Sei sulla pagina 1di 15

Journal of Controlled Release 254 (2017) 92–106

Contents lists available at ScienceDirect

Journal of Controlled Release


journal homepage: www.elsevier.com/locate/jconrel

Targeting tumor associated macrophages (TAMs) via nanocarriers MARK


a,1 a,1 a a a
Yuvraj Singh , Vivek Kumar Pawar , Jaya Gopal Meher , Kavit Raval , Animesh Kumar ,
Richa Shrivastavab, Smrati Bhadauriab, Manish K. Chourasiaa,⁎
a
Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
b
Toxicology Division, CSIR-Central Drug Research institute, Lucknow 226031, India

A R T I C L E I N F O A B S T R A C T

Keywords: Recruitment of inflammatory cells to tumor has been well documented, with the most frequent inhabitants being
TAMs macrophages termed as tumor associated macrophages, (TAMs). Their presence was thought to be an evidence of
Nanocarriers immune system initiating a fight response towards the tumor, i.e. immune surveillance. This is the case too
Active targeting initially, when TAMs majorly exhibit an M1 phenotype, but their continued presence in tumor microenviron-
Tumor environment
ment brings about their polarization to M2 phenotype, which not only participate in continued sustenance of
Immunosuppression
Recruitment
existing tumor but also open up deleterious avenues for further progression and metastasis of cancer. Current
Polarization perspective is built around this very premise and focuses specifically on TAMs and how they are being targeted
by researchers working in annals of nanomedicine. To do so, we dwell into tumor microenvironment and focus
on nanotechnology based drug delivery aspects which have either been already or can be potentially employed
in the future to target tumor associated macrophages for improved immunoadjuvant therapy of cancer.

1. Introduction of macrophages present in tumor stroma [conveniently christened as


tumor associated macrophages (TAMs)]. TAMs present two subtypes i.e.
Balance of homoeostatic machinery is an important regulator of classically activated macrophages (M1) and alternatively activated
human well-being. Even under exposition of harshest insults, home- macrophages (M2). The process of phenotypic conversion of one sub
ostasis attempts to preserve body's integrity, i.e. it tries to restore basal type into another is termed as polarization of TAMs. M1 cells have
structural and functional levels. An immune response can be purported strong antigen presenting capability and facilitate immunological
as the perfect example, wherein, homeostatic machinery actively tries response against tumor. They raise proinflammatory cytokines such
to cure the human body when faced with stress. Immune response, as TNFα, IL-12, and IL-23 and enhance release of reactive oxygen
regardless of stimulant, scope, or context, is incessantly ubiquitous. species (ROS) and nitric oxide (NO) which provides an antitumor
However, there are circumstances when an intended immune response response. Moreover, they also express high levels of major histocom-
jeopardizes entire pretext for which that response was generated in the patibility complex: class I and class II molecules, raising probability of
first place; case in point being etiologic role of immune system in recognizing tumor specific antigens. Contrarily M2 subset has strong
progression of cancer. Immunoreactivity towards carcinoma cells is anti-inflammatory activity. It enhances release of IL-4, IL-10, and IL-13
expressed by lymphoid infiltration into tumor microenvironment and and has poor antigen presenting characteristics which suppresses T cells
regional lymphatic alterations, specifically phenotypic differentiation and generation of ROS and/or NO. IL-4 and IL-10 have negative

Abbreviations: TAMs, Tumor associated macrophages; Akt, Protein kinase B; CAT, Catalase; CCL, Chemokine (C-C motif) ligand; CD, Cluster of differentiation; COX, Cycloxygenase; CpG,
Unmethylated sequences of DNA that have immunostimulatory properties; CSF, Colony-stimulating factor; CXCL, Chemokine (C-X-C motif) ligand; CXCL10, C-X-C motif chemokine 10
also known as IP10; CXCL9, Chemokine (C-X-C motif) ligand 9; CXCR1, Interleukin 8 receptor α; CXCR2, Interleukin 8 receptor β; EGF, Epidermal growth factor; EPR, Enhanced
permeability and retention; ERK, Extracellular signal–regulated kinases; GPx1, Glutathione peroxidase 1; HIF, Hypoxia-inducible factors; IKK-β, Inhibitor of nuclear factor kappa-B kinase
subunit beta; IL, Interleukin; IP10, Interferon gamma-induced protein 10; JNK, c-Jun N-terminal kinases; M1, M1 subtype of tumor associated macrophage; M2, M2 subtype of tumor
associated macrophage; MAPK, Mitogen-activated protein kinase; M-CSF, Macrophage colony-stimulating factor also known as CSF1; MDC, Macrophage-derived chemokine also known
as CCL22; MIG, Monokine induced by gamma interferon; MIP-1 β, A protein which in humans is encoded by the CCL4 gene; MMP, Matrix metalloproteinase; MRI, Magnetic resonance
imaging; MYO3A, A gene which encodes for the protein Myosin IIIA; NFκB, Nuclear factor kappa-light-chain-enhancer of activated B cells; NO, nitric oxide; p38, Class of mitogen-
activated protein kinases; PD-L1, Programmed cell death 1; PEG, Polyethyleneglycol; PET, Positron emission tomography; PI3K, Phosphoinositide 3-kinase; ROS, Reactive oxygen species;
SESN 3, A protein; SOD, Superoxide dismutase; STAT, Signal transducer and activator of transcription; TGF, Transforming growth factor; TRAIL, TNF-related apoptosis-inducing ligand;
TNFα, Tumor necrosis factor alpha; VEGF, Vascular endothelial growth factor; VEGFR, Vascular endothelial growth factor receptor

Corresponding author at: Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India.
E-mail address: manish_chourasia@cdri.res.in (M.K. Chourasia).
1
Contributed equally.

http://dx.doi.org/10.1016/j.jconrel.2017.03.395
Received 21 February 2017; Received in revised form 29 March 2017; Accepted 30 March 2017
Available online 01 April 2017
0168-3659/ © 2017 Elsevier B.V. All rights reserved.
Y. Singh et al. Journal of Controlled Release 254 (2017) 92–106

Table 1
Effect of M1 and M2 polarization of TAMs on expression of various receptors and production of cytokines and chemokines.

Cellular component M1 phenotypic TAMs M2 phenotypic TAMs Effector Cytokines References

Receptors Membrane scavenger receptor A and receptor B ↓↓ ↑↑ IL-10 [5,6]


Membrane CD14 ↓ ↑↑ IL-10 [7]
Membrane scavenger receptor CD163 and Fcε receptors (CD23) ↓↓ ↑↑ IL-4 and IL-13 [8,9]
Membrane mannose receptor CD206 ↓↓ ↑↑ IL-4, IL-10 and IL-13 [10,11]
Membrane Toll-like receptor 2 (CD282) and 4 (CD284) ↑↑ ↓ IFNγ [12]
Membrane Fcγ receptors (CD16, CD32, CD64) ↑↑ ↓ IFNγ [13]
Membrane CD80 ↑↑ ↓ IFNγ [14]
Cytokine decoy Interleukin-1 R type II (CD121b) ↓↓ ↑↑ IL-4, and IL-13 [15,16]
C-C chemokine receptor type 7 (CCR7) ↑↑ ↓↓ IFNγ [17]
C-C chemokine receptor type 2 (CCR2) ↓ ↑↑ IL-10 [18]
Interleukin 8 receptor α and β (CXCR1 and CXCR2) ↓ ↑↑ IL-4 and IL-13 [19]
Cytokines TNF-α, IL-1, IL-12 and Type I IFN ↑↑ ↓↓ IFNγ [20,21]
IL-1ra ↓ ↑↑ IL-4, IL-10 and IL13 [22]
IL-10 ↓ ↑↑ IL-4, and IL-13 [23]
Chemokines Chemokine (C-X-C motif) ligand 8 9, 10 and 11 (CXCL8, CXCL9, CXCL10 and ↑↑ ↓↓ IFNγ [24–27]
CXCL11)

Chemokine (C-C motif) ligand 2, 3, 4, and 5 (CCL2, CCL3, CCL4 and CCL5)
Chemokine (C-C motif) ligand 17, 22 and 24 (CCL17, CCL22 and CCL24) ↓↓ ↑↑ IL-4 and IL-13 [23,28]
Chemokine (C-C motif) ligand 18 (CCL18) ↓↓ ↑↑ IL-4, IL-10 and IL-13 [29]

