needed to encompass the tumor bed. The use of radiation include lymphedema, rib fracture,
interstitial implants for the boost treatment has brachial plexopathy, pulmonary fibrosis, car-
been supplanted in recent years. cinogenesis, and contralateral breast cancer.
Three-dimensional treatment planning is a Standard surgical therapy for invasive breast
major development in the delivery of radiother- cancer includes level I and II axillary dissection,
apy. As the contour of the breast is irregular, which has a finite risk of arm edema of 10 to 15
there is potential for significant inhomogeneity percent.129,130 The addition of radiotherapy may
of dose. Using CT scanning of the treatment increase the risk to as high as 20 percent. The
volumes, technology exists to determine dose measured incidence of arm edema likely differs
delivery in three dimensions with modifica- from the incidence of clinically significant arm
edema. One study showed a direct relationship
between the incidence of arm edema and the
number of lymph nodes dissected.131 There is
less risk of lymphedema in a level I and level II
selective dissection than in a full axillary dis-
section. It is hoped that sentinel node biopsy
will eliminate this complication for patients
with histologically negative nodes. Any risk of
lymphedema after sentinel node biopsy has yet
to be determined. Brachial plexopathy is a com-
plication in patients receiving supraclavicular
Figure 13–1. Cross-sectional representation of tangential
radiation portals for breast conservation therapy. radiation. The delivered dose is the significant
Breast Cancer and Radiation Therapy 231

risk factor as the incidence is reported to be < 2 than is the general population. Whether radio-
percent if 50 Gy is delivered, versus 5 percent if therapy further increases that risk is the subject
> 50 Gy is given.122 Rib fracture possibly related of debate. Current technology virtually elimi-
to beam energy has been reported after breast nates dose to the contralateral breast with tan-
and chest wall radiotherapy. The incidence is gential radiotherapy. However, the half-beam
very low (< 0.5 percent), but 4 MV photons may block technique described earlier allowed for
be associated with a higher incidence (2.2 up to 400 to 500 cGy to be delivered to the con-
percent) than are higher energy beams. 122 tralateral breast. Hazard analysis of historical
The chief rationale for the use of BCT over series revealed a small increased risk in irradi-
mastectomy is cosmesis. Poor cosmesis should ated patients compared to patients treated with
be considered a complication of treatment. In surgery alone.139
general, 75 to 85 percent of patients and treating
physicians report good to excellent cosmesis LOCAL RECURRENCE
after lumpectomy and radiotherapy. Factors that
may affect cosmesis include delivery of concur- Local recurrence has been shown to predict for
rent chemotherapy, type and extent of surgery, subsequent distant metastases, having an
and the dose of radiotherapy delivered.132,133 impact on survival.140 These data must be con-
Two series have shown that radiotherapy sidered in “very low risk” patients evaluated for
delivered after breast augmentation can lead to omission of radiotherapy. Salvage treatment
capsular contracture in 30 percent of patients. with mastectomy after ipsilateral breast recur-
Although radiation following breast recon- rence in patients treated with BCT offers sub-
struction is very well tolerated,134,135 the sequent disease-free survival in 40 to 70 per-
reported contracture rate for the general popu- cent of affected patients. Axillary recurrences
lation is 5 to 10 percent. Subcutaneous implan- are rare but subsequent survival is lower (25 to
tation is associated with a higher risk of con- 30 percent).141 Failure in the supraclavicular
tracture than are subpectoral implants. fossa has a dismal prognosis.142 Short interval
Postmastectomy radiotherapy after transverse to recurrence is likely to be predictive of subse-
rectus abdominus myocutaneous (TRAM) flap quent distant disease.143
breast reconstruction can be delivered effec- There is a question whether development of
tively without cosmetic compromise.136 cancer in a treated breast many years after BCT
The carcinogenic potential of ionizing represents relapse or a new primary cancer. It
radiotherapy is rare and the latent period long. has been postulated that the location of the
Follow-up intervals > 20 years are usually recurrence in relation to the primary, the time
necessary to see carcinogenic effects. Most interval between incidents, and the presence or
large retrospective series report scattered absence of diffuse disease in the surrounding
cases of sarcoma developing in the treated stroma may distinguish between recurrence and
field years after treatment. Lymphangiosar- a new primary. The presence of a lesion near
coma has been reported in patients who expe- the original tumor site with disease in the sur-
rience significant lymphedema in the treated rounding stroma presenting relatively shortly
breast or ipsilateral arm. Osteosarcomas are after initial diagnosis more likely represents
found to be most common within the treated recurrent disease than does a remote lesion pre-
field, with an overall incidence of sarcoma senting in normal surrounding tissue after a
development of 0.2 percent and a median long interval.144 Differences in DNA content
latency of 11 years.137,138 detectable by flow cytometry are more likely in
Patients with breast cancer are at higher risk a second primary malignancy than in a recur-
for developing a contralateral breast cancer rent tumor. The subsequent treatment of the dis-
232 BREAST CANCER

ease may differ, particularly in patients exposed REFERENCES


to systemic treatment. 1. Fletcher GH. Local results of irradiation in the
Standard therapy for patients experiencing primary management of localized breast can-
recurrence in an irradiated breast is mastectomy. cer. Cancer 1972;29:545–51.
However, there are limited data on the retreat- 2. National Institutes of Health Consensus Develop-
ment Panel. Consensus statement: treatment of
ment of patients with further excision and reirra-
early-stage breast cancer. J Natl Cancer Inst
diation; 62 percent of patients were salvaged and Monogr 1992;11:15.
treatment was well tolerated, as 15 out of 16 3. Cuzick J, Stewart H, Peto R, et al. Overview of
patients received 5000 cGy with no reported randomized trials of adjuvant radiotherapy in
deleterious effect.145 breast cancer. Recent Results Cancer Res
The majority of patients who experience 1988;111:109–29.
4. Cuzick J, Stewart H, Rutqvist L, et al. Cause-spe-
recurrence after local excision and no radio- cific mortality in long-term survivors of breast
therapy are treated with and often choose mas- cancer who participated in trials of radiother-
tectomy over re-excision and radiotherapy as apy. J Clin Oncol 1994;12:447–53.
salvage treatment. If the overall goal of breast 5. Recht A, Bartelink H, Fourquet A, et al. Postmas-
conserving treatment is breast retention, ther- tectomy radiotherapy: questions for the twenty-
first century. J Clin Oncol 1998;16:2886–9.
apy should optimize the patient’s wish, even at 6. Lipsztein R, Dalton JF, Bloomer WD. Sequelae of
the expense of potential over-treatment. breast irradiation. JAMA 1985;253:3582–4.
7. Garfinkel L, Boring C, Heath C Jr. Changing
CONCLUSION trends: an overview of breast cancer incidence
Breast conservation therapy consisting of local and mortality. Cancer 1994;74 Suppl:222–7.
8. Bland KI, Menck HR, Scott-Connor CE, et al. The
excision and breast radiotherapy should be the National Cancer Data Base 10-year survey of
treatment of choice for the majority of patients breast cancer treatment at hospitals in the
with early invasive and in situ carcinoma. United States. Cancer 1998;83:1263–73.
Radiotherapy to the breast has been shown to be 9. Bjurstrum N, Bjorneld L, Duffy SW, et al. The
effective in improving local and regional con- Gothenberg Breast Screening Trial. First results
on mortality, incidence, and mode of detection
trol, with minimal complications. The incidence
for women 39–49. Cancer 1997;80:2091–9.
of intraductal and early stage breast cancer may 10. Garne J, Aspegren K, Balldin G, Ranstam J.
continue to increase as mammographically Increasing incidence of and declining mortality
detected lesions become more predominant. from breast cancer. Cancer 1997;79:69–74.
Radiotherapy delivery may become more selec- 11. Nattinger AB, Gottleib MS, Hoffman RG, et al.
tive as methods are improved to identify Minimal increase in the use of breast-conserv-
ing surgery from 1986 to 1990. Med Care,
patients at high and low risk for local recur- 1996;34:479–89.
rence. However, with the very small risk of 12. Kotwall CA, Covington DL, Rutledge R, et al.
complication from treatment, the number of Patient, hospital, and surgeon factors associ-
patients not receiving radiotherapy should ated with breast conserving surgery. A
remain very small. Recent data regarding post- statewide analysis in North Carolina. Ann Surg
1996;224:419–26.
mastectomy radiotherapy indicate that sub-
13. Morrow M, Bucci C, Rademaker A. Medical con-
groups of patients with locally advanced disease traindications are not a major factor in the
achieve a survival benefit from the addition of under utilization of breast conserving therapy.
postmastectomy radiation. Modern radiotherapy J Am Coll Surg 1998;186:269–74.
techniques minimize cardiac injury. Identifying 14. Fisher B, Anderson S, Redmond CK. Reanalysis
patients with significant risk for local failure, and results after 12 years of follow-up in a ran-
domized clinical trial comparing total mastec-
optimizing local treatment in these patients, and tomy with lumpectomy with or without irradi-
evaluating the impact on overall cure are areas ation in the treatment of breast cancer. N Engl
for current and future investigation. J Med 1995;333:1456–61.
Breast Cancer and Radiation Therapy 233

15. Lichter AS, Lippmann ME, Danforth DN Jr, et al. relapse rates after undergoing breast conserva-
Mastectomy versus breast conserving therapy tion therapy. J Am Coll Surg 1998;187:1–8.
in the treatment of stage I and II carcinoma of 27. Matthews RH, McNeese MD, Montague ED,
the breast: a randomized trial at the National Oswald MJ. Prognostic implications of age in
Cancer Institute. J Clin Oncol 1992;10:976–83. breast cancer patients treated with tumorec-
16. Jacobson JA, Danforth DN, Cowan KH, et al. Ten- tomy and irradiation or with mastectomy. Int J
year results of a comparison of conservation with Radiat Oncol Biol Phys 14:659–63.
mastectomy in the treatment of stage I and II 28. Donegan WL, Perez-Mesa CM, Watson FR. A bio-
breast cancer. N Engl J Med 1995;332:901–11. statistical study of locally recurrent breast car-
17. Veronesi U Luini A, Galimberti V, Zurrida S. Con- cinoma. Surg Gyn Obstet 1966;122:329.
servation approaches for the management of 29. Fowble B. Radiotherapeutic considerations in the
stage I/II carcinoma of the breast: Milan Cancer treatment of primary breast cancer. J Natl Can-
Institute Trials. World J Surg 1994;18:70–5. cer Inst Monogr 1992;11:49–58.
18. Van Dongen JA, Bartelink H, Fentiman IS, et al. 30. Recht A, Harris JR. Selection of patients with early
Randomized clinical trial to assess the value of stage breast cancer for conservative surgery
breast-conserving therapy in stage I and II and radiation. Oncology 1990;4:23–30.
breast cancer: EORTC 10801 trial. J Natl Can- 31. Leopold K, Recht A, Schnitt SJ, et al. Results of
cer Inst 1992;11:15–8. conservative surgery and radiation therapy for
19. Blichert-Toft M, Rose C, Anderson JA, et al. Dan- multiple synchronous cancers of one breast. Int
ish randomized trial comparing breast conser- J Radiat Oncol Biol Phys 1989;16:11–16.
vation therapy with mastectomy: six years of 32. Fowble B, Yeh I, Schultz DJ, et al. The role of mas-
life table analysis. J Natl Cancer Inst Monogr tectomy in patients with stage I–II breast can-
1992;11:19–25. cer presenting with gross multifocal or multi-
20. Arriagada R, Le MG, Rochard F, Contesso G. Con- centric disease or diffuse calcifications. Int J
servative treatment versus mastectomy in early Radiat Oncol Biol Phys 1993;27:567–73.
breast cancer: Patterns of failure with 15 years of 33. Kurtz J. Factors influencing the risk of local recur-
follow-up data. Institut Gustave-Roussy Breast rence in the breast. Eur J Cancer 1992;28:660–6.
Cancer Group. J Clin Oncol 1996;14:1558–64. 34. Schnitt SJ, Connolly J, Khettry V, et al. Pathologic
21. Early breast cancer trialists’ Collaborative Group. findings on reexcision of the primary site in
Effects of radiotherapy and surgery in early breast cancer patients considered for treatment
breast cancer. N Engl J Med 1995;333:1444–55. by primary radiation therapy. Cancer 1997;59:
22. Winchester DJ, Menck HR, Winchester DP. The 675–81.
National Cancer Data Base report on the 35. Hurd TC, Sneige N, Allen PK, et al. Impact of
results of a large non-randomized comparison extensive intraductal component on recurrence
of breast preservation and modified radical in patients with stage I or II breast cancer
mastectomy. Cancer 1997;80:162–7. treated with breast conservation therapy. Ann
23. Cowen D, Jacquemier J, Houvenaeghel G, et al. Surg Oncol 1997;4:119–24.
Local and distant recurrence after conservative 36. Schnitt SJ, Abner A, Gelman R, et al. The rela-
management of “very low risk” breast cancer tionship between microscopic margins of
are dependant events: a 10-year follow-up. Int resection and the risk of local recurrence in
J Radiat Oncol Biol Phys 1998;41: 801–7. patients with breast cancer treated with breast-
24. Elkhuizen PH, Van de Vijver MJ, Hermans J, et al. conserving surgery and radiation therapy. Can-
Local recurrence after breast conserving ther- cer 1994;74:1746–51.
apy for invasive cancer; high incidence in 37. Moran M, Haffty BG. Lobular carcinoma in situ
young patients and association with poor sur- as a component of breast cancer: the long-term
vival. Int J Radiat Oncol Biol Phys 1998;40: outcome in patients treated with breast-con-
859–67. serving therapy. Int J Radiat Oncol Biol Phys
25. Fowble BL, Schultz DJ, Overmoyer B, et al. The 1998;40:353–8.
influence of young age on outcome in early 38. Schnitt SJ, Connolly JL, Recht A, et al. Influence
stage breast cancer. Int J Radiat Oncol Biol of infiltrating lobular histology on local tumor
Phys 1994;30:23–33. control in breast cancer patients treated with
26. Kim SH, Simkovich-Heerdt A, Tran KN, et al. conservative surgery and radiotherapy. Cancer
Women 35 years of age have higher locoregional 1989;64:448–54.
234 BREAST CANCER

39. Haffty BG, Wilson LD, Smith R, et al. Subareolar 53. Guidi AJ, Connolly JL, Harris JR, Schnitt SJ. The
breast cancer: long-term results with conserva- relationship between shaved margin and inked
tive surgery and radiation therapy. Int J Radiat margin status in breast excision specimens.
Oncol Biol Phys 1995;33:53–7. Cancer 1997;9:1568–73.
40. Bussieres E, Guyon F, Thomas L, et al. Conserva- 54. Holland R, Veling SH, Mravunac M, Hendriks JH.
tion treatment in subareolar breast cancers. Eur Histologic multifocality of Tis, T1-2 breast car-
J Surg Oncol 1996;22:267–70. cinomas. Implications for clinical trials of
41. Dale PS, Giulliano AE. Nipple-areolar preservation breast conserving surgery. Cancer 1985:56:
during breast-conserving therapy for subareolar 979–90.
breast carcinomas. Arch Surg 1996;131:430–3. 55. Wazer DE, Schmidt-Ullrich RK, Ruthazer R, et
42. Pierce LJ, Haffty BG, Solin LJ, et al. The conserva- al. Factors determining outcome for breast-
tive management of Paget’s disease of the breast conserving irradiation with margin-directed
with radiotherapy. Cancer 1997;80:1065–72. dose escalation to the tumor bed. Int J Radiat
43. Nixon AJ, Schnitt SJ, Gelman R, et al. Relation- Oncol Biol Phys 1998;40:851–8.
ship of tumor grade to other pathologic features 56. Fagundes MA, Fagundes HM, Brito CS, et al.
and to treatmetn outcome of patients with early Breast-conserving surgery and definitive radi-
stage breast cancer. Cancer 1996;78:1426–31 ation: a comparison between quadrantectomy
44. Chabner E, Nixon A, Gelman R, et al. Family his- and local excision with special focus on loco-
tory and treatment outcome in young women regional control and cosmesis. Int J Radiat
after breast-conserving surgery and radiation Oncol Biol Phys 1993;27:553–60.
therapy for early-stage breast cancer. J Clin 57. Veronesi U, Volterrani F, Luini A, et al. Quadran-
Oncol 1998;16:2045–51. tectomy versus lumpectomy for small breast
45. Malone KE, Daling JR, Weiss NS, et al. Family his- cancer. Eur J Cancer 1990;26:671–3.
tory and survival of young women with invasive 58. Ross J, Hussey D, Mayr N, Davis CS. Acute and
breast carcinoma. Cancer 1996;78:1417–25. late reactions to radiation therapy in patients
46. Porter D, Cohen B, Wallace M, et al. Breast can- with collagen vascular diseases. Cancer 1993;
cer incidence, penetrance and survival in prob- 71:3744–52.
able carriers of BRCA1 gene mutation in fam- 59. Antypas C, Sandilos P, Kouvaris J, et al. Fetal dose
ilies linked to BRCA1 on chromosome during breast cancer radiotherapy. Int J Radiat
17q12-21. Br J Surg 1994;81:1512–15. Oncol Biol Phys 1998;40:995–9.
47. Anscher MS, Jones P, Prosnitz LR, et al. Local fail- 60. Clark RM, Wilkinson RH, Miceli PN, MacDonald
ure and margin status in early stage breast carci- WD. Breast cancer: experiences with conserva-
noma treated with conservative surgery and tion therapy. Am J Clin Oncol 1987;10:461–8.
radiation therapy. Ann Surg 1993;218:22–8. 61. Romestaing P, Lehingue Y, Carrie C, et al. Role of
48. Ryoo MC, Kagan AR, Wollin M, et al. Prognostic 10-Gy boost in the conservative treatment of
factors for recurrence and cosmesis in 393 early breast cancer: results of a randomized
patients after radiotherapy. Radiology 1989; clinical trial in Lyon, France. J Clin Oncol
172:555–9. 1997;15:963–8.
49. Heimann R, Powers C, Halpern HJ, et al. Breast 62. Solin LJ, Fowble BL, Schultz DJ, Goodman RL.
preservation in stage I and II carcinoma of the The significance of pathology margins of
breast. The University of Chicago experience. tumor excision on the outcome of patients
Cancer 1996;78:1722–30. treated with definitive irradiation for early
50. Spivack B, Khanna MM, Tafra L, et al. Margin stage breast cancer. Int J Radiat Oncol Biol
status and local recurrence after breast-con- Phys 1991;21:279–87.
serving surgery. Arch Surg 1994;129: 952–6. 63. Smitt MC, Nowels KW, Zdeblick MJ, et al. The
51. Gage I, Schnitt SJ, Nixon AJ, et al. Pathologic importance of the lumpectomy surgical margin
margin involvement and the risk of recurrence status in long-term results of breast conserva-
in patients treated with breast-conserving tion. Cancer 1995;76:259–67.
surgery. Cancer 1996;78:1921–8. 64. Fisher B, Digham J, Mamounas E, et al. Sequential
52. DiBiase SJ, Komarnicky LT, Schwartz GF, et al. methotrexate and fluorouracil for the treatment
The number of positive margins influences the of node-negative breast cancer patients with
outcome of women treated with breast preser- estrogen receptor-negative tumors: eight-year
vation of early stage breast carcinoma. Cancer results from the National Surgical Adjuvant
1998;82:2212–20. Breast and Bowel Project (NSABP) B-13 and
Breast Cancer and Radiation Therapy 235

first report of findings from NSABP B-19 com- 5-fluorouracil and radiation therapy. Int J
paring methotrexate and fluorouracil with con- Radiat Oncol Biol Phys 1997;39:1059–68.
ventional cyclophosphamide, methotrexate, and 75. Berns M, Klijn J, van Putten WL, et al. P53 pro-
fluorouracil. J Clin Oncol 1996;14:1982–92. tein accumulation predicts poor response to
65. Fisher BJ, Parera FE, Cooke AL, et al. Long-term tamoxifen therapy of patients with recurrent
follow-up of axillary node positive breast can- breast cancer. J Clin Oncol 1998;16:121–7.
cer patients receiving adjuvant systemic ther- 76. Gorski DH, Mauceri HJ, Salloum RM, et al.
apy alone: patterns of recurrence. Int J Radiat Potentiation of the antitumor effect of ionizing
Oncol Biol Phys 1997;38:541–50. radiation by brief concomitant exposures to
66. Fisher BJ, Perera FE, Cooke AL, et al. Long-term angiostatin. Cancer Res 1998;58:5686–9.
follow-up of axillary node positive breast can- 77. Heimann R, Ferguson D, Powers C, et al. Angio-
cer patients receiving adjuvant tamoxifen genesis as a predictor of long-term survival for
alone: patterns of recurrence. Int J Radiat patients with node negative breast cancer. J
Oncol Biol Phys 1998;42:117–23. Natl Cancer Inst 1998;88:1764.
67. Cooke AL, Parera F, Fisher B, et al. Tamoxifen 78. Clark RM, Whelan T, Levine M, et al. Random-
with and without radiation after partial mastec- ized clinical trial of breast irradiation follow-
tomy in patients with involved nodes. Int J ing lumpectomy and axillary dissection for
Radiat Oncol Biol Phys 1995;31:777–81. node-negative breast cancer. An update. J Natl
68. Dalberg K, Johansson H, Johansson U, Rutqvist Cancer Inst 1996;88:1659–64.
LE. A randomzed trial of long term adjuvant 79. Veronesi U. Breast cancer trials on conservative
tamoxifen plus postoperative radiation therapy surgery. Eur J Surg Oncol 1995;21:231–3.
versus radiation therapy alone for patients with 80. Veronesi U, Luini A, Del Vecchio M, et al. Radio-
early stage breast cancer treated with breast therapy after breast preserving surgery in
conserving surgery, Stockholm Breast Cancer women with localized cancer of the breast. N
Study Group. Cancer 1998;82:2204–11. Engl J Med 1993;328:1587–91.
69. Forrest P, Stewart H, Everington D, et al. Ran- 81. Liljegren G, Holmberg L, Adami HO, et al. Sector
domized controlled trial of conservation ther- resection with or without postoperative radio-
apy for breast cancer: 6-year analysis of the therapy for stage I breast cancer. Five year
Scottish trial. Scottish Cancer Trials Breast results of a clinical trial. J Natl Cancer Inst
Group. Lancet 1996;348:708–13. 1994;86:717–22.
70. Turner BC, Haffty BG, Narayanam L, et al. Insulin- 82. Nemoto T, Patel JK, Rosner D, et al. Factors
like growth factor-I receptor overexpression affecting recurrence in lumpectomy without
mediates cellular radioresistance and local irradiation for breast cancer. Cancer 1991;67:
breast cancer recurrence after lumpectomy and 2079–82.
radiation. Cancer Res 1997;57:3079–83 83. Morrow M, Harris JR, Schnitt SJ. Local control
71. Haffty BG, Brown F, Carter D, Flynn S. Evaluation following breast-conserving surgery for inva-
of HER-2 neu oncoprotein expression as a sive breast cancer: results of clinical trials. J
prognostic indicator of local recurrence in con- Natl Cancer Inst 1995;87:1669–73.
servatively treated breast cancer: a case-control 84. Liljegren G, Lindgren A, Bergh J, et al. Risk fac-
study. Int J Radiat Oncol Biol Phys 1996;35: tors for local recurrence after conservative
751–7. treatment in stage I breast cancer. Definition of
72. Sjogren S, Inganas M, Lindgren A, et al. Prognos- a subgroup not requiring radiotherapy. Ann
tic and predictive value of c-erbB-2 overex- Oncol 1997;8:235–41.
pression in primary breast cancer, alone and in 85. Schnitt SJ, Hayman J, Gelman R, et al. A prospec-
combination with other prognostic markers. J tive study of conservative surgery alone in the
Clin Oncol 1998;16:462–9. treatment of selected patients with stage I
73. Gaffney DK, Brohet KM, Lewis CM, et al. breast cancer. Cancer 1996;77:1094–1100.
Response to radiation therapy and prognosis in 86. Solin LJ, Schultz DJ, Fowble BL. Ten year results
breast cancer patients with BRCA1 and BRCA2 of the treatment of early-stage breast carci-
mutations. Radiother Oncol 1998;47:129–36. noma in elderly women using breast-conserv-
74. Formenti SC, Dunnington G, Uzieli B, et al. Orig- ing surgery and definitive breast irradiation.
inal p53 status predicts for pathological Int J Radiat Oncol Biol Phys 1995;33:45–51
response in locally advanced breast cancer 87. Merchant TE, McCormick B, Yahalom J, Borgen
treated preoperatively with continuous infusion P. The influence of older age on breast cancer
236 BREAST CANCER

treatment decisions and outcome. Int J Radiat 100. Silverstein MJ, Lagios MD, Martino S, et al. Out-
Oncol Biol Phys 1996;34:565–70. come after invasive local recurrence in patients
88. Holland R, Hendricks JH, Vebeek AL, et al. with ductal carcinoma in situ of the breast. J
Extent, distribution and mammographic histo- Clin Oncol 1998;16:1367–73.
logical correlations of breast ductal carcinoma 101. Auquier A, Rutqvist LE, Host H, et al. Post-mas-
in situ. Lancet 1990;335:519–22. tectomy megavoltage radiotherapy: the Oslo and
89. Fisher B, Anderson S. Conservative surgery for Stockholm trials. Eur J Cancer 1992;28:433–7.
the management of invasive and non-invasive 102. Gagliardi G, Lax I, Soderstrom S, et al. Predic-
carcinoma of the breast: NSABP trials. tion of excess risk of long-term cardiac mortal-
National Surgical Adjuvant Breast and Bowel ity after radiotherapy of stage I breast cancer.
Project. World J Surg 1994;18:63–9. Radiother Oncol 1998;46:63–71.
90. Fisher B, Constantino J, Redmond C, et al. 103. Kuske R. Adjuvant chest wall and nodal irradia-
Lumpectomy compared with lumpectomy and tion: maximize cure, minimize late toxicity
radiation therapy for the treatment of intraductal [editorial]. J Clin Oncol 1998;16:2579–82.
breast cancer. N Engl J Med 1993;328:1581–6. 104. Arriagada R, Rutqvist LE, Mattson A, et al. Ade-
91. Fisher B, Dignam J, Wolmark N, et al. Lumpec- quate locoregional treatment for early breast
tomy and radiation therapy for the treatment of cancer may prevent secondary dissemination. J
intraductal breast cancer: findings from the Clin Oncol 1995;13:2869–78.
National Surgical Adjuvant Breast and Bowel 105. Overgaard M, Hansen PS, Overgaard J, et al.
Project B-17. J Clin Oncol 1998;16:441–52. Postoperative radiotherapy in high-risk pre-
92. Fisher E, Constantino J, Fisher B. Pathologic find- menopausal women with breast cancer who
ings from the National Surgical Adjuvant receive adjuvant chemotherapy. N Engl J Med
Breast Project (NSABP) protocol B-17. Intra- 1997;337:949–55.
ductal carcinoma (ductal carcinoma in situ). 106. Ragaz J, Jackson SM, Le N, et al. Adjuvant radio-
Cancer 1995;75:1310–9. therapy and chemotherapy in node-positive
93. Fowble B, Hanlon AL, Fein DA, et al. Results of premenopausal women with breast cancer. N
conservative surgery and radiation for mam- Engl J Med 1997;337:956–62.
mographically detected ductal carcinoma in 107. Recht A, Bartelink H, Fourquet A, et al. Post-
situ (DCIS). Int J Radiat Oncol Biol Phys mastectomy radiotherapy: questions for the
1997;38:949–57. twenty-first century. J Clin Oncol 1998;16:
94. Solin LJ, Kurtz J, Fourquet A, et al. Fifteen-year 2886–9.
results of breast conserving surgery and defin-
108. Fowble B, Gray R, Gilchrist K, et al. Identifica-
itive breast irradiation for the treatment of duc-
tion of a subgroup of patients with breast can-
tal carcinoma in situ of the breast. J Clin Oncol
cer and histologically positive axillary lymph
1996;14:754–63.
nodes receiving adjuvant chemotherapy who
95. Vicina FA, Lacerna MD, Goldstein NS, et al.
may benefit from postoperative radiotherapy. J
Ductal carcinoma in situ detected in the mam-
Clin Oncol 1988;6:1107–17.
mographic era: an analysis of clinical, patho-
logic, and treatment related factors affecting 109. Fowble B. Post-mastectomy radiotherapy: then
outcome with breast-conserving therapy. Int J and now. Oncology 1997;11:213–31.
Radiat Oncol Biol Phys 1997;39:627–35. 110. Kozeniowski S. “One to three” or “four or
96. Silverstein MJ, Lagios MD, Craig PH, et al. A more”? Selecting patients for post-mastectomy
prognostic index for ductal carcinoma in situ of radiation therapy. Cancer 1997;80:1357–8.
the breast. Cancer 1996;77:2267–74. 111. Fisher BF, Perera F, Cooke A, et al. Extracapsular
97. Silverstein MJ, Lagios MD. Use of predictors to axillary node extension in patients receiving
plan therapy for DCIS of the breast. Oncology adjuvant systemic therapy: an indication for
1997;11:393–406. radiotherapy? Int J Radiat Oncol Biol Phys
98. Lagios MD, Silverstein MJ. Ductal carcinoma in 1997;38:551–9.
situ. The success of breast conservation ther- 112. Donegan WL, Stine SB, Samter TG. Implications
apy: a shared experience of two single institu- of extracapsular nodal metastases for treatment
tional non-randomized prospective studies. and prognosis of breast cancer. Cancer 1993;
Surg Oncol Clin North Am 1997;6:385–92. 72:778–82.
99. Winchester DP, Strom EH. Standards for diagno- 113. Krag D, Weaver D, Ashikaga T, et al. The sentinel
sis and management of ductal carcinoma in node in breast cancer. N Engl J Med 1998;339:
situ of the breast. CA-A 1998;48:108–28. 941–6.
Breast Cancer and Radiation Therapy 237