downstream effect on secretion of regulatory cytokines like IP-10 Tymoszuk et al. have demonstrated that both monocyte recruitment
(CXCL10), and MIG (CXCL9) which stall Th1 immune response; and local macrophage proliferation determines TAMs pool size in
whereas IL-4 and IL-10 released by them upregulates secretion of Th2 HER2/Neu-positive mammary carcinomas [35]. Recruitment in tumor
immune response mediators like eotaxin-2 (CCL24), CCL18 and MDC stroma is instigated by various factors like tumor induced local hypoxia,
(CCL22) [1,2]. Known list of effectors, chemokines and receptors prevalent acidosis, cytokines, etc. (Fig. 1). For instance, the soluble
modulated by TAMs is very large, however, some selected ones which chemokine CCL2, whose expression itself is regulated by activin A, is a
sub serve role in cancer dynamics are listed in Table 1. known facilitator of monocyte or macrophage accumulation in inflam-
Accumulation of TAMs in tumor stroma has significant physiologi- matory sites in vicinity of prostate [36] and breast cancer [37,38].
cal implications [3,4]. They initially inhibit, but later due to ‘self and/or Apart from its chemotactic action, CCL2 also partakes in signaling
cross’ talk promote tumor progression in various ways including which leads to polarization of monocytes to M2 subtype instigating Th2
retardation of antitumor immunologic response, stimulating angiogen- immune response [39]. Mizutani K et al. have demonstrated CCL2
esis, etc. It is therefore no wonder that scientists have tried to target this overexpressive luciferase-tagged PC3 cells attract monocytes in both in
complex interplay, “of originally intended to be good immune cells, vitro and in vivo conditions using a xenograft model. They were even
gone awry” to provide immunotherapy of cancer. Current perspective is able to significantly reduce tumor burden by administering CCL2
built around this very premise and focuses specifically on TAMs and neutralizing antibodies, which prevented recruitment of macrophages
how they are being targeted by researchers working in annals of in tumor microenvironment [40]. Other chemokines such as CXCL1,
nanomedicine. To do so, we dwell deep into tumor microenvironment CXCL8, CXCL12, CXCL15, CCL5, CCL17 and CCL22 are also observed in
and try to locate chinks in tumor armory, which might be used to tumor environment, playing variety of overt, permissive or occult roles
modulate activity of TAMs. We then focus on nanotechnology based in initial inflammation, tumor progression, recruitment and polariza-
drug delivery aspects which have either been already or can be tion of TAMs [41–43].
potentially employed in the future to target tumor associated macro- Almost all oxygen-breathing species express a highly conserved
phages for improved immunoadjuvant therapy of cancer. transcriptional complex, hypoxia-inducible factor (HIF-1) that responds
to decrease in available intracellular oxygen. In general, HIFs are vital
to embryonic development and tissue repair. However, several reports
2. Recruitment of TAMs suggest HIF-1 is deregulated in cancers due to distorted tumor milieu
which makes chronic inflammation self-perpetuating and provides axis
Peyton Rous first coined the term “subthreshold neoplastic states” for further sustenance and metastasis of cancer [44–46]. HIF activity is
for chemical and viral carcinogenesis induced somatic changes because required in angiogenesis which is mandatory to nurture rapidly
they involve irreversible DNA alterations [30]. But it was Rudolf dividing cancer cells, and consequently HIF inhibitors like acriflavine
Virchow who visualized recruitment of leukocytes in tumorous tissues and phenethyl isothiocyanate are under the scanner of clinical inves-
and proposed a link between tumor and inflammation [31]. Later Karin tigation as probable anticancer drug candidates. HIF enhances expres-
substantiated Virchow's notion by elucidating the role of inflammation sion of cytokines CXCR4 and CXCL12 which not only attract circulatory
in tumorigenesis, particularly involvement of NFκB. This study even- monocytes but also induce their polarization to M2 subtype by ERK
tually linked immunity and inflammation to cancer development and signaling [47–50]. Tripathi et al. have elucidated the mechanism for
progression; as all known targets of NFκB inhibitor, IKK-β, such as TNF- recruitment of TAMs to hypoxic regions. They identified and described
α, IL-1, IL-6 were associated with carcinogenesis [32]. These observa- role of hypoxic cancer cell derived cytokines Oncostatin M and Eotaxin-
tions strengthened the link between cancer related inflammation, the 1 in promoting recruitment of macrophages and their phenotypic
seventh hallmark of cancer, to genetic instability [33]. Thus recruit- switching to M2 subtype in solid tumors [51]. In a different study
ment of inflammatory cells to tumor became well documented with the Wen Z et al. have shown that even transient elevation of 5-lipoxygenase
most frequent inhabitants being tumor associated macrophages. Their (a metabolite produced by hypoxic cancer cells) enhances infiltration of
presence was thought to be an evidence of body initiating a fight TAMs. This hastening in migratory tendency of TAMs in response to
response towards tumor and referred to as immune surveillance [34]. In local 5-lipoxygenase levels is mediated by matrix metalloproteinase
terms of lineage, TAMs are derived either directly from circulatory (MMP)-7 via p38 pathway. The mechanism is so prudent that in in vivo
monocytes or pre-differentiated macrophages invading the periphery.

93
Y. Singh et al. Journal of Controlled Release 254 (2017) 92–106

Fig. 1. Concept of tumor associated macrophage and its physiologic implications. Any macrophage found inside tumor stroma is nominated as a tumor associated macrophage (TAMs).
These macrophages undergo phenotypic differentiation into either M1 or M2 subtype to produce opposing effects on their milieu. M1 cells produce antitumor response; whereas M2 cells
are strongly pro-tumor and tend to produce angiogenesis, immunosuppression and metastasis of cancer cells. Hypoxic tumor cells, certain growth factors, cytokines and chemokines are
strong stimuli for recruitment and activation of TAMs.

studies, 5-lipoxygenase inhibitor (Zileuton) effectively wards off infil- discussed next. To aptly signify observations made in the following
tration of TAMs [52]. Hypoxic regions in tumor microenvironment are sections, a list of ongoing clinical trials trying to establish or mitigate
therefore an established stimuli for recruitment of TAMs [53] and role of TAMs in varying cancers has also been compiled in Table 2.
appear to be a loci of substantial significance in immunotherapy of
cancer for the foreseeable future. Semaphorin 3A released from tumor
3. TAMs promote angiogenesis
hypoxic regions is yet another chemoattractant for TAMs. It works by
phosphorylating vascular endothelial growth factor receptor-1
Angiogenesis is a physiological process which generates new blood
(VEGFR1), Neuropilin-1 (Nrp1) and PlexinA1/PlexinA4 holoreceptors
vessels form pre-existing ones and is vital in growth, development, and
[54].
wound healing; however it is also fundamental in transforming a benign
In animals, intermittent hypoxia activates cyclooxygenase-2 (COX-
tumor to a malignant one. M2 phenotype of TAMs are steeply pro-
2) pathway. The consequent increase in prostaglandin E2 (PGE2)
angiogenic [58]. In a study conducted by Lie et al., TAMs were found to
accelerates tumor progression by inducing phenotypic switching of
be major agents of angiogenesis in middle T oncoprotein mice [59].
TAMs to M2 subtype. Campillo N et al. have used celecoxib, a COX-2
They enhance secretion of vascular endothelial growth factor (VEGF),
inhibitor, to prevent intermittent hypoxia induced lung tumor malig-
which is a major contributor towards angiogenesis in various cancers
nancy in a mouse model. They found that TAMs isolated from celecoxib
[60,61]. Among VEGF, the most potent and widely reported factor is
treated mouse themselves had innate ability to reduce proliferation of
VEGF-A [62]. VEGF-A and colony-stimulating factor-1 (CSF-1) have
naïve Lewis lung cancer cells while the opposite occurred when TAMs
been attributed as possible perpetrators of TAMs migration. Linde N
isolated from untreated or placbo treated mice were used. In a different
et al. found that implantation of VEFG-A in non-cancerous skin cells
study Dubey P et al. have also demonstrated role of COX-2 in
induced tumor formation during in vivo experiments [63]. Secretion of
accelerating polarization of TAMs using an antagonist flunixin meglu-
VEGF-A is regulated by hypoxia inducible transcription factors (HIF-1α
mine [55].
and HIF-2α) in tumor hypoxic regions [64,65]. However HIF-1 is not
Grimshaw MJ et al. have investigated inhibition of monocyte and
the only regulator and several other cytokines like IL1β [66], TGFβ1
macrophage chemotaxis under influence of hypoxia and inflammation
[67] and CSF-1 (M-CSF) [68] also have a say. Effect of cytokine
accompanying tumor by mitogen-activated protein kinase (MAPK)
Oncostatin M on angiogenesis has been studied by Vasse et al., on
inhibitors. It was observed that under normal circumstances without
endothelial cells in a rabbit corneal model. It was found that in vivo
inhibitor pretreatment, level of MAPK mRNA was drastically enhanced
blockade of Oncostatin M synergistically potentiated anti-angiogenic
in migratiory monocytes/macrophages whereas level of phosphorylated
effect of bevacizumab, an existing angiogenesis inhibitor [69]. Another
MAPK was much less [56] which sets up MAPK inhibitors as probable
major contributor to angiogenesis is a class of enzyme termed as matrix
immunoadjuvants capable of iterating macrophage infiltration in tumor
metalloproteinase (MMP). MMPs degrade proteins responsible for
stroma. For a more detailed overview on the bridge between TAMs and
turgidity of extracellular matrix of existing endothelial vessels. This
hypoxia, and the resulting therapeutic approaches, readers are encour-
proteolysis allows individual endothelial cells to infiltrate tumor stroma
aged to go through a perspective by Silva et al. [57]. Once recruited to
and combine with preexisting VEGF-A to form new capillaries as seen in
tumor environment, TAMs, especially the M2 subtype produce myriad
sprouting angiogenesis. Since TAMs facilitate expression of MMPs,
of responses, which both in the short as well as long run promote tumor
especially MMP-1 [70], MMP-7 [71] and MMP-9 [72], their overriding
growth and metastasis. Some of the prominent responses of macro-
role in promoting angiogenesis and tumor growth becomes extremely
phage recruitment and polarization in tumor microenvironment are
apparent. Once formed, tumor endothelium releases angiopoietin-2

94
Table 2
On-going clinical trials investigating role of tumor associated macrophages in varying cancers.

Drug Disease Target Method Study phase Clinical trials. Status Sponsor
Y. Singh et al.

gov identifier

– Lung Cancer Impact of M1/M2 tumor associated Treatment response and mortality, clinical Observational NCT00690261 Recruiting National Taiwan
macrophage polarization on cancer presentation participants University Hospital
progression and prognosis prediction
– Advanced non-small cell Correlate response and outcomes in Response to treatment of advanced NSCLC Retrospective NCT01551251 Completed Chang Gung Memorial
lung cancer advanced non-small cell lung cancer after with high or low TAMs, CD68 antibody Hospital
first line treatment staining
– Wilms Tumor and Other Relationships between tumor-associated Tumor stage, presence of vascular invasion, Observational NCT01493817 Completed Children's Oncology
Childhood Kidney Tumors macrophages and clinicopathological tumor progression, and survival. Paraffin- Group
factors in Wilms Tumor embedded specimens are analyzed for
macrophage markers
Ferumoxytol followed by MM- Solid Tumors Determine biodistribution of MM-398 and Measure tumor levels of irinotecan and SN- Interventional NCT01770353 Recruiting Merrimack
398 administered i.v. ER/PR Positive Breast feasibility of Ferumoxytol as magnetic 38, safety profile of MM-398 in the presence phase I pilot study participants Pharmaceuticals
Cancer resonance imaging to measure tumor of ferumoxytol, Tumor response rate
Triple Negative Breast associated macrophages and to predict measured by RECIST, Cmax, AUC
Cancer patient response to treatment
Metastatic Breast Cancer
With Active Brain
Metastasis
Multitargeted Tyrosine Kinase Stage I, Stage II, Stage III Targeting the prostatic tumor Grade of specific types of toxicity, MTD of Interventional NCT02472275 Recruiting Barbara Ann Karmanos
Inhibitor PLX3397 Prostate Adenocarcinoma microenvironment with PLX3397, a multitargeted tyrosine kinase inhibitor Phase I participants Cancer Institute
tumor-associated macrophage inhibitor in PLX3397, Change in TAMs levels as measured
men with unfavourable risk prostate by immunohistochemistry, Change in PSA
cancer undergoing radiation therapy and levels
androgen deprivation therapy

95
– Neuroblastoma Identify subgroups of rNB patients who Gene panel sequencing of tumor specimens, Observational NCT02868268 Recruiting New Approaches to
have potentially targetable genetic (ALK, Identify genomic alterations, presence/ participants Neuroblastoma Therapy
MAPK pathway, Metabolic-related genes) absence of immunologic biomarkers Consortium
and/or immunologic (tumor-associated
macrophage infiltration and/or
programmed death ligand [PD-L1]
expression) biomarkers in rNB
GM-CSF (sargramostim) Stage II or Stage III Colon Efficiency of GM-CSF and combination Safety of neoadjuvant and adjuvant Interventional NCT00262808 Completed University of Rochester
Cancer chemotherapy work in treating patients sargramostim (GM-CSF), proportion of Phase II
who are undergoing surgery patients with a change in tumor-associated
macrophage, VEGF expression, overall
survival
cSLO imaging Ocular Melanoma Determine whether a novel imaging Identification of macrophages Observational NCT02909517 Recruiting St. Michael's Hospital,
technique can identify presumptive tumor participants Toronto
associated macrophages (TAMs) in
patients with ocular tumors to establish
role of inflammation
– In vitro model of Impact of macrophages on de-novo Proliferation and migration of PANC1 cell Intervention NCT01921699 Unknown Rambam Health Care
pancreatic ductal chemoresistance of adenocarcinoma cells line in the tested groups, direct cell-to-cell Campus
adenocarcinoma interaction or soluble factors participate in
carcinoma MIC
– Malignant Mesothelioma Diagnostic and prognostic role of new Purification of tumor associated macrophages Observational NCT02300883 Ongoing Armando Santoro, MD
inflammation biomarkers (TAMs) and tumor cells from pleural
effusions,
mass spectrometry analysis of TAMs and
tumor cells. Confocal microscopy analysis of
macrophages, tumor cells
AMG 820 Advanced Solid Tumors Sequential dose escalation and expansion Evaluating the safety, tolerability and Interventional NCT01444404 Completed Amgen
(human IgG2 c-fms study of AMG 820 in patients pharmacokinetics of AMG 820, change in Phase I
(continued on next page)
Journal of Controlled Release 254 (2017) 92–106
Table 2 (continued)

Drug Disease Target Method Study phase Clinical trials. Status Sponsor
gov identifier
Y. Singh et al.

antagonistic antibody) tumor associated macrophages (TAMs) as


assessed by tumor biopsy
– Colorectal Cancer, Defining the scopes of specific Transcriptomal and molecular Observational NCT00979277 Unknown National University
Nasopharyngeal, immunotherapy strategies to overcome characterization of tumor associated Hospital, Singapore
Hepatocellular and Renal tumor-induced immunosuppression and monocytes/macrophages
cell carcinoma induce monocyte/macrophage-mediated
antitumor response
GM-CSF Hematologic Malignancies Study of low-dose cyclophosphamide, Immunological efficacy as indicated by T-cell Interventional NCT02709993 Yet to open Kiromic, LLC
tumor associated peptide antigen-pulsed cytokine levels Phase II
dendritic cell therapy and low dose
granulocyte- macrophage colony
stimulating factor, as consolidation
treatment
Emactuzumab and RO7009789 Solid Tumors To assess safety, pharmacokinetics, Total tumor-associated macrophages (TAMs) Interventional NCT02760797 Recruiting Hoffmann-La Roche
pharmacodynamics and therapeutic in paired-tumor biopsies Phase I participants
activity of emactuzumab and RO7009789
administered in combination
– Paediatric Solid Tumors Characterize the molecular, cellular and Evaluate tumor-associated macrophages Observational NCT01050296 Recruiting St. Jude Children's
genetic properties of primary and (TAMs) post-therapy to locate gene participants Research Hospital
metastatic neuroblastoma, osteosarcoma, expression patterns, markers and functional
retinoblastoma, Ewing sarcoma family of correlation of the tissue repair properties of
tumors, soft tissue sarcomas, macrophages
adrenocortical tumors and liver
malignancies
MCS110, Carboplatin, Advanced Triple Negative Determine whether MCS110 antibody Tumor associated macrophages (TAMs) and Interventional NCT02435680 Recruiting Novartis

96
gemcitabine Breast Cancer (TNBC) therapy improves the efficacy of Tumor infiltrating lymphocyte (TIL) content Phase II participants Pharmaceuticals
With High TAMs carboplatin and gemcitabine (carbo/gem) in pre- and post-dose tumor biopsies
Metformide Hydrochloride/ Oral Cavity Neoplasm How well ACTOplus met XR works in Change in treg, T4, T8, tumor-associated Interventional NCT02917629 Yet to open National Cancer
Pioglitazone Hydrochloride Oropharyngeal Neoplasm treating patients with stage I-IV oral macrophage-CD68 positive, PD1, PD-L1 Phase II Institute
Extended-Release Tablet cavity or oropharynx cancer that are expression, assessed in tumor tissue by IHC
undergoing definitive treatment
Paclitaxel Advanced Solid Tumors Safety, tolerability, pharmacokinetics and Pharmacodynamic marker: tumor associated Interventional NCT01494688 Ongoing Hoffmann-La Roche
RO5509554 pharmacodynamics of RO5509554 in macrophages (TAMs) expressing cells in Phase I
participants surrogate/tumor tissue
Breast Cancer Trans-tissular migration of macrophages Surgical samples of human primitive breast in Interventional NCT02876497 Yet to open Institut Claudius Regaud
associated to human breast cancer terms of histology. Isolation, differentiation
of human monocyte-derived Macrophages
(Mphs) and determination of the Macrophage
(Mph) migration mode,
Anti-Macrophage Migration Malignant Solid Tumors Safety, tolerability, pharmacokinetics, Number of participants who develop binding Interventional NCT01765790 Completed Baxalta US Inc.
Inhibitory Factor (Anti-MIF) and pharmacodynamics of anti-MIF and/or neutralizing anti-anti-macrophage Phase I
Antibody antibody migration inhibitory factor (anti-MIF)
antibodies following treatment with anti-MIF
Vaccine MAGE-3.A1 peptide, or Metastatic Melanoma Study of peptide vaccination associated Tumor response will be assessed in Phase I NCT01191034 Unknown Cliniques universitaires
the NA17.A2 peptide + IL-2, with tumoral immunomodulation with accordance with the modified RECIST version Phase II Saint-Luc- Université
IFN-α and GMCSF, proinflammatory cytokines Catholique de Louvain
Imiquimod
PD 0360324 Cyclophosphamide Malignant Neoplasm of Pre-conditioning effects of anti- Change in density of cluster of differentiation Interventional NCT02948101 Yet to open M.D. Anderson Cancer
Female Genital Organs macrophage therapy using PD 0360324 in 8 (CD8+) T cells in tumor, safety of PD Phase II Center Pfizer
recurrent platinum-resistant epithelial 0360324 followed by cyclophosphamide
ovarian cancer
Ferumoxytol-Enhanced MRI Brain Tumors or Other Monitoring extent of inflammation Obtain measurements of iron concentration Interventional NCT02452216 Recruiting Michael Iv
Conditions of the CNS associated with pathologies in the central (indicative of ferumoxytol uptake), participants
nervous system may be helpful for Determine number of macrophages in
(continued on next page)
Journal of Controlled Release 254 (2017) 92–106
Y. Singh et al. Journal of Controlled Release 254 (2017) 92–106

(Tie2) overexpressive monocytes (TEMs), which despite counter evi-


dence, seem to further endorse tumor angiogenesis [73–75] by enhan-

Centre Leon Berard


cing secretion of IL-10 [76], stimulating overexpression of mannose

Duke University
receptors [77] and down regulating TNF-α and IL-12. TEMs also
produce high levels of MMP9 and VEGF-A which continue the vicious
Sponsor

cycle of angiogenesis, recruitment, and polarization [78].

4. TAMs exhibit immunosuppressive activities


participants

participants
Recruiting

Recruiting
Retardation of inherent antitumor immune response due to pheno-
Status

typic polarization of TAMs is a major concern during chronic stages of


cancer. For example, suppression of immune response by TAMs is one
of the major reasons responsible for current incurable status of Non-
NCT02777710

NCT02132182
Clinical trials.
gov identifier

Hodgkin lymphoma. TAMs produce IL-6 and IL-10 leading to over


expression of B7-H4 (a recently discovered immunosuppressive mole-
cule), which inhibits host anti-tumor immunity [79]. Additionally, M2
subtype of TAMs have capability to alter antigen presentation and T-cell
mediated immune response by enhancing secretion of IL-10 and TGF-β
[80]. Whilst attempting to delineate implications of macrophage
Interventional

Observational

polarization in squamous cell carcinoma of oral cavity, Costa NL


Study phase

et al. observed significantly higher percentage of M2 subtype of TAMs


Phase I

(accompanied with higher levels of IL-10 and TGF-β) in cancer inflicted


patients than in healthy individuals [81]. IL-10 decreases synthesis of
granulocyte-macrophage colony-stimulating factor (GM-CSF) which in
resected/biopsied samples at histopathology

turns reduces T-cells production and the subsequently expected anti-


Variations in the intensity of cell surface
receptor expression, Disease progression

tumor immune response [82]. An eicosanoid, PGE2 and chemotactic


factors CCL17, CCL18, and CCL22, derived from TAMs and IL-10 play
important role in induction of T regulatory cells [83]. IL-10 in
pharmacokinetic evaluation,

conjunction with TNF-α also participates in production of a chemokine


named programmed cell death 1 (PD-L1) which has deleterious effects
Dose-limiting toxicities
objective response rate

on cytotoxic T-cell production and functionality [84]. In murine tumor


models, suppression of CD8+ T cell proliferation by TAMs is dependent
on production of ROS. This suppression has been offered as a possible
explanation for failure of cytotoxic T lymphocytes in eliminating
Method

tumors [85].

5. TAMs promote tumor invasion and metastasis


without sarcoma to exploit potential anti-
well as monitoring treatment response of

anti-tumor T-cell response following PD-

between monocytes in patients with and


microenvironment of M2 macrophages,
diagnostic and prognostic purposes as

Phenotypic and functional differences


thus favoring induction of a cytotoxic
CSF1R blockade to deplete the tumor

tumor capabilities of monocytes and


current and future immunotherapies

TAMs play major role in metastasis of cancer by infiltrating lymph


L1 blockade with an anti-PD-L1

nodes draining tumor. In a study undertaken specifically to investigate


role of TAMs in malignant breast cancer metastasis, it was found that
TAMs infiltration density had a negative correlation with 5-year
monoclonal antibody

survival rates of breast cancer patients [86,87]. Fellani et al. have


looked into clinicopathological significance of M1 and M2 macrophages
macrophages

in cutaneous melanoma, by monitoring accumulation of M1 and M2


TAMs in tumor stroma, intratumoral nests, and distant invasive sites.
Target

They observed that population of M2 biased TAMs was overwhelmingly


greater than M1 TAMs in all stages of melanoma progression, thereby
favoring neoplastic growth and rapid dissemination of metastatic cells
[88]. Tumor cells secrete CSF-1 and TAMs release epidermal growth
factor (EGF) and TNF-α to simultaneously control migration of cells to
Pancreatic Cancer
Colorectal Cancer

and from tumor microenvironment [89]. Activation of EGF receptors


Osteosarcoma

increase invasion, invadopodia formation and matrix degradation [90].