114. Fisher B, Bryant J, Wolmark N, et al. Effect of left-side breast cancer treated with adjuvant
preoperative chemotherapy on the outcome of radiotherapy. Int J Radiat Oncol Biol Phys
women with operable breast cancer. J Clin 1998;41:809–15.
Oncol 1998;16:2672–85. 127. Rutqvist LE, Liedberg A, Hammar N, Dalberg K.
115. Fisher B, Brown A, Mamounas E, et al. Effect of Myocardial infarction among women with
preoperative chemotherapy on local-regional early-stage breast cancer treated with conserv-
disease in women with operable breast cancer: ative surgery and breast irradiation. Int J
findings from National Surgical Adjuvant Radiat Oncol Biol Phys 1998;40:359–63.
Breast and Bowel Project B-18. J Clin Oncol 128. Nixon AJ, Manola J, Gelman R, et al. No long-
1997;15:2483–93. term increase in cardiac related mortality
116. Jacquillat C, Weil M, Baillet F, et al. Results of after breast-conserving surgery and radiation
neoadjuvant chemotherapy and radiation ther- therapy using modern techniques. J Clin Oncol
apy in the breast conserving treatment of 250 1998;16:1374–9.
patients with all stages of infiltrative breast 129. Hoe AL, Iven D, Royle GT, Taylor I. Incidence of
cancer. Cancer 1990;66:119–29. arm swelling following axillary clearance for
117. Brenin DR, Morrow M. Breast conservation breast cancer. Br J Surg 1992;79:261–2.
surgery in the neo-adjuvant setting. Semin 130. Siegel BM, Mayzel KA, Love SM. Level I and II
Oncol 1998;25:13–18. axillary dissection in the treatment of early-
118. Ellis P, Smith I, Ashley S, et al. Clinical, prog- stage breast cancer. An analysis of 259 consec-
nostic and predictive factors for primary utive patients. Arch Surg 1990;125:1144–7.
chemotherapy in operable breast cancer. J Clin 131. Kiel KD, Rademaker AW. Early-stage breast can-
cer: arm edema after wide excision and breast
Oncol 1998;16:107–14.
irradiation. Radiology 1996;198:279–83.
119. Mumtaz H, Davidson T, Spittle M, et al. Breast
132. Taylor ME, Perez CA, Halverson KJ, et al. Fac-
surgery after neoadjuvant treatment. Is it nec-
tors influencing cosmetic results after conser-
essary? Eur J Surg Oncol 1996;22:335–41.
vation therapy for breast cancer. Int J Radiat
120. Bucholz TA, Austin-Seymour MM, Moe RE, et
Oncol Biol Phys 1995;31:756–64.
al. Effect of delay in radiation in the combined
133. Olivotto IA, Weir LM, Kim-Sing C, et al. Late
modality treatment of breast cancer. Int J
cosmetic results of short fractionation for breast
Radiat Oncol Biol Phys 1993;26:23–5. conservation. Radiother Oncol 1996;41:7–13.
121. Recht A, Come SE, Henderson IC, et al. The 134. Victor SJ, Brown DM, Horwitz EM, et al. Treat-
sequencing of chemotherapy and radiation ther- ment outcome with radiation therapy after
apy after conservative surgery for early-stage breast augmentation or reconstruction in
breast cancer. N Engl J Med 1996;334:1356–61. patients with primary breast carcinoma. Can-
122. Pierce S, Recht A, Lingos T, et al. Long-term cer 1998;82:1303–9.
radiation complications following conservative 135. Ryu J, Yahalom J, Shank B, et al. Radiation ther-
surgery (CS) and radiation therapy (RT) in apy after breast augmentation or reconstruction
patients with early stage breast cancer. Int J in early or recurrent breast cancer. Cancer
Radiat Oncol Biol Phys 1992;23:915–23. 1990;66:844–7.
123. Fein DA, Fowble BL, Hanlon AL, et al. Does the 136. Hunt KK, Baldwin BJ, Strom EA, et al. Feasibil-
placement of surgical clips within the excision ity of postmastectomy radiotherapy after
cavity influence local control for patients treated TRAM flap breast reconstruction. Ann Surg
with breast-conserving surgery and irradiation. Oncol 1997;4:377–84.
Int J Radiat Oncol Biol Phys 1996;34:1009–17. 137. Karlsson P, Holmberg E, Johansson KA, et al.
124. Geynes G, Gagliardi G, Lax I, et al. Evaluation of Soft tissue sarcoma after treatment for breast
irradiated heart volumes in stage I breast can- cancer. Radiother Oncol 1996;38:25–31.
cer patients treated with postoperative adjuvant 138. Pendlebury SC, Bilous M, Langlands AO. Sarco-
radiotherapy. J Clin Oncol 1997;15:1348–53. mas following radiation therapy for breast can-
125. Hughes-Davies L, Sacks D, Rescigno J, et al. cer: a report of three cases and a review of the
Serum cardiac troponin T levels during treat- literature. Int J Radiat Oncol Biol Phys 1995;
ment of early-stage breast cancer. J Clin Oncol 31:405–16.
1995;13:2582–4. 139. Boice JD, Harvey EB, Blettner M, et al. Cancer in
126. Cowen D, Gonzague-Casabianca L, Brenot-Rossi the contralateral breast after radiotherapy for
I, et al. Thallium-201 perfusion scintigraphy in breast cancer. N Engl J Med 1992;326:781–5.
the evaluation of late myocardial damage in 140. Touboul E, Buffat L, Belkacemi Y, et al. Local
238 BREAST CANCER

recurrences and distant metastases after breast- eral breast tumor recurrence as a predictor of
conserving surgery and radiation therapy for distant disease: implications for systemic ther-
early breast cancer. Int J Radiat Oncol Biol apy at the time of local relapse. J Clin Oncol
Phys 1999;43:25–38. 1996;14:52–7.
141. Leborgne F, Leborgne JH, Ortega B, et al. Breast 144. Haffty BG, Carter D, Flynn SD, et al. Local
conservation treatment of early stage breast recurrence versus new primary: clinical analy-
cancer: patterns of failure. Int J Radiat Oncol sis of 82 breast relapses and potential applica-
Biol Phys 1995;31:765–75. tions for genetic fingerprinting. Int J Radiat
142. Willner J. The role of radiation therapy in the Oncol Biol Phys 1993;27:575–83.
multidisciplinary management of recurrent and 145. Mullen E, Deutsch M, Bloomer WD. Salvage
metastatic breast cancer [correspondence]. radiotherapy for local failures of lumpectomy
Cancer 1995;75:902–3. and breast irradiation. Radiother Oncol 1997;
143. Haffty BG, Reiss M, Beinfield M, et al. Ipsilat- 42:25–9.
14
Carcinoma of
the Breast in Men
PHILIP N. REDLICH, MD, PHD
WILLIAM L. DONEGAN, MD

Breast cancer is not entirely a disease of He described the poor prognosis associated with
women. 0.7 percent of all cases of breast cancer high histologic grade, cutaneous ulceration, and
in the United States occur in males. Men axillary node involvement. Postoperative mor-
account for 0.9 percent of deaths from breast tality was 6.1 percent; only nineteen percent of
cancer. About 0.2 percent of cancers in males 111 cases with complete follow-up survived 5
arise in the breast. An estimated 1,300 new years. Wainwright concluded that the prognosis
cases of breast cancer in men are expected in the was not as good in men as in women.
United States, in 1999, and 400 men will die of Reports from numerous countries document
the disease. Breast cancer causes 0.14 percent of the pervasiveness of male breast cancer.
deaths from cancer in men.1 Because of the low incidence of breast cancer in
The earliest record of cancer of the breast, men, information on the subject is based largely
which dates from the Edwin Smith surgical on case reports and retrospective analyses of
papyrus, describes the disease in a man. This data collected in medical centers and tumor reg-
Egyptian antiquity, written circa 3000 to 2500 istries over many years. Few individual physi-
BC, indicates that no known treatment was suc- cians have personal experience with more than a
cessful for bulging tumors of the breast.2 Holleb small number of cases. Clinical trials of treat-
attributes the first documented case of male ment are nonexistent for males, and in most
breast cancer to John of Arderne in England in respects, advances in treatment are translations
the fourteenth century;3 Meyskens attributes it from lessons learned about the disease in
to William Fabry of Germany in the sixteenth women, the prevalence of which has enabled
century.4 Clinical descriptions of male breast controlled studies. As a consequence, guidelines
cancer began to appear in medical journals in for treatment of women are being used for
France and England in the early nineteenth cen- males, and men are now receiving less radical
tury. Considered a curiosity, male breast cancer operations and more effective systemic adjuvant
received little attention until later that century therapy. Similarly, the role of hereditary factors
when collections of cases began to appear in the and gene mutations are being explored in males.
literature.5 In 1883, Porier published a detailed It has recently come to be appreciated that
description of the clinical evolution of breast breast cancer is similar in both sexes. Early
cancer in males that leaves little room for reports emphasized differences, but accumulat-
improvement. In 1927, Wainwright in Pennsyl- ing information makes it clear there are more
vania was able to report on 418 collected cases.5 similarities than differences; this change is

239
240 BREAST CANCER

important not only for discovering causation patterns known for women (Table 14–1). Case
but for application of treatment. While the eti- control studies associate high risk variously with
ology of breast cancer in both sexes is high socioeconomic status, higher levels of edu-
unknown, there is little reason to believe that it cation, Ashkenazi Jewish descent, childlessness,
is different. Men and women are subject to sim- obesity, limited exercise, tallness, and consump-
ilar environmental exposures. The pathology tion of red meat.14,15 Linkage between male
and clinical courses are parallel, and in similar breast cancer and exposure to low frequency
circumstances, men and women prove equally magnetic fields has not been confirmed.16
curable. The older age of men at diagnosis, sub- From 11 to 27 percent of affected males
areolar origin of the tumor, and presentation in report a family history of breast cancer.17,18–20
more advanced stages with poorer overall prog- Families with high rates of breast cancer some-
nosis can be attributed to the small size of the times include affected males; multiple males may
male breast and the scant notice it receives. be affected and males in more than one genera-
Screening for early detection does not exist for tion of such families have developed breast can-
men, but public and professional awareness of cer.21 Female descendents of males with breast
breast cancer in men, and appropriate applica- cancer are at increased risk, indicating transmis-
tion to men of the intensive research of the dis- sion through the male line. In males, inheritance
ease in women, should result in progress. of breast cancer risk has been linked to germline
mutations in the BRCA2 gene on chromosome
EPIDEMIOLOGY 13q12-13. Between 35 and 45 percent of familial
breast cancer can be accounted for by BRCA2
The age-specific incidence and mortality of mutations, often including families in which
breast cancer rise steadily in males beginning in both males and females are affected. For females
the third decade.6 The disease has been diag- who are members of high-risk families, muta-
nosed in teenagers, but cases usually begin to tions in BRCA1 or BRCA2 carry an estimated 56
appear in the fourth decade of life, with the to 85 percent lifetime risk of breast cancer. Lim-
average age at diagnosis in large series cluster- ited figures indicate that from 4 to 43 percent of
ing around 65 years, 5 to 8 years older than the males with breast cancer carry various mutations
average age of women at diagnosis.7,8 Wide age on chromosome 13q. Family history of breast
ranges are reported, from 23 to 97 years.9 In cancer is usually present in reported series of
reports from various countries, the incidence in male breast cancer patients with BRCA2 muta-
men parallels that of women.10,11 High rates are tions but the frequency of a positive family his-
reported in England and Wales and low rates in tory ranges up to 85 percent.18,22,23 Mutations in
Japan and Finland. Black races in subSaharan the androgen-receptor (AR) gene associated
Africa have a high frequency of affected males, with androgen insensitivity syndrome are also
often attributed to a high prevalence of liver linked to male breast cancer.24,25
disease which leads to alterations in estrogen Men with Klinefelter’s syndrome (obesity,
metabolism. Males account for 7 percent of hypogonadism, aspermatogenesis, increased
cases of breast cancer in Tanzania12 and 9 per- urinary gonadotropins, and gynecomastia),
cent of cases in Nigeria.13 The lower average identified by an XXY karyotype are estimated
age at diagnosis in African countries also sug- to have a 20- to 50-fold increase in risk for
gests an earlier onset. breast cancer and a 3 percent lifetime risk.26–29
Factors identified with high risk for men are Nevertheless, cases of Klinefelter’s syndrome
fragmentary and sometimes controversial but are not regularly found in reported series of
suggest genetic, hormonal, and environmental males with breast cancer; the frequency varies
influences. In many respects, they reflect the risk widely, from 0 to 7.5 percent.
Carcinoma of the Breast in Men 241

Endocrine abnormalities are not often found Table 14–1. ASSOCIATIONS WITH INCREASED RISK
FOR BREAST CANCER IN MEN
in males with breast cancer, but available infor-
mation suggests some role for excess estrogen or Genetic
First-degree relatives with breast cancer
a deficiency of androgen. High levels of endoge- Ashkenazi Jewish descent
nous estrogens may result from obesity and liver BRCA2 gene mutations
cirrhosis, which are often associated with male Klinefelter’s syndrome
Androgen insensitivity
breast cancer in Denmark and in African coun- Environmental exposures
tries.30,31 Testicular function declines with aging Ionizing radiation
Estrogens
as the incidence of breast cancer rises. Breast Occupational exposures
cancer has been reported in three orchiec- Soap and perfume workers
tomized male transsexuals treated with estrogen Blast furnace workers and steelworkers
Reduced testicular function
to enhance breast development.32,33 Crichlow Mumps orchitis
cites four cases of breast cancer in men treated Inguinal herniorrhaphy
Undescended testes
with estrogens for prostate cancer; more fre- Gynecomastia
quent than primary breast carcinoma, however, Hyperprolactinemia
among men with prostatic carcinoma is metasta- Head trauma
Hyperprolactinemia
tic involvement of the breast.34 In one series, no Other
breast cancers were seen in over 4,000 males High body weight early in life
High socioeconomic status
treated with estrogens for prostate cancer, but Higher education
durations of exposure may have been relatively Childlessness
short. Androgen deficiency is suggested by the
frequent histories of orchitis, inguinal hernior-
rhaphy, mumps infections in adulthood, orchiec- prior history of breast irradiation in 3.1 percent
tomy, and testicular injury among men with of 229 men with breast cancer.9
breast cancer.35 Impaired testicular function may Ductal and lobular development of the male
result from occupational exposure to high envi- breast from genetic, environmental, or endoge-
ronmental temperatures and chemicals, which nous causes may place it at increased risk for
is reported by many affected men. carcinogenesis. Up to 40 percent of breast can-
There have been a number of reports associ- cers in males are associated with gynecomastia.
ating male breast cancer with chronic hyper- This relationship is inconclusive in view of the
prolactinemia. Such cases have included bilat- high frequency of gynecomastia in adult males.
eral breast involvement36a and a history of Noteworthy, however, is the parallel increase of
prolactinoma and head injury.36b The precise breast cancer and gynecomastia in men with
role of prolactin and any associated endocrine aging and the derivation of cancers from ductal
disturbances is undetermined. and lobular elements when present. Ductal hyper-
Case reports document primary breast can- plasia is often seen in association with ductal
cer in men after exposure of the breast to ioniz- carcinoma in males, and in situ and invasive lob-
ing radiation, in one case to treat pubertal ular carcinoma has been seen with Klinefelter’s
gynecomastia.37,38 Radiation is known to be car- syndrome39 and after chronic cimetidine stimu-
cinogenic for the breasts of women, particularly lation of the male breast.40 The presence of
with exposure early in life. Women exposed to gynecomastia and the influences that produce it
atomic radiation or to multiple fluoroscopies in are often indistinguishable.
the course of treatment for tuberculosis, or who Of importance in the epidemiology of breast
have been irradiated for mastitis or treated with cancer in men is freedom of men from the
radiation for Hodgkin’s disease, are known to be unique reproductive functions of women that
at increased risk. Reid and colleagues found a are so prominent in risk for breast cancer. The
242 BREAST CANCER

absence of these promoters is potentially useful pure form or mixed with an invasive compo-
in providing a less cluttered view of the disease. nent.42,43 Ductal carcinoma in situ (DCIS) com-
The fact remains that in the majority of men or prises approximately 5 percent of all cases of
women, no special risk feature is evident other male breast carcinoma but ranges as high as 17
than age. Avoiding potential mammary carcino- percent in reported series.43 The median age of
gens and aspiring to a low-risk profile are some occurrence of DCIS is usually the late 50s to
lessons in prevention derived from studies of mid-60s but has been reported in men under the
breast cancer in males. Fortunately, ionizing age of 40 years. The most frequent histologic
radiation is no longer used to treat pubertal pattern is the papillary subtype, with the major-
gynecomastia, acne, and other benign condi- ity of cases being of low or intermediate grade.
tions of youth. Hormonal stimulation of the In a recent review of Paget’s disease, this histo-
male breast and obesity are avoidable. Identifi- logic type is characterized as presenting in the
cation of individuals with an inherited high risk fifth to sixth decade of life and being associated
for breast cancer through genetic testing can with a palpable mass in 50 percent of cases.44
permit more informed decisions about prophy- Virtually all known histologic types of inva-
lactic mastectomy for men.41 sive breast cancer have been identified in men.
Invasive ductal carcinomas predominate,
PATHOLOGY accounting for up to approximately 90 percent
of cases. Also, special histologic types have
The same histologic types of breast cancer been noted. Both invasive lobular carcinoma
occur in men and women but the frequencies of and lobular carcinoma in situ (LCIS) have been
these types vary (Table 14–2). Noninvasive duc- reported in men but are much less common
tal carcinoma has been described either in a than in women. Sarcomas comprise a minority
of reported invasive breast cancers; there have
Table 14–2. HISTOLOGIC TYPES OF been a variety of types noted (see Table 14–2).
BREAST CANCER IN MEN Metastatic cancer to the breast must be
Noninvasive Carcinoma included in the differential diagnosis of breast
Ductal carcinoma in situ masses. Lung carcinoma has been reported to
Lobular carcinoma in situ
Paget’s disease metastasize to the male breast.45
Papillary carcinoma in situ
Invasive Carcinoma
Argyrophilic neuroendocrine carcinoma CLINICAL PRESENTATION
Colloid carcinoma AND EVALUATION
Inflammatory carcinoma
Intracystic papillary carcinoma
Invasive ductal carcinoma The clinical features of male breast cancer have
Invasive lobular carcinoma been well described in the literature9,20,46–54 and
Invasive papillary carcinoma
Medullary carcinoma
recently summarized.55 Signs and symptoms of
Mucinous carcinoma male breast cancer are shown in Table 14–3.
Oncocytic carcinoma The mean age of patients presenting with this
Secretory carcinoma
Sarcoma disease as noted above, is usually in the late 50s
Phyllodes tumor to mid-60s, with a range from the mid-20s to
Fibrosarcoma
Leiomyosarcoma
the early 90s. The most common presenting
Lymphosarcoma complaints are related to a breast mass, usually
Myxoliposarcoma occurring in > 70 percent of cases, and axillary
Osteosarcoma
Spindle cell sarcoma adenopathy, occurring in 30 to 50 percent of
cases. For pure DCIS, a subareolar mass and
Adapted from Donegan WL, Redlich PN. Breast cancer in men.
Surg Clin North Am 1996;76:343–63. nipple discharge were the two most common
Carcinoma of the Breast in Men 243

Table 14–3. SIGNS AND SYMPTOMS Table 14–4. STAGE OF DISEASE AT PRESENTATION
OF MALE BREAST CANCER
Stage Frequency (%)
Frequent
Breast mass 0 0–17
Axillary adenopathy I 10–40
Nipple retraction II 20–40
Nipple discharge III 15–40
IV 10–15
Retraction of skin
Ulceration of nipple or skin
Less Frequent
Fixation to muscle Evaluation of breast lesions includes the use
Breast pain of mammography, ultrasonography, fine needle
Inflammatory skin changes
Skin discoloration aspiration cytology (FNAC), needle core biopsy,
and open biopsy. Characteristics of male breast
cancer on mammography include a mass eccen-
symptoms in a recent series, occurring in 58 tric to the nipple, spiculated margins, and
and 35 percent of patients, respectively.43 In microcalcifications (Figure 14–1).56–58 Malig-
virtually all series, there is a report of signifi- nancy must be differentiated from gynecomas-
cant delay in diagnosis of breast cancer in men. tia, which often presents as an area of increased
In early series, the mean duration of symptoms density positioned symmetrically in the retroare-
was > 14 months. In recent series, the mean ola region, but may obscure tumors.56 Secondary
duration is declining to a range of 3 to 6 radiologic signs of malignancy include architec-
months.48,50,52,54 There is a history of trauma in tural distortion, nipple and skin changes, and
many series, ranging from 5 to 10 percent of enlarged axillary nodes.56 The ultrasound fea-
cases. Many series report the presence of
gynecomastia associated with this disease in 7
to 23 percent of cases.9,20,50 The mass is cen-
trally located in the majority of cases and has
an average diameter of 2.5 to 3.0 cm, with a
range of 0.5 to 12 cm. Bilaterality of the disease
is present in usually < 1 percent of cases,
although in one series,50 7 percent of patients
were found to have bilateral disease. Clinically
suspicious axillary adenopathy is often found
in these patients, ranging as high as 55 percent.
The accuracy of the clinical exam is question-
able, however, and pathologically proven
metastases are usually more frequent. In some
series, histologically proven axillary metastases
occur as often as 70 percent of the time but
more frequently are in the 40 to 60 percent
range. The stage of disease at presentation is
somewhat variable between reported series and
may not be entirely comparable from series to Figure 14–1. Mammography of a male patient with breast
cancer. This 83 year-old man presented with a 4-month his-
series due to the large time spans involved and tory of a right-breast mass. He had a 4.5 cm tumor, T2N1M0
modification of staging systems over time. (stage IIB), treated by modified radical mastectomy and adju-
vant tamoxifen. He is free of disease 3 years later. Shown are
Stratification of patients by TNM stage at the the medial-lateral-oblique views of both breasts, the tumor
time of presentation is presented in Table 14–4. being in the right breast on the left side of the figure.
244 BREAST CANCER

tures of male breast cancer compared to other Observed survival of men is regularly inferior to
benign entities, including gynecomastia, lipoma, that of women. The Winchesters and colleagues
and fat necrosis have been reported.58 Male reported on 4,755 cases of male breast cancer
breast cancer appears as a hypoechoic lesion obtained through the National Cancer Data Base
with irregular margins with architectural distor- and compared them with 624,174 cases of breast
tion of surrounding normal breast tissue and cancer in women.7 The mean age of men, 64.7
subcutaneous fat. Ultrasound should be regarded years, was older than that of women, which was
as complimentary to mammography in the eval- 60.9 years. Similar distributions of tumor grades
uation of the male breast. were found. Men presented in more advanced
The first diagnostic step in the evaluation of stages than women, and 5-year survival was sig-
a male breast mass is often FNAC. Cytologic nificantly lower. However, when adjusted for age
features of male breast cancer are similar to and comorbidity, survival was equivalent.
those seen in the female and allow this modality The survival of men with breast cancer
to be a reliable means of assessment.59 Difficul- compared to women with breast cancer has
ties encountered using FNAC include epithelial been addressed in many series. An overall
hyperplasia associated with gynecomastia and worse prognosis for men has been identified by
the differentiation between primary and a number of authors.51,54,65 Other authors sug-
metastatic lesions of the breast.59 Combined gest that the prognosis in male breast cancer is
physical examination and FNAC for the evalua- no worse than that for women with comparable
tion of palpable breast masses in males has been disease.7,47,50,66 Guinee and colleagues sug-
studied.60 This combination was found to be gested that the prognosis is the same for male
diagnostically accurate and resulted in a reduc- and female patients when stratified on the basis
tion of patient charges compared to routine open of histologically positive nodes.49 There has
biopsy. In another series reviewing the diagnos- been a similar prognosis for male and female
tic evaluation of over 700 male patients, the role patients when analyzed by disease-specific sur-
of palpation, mammography, cytology, and vival, tumor size, and axillary node involve-
ultrasound was evaluated. The combined palpa- ment reported by other authors as well.50,52,66
tion and mammography demonstrated a very There are a number of influences unrelated to
high sensitivity for an accurate diagnosis.61 breast cancer itself that contribute to the unfa-
Accurate diagnosis by cytology requires an vorable comparison of men and women. Among
experienced cytologist, the absence of which them are the older age at diagnosis and their
mandates either needle core biopsy or ulti- shorter life expectancy after 65 years of age due
mately open biopsy of lesions. Open biopsy to comorbid disease; men have higher rates of
should be performed in all lesions where uncer- death from heart disease, second cancers, and
tainty exists regarding the diagnosis, both to stroke.55 These confounding variables bias com-
confirm the diagnosis and obtain tissue for parisons of observed survival and disease-free
estrogen and progesterone receptor measure- survival in favor of women.7 More valid compar-
ments. Receptor status is important as a prog- isons require the use of survival adjusted for nat-
nostic indicator for survival and as an indicator ural mortality (adjusted survival) or of disease-
for response to hormonal manipulation.54,62 specific survival (DSS).7 Adjusted five-and
ten-year survivals reported by Joshi and col-
PROGNOSTIC FACTORS leagues67 were 76 and 42 percent, respectively
and DSS of 74 percent at 5 years and 51 percent
Overall survival for men with breast cancer in at 10 years were reported by Cutuli and col-
large series ranges from 53 to 70 percent at 5 leagues.52 Further detrimental to the survival of
years and 38 to 53 percent at 10 years.52,63,64 men is the high frequency of locally and region-
Carcinoma of the Breast in Men 245

ally advanced disease and of disseminated dis- survival for male breast cancer. Crichlow
ease, features in keeping with delay in diagnosis reported 5-year survivals of 79 and 28 percent for
and not necessarily with inherently aggressive 143 patients without and with pathologic axil-
cancers.7,67 Skin involvement is often present lary metastases, respectively.29 In a review of
and involved axillary nodes are found in 45 to 397 nondisseminated cases, the 5-year DSS of
65 percent of men with axillary dissections. 77 and 51 percent for cases with histologically
When factors of stage and comorbidity are uninvolved and involved nodes, respectively,
taken into account, however, investigators find were reported.52 As in women, the absolute
little or no difference between the prognosis of number of involved nodes is inversely related to
males and females with breast cancer.44,50,52,66,67 survival. In 335 collected cases, Guinee and
The patient’s TNM stage at diagnosis is colleagues demonstrated that the 5-year sur-
important prognostically for men, and outcome vival for those with negative nodes was 90 per-
by stage is largely not influenced by variations cent, for one to three positive nodes was 73 per-
in local treatment (Figure 14–2). The current cent, and for four or more positive nodes 55
TNM staging system, derived from studying percent.49 Others have found the same relation-
breast cancer in women, may not be entirely ship53,54 (Figure 14–3). Unequal numbers of
appropriate for men. Male cancers average 2.0 involved nodes contribute to varied prognoses
to 2.9 cm in diameter, but the diminutive reported for node-positive cases (see Figure
breasts of men allow even small tumors to read- 14–3). Skin and nipple involvement are identi-
ily reach underlying muscle and overlying skin fied with adverse survival (Figure 14–4).67 Skin
or nipple.8,67 Pivot and colleagues found skin or ulceration becomes insignificant, however,
muscle involvement in 45 percent of 85 cases; when tumor size is taken into consideration,
skin involvement was directly correlated with and fixation to skin and chest wall are not
tumor size.68 Forty-five percent of tumors < 2 important prognosticially when size and nodal
cm in diameter had produced nodal metastases, status are taken into account.49 There has been
and all tumors > 5 cm in diameter had produced a statistically significant difference in survival
nodal metastases. There was a similar prognosis based on histologic grade reported in one series,
for T3 and T4 tumors. These authors proposed a a worse survival associated with grade III ver-
reduced T1, T2, and T4 classification for men. sus grade II disease.53
Among traditional prognosticators, the pres-
ence of axillary metastases and primary tumor
size are the most important features in undis-
seminated cases.51,69,70,71 There is a direct corre-
lation between the size of the primary tumor and
the involvement of axillary lymph nodes that
links these two clinical features.68,71 Guinee and
associates found a progressive drop in 5-year
survival from 94 percent for cases with tumors 0
to 10 mm in diameter to 39 percent in cases with
tumors > 51 mm in diameter.49 Tumor size is
important prognostically, independent of axillary
node status. In node-negative cases, the relative Figure 14–2. Survival of men with breast cancer stratified by
TNM stage. Differences between the following stages reached
risks of death associated with T0-T1, T2, and T3- statistical significance: 0 vs III, 0 vs IV, I vs II, I vs III, I vs IV, II
T4 cases were 1.0, 2.0, and 3.2, respectively.52 vs III, II vs IV, and III vs IV. The number of cases is shown in
The presence of axillary metastases is the parentheses. Reprinted with permission from Donegan WL,
Redlich PN, Lang PJ, Gall MT. Carcinoma of the breast in
single-most important prognostic indicator of males: a multi-institutional survey. Cancer 1998;83:498–509.
246 BREAST CANCER