Elaine Y. Lin et al. have provided evidences which frame CSF-1 to be
Disease

responsible for metastatic growth of tumor by promoting infiltration of


TAMs and regulating their functions [91]. Involvement of TNF-α/NF-κB
dependent pathway in cancer cell migration and invasion is well
established [92]. TNF-α induces expression of macrophage migration
inhibitory factor to render cancer cells invasive [93]. TNF-α is also
involved in MMPs signaling which further facilitates invasion of tumor
Table 2 (continued)

cells. Lee SJ et al. explored MMP9 activation by TNF-α in human


Durvalumab

urinary bladder cancer (HT1376 cells) and deduced involvement of


Pexidartinib

extracellular signal-regulated kinase (ERK1/2), p38 MAP kinase and


JNK pathways [94]. They were able to block TNF-α dependent
Drug

activation of MMP-9 by administering an antitumor nucleoside cordy-


cepin, which is a known inhibitor of kinase pathways and thereby have

97
Y. Singh et al. Journal of Controlled Release 254 (2017) 92–106

come up with a probable strategy to stall invasive tendency of at least anticancer efficacy by reducing dose dependent side effects. In a recent
the investigated cancer. VEGF also activates MMP to induce enhanced study, simvastatin was encapsulated in a long circulating nanometric
vascularization and opens up new pathways for migration of invasive liposomal formulation and administered intravenously in B16F10
cancer cells [95]. Belotti D et al. found that VEGF specifically activates tumor bearing animals. Liposomal simvastatin significantly reduced
MMP-9 to cause invasion of ovarian tumor cells in mice bearing 1A9- tumor growth compared to free simvastatin with the purported activity
VS-1 tumor and treatment of animals with an anti-VEGF antibody directly dependent on local infiltrative TAMs population. Liposomal
(bevacizumab) inhibits VEGF dependent activation of MMP-9 and simvastatin furnished anticancer activity by modulating malondialde-
down-stream processes [96]. hyde levels, nitrites levels, catalase activity, and HIF-1α production, all
Tjiu et al. have investigated TAMs induced invasion of human basal of which have established roles in the dynamics of TAMs and tumor
cell carcinoma. To do so, M2-polarized TAMs derived from peripheral [121]. Until now iron supplement ferumoxytol and other iron oxide
blood monocytes were cocultured with basal cell carcinoma cells in nanoparticles have been used for treating iron deficiency or as contrast
coculture inserts so as to prevent direct cell to cell contact. It was found agents for magnetic resonance imaging and as drug carriers. However,
that TAMs induced cyclooxygenase-2 dependent release of MMP-9, recent studies have shown their role in inducing polarization of
VEGF-A, basic fibroblast growth factor which increased angiogenesis macrophages towards pro-inflammatory M1 subtype in tumor tissues.
and subsequent invasion of basal cell carcinoma cells [97].TAMs also Macrophages exposed to ferumoxytol displayed increased mRNA
promote metastasis via paracrine signaling. Baghel et al. have reported associated with pro-inflammatory Th1-type responses. Intravenous
the role of MIP-1 β in potentiating invasiveness of both metastatic and treatment of ferumoxytol in adenocarcinoma afflicted mice enhanced
poorly metastatic cells which is dependent on MYO3A. Hence MIP1β- the presence of pro-inflammatory M1 macrophages in the tumor tissues
MYO3A axis could be targeted for devising anti-metastatic therapeutics. and thus could potentiate macrophage-modulating cancer immu-
Cytokines found in mileu of TAMs also prompt invasion of cancer cells, notherapies. Inhibiting M2 polarization of TAMs is also an approach
with CCL18 being an example whose expression in breast cancer cells for cancer therapy. Resveratrol (3,5,4′-trihydroxy-trans-stilbene), a
can be positively correlated to tumor invasion [98]. Other TAMs natural plant phenol produced in response to attack by pathogens has
derived cytokines like, IL-6 and CCL4, have been shown to induce been found to inhibit activation of M2 macrophages. Whilst trying to
STAT3 activation in small cell lung cancer cells which leads to invasion, decode the underlying mechanisms in a lung cancer model, Liwei Sun
spheroid formation, acceleration of tumorogenesis in distant locations et al. discovered that resveratrol significantly reduced STAT3 activation
and chemoresistance. Downregulation of IL-6 receptor by anti-IL-6 and F4/80 positive cells in and around tumor. They exploited this
receptor antibody supresses STAT activation and consequently the cell- response to inhibit lung cancer growth, in vitro and in vivo [122].
cell interaction between TAMs and cancer cells via intermediary
cytokines can be a target for anti-tumor immunotherapy [99]. 7. Modification of TAM functionality using nanocarriers

6. Targeting tumor microenvironment by phamacotherapy Reversal of M2 subtype to M1 is a viable strategy for chemoimmu-
notherapy of cancer. Polarization of TAMs can be reversed by modulat-
From the previous sections it is clear as to how accumulation of ing Th1 cytokine profile and balancing expression of proangiogenic and
TAMs in tumor milieu influences its sustenance, progression and antiangiogenic factors. Working in this direction Huang et al. encapsu-
invasiveness. This has prompted researchers to try and plug relevant lated CpG, anti-IL-10 and anti-IL-10 receptor oligonucleotides in a
gaps via immunotherapy. Unfortunately though, clinical implications galactosylated cationic dextran nano-complex which had specific action
are still tricky, as stand-alone immune modulation has been found to be directed against TAMs. The nano-complex was fabricated to sense
capable of only priming the body to fight tumor, and not actually subtle variation in pH associated with tumor microenvironment
eradicate it. The next plausible maneuver in clinically tackling cancer triggering release of its encapsulated oligonucleotides. When tested in
therefore seems adaptation of a combinatorial strategy amalgamating an allograft hepatoma murine model, the delivery system directly
circumstantially effective chemotherapy with a facilitatory immuno- accumulated in TAMs and caused its phenotypic reversion by inducing
modulative regimen to obtain a focused counter measure free from secretion of Th1 cytokine IL-12 and limiting Th2 cytokine IL-10.
incidences of adverse effects or subpar efficacy. Nanomedicine and Expression of M2 related genes (Arg1, Ym1, Msr2, FIZZ1, Mgl1 and
colloidal carrier based drug delivery has recently come to the fore with Mgl2) was reduced. Further, expression of proangiogeneic factors
respect to anticancer therapy [100,101]. Nanocarriers owing to their including VEGF and MMP9 was also suppressed which cumulatively
size inadvertently accumulate in irregularly fenestrated tumor micro- resulted in significant tumor regression [123]. Wang et al. have
environment (EPR effect) paving way for preferential tumoral localiza- developed tumor environment sensitive poly-β-amino ester nanoparti-
tion of chemotherapeutic agents and utilization of ligand based active cles and used them to deliver IL-12 for reeducating TAMs without
targeting with several products gaining clinical approval [102,103]. causing any undue cytotoxicity. In in vivo studies, reversing of TAM
This opens doors for selectively killing [104] or reversing polarization polarization was confirmed by identifying relative count of CD206
of macrophages infiltrating tumor environment, leaving peripheral or (marker for M2) and CCR7 (marker for M1) positive cells using confocal
circulatory monocytes/macrophages unharmed [105,106]. Due to their microscopy and flow cytometry. Biodistribution studies employing
miniscule size, nanocarriers furnish novel patterns of drug delivery surrogates suggested that enhanced therapeutic efficacy of IL-12 loaded
inside cancer cells, by presenting maximal payload directly at intracel- nanoparticles was due to their selective tumoral accumulation and
lular locations like nucleus or microtubule [107–110]. They do so by capability to extend circulation time of IL-12 [124]. Banciu et al. whilst
modulating binding capacity and penetrability of cell membrane and investigating probable role of TAMs in anticancer activity of Doxil® (an
different cellular organelles. Nanocarriers also mold inherent pharma- approved pegylated liposomal formulation of doxorubicin hydrochlor-
cokinetic properties of drugs they carry by controlling their presenta- ide) found that it had little or no effect on level of proangiogeneic and
tion to systemic dispositional forces, thereby catalyzing drug's efficacy antiangiogenic factors [125]. Taking note, they prepared polyethylene
and overall safety [111–114] [115–117]. For instance, HMG-CoA glycol functionalized dipalmitoylphosphatidylcholine liposomes loaded
reductase inhibitors (statins), at certain higher doses modulate various with prednisolone phosphate, which were capable of preferentially
intracellular pathways to exhibit quantifiable anticancer activity accumulating in tumor microenvironment and remodel TAMs polarity.
[118,119]. Simvastatin indirectly increases intracellular oxidative Administration of prednisolone phosphate loaded liposomes in B16F10
stress and induces apoptosis by quenching free radical scavengers induced tumor bearing mice significantly reduced expression of proan-
(Mn-SOD, CAT, GPx1, and SESN 3) [120]. Targeted delivery of statins giogeneic factors (GM-CSF, M-CSF, IL-1α, IL-1β, IL-6, IL-9 and TNF-α)
to tumor microenvironment can therefore theoretically improve their [126].