found 5-year survivals of 60, 40, and 5 percent


in men with tumor grades of I, II, and III,
respectively.75 For diploid tumors, Pich and col-
leagues found a median survival of 77 months
and only 38 months for aneuploid tumors73
Mutation of p53 cellular protein shortens
median survival and disease-free survival.73,76
The mean S-phase fraction of male breast can-
cers (7.2%) approximates that of females.77
Winchester and colleagues found high S-phase
Figure 14–3. Survival by men with breast cancer stratified by
fraction (SPF) to be a significant indicator of
number of axillary nodes with metastases. Differences
between 0 vs 4+ positive nodes and 1–3 vs 4+ positive nodes poor disease-free survival.70 In a study of 27
reached statistical significance. The number of cases is shown male breast cancers, Pich and associates found
in parentheses. Reprinted with permission from Donegan WL,
Redlich PN, Lang PJ, Gall MT. Carcinoma of the breast in that strong staining for argyrophilic nuclear
males: a multi-institutional survey. Cancer 1998;83:498–509. organizer regions and for proliferating cell
nuclear antigen were correlated with inferior
The influence of steroid receptors on the survival.74 The frequency of tumor markers
prognosis of men is controversial. Estrogen with and without established prognostic signif-
receptor (ER) positivity, which is regularly icance are shown in Table 14–5.
more frequent in men than in women, is a weak
but favorable prognostic sign for women. In TREATMENT OF LOCALIZED DISEASE
men, ER positivity has been associated with
both increased and decreased survival.54,63,72 The treatment of male breast cancer localized
High tumor grade and aneuploidy are both to the breast and axillary nodes is mastectomy
associated with shortened survival.63,73 Pich with axillary lymph node dissection.55 In sev-
and colleagues found median survival signifi- eral recent series, modified radical mastectomy
cantly less for grade III than for grade II was the most common procedure performed,
tumors.74 Visfelt and Scheike graded 150 male with from 34 to 76 percent of patients treated in
breast carcinomas according to the degree of this fashion.8,20,50–52,54,65 In one multi-institu-
tubule formation, mitoses, and atypia and tional survey, 82 percent of patients diagnosed
since 1986 were treated by modified radical
mastectomy.54 Of 242 patients treated in the
Department of Veterans’ Affairs, 51 percent
underwent modified radical mastectomy.8
Other surgical procedures reported in these
patients include radical mastectomy, simple
mastectomy, and lumpectomies.
The use of radical mastectomy has decreased
markedly in recent decades. No significant dif-
ference in outcome for patients who underwent
radical mastectomy compared to modified radi-
cal mastectomy was found in a number of
series.20,78 The effect of the extent of mastec-
Figure 14–4. Direct involvement of the nipple in a man with tomy on the local regional recurrence rate is
breast carcinoma. The nipple is fixed to the underlying tumor, unclear, but a trend toward a lower local recur-
endurated, and retracted. The site of skin biopsy is marked by
a stitch. rence rate was identified for patients undergo-
Carcinoma of the Breast in Men 247

ing mastectomy versus lumpectomy.52 Axillary ifen.46,50,80,81 Orchiectomy has been reported in a
dissection is considered part of the local– few series but is usually limited to only 3 per-
regional treatment of breast cancer. Cutuli and cent of cases.9,20,50 Tamoxifen is generally well
colleagues reported a statistically significant tolerated by men, but side effects have been
difference in the regional nodal recurrence rate reported that, on occasion, may lead to termina-
of patients undergoing axillary dissection com- tion of treatment.80,81 Combination chemotherapy
pared to those without dissection—1.2 percent administered in an adjuvant setting has been
of 320 patients with axillary dissection had a used and reported by a number of authors.9,52,54,55
regional recurrence, compared to 13 percent of Treatment with cyclophosphamide, methotrex-
77 patients without axillary dissection.52 ate, and 5-fluorouracil (CMF) has been reported
Postoperative radiation therapy is frequently from the National Cancer Institute in a series of
used as adjuvant therapy for male breast cancer. 24 patients with stage II disease. The projected
Its use, however, varies widely.55 Some series 5-year survival rate was > 80 percent, represent-
suggest a decrease in the local–regional recur- ing an improvement over survival rates reported
rence rate with the use of postoperative radia- in other series.82 In another report, 5-fluo-
tion therapy,52,69 whereas no efficacy was noted rouracil, doxorubicin, and cyclophosphamide
in other series.20,78 Chest wall radiation offers (FAC) or CMF was administered in the adjuvant
no survival benefit.20,55,69 Overall, postopera- setting, with the projected 5-year survival > 85
tive radiotherapy may reduce the local recur- percent.83 Adverse effects of these chemother-
rence rate and should be considered as part of apy regimens have been reported, limiting the
the overall treatment plan for cases at high risk ability of patients to tolerate all the planned
for local or regional recurrence.52,79 treatments. In a recent series, Donegan and col-
Systemic adjuvant therapy, either chemo- leagues evaluated the effect of systemic adju-
therapy, hormonal therapy, or both, is used for vant therapy, either chemotherapy, hormonal
male patients based on the experience in female therapy, or combinations of both, on survival.54
patients. The modality most often used for post- No improvement in overall survival was demon-
operative adjuvant hormonal therapy is tamox- strated; however, further analysis revealed

Table 14–5. TUMOR MARKERS IN MALE BREAST CANCERS

Marker Frequency (%) Prognosis Reference

ER+ 64–93 Controversial Rayson,94b Willsher,95 Joshi,67 Bruce,63 Cutuli52


PR+ 73–96 Rayson,94b Willsher,95 Joshi,67 Cutuli52
pS2 50 Bruce63
Cathepsin D 46 Bruce63
AR+ 95 Bruce63
HER2/neu+ 21–45 Unfavorable Rayson,94b Bines,96 Willsher,95 Joshi,67 Bruce63
p53+ 21–58 Unfavorable Rayson,94b Bines,96 Willsher,95 Dawson,97 Joshi67
Bruce,63 Anelli,76 Pich73
MIB-1+ 38–40 Unfavorable Rayson,94b Willsher95
bcl-2+ 93 Rayson94b
EGFR+ 20 Willsher95
Mean SPF 7.2 (mean) Highly unfavorable Jonasson,77 Winchester70
Grade III 33–73 Unfavorable Willsher,95 Jonasson,77 Bruce,63 McLachlan53
Aneuploidy 57 Unfavorable Jonasson,77 Pich73
Ki-ras 12 Dawson97
AgNOR+ Highly unfavorable Pich74
pcna Highly unfavorable Pich74

ER = estrogen receptor protein; PR = progesterone receptor protein; AR = androgen receptor; EGFR = epidermal growth factor receptor;
pS2 = estrogen-dependent protein; HER2/neu = transmembrane oncoprotein; Grade III = histologic grade; AgNOR = argyrophilic nucleolar
organizer regions; pcna = proliferating cell nuclear antigen.
248 BREAST CANCER

improved DSS in a subset of patients. Disease CHANGES OVER TIME


specific survival was improved in patients who
were axillary lymph node positive with estrogen During the last century, clinical acumen, classi-
receptor positive tumors, although the number fication systems, techniques of pathologic exam-
of patients with information regarding such sys- ination and methods for reporting the results of
temic therapy was small.54 treating breast cancer have undergone extensive
changes. True comparability of reporting
between periods is unlikely, particularly when
TREATMENT OF METASTATIC DISEASE
data are few and incomplete, as is the case for
Many men with breast cancer present with male breast cancer. Wainwright’s review of col-
metastatic disease, ranging from 11 to 16 per- lected cases diagnosed before 1927 provides one
cent in several recent series.20,46,47,50,54 The pat- baseline against which to evaluate progress. By
tern of spread in male patients is similar to that modern standards, it provides clear signs of
seen in female patients including local regional delayed diagnosis and advanced disease.5 The
recurrences and metastases to bone, lung, liver, average symptomatic interval prior to diagnosis
skin, and other areas.9,20,50,51,53 The first-line pal- was 2.4 years. The modal age of cases was 60 to
liative therapy used by most authors is hormonal 64 years, with a mean of 52.6 years. Skin ulcer-
therapy, most often with tamoxifen.9,50,84,85 Reid ation was observed in 38 percent of cases, and
and colleagues reported the use of hormonal 68.9 percent of patients had involved axillary
therapy in 73 percent of patients treated for lymph nodes. Overall 5-year survival was only
metastatic disease.9 There has been a response 19 percent.20 While the age of men at the time of
rate of 25 to 58 percent to tamoxifen therapy presentation has not declined convincingly,
reported in various series, the mean duration of recent reports document shorter median sympto-
response being 9 to 12 months, with few matic intervals of 3 to 8 months, suggesting an
reported side effects. The response to tamox- earlier diagnosis.17,94a Average tumor sizes stub-
ifen seems to correlate with estrogen receptor bornly remain at 2.0 to 2.5 cm, but skin ulcera-
status in that patients with receptor negative tion is now reported in only 12 to 13 percent of
tumors show no response.85,86 Historically, cases.34,49 The rate of dissemination at diagnosis
other hormonal treatments have been does not exceed that for females (in most reports
employed, including orchiectomy, adrenalec- 7 to 14 percent) and 2.9 to 5.6 percent of cases
tomy, and hypophysectomy. Response rates for are now being diagnosed in situ—both favorable
orchiectomy are in the 30 to 60 percent signs even in the absence of a concerted public
range.47,87–89 Response to secondary endocrine screening effort.49,55 The frequency of metastasis
procedures have been reported in > 50 percent to axillary nodes generally remains high (45 to
of cases.87 Other hormonal manipulations, 51 percent), but in some reports the rate is as low
including androgen therapy and gonadotropin- as 33 percent.34,52,55
releasing hormone agonist analogue therapy Favorable changes in managment of men have
have been reported.55 Systemic chemotherapy been less radical surgery and more frequent use of
may be considered as a second-line palliative systemic adjuvant chemohormonal therapy. Men
treatment, with an overall response rate of 30 to are now treated with modified radical mastec-
40 percent.28,62,87,90–92 Combination chemother- tomy more often than with radical mastectomy,
apy, such as CMF or doxorubicin-containing with no apparent detrimental effect and with
regimens, have been reported in various series improved cosmetic outcome. Chemotherapy,
and may be useful in patients who have failed tamoxifen, or both, targeted at micrometastases,
prior therapies.62,87,91,93 are an increasingly frequent component of pri-
mary treatment, with expectations that the bene-
Carcinoma of the Breast in Men 249

fits will parallel those for women. The indica- therapy, especially in view of the high incidence
tions are that these changes are improving the of estrogen-receptor positivity of breast cancer
outlook for cure or extended survival of men. in men compared to women. Important prog-
Current 5-year survivals of 47 to 51 percent over- nostic variables include stage, tumor size, and
all, and as high as 76 percent in undisseminated axillary node involvement. Also, survival
cases, compare favorably with results reported decreases with increasing number of axillary
early in the century. Some investigators have sug- nodes containing metastatic disease. Survival
gested that changes in therapy have resulted in lit- trends suggest improvement in the last one to
tle improvement in symptomatic interval, stage at two decades, although the length of this
diagnosis, or survival for men with breast cancer. improvement is limited by the older male popu-
In a report from the Mayo Clinic comparing the lation with this disease. Though the survival of
results from the period 1933 to 1958 with the men is reported to be similar to women com-
period 1959 to 1983, fewer radical operations and pared stage for stage, comorbidities in men,
a higher frequency of adjuvant systemic therapy such as heart disease, strokes, and second
were found in the more recent period, but not an malignancies, contribute to an overall survival
improvement in median survival (5.5 years vs 6.3 that may be inferior to that found in women.
years, respectively) or in 5-year disease-free sur-
vival of males with breast cancer (52 vs 47 per- REFERENCES
cent).20 In a more recent 10-year span from 1986
to 1995 compared with the period 1953 to 1985, 1. Landis SH, Murray T, Bolden S, Wingo PA. Can-
cer Statistics, 1999. CA Cancer J Clin 1999;
investigators at the Medical College of Wiscon-
49:8–31.
sin noted a change of surgical treatment to mod- 2. Breasted JH. The Edwin Smith surgical papyrus.
ified radical mastectomy and increased use of Vol. I. Chicago: University of Chicago Press;
systemic therapy and found improvement in 1930. p. 403–6.
5-year observed survival of males treated in 3. Holleb AI, Freeman HP, Farrow JH. Cancer of the
Wisconsin hospitals, from 42 to 59 percent male breast. I and II. N Y State J Med 1968;68:
(p = .055).54 Stage for stage, the prognosis for 544,656.
men is comparable to, or somewhat inferior to, 4. Meyskens FL Jr, Tormey DC, Neifeld JP. Male
breast cancer: a review. Cancer Treat Rev
that for women. Truly accurate comparisons,
1976;3:83–93.
however, are challenged by the need to control 5. Wainwright JM. Carcinoma of the male breast.
for multiple tumor-related and nontumor-related Arch Surg 1927;14:836–59.
influences on outcome. 6. Cutler SJ, Young JL Jr (eds). Third National Can-
cer Survey: Incidence Data. National Cancer
Institute Monograph 41. March 1975. DHEW
SUMMARY
Publication No (NIH) 75–787. US Department
of Health, Education, and Welfare. Public
Breast cancer in men is infrequent and occurs in
Health Service. National Institutes of Health.
an older population than does the disease in National Cancer Institute, Bethesda, Maryland
women. The disease in men is remarkably simi- 20014. US Government Printing Office. Wash-
lar to the disease in women. Men present with ington C.C. 20402. p. 106–107.
advanced stages of disease more often than 7. Winchester DJ, Bilimoria MM, Scott-Conarr CEH,
women, but a trend toward earlier diagnosis is Menck HR, Winchester DP. Male breast cancer:
evident. There is an increased awareness of the presentation, treatment, and survival (In press).
8. Schultz MZ, Coplin M, Radford D, Virgo KS,
importance of family history and an association
Johnson FE. Outcome of male breast cancer
with BRCA2 mutations. Analysis of treatment (MBC) in the Department of Veterans Affairs
trends demonstrates the use of less radical (DVA). Proc Annu Meet Am Soc Clin Oncol
surgery and increased use of systemic adjuvant 1996;15:A257.
250 BREAST CANCER

9. Reid C, Pintilie M, Goncalves S, et al. A review of breast carcinoma. J Clin Endocrinol Metab
229 male breast cancer patients presenting to 1996;81:3404–7.
the Princess Margaret Comprehensive Cancer 25. Poujol N, Lobaccaro JM, Chiche L, et al. Func-
Centre. Proc Annu Meet Am Soc Clin Oncol tional and structural analysis of R607Q and
1997;16:A476. R608K androgen receptor substitutions associ-
10. Ewertz M, Homnberg L, Kajjalaninen S, et al. ated with male breast cancer. Mol Cell
Incidence of male breast cancer in Scandinavia Endocrinol 1997;130:43–51.
1943–1982. Int J Cancer 1989;43:27–31. 26. Klinefelter HF Jr, Reifenstein EC Jr, Albright F.
11. Schottenfeld D, Lilienfeld AM, Diamond H. Some Syndrome characterized by gynecomastia,
obervations on the epidemiology of breast can- aspermatogenesis without A-leydigism and
cer among males. Am J Pub Health increased excretion of follicle-stimulating hor-
1963;52:890–7. mone. J Clin Endocrinol 1942;2(11):615–27.
12. Amir H, Makwaya CK, Moshiro C, Kwesigabo G. 27. Hultborn R, Hanson C, Kopf I, et al. Prevalence of
Carcinoma of the male breast: a sexually trans- Klinefelter’s syndrome in male breast cancer
mitted disease? East Afr Med J 1996;73:187–90. patients. Anticancer Res 1997;17(6D):4293–7.
13. Hassan I, Mabogunje O. Cancer of the male breast 28. Volm MD, Gradishar WJ. How to diagnose and
in Zaria, Nigeria. East Afr Med J 1995;72: manage male breast cancer. Contemp Oncol
457–8. 1994;4:17–28.
14. Hsing AW, McLaughlin JK, Cocco P, et al. Risk 29. Crichlow RW. Breast cancer in males. Breast, Dis-
factors for male breast cancer (United States). eases of the Breast 1976;Oct/Dec:2(4):12–16.
Cancer Causes Control 1998;9:269–75. 30. Casagrande JT, Hanisch R, Pike MC, et al. A case-
15. D’Avanzo B, LaVecchia C. Risk factors for male control study of male breast cancer. Cancer
breast cancer. Br J Cancer 1995;71:1359–62. Res 1988;48:1326–30.
16. Stenlund C, Floderus B. Occupational exposure to 31. Srensen HT, Friis S, Olsen JH, et al. Risk of breast
magnetic fields in relation to male breast cancer cancer in men with liver cirrhosis. Am J Gas-
and testicular cancer: a Swedish case-control troenterol 1998;93:231–3.
study. Cancer Causes Control 1997;8:184–91. 32. Symmers WC. Carcinoma of the breast in trans-
17. Donegan WL, Perez-Mesa CM. Carcinoma of the sexual individuals after surgical and hormonal
male breast. Arch Surg 1973;106;273–9. interference with the primary and secondary
18. Olsson H, Andersson H, Johansson O, et al. Popu- sex characteristics. Br Med J 1968;2:83–5.
lation-based cohort investigations of the risk 33. Pritchard TJ, Pankowsky DA, Crowe JP, et al.
for malignant tumors in first-degree relatives Breast cancer in a male-to-female transsexual.
and wives of men with breast cancer. Cancer JAMA 1988;259:2278–80.
1993;71:1273–8. 34. Crichlow RW, Kaplan EL, Keamey WH. Male
19. Friedman LS, Gayther SA, Kurosaki T, et al. Muta- mammary cancer: an analysis of 32 cases. Ann
tion analysis of BRCA1 and BRCA2 in a male Surg 1972:175;489–94.
breast cancer population. Am J Hum Genet 35. Mabuchi K, Bross DS, Kessler II. Risk factors for
1997;60:313–9. male breast cancer. J Natl Cancer Inst 1985;74:
20. Gough DB, Donohue JH, Evans MM, et al. A 50- 371–5.
year experience of male breast cancer: is out- 36a. Karamanakos P, Apostolopoulos V, Fafouliotis S,
come changing? Surg Oncol 1993:2:325–33. et al. Synchronous bilateral primary male
21. Everson RB, Fraumeni JF Jr, Wilson RE. Familial breast carcinoma with hyperprolactinemia.
male breast cancer. Lancet 1976 Jan 3; 9–14. Acta Oncol 1996:35:757–9.
22. Couch FJ, Farid LM, DeShano ML, et al. BRCA2 36b. Volm MD, Talamonti MS, Thangavelu M, Gradishar
germline mutations in male breast cancer cases WK. Pituitary adenoma and bilateral male
and breast cancer families. Nat Genet 1996; breast cancer: an unusual association. J Surg
13:123–5. Oncol 1997:64:74–8.
23. Haraldsson K, Loman N, Zhang QX, et al. BRCA2 37. Thompson DK, Li FP, Cassady Jr. Breast cancer in
germ-line mutations are frequent in male a man 30 years after radiation for metastatic
breast cancer patients without a family history osteogenic sarcoma. Cancer 1979;44:2362–5.
of the disease. Cancer Res 1998;58:1367–71. 38. Lowell DM, Marineau RG, Luria SB. Carcinoma
24. Hiort O, Naber SP, Lehners A, et al. The role of of the male breast following radiation. Cancer
androgen receptor gene mutations in male 1968;22:581–6.
Carcinoma of the Breast in Men 251

39. Sanchez AG, Villanueva AG, Redondo C. Lobular 55. Donegan WL, Redlich PN. Breast cancer in men.
carcinoma of the breast in a patient with Kline- Surg Clin North Am 1996;76:343–63.
felter’s syndrome. Cancer 1986;57:1181–3. 56. Dershaw DD, Borgen PI, Deutch BM, Liberman L.
40. San Miguel P, Sancho M, Enriquez JL, et al. Lob- Mammogrphic findings in men with breast
ular carcinoma of the male breast associated cancer. AJR 1993;160:267–70.
with the use of cimetidine. Virchows Arch 57. Cooper RA, Gunter BA, Ramanurthy L. Mam-
1997;430:261–3. mography in men. Radiology 1994;191:651–6.
44. Daltry IR, Eeles RA, Kissin MW. Bilateral pro- 58. Stewart RAL, Howlett DC, Hern FJ. Pictorial
phylactic mastectomy: not just a woman’s review: the imaging reatures of male breast
problem! Breast 1998;7:236–7. disease. Clin Radiol 1997;52:739–44.
42. Cutuli B, Dilhuydy JM, DeLafontan B, et al. Duc- 59. Sneige N, Holder PD, Katz RL, et al. Fine-needle
tal carcinoma of the male breast: analysis of 31 aspiration cytology of the male breast in a can-
cases. Eur J Cancer 1997;33:35–8. cer center. Diagn Cytopathol 1993;9:691–7.
43. Hittmair AP, Lininger RA, Tavassoli FA. Ductal 60. Vetto J, Schimdt W, Pommier R, et al. Accurate and
carcinoma in situ (DCIS) in the male breast. cost effective evaluation of breast masses in
Cancer 1998;83:2139–49. males. Am J Surg 1998;175:383–7.
44. Desai DC, Brennan EJ Jr, Carp NZ. Paget’s disease 61. Ambrogetti D, Ciatto S, Catarzi S, Muraca MG.
of the male breast. Am Surg 1996;62: The combined diagnosis of male breast
1068–72. lesions: a review of a series of 748 consecutive
45. Muttarak M, Nimmonrat A, Chaiwun B. Metasta- cases. Radiol Med 1996;91:356–9.
tic carcinoma of the male and female breast. 62. Bezwoda WR, Hesdorffer C, Dansey R, et al.
Australas Radiol 1998;42:16–9. Breast cancer in men: clinical features, hor-
46. Ribeiro, G. Male breast carcinoma—A review of mone receptor status and response to therapy.
301 cases from the Christies Hospital & Holt Cancer 1987;60:1337–40.
Radium Institute, Manchester. Br J Cancer 63. Bruce DM, Heys SD, Payne S, et al. Male breast
1985;51:115–9. cancer: clinico-pathological features, immuno-
47. van Geel AN, van Slooten EA, Mavrunac M, Hart cytochemical characteristics and prognosis.
AAM. A retrospective study of male breast Eur J Surg Oncol 1996;22:42–6.
cancer in Holland. Br J Surg 1985;72:724–7. 64. Caton J, Rearden T, Ellis R. Male breast cancer: the
48. Hultborn R, Friberg S, Hultborn KA. Male breast Department of Defense (DOD) experience. Proc
carcinoma. I. A study of the total material Annu Meet Am Soc Clin Oncol 1995;14:A140.
reported to the Swedish Cancer Registry 65. Ciatto S, Iossa A, Bonardi R, Pacini P. Male breast
1958–1967 with respect to clinical and histo- carcinoma: review of multicenter series of 150
pathologic parameters. Acta Oncol 1987;26: cases. Tumori 1990;76:555–8.
241–56. 66. Borgen PI, Senie RT, McKinnon WM, Rosen, PP.
49. Guinee VF, Olsson H, Moller T, et al. The progno- Carcinoma of the male breast: analysis of prog-
sis of breast cancer in males. Cancer 1993; nosis compared with matched female patients.
71:154–61. Ann Surg Oncol 1997;4:385–8.
50. Borgen PI, Wong GY, Vlamis V, et al. Current man- 67. Joshi MG, Lee AK, Loda M, et al. Male breast car-
agement of male breast cancer: a review of 104 cinoma: an evaluation of prognostic factors
cases. Ann Surg 1992;215:451–9. contributing to a poorer outcome. Cancer
51. Salvadori B, Saccozzi R, Manzari A, et al. Progno- 1996;77:490–8.
sis of breast cancer in males: an analysis of 170 68. Pivot X, Llombart-Cussac A, Rhor-Albarddo A, et
cases. Eur J Cancer 1994;30A:930–5. al. Clinical staging for male breast cancer: an
52. Cutuli B, Lacroze M, Dilhuydy JM, et al. Male adaptation of the international classification
breast cancer: results of the treatments and (TNM). Proc Annu Meet Am Soc Clin Oncol
prognostic factors in 397 cases. Eur J Cancer 1997;16:A655.
1995;31A:1960–4. 69. Lartigau E, El-Jabbour JN, Dubray B, Dische S.
53. McLachlan SA, Erlichman C, Liu FF, et al. Male Male breast carcinoma: a single centre report of
breast cancer: an 11 year review of 66 patients. clinical parameters. Clin Oncol 1994;6:162–6.
Breast Cancer Res Treat 1996;40:225–30. 70. Winchester DJ, Goldschmidt RA, Khan SJ, et al.
54. Donegan WL, Redlich PN, Lang PJ, Gall MT. Car- Flow cytometric and molecular prognostic
cinoma of the breast in males: a multi-institu- markers in male breast carcinoma patients
tional survey. Cancer 1998;83:498–509. [abstract]. Presented at the 46th Annual Cancer
252 BREAST CANCER

Symposium of the Society of Surgical Oncol- 84. Becher R, Höffken K, Pape H, Schmidt CG.
ogy; Los Angeles; 1993; March 18–21. Tamoxifen treatment before orchiectomy in
71. Yap HY, Tashima CK, Blumenschein GR, Eckles advanced breast cancer in men. N Engl J Med
NE. Male breast cancer: a natural history study. 1981;305:169–70.
Cancer 1979;44:748–54. 85. Ribeiro GG. Tamoxifen in the treatment of male
72. Winchester DJ. Male breast cancer. Semin Surg breast carcinoma. Clin Radiol 1983;34:625–8.
Oncol 1996;12:364–9. 86. Patterson JS, Batersby LA, Back BK. Use of
73. Pich A, Margaria E, Chiusa L, et al. DNA ploidy tamoxifen in advanced male breast cancer.
and p53 expression correlate with survival and Cancer Treat Rep 1980;64:801–4.
cell proliferative activity in male breast carci- 87. Jaiyesimi IA, Buzdar AU, Sahin AA, et al. Carci-
noma. Hum Pathol 1996;27:676–82. noma of the male breast. Ann Intern Med
74. Pich A, Margaria E, Chiusa L. Proliferative activity 1992;117;771–7.
is a significant prognostic factor in male breast 88. Kantarjian H, Yap H-Y, Hortobagyi G, et al. Hor-
carcinoma. Am J Pathol 1994;145:481–9. monal therapy for metastatic male breast can-
75. Visfelt J, Scheike O. Male breast cancer. I. Histo- cer. Arch Intern Med 1983;143:237–40.
logic typing and grading of 187 Danish cases. 89. Patel JK, Nemoto T, Dao TL. Metastatic breast
Cancer 1973;32:985–90. cancer in males: assessment of endocrine ther-
76. Anelli A, Anelli TF, Youngson B, et al. Mutations apy. Cancer 1984;53:1583–5.
of the p53 gene in male breast cancer. Cancer 90. Kraybill WG, Kaufman R, Kinne D. Treatment of
1995;75:2233–8. advanced male breast cancer. Cancer 1981;47:
77. Jonasson JG, Agnarsson BA, Thorlacius S, et al. 2185–9.
Male breast cancer in Iceland. Int J Cancer 91. Sandler B, Carman C, Perry RR. Cancer of the
1996;65:446–9. male breast. Am Surg 1994;60:816–20.
78. Hultborn R, Friberg S, Hultborn DA, et al. Male 92. Yap HY, Tashima CK, Blumenschein GR, et al.
breast carcinoma. II. A study of the total mate- Chemotherapy for advanced male breast can-
rial reported to the Swedish Cancer Registry cer. JAMA 1980;243:1739–41.
1958–1967 with respect to treatment, prognos- 93. Lopez M, DiLauro L, Papaido P, et al. Chemother-
tic factors and survival. Acta Oncol 1987;26: apy in metastatic male breast cancer. Oncology
327–41. 1985;452:205–9.
79. Schuchardt U, Seegenschmiedt MH, Kirschner MJ, 94a. Stierer M, Rosen H, Weitensfelder W, et al. Male
et al. Adjuvant radiotherapy for breast carci- breast cancer: Austrian experience. World J
noma in men: a 20-year clinical experience. Surg 1995;19:687–92.
Am J Clin Oncol 1996;19:330–6. 94b. Rayson D, Erlichman C, Wold LE, et al. Molecu-
80. Anelli TFM, Anelli A, Tran KN et al. Tamoxifen lar markers in male breast cancer. Proc Annu
administration is associated with a high rate of Meet Am Soc Clin Oncol 1997;16:A477.
treatment-limiting symptoms in male breast 95. Willsher PC, Leach IH, Ellis IO, et al. Male breast
cancer patients. Cancer 1994;74:74–7. cancer: pathological and immunohistochemi-
81. Ribeiro G, Swindell R. Adjuvant tamoxifen for cal features. Anticancer Res 1997;17:2335–8.
male breast cancer. Br J Cancer 1992;65:252–4. 96. Bines J, Goss B, Hussong J, et al. c-erbB2 and p53
82. Bagley CS, Wesley MN, Young RC, Lippman ME. overexpression as predictors of survival in
Adjuvant chemotherapy in males with cancer patients with male breast cancer. Proc Annu
of the breast. Am J Clin Oncol 1987;10:55–60. Meet Am Soc Clin Oncol 1997;16:A558.
83. Patel, HZ, Buzdar AU, Hortobagyi GN. Role of 97. Dawson PJ, Schroer KR, Wolman SR. Ras and p53
adjuvant chemotherapy in male breast cancer. genes in male breast cancer. Mod Pathol 1996;
Cancer 1989;64:1583–5. 9:367–70.
15
Estrogen Replacement Therapy
for Breast Cancer Survivors
WENDY R. BREWSTER, MD
PHILIP J. D ISAIA, MD

Ninety percent of women will live to the cli- agents and other drugs that cause amenorrhea
macteric age, compared to only 30 percent 200 in 84 percent of women aged 35 to 44 years.
years ago. Attrition and aging of ovarian folli- Other studies indicate that this treatment causes
cles results in termination of the maturation of permanent ovarian failure in 86 percent of
granulosa cells, which are responsible for estro- women > 40 years of age.2 As a result, a larger
gen production. Sources of estrogen in the pre- number of women will potentially be rendered
menopausal woman are several fold, including menopausal in the fourth, and fifth decades of
direct production of estradiol by the ovaries as their lives, which has serious consequences in
well as the extraglandular aromatization in adi- terms of the risk of cardiovascular disease and
pose cells of androstenedione created in the osteoporosis.
adrenal glands and ovary. The hallmark of The major concern of many physicians in
menopause is a drop in ovarian production of prescribing estrogen-replacement therapy
estriol and testosterone. Peripheral aromatiza- (ERT) for breast cancer survivors is the theory
tion of other steroids not produced by the that metastatic quiescent tumor foci might be
ovaries is an additional source of estrogen in all activated and the “fire” of breast cancer ignited
women. However, this source is not sufficient by the “fuel” estrogen. Other fears are that
in most women to prevent the symptoms char- estrogen might cause a second primary in the
acteristic of estrogen deprivation. already environmentally/genetically primed
Given the current population, 30 million contralateral breast or might change breast den-
women in the United States will spend approx- sity and mask new mammographic findings.
imately 40 percent of their lifetime in the post- These concerns are in part based upon epi-
menopausal period. These women have a life- demiologic studies demonstrating a relation-
time risk of one in eight of developing breast ship between duration of postmenopausal estro-
cancer. Thus, a considerable number of Ameri- gen replacement and breast cancer.3,4 Also,
can women are likely to have a history of breast surgical oophorectomy is beneficial in a subset
cancer treatment and at the same time be poten- of premenopausal breast cancer patients, and
tial candidates for hormone-replacement ther- estrogen withdrawal has also been observed to
apy. In the last decade, indications for promote regression of metastatic breast cancer
chemotherapy as adjuvant treatment to surgery lesions.5 Despite very limited clinical data to
have widened and now encompass many more support these concerns, it remains standard
premenopausal women.1 Adjuvant therapy for practice to prohibit breast cancer survivors
breast cancer includes the use of alkylating from receiving ERT.

253
254 BREAST CANCER

The well-substantiated benefits of estrogen raises the serum level of high-density lipoprotein
replacement therapy must be balanced against cholesterol, especially the HDL2 subfraction,
theoretical concerns. Arguments in support of and lowers the serum level of low-density
the safety of ERT are based on several natural lipoprotein cholesterol.6 Other less well-studied
experiments and observations, discussed in factors that may influence cardiovascular health
detail below. during treatment with estrogen, with or without
progestin, include beneficial effects on the circu-
BENEFITS OF ESTROGEN lation, blood pressure, coagulation, and fibrinol-
REPLACEMENT THERAPY ysis.7,8 Estrogen also has vasodilating properties
mediated by the generation of prostacyclin in
Over the last two decades, overwhelming evi- the cell membrane.
dence has been accrued demonstrating that Many epidemiological studies have found
postmenopausal estrogen replacement protects that postmenopausal women who use estrogen
against ischemic heart disease, osteoporosis, are at a much lower risk for coronary disease
deterioration in cognitive function, colorectal than are nonusers. Observational studies sug-
cancer, and provides relief from vasomotor gest a 50 percent reduction in the risk of coro-
symptoms and urogenital atrophy. Multimodal- nary heart disease among healthy post-
ity screening has resulted in an increase in the menopausal women taking oral estrogen.9
incidence of breast cancer diagnoses; this In 1981, Henderson and colleagues
increase, however, reflects more frequent detec- recruited over 8,000 women from a retirement
tion of early-stage breast cancer. Because community in Laguna Hills, California called
breast cancer survival is inextricably linked to Leisure World. This is a stable community and
early diagnosis, there are now more breast can- very few individuals were lost to follow-up. Of
cer survivors than ever. Morbidity and mortal- this cohort, 57 percent reported estrogen use,
ity associated with estrogen deprivation present 14 percent were current users at the time of the
serious health concerns. The risk/benefit ratio questionnaire, and 43 percent reported previous
of estrogen replacement therapy (ERT) is an use. The incidence of mortality from acute
appropriate consideration for all patients. myocardial infarction was statistically lower
among current users and those who had used
Coronary Artery Disease estrogen in the past compared to nonusers. The
relative risk was 0.59, with the 95 percent con-
Cardiovascular disease is the leading cause of fidence interval (CI) of 0.42 to 0.82.
mortality among women in the United States. Hunt10 reported on a cohort of 4,544 women
The number of deaths from diseases of the cir- who had taken hormone replacement therapy
culatory system in women in the United States continuously for at least 1 year at the time of
is greater than the number who die from can- recruitment. When compared with the general
cers of the breast, reproductive tract, and mater- female population, mortality rates for ischemic
nal morbidity combined. It is only during the heart disease among the cohort were signifi-
reproductive years that more women die from cantly lower, with a relative risk of 0.41 and a
malignancy than from cardiovascular disease. 95 percent CI of 0.2 to 0.61. Bush11 evaluated a
This is reversed past 60 years of age. cohort of 2,270 women, 593 of whom were
The endocrine influences of factors thought estrogen users. The age-adjusted relative risk of
to be contributors to the risk of cardiovascular death from cardiovascular disease was 0.34,
disease have been studied extensively. The liter- with the 95 percent CI of 0.12-0.81.
ature is vast and has been well summarized in Stampfer12 evaluated postmenopausal
several recent reviews. Unopposed estrogen estrogen therapy and cardiovascular disease in
Estrogen Replacement Therapy for Breast Cancer Survivors 255

the Nurses’ Health Study, with a 10-year fol- diovascular incidents were observed for HRT
low-up. Women currently using post- over ERT.
menopausal hormone therapy accounted for Grodstein14 evaluated the effect of com-
21.8 percent of the total follow-up time of bined estrogen and progestin use and the risk of
337,854 person-years. There was a reduction in cardiovascular disease in the Nurses’ Health
the age-adjusted relative risk of fatal cardio- Study. Among the 59,337 women enrolled,
vascular disease among current hormone there were 770 casualties of myocardial infarc-
users. In the same study, the age-adjusted risk tion or deaths from coronary artery disease.
of major coronary artery disease among cur- There was a marked decrease in the risk of
rent estrogen users was about half that of major coronary artery disease among women
women who had never used estrogen, with a who took estrogen with progestin compared to
relative risk of 0.51 p < .0001. For former that for women who did not use hormones. The
users, the age-adjusted relative risk (RR) was multivariate-adjusted relative risk was 0.39,
0.91. When this was adjusted for other risk fac- with the 95 percent CI of 0.19 to 0.78.
tors, the relative risk was 0.83. The relative risk
of fatal cardiovascular disease was decreased Osteoporosis
in both current and former users.
The above studies were all based on post- Postmenopausal women are at risk for loss of
menopausal estrogen use only. Given the fact cancellous bone in the vertebrae and other long
that current medical recommendations call for bones, which places them at increased risk for
the addition of a progestin to estrogen therapy fracture. Bone mineral density decreases
in nonhysterectomized women, there is the rapidly within 5 years of menopause due to
valid concern that progestin therapy may negate estrogen deficiency. This ultimately results in
the benefits gained by estrogen (Table 15–1). microarchitectural deterioration and a progres-
The investigators in the Postmenopausal sive increased fracture risk. Postmenopausal
Estrogen/Progestin Interventions (PEPI) trial untreated women may lose 35 percent of their
examined this issue.13 They found, as had been cortical bone and up to 50 percent of their tra-
confirmed in numerous previous studies, that becular bone. It is estimated that 1.2 million
unopposed estrogen decreased the risk factors major fractures per year in the United States in
for cardiovascular disease. However, estrogen women are related to osteoporosis. Fifteen per-
given with medroxyprogesterone acetate or cent of postmenopausal women will suffer
micronized progesterone hormone-replace- wrist fractures, and an even larger number will
ment therapy (HRT) was associated with lower incur spinal compression fractures. Compres-
fibrinogen levels and improved lipoprotein sion fractures of the vertebral bones may result
profiles. No adverse effects on the rate of car- in loss of stature, pulmonary restriction, and

Table 15–1. EPIDEMIOLOGIC STUDIES OF THE CARDIOVASCULAR BENEFITS OF


POSTMENOPAUSAL ESTROGEN AND PROGESTERONE USE

Study Design Number Results

Falkeborn et al.41 Prospective 227 MI cases RR = 0.74 ever estrogen only


23,174 women RR = 0.50 ever combined therapy
Psaty et al42 Case-control 502 MI cases RR = 0.69 estrogen alone
1,193 controls RR = 0.68 current combined therapy
Grodstein et al14 Prospective 770 MI cases RR = 0.60 current estrogen alone
59,337 women RR = 0.39 current combined therapy

MI= myocardial infarction; RR = relative risk.


256 BREAST CANCER

decreased ambulation. An estimated 40 percent subjects were initially free of Alzheimer’s dis-
of the women who will live to the age of 80 ease, Parkinson’s disease, and stroke and were
years will develop spinal fractures and 33 per- part of a longitudinal study of aging and health
cent will experience a hip fracture. in a New York community. Overall, 158 (12.5
Of concern is the morbidity and mortality percent) reported taking estrogen after the
associated with hip fractures in older women. onset of menopause. The age of onset of
Within this group, 12 to 20 percent will die Alzheimer’s disease was significantly later in
within 6 months of the fracture, and half of the women who had taken estrogen than in those
survivors require long-term nursing care. who did not, 78 years versus 73 years. Even
Osteoporotic fractures in the United States after adjustment for differences in education
resulted in health care costs of $7 billion in and ethnic origin, the relative risk of
1986. This is estimated to increase to as much Alzheimer’s disease was significantly reduced
as $62 billion by the year 2020. in estrogen users over nonusers: 0.4, with a 95
percent CI of 0.22 to 0.85.
Alzheimer’s Disease Even among postmenopausal women who
are not demented, ERT may help maintain cog-
As the population ages, Alzheimer’s disease has nitive function.17 Estrogen appears to have a
emerged as a major health problem. After the specific effect on verbal memory skills in
age of 65 years, the prevalence of dementia and healthy postmenopausal women.18,19
Alzheimer’s disease doubles every 5 years; 30 The emotional, physical, social, and finan-
to 50 percent of women older than 83 years cial costs of Alzeimer’s disease to patients,
may suffer from dementia of some sort. families, caregivers, and society are tremen-
Laboratory studies suggest that estrogen dous. The estimated total cost of the disease in
may affect Alzheimer’s disease through several 1991 was estimated to be $173,932 per case.
mechanisms. Estrogen has been shown to The estimated prevalence cost for both men and
improve regional cerebral blood flow and to women for that year was $67.3 billion.20 The
increase glucose utilization. It can also stimu- economic cost of care alone is greater than the
late neurite growth and synapse formation in cost of care for heart disease and cancer com-
vitro. Under some circumstances, estrogen may bined. If the use of estrogen could delay the
modify neural sensitivity to neurotrophin and onset of Alzheimer’s disease by several years,
play a role in the reparative neuronal response there would be a substantial saving in both
to injury. One key histologic feature of emotional and financial costs.
Alzheimer’s disease is the deposition of beta-
amyloid protein in cores of neuritic plaques. Colorectal Cancer
Estrogen may promote the breakdown of the
amyloid precursor protein to fragments less There have been > 20 retrospective studies of
likely to accumulate as beta amyloid. Acetyl- the risk of colon cancer and ERT, with more
choline is a key neurotransmitter in learning than 70 percent of these reports illustrating a
and memory. Estrogen affects several neuro- statistically significant reduction in incidence
transmitter systems, including the cholinergic with users versus nonusers. One proposed
system. Finally, estrogen may modify inflam- mechanism affecting this protection is that
matory responses postulated to participate in estrogen reduces the concentration of bile
neuritic plaque formation.15 acids, and may limit carcinogenic action to the
Tang and colleagues examined the effect of colon mucosa. It has been demonstrated that
a history of estrogen use on the development of bile acid concentrations are higher in colon
Alzheimer’s disease in 1,200 women.16 These cancer cases than in control subjects, and it is
Estrogen Replacement Therapy for Breast Cancer Survivors 257

known that estrogen decreases bile acid synthe- thirds of women experiencing symptoms classi-
sis and secretion.21 Estrogen receptors are pre- fied as moderate to severe.24 This effect may be
sent in both normal and cancerous colon compounded by tamoxifen therapy, which also
mucosal cells, and there is laboratory evidence leads to vasomotor instability.
to suggest that estrogen may inhibit the growth
of colon cancer cells.22 Urogenital Atrophy
Calle and colleagues23 investigated the rela-
tionship between postmenopausal estrogen use Because the vagina and urethra share a com-
and fatal colorectal cancer in a large prospective mon embryologic origin, it is believed that
study of adults in the United States. Eight hun- estrogen deficiency causes atrophy of both
dred and seventy-nine colon cancer case patients structures. Atrophy of the vaginal epithelium
were compared to 421,476 noncase subjects. may cause vaginal itching, dryness, and dys-
Ever use of ERT was associated with a signifi- pareunia, with resulting inflammation. One
cantly decreased risk of fatal colon cancer (RR = effect of estrogen deficiency is to cause
0.71; 95% CI = 0.61 to 0.83). Reduction in risk changes in the vaginal pH, which predispose
was strongest among current users (RR = 0.55; women to urinary tract infections that cause
95% CI = 0.40 to 0.76) compared to former urgency, incontinence, frequency, nocturia, and
estrogen users. There was a significant trend of dysuria. The loss of estrogen on periurethral tis-
decreasing risk with increasing years of estrogen sues will contribute to pelvic laxity and stress
use among all users (p = .0001). Those women incontinence. Recurrent urinary tract infections
who used estrogen for ² 1 year had a RR = 0.81, can be prevented with systemic estrogen ther-
whereas users of ³ 11 years had a RR of 0.54 apy, and low-dose topical estrogen is effective
(95 percent CI = 0.39 to 0.76). These associa- in managing atrophic vaginitis. Estrogen pro-
tions were not altered in multivariate analyses vides relief of these symptoms and may protect
controlling for age, race, parental history of against recurrent urinary tract infections.
colon cancer, body mass index, exercise, parity,
type of menopause, age of menopause, oral con- EXPOSURE TO EXOGENOUS OR
traceptive pill use, aspirin use, and smoking. ENDOGENOUS ESTROGEN DURING
BREAST CANCER DEVELOPMENT
Vasomotor Instability
The decision whether or not to take hormone
The menopausal state most commonly produces replacement remains difficult for the post-
vasomotor instability and genital organ atrophy. menopausal woman because of conflicting risks
Vasomotor symptoms affect 70 percent of post- and benefits and is even more difficult for the
menopausal women but only about 30 percent breast cancer survivor for whom there is even
seek medical assistance. For 25 percent of less data. One can therefore analyze situations
menopausal women, these symptoms may per- in which women are inadvertently exposed to
sist for > 5 years and may be lifelong in others. exogenous or endogenous estrogen at a time
Vasomotor instability is more commonly termed when they may have been harboring subclinical
“hot flushes” or “hot flashes.” The frequency, breast cancer. Does such exposure adversely
severity, or diurnal variation with which hot affect survival outcome for these patients?
flushes occur can result in significant disrup- Such situations include those in which the
tions of sleep and daytime function. Menopausal diagnosis of breast cancer is made in post-
symptoms are the most common side effect menopausal women receiving ERT at the time of
associated with the use of adjuvant chemother- diagnosis or in whom the diagnosis is made in
apy for breast cancer, with approximately two- pregnancy or during lactation, or in those women
258 BREAST CANCER

with a history of oral contraceptive pill use The physiologic changes and engorgement
around the time of diagnosis of breast cancer. that occur in the breast during pregnancy often
hinder early detection of breast cancer. This
Breast Cancer in Women on results in a diagnosis at more advanced stages
Estrogen Replacement Therapy in pregnant and lactating women. Comparisons
to nonpregnant women matched for similar age
Bergkvist and co-workers25 compared 261 stage of breast cancer and reproductive capac-
women who developed breast cancer while on ity do not suggest a worse prognosis for the
ERT to 6,617 breast cancer patients who had no pregnant patients with breast cancer.28,29 von
recorded treatment with estrogen. The relative Schoultz30 performed a comparison of women
survival rate over an 8-year period was higher in diagnosed with breast cancer 5 years before
the breast cancer patients who had previously pregnancy to women without a pregnancy dur-
received ERT. This corresponded to a 32 percent ing the same time period. There was no survival
reduction in excess mortality. Gambrell,26 in a disadvantage to the women who were pregnant
prospective study, also evaluated the effect on 5 years prior to the diagnosis of breast cancer.
survival in breast cancer patients diagnosed while These and other studies have discouraged the
on ERT. Mortality was 22 percent among those practice of prohibiting breast cancer survivors
diagnosed with breast cancer while on ERT com- from becoming pregnant on clinical grounds.
pared to 46 percent among those who had never Subsequent pregnancies do not negatively
received hormone replacement. Henderson and affect survival outcomes.
colleagues27 observed a 19 percent reduction in Anderson and colleagues31 reported their
breast cancer mortality among 4,988 previous experience at the Memorial Sloan Kettering
ERT users, compared to 3,865 nonusers who Cancer Center with breast cancer in women
subsequently developed this disease. < 30 years of age. Two hundred and twenty-
seven cases were identified, of whom 22 had
Breast Cancer pregnancy-associated breast cancer. The
Associated with Pregnancy authors confirmed that pregnancy-associated
breast cancers were usually larger and present
Pregnancy coincident with, or subsequent to, the in more advanced stages at the time of diagno-
detection of breast cancer provides another excel- sis, compared to a similar group who were not
lent opportunity to evaluate the outcome of breast pregnant. However, the survival probability for
cancer patients inadvertently exposed to high lev- women with early stage disease was indepen-
els of estrogen at times when they were harboring dent of pregnancy status.
occult disease. During pregnancy, the serum lev- The experience of women who have com-
els of estriol increase 50-fold. Only 0.5 to 4 per- pleted term pregnancies after treatment of
cent of all breast cancers are diagnosed during antecedent breast cancer is another situation
pregnancy. Because the average breast cancer that deserves analysis. There are inherent biases
remains occult in the breast some 5 to 8 years associated with evaluation of this particular
prior to diagnosis, some authors include in this group of subjects. This cohort is representative
category women in whom a diagnosis of breast of the young women who did well after primary
cancer has been made within 12 months of deliv- breast cancer therapy; since pregnancy data is
ery. The outcome in women with subclinical not uniformly coded in cancer registry data-
breast cancers exposed to elevated levels of pro- bases, the true denominator of postbreast can-
gesterone and estrogen under these circumstances cer pregnancies is unknown. Clark32 reported a
could provide insight into the influence of these 71 percent 5-year survival in a series of 136
hormones on the malignant disease process. women with pregnancies after breast cancer
Estrogen Replacement Therapy for Breast Cancer Survivors 259

(stages I to III). Equivalent survival outcomes als of the German Breast Cancer Study Group.
were reported by von Schoultz30 for breast can- One hundred and thirty-seven OCP users (32.5
cer patients with no subsequent pregnancy, percent) were younger than those who did not
compared to those who became pregnant within use OCP (mean age 41.5 years versus 45 years).
5 years of their diagnosis. Noteworthy was the fact that the percentage of
patients with smaller tumors was higher in the
Breast Cancer in group of OCP users. No significant effect of
Oral Contraceptive Pill Users OCP use on either disease-free or overall sur-
vival could be demonstrated in univariate and
Given the long natural history of this neoplasm, multivariate analyses after adjustment for tumor
it is certain that a large number of patients sub- size and other prognostic factors.
sequently diagnosed with breast cancer have
used oral contraceptive pills (OCP) during the STUDIES ON
genesis and progression of their malignant dis- ESTROGEN-REPLACEMENT THERAPY
ease process; they are another group that IN BREAST CANCER SURVIVORS
deserves examination.
Rosner33 evaluated 347 women < 50 diag- DiSaia36 reported on 71 breast cancer survivors
nosed with breast cancer, of whom 112 were who received ERT. There was no exclusion based
OCP users. The distribution of tumor size, on time interval from diagnosis, stage, age,
estrogen-receptor status, and family and repro- receptor status, or lymph node status. Women
ductive history was the same between the two received combination therapy with progestin only
cohorts. There was no difference in disease-free if they had not previously undergone hysterec-
survival or survival between the two groups. tomy. Later, the author reported a comparison of
Women who used OCP within a year of diag- 41 of these ERT survivors to 82 non-ERT breast
nosis of their breast cancer had a similar sur- cancer subjects, matched for both age and stage
vival to those who had discontinued use > 1 of disease.37 Survival analysis did not indicate a
year before. There was no difference in survival significant difference between the two groups.
among those who used OCP ³ 10 years prior to An updated series of 145 patients who received
their diagnosis of breast cancer. ERT for at least 3 months after diagnosis has
Schonborn and colleagues34 evaluated the identified 13 recurrences. The duration of estro-
influence of a positive history of OCP use on gen use prior to the diagnosis of recurrent breast
survival. Four hundred and seventy-one breast cancer ranged from 4 months to 11.5 years (Fig-
cancer patients were investigated. Two hundred ures 15–1 and 15–2).
and ninety-seven patients (63 percent) had used Other authors have reported their experi-
OCP during any period of their life, and 92 (20 ence of ERT in breast cancer survivors. Eden38
percent) still used them at the time of diagnosis. reported six recurrences among 90 women
Sixty months after diagnosis, the OCP users receiving ERT. These ERT users were matched
had a significantly increased overall survival two to one with control subjects with no history
(p = .037). Survival rates amounted to 79.5 per- of hormone use after diagnosis of breast cancer.
cent and 70.3 percent for OCP users and The recurrence rate was 7 percent in the ERT
nonusers, respectively. users and 30 percent in the non-ERT users.
Sauerbrei35 investigated the relationship Bluming39 reported on 155 breast cancer
between OCP use and standard prognostic fac- patients who received ERT for between 1 and
tors, and the effect of OCP use on disease-free 56 months, among whom 7 recurrences were
survival and overall survival, in 422 pre- identified. The only published prospective ran-
menopausal node-positive patients from two tri- domized trial is being undertaken by Vas-
260 BREAST CANCER

100
90
Stage 89
0
80
I/II

Number of Patients
70 III/IV
60 Unknown

50
40
30 24
20 13 13
10 6
0 50 100 150 0 0 0
Months 0
Recurrence No Recurrence
Figure 15–1. Subjects with recurrent breast cancer. The inter-
val between initiation of use of estrogen replacement therapy Figure 15–2. Comparison by known stage of subjects who
and diagnosis of recurrent breast cancer. received estrogen replacement therapy.

silopoulou-Sellin.40 Subjects are randomized to CONCLUSION


either a placebo or ERT without a progesta- The fear that administration of estrogen to
tional agent. Ninety women have been random- women with a history of breast cancer will
ized and 49 have received ERT for a minimum result in the activation of quiescent metastatic
of 2 years. No breast cancer recurrences have foci, as well as the climate of medical litigation,
been observed in the ERT arm. The single are the basis of much of the reluctance of physi-
recurrence was in the placebo arm. cians to prescribe this agent. The standard of
The series discussed above are representative care no longer supports prophylactic oophorec-
of the reported experience of ERT in 499 female tomy in young women who do not become
breast cancer survivors (Table 15–2). This group amenorrheic after cytotoxic therapy. In addition,
of women is very heterogeneous with respect to many women continue to menstruate regularly
breast cancer stage, the interval between diagno- after treatment and may even complete pregnan-
sis of breast cancer and initiation of ERT, the cies. If castration and pregnancy termination are
hormonal combinations prescribed, estrogen- not routinely recommended, why then should
receptor status, and finally in the duration of use the replacement of estrogen at a much lower
of estrogen. Despite these limitations, it remains dose than is physiologic be prohibited?
obvious that the use of estrogen is not associated Fifty-year-old women have a 13 percent life-
with a rash of occurrences. Overall, the data do time probability of developing breast cancer and
not suggest that ERT has an adverse effect on a 3 percent probability of dying from it; they
breast cancer outcome. have a 46 percent chance of developing coronary

Table 15–2. ESTROGEN-REPLACEMENT THERAPY IN BREAST CANCER SURVIVORS

Study No. of Patients Stage of Disease Duration of ERT Recurrences

Stoll43 Unknown Early stage 3–6 mo None


Wile et al44 25 All stages 24–82 mo 3
Powles et al45 35 All stages 1–44 mo 8
Eden et al38 90 Local 4–144 mo 6
Vassilopoulou-Sellin et al40 49 0–III 24–142 mo 0
(ER negative only) (oral or vaginal estrogen only)
Bluming et al39 155 Local 1–56 mo 7
Brewster et al* 145 All stages 3–144 mo 13

*In press.
ERT=estrogen-replacement therapy; ER=estrogen replacement.
Estrogen Replacement Therapy for Breast Cancer Survivors 261

heart disease and a 31 percent probability of 9. Grady D, Rubin SM, Petitti DB, et al. Hormone
dying from it. While breast cancer claims 43,000 therapy to prevent disease and prolong life in
postmenopausal women. Ann Intern Med
lives per year in the United States, coronary
1992;117:1016–37.
heart disease will kill approximately 233,000 10. Hunt K, Vessey M, McPherson K, Coleman M.
women annually. Nearly 65,000 women die each Long-term surveillance of mortality and can-
year from the complications of hip fracture. cer incidence in women receiving hormone
No guarantee can be made that ERT will be replacement therapy. Br J Obstet Gynaecol.
1987;94:620–35.
accompanied with freedom from recurrent 11. Bush TL, Barrett-Connor E, Cowan LD, et al. Car-
breast cancer, because some women will have diovascular mortality and noncontraceptive use
recurrent disease coincident with renewed hor- of estrogen in women. Circulation 1987;75:
monal exposure. However, should one discuss 1102–9.
the risks and benefits of estrogen replacement 12. Stampfer MJ, Colditz GA, Willet WC, et al. Post-
menopausal estrogen therapy and cardiovascular
therapy with patients to help them make an disease. Ten-year follow-up from the Nurses’
informed decision? Health Study. N Engl J Med 1991;325:756–62.
13. The Writing Group for the PEPI Trial. Effects of
REFERENCES estrogen or estrogen/progestin regimens on
heart disease risk factors in postmenopausal
1. Early Breast Cancer Trialists’ Collaborative women. The Postmenopausal Estrogen/Prog-
Group. Systemic treatment of early breast can- estin Interventions (PEPI) Trial. JAMA 1995;
cer by hormonal, cytotoxic, or immune ther- 274:199–208.
apy: 133 randomized trials involving 31,000 14. Grodstein F, Stampfer MJ, Colditz GA, et al. Post-
recurrences and 24,000 deaths among 75,000 menopausal hormone therapy and mortality. N
women. Lancet 1992;339:1–15, 71–85. Engl J Med 1997;336:1769–75.
2. Mehta RR, Beattie CW, Das Gupta T. Endocrine 15. Paganini-Hill A, Henderson VW. Estrogen replace-
profile in breast cancer patients receiving ment therapy and risk of Alzheimer’s disease.
chemotherapy. Breast Cancer Res Treat 1991; Arch Intern Med 1996;156:2213–17.
20:125–32 16. Tang MX, Jacobs D, Stern Y, et al. Effect of
3. Colditz GA, Hankinson SE, Hunter DJ, et al. The oestrogen during menopause on risk and age at
use of estrogens and progestins and the risk of onset of Alzheimer’s disease. Lancet 1996;348:
429–32.
breast cancer in postmenopausal women. N Engl
17. Jacobs DM, Tang MX, Stern Y, et al. Cognitive
J Med 1995;332:1589–93.
function in nondemented women who took
4. Stanford JL, Weiss NS, Voigt LF, et al. Combined
estrogen after menopause. Neurology 1998;50:
estrogen and progestin hormone replacement
368–73.
therapy in relation to risk of breast cancer in
18. Kampen DL, Sherwin BB. Estrogen use and ver-
middle age women. JAMA 1995;274:137–41. bal memory in healthy postmenopausal
5. Dhodapkar MV, Ingle JN, Ahmann DL. Estrogen women. Obstet Gynecol 1994;83:979–83.
replacement therapy withdrawal and regression 19. Rice MM, Graves AB, McCurry SM, Larson EB.
of metastatic breast cancer. Cancer 1995;75: Estrogen replacement therapy and cognitive
43–6. function in postmenopausal women without
6. Sacks FM, Walsh BW. The effects of reproductive dementia. Am J Med 1997;103:26S–35S.
hormones on serum lipoproteins: unresolved 20. Ernst RL, Hay JW. The US economic and social
issues in biology and clinical practice. Ann NY costs of Alzheimer’s disease revisited. Am J
Acad Sci 1990;592:272–85. Public Health 1994;84:1261–4.
7. Gebera OC, Mittleman MA, Walsh BW, et al. Fib- 21. Davis RA, Fern F Jr. Effects of ethinyl estradiol
rinolytic potential is significantly increased by and phenobarbital on bile acid synthesis and
oestrogen treatment in postmenopausal women biliary bile acid and cholesterol excretion. Gas-
with mild dyslipidemia. Heart 1998;80:235–9. troenterology 1976;70:1130–5.
8. Koh KK, Mincemoyer R, Bui MN, et al. Effects of 22. Lointier P, Wildrick DM, Bowman BM. The
hormone-replacement therapy of fibrinolysis effects of steroid hormones on a human cancer
in postmenopausal women. N Engl J Med cell line in vitro. Anticancer Res 1992;12:
1997;336:683–90. 1327–34.
262 BREAST CANCER