98
Y. Singh et al. Journal of Controlled Release 254 (2017) 92–106

Bisphosphonates have been reported to act and modify phenotypic 9. Magic bullets: Actively targeting TAMs
status of TAMs to render them anti-tumor [127–129]. Zoledronic acid is
the most potent bisphosphonate; but its anticancer activity is limited by M1 and M2 macrophages have differing phenotypes, which not only
short plasma half-life and rapid accumulation in bones. This is where alters their functionality but also induces in them several structural
nanotechnology comes into fray. Zoledronic acid was encapsulated in differences. The resultant of such variations is especially felt in relative
pegylated nanoparticles to improve its accumulation in tumor micro- presence/absence of surface receptors, which participate in uptake
environment. In, in vivo studies, normal drug had no anticancer activity processes like phagocytosis, endocytosis, pinocytosis etc. Since, nanoar-
whereas drug carrying pegylated nanoparticles possessed substantial riers also exploit similar pathways for uptake; a cursory inspection into
anticancer activity and prolonged animal survival. TAMs population intracellular pharmacokinetics of administered nanoparticles might
and neoangiogenesis was also reduced [130]. Another phosphonate provide clues towards selecting appropriate construct material, dosing
analogue, clodronate, enhances secretion of Th1 cytokine and inhibits frequency, etc. For instance, Jones et al. devised an intravital micro-
secretion of Th2 cytokines, angiogenic factors, but its use is again scopy based assay to determine effect of immune system on nanopar-
limited by inherent pharmacokinetic constraints [131,132] necessitat- ticle clearance. But their results were contradictory; in vivo studies
ing development of liposomal clodronate [133,134]. Zeisberger SM suggested that nanoparticles were cleared more slowly in Th1 biased
et al. have successfully employed liposomal clodronate formulation to mice in comparison to Th2 biased mice, whereas the trend was opposite
reeducate TAMs for beneficial activity in cancer. Immunohistochemical in vitro, where macrophage polarization from M1 to M2 increased
evaluation showed liposomal clodronate drastically reduced F4/80 nanoparticle uptake and retention [142]. In a different study Hopp-
positive cells in comparison to free clodronate [135]. In a different stadter et al. obtained conformity in in vitro and in vivo results. Uptake
study Fritz et al. showed that liposomal clodronate specifically hampers of investigated fluorescent silica nanoparticles was greater in M2
TAMs population in mouse lung adenocarcinomas by reducing insulin- polarized macrophages in comparison to M1 polarized macrophages
like growth factor-I, CXCL1, IL-6, and CCL2 levels in pulmonary fluids [143].
[136]. A recent study by Pal et al. has confirmed functionality of noble Wang et al. attempted to detail effect of nanoparticle surface
metal (gold and silver) nanoparticles in modulation of TAMs [137]. The modification and macrophage polarization on cellular uptake. They
study addresses functional characterization of TAMs isolated from constructed different batches of particles using p(N-isopropylacryla-
murine fibrosarcoma induced by a chemical carcinogen, 3-methylcho- mide-co-acrylic acid) (p-(NIPAm-co-AAc)) copolymer and functiona-
lanthrene. The fabricated gold and silver nanoparticles were able to lized them with thirteen functional groups. Quantitative structure
modulate reactive oxygen species and reactive nitrogen species produc- activity relationship software deduced that surface charge, number of
tion in tumor cells and also activate proinflammatory signaling hydrogen atoms and number of 2° carbon atoms in nanoparticle
cascades resulting in a polarization of TAMs from M2 to M1. construct had a telling say in decreasing complement activation and
overall uptake in M2 polarized macrophages [144]. Fuchs AK et al. also
8. Direct killing of TAMs investigated the role of surface decoration of delivery vectors on fate of
macrophage subset. They functionalized polystyrene nanoparticles with
There is an alternative school of thought which prefers complete carboxyl and amine groups. The decorated nanoparticles did not affect
eradication of susceptible M2 subtype from tumor stroma, instead of expression of the M1 markers CD86, NOS2, TNF-α, and IL-1β. In
reaping advantages from its reversion to M1 cells, as theoretically, there contrast, both carboxyl and amine functional groups impaired expres-
is always a chance of a phenotypic re-reversal after discontinuation of sion of scavenger receptor CD163 and CD200R, and release of IL-10
therapy. Wu et al. conducted a cytotoxicity experiment on a co-culture thus suggesting a probable role for functionalized nanoparticles as a
of Hut78tumor cells and M2 macrophages and found that liposomal tool in selective reprogramming of M1/M2 polarization [145]. It
clodronate selectively kills TAMs and does not affect viability of Hut78 therefore seems rational that drug loaded nanoparticles should be
tumor cells. Implications of selective TAMs eradication were investi- decorated with phenotype receptive ligands to vindicate notion of
gated in animals having cutaneous T-cell lymphoma. Intravenous Ehlrich's magic bullet, i.e. specific activity with little/no off target
administration of liposomal clodronate reduced TAMs (measured by effects. Following section details some bio-inspired ligands which have
number of F4/80 positive cells) and inhibited angiogenesis (quantified been employed to target TAMs.
by CD31 and podoplanin antibodies in tissue sections) which ultimately
led to substantial tumor reduction [138]. Other examples include 9.1. Carbohydrates
packaging of doxorubicin in a TAMs targeted delivery system to
produce a gross cytotoxic response. Biodistribution studies revealed Scavenger receptors like lectin (CD206/mannose receptors) are
that doxorubicin loaded system selectively accumulated in tumor and overexpressed selectively on M2 subtype of TAMs. CD206 receptors
caused massive reduction in TAMs population and overall B16F10 respond to mannose and fucose residues on any trailing entity,
tumor burden [139]. F. Piaggio et al. and co-workers have developed initiating its endocytosis via clathrin mediated pathway. This has
clodronate-containing liposomes for intravenous administration. These prompted extensive utilization of carbohydrate moieties (Mannose,
novel systems were investigated in vitro to inhibit proliferation and dextrans and galactose) for decoration of drug and nucleotide loaded
induce apoptosis of a macrophage-like cell line in a dose and time- nanoparticles to target TAMs via CD206 receptors [139,146]. In a
dependent manner. Upon administration in B16/F10 subcutaneous recent study by Yu et al., a triblock copolymer was functionalized with
melanoma-bearing mice model, it significantly lowered the plasma mannose using click chemistry. The synthesized mannose copolymer
levels of IL-10, Mo KC, TNF-α and VEGF [140]. Folate receptors are was used to develop a micellar system for delivery of siRNA to TAMs.
over expressed on numerous cancer cells as well as on adjoining TAMs, Mannose functionalization significantly enhanced macrophage uptake
which renders them as a convenient target for cancer treatment. Luo T of micelles and produced knockdown of target gene [147]. The same
et al. have successfully exploited this receptor-cancer axis to deliver a group also mannosylated nanoparticles for delivering siRNA to TAMs.
combinatorial pay load of oxygen and paclitaxel in xenograft mouse Depending upon the experimental model, the said nanoparticles
ovarian cancer model using ultrasound guided microbubbles. It was accumulated selectively in ovarian tumor and lung tumor, undergoing
found that, microbubbles could penetrate deep into tumor ascites fluid rapid internalization by targeted tumor associated macrophages to stop
and tumor nodules, and induce apoptosis and reduce expression of cancer metastasis without producing any side effect on liver and kidney
VEGF. Since this system was cytotoxic to both cancer cells and TAMs, it functions [146]. Proficiency of mannose ligated delivery system has
can provide an approach of dual intervention at both cytotoxic and been validated by PET imaging. A dual imaging mannosylated liposo-
immunologic level [141]. mal system was prepared by tagging 64Cu to a fluorescent dye

99
Y. Singh et al. Journal of Controlled Release 254 (2017) 92–106

encapsulated in a lipidic membrane. PET and fluorescent imagining probe, when tested in a 4T1 tumor bearing animal model specifically
reveled that liposomes specifically accumulate in TAMs upon adminis- tags onto TAMs, generating pertinent in vivo images [158]. In another
tration in an animal model with lung adenocarcinoma [148]. Zhu et al. study, Movahedi et al. used mannose receptor targeted nanobodies
have amalgamated advantages of mannose receptor based macrophage (single domain antigen binding fragments prepared from Camelidae
recognition with a sheddable PEG cloak to come up with a full proof heavy-chain antibodies). 99mTc tagged nanobodies specifically label
method of ensuring targeted delivery of doxorubicin. They coupled PEG TAMs in tumor microenvironment [159] and should be tested in
to mannosylated nanoparticle template with a pH sensitive hydrazone delivery of cytotoxics for therapeutic benefit. Hemoglobin scavenger
bond. It was assumed that upon intravenous administration, PEG receptor CD163 are also overexpressed on TAMs and are involved in
component would prevent unwanted reticuloendothelial uptake of progression of cancer [160]. Therapeutic significance of this receptor
nanoparticles until they reach slightly acidic tumor environment. This has been illustrated by Etzerodt et al. They ligated CD163 responsive
pH change would trigger loss of hydrazone bond and shedding of PEG monoclonal antibody on pegylated liposomes to enhance its TAMs
cloak, exposing mannosylated nanoparticle surface (capable of inter- uptake and efficacy of loaded drug [161].
acting with scavenger receptors on TAMs). Results suggested that
payload accumulated explicitly in tumor and TAMs, away from 9.3. Proteins and peptides
unnecessary sites like heart or liver to significantly reduce tumor
volume in B16F10 tumor bearing mice [139,149]. Zhan et al. have Proteins have specific three dimensional confirmations, which is
conjugated glucomannan to a bisphosphonate (alendronate) for specific responsible for their selective affinity towards a substrate. This
targeting and elimination of TAMs in a subcutaneous S180 tumor- specificity can be exploited for development of targeted delivery
bearing mice model. Glucomannan has very high affinity for mannose systems. Legumain, an asparaginyl endopeptidase, found in plant,
receptors and consequently its conjugation allows alendronate to parasites and mammals is overexpressed in variety of solid tumors
overcome its inherent tumor environment susceptibility and instead leading to their invasion/metastasis [162–164]. More importantly, it is
be rapidly taken up by TAMs to elicit specific immunoactivity which overexpressed on TAMs; a fact which has been used in development of
leads to suppression of angiogenesis and tumor [150]. Another member TAMs targeted vaccine for eradication of tumor growth and metastasis
of lectin subfamily of receptors overexpressed on TAMs includes [165–168]. A doxorubicin based prodrug, responsive to legumain
Sialoadhesin (Siglec-1) receptor, which correspond specifically to expression has also been developed to suppress TAMs and related
carbohydrates possessing sialic acid residue (O - or - N substituted tumor growth [169]. In a recent study, a dual approach based upon
derivatives of neuraminic acid). Siglec-1 is an endocytic receptor, and legumain and cell penetrating peptide was employed to develop a
therefore is ideal for receptor oriented nanocarrier delivery. Zhou et al. liposomal formulation for improved efficacy of doxorubicin by target-
have exploited this receptor using sialic derivative of cholesterol as a ing tumor associated macrophages [170]. In a conceptually similar
targeting moiety for epirubicin loaded liposomes. In vitro and in vivo cell study, Zhang X and fellow researchers have loaded hydrazinocurcumin,
uptake studies suggested selective accumulation of these liposomes in a synthetic analogue of curcumin in legumain-targeting liposomal
TAMs which in turn resulted in depletion of macrophage population nanoparticles and investigated their effects on suppression of tumor
and several fold enhancement in antitumor activity of epirubicin [151]. in vivo. When fabricated nanoparticles were intravenously administered
Dextran's (complex branched glucan) has also been used to target in Balb/C mice model afflicted with breast cancer, they were able to
TAMs for purpose of imaging and delivery of small molecules. considerably “re-educate” M2-like macrophages and caused suppres-
Weissleder et al. synthesized a library of 146 nanoparticles using sion of tumor growth, angiogenesis and metastasis [171].
different synthetic small molecules and among them dextran coated Peptides are small amino acid sequences possessing linear or cyclic
iron oxide nanoparticles (CLIO680 and AMTA680) exhibited highest structures. Their small molecular size facilitates easy synthesis and
affinity towards TAMs [152]. CLIO680 and AMTA680 nanoparticles provides environmental stability. Recently, Cieslewicz et al. created a
were consequently conjugated with a fluorochrome (VT680) to speci- peptide sequence M2pep (YEQDPWGVKWWY). M2pep demonstrated
fically label TAMs for imaging applications [153]. Daldrup-Link et al. high affinity towards TAMs in mixed cells population and has been used
have used carboxydextran coated iron oxide nanoparticles for magnetic as targeting vector for intracellular delivery of an apoptotic peptide.
resonance imaging of TAMs. In an experimental model treated with When tested in vivo, the system specifically binds to TAMs and delays
CSF1 monoclonal antibodies to kill TAMs, there was quantifiable mortality in tumor bearing mice [172]. M2pep has also been used to
reduction in accumulation and the consequently generated MRI signal decorate a nanoparticle system for delivery of siRNA to TAMs in order
by carboxydextran coated iron oxide nanoparticles showcasing efficacy to inhibit expression of VEGF and associated tumor burden [173]. In
of adapted treatment [154]. Glactose type lectin receptors are also another study, Yan el al, synthesized a Y shaped peptide [Ala-Ala-Asn-
highly expressed over surface of M2 phenotype of TAMs and are worth Leu-His-Lys-(His-Lys)2] for targeting ligumain overexpressed on TAMs.
a try as targeting moiety. These receptors recognize terminal GalNAc The synthesized peptide and paramagnetic Fe3O4 nanoparticles were
residue on glycans and mediate endocytosis [155–157] (Fig. 2). conjugated on to a carbon nanotube and used for selective MRI imaging
of TAMs. Interestingly, the histidine residue present in Y peptide
9.2. Antibodies facilitated endosomal escape of nanotubes due to proton sponge effect
[174]. Lyp-1 peptide can recognize lymphatic vessel, tumor cells and
Antibody (immunoglobulin; Ig) is a Y shaped protein which can TAMs [175,176]. Lyp-1 has been used to decorate a liposomal
identify specific regions on a target antigen. Each tip of Y has a formulation for specific binding with TAMs so as to inhibit growth of
paratope which recognizes epitope available on the antigen. The metastatic tumor [177]. RVG peptide derived from rabies virus
hypervariable region, also known as Fab(ab')2 fragment, on the Y tip glycoprotein has been used for active targeting of TAMs of glioma
has large number of slightly variable biding sites which further enlarges region [178]. For instance, RVG peptide has been conjugated on to shell
the catalogue of recognizable antigens. Fc fragment present at the base of biodegradable nanoparticles containing paclitaxel to treat malignant
of Y however is very specific. Antibody ligation on nanoparticle surface glioma. Owing to their spherical shape, small size, narrow distribution,
thus in principle could facilitate site specific internalization if the and ligand conjugated surface, the said nanoparticles were able to
tethered antibody recognizes any antigen present on target cell. penetrate the normally impermeable blood brain barrier and accumu-
However, physiological stability and capability to trigger unintended late in TAMs populating the glioma, keeping away from regular neurons
immune response makes antibody usage tricky. Sun et al. have to provide safe and effective anti-glioma therapy [179]. Table 3
synthesized near-infrared fluorescence responsive probe by conjugating summarizes various ligands and the capacities in which they have been
anti-mouse CD206 antibody with DyLight680 succinimidyl ester. The used to target TAMs.