23. Calle EE, Miracle-McMahill HL, Thun MJ, Heath effect of oral contraceptive use on the progno-
CW Jr. Estrogen replacement therapy and the sis of node positive breast cancer patients. Ger-
risk of fatal colorectal cancer in a prospective man Breast Cancer Study Group. Eur J Cancer
cohort of postmenopausal women. J Natl Can- 1998 Aug;34(9):1348–51.
cer Inst 1995;87:517–23. 36. DiSaia PJ, Odicino F, Grosen EA, et al. Hormone
24. Canney PA, Hatton MQF. The prevalence of replacement therapy in breast cancer [letter].
menopausal symptoms in patients treated for Lancet 1993;342:1232.
breast cancer. Clin Oncol 1994;6:297–9. 37. DiSaia PJ, Grosen EA, Kurosaki T, et al. Hormone
25. Bergkvist L, Adami HO, Persson I, et al. Progno- replacement therapy in breast cancer survivors.
sis after breast cancer diagnosis in women A cohort study. Am J Obstet Gynecol 1996;
exposed to estrogen and estrogen progesterone 174:1494–8.
replacement therapy. Am J Epidemiol 1992; 38. Eden JA, Bush T, Nand S, Wren BG. A case con-
130:221–8. trol study of combined continuous estrogen-
26. Gambrell DR. Proposal to decrease the risk and progestin replacement therapy among women
improve the prognosis in breast cancer. Am J with a personal history of breast cancer.
Obstet Gynecol 1984;150:119–28. Menopause 1995;2:67–72.
27. Henderson BE, Paganini-Hill A, Ross RK. 39. Bluming AZ, Waisman JR, Dosik GM. Hormone
Decreased mortality in users of estrogen replace- replacement therapy (HRT) in women with pre-
ment therapy. Arch Intern Med 1991;151:75–8. viously treated primary breast cancer. Update III.
28. Holleb AI, Farrow JH. The relation of carcinoma Proc Am Soc Clin Oncol 1997;16A:463,131a.
of the breast and pregnancy in 283 patients. 40. Vassilopoulou-Sellin R, Therriault R, Klein MY.
Surg Gynecol Obstet 1962;115:65. Estrogen replacement therapy in women with
29. Nugent P, O’Connell TX. Breast cancer and preg- prior diagnosis and treatment for breast cancer.
nancy. Arch Surg 1985;120:1221–4. Gynecol Oncol 1997;65:89.
30. von Schoultz E, Johansson H, Wilking N, 41. Falkeborn M, Persson I, Adami HO, et al. The risk
Rutqvist LE. Influence of prior and subsequent of acute myocardial infarction after oestrogen
pregnancy on breast cancer prognosis. J Clin and oestrogen-progestogen replacement. Br J
Oncol 1995;13:430–4. Ob Gyn 1992;99:821–8.
31. Anderson BO, Petrek JZ, Byrd DR, et al. Preg- 42. Psaty BM, Heckbert SR, Atkins D, et al. The risk
nancy influences breast cancer stage at diagno- of myocardial infarction associated with the
sis in women 30 years of age and younger. Ann combined use of estrogens and progestins in
Surg Oncol 1996;3:204. postmenopausal women. Arch Intern Med
32. Clark RM, Chua T. Breast cancer and pregnancy: 1994;154:1333–9.
the ultimate challenge. Clin Oncol (R Coll 43. Stoll BA. Hormone replacement therapy in
Radiol) 1989;1:11–8. women treated for breast cancer. Eur J Cancer
33. Rosner D, Lane W. Oral contraceptive use has no Clin Oncol 1989;25:1909–13.
adverse effect on the prognosis of breast can- 44. Wile AG, Opfell RW, Margileth DA. Hormone
cer. Cancer 1986;57:591–6. replacement therapy in previously treated breast
34. Schonborn I, Nischan P, Ebeling K. Oral contra- cancer patients. Am J Surg 1993;165:372–5.
ceptive use and the prognosis of breast cancer. 45. Powles TP, Casey S, O’Brien M, Hickish T. Hor-
Breast Cancer Res Treat 1994;30:283–92. mone replacement after breast cancer. Lancet
35. Sauerbrei W, Blettner M, Schmoor C, et al. The 1993;342:60.
16
Surveillance of the
Breast Cancer Patient
JANARDAN D. KHANDEKAR, MD

In recent years, extensive clinical trials have DEFINITIONS


established the roles for conservative surgery,
radiation, and adjuvant chemo/hormonal ther- It will be useful to review some of the terms
apy in the primary therapy of breast cancer. It used in analyzing the surveillance data. The
may seem self-evident that repeated postopera- decision matrix is a term most commonly
tive contact between cancer patients and their applied to the simple decision of whether the
physicians, that is, follow-up, is a good thing. disease is present (D+) or absent (D-) when
Follow-up practice patterns vary greatly, with the test is abnormal, that is, positive (T+) or
some oncologists frequently following their normal, that is negative (T–). When these two
breast cancer patients with various intensive binary results are plotted on a 2 ´ 2 table, four
investigation and others only doing sporadic possible combinations form the ratios shown
follow-up. The possible beneficial effects of below.1–3
follow-up include: The true positive (TP) ratio represents the
proportion of positive tests in all patients who
1. Management of postsurgical complications. have the disease. The ratio therefore expresses
This is essential and need not be elaborated the sensitivity of the test and can be express-
upon here. ed as:
2. Early detection of recurrence or of new
primaries. diseased subjects with positive test
sensitivity = ´ 100
3. Reassuring patients. This can be a double- all diseased subjects tested
edged sword, as some patients are reassured
by the process while others are made anx- More simply stated, sensitivity is deter-
ious by an impending visit to the physician. mined by the false-negative (FN) ratio, which is
4. Measurement of quality control of out- the proportion of negative tests in all patients
comes. Participation in clinical trials or with the disease. The TP and the FN ratio then
American College of Surgeons-sponsored represent the sensitivity of the test.
audits can be helpful. The false-positive (FP) ratio is the propor-
tion of positive tests in all patients who do
This chapter evaluated criteria for follow-up not have the disease. The true-negative (TN)
and provides background on the principal of ratio is the proportion of negative tests in all
screening and economic analysis. patients who do not have the disease. The FP

263
264 BREAST CANCER

and TN ratios then express specificity, defin- The PV of a negative test can be calculated
ed as: as follows:

nondiseased subjects with negative test number (or proportion) of


sensitivity = ´ 100 nondiseased persons with positive test
all nondiseased subjects tested PV = ´ 100
total number (or proportion) of
The sensitivity of a test under consideration persons with positive test
is usually determined by evaluating its efficacy
against a known standard. Depending on the sen- The usefulness of a diagnostic test is there-
sitivity of that standard, the sensitivity can be fore directly proportional to the prevalence of
spuriously high or low. For example, the sensi- the target disease in the population. The FP rate
tivity of a bone scan is usually evaluated in rela- of a test is usually constant and is often related
tion to radiography, a technique which itself is to the test and not the disease itself. Therefore,
not very sensitive. Therefore, the sensitivity of when the prevalence of the target disease is low,
the bone scan is very high, in the vicinity of the PV of the positive test is also low since FP
about 99 percent. However, if in the future even is constant. On the other hand, if there is a
more sensitive tests for detecting bone metasta- higher prevalence of the disease, the PV of a
sis become available, such as use of either better positive test will also be high.
imaging techniques and/or polymerase chain The evaluation of various diagnostic tests
reaction-based molecular assays, the sensitivity are also affected by various biases that signifi-
of the bone scan will decrease. The specificity of cantly affect the interpretation of the data.
the test is also dependent upon the FP ratio. Gen- These are outlined below.
erally, the FP rate increases with more data, and
the initial specificity of a given diagnostic test Lead-Time Bias
decreases with time. These factors then govern
The apparent increased duration of survival sim-
the sensitivity and specificity of various diag-
ply reflects a longer time that the recurrence was
nostic tests. When sensitivity and specificity of a
clinically known but there is no true gain in
test are determined, it is possible to calculate the
longevity. In other words, there is an illusion of
predictive value (PV).
an increased survival because of the longer dura-
The positive PV is defined as the likeli-
tion of observation but there is no impact on
hood that a subject yielding a positive test
mortality rate. It is because of this consideration
actually has the disease. Conversely, a nega-
that reduction of mortality rate has become the
tive PV indicates the likelihood that a subject
“gold standard” in evaluating the impact of a
with a negative test does not have the disease.
diagnostic or therapeutic intervention.
This likelihood is related to the actual preva-
lence of disease in the total population. More
Length-Time Bias
simply stated, PV for positive tests can be
defined as the percentage of time that a posi- An event such as cancer detected in an asymp-
tive test will detect the diseased individual. tomatic phase often has an indolent course and
The PV of a positive test can be calculated as is therefore detected at the time of evaluation
follows:4 rather than between the visits. Cancers that are
aggressive will often present with symptoms in
number of diseased subjects (or proportion) the intervening visits, creating the illusion that
with positive test more intensive surveillance would have
PV = ´ 100
total number (or proportion) of subjects resulted in detecting of the disease earlier and a
with positive test better outcome.
Surveillance of the Breast Cancer Patient 265

EVALUATION OF VARIOUS TESTS terns of recurrence and methods of detection.7


USED IN BREAST CANCER In 79 of 87 patients, recurrence was detected
by symptoms such as pain or shortness of
The American Society of Clinical Oncology breath, while physical examination detected an
(ASCO) established an expert panel to evaluate additional 5 patients with recurrence. Review
the use of various tests in breast cancer.5 The of the literature indicates that only 12 to 22
panel modified the scale developed by the percent of recurrences occur in truly asympto-
Canadian Taskforce on Periodic Health Exami- matic women.8–14 In the prospective intergroup
nation to evaluate various tests (Table 16–1).6 for cancer care evaluation trial (GIVIO), 31
There are only two prospective randomized tri- percent of the recurrences were detected in
als that have evaluated the impact of a multi- asymptomatic patients in the intensely investi-
tude of surveillance tests on the overall survival gated group compared to 21 percent of recur-
and quality of life in breast cancer patients. rences in the control group.11 However, there
They fulfill the criteria of providing Level 1, was no effect on survival in patients detected in
that is, highest level of evidence. However, the asymptomatic phase. It has been argued
beginning with studies published in 1979 by that patients with local recurrence only those
Winchester and colleagues,7 a large database who undergo aggressive therapy have a 50 per-
has been developed that has retrospectively cent 5-year survival, and may therefore have an
analyzed the value of various diagnostic tests in improved outcome. However, it can be argued
the follow-up of breast cancer patients. These that patients with local recurrence may have
studies have tried to answer some of the fol- more biologically inert disease, as these were
lowing questions: retrospective studies. Dewar and Kerr in an
English study reported that of 546 breast can-
1. Do the available tests diagnose early asymp- cer patients followed with 6,863 clinic visits,
tomatic recurrence in breast cancer? If so, only 1 percent of the visits were associated
which tests are useful? with recurrences that were curable.14 These
2. Does early detection and recurrence result authors have therefore questioned even routine
in better therapy and thus improve quality physical examination. In their studies, recur-
and quantity of survival? (This is the most rences were found five times more often dur-
important question). ing spontaneous visits than during routine vis-
3. What is the cost-benefit analysis for the pos- its, illustrating the lead-time bias. Dewar and
sible improved quality or quantity of life? Kerr suggest that negative physical examina-
tions may give false assurance to patients,
Winchester and colleagues analyzed 87 leading to an even further delay in diagnosis of
patients with recurrent breast cancer for pat- a recurrence.

Table 16–1. MODIFIED CANADIAN CRITERIA FOR EVALUATING DIAGNOSTIC TEST EVIDENCE

Level of Evidence Type of Evidence for Recommendation

Level 1 Meta-analysis or large high-powered concurrently controlled studies with a primary objective to evaluate
(highest level) the utility of a given test
Level 2 Prospective clinical trials designed to test given hypothesis
Level 3 Large size retrospective trials
Level 4 Similar to Level 3, but even less reliable. Comparative and correlative descriptive and case studies can
be included.
Level 5 Case reports and clinical examples

Adapted from Khandekar JD. Preoperative and postoperative follow up of cancer. In: Winchester DJ, Scott Jones R, Murphy GP, editors. Surgical
oncology for the general surgeon. Philadelphia, PA: Lippincott, Williams and Wilkins; 1999.p. 43–54.
266 BREAST CANCER

Scanlon and colleagues carefully analyzed eases and/or can develop new problems. There-
the information from the last examination to fore, an occasional blood test may be warranted.
recurrence in 93 patients.8 They reported that
43 percent of recurrences were detected within Tumor Markers: CA-15-3 and CEA
3 months of the last examination, 64 percent
within 6 months, and 94 percent within 1 year. Hayes and colleagues defined the marker that
They therefore recommended that examina- represents qualitative or quantitative alteration or
tions be conducted every 3 months for the first deviation from normal of a molecule, substance,
2 to 3 years after primary therapy for breast or process that can be detected by some type of
cancer, and then at a reduced interval. assay.16 This includes measurement of a gene,
The vast majority of recurrences are RNA, a product such as protein, carbohydrate, a
detected by history and physical examination. lipid, or a process such as vascular density.
Although the impact of such detection on over- Markers used in follow-up are considered below.
all survival is unknown, it may have the psy-
chological benefit of reassuring the patient of CA-15-3
having had a contact with their physician. Since
the cost of such surveillance is approximately The CA-15-316 marker measures the serum level
$150 per annum per patient and allows evalua- of a mucin-like membrane glycoprotein that is
tion of other parameters such as the effects of shed from the tumor cells into the blood stream.
primary therapy, physical and psychosocial Recently, the Food and Drug Administration
rehabilitation, and detection of contralateral approved the Truquant assay which uses a mon-
primary, the current author agrees with ASCO’s oclonal antibody, CA-27-29, to measure CA-15-
recommendation that the patients should be 3-like antigen. This marker is elevated only in
seen at 3-month intervals for the first 2 years patients with advanced disease.16 The level of
then every 6 months for the next 3 years.5 CA-15-3 is highest in patients with liver or bone
metastasis. The ASCO panel evaluated 12 stud-
Chemistry ies reporting on the value of CA-15-3 in detect-
ing asymptomatic recurrent breast cancer. Of
An abnormal chemical evaluation has been the these studies, only seven could be properly ana-
first evidence of recurrent breast cancer in lyzed.5 Of 1,672 patients followed in these trials,
approximately 1 to 12 percent of patients.7,14 An 352 developed recurrence. About two-thirds of
abnormal blood count is rarely seen as a first these were detected by an elevated CA-15-3
indicator of recurrent breast cancer. Hannisdal before other parameters revealed recurrence.
and colleagues reviewed their experience with The mean lead-time from marker elevation to
430 patients.15 In 8 of 430 patients, an elevated clinical diagnosis was 5 to 7 months. However,
erythrocyte sedimentation rate, gamma glu- the sensitivity of the test was only 57 to 79 per-
tamyl transferase, and alkaline phosphatase val- cent. It is also not known whether such early
ues heralded recurrences. The sensitivity and detection leads to improved survival. If and
specificity of these tests for relapse was 55 and when new therapies for the treatment of metasta-
91 percent, respectively.15 The ASCO panel rec- tic breast cancer are developed, the role of CA-
ommends no laboratory tests in the follow-up.5 15-3 may need re-evaluation.
However, the chemotherapeutic drugs used as
adjuvant therapy can be leukemogenic, and CEA
periodic blood tests may have to be undertaken
to detect these changes. Further, many patients The value of CEA5 in detecting recurrent breast
with breast cancer have other concurrent dis- cancer is even less than that of CA-15-3. At
Surveillance of the Breast Cancer Patient 267

present, there are no indications for using CEA Bone Scans


in routine surveillance of breast cancer patients.
In 1979, it was proposed on the basis of Bayes
decision analysis that since there is no evidence
Mammography
that early detection and therapy for metastatic
Patients with breast cancer are at higher risk for breast cancer alters the clinical course of the
developing contralateral breast cancer. Further, patient, bone scans should be performed only in
patients with breast cancer who have undergone symptomatic patients. Since then, several stud-
conservative therapy with lumpectomy or radi- ies have confirmed this recommendation. The
ation are at risk for ipsilateral recurrence. Cur- Eastern Cooperative Group confirmed the find-
rently, it is recommended that patients who ing in the mid-1980s.18 The National Surgical
have undergone unilateral mastectomy should Adjuvant Breast and Bowel Project (NSABP)
undergo contralateral mammography on a followed 1,989 patients on the B-09 arm of
yearly basis. Patients who have been treated their study.19 Of these patients, 779 had treat-
with conservative techniques and develop local ment failure, of whom roughly one-fifth had
recurrence can be salvaged by appropriate recurrences limited to bone. Only 52 (0.6%)
treatment such as mastectomy. In the GIVIO patients had screening scans that were useful in
trial,11 patients randomized to intensive follow- detecting lesions in asymptomatic patients. The
up had an 11.4 percent incidence of contralat- NSABP changed their recommendation about
eral breast cancer, versus 6.6 percent in the surveillance of breast cancer patients in 1994
group with only routine follow-up. Local ther- based on this study.
apy for ipsilateral breast cancer recurrence or In the GIVIO trial, in which patients were
detection of a new primary in the contralateral randomized between intensive follow-up versus
breast will improve quality and quantity of life. observation only, compliance was > 80 percent
in both groups.11 At a median follow-up of 71
Routine Chest Radiography
months, there was no difference in overall sur-
vival in the two groups. The study also showed
Several investigators have studied the value of no impact on the quality of life because of
annual chest radiography in patients with breast intensive intervention. Another Italian trial, that
cancer. In general, only 0.2 to 4 percent of radi- of Del Turco and colleagues,12 evaluated 1,243
ographs were abnormal in truly asymptomatic consecutive patients with intensive intervention
patients. As pointed out by Loprinzi,17 the only versus minimum follow-up. In this study, there
value of routine annual chest radiography is to was increased detection of isolated intratho-
detect lymphangitic pulmonary metastasis racic and bone metastasis in the intensive fol-
before it causes significant pulmonary symp- low-up group compared to clinical follow-up
toms and thus impairs quality of life. Although group. No difference was observed, however,
the ASCO panel does not recommend annual for other sites, and 5-year overall mortality was
chest radiography,6 the current author agrees 18.6 versus 19.5 percent (statistically insignifi-
with Loprinzi17 that it may be beneficial up to 3 cant difference) between the two groups.
years following primary therapy. It is unlikely The bone scan has a false-positive rate of
that this will improve survival, but it can be approximately 15 percent.2 If the prevalence of
helpful in preserving quality of life. Patients metastasis is low, the predictive value of a pos-
with aggressive breast cancer tend to recur in itive test will be low and the patient may be
the first 2 years following primary therapy, subjected to additional unnecessary interven-
meaning that the annual radiograph can be tions. On the basis of Baye decision analysis, it
ceased after 3 years of follow-up.3 was pointed out that routine bone scans in sur-
268 BREAST CANCER

veillance of a breast cancer patient will lead to IMPLICATIONS FOR


a low predictive value of a positive test and HEALTH-CARE COSTS
unnecessary and expensive interventions.2 It is
therefore recommended that bone scans be per- In response to spiraling health-care costs, man-
formed only in breast cancer patients who are aged care was aggressively introduced.20 Man-
symptomatic with bone pain or with significant aged care is under intense pressure these days,
elevations in their alkaline phosphatase. and its future is uncertain. However, the silver
lining of managed care has been that it has
Imaging Studies of the Liver forced clinicians to critically evaluate their prac-
tices. This has resulted in developing pathways
There have been no prospective studies com- and guidelines for various illnesses. Cohen and
paring sensitivity and specificity of CT and colleagues argue that physicians have a respon-
MRI evaluations in the postoperative surveil- sibility to assure the delivery of appropriate
lance of breast cancer patients. However, on health care without sacrificing the quality of
the basis of evaluation in preoperative breast care.21 The upper limit for an acceptable cost-
and lung cancers, it is safe to conclude that in effectiveness ratio remains controversial. More-
the absence of symptoms and abnormal liver over, it is important to bear in mind that not only
function tests, these imaging techniques will quantity of life but the impact of an intervention
be of little or no value. The Italian GIVIO on the quality of life should be measured.22
investigators performed liver echography in Measurement of quality of life is still somewhat
their intervention group. In that study, 6.5 per- subjective, but as new tools are developed, it
cent of patients had their first recurrence diag- must be incorporated into cost analyses.
nosed by liver echography, versus 6.1 percent In two prospective clinical trials conducted
in the control group, who had echography in Italy,11,12 no cost-effective analysis was
because of abnormal examination and/or available. Schapira attributed savings of $636
hepatic function tests.11 Based on these stud- million in 1990 costs and projected a $1 billion
ies, it can be concluded that routine imaging saving in the year 2,00023 when a minimal fol-
techniques for detecting liver metastasis are low-up schema, as described here, is
not warranted. employed. However, it is unclear whether his
Table 16–2 summarizes recommendations analysis includes additional expenses incurred
for follow-up of breast cancer patients who as the result of FP tests, which can lead to
have completed their primary therapy, and, additional interventions. The negative psycho-
when appropriate, adjuvant chemotherapy. logical impact of an FP test cannot be quanti-

Table 16–2. PROPOSED FOLLOW-UP SCHEDULE

Procedure Low-Risk* High-Risk†

History and physical examination 3 mo ´ 2 yr and then 3 mo ´ 2 yr


6 mo ´ 3 yr then yearly 6 mo ´ 3 yr then yearly
Complete blood count and chemistry Every 6 mo ´ 2 yr 3 mo ´ 2 yr
and then yearly 6 mo ´ 3 yr then yearly
Markers: CEA, CA-15-3 — —
Mammogram Yearly Yearly
Chest radiograph Yearly Yearly
Scans (bone, liver) — —

*Patients with negative nodes and/or positive receptors.



Patients with positive nodes and/or received chemotherapy.
Adapted from Khandekar JD. Preoperative and postoperative follow up of cancer. In: Winchester DP, Scott Jones R, Murphy GP, editors.
Surgical oncology for the general surgeon. Lippincott Williams and Wilkins; 1999. p.
Surveillance of the Breast Cancer Patient 269

fied at this time, but needs to be evaluated in nosis of local and regional recurrences as well
future analyses. as new contralateral breast cancers, which can
In summary, the minimal surveillance be cured. Although controversial, the current
schema proposed has considerable implications author continues to believe that an annual radi-
for health-care costs. Reallocation of health- ograph for the first 3 years after primary treat-
care resources to areas that will lead to ment and occasional blood tests are indicated.
improved survival and quality of life is an On the other hand, more expensive tests such as
important aspiration for health-care policy. bone scans, ultrasound, and CT scans of the
chest, brain, and liver, and measurement of
LEGAL IMPLICATIONS tumor markers, are not indicated.
At this time, there is no evidence that
In our litiginous society, there is apprehension metastatic disease, when detected early, can be
about the legal consequences of a delayed diag- cured by present techniques. However, the rec-
nosis, even for the metastatic disease. It is ommendation of minimal follow-up may need
important to educate the public as well as the to be altered if early intervention in recurrent
legal profession on the differentiation between breast cancer will lead to improved survival.
diagnosis of a primary breast cancer and that of Patients who are in clinical trials should have a
a metastatic disease. If in the future better and more intensive follow-up to differentiate the
more effective treatments become available that disease-free interval and overall survival in the
change the natural history of the disease, detec- control and experimental groups. However, the
tion at an earlier time may become important. clinical trial should keep these diagnostic tests
The guidelines developed by associations such to a minimum so that managed care and other
as ASCO5 and the Society of Surgical Oncol- healthcare providers do not object to extra
ogy21 are helpful for physicians protecting expenses. To improve therapy, it is important
against these legal threats. that an increasing number of patients be
enrolled in clinical trials. Therefore, true collab-
CONCLUSION oration needs to be developed between various
cooperative groups and healthcare providers. It
It is only in recent years, primarily because of is important to improve the health-care of
economic pressure, that guidelines for surveil- patients while giving appropriate consideration
lance have been developed and adapted. In to the problem of escalating health-care costs.
1999, about $1.3 trillion will be spent on health
care.24 Several studies as well as the analysis REFERENCES
presented here indicate that a minimal surveil-
lance approach is clearly warranted.17,25 One 1. McNeil BJ, Keller E, Adelstein SJ. Primer on cer-
can argue that there need not be any follow-up tain elements of medical decision-making. N
Engl J Med 1975;293:211.
for breast cancer patients after primary inter-
2. Khandekar JD. Role of routine bone scans in oper-
vention. Although routine history and physical able breast cancer: an opposing viewpoint.
examinations may not have a direct benefit in Cancer Treat Rep 1979;63:1241.
terms of survival, they have an immense psy- 3. Khandekar JD. Preoperative and postoperative fol-
chological effect. Most patients need reassur- low-up of cancer. In: Winchester DP, Scott Jones
ance, which leads to self-confidence. Visits also R, Murphy GP, editors. Surgical oncology for
the general surgeon. Philadelphia, PA: Lippin-
provide time to educate patients and discuss
cott, Williams & Wilkins; 1999. p. 43–54.
psychosocial and physical rehabilitation—it is 4. Vecchio TJ. Predictive value of a single diagnostic
imperative that patients do not feel abandoned test in unselected populations. N Engl J Med
by their physicians. These visits also allow diag- 1966;274:1171.
270 BREAST CANCER