100
Y. Singh et al. Journal of Controlled Release 254 (2017) 92–106

Fig. 2. Utilization of phenotypic differentiators for selective targeting of M2 subtype of tumor associated macrophages. Scavenger receptors such as CD206, CD163 are found only on M2
cells and not on M1 cells. Researchers have therefore come up with full proof surface decorated nanocarriers loaded with cytotoxic or immunomodulatory drugs which recognize these
receptors to either kill or reeducate TAMs, sparing other unwanted organs or cells from cytotoxic action. Nanocarriers often exploit subtle variations such as hypoxia; altered pH,
enhanced permeability and retention effect to selectively accumulate in tumor environment. Thereafter receptor mediated endocytosis is initiated which involves recognition and binding
of specific ligands (carbohydrates, proteins, antibodies) appended on nanocarriers by receptors present on cell surface. The payload is subsequently endocytosed and delivered to
intracellular machinery to produce further effects.

10. Future prospects glioma patients and is currently clinical under clinical investigation
[193]. Immunomodulatory compounds like thymosin and β-glucan
With improved knowledge of genesis, sustenance, and propagation have also been explored to reeducate TAMs and increase immune
of cancer there has been a spurt of probable targets especially with response in patients suffering from cancer [194,195]. Direct killing of
respect to signaling involved in TAMs. It has been reported that TAMs has also been tried using trabectedin, which causes apoptosis of
recruitment of TAMs in tumor microenvironment is facilitated by macrophages through TNF-related apoptosis-inducing ligand (TRAIL)
certain chemoattractants like CCL2 [180] and CCL5 [181]. CNTO88 receptors [196]. PF-04136309, a CD192 antagonist, prevents recruit-
(carlumab), an antibody of CCL2, currently under clinical trial has ment of CD192 + monocytes from bone marrow to tumors in a mouse
demonstrated strong anticancer activity in preliminary testing [182]. model bearing pancreatic cancer leading to depletion in TAMs popula-
CSF-1 can be targeted using antisense oligonucleotides [183,184], tion, which in turn reduces tumor growth and metastasis. In hypoxic
monoclonal antibodies [185] and tyrosine kinase inhibitors [186,187] regions of hepatocellular carcinoma, LY294002, a protein inhibitor, has
to modulate recruitment of TAMs in tumor microenvironment [188]. shown credible anti-tumor activity by interfering in HIF-1 expression
Th1 cytokine IFNγ [189], toll like receptor agonists [190,191] and and subsequent activation of PI3K/Akt headed epithelial-mesenchymal
CD40 molecule activated antibodies (anti-CD40 mAb) [192] can all be transition (EMT metastasis) and chemoresistance [197]. Overall hypox-
used to reeducate TAMs to furnish an antitumor immune response. ia has been associated with malignant phenotype and poor prognosis of
WP1066, an inhibitor of STAT3 can induce TH1 biased cytokines like cancer cells [198]. It is studied as a key target due to the fact that
IL-12 and receptors CD80, CD86 in peripheral as well as tumor cancerous cells acquire a partial oxygen pressure which is at least 33%
associated macrophages, can reverse immune tolerance in malignant less than normal live cells [199]. Hypoxia induces abnormal vascular-

Table 3
Examples of ligands utilized for targeting TAMs through nanocarriers.

Ligand Class Target Active moiety and imaging agent Nanocarrier system References

Mannose Carbohydrate CD206 receptors siRNA Micelles [147]


Mannose Carbohydrate CD206 receptors Doxorubicin Nanoparticles [136]
Mannose Carbohydrate CD206 receptors 64
Cu and a fluorescent dye Liposomes [148]
Dextran Carbohydrate CD206 receptors Fluorochrome VT680 Nanoparticles [153]
Carboxydextran Carbohydrate CD206 receptors Iron oxide for MRI Nanoparticles [154]
Galactosylated cationic dextran Carbohydrate Glactose type lectin receptors Oligonucleotide Nanoparticles [123]
Anti-mouse CD206 antibody Antibody CD206 receptors DyLight680 succinimidyl ester – [158]
Nanobodies Antibody fragments CD206 receptors 99m
Tc – [159]
Monoclonal antibody Antibody Hemoglobin scavenger receptor CD163 Liposomes [161]
Legumain Proteins Legumain receptors Hydrazinocurcumin, Doxorubicin Liposomes [170,171]
M2pep Peptide – siRNA Nanoparticles [173]
Y shaped peptide Peptide – Anti-apoptotic peptide and iron oxide Carbon nanotube [174]
Lyp-1 Peptide Lymphatics and tumor cells Fluorescein and doxorubicin Liposomes [177]