5. ASCO Special Article. Recommended breast can- 16. Eskelinen M, Hippelainen M, Carlsson L, et al. A
cer surveillance guidelines. J Clin Oncol 1997; decision support system for predicting a recur-
15:2149. rence of breast cancer; a prospective study of
6. Canadian Medical Association. The Canadian task serum tumor markers TAG 12, CA 15-3 and
force on the periodic health examination. Can MCA. Anticancer Res 1992;12:1439–42.
Med Assoc J 1979;121:1193. 17. Loprinzi CL. It is now the age to define the appro-
7. Winchester DP, Sener SF, Khandekar JD, et al. priate follow-up of primary breast cancer
Symptomatology as an indicator of recurrent or patients. J Clin Oncol 1994;12:881.
metastatic breast cancer. Cancer 1979;43:956. 18. Pandya KJ, McFadden ET, Kalish LA, et al. A ret-
8. Scanlon EF, Oviedo MA, Cunnigham MP, et al. Pre- rospective study of earliest indicators of recur-
operative and follow-up procedures on patients rence in patients on Eastern Cooperative
with breast cancer. Cancer 1980;46:977. Oncology Group adjuvant chemotherapy trials
9. Gerber FH, Goodreau JJ, Kirchner PT, Fouty WF. for breast cancer. A preliminary report. Cancer
Efficacy of preoperative and postoperative 1985;55:202.
bone scanning in the management of breast 19. Wickerham L, Fisher B, Cronin W. The efficacy of
carcinoma. N Engl J Med 1977;297:300. bone scanning in the follow-up of patients with
10. Burke W, Daly M, Garber J, et al. Recommenda- operable breast cancer. Breast Cancer Res
tions for follow-up care of individuals with an Treat 1984;4:303.
inherited predisposition to cancer. II. BRCA1 20. Bodenheimer T. The American health care system:
and BRCA2. JAMA 1997;277:997. physicians and the changing medical market-
11. The GIVIO Investigators. Impact of follow-up test- place. N Engl J Med 1999;340:584–8.
ing on survival and health-related quality of life 21. Cohen AM, Bland KI, Gardner B, Winchester DP.
in breast cancer patients. A multicenter random- Society of Surgical Oncology and Practice
ized controlled trial. JAMA 1994;271:1587. Guidelines. Oncology 1997;11:869.
12. Del Turco MR, Palli D, Cariddi A, et al. Intensive 22. Cella D, Fairclough DL, Bonomi PB, et al. Qual-
diagnostic follow-up after treatment of primary ity of life (QOL) in advanced non-small cell
breast cancer. A randomized trial. JAMA 1994; lung cancer (NSCLC) results from Eastern
271:1593. Cooperative Oncology Group (ECOG) study
13. Tomin R, Donegan WL. Screening for recurrent E5592. Proc ASCO 1997;16(4):2a.
breast cancer: its effectiveness and prognostic 23. Schapira DV, Urbrn N. A minimalist policy for
value. J Clin Oncol 1987;5:62. breast cancer surveillance. JAMA 1991;265:
14. Dewar JA, Kerr GR. Value of routine follow-up of 380.
women treated for early carcinoma of the 24. Modern Healthcare. Economic data: some things
breast. BMJ 1985;291:1464. never change. Modern Healthcare 1999;29:16.
15. Hannisdal E, Gundersen S, Kvaloy S, et al. Follow- 25. Schapira DV. Breast cancer surveillance: a cost-
up of breast cancer patients stage I-II: a baseline effective strategy. Breast Cancer Res Treat
strategy. Eur J Cancer 1993;29A:992–7. 1993;25:107.
17
Treatment of Metastatic
Breast Cancer
DOUGLAS E. MERKEL, MD

Despite the improvements in prognosis achieved The most common sites of metastatic involve-
for many patients with breast cancer, approxi- ment are bone, lungs, and liver.3 While both infil-
mately 46,000 women die of this disease each trating ductal carcinoma and infiltrating lobular
year. The increase in incidence of breast cancer carcinoma will relapse at the same rate over time
seen through the early 1990s has been success- based on their size and degree of nodal involve-
fully offset by two factors: widespread applica- ment, a predilection for certain sites of involve-
tion of screening mammography, permitting ment can be related to histology. Infiltrating lob-
more frequent early diagnosis and, more recently, ular carcinomas are more likely to recur in bone
the decrease in recurrence and mortality rates marrow, peritoneum, pelvic organs, and
achieved through the now standard application of meninges than are infiltrating ductal carcinomas.
effective systemic adjuvant therapy.1 Unfortu- On the other hand, lung metastases are more
nately, similar gains have not been achieved for common with infiltrating ductal cancers.4–6
women who present with metastatic breast cancer When metastatic breast carcinoma is first
or for those with distant disease relapse after ini- diagnosed, a brief staging workup is indicated to
tial treatment. For these women, palliation of determine the extent of disease and thus treat-
symptoms and some prolongation of survival is ment priorities. In the absence of symptoms,
possible but there is no known curative treatment. chest radiography, abdominal CT scan, and
In fact, the death rate for this disease has bone scan are needed. Any abnormalities on
remained stubbornly constant over decades.2 bone scan should be further pursued with bone
Metastatic breast cancer is the second most com- radiographs, to confirm metastatic disease and
mon cause of cancer death among women. determine whether it is lytic or blastic in nature.
While only 6 percent of patients present ini- The finding of lytic disease, particularly in
tially with metastatic breast cancer,2 metastases weight-bearing bones, has specific palliative
will eventually develop in at least 30 percent of implications. In addition to the above studies,
patients with node-negative primary breast can- any symptoms should be fully investigated.
cer and 50 percent of those with positive nodes Prior to beginning treatment for metastatic
at diagnosis. The event rate for recurrence is rel- disease, biopsy to confirm clinical suspicions
atively constant over the first 10 years for should be considered mandatory in all but the
women who receive adjuvant therapy, that is, most unusual of circumstances. The diagnosis
each year healthy survivors face the same risk of incurable metastatic disease has obvious and
for recurrence as they did in the preceding year.1 profound prognostic implications, and often

271
272 BREAST CANCER

commits a patient to lifelong systemic treat- BRAIN METASTASES


ment. The possibility of a benign lesion must
therefore be excluded. In a disease such as Brain metastases are diagnosed in 16 to 25
breast cancer, which may recur many years percent of all women with breast cancer, but the
after initial treatment, the possibility of a sec- brain is seldom the first site of relapse.15 Thus,
ond primary carcinoma must also be consid- while brain imaging with gadolinium-enhanced
ered and excluded. In addition to confirming MRI is not part of the routine initial workup for
the diagnosis of metastatic cancer, the tumor newly diagnosed metastatic breast cancer, any
should be analyzed for estrogen receptor, prog- patient with new neurologic complaints should
esterone receptor, and HER2/c-erbB2 protein,7 be promptly evaluated. Most commonly, a pro-
as these results will largely determine the gressively worsening headache develops over
options for systemic therapy. days to weeks. Other common clinical features
The prognosis for metastatic breast cancer is of brain metastases include behavioral or cogni-
related to a number of variables, perhaps most tive changes, focal weakness, ataxia, speech dis-
importantly to the disease-free interval, or the orders, and seizures.16
duration of time between initial diagnosis and Papilledema is present in only 15 percent of
recurrence. This duration provides some mea- patients, and the screening neurologic exam may
sure of the growth rate of the cancer; longer sur- be negative. Any of the above symptoms are thus
vivals are reported when the disease-free inter- indications for scheduling a gadolinium-
val exceeds several years.8 The extent of enhanced MRI. Computed tomography (CT)
metastatic involvement, or the number of scan is less sensitive and more likely to result in
involved sites, also has an impact on survival, as equivocal or false-positive findings. These scans
does location.9 There is a particularly good cannot detect meningeal involvement and should
prognosis observed for patients with a single only be obtained where MRI is unavailable.17
metastatic focus amenable to surgery or radio- Corticosteroids, usually dexamethasone at a
therapy.10 Survival in excess of 2 years is also dosage of 4 mg every 6 hours, can produce
common when the disease is limited to bone but immediate but shortlived improvement in neu-
is not expected when there is visceral involve- rologic symptoms and are indicated as initial
ment.11 Finally, improved survival is reported treatment in all patients with strongly suspected
for estrogen receptor-positive metastatic breast or newly diagnosed brain metastases.18 Anti-
cancer,12 although other markers such as ploidy, convulsants, however, should be reserved for
S-phase fraction, and HER2 status are not infor- the 20 to 30 percent of patients who suffer focal
mative.13 The relationship of estrogen receptor or generalized seizures.19
status and prognosis may not be independent of Treatment of brain metastases may consist
other factors, however, as these metastases are of either surgical extirpation, whole brain
also more likely to be found in bone and soft tis- radiotherapy, or stereotactic “gamma-knife”
sue, and to occur at a longer disease-free inter- radiosurgery. As retrospective analyses and a
val, than those lacking estrogen receptors.14 single randomized trial have demonstrated
As many patients will present initially improved neurologic control and survival for
with only a single site of metastasis, or a patients undergoing surgery for brain metas-
dominant lesion, treatment considerations for tases, resection must be the first consideration
specific sites of metastases will be surveyed for all appropriate patients.20 The most appro-
below. Systemic measures designed to palli- priate candidates for resection have single,
ate symptoms and offer some hope of delay- accessible lesions, particularly those that are
ing the progression of metastatic disease will relatively bulky and thus unlikely to respond
then be discussed. completely to radiotherapy. The surgical candi-
Treatment of Metastatic Breast Cancer 273

date should also be one whose other sites of ally in the setting of disseminated, progressive
metastatic disease are responding, or are likely disease.25 As mentioned above, this complica-
to respond to systemic therapy; for those whose tion is more commonly observed in patients
expected survival is limited, surgical interven- with infiltrating lobular cancer. The majority of
tion has little or no advantage over radiotherapy. patients will present with neurologic signs
Radiation therapy is indicated as initial pal- referable to some combination of cerebrum,
liative treatment for all other patients, for exam- cranial nerves, and spinal cord, although the
ple, those with multiple lesions or poorly con- patient may complain only of a single symp-
trolled systemic disease. Median survival for tom.26 The single-most common complaint is
patients treated in this fashion is 3 to 6 months weakness of the legs, perhaps accompanied by
but a majority receive symptomatic benefit.21 pain or paresthesias. Cranial nerve involvement
Those who survive for ³ 1 year or longer after can produce diplopia, facial numbness or weak-
whole brain radiotherapy are at risk for a variety ness, and hearing loss. Involvement of the cere-
of complications ranging from subtle cognitive bral cortex is heralded by headache, impaired
deficits to leukoencephalopathy manifesting as memory, lethargy, and nausea.
progressive dementia with ataxia. This is pri- Definitive diagnosis of leptomeningeal car-
marily a concern in good-risk patients and has cinomatosis is difficult, as initial cytologic
made the use of adjuvant whole brain radiother- examination of the cerebrospinal fluid (CSF) is
apy following surgical removal of a solitary falsely negative in up to 46 percent of
brain metastasis controversial. patients.27 Elevated CSF protein levels and
Stereotactic radiosurgery is a new technique monocytosis may be observed, and repeated
that delivers a single, large, tightly focused sampling may yield positive cytology. Gadolin-
dose of radiation to a metastatic site, using mul- ium-enhanced MRI of any areas of clinical
tiple beams. This technique is highly effective involvement should be obtained, both to rule
for tumors < 3 cm, can be performed on an out- out parenchymal brain metastases or epidural
patient basis, and appears to result in far less cord compression and to detect enhancing,
risk of long-term damage to surrounding nor- nodular meningeal enhancement—this may be
mal tissue.22 While the current treatment of seen along the convexity of the cerebrum, along
choice for recurrent disease after whole brain the brain stem, or involving spinal nerve roots
radiotherapy, and for patients with surgically in up to 70 percent of patients.28
inaccessible lesions, this technique may in time Treatment of leptomeningeal carcinomatosis
replace primary surgery for some patients. is difficult, as it often arises in the midst of pro-
Perhaps surprisingly, brain metastases from gressive systemic breast cancer and there has
breast cancer have been reported to respond been no optimal approach established. Radia-
favorably to systemically administered chemo- tion therapy is usually administered to areas of
therapy23 or tamoxifen.24 While currently no tri- bulky or symptomatic disease, although studies
als have established this as frontline therapy for to establish this practice are lacking. Radiation
brain metastases, this approach can certainly be therapy to the entire neuraxis is to be avoided as
tried in patients who relapse following whole- it can result in severe and prolonged myelosup-
brain radiotherapy, or those who decline to pression, thus preventing the subsequent admin-
undergo it. istration of systemic chemotherapy.
As the entire neuraxis is potentially at risk
LEPTOMENINGEAL CARCINOMATOSIS for leptomeningeal spread, direct CSF installa-
tion of chemotherapy is also indicated. Because
Involvement of the leptomeninges occurs in up of improved distribution of drug throughout the
to 5 percent of patients with breast cancer, usu- CSF, intraventricular administration via an
274 BREAST CANCER

Ommaya reservoir is preferred over lumbar The purpose of chest tube placement and
puncture. Methotrexate 12 mg two or three suction drainage is to empty the pleural space
times weekly has been used most often, with to permit approximation of the visceral and
improvement reported in 60 to 80 percent of parietal pleura. When chest tube output is min-
patients.26,29 The most common complication is imal, any of a variety of topical irritants is
transient aseptic meningitis, manifesting as instilled and the patient repositioned every 15
headache, fever, and stiff neck. Particularly with minutes for 2 hours to distribute the irritant
simultaneous cranial radiotherapy, a necrotizing throughout the pleural space. The goal is to cre-
leukoencephalopathy with impaired mentation ate adhesions between the irritated visceral and
and focal defects may develop.29 Leakage of parietal pleura to prevent subsequent massive
methotrexate outside of the CSF may result in reaccumulation of fluid with atelectasis. There
mucositis or myelosuppression but may be have been a variety of agents employed, includ-
counteracted by concurrent administration of ing talc slurry, tetracycline, bleomycin, and
oral or intravenous folinic acid. The median sur- other chemotherapeutic agents. In a random-
vival for patients who develop carcinomatosis ized trial comparing the first three agents, an
meningitis is 3 to 6 months, although respon- insufficient number of patients was accrued; in
ders may live in excess of 1 year. the absence of a direct comparison, talc appears
to have the highest success rate.31
MALIGNANT EFFUSIONS Pericardial effusions are not uncommon and
may eventually occur in up to 25 percent of all
Breast cancer is the most common cause of women with metastatic breast cancer.32 The pre-
malignant pleural effusions in women. They are senting complaint is typically exertional dysp-
more commonly seen ipsilateral to the primary nea. Chest radiography and resting arterial oxy-
tumor, suggesting that the effusion sometimes gen saturation may both be normal, requiring
arises via direct extension through the chest wall this diagnosis to be specifically considered in
or through involvement of internal mammary the dyspneic patient.33 As pericardial effusions
lymph nodes. While 80 percent of malignant occur not infrequently in conjunction with
pleural effusions arise in the presence of other malignant pleural effusions but go unrecognized
sites of metastatic involvement,30 they are usu- on chest radiography, pericardial effusions
ally symptomatic and require specific treatment. should also be considered whenever pleural
In a previously untreated patient with newly effusions are diagnosed. Physical exam may
metastatic breast cancer, an attempt may be show tachycardia, an absent precordial cardiac
made to relieve the malignant pleural effusion impulse, a pericardial friction rub, atrial fibrilla-
with therapeutic thoracentesis and initiation of tion, and pulsus paradoxus. Electrocardiogram
systemic chemotherapy or hormonal therapy. In will show decreased voltages in the precordial
this setting, a positive response to systemic leads. Definitive diagnosis of pericardial effu-
therapy is likely and may be sufficiently rapid sion requires echocardiography, which may also
to prevent reaccumulation of fluid. In patients demonstrate cardiac tamponade with diastolic
with previously treated metastatic disease, collapse of the right atrium and ventricle.34
however, the likelihood of objective response to Patients with symptomatic or hemodynami-
any systemic therapy is certainly < 50 percent; cally significant pericardial effusions should
definitive treatment with chest tube drainage undergo immediate drainage. Immediate catheter
and sclerosis is recommended. Failure to ade- drainage can prevent cardiovascular collapse in
quately manage a malignant pleural effusion patients with tamponade but does not provide
can result in a trapped lung, with permanent definitive treatment. The creation of a subxiphoid
dyspnea, cough, and pain. pericardial window is a relatively simple surgical
Treatment of Metastatic Breast Cancer 275

solution with a high success rate.35 Open thora- minemia, however. Diuretics are seldom help-
cotomy with pericardial stripping has a much ful in managing malignant ascites.
higher morbidity and is required only for rare
patients with constrictive pericarditis. BONE METASTASES
Malignant ascites can develop as a manifes-
tation of peritoneal metastases, occurring more Bone is the most frequent site of metastatic
frequently in patients with infiltrating lobular spread, with autopsy series revealing skeletal
carcinoma. Symptoms include bloating, disten- involvement in 85 percent of all breast cancer
sion, early satiety, and shortness of breath. Both patients. The axial skeleton is most commonly
ultrasound and CT scan can demonstrate affected, for example, the pelvis, spine, ribs,
ascites, with the latter also revealing peritoneal skull, and proximal long bones (Figure 17–1).36
studding or omental thickening in some Constant, dull, progressive pain is the usual pre-
patients. While the most satisfactory control of sentation, and any such complaint should be
malignant ascites is achieved with effective investigated radiographically, particularly if the
systemic therapy, this is often not possible pain is unrelieved by rest. Plain radiographs
where ascites occurs as a late complication of most often show lytic lesions, although 15 per-
advanced disease. Therapeutic paracentesis cent of breast carcinomas are associated with
may provide transient relief of symptoms. blastic lesions and both patterns may be evi-
Repeated drainage of several liters of ascitic dent.37 Technetium bone scans are more sensi-
fluid may result in hypotension or hypoalbu- tive but less specific than plain films, as bone

Figure 17–1. A bone scan with multiple focal areas of increase radionuclide uptake (e.g., spine, skull)
responding over time to hormonal therapy.
276 BREAST CANCER

loss of up to one-third may occur before becom- bone metastases. Bisphosphonates are a class
ing visible. The first discovery of a bone metas- of drugs related to pyrophosphate that bind to
tasis in a patient should prompt a bone scan to hydroxyapatite crystals, stabilizing bone and
determine the extent of disease. Unfortunately, inhibiting reabsorption. The bisphosphonate
available serum markers of osteoblastic activity pamidronate, administered intravenously, has
such as alkaline phosphatase or tumor markers been shown to produce sclerosis or stabilization
such as CA15-3 antigen are insufficiently sensi- of lytic metastases in 50 percent of breast can-
tive to rule out bone metastases in a patient with cer patients.41 This results in a decrease in the
bony pain.37 rate of pathologic fractures, in the need for
External beam radiation therapy is the main- radiotherapy, and in the use of analgesics,42
stay of palliating the pain of bone metastases suggesting an important palliative role for
and will provide at least some relief in 90 per- pamidronate. Similar trials using orally
cent of patients.38 Relief may be experienced absorbed bisphosphonates such as alendronate
early on as a result of decreasing periosteous have also been completed.
inflammation, with maximal palliation within 3 Strontium 89 is an emitter of b-radiation
weeks. The main side effect of radiation ther- that is taken up by sites of active bone destruc-
apy to the axial skeleton is myelosuppression, tion, and can be administered intravenously.
which may be cumulative and prolonged when Improvement in bone pain is reported by 80
large fields or multiple sites are treated. As this percent of patients,43 and toxicity is largely lim-
may preclude effective dosing of chemother- ited to myelosuppression. Indications include
apy, radiation should be reserved for sites of widespread, painful bony metastases in patients
severe or dominant symptoms, to prevent frac- who will not be candidates for chemotherapy
ture of long bones, or in patients unsuitable for, and recurrent pain in sites already treated by
or unlikely to respond to, chemotherapy. external beam radiotherapy.
Surgical stabilization is required in patients
with impending fractures of the femur or for LOCAL RECURRENCE
occasional patients with extensive and painful
humeral lesions. The proximal femur is at par- Recurrence of cancer within a breast after
ticular risk of pathologic fracture due to the high lumpectomy and radiation therapy has different
mechanical stresses of ambulation; an aggres- implications from a recurrence involving the
sive approach to prevention is appropriate due skin or chest wall following mastectomy.44
to the catastrophic effects of this complication. Treatment involves mastectomy and often a
Prediction of an impending fracture is not course of systemic “pseudoadjuvant” chemo-
entirely accurate, but the usual criteria for pro- therapy or change in hormonal adjuvant ther-
phylactic stabilization are cortical destruction of apy. No randomized trials have addressed this
> 50 percent or proximal lesions > 1 inch.39 issue, however.
Lytic lesions are more prone to fracture than is Initial recurrence in the skin overlying a
blastic disease. Depending on location, a variety mastectomy site is associated with synchronous
of orthopedic approaches may be required; the presentation of distant metastatic disease in
use of methylmethacrylate cement permits early one-third of patients. Discovery of local recur-
reambulation.40 Following fixation, adjuvant rence should therefore prompt restaging with a
radiotherapy to the involved bone is usually bone scan and CT scan of chest and liver.45 If
indicated to prevent progressive destruction of distant metastasis are not discovered, the skin
bone, with resulting destabilization. recurrence, usually in the form of one or sev-
Recently, medical therapy has been able to eral dermal or subdermal nodules, should
more directly address the pathophysiology of receive local treatment. Complete excision of
Treatment of Metastatic Breast Cancer 277

isolated, small nodules should be attempted. must involve weighing the potential improve-
Wide excision with partial or full-thickness ment in quality of life against the expected tox-
chest wall resection are seldom indicated, how- icities of treatment.
ever, due to morbidity and a 50 percent failure
rate.46 Rather, radiation therapy delivered to the HORMONAL THERAPY
entire chest wall at a dose of 45 to 50 cGy, with
a boost to the site of recurrence, should be con- In this context, hormonal therapy is almost
sidered standard therapy. This will yield 5-year always preferred as initial therapy for women
local control rates of 85 percent if the tumor is with estrogen receptor-positive metastatic breast
first excised or 63 percent if radiation is given cancer. Whenever possible, and particularly for
without excision.47 women who relapse after adjuvant hormonal
Nearly all patients with isolated local recur- therapy, this decision should be based on recep-
rence will subsequently develop distant metas- tor assays performed on a biopsy of the metasta-
tases. Only 30 percent remain free from distant tic disease. While in the absence of intervening
metastases after 5 years, with the disease-free therapy the receptor status of metastatic disease
interval between mastectomy and skin recur- is predicted by that of the primary tumor, estro-
rence the most important predictive factor. gen receptor becomes negative in one-third of
Only 20 percent of those suffering local recur- patients and progesterone receptor becomes neg-
rence within 2 years will be free of distant dis- ative in one-half of patients who receive tamox-
ease 3 years later, compared to 36 percent of ifen in the interval before relapse.50
those who first recurred > 2 years after mastec- There will be a complete or partial response
tomy.48 This suggests that most patients could obtained by initial hormonal therapy in over
potentially benefit from systemic therapy fol- three-quarters of women with estrogen receptor
lowing local recurrence. The benefits of postre- and progesterone receptor-positive metastatic
currence hormonal therapy with tamoxifen disease and no prior therapy.51 If the estrogen
have been established in a randomized trial for receptor assay is negative, the response rate
patients with estrogen receptor-positive drops to less than half, and to one-third if the
tumors.49 Unfortunately, similar benefits have progesterone receptor assay is negative. If nei-
not been established for chemotherapy in ther receptor is detected, response is seen in < 10
receptor-negative patients. percent of patients; in this circumstance,
chemotherapy is often a preferable option. Hor-
SYSTEMIC THERAPY monal therapy is also contraindicated in women
with lymphangitic carcinomatosis or extensive
When metastatic breast cancer presents clini- metastases to the liver, due to the need for a rapid
cally as an isolated, symptomatic site, specific response. Finally, if biopsy of a metastatic site is
palliative measures, discussed above, are indi- not possible, the decision to employ hormone
cated. Many patients, however, present with therapy can be based on those clinical criteria
visceral or multi-site disease, or will be found (eg, a long disease-free interval, disease limited
to have additional metastases upon restaging. to bone or soft tissue, and elderly patients) asso-
While widely metastatic breast cancer is incur- ciated with the receptor-positive phenotype.
able, lessening of the symptomatic burden and
prolongation of survival are possible for most Estrogen Antagonists
women through judicious use of systemic hor-
mone therapy and chemotherapy. It is important The likelihood of response to initial hormonal
to understand that the goal of systemic therapy therapy is similar for several classes of drugs,
is ultimately palliation, so that every decision and so initial treatment can often be selected
278 BREAST CANCER

based on their side-effect profiles. In previ- may occur due to outgrowth of receptor-
ously untreated patients, or for those several negative clones within a heterogenous popula-
years removed from adjuvant hormonal ther- tion or to acquired, specific resistance to estro-
apy, competitive inhibitors of estrogen binding gen antagonists. Once a responding tumor
are usually the first choice. progresses on tamoxifen, other agents in this
The oldest and most widely prescribed estro- class have little activity. Indeed, a fraction of
gen antagonist is tamoxifen,52 but toremifene such patients will briefly improve when tamox-
has also been approved for this indication. ifen is withdrawn, suggesting that changes in
Raloxifene is currently marketed for prevention the receptor or the cellular estrogen-response
of osteoporosis and may also have some effi- machinery has led to the drug behaving as an
cacy against metastatic breast cancer, although estrogen agonist.57
further study is clearly required.53 Tamoxifen
appears to be effective for both pre- and post- Aromatase Inhibitors
menopausal women with advanced, receptor-
About half of women who initially respond to
positive disease.54 Common side effects of
tamoxifen will also respond to second-line hor-
tamoxifen include hot flashes (particularly in
monal therapy. Randomized trials have sug-
perimenopausal women), disruption of men-
gested somewhat greater efficacy, lesser side
strual cycles, and vaginal dryness or dis-
effects, and perhaps slight improvement in sur-
charge.55 In addition, weight gain and mild fluid
vival when specific aromatase inhibitors are
retention are frequent, with nocturnal leg
compared to oral progestins in this setting.58,59
cramps not uncommonly reported. Patients with
The new generation of aromatase inhibitors—
bone metastasis may suffer a syndrome of
anastrozole, letrozole, and others not yet
“tumor flare,” typically 7 to 10 days after initia-
approved for use—have replaced the older drug
tion of tamoxifen. This is seen in 1 to 3 percent
aminoglutethimide due to much improved
of patients and consists of increased pain at sites
safety profiles. Anastrozole and letrazole work
of metastases; it may lead to hypercalcemia. As
by binding competitively to the porphyrin
this is predictive of subsequent response to
nucleus of the aromatase enzyme, which is
tamoxifen, therapy should be continued, with
responsible for estrogen production from
supportive measures as needed. Approximately
androstenedione. This extraovarian pathway is
1 percent of healthy patients on tamoxifen will
important only in postmenopausal women,
develop deep-vein thrombosis, although women
therefore anastrozole and letrazole should be
with metastatic breast cancer also have an
used only after menopause. The most common
increased incidence of thromboembolic dis-
side effects seen with these drugs are headache
ease.56 Other, rare complications such as
and mild nausea. Prior to the development of
cataract formation or an increased incidence of
these agents, aminoglutethimide had been
endometrial cancer are seldom of concern to
employed as an aromatase inhibitor, but has
women with metastatic disease.
now fallen into disuse because of its high fre-
The average duration of response to initial
quency of unacceptable side effects, including
hormone therapy is approximately 1 year.
rash, lethargy, and ataxia.
Women whose disease stabilizes on tamoxifen
appear to do as well as those achieving objec- Progestins
tive remissions. While responses lasting for
years are not uncommon (particularly if there Before the development of the newer aromatase
has been a long disease-free interval), eventu- inhibitors, second-line hormonal therapy for
ally most tumors will develop resistance to most women consisted of progestins, usually
tamoxifen, leading to clinical progression. This oral megestrol acetate or parenteral medrox-
Treatment of Metastatic Breast Cancer 279

yprogesterone acetate.60,61 Up to half of women CHEMOTHERAPY


receiving these drugs will respond with
improvement or stabilization of their disease. Systemic chemotherapy is often indicated to
Unlike other hormonal agents, there is evidence control disseminated breast cancer and relieve
of a dose-response effect with progestins, symptoms. While prolonged remissions may be
although their mechanism of action is unknown. achieved, there is no evidence that metastatic
These drugs also produce an increased sense of breast cancer can be cured by chemotherapy.
well-being, improved appetite, and suppress hot Thus, the ultimate goals are again palliative,
flashes. Unfortunately, the side effects of and the toxicity of chemotherapy must be care-
chronic weight gain, fluid retention, and dysp- fully weighed against a realistic appraisal of
nea make them unacceptable to many. benefits. With this caveat, chemotherapy is
commonly indicated as frontline therapy for
metastases to liver or lung, those arising from
Ovarian Ablation estrogen receptor-negative tumors, and those
For premenopausal women with receptor-posi- that fail to respond to initial or subsequent hor-
tive disease, medical or surgical castration is also monal treatments.
an effective approach to hormonal therapy. The There are a wide variety of chemothera-
endocrinologic effect of castration is achieved by peutic agents that show some activity against
two analogs of gonadotropin-releasing hormone, metastatic breast cancer. Response rates are
goserelin and leuprolide, which suppress follicle- affected by site of disease, with soft tissue
stimulating hormone and luteinizing hormone, metastases typically most responsive, and
and thus estrogen production by the ovary.62,63 liver metastases least responsive, to many
Either agent will achieve the same benefit as agents. Prior treatment history has a major
oophorectomy, that is, a 45 percent likelihood of effect on the likelihood of response, due to the
disease regression or stabilization, but require phenomenum of pleiotropic drug resistance,
parenteral administration on a monthly or tri- which occurs when cancer cells undergoing
monthly basis. Side effects are limited to pain at treatment become resistant not only to that
the injection site and menopausal symptoms particular agent but also to unrelated classes
such as hot flashes, mood swings, and dry skin. of cytotoxic drugs. Attempts to overcome drug
Once disease progresses after either medical or resistance have included the use of chemother-
surgical castration, the alternate approach has apeutic agents in combination and at increased
little chance of benefit. Obviously, castration by dose intensity. These approaches have resulted
either technique can only be of benefit to pre- in higher response rates, but the average dura-
menopausal patients, where the ovary is the pri- tion of response to initial chemotherapy
mary site of estrogen production. remains < 1 year. High dose chemotherapy
There have been a number of studies that with bone marrow or stem cell rescue has not
have attempted to combine hormonal agents for demonstrated any survival advantage when
more effective control of metastatic disease.64 In compared to conventional regimens in strictly
general, a small increase in response rate is seen randomized prospective trials and should
with combinations, but time-to-progression is remain investigational. The use of alternative
not improved over the use of the same agents agents after progression of disease is marked
employed sequentially and there is clearly no by lower response rates and shorter durations
survival advantage. Additional toxicity is often of response, so much so that patients rarely
reported when hormonal agents are used in benefit from more than three sequential
combination; since the goal of all such therapy chemotherapy regimens. The major classes of
is palliative, this approach is not recommended. useful cytotoxic agents are reviewed below.
280 BREAST CANCER