101
Y. Singh et al. Journal of Controlled Release 254 (2017) 92–106

ization at tumor site creating a barricade in drug access and results in [14] C.A. Ambarus, S. Krausz, M. van Eijk, J. Hamann, T.R.D.J. Radstake,
K.A. Reedquist, P.P. Tak, D.L.P. Baeten, Systematic validation of specific pheno-
variable bioavailability which in turn may cause therapeutic chemo- typic markers for in vitro polarized human macrophages, J. Immunol. Methods
resistance of anticancer agents. Certain drugs such as melphalan, 375 (2012) 196–206.
bleomycin, etoposide, etc., and radiation therapy necessarily require [15] F. Colotta, S. Saccani, J.G. Giri, S.K. Dower, J.E. Sims, M. Introna, A. Mantovani,
Regulated expression and release of the IL-1 decoy receptor in human mono-
molecular oxygen for maximal cytotoxicity, and therefore produce sub nuclear phagocytes, J. Immunol. (Baltimore, Md.) 156 (1996) (1950) 2534–2541.
maximal effects in vivo due to prevalent hypoxia [200]. Therefore [16] F.O. Martinez, S. Gordon, M. Locati, A. Mantovani, Transcriptional profiling of the
researchers have tried various strategies to directly address tumor human monocyte-to-macrophage differentiation and polarization: new molecules
and patterns of gene expression, J. Immunol. (Baltimore, MD) 177 (2006) (1950)
hypoxia and the subsequently expected downstream effectuality, i.e. 7303–7311.
‘recruitment of TAMs’ [201]. Examples include development of bio- [17] M. Jaguin, N. Houlbert, O. Fardel, V. Lecureur, Polarization profiles of human M-
reductive pro-drugs that are activated by enzymatic reduction specifi- CSF-generated macrophages and comparison of M1-markers in classically acti-
vated macrophages from GM-CSF and M-CSF origin, Cell. Immunol. 281 (2013)
cally in hypoxia affected region and inhibition of molecular events that
51–61.
favor proliferation of hypoxic cells [202]. Chemists have developed [18] L. Sun, M.C. Louie, K.M. Vannella, C.A. Wilke, A.M. LeVine, B.B. Moore,
various bio-reductive pro-drugs from five chemical moieties (nitro T.P. Shanley, New concepts of IL-10-induced lung fibrosis: fibrocyte recruitment
groups, quinones, aromatic N-oxides, aliphatic N-oxides and transition and M2 activation in a CCL2/CCR2 axis, Am. J. Physiol. Lung Cell. Mol. Physiol.
300 (2011) L341–L353.
metals) which have potential to undergo enzymatic metabolism under [19] W. Fang, L. Ye, L. Shen, J. Cai, F. Huang, Q. Wei, X. Fei, X. Chen, H. Guan,
hypoxic condition although the expression level of the activating W. Wang, X. Li, G. Ning, Tumor-associated macrophages promote the metastatic
enzyme remains a critical parameter. For instance, aromatic N-oxide potential of thyroid papillary cancer by releasing CXCL8, Carcinogenesis 35
(2014) 1780–1787.
tirapazamine is 200 fold more toxic to hypoxic cells than norm-oxic [20] A. Sica, A. Mantovani, Macrophage plasticity and polarization: in vivo veritas, J.
cells and the activation is governed by single electron reduction process Clin. Invest. 122 (2012) 787–795.
that damages the DNA replication fork. [21] A. Kroner, A.D. Greenhalgh, J.G. Zarruk, R. Passos Dos Santos, M. Gaestel,
S. David, TNF and increased intracellular iron alter macrophage polarization to a
It is our firm belief that, if packaged in a rationally devised detrimental M1 phenotype in the injured spinal cord, Neuron 83 (2014)
nanotechnology platform, the aforementioned chemo-attractants and 1098–1116.
endogenous/exogenous compounds would open up even more antic- [22] A. Mantovani, Macrophage diversity and polarization: in vivo veritas, Blood 108
(2006) 408–409.
ancer avenues by targeting TAMs more efficiently.
[23] S.K. Biswas, A. Mantovani, Macrophage plasticity and interaction with lymphocyte
subsets: cancer as a paradigm, Nat. Immunol. 11 (2010) 889–896.
Acknowledgements [24] S. David, A. Kroner, Repertoire of microglial and macrophage responses after
spinal cord injury, Nat. Rev. Neurosci. 12 (2011) 388–399.
[25] E. Cassol, L. Cassetta, C. Rizzi, M. Alfano, G. Poli, M1 and M2a polarization of
We would like to thank CSIR for providing financial assistance with human monocyte-derived macrophages inhibits HIV-1 replication by distinct
Network Project titled “Advanced Drug Delivery Systems” [CSC0302]. mechanisms, J. Immunol. 182 (2009) 6237–6246.
This is CSIR-CDRI communication 9473. [26] H. Qin, A.T. Holdbrooks, Y. Liu, S.L. Reynolds, L.L. Yanagisawa, E.N. Benveniste,
SOCS3 deficiency promotes M1 macrophage polarization and inflammation,
Journal of immunology (Baltimore, Md.) 189 (2012) (1950) 3439–3448.
References [27] M. Benoit, B. Desnues, J.-L. Mege, Macrophage polarization in bacterial infections,
J. Immunol. 181 (2008) 3733–3739.
[28] T. Katakura, M. Miyazaki, M. Kobayashi, D.N. Herndon, F. Suzuki, CCL17 and IL-
[1] R. Bonecchi, S. Sozzani, J.T. Stine, W. Luini, G. D'Amico, P. Allavena, D. Chantry, 10 as effectors that enable alternatively activated macrophages to inhibit the
A. Mantovani, Divergent effects of interleukin-4 and interferon-gamma on generation of classically activated macrophages, J. Immunol. 172 (2004)
macrophage-derived chemokine production: an amplification circuit of polarized 1407–1413.
T helper 2 responses, Blood 92 (1998) 2668–2671. [29] I.U. Schraufstatter, M. Zhao, S.K. Khaldoyanidi, R.G. DiScipio, The chemokine
[2] V. Kodelja, C. Muller, O. Politz, N. Hakij, C.E. Orfanos, S. Goerdt, Alternative CCL18 causes maturation of cultured monocytes to macrophages in the M2
macrophage activation-associated CC-chemokine-1, a novel structural homologue spectrum, Immunology 135 (2012) 287–298.
of macrophage inflammatory protein-1 alpha with a Th2-associated expression [30] P. Rous, J.G. Kidd, Conditional neoplasms and subthreshold neoplastic states: a
pattern, J. Immunol. (Baltimore, Md.) 160 (1998) (1950) 1411–1418. study of the tar tumors of rabbits, J. Exp. Med. 73 (1941) 365–390.
[3] J.A. Joyce, J.W. Pollard, Microenvironmental regulation of metastasis, Nat. Rev. [31] F. Balkwill, A. Mantovani, Inflammation and cancer: back to Virchow? Lancet 357
Cancer 9 (2009) 239–252. (2001) 539–545.
[4] B.Z. Qian, J.W. Pollard, Macrophage diversity enhances tumor progression and [32] M. Karin, Nuclear factor-kappaB in cancer development and progression, Nature
metastasis, Cell 141 (2010) 39–51. 441 (2006) 431–436.
[5] N. Wang, H. Liang, K. Zen, Molecular mechanisms that influence the macrophage [33] F. Colotta, P. Allavena, A. Sica, C. Garlanda, A. Mantovani, Cancer-related
M1–M2 polarization balance, Front. Immunol. 5 (2014) 614. inflammation, the seventh hallmark of cancer: links to genetic instability,
[6] Y. Chen, F. Wermeling, J. Sundqvist, A.B. Jonsson, K. Tryggvason, T. Pikkarainen, Carcinogenesis 30 (2009) 1073–1081.
M.C. Karlsson, A regulatory role for macrophage class A scavenger receptors in [34] S. Sousa, R. Brion, M. Lintunen, P. Kronqvist, J. Sandholm, J. Mönkkönen, P.-
TLR4-mediated LPS responses, Eur. J. Immunol. 40 (2010) 1451–1460. L. Kellokumpu-Lehtinen, S. Lauttia, O. Tynninen, H. Joensuu, Human breast
[7] S. Tundup, L. Srivastava, T. Nagy, D. Harn, CD14 influences host immune cancer cells educate macrophages toward the M2 activation status, Breast Cancer
responses and alternative activation of macrophages during Schistosoma mansoni Res. 17 (2015) 101.
infection, Infect. Immun. 82 (2014) 3240–3251. [35] P. Tymoszuk, H. Evens, V. Marzola, K. Wachowicz, M.-H. Wasmer, S. Datta,
[8] D. Niino, Y. Komohara, T. Murayama, R. Aoki, Y. Kimura, K. Hashikawa, J. Kiyasu, E. Müller-Holzner, H. Fiegl, G. Böck, N. van Rooijen, I. Theurl, W. Doppler, In situ
M. Takeuchi, N. Suefuji, Y. Sugita, M. Takeya, K. Ohshima, Ratio of M2 proliferation contributes to accumulation of tumor-associated macrophages in
macrophage expression is closely associated with poor prognosis for angioimmu- spontaneous mammary tumors, Eur. J. Immunol. 44 (2014) 2247–2262.
noblastic T-cell lymphoma (AITL), Pathol. Int. 60 (2010) 278–283. [36] R.D. Loberg, C. Ying, M. Craig, L. Yan, L.A. Snyder, K.J. Pienta, CCL2 as an
[9] G. Liu, H. Ma, L. Qiu, L. Li, Y. Cao, J. Ma, Y. Zhao, Phenotypic and functional important mediator of prostate cancer growth in vivo through the regulation of
switch of macrophages induced by regulatory CD4 +CD25 + T cells in mice, macrophage infiltration, Neoplasia (New York, N.Y.) 9 (2007) 556–562.
Immunol. Cell Biol. 89 (2011) 130–142. [37] T. Kitamura, B.-Z. Qian, D. Soong, L. Cassetta, R. Noy, G. Sugano, Y. Kato, J. Li,
[10] R.A. Ezekowitz, S. Gordon, Alterations of surface properties by macrophage J.W. Pollard, CCL2-induced chemokine cascade promotes breast cancer metastasis
activation: expression of receptors for Fc and mannose-terminal glycoproteins and by enhancing retention of metastasis-associated macrophages, J. Exp. Med. 212
differentiation antigens, Contemp. Top. Immunobiol. 13 (1984) 33–56. (2015) 1043–1059.
[11] K.M. Choi, P.C. Kashyap, N. Dutta, G.J. Stoltz, T. Ordog, T.S. Donohue, A.J. Bauer, [38] B.-Z. Qian, J. Li, H. Zhang, T. Kitamura, J. Zhang, L.R. Campion, E.A. Kaiser,
D.R. Linden, J.H. Szurszewski, S.J. Gibbons, G. Farrugia, CD206-positive M2 L.A. Snyder, J.W. Pollard, CCL2 recruits inflammatory monocytes to facilitate
macrophages that express heme oxygenase-1 protect against diabetic gastroparesis breast-tumour metastasis, Nature 475 (2011) 222–225.
in mice, Gastroenterology 138 (2010) 2399–2409. e2391. [39] E. Sierra-Filardi, C. Nieto, A. Dominguez-Soto, R. Barroso, P. Sanchez-Mateos,
[12] J. Xu, Y. Yang, J. Sun, Y. Ding, L. Su, C. Shao, B. Jiang, Expression of toll-like A. Puig-Kroger, M. Lopez-Bravo, J. Joven, C. Ardavin, J.L. Rodriguez-Fernandez,
receptors and their association with cytokine responses in peripheral blood C. Sanchez-Torres, M. Mellado, A.L. Corbi, CCL2 shapes macrophage polarization
mononuclear cells of children with acute rotavirus diarrhoea, Clin. Exp. Immunol. by GM-CSF and M-CSF: identification of CCL2/CCR2-dependent gene expression
144 (2006) 376–381. profile, J. Immunol. (Baltimore, Md.) 192 (2014) (1950) 3858–3867.
[13] B. Mallavia, A. Oguiza, O. Lopez-Franco, C. Recio, G. Ortiz-Muñoz, I. Lazaro, [40] K. Mizutani, S. Sud, N.A. McGregor, G. Martinovski, B.T. Rice, M.J. Craig,
V. Lopez-Parra, J. Egido, C. Gomez-Guerrero, Gene deficiency in activating Fcγ Z.S. Varsos, H. Roca, K.J. Pienta, The chemokine CCL2 increases prostate tumor
receptors influences the macrophage phenotypic balance and reduces athero- growth and bone metastasis through macrophage and osteoclast recruitment,
sclerosis in mice, PLoS One 8 (2013) e66754. Neoplasia (New York, N.Y.) 11 (2009) 1235–1242.