Anthracyclines Taxanes

Doxorubicin has long been considered the The complex, semisynthetic paclitaxel and the
benchmark drug for treatment of metastatic synthetic docetaxol have recently established
breast cancer, with a single-agent response rate themselves to be of equal or greater single-
of 40 to 50 percent.65 Increases in the dose of agent efficacy than doxorubicin and maintain
doxorubicin, sometimes given with granulocyte significant activity in patients previously
colony-stimulating factor support, can yield treated with doxorubicin.71,72 Paclitaxel admin-
response rates as high as 80 percent, but at the istered by 24-hour infusion has achieved
price of increasing toxicity.66 As this higher response rates as high as 60 percent, but the
response rate does not result in any noticeable optimum dose and schedule for this drug have
improvement in survival, dose-intense schedules not been established. Toxicities include neu-
cannot be recommended at present. Common tropenia, a delayed arthralgias/myalgia syn-
toxicities include moderate nausea, mucositis, drome occurring 48 hours after administration,
neutropenia, and a cumulative dose-related risk and a peripheral neuropathy with higher cumu-
of congestive cardiomyopathy. Despite these lative doses. Bradycardia is observed but is sel-
toxicities, doxorubicin, often given in combina- dom clinically significant. Frequent type I
tion, has become the standard frontline hypersensitivity reactions require premedica-
chemotherapy for metastatic breast cancer. Sev- tion with steroids and antihistamines. These
eral randomized trials have established that dox- toxicities are lessened by administration on a
orubicin-containing combinations are superior weekly rather than triweekly schedule.
to similar regimens lacking an anthracycline. Docetaxol has recently been introduced for
Mitoxantrone is a potentially less toxic deriv- treatment of metastatic breast cancer, where it
ative of doxorubicin that is also widely used for has demonstrated a higher response rate and
palliative treatment of metastatic disease. It is more durable remissions than doxorubicin.72
clearly less emetogenic and appears to have less Toxicity consists of neutropenia and a capil-
cardiotoxicity than the parent compound, lary leak syndrome, resulting in peripheral
although cardiotoxicity is cumulative and addi- edema and pleural or pericardial effusions,
tive to that induced by prior doxorubicin expo- which are preventable with a 3-day course of
sure.67 In direct comparison to doxorubicin, corticosteroids.73 These taxanes have been
either alone or in combination, response rates combined by a number of investigators, but
were lower for mitoxantrone, although overall thus far both activity and toxicity appears to
survival was not compromised.68 The most effec- be additive rather than synergistic. An excep-
tive use of mitoxantrone may be in combination tion is the combination of doxorubicin and
with 5-fluorouracil and folinic acid, which is paclitaxel, which appears to produce a very
associated with a response rate of up to 65 per- high response rate but results in cardiotoxicity
cent and quite manageable toxicity.69 Another at lower than expected cumulative doxoru-
approach to lessening the toxicity of doxorubicin bicin doses.74
is to encapsulate the drug in lipid liposomes.70
This strategy permits more selective tissue Alkylating Agents
uptake, resulting in less nausea, neutropenia, and
cardiotoxicity, although various cutaneous reac- Cyclophosphamide has reasonable single-agent
tions are seen with the currently available formu- activity but is usually used in combination with
lation. Studies using liposomal doxorubicin are other agents such as doxorubicin or methotrexate
still in progress and the drug is not currently and fluorouracil. Toxicity is limited at conven-
approved for treatment of breast cancer. tional doses to neutropenia, moderate nausea,
Treatment of Metastatic Breast Cancer 281

mucositis, and occasional hemorrhagic cystitis. Other Agents


Ifosfamide, an analog of cyclophosphamide,
Cisplatin is an active single agent in previously
appears to have similar efficacy, and is not
untreated metastatic breast cancer, but dosage
entirely cross-resistant to cyclophosphamide. In
is inconvenient due to the need for prolonged
addition to neutropenia, toxicity includes fre-
hydration to prevent nephrotoxicity; also, the
quent hemorrhagic cystitis (requiring the use of
drug has a poor response rate in previously
a urothelial protective agent), interstitial nephri-
tis, and temporary encephalopathy. These toxic- treated patients. Gemcitabine may be non-
ities have limited the use of ifosfamide. crossresistant with anthracyclines and taxanes,
with toxicity limited to fatigue and mild
myelosuppression, suggesting that this drug
Antimetabolites
may find a role in previously treated patients.79
Five-fluorouracil, a pyrimidine analog that Further study is required, however, and neither
binds to thymidylate synthase, is widely this drug nor cisplatin has been approved for
employed in the treatment of breast cancer. treatment of breast cancer. Finally, capecitabine,
Intermittent bolus administration, as is found in an oral drug, has recently been approved for
many classic cytotoxic combinations, is prob- use in previously treated patients with metasta-
ably the least effective schedule for this cell- tic breast cancer. Once absorbed, this drug is
cycle active agent, given its short half-life. converted to fluorouracil, explaining its similar
Continuous infusion of 5-fluorouracil will yield toxicity spectrum.
responses in even heavily pretreated patients, The higher response rates seen with combi-
with manageable toxicity consisting largely of nation chemotherapies often justify their initial
palmar/planter dermatitis.75 Alternatively, the use over single agents. After progression on
intracellular binding of fluorouracil to frontline treatment, however, the toxicity of
thymidylate synthase can be stabilized by coad- multiagent therapy may make adequate dosing
ministration of folinic acid. Again, higher impossible and obviate any advantage seen with
response rates are seen, although neutropenia, this approach. Thus, the sequential use of single
mucositis, and diarrhea become significant.76 agents, especially after initial treatment, may
Methotrexate, a folic acid analog, has a low provide a higher quality of life, equal palliative
single-agent response rate in metastatic breast benefit, and no compromise of overall survival.
cancer but is occasionally useful, primarily to In patients who achieve a complete response
provide biochemical synergy with fluorouracil. or whose disease stabilizes after a partial
response, the question of duration of chemo-
Vinca Alkaloids therapy arises. Several small studies have sug-
Whereas vincristine has little activity against gested that the time-to-treatment failure is
breast cancer, vinblastine yields response rates extended by several months and that quality of
as high as 37 percent when given by 120-hour life is improved by continuing with maintenance
infusion.77 Toxicity is limited to myelosuppres- therapy once a response is achieved, rather than
sion but the schedule is inconvenient. Vinblas- withholding further therapy until relapse.80
tine was given as a bolus in many earlier com-
binations but probably added little. Vinorelbine, IMMUNOTHERAPY
a newer vinea derivative, yields a single-agent
response rate of 35 to 40 percent when given as Earlier studies in which the nonspecific
a weekly bolus.78 Toxicity consists of neutrope- immunostimulants bacille Calmette-Guerin or
nia, peripheral neuropathy, myalgias, and short- levamisole were added to chemotherapy showed
lived pain at sites of metastatic disease. no advantage to this procedure. In the last 2
282 BREAST CANCER

years, however, studies using specific nation plus antibody treatment and in 12 percent
immunotherapy with the humanized murine receiving paclitaxel plus antibody. The mecha-
monoclonal antibody trastuzumab have yielded nism of this synergy, manifest both in increased
promising results. This antibody recognizes and response rates and cardiotoxicity, is not yet
binds to a transmembrane tyrosine kinase coded understood, and a wide variety of trials have
for by the c-erbB2 or HER2 gene, which is begun to define the role of trastuzumab in the
amplified and/or overexpressed in up to one- treatment of metastatic breast cancer.
third of all breast cancer specimens (Figure
17–2). When given to a heavily pretreated group CONCLUSION
of patients whose tumors overexpressed this
gene product, trastuzumab produced a 16 per- Metastatic breast cancer is responsible for over
cent objective response rate.81 Toxicity was min- 40,000 deaths of American women each year,
imal, consisting of fever and chills after the first with most of these women having lived with
weekly infusion, and mild pain, asthenia, nausea, metastatic disease for 2 or more years prior to
diarrhea, and dyspnea. In addition, 5 percent of their death. During this time, many symptoms
patients had evidence of cardiac dysfunction. can be palliated or avoided by addressing both
Trastuzumab was also studied in a placebo- local problematic sites with surgery or radia-
controlled study involving women simultane- tion therapy and the overall course of the dis-
ously receiving chemotherapy with either dox- ease with hormonal therapy or chemotherapy.
orubicin-cyclophosphamide or paclitaxel.82 At all times, the impact of a therapeutic inter-
When added to doxorubicin-cyclophosphamide, vention on quality of life must be weighed, as
the response rate increased from 43 to 52 percent, many options, particularly second- or third-line
with a 3.6-month prolongation of responses. therapies, offer little chance of prolonging life.
When trastuzumab was added to paclitaxel in Improvements in breast cancer prevention,
patients who had prior exposure to doxorubicin, early detection, and postsurgical adjuvant ther-
response rate increased from 16 to 42 percent, apy are likely to reduce the overall mortality of
and duration of response from 4.4 to 11 months. breast cancer by reducing the number of women
Unfortunately, cardiotoxicity was seen in 27 per- who suffer metastatic recurrence. Those women
cent of women receiving the doxorubicin combi- who nonetheless are forced to contend with
metastases will have an increasing number of
options in coming years. Newer chemothera-
peutic drugs, hormonal agents, and immuno-
logic approaches offer the hope of more selec-
tive, less toxic, and ultimately more effective
treatments for metastatic breast cancer.

REFERENCES
1. Early Breast Cancer Trialists’ Collaborative
Group. Tamoxifen for early breast cancer: an
overview of the randomised trials. Lancet
1998;351:1451–67.
2. Landis S, Murray T, Bolden S, et al. Cancer sta-
tistics, 1999. CA Cancer J Clin 1999;49:8–31.
3. Valagussa P, Bonadonna G, Veronesi U. Patterns
Figure 17–2. A breast cancer showing overexpression of
membrane associated HER-2 protein. Metastatic breast can-
of relapse and survival following radical mas-
cer with this phenotype demonstrates a 16% response rate tectomy. Cancer 1978;41:1170.
to trastuzumab when employed as a single agent. 4. Borst MJ, Ingold JA. Metastatic patterns of inva-
Treatment of Metastatic Breast Cancer 283

sive lobular versus invasive ductal carcinoma 18. Galicich JH, French LA, Melby J. Use of dexa-
of the breast. Surgery 1993;114:637. methasone in treatment of cerebral edema asso-
5. Harris M, Howell A, Chrissohou M, et al. A com- ciated with brain tumors. Lancet 1961;1:46.
parison of the metastatic pattern of infiltrating 19. Cohen N, Strauss G, Lew R, et al. Should prophy-
lobular carcinoma and infiltrating duct carci- lactic anticonvulsants be administered to
noma of the breast. Br J Cancer 1984;50:23. patients with newly diagnosed cerebral metas-
6. Lamovec J. Metastatic pattern of infiltrating lobu- tases? a retrospective analysis. J Clin Oncol
lar carcinoma of the breast: an autopsy study. J 1988;6:1621.
Surg Oncol 1991;48:28. 20. Patchell R, Tibbs P, Walsh J, et al. A randomized
7. Van De Vijver M, Mooi W, Wisman P, et al. trial of surgery in the treatment of single
Immunohistochemical detection of the neu metastases to the brain. N Engl J Med 1990;
protein in tissue sections of human breast 322:494–500.
tumors with amplified neu DNA. Oncogene 21. Borgelt B, Gelber R, Kramer S, et al. The palliation
1988;2:175–8. of brain metastases: final results of the first of
8. Rouesse J, Friedman S, Guash-Jordan I, et al. Sur- two studies by the Radiation Therapy Oncology
vival effect of systemic therapy on patients Group. Int J Radiat Oncol Biol Phys 1980;6:1.
developing metastatic breast carcinoma. Breast 22. Flickinger JC, Kondziolka D, Lansford LD, et al. A
Cancer Res Treat 1990;15:13. multi-institutional experience with stereotactic
9. Perez JE, Machiavelli M, Leone BA, et al. Bone- radiosurgery for solitary brain metastases. Int J
only versus visceral-only metastatic pattern in Radiat Oncol Biol Phys 1994;28:979.
breast cancer: analysis of 150 patients. Am J 23. Rosner D, Taukuma N, Lane W. Chemotherapy
Clin Oncol 1990;13:294. induces regression of brain metastases in
10. Blumenschein GR, Pinnamaneni K, Buzdar AU. breast carcinoma. Cancer 1986;58:832.
Combined regional and systemic therapy in 24. Colomer R, Cosos D, Del Campo JM, et al. Brain
breast cancer patients with an isolated metasta- metastases from breast cancer may respond to
sis with or without prior chemotherapy. In: endocrine therapy. Breast Cancer Res Treat
Jones SE, Salmon SE, editors. Adjuvant ther- 1988;12:83.
apy of cancer. IV. Orlando: Grune and Stratton; 25. Tsukada Y, Fouad A, Pickren JW, et al. Central
1984. p. 311. nervous system metastasis from breast carci-
11. Vogel C, Azevedo S, Hilsenbeck S, et al. Survival noma: autopsy study. Cancer 1983;52:2349.
after first recurrence of breast cancer. Cancer 26. Ongerboer de Visser BW, Somers R, Nooyen WH,
1992;70:129–35. et al. Intraventricular methotrexate therapy of
12. Clark G, Sledge G, Osborne C, et al. Survival leptomeningeal metastasis from breast carci-
from first recurrence: relative importance of noma. Neurology 1983;33:1565.
prognostic factors in 1,015 breast cancer 27. Wasserstrom WR, Glass JP, Posner JB. Diagnosis
patients. J Clin Oncol 1987;5:55–61. and treatment of leptomeningeal metastases
13. Blanco G, Holli K, Heikkinen O, et al. Prognostic from solid tumors: experience with 90 patients.
factors in recurrent breast cancer: relationships Cancer 1982;49:759.
to site of recurrence, disease-free interval, 28. Sze G, Soletsky S, Bronen R, et al. MR imaging
female sex steroid receptors, ploidy and histo- of the cranial meninges with emphasis on con-
logical malignancy grading. Br J Cancer trast enhancement and meningeal carcinomato-
1990;62:142–6. sis. AJNR Am J Neuroradiol 1989;10:965.
14. Qazi R, Chuang JL, Drobyski W. Estrogen recep- 29. Yap HY, Yap BS, Rasmussen S, et al. Treatment
tors and the pattern of relapse in breast cancer. for meningeal carcinomatosis in breast cancer.
Arch Intern Med 1984;144:2365–7. Cancer 1982;49:219.
15. DiStefano A, Yap HY, Hortobagyi GN, et al. The 30. Raju RN, Kardinal CG. Pleural effusion in breast
natural history of breast cancer patients with carcinoma: analysis of 122 cases. Cancer 1981;
brain metastases. Cancer 1979;44:1913. 48:2524.
16. Posner JB. Neurologic complications of systemic 31. Hausheer FH, Yarbro JW. Diagnosis and treatment
cancer. Med Clin North Am 1979;63:783. of malignant pleural effusion. Semin Oncol
17. Davis PC, Hudgins PA, Peterman SB, et al. Diag- 1985;12:54.
nosis of cerebral metastases: double-dose 32. Hagemeister FB, Buzdar AU, Luna MA, et al.
delayed CT vs contrast-enhanced MR imaging. Causes of death in breast cancer. Cancer 1980;
AJNR Am J Neuroradiol 1991;12:293. 46:162.
284 BREAST CANCER

33. Cham WC, Freiman AH, Carstens PHB, et al. cancer [abstract]. Radiother Oncol 1992;24
Radiation therapy of cardiac and pericardial Suppl:S41.
metastases. Ther Radiol 1975;114:701. 48. Aberizk WJ, Silver B, Henderson IC, et al. The
34. Gillam LD, Guyer DE, Gibson TC, et al. Hydro- use of radiotherapy for treatment of isolated
dynamic compression of the right atrium: a locoregional recurrence of breast carcinoma
new echocardiographic sign of cardiac tam- after mastectomy. Cancer 1986;58:1214.
ponade. Circulation 1983;68:294. 49. Borner M, Bacchi M, Goldhirsch, et al. First iso-
35. Vaitkus PT, Herrmann HC, LeWinter MM. Treat- lated locoregional recurrence following mas-
ment of malignant pericardial effusion. JAMA tectomy for breast cancer: results of a phase III
1994;272:59. multicenter trial comparing systemic treatment
36. Tubiana-Hulin M. Incidence, prevalence and dis- with observation after excision and radiation. J
tribution of bone metastases. Bone 1991;12 Clin Oncol 1994;12:2071.
Suppl 1:S9 50. Hull D, Clark G, Osborne K, et al. Multiple estro-
37. O’Brien DP, Horgan PG, Gough DB, et al. CA 15- gen receptor assays in human breast cancer.
3: a reliable indicator of metastatic bone dis- Cancer Res 1983;43:413–6.
ease in breast cancer patients. Ann R Coll Surg 51. Wittliff JL. Steroid-hormone receptors in breast
1992;74:9. cancer. Cancer 1984;53:630.
38. Poulsen HS, Nielsen OS, Klee M, et al. Palliative 52. Jaiyesimi IA, Buzdar AU, Decker DA, Hortobagyi
irradiation of bone metastases. Cancer Treat GN. Use of tamoxifen for breast cancer: twenty
Rev 1989;16:41. eight years later. J Clin Oncol 1995;13:513–29.
39. Harrington KD. Orthopaedic management of 53. Gradishar WJ, Jordan VC. Clinical potential of new
metastatic bone disease. St Louis: CV Mosby; antiestrogens. J Clin Oncol 1997;15:840–52.
1988. p. 7. 54. Sunderland MC, Osborne CK. Tamoxifen in pre-
40. Yazawa Y, Frassica FJ, Chao EYS, et al. Metasta- menopausal patients with metastatic breast can-
tic bone disease: a study of the surgical treat- cer: a review. J Clin Oncol 1986;9:1283–97.
ment of 166 pathologic humeral and femoral 55. Fisher B, Costantino J, Wickerham D, et al.
fractures. Clin Orthop 1990;251:213. Tamoxifen for prevention of breast cancer:
41. Morton AR, Cantrill JA, Pillai GV, et al. Sclerosis report of the national surgical adjuvant breast
of lytic bone metastases after disodium amino- and bowel project p-1 study. J Natl Cancer Inst
hydroxypropylidene bisphosphonate (APD) in 1998;90:1371–88.
patients with breast carcinoma. BMJ 1988; 56. McDonald CC, Alexander FE, Whyte BW, et al.
297:772. Cardiac and vascular morbidity in women
42. van Holten-Verzantvoort ATM, Kroon HM, receiving adjuvant tamoxifen for breast cancer
Bijvoet OLM, et al. Palliative pamidronate in a randomized trial. The Scottish Cancer Tri-
treatment in patients with bone metastases als Breast Group. BMJ 1995;311:977–80.
from breast cancer. J Clin Oncol 1993;11:491. 57. Howell A, Dodwell DJ, Anderson H, Redford J.
43. Robinson RG, Blake GM, Preston DF, et al. Stron- Response after withdrawal of tamoxifen and
trium-89: treatment results and kinetics in progestins in advanced breast cancer. Ann
patients with painful metastatic prostate and Oncol 1992;3:611–7.
breast cancer in bone. Radiographic 1989;9:271. 58. Dombernowsky P, Smith I, Falkson G, et al. Letro-
44. Recht A, Schnitt SJ, Connolly JL, et al. Prognosis zole, a new oral aromatase inhibitor for
following local or regional recurrence after advanced breast cancer: double-blind random-
conservative surgery and radiotherapy for early ized trial showing a dose effect and improved
stage breast carcinoma. Int J Radiat Oncol Biol efficacy and tolerability compared with mege-
Phys 1989;16:3. strol acetate. J Clin Oncol 1998;16:453–61.
45. Andry G, Suciu S, Vico P, et al. Locoregional 59. Buzdar A, Jonat W, Howell A, et al. Anastrozole
recurrences after 649 modified radical mastec- versus megestrol acetate in the treatment of
tomies: incidence and significance. Eur J Surg postmenopausal women with advanced breast
Oncol 1989;15:476. carcinoma. Cancer 1998;83:1142–52.
46. Dahlstrom KK, Andersson AP, Andersen M, et al. 60. Muss H, Case D, Cappizzi RL, et al. High versus
Wide local excision of recurrent breast cancer standard dose megesterol acetate in women
in the thoracic wall. Cancer 1993;72:774. with advanced breast cancer: a phase III trial of
47. Kenda R, Lozza L, Zucali R. Results of irradiation the Piedmont Oncology Association. J Clin
in the treatment of chest wall recurrent breast Oncol 1990;8:1797–1805.
Treatment of Metastatic Breast Cancer 285

61. Cavalli F, Goldhirsch A, Jungi F, et al. Randomized 72. Chan S, Friedrichs K, Noel D, et al. A randomized
trial of low versus high dose medroxyproges- phase III study of taxotere (T) versus doxoru-
terone acetate in the induction treatment of bicin (D) in patients (pts) with metastatic
postmenopausal patients with advanced breast breast cancer (MBC) who have failed an alky-
cancer. J Clin Oncol 1984;2:414–9. lating containing regimen: preliminary results.
62. Taylor CW, Green S, Dalton WS, et al. Multicenter ASCO 1997;540.
randomized clinical trial of goserelin versus sur- 73. Riva A, Fumoleau P, Roche H, et al. Efficacy and
gical ovariectomy in premenopausal patients with safety of different corticosteroid (C) premed-
receptor-positive metastatic breast cancer: an ications (P) in breast cancer (BC) patients
intergroup study. J Clin Oncol 1998;16:994–9. (PTS) treated with taxotere (T). Proc Am Soc
63. Harvey HA, Lipton A, Max DT, et al. Medical Clin Oncol 1997;16:188a.
castration produced by the GnRH analogue 74. Gianni L, Munzone E, Capri G, et al. Paclitaxel by
leuprolide to treat metastatic breast cancer. J 3-hour infusion in combination with bolus
Clin Oncol 1985;3:1068–72. doxorubicin in women with untreated metasta-
64. Ingle JN, Green SJ, Ahmann DL, et al. Random- tic breast cancer: high antitumor efficacy and
ized trial of tamoxifen alone or combined with cardiac effects in a dose-finding and sequence-
aminoglutethimide and hydrocortisone in finding study. J Clin Oncol 1995;13:2688–99.
women with metastatic breast cancer. J Clin 75. Huan S, Pazdur R, Singhakowinta A, et al. Low-
Oncol 1986;4:958–64. dose continuous infusion 5-fluorouracil. Can-
65. Taylor SG, Gelber RD. Experience of the Eastern cer 1989;63:419–22.
Cooperative Oncology Group with doxoru- 76. Swain SM, Lippman ME, Egan EF, et al. Fluo-
bicin as a single agent in patients with previ- rouracil and high-dose leucovorin in previously
ously untreated breast cancer. Cancer Treat treated patients with metastatic breast cancer. J
Rep 1982;66:1594. Clin Oncol 1989;7:890–9.
66. Bronchud MH, Howell A, Crowther D, et al. The 77. Fraschini G, Yap HY, Hortobagyi N, et al. Five-day
use of granulocyte colony-stimulating factor to continuous-infusion vinblastine in the treat-
increase the intensity of treatment with dox- ment of breast cancer. Cancer 1985;56:225–9.
orubicin in patients with advanced breast and 78. Romero A, Rabinovich MG, Vallejo CT, et al.
ovarian cancer. Br J Cancer 1989;60:121. Vinorelbine as first-line chemotherapy for
67. Myers CE, Chabner BA. Anthracyclines. In: metastatic breast carcinoma. J Clin Oncol
Chabner B, Collins JM, editors. Cancer 1994;12(2):336–41.
chemotherapy: principles and practice. 79. Carmichael J, Possinger K, Phillip P, et al.
Philadelphia: JB Lippincott; 1990. p. 356. Advanced breast cancer: a phase II trial with
68. Cowan JD, Neidhart J, McClure S, et al. Random- gemcitabine. J Clin Oncol 1995;13:2731–6.
ized trial of doxorubicin, bisantrene, and 80. Muss HB, Case LD, Richards F. Interrupted ver-
mitoxantrone in advanced breast cancer: a sus continuous chemotherapy in patients with
Southwest Oncology Group study. J Natl Can- metastatic breast cancer. N Engl J Med 1991;
cer Inst 1991;83:1077. 325:1342.
69. Hainsworth JD, Andrews MG, Johnson DH, et al. 81. Cobleigh MA, Vogel CL, Tripathy D, et al. Effi-
Mitoxantrone, fluorouracil, and high-dose leu- cacy and safety of herceptin (humanized anti-
covorin: an effective, well-tolerated regimen HER2 antibody) as a single agent in 222
for metastatic breast cancer. J Clin Oncol 1991; women with HER2 overexpression who
9:1731. relapsed following chemotherapy for metasta-
70. Lyass O, Uziely B, Heching NI, et al. Doxil in tic breast cancer. Proc Am Soc Clin Oncol
metastatic breast cancer (MBC) after prior 1998;17:97a.
chemotherapy: therapeutic results in two con- 82. Slamon D, Leyland-Jones B, Shak S, et al. Addition
secutive studies. Proc Am Soc Clin Oncol 1998. of herceptin (humanized anti-HER2 antibody)
71. Reichman BS, Seidman AD, Crown JPA, et al. to first line chemotherapy for HER2 overex-
Paclitaxel and recombinant human granulocyte pressing metastatic breast cancer (HER2+/
colony-stimulating factor as initial chemother- MBC) markedly increases anticancer activity: a
apy for metastatic breast cancer. J Clin Oncol randomized, multinational controlled phase III
1993;11:1943–51. trial. Proc Am Soc Clin Oncol 1998b;17:98a.
Index
Adenoid cystic carcinoma, 102–103 Atypical cells, 93 “long throw,” 71
Adenopathy, 81–82 pagetoid spread of, 93 of microcalcifications, 74
Adenosis, 52 Atypical hyperplasia needle localization, 65–68
calcifications of, 49 as biomarker, 23 open surgical, indication for, 74,
Adjuvant therapy, 201–218, 253 as indicator for mastectomy, 33, 85 84–85
benefits versus toxicity, 212–214 as risk factor, 19 scarring, 65
candidates for, 202 “Atypical vascular lesions,” 108 specimen handling, 92, 97–98,
chemotherapy, 204–208 Augmented breast 144
complications, 213 evaluation of, 58–60 specimen orientation, 144
guidelines, 208, 214 Autologous reconstruction, 181–193 stereotactic equipment, 68–79
hormone receptor status, 212 advantages, 182 stereotaxis and, 66
myelosuppression, 213 lateral thigh flaps, 191–192 techniques, 66–79
patient age, 212 latissimus dorsi flap, 188–189 vacuum-assisted, 75–77
principles of, 201 Rubens flap, 189–190 wire localization, 65–68
regimens, 205 superior gluteal flap, 190–191 wound closure, 68
side effects, 213–214 TRAM flap, conventional, Bisphosphonates, 276
trials, 204–209 182–185 Bloom’s syndrome, 3
Adriamycin, cardiac morbidity, 219 TRAM flap, free, 185–188 Bone marrow transplantation, 161,
Age of onset, 3, 5, 8 Axilla, management of, 146–148 204, 207
Alcohol consumption, 31 dissection of, 146–147 Bone metastases, 271, 276
Alkylating agents, 280–281 side effects, 146–147 bisphosphonates, 276
Aminoglutethimide, 210 staging, 147 impending fractures, 276
Anastrozole, 210 Axillary adenopathy, 43 pamidronate, 276
Androstenedione, 24 Axillary irradiation, 159 radiation therapy, 276
Aneuploidy, as biomarker, 23 Axillary lymph nodes strontium 276
Angiogenesis, 226 metastases in, 201–202 Bone scans, 271, 275
Angiosarcoma outcome, 202 for follow-up, 267–268
postradiotherapy, 108–109 Axillary node dissection, 157–159 Brachial plexopathy, 231
primary, 107 arguments against, 158 Brain metastases, 272–273
prognosis, 109 clinical features, 272
Anthracycline, 158–159, 206, 280 Bilateral reconstruction, 192–193 radiation therapy, 272, 273
Antiestrogens, 27–28 mesh, 193 stereotactic radiosurgery, 272–273
Antimetabolites, 281 Biomarkers treatment, 272
Apocrine carcinoma, 101–102 assessment, 30–31 BRCA1/BRCA2 genes, 1, 21, 166
Architectural distortion, 43, 57, 84 intermediate, 23, 91 breast cancer risk, 2
progressive, 58 Biopsy description, 21
Areolar thickening, 43 adequacy of excision, 97 male carriers, 2
Argyrophilic cytoplasmic granules, automated Tru-cut™ type, 71–72 mutations, 1–5
104 devices, 70–72 other cancers, 2
Aromatase inhibitors, 210, 278 fibroadenoma, 75 ovarian cancer risk, 2, 8–9
Ashkenazi Jews, 2, 5 fine-needle aspiration (FNA), and p53, 122
BRCA1 mutation, 21 70–71, 81–82 pathophenotype, 7–8
risk, 2 frozen section, 97 probability, 3
Ataxia-telangiectasia, 3, 22 image-guided, 65–87 prophylactic mastectomy, 33

287
288 INDEX

psychological assessment, 13 in DCIS, 54, 134 histology, 90


radiation and, 8 detection of, 47–48 Comparisons of survival, 123–124
risk, 11 distribution, 49 Computer-aided diagnosis, 61
Breast conservation therapy (BCT), dystrophic, 52 Cosmesis
221–225, 232 fat necrosis, 51–52, 57 importance of, 219
age, 223 in fibroadenoma, 51 optimal, 142, 172
chemotherapy with, 225 fine, 53 tumor size and, 141–142
contraindication to, 220, 224 indeterminate, 52–55 Cowden’s disease, 3, 21–22
defect correction, 193–195 linear, 53, 54 Cox model, 124
mastectomy versus, 221 lobular, 52 Cox multivariate model, 125
nipple-areolar complex, 222 malignant, 52–55 Cribriform DCIS, 91
Paget’s disease, 222 number, 49 invasive, 96
patient variables, 222–223 pleomorphic, 53 Cyclin, 117
radiation therapy and, 8 on preradiation mammography, Cyclophosphamide, 166, 203, 281
rare malignancies and, 99 134 Cyclophosphamide, methotrexate,
recurrence after, 133, 158, 232 rod-like, 52 5-fluorouracil (CMF), 154, 203,
recurrence risk factors, 221, 222, secretory, 51, 52 207
223 shape, 49 for men, 247
self-esteem, 219 size, 48–49 meta-analyses, 205–206
Breast preservation therapy, 141–144 skin, 50 toxicity, 213
central lesions and, 142 tubular pattern, 50, 51, 52 Cylindroma, 102
contraindications for, 142 underestimation of, 53 Cyst, 44
contraindications for, 155 vascular, 50 aspiration, 80–84
ductal versus lobular, 143 See also Microcalcifications calcium in, 50–51
efficacy of, 141 Cancer and Steroid Hormone sonography of, 44, 80–82
incision placement, 145 (CASH) Study, 25 Cystosarcoma phyllodes, 46, 104
after induction chemotherapy, Capecitabine, 281 Cytokine support, 203, 207
154–159 Carcinoembryonic antigen (CEA), 118 Cytotrophoblast, 104
Paget’s disease, 142 for follow-up, 266–267 Cytoxan, adriamycin, 5-fluorouracil
patient selection, 141–143 Carcinosarcoma, 101 (CAF), 121, 207–208
recurrence risk factors, 144 Cardiovascular disease ex-CAF, 203
recurrence study, 144 estrogen replacement therapy and, trial, 212
Breast-gastrointestinal tract cancer 26, 254
syndrome, 3 HRT and, 27 Decision matrix, 263
Chemoprevention, 19, 27–32 Dietary interventions, 31–32, 34
c-erbB-2, 119–121, 282 agents, 27–32 Digitization versus film, 71–72
abnormal, 120 definition, 19 Dimpling, 43
assays, 120–121 strategies, development of, 19 Docetaxel, 159, 161, 203
CAF and, 121 studies, 19–21 Doxorubicin, 166, 203, 206–207, 212
description, 120 trials, 27–31 complications, 213
overexpression, 23, 89, 120, 122 Chemoresistance, 154 side effects, 213
as predictive marker, 121, 202 Chemosensitivity, predicting, 166 Drug resistance, predicting, 166
scoring, 120 Chest wall recurrence, 149 Ductal carcinoma in situ (DCIS),
as therapeutic target, 121 Cholesterol, tamoxifen and, 28 143, 171
CA-15-3, 266 Chondrosarcoma, 107 adjuvant systemic therapy, 211
Calcifications, 47–55 Choriocarcinomatous differentiation, as biomarker, 23
in acini, 52 104 classification, 90–92
adenosis, 49 Cisplatin, 166, 281 comedo type, 54, 90, 132, 134
analysis of, 48–55 “Clinging carcinoma,” 90 cribriform, 91
benign, 50–52 Colloid carcinoma, 95 endocrine differentiation in, 104
biopsy of, 49 histopathology, 95 follow-up, 136–137
branching, 53, 54 presentation of, 45, 46 grading, 90–92
characteristics, 48 Comedo carcinoma, 49, 53, 54 histopathology of, 89–93
clustered, 52 DCIS, 90 incidence, 131–132
INDEX 289

with LCIS, 93 cholesterol, 254 dose-intensive, 155–157


lumpectomy with radiation, colorectal cancer, 256–257 induction, 161
132–135, 211, 227 coronary artery disease, 254–255 for men, 247
in men, 242 hip fractures, 256 paclitaxel and, 161
microcalcifications, 47, 53 menopause and, 26 standard, 155
micropapillary DCIS, 90–91 osteoporosis, 255–256 Follow-up, 263–270
mortality rate, 136 progestin, 255 alkaline phosphatase, 268
multicentric, 59 recurrence rates, 259 benefits of, 263
non-comedo, 55, 134 studies, 259–260 bone scans, 267–268
occult multicentric, 132 survival analysis, 259 chemical evaluation, 266
papillary, 91 urogenital atrophy, 257 chest radiography, 267
pathology report, 92 vasomotor instability, 257 detecting recurrent, 265
radiotherapy for, 226–227 Estrogen health-care costs, 268–269
recurrence, 132–136, 227 postmenopausal deficiency, 25 legal implications, 269
recurrent, 58 risk and, 23–27 mammography, 267
re-excision, 136 Estrogen withdrawal, 253 managed care, 268
solid, 91 Estrone, 24 quality of life, 268
standard of care, 137 Exogenous hormones, 24–27 schedule, 268
surgical management, 131–138 Extensive intraductal component timing, 266
survival rates, 135 (EIC), 55, 59, 222 tumor markers, 266
tamoxifen and fenretinide in, Extracapsular extension, 228–229 Formestane, 210
30–31 Frond-like pattern, 100
total mastectomy for, 132 False negative ratio, 263
treatment, 132–136, 141 False positive ratio, 263–264 Gail Model, 13, 20
Van Nuys system, 91–92, 134–135 Family history, 16, 21–22 Galactocele, 44
Ductal casts, 53, 54 clinical features, 3 Galactography, 55–56
Ductography, 55–56 as indicator for mastectomy, 33 Gemcitabine, 281
pedigree, 4, 5, 6 Genetic counseling, 9–13, 16
Earlier detection, 19, 220 risk assessment, 3 algorithm, 10
Early onset, 33 Fat necrosis, 44, 51–52, 53, 57 depression rates, 12
Edema Fenretinide, 30 discrimination, 9–10
arm, 230 Fibroadenolipoma, 44 patient reaction, 9–13
breast, 43, 57 Fibroadenoma, 43, 103, 104–105 Genetic predisposition, 19
recurrent, 57 biopsy of, 75 Genetic therapies, 14–15
Elston scheme, 116–117 calcifications in, 51, 52 Genetics, 1–16
Endocrine differentiation, 104 follow-up, 84–85 Genistein, 31–32
Endocrine metaplasia, 104 Fibrosarcoma, 107–108 Gonadotropin-releasing hormone,
Endogenous hormones, 23–24 Fibrous histiocytoma, 107–108 209–210
Endometrial cancer, 26 Fine-needle aspiration (FNA), 70 Goserelin, 210, 211
Epidermal growth factor receptor limitation, 140 Growth factors/receptors, 119
(EGFR), 119, 121–122 pitfalls of, 71
overexpression, 122 sensitivity of, 71 Halstedian theory, 202
Epirubicin, 206 specificity of, 71 Hamartoma, 44
Epithelial hyperplasia, 105 stereotactic guidance, 70 Headache, 273
Essential oils, 31 ultrasound-guided, 81–82 Hemangiopericytoma, 107
Estrogen receptor status, 28, versus core biopsy, 140 Hematoma, 108, 140
116–117, 203 Fish-oil supplements, 31 aspiration/biopsy, 81
Estrogen replacement therapy, 26–27, Flow cytometry, 117 on mammogram, 57
253–262 Fluid collection, on mammogram, 57 HER-2/neu, 119, 140
Alzheimer’s disease, 256 Fluorodeoxyglucose, 61 antibodies against, 166
benefits, 254–257 Fluorouracil, 203, 204, 212 Hereditary breast cancer, 1–2
benefit versus risk, 261 5-Fluorouracil, 281 characteristics of, 21–22
breast cancer on, 258 5-Fluorouracil, doxorubicin, high S-phase, 7
cardiovascular disease, 254–255 cyclophosphamide (FAC), 154 mammography, 15
290 INDEX

management of, 14–15 symmetry, 177 Leptomeningeal carcinomatosis, 273


“other,” 7 ultrasonographic evaluation of, 59 Lesion
patient education, 15 Implants, adjustable, 177–179 fat containing, 44
self examination, 15 prior radiation, 178, 190 hyperplasia, 23
surveillance, 15 Implants, expanders, 177, 178–181, intraductal, 46
testing, 16 189 metastatic, 47
types, 7–8 bilateral, 192 nonpalpable, solid, 81
Hereditary breast-ovarian cancer In situ carcinoma precancerous, 23
syndrome, 1, 21 presentation of, 43 preinvasive, 23
genetics of, 1, 3 “Indian files,” 94 risks, 23
management of, 14 Induction chemotherapy, 154–160 Letrozole, 210
pedigree, 6 Infiltrating breast carcinoma, 93–97 Leuprolide, 210
Heterocyclic amines, 31 Infiltrating ductal carcinoma Li-Fraumeni syndrome, 3, 4, 22, 122
Hodgkin’s disease, 22 architectural patterns, 94 Limonene, 31
Hormonal therapy cytology, 94–95 Lipoma, 44
aromatase inhibitors, 278 EGFR in, 122 Liposarcoma, 107
estrogen antagonists, 278 endocrine differentiation in, 104 Lobular acini, 93
metastatic breast cancer, 277–279 histopathology, 94 “Lobular cancerization,” 93
ovarian ablation, 279 imaging of, 44–45, 58, 59 Lobular carcinoma in situ (LCIS)
progestins, 278–279 Infiltrating lobular carcinoma, 45, 94 as biomarker, 23
trials, 208–211 Inflammatory breast cancer, 159–161 with DCIS, 93
Hormone replacement therapy Intracystic carcinoma, 99 definition, 92–93
(HRT), 26–27 Intracytoplasmic lumens, 93 endocrine differentiation in, 104
benefits, 27 Intraductal carcinoma, 222 histopathology, 92–93
breast cancer–related deaths and, Invasive breast carcinoma in men, 242
27 biomarkers of, 23 recurrent, 222
relative risk, 26–27 categories, 93–96 Lobular neoplasia, 7
studies, 26 grading, 96–97 Locally advanced breast cancer,
Hysterectomy, prophylactic, 33–34 histopathology, 93–97 153–169
with oophorectomy, 33–34 Invasive cribriform carcinoma, 96 axillary node dissection, 156–157
Invasive ductal carcinoma, 58, 97 breast conservation, 153, 157–159
Ifosfamide, 281 Invasive lobular carcinoma, 106, 143 definition, 153
Immunotherapy, 282 mitotic rate in, 117 goals, 153
Implants, 174–181 irradiation, 158
advantages, 181–182 Juvenile carcinoma, 103 multimodality therapy for, 229
bilateral, 192 radiotherapy for, 227–229
capsular contracture, 175, 180, 231 Kaplan-Meier survival curves, recurrence, 158
capsular fibrosis, 181 123–124, 160 survival, 158
delayed reconstruction, 177 Keratin pearls, 103 TNM, 153
device failure, 181 Keratohyaline granules, 103 Log-rank test, 123
disadvantages, 181 Ki-67, 117 Lumpectomy, 66, 221, 226, 227
dual chamber, 174, 178 boost irradiation, 224–225
evaluation, 58–60 Lactating women, imaging, 42, 80 cosmetic outcome, 224
extracapsular extravasation, 176 Lateral thigh flaps, 191–192 for DCIS, 135–136
irradiation and, 162 Latissimus dorsi flap, 162, 163, with radiation, 132–135
lawsuits, 176–177 188–189 recurrence risk factors, 144–145
mammography and, 176 advantage, 162 for Stage I, II, 141
MRI evaluation of, 60 for defect repair, 195 Lymph node involvement, 147
nonadjustable, 174 disadvantage, 162 Lymph node metastases
position, 177 expander, 189 as marker, 115
primary reconstruction, 177 indications for, 188 Lymphatic drainage patterns, 147
rupture, 175–176, 181 Lead-time bias, 264 Lymphedema, 147
saline, 174,181 Leiomyosarcoma, 107 Lymphocytic infiltrate, 8
silicone gel, 174, 175–176, 181 Length-time bias, 264–265 periductal, 90
INDEX 291

Lymphoplasmacytic infiltrate, 96 risk factors, 240–242 spiculated, 43, 44


Lymphoscintigraphy, 148 sarcomas, 242 status, 133–134, 223
Lynch syndrome, 8 similarities to women, 239–240 ultrasound of, 44
skin involvement, 245 Markers, 115–125, 119
Magnetic resonance imaging (MRI), survival, 244–249 evaluation of new, 114, 125
60–61 tamoxifen, 247–248 goal of using, 118
of benign lesions, 60 testicular function, 241 guidelines, 115
evaluation of implants, 60 TNM stage, 243, 245 predictive, 114, 119
false positives, 60 transsexuals, 241 prognostic, 114, 118–119, 125, 225
limitations of, 60–61 treatment, 246–248 statistical issues, 122–125
of malignant lesions, 60 tumor markers, 247 studies, 118–119
measuring tumor response, 166 types, 242 trials, 122–123
of occult in situ carcinoma, 60 ultrasound, 243–244 types, 114
sensitivity of, 60 Malignancy Mass, palpable, 42
specificity, 60 classification of, 89–98 Mass
Males, breast cancer in, 21, 228, secondary signs, 43 analysis of, 43–44
239–252 signs of, 43 discrete, 43
age, 240 sonographic features of, 44 hypoechoic, 44
androgen deficiency, 241 Malignant ascites, 275 intracystic, 46, 47
androgen-receptor gene, 240 Malignant pleural effusions, 274–275 irregular, 43
axillary metastases, 245 Mammography mammography of, 43–44
biopsy, 244 assessment categories, 42 margin of, 43
BRCA1/BRCA2, 240 baseline, 68 shape, 43
comorbidities, 249 with biopsy, 66–68 Mastectomy defect, 193–195
DCIS, 242 breast cancer abnormalities, 43 classification, 194
ductal hyperplasia, 241 decision categories, 42 flap choice, 195
EGFR in, 122 diagnostic, 41 inferior, 194
epidemiology, 240–242 follow-up, 267 inferior pole, 195
estrogen metabolism in, 24 implants and, 58–60 recurrence risk, 193
estrogen receptor status, 246 localization, 66 supra-areolar, 195
evaluation, 244 for long-term follow-up, 56–57 surveillance, 195
family history, 240 magnification, 41, 55, 56–57 technique, 194
gynecomastia, 241, 243–244 for monitoring, 56 upper quadrant, 194
head injury, 241 postoperative, 56 Mastectomy, prophylactic, 32–33,
history of, 239 postradiation, 56–60 146
hyperprolactinemia, 241 radiation risk, 22 bilateral, 32, 146
incidence, 240 radiotherapy, 68 candidates for, 33
Klinefelter’s syndrome, 24, 240, recommendations, 41 indications for, 33
241 reporting, 42 unsuspected cancers found, 33
LCIS, 242 screening, 41–44 Mastectomy, radical, 146, 221
liver disease and, 240 specimen, 92 Mastectomy, skin-sparing, 172,
localized, 246–248 spot compression, 41 173–177
lung carcinoma, 242 Margin distance, optimal, 144 complications, 174
mammography, 243 Margin involvement, 144 esthetic results, 174
metastatic, 248 Margins, biopsy indications for, 173
nipple discharge, 242 clear, 223 recurrence with, 174
node number, 245, 246 intraoperative inking of, 66–67, Mastectomy, subcutaneous
Paget’s disease, 242 223 defined, 32
papillary carcinoma in, 99 negative, 223 prophylactic, 33
pathology, 242 radiation boost, 225 Mastectomy, total, 144–145
presentation, 242–244 Margins, tumor for DCIS, 132
prognosis, 239, 244, 249 assessment of, 43, 66, 92 defined, 32
prostatic carcinoma, 241 ill-defined, 43 incision placement, 145
receptor status, 244, 248 imaging, 45 indications for, 144
292 INDEX

with reconstruction, 145 Multicentric disease, 143, 155, Osteosarcoma, 108


skin sparing, 145 221–222 Ovarian carcinoma, 8–9
Medroxyprogesterone, 210 Multiple myeloma, 49 BRCA1/BRCA2 mutations and, 8
Medullary carcinoma, 95–96 Multivariate analyses, 125 surveillance, 15
atypical, 96 Mutation search, 2–3
EGFR in, 122 p53, 19, 140
histopathology, 95–96 Necrosis, 91, 95 adenovirus for, 166
presentation of, 45, 46 Necrotic neoplasm, 44 as biomarker, 23, 122, 202
Melphalan, 203, 212 Negative predictive value, 264 mutations, 122
Menstrual factors, 24 Neoadjuvant therapy, 211 tamoxifen and, 225
Mesh, 187–188 Nicotine use, 184 Paclitaxel, 159, 161, 203
prolene, 193 Nipple retraction, 43 Paget’s disease, 43, 142, 222
Metaplastic carcinoma, 95, 103, 107 Nipple thickening, 43 in men, 242
histopathology, 101 Nipple-areolar reconstruction, Pamidronate, 276
Metastatic, treatment, 271–285 195–196, 222 Papillary carcinoma, 44–45, 47, 95,
biopsy, 271–272 Nipple discharge 99–101
bone involvement, 271, 276 ductography, 55–56 Papillary DCIS, 91
bone scans, 271, 275 galactography, 55–56 Parenchyma, irregular, 57
brain, 272–273 in males, 242 Pectoralis muscle, invasion of, 146
chemotherapy, 279–281 malignant, 99 Pericardial effusions, 274
immunotherapy, 281 serosanguinous, 45, 56 tamponade, 274
leptomeningeal carcinomatosis, “No special type” (NST) invasive Perillic acid, 31
273–274 carcinomas, 7, 94, 97 Perillyl alcohol, 31
local, 276–279 Node number, 154 Peritoneal metastases, 275
maintenance, 281 Noninvasive intracystic carcinoma, 99 Phyllodes tumor, 46–47, 48,
malignant effusions, 274–275 Normal breast anatomy, 81 103–106
peritoneal, 275 Nuclear grading, 116 classification, 105
prognosis, 272 Nuclear medicine techniques, 61 differentiation, 106
recurrence rate, 271 Nulliparity, 25 excision, 106
sites, 271 histopathology, 105
Methotrexate, 121, 156, 203, 281 Obesity, postmenopausal incidence, 104–105
Methotrexate, 5-fluorouracil, as risk factor, 24 presentation, 104
vinblastine (MFVb), 155 Omega-3 polyunsaturated fatty acids, Polarity, cell, 90–91
Microcalcifications 31 Positive predictive value, 264
follow-up, 84 Omega-6 polyunsaturated fatty acids, Positron emission tomography
as indicator for mastectomy, 33 31 (PET), 61, 147
monitoring, 56–57 Oncogenes, 119 as predictor of response, 166
visualization of, 41 Oophorectomy, 203, 209–210 sensitivity, 61
Microlumens, secondary, 91 prophylactic, 11, 13, 15, 33–34 specificity, 61
Micropapillary DCIS, 90–91 protective effect of, 23–24 Postexcision monitoring, 56
Milk of calcium, 51 trials, 209–210 Postradiotherapy angiosarcoma, 108
Mitotic count, 7–8 Oral contraceptives, 259 Predictive value, 264
as marker, 117 as risk factor, 24–25, 27 Pregnancy, 258–259
Monoclonal antibodies, 119, 121, 166 studies, 24–25 abortion as risk factor, 24
Monomorphism, 90–91 survival, 259 BCT, 224
Monoterpenes, 31 Osseous metaplasia, 101 imaging, 42, 80
Mortality, decline in, 19 Osseous trabeculae, 107 protective factor of, 24
Mortality rate, 264 Osteogenic sarcoma, 107 radiation, 224
Mucin globules, 93 Osteoporosis Prevalence of disease, 264
Mucinous carcinoma, 46 estrogen replacement therapy Primary lymphoma, 106
endocrine differentiation in, 104 and, 26 Probability (p) values, 124
histopathology, 95 tamoxifen and, 28–29 significance, 125
INDEX 293

Progestins, 26, 255, 278–279, routine for follow-up, 267 Sarcoma, breast, 107–108
trials, 210 Raloxifene in men, 242
Progression, 23 as chemoprevention, 29, 34 Sarcoma, postradiotherapy, 108
Proliferating cell nuclear antigen, 117 with tamoxifen, 30 Scarff-Bloom-Richardson system,
Proliferation rate, 115 trials, 29–30 96–97
as marker, 117 ras Gene, 119 Scintigraphic imaging, 61
Proliferative breast disease, 23 Reconstructive surgery, 161–166, Sclerosing adenosis, 52
Prophylactic mastectomy, 13–14 171–200 Sclerosing papillomatosis, 46
attitudes toward, 12 adjuvant chemotherapy, 172 Secretory carcinoma, 103–104
Proportional hazard statistical autologous, 181–193 Seizures, 272
models, 124 bilateral, 192–193 Selective estrogen receptor
Pseudocalcification, 50 delayed, 145, 172, 185 modulators (SERMs), 29–30, 34,
Pseudosarcomatous metaplasia, 101 expanders, 177, 178–181, 189 213
Pseudosarcomatous stroma, 101 flaps, 182–193 Sentinel lymph node
immediate, 171–173, 179–181, biopsy, 159, 231
Radial scar, 46, 48 185, 195 mapping, 229
Radiation, 219–238 implants, 145, 162, 173, 174–181 Sentinel lymphadenectomy, 147–148
for bone metastases, 276 mastectomy defect, 193–195 advantages, 148
boost doses, 223, 230 nipple-areolar, 195–196, 222 isosulfan blue, 148
brain, 273 radiation with, 173 radiocolloid, 148
BRCA1/BRCA2 mutations and, 8 Rectus abdominis myocutaneous Seroma
carcinogenic potential, 231 flaps, 162 aspiration/biopsy, 81
carcinogenic risk, 22 Recurrence on mammogram, 57
cardiac injury, 230, 232 after BCT, 221–225 Shadowing, 44
cardiac toxicity, 227 cytologic factors, 225–226 Signs of malignancy, 43
chemotherapy and, 229 detecting, 265 Skin changes, 108
chest wall, 227–229 mammographic indications of, Skin thickening, 43, 57–58
collagen vascular diseases, 224 57–58 Small-cell undifferentiated
complications, 230–231 metastatic, 276–282 carcinoma, 104
contralateral breast cancer, 231 risk factors for, 221 Smoking, 31, 184
for DCIS, 226–227 Relapse Solid DCIS, 91
environmental, 22 patterns of, 202–203 histopathology, 91
factors, 224 radiation therapy, 202 Sonography
local recurrence, 231 risk of, 201–202 features of malignancy, 44
for locally advanced breast survival, 202 of masses, 44
cancer, 227–230 Reporting, standardizing, 98 Soy-based products, 31
lumpectomy and, 221 9-cis-Retinoic acid, 29–30 “Special type,” tumors, 94–96
orthovoltage, 228 Retinoids, as chemoprevention, 28, Spindle-cell carcinoma, 101
post complete response, 229–230 30, 34 Squamous cell carcinoma, 101
postmastectomy, 231 Rhabdomyosarcoma, 107 histopathology, 103
pregnancy and, 224 “Rigid bridges,” 91 Squamous metaplasia, 103, 105
pulmonary injury, 230 Risk assessment, 12, 19–27 Stage I and II
recurrence rates, 221 absolute risk, 20–21 adjuvant therapy, 149
sarcomas, 231 family history, 16, 21–22 diagnostic evaluation, 139–140
stereotactic, 273–274 genetic counseling, 9–13 fine-needle aspiration, 140
tangential, 230, 230 predisposition testing, 9 recurrence, 149
techniques, 229–230 quantification, 20 staging, 140, 141
toxicity of, 220 Risk biomarkers, 20 stereotactic core biopsy, 139
Radiation-induced DNA damage, 14 Risk factors, 19 therapy, 140–149
Radiation injury, 142 Risk, modulation of, 32 treatment algorithm, 141
Radiography “Roman bridges,” 90 tumor size, 141–142
magnification, 48 Rubens flap, 189–190, 191 Stem cell transplant, 207–208, 215
294 INDEX

Stereotactic biopsy, 68–79 contralateral breast cancer, 28 benefits of, 190


anesthetic, 72 endometrial cancer, 28–29 bilateral, 192
automated Tru-cut™, 71–72 estrogen receptor status and, 116 bipedicled, 188
complications, 77 gastrointestinal malignancies, 28 complications, 187–188
dedicated prone, 69–70, 69 invasive breast cancers, 29 for defect repair, 195
difficulties in, 77–79 long-term treatment, 28 disadvantage, 186
digital imaging, 71–72 for men, 247–248 fat necrosis, 188
equipment, 68–72 noninvasive breast cancer, 29 free versus pedicled, 186–188
indications for, 84 raloxifene with, 29–30 hernias, 186–187
pull-back depth, 73, 74 resistance, 122 high-risk patients, 185, 188
scout image, 72 side effects of, 29, 213–214 mesh, 187–188
stroke margin, 77, 79 studies, 28–29 modifications, 187
targeting errors, 77, 78 term, 214 Trastuzumab, 282
technique, 72–75 trials, 208–211 Tru-cut™ biopsy, 71–73
upright add-on, 68 uterine cancer with, 213–214 True negative ratio, 263
Stereotaxis, 66, 70 vasomotor instability, 257 True positive ratio, 263
standardization of, 71 Taxanes, 203, 280 Tubular carcinoma, 95–96
Steroid receptors, 116–117 Taxol resistance, 121 histopathology, 95
as markers, 116 Technetium-99M (Tc-99m), 61 presentation of, 46, 47
positivity, 116 Test evaluation, 265–269 Tubular-lobular group, 7
progesterone receptors, 116–117 Test sensitivity, 263–264 Tubule formation, 7
scoring systems, 116 Test specificity, 264 Tumor
Strontium 89, 276 Therapeutic modeling, 117 estrogen receptor status, 28–29
Study of Tamoxifen and Raloxifene Thiotepa, 203, 204 grade, 92, 116, 202, 222
(STAR), 27, 30 Thymidine, 117 size, 89, 92, 115, 202, 222
Subtyping, 117 TNM (tumor, nodes, metastases) subtyping, 117
Superior gluteal flap, 190–191 staging system, 140, 141, 153 Tumor cells
Surrogate end-point biomarkers Toremifene, 210 linear files, 94
(SEBs), 19–20, 23 TRAM, conventional, 182–185 “targetoid,” 94
trials, 30–31 bilateral, 192 Tumor perimeter, 7–8
Surveillance, 12, 263–270 blood flow in, 182
data analysis, 263–264 complications, 184–185 Ultrasonography, 41, 79–80
Hodgkin’s disease, 22 description, 182 of augmented breast, 59
Surveillance, Epidemiology, and End macromastia, 185 whole breast, 80
Results (SEER) program, 25–26 outcome, 186 Ultrasound-guidance
Survival, apparent increase, 264 patient selection, 184 advantages, 84
Syncytiotrophoblast, 104 perfusion in, 182–184 core-needle biopsy, 81, 83
Systemic therapy perioperative conditions, 183–184 cyst aspiration, 81–84
improving, 166–167 postoperative monitoring, 183 selecting, 84
metastatic breast cancer, 277 pre-existing scars, 184 Univariate analyses, 124–125
radiation of, 173
Tamoxifen, 203, 225 strategy, 184 Vacuum-assisted biopsy, 75–77
as adjuvant treatment, 146 technique, 183 versus core-needle, 76–77
benefits, 214 zones, 183 Van Nuys system, 91–92, 134–135,
bone mineral density, 28–29 TRAM, free, 185–188 227
cardiac events, 29 advantages, 185, 187, 192 Vertical rectus abdominis
as chemopreventive agent, 27–29, bilateral, 192 myocutaneous (VRAM), 162–164
34, 146 contraindications to, 186 Vinblastine, 156, 203, 281
chemotherapy plus, 210–211 outcome, 186 Vinca alkaloids, 281
cholesterol, 28 technique, 185 Vincristine, doxorubicin,
clotting factors, 28 Transverse rectus abdominis cyclophosphamide, prednisone
colorectal cancer, 28 myocutaneous (TRAM) flap, 33, (VACP), 155
complications of, 29 162–164 Vinorelbine, 281

Potrebbero piacerti anche