102
Y. Singh et al. Journal of Controlled Release 254 (2017) 92–106

[41] R. Bonecchi, F. Facchetti, S. Dusi, W. Luini, D. Lissandrini, M. Simmelink, [65] K.L. Talks, H. Turley, K.C. Gatter, P.H. Maxwell, C.W. Pugh, P.J. Ratcliffe,
M. Locati, S. Bernasconi, P. Allavena, E. Brandt, F. Rossi, A. Mantovani, S. Sozzani, A.L. Harris, The expression and distribution of the hypoxia-inducible factors HIF-
Induction of functional IL-8 receptors by IL-4 and IL-13 in human monocytes, J. 1α and HIF-2α in normal human tissues, cancers, and tumor-associated macro-
Immunol. (Baltimore, Md.) 164 (2000) (1950) 3862–3869. phages, Am. J. Pathol. 157 (2000) 411–421.
[42] G. Ghilardi, M.L. Biondi, A. La Torre, L. Battaglioli, R. Scorza, Breast cancer [66] Y.J. Jung, J.S. Isaacs, S. Lee, J. Trepel, L. Neckers, IL-1beta-mediated up-regulation
progression and host polymorphisms in the chemokine system: role of the of HIF-1alpha via an NFkappaB/COX-2 pathway identifies HIF-1 as a critical link
macrophage chemoattractant protein-1 (MCP-1)−2518 G allele, Clin. Chem. 51 between inflammation and oncogenesis, FASEB J. 17 (2003) 2115–2117.
(2005) 452–455. [67] S.H. Jeon, B.C. Chae, H.A. Kim, G.Y. Seo, D.W. Seo, G.T. Chun, N.S. Kim, S.W. Yie,
[43] D. Milliken, C. Scotton, S. Raju, F. Balkwill, J. Wilson, Analysis of chemokines and W.H. Byeon, S.H. Eom, K.S. Ha, Y.M. Kim, P.H. Kim, Mechanisms underlying TGF-
chemokine receptor expression in ovarian cancer ascites, Clin. Cancer Res. 8 beta1-induced expression of VEGF and Flk-1 in mouse macrophages and their
(2002) 1108–1114. implications for angiogenesis, J. Leukoc. Biol. 81 (2007) 557–566.
[44] J.M. Roda, L.A. Sumner, R. Evans, G.S. Phillips, C.B. Marsh, T.D. Eubank, Hypoxia- [68] S.L. Rego, R.S. Helms, D. Dreau, Breast tumor cell TACE-shed MCSF promotes pro-
inducible factor-2alpha regulates GM-CSF-derived soluble vascular endothelial angiogenic macrophages through NF-kappaB signaling, Angiogenesis 17 (2014)
growth factor receptor 1 production from macrophages and inhibits tumor growth 573–585.
and angiogenesis, J. Immunol. (Baltimore, Md.) 187 (2011) (1950) 1970–1976. [69] M. Vasse, J. Pourtau, V. Trochon, M. Muraine, J.-P. Vannier, H. Lu, J. Soria,
[45] J.Y. Yhee, C.H. Yu, J.H. Kim, K.S. Im, N.H. Kim, B.W. Brodersen, A.R. Doster, C. Soria, Oncostatin M induces angiogenesis in vitro and in vivo, Arterioscler.
J.H. Sur, Angiogenesis and expression of vascular endothelial growth factor, Thromb. Vasc. Biol. 19 (1999) 1835–1842.
tumour necrosis factor-alpha and hypoxia inducible factor-1alpha in canine renal [70] C. Murdoch, C.E. Lewis, Macrophage migration and gene expression in response to
cell carcinoma, J. Comp. Pathol. 147 (2012) 129–138. tumor hypoxia, Int. J. Cancer 117 (2005) 701–708.
[46] J.M. Roda, Y. Wang, L.A. Sumner, G.S. Phillips, C.B. Marsh, T.D. Eubank, [71] B. Burke, A. Giannoudis, K.P. Corke, D. Gill, M. Wells, L. Ziegler-Heitbrock,
Stabilization of HIF-2alpha induces sVEGFR-1 production from tumor-associated C.E. Lewis, Hypoxia-induced gene expression in human macrophages: implications
macrophages and decreases tumor growth in a murine melanoma model, J. for ischemic tissues and hypoxia-regulated gene therapy, Am. J. Pathol. 163
Immunol. (Baltimore, Md.) 189 (2012) (1950) 3168–3177. (2003) 1233–1243.
[47] J. Zhang, J. Cao, S. Ma, R. Dong, W. Meng, M. Ying, Q. Weng, Z. Chen, J. Ma, [72] G. Bergers, R. Brekken, G. McMahon, T.H. Vu, T. Itoh, K. Tamaki, K. Tanzawa,
Q. Fang, Q. He, B. Yang, Tumor hypoxia enhances non-small cell lung Cancer P. Thorpe, S. Itohara, Z. Werb, D. Hanahan, Matrix metalloproteinase-9 triggers
Metastasis by selectively promoting macrophage M2 polarization through the the angiogenic switch during carcinogenesis, Nat. Cell Biol. 2 (2000) 737–744.
activation of ERK signaling, Oncotarget 5 (2014) 9664–9677. [73] V. Martin, D. Liu, J. Fueyo, C. Gomez-Manzano, Tie2: a journey from normal
[48] K.L. Talks, H. Turley, K.C. Gatter, P.H. Maxwell, C.W. Pugh, P.J. Ratcliffe, angiogenesis to cancer and beyond, Histol. Histopathol. 23 (2008) 773–780.
A.L. Harris, The expression and distribution of the hypoxia-inducible factors HIF- [74] M. Felcht, R. Luck, A. Schering, P. Seidel, K. Srivastava, J. Hu, A. Bartol,
1alpha and HIF-2alpha in normal human tissues, cancers, and tumor-associated Y. Kienast, C. Vettel, E.K. Loos, S. Kutschera, S. Bartels, S. Appak, E. Besemfelder,
macrophages, Am. J. Pathol. 157 (2000) 411–421. D. Terhardt, E. Chavakis, T. Wieland, C. Klein, M. Thomas, A. Uemura, S. Goerdt,
[49] A.L. Doedens, C. Stockmann, M.P. Rubinstein, D. Liao, N. Zhang, D.G. DeNardo, H.G. Augustin, Angiopoietin-2 differentially regulates angiogenesis through TIE2
L.M. Coussens, M. Karin, A.W. Goldrath, R.S. Johnson, Macrophage expression of and integrin signaling, J. Clin. Invest. 122 (2012) 1991–2005.
hypoxia-inducible factor-1 alpha suppresses T-cell function and promotes tumor [75] H. Huang, J.Y. Lai, J. Do, D. Liu, L. Li, J. Del Rosario, V.R. Doppalapudi, S. Pirie-
progression, Cancer Res. 70 (2010) 7465–7475. Shepherd, N. Levin, C. Bradshaw, G. Woodnutt, R. Lappe, A. Bhat, Specifically
[50] T. Schioppa, B. Uranchimeg, A. Saccani, S.K. Biswas, A. Doni, A. Rapisarda, targeting angiopoietin-2 inhibits angiogenesis, Tie2-expressing monocyte infiltra-
S. Bernasconi, S. Saccani, M. Nebuloni, L. Vago, A. Mantovani, G. Melillo, A. Sica, tion, and tumor growth, Clin. Cancer Res. 17 (2011) 1001–1011.
Regulation of the chemokine receptor CXCR4 by hypoxia, J. Exp. Med. 198 (2003) [76] S.B. Coffelt, A.O. Tal, A. Scholz, M. De Palma, S. Patel, C. Urbich, S.K. Biswas,
1391–1402. C. Murdoch, K.H. Plate, Y. Reiss, C.E. Lewis, Angiopoietin-2 regulates gene
[51] C. Tripathi, B.N. Tewari, R.K. Kanchan, K.S. Baghel, N. Nautiyal, R. Shrivastava, expression in TIE2-expressing monocytes and augments their inherent proangio-
H. Kaur, M.L. Bhatt, S. Bhadauria, Macrophages are recruited to hypoxic tumor genic functions, Cancer Res. 70 (2010) 5270–5280.
areas and acquire a pro-angiogenic M2-polarized phenotype via hypoxic cancer [77] S.B. Coffelt, Y.-Y. Chen, M. Muthana, A.F. Welford, A.O. Tal, A. Scholz, K.H. Plate,
cell derived cytokines Oncostatin M and Eotaxin, Oncotarget 5 (2014) 5350–5368. Y. Reiss, C. Murdoch, M. De Palma, C.E. Lewis, Angiopoietin 2 stimulates TIE2-
[52] Z. Wen, H. Liu, M. Li, B. Li, W. Gao, Q. Shao, B. Fan, F. Zhao, Q. Wang, Q. Xie, expressing monocytes to suppress T cell activation and to promote regulatory T
Y. Yang, J. Yu, X. Qu, Increased metabolites of 5-lipoxygenase from hypoxic cell expansion, J. Immunol. 186 (2011) 4183–4190.
ovarian cancer cells promote tumor-associated macrophage infiltration, Oncogene [78] Y. Zhu, C. Lee, F. Shen, R. Du, W.L. Young, G.Y. Yang, Angiopoietin-2 facilitates
34 (2015) 1241–1252. vascular endothelial growth factor-induced angiogenesis in the mature mouse
[53] E. Van Overmeire, D. Laoui, J. Keirsse, J.A. Van Ginderachter, Hypoxia and tumor- brain, Stroke 36 (2005) 1533–1537.
associated macrophages: a deadly alliance in support of tumor progression, [79] F. Che, X. Heng, H. Zhang, Q. Su, B. Zhang, Y. Chen, Z. Zhang, Y. Du, L. Wang,
Oncoimmunology 3 (2014) e27561. Novel B7-H4-mediated crosstalk between human non-Hodgkin lymphoma cells
[54] A. Casazza, D. Laoui, M. Wenes, S. Rizzolio, N. Bassani, M. Mambretti, and tumor-associated macrophages leads to immune evasion via secretion of IL-6
S. Deschoemaeker, J.A. Van Ginderachter, L. Tamagnone, M. Mazzone, Impeding and IL-10, Cancer Immunol. Immunother. (2017). http://dx.doi.org/10.1007/
macrophage entry into hypoxic tumor areas by Sema3A/Nrp1 signaling blockade s00262-017-1961-7.
inhibits angiogenesis and restores antitumor immunity, Cancer Cell 24 (2013) [80] J.G. Quatromoni, E. Eruslanov, Tumor-associated macrophages: function, pheno-
695–709. type, and link to prognosis in human lung cancer, Am. J. Transl. Res. 4 (2012)
[55] P. Dubey, R. Shrivastava, C. Tripathi, N.K. Jain, B.N. Tewari, M.U. Lone, 376–389.
K.S. Baghel, V. Kumar, S. Misra, S. Bhadauria, M.L. Bhatt, Cyclooxygenase-2 [81] N.L. Costa, M.C. Valadares, P.P. Souza, E.F. Mendonca, J.C. Oliveira, T.A. Silva,
inhibition attenuates hypoxic cancer cells induced m2-polarization of macro- A.C. Batista, Tumor-associated macrophages and the profile of inflammatory
phages, Cell. Mol. Biol. (Noisy-le-Grand, France) 60 (2014) 10–15. cytokines in oral squamous cell carcinoma, Oral Oncol. 49 (2013) 216–223.
[56] M.J. Grimshaw, F.R. Balkwill, Inhibition of monocyte and macrophage chemotaxis [82] T.M. Walker, C.J. Burger, K.D. Elgert, Tumor growth alters T cell and macrophage
by hypoxia and inflammation–a potential mechanism, Eur. J. Immunol. 31 (2001) production of and responsiveness to granulocyte-macrophage colony-stimulating
480–489. factor: partial dysregulation through interleukin-10, Cell. Immunol. 154 (1994)
[57] V.L. Silva, W.T. Al-Jamal, Exploiting the cancer niche: tumor-associated macro- 342–357.
phages and hypoxia as promising synergistic targets for nano-based therapy, J. [83] L. Yang, Y. Zhang, Tumor-associated macrophages: from basic research to clinical
Control. Release 253 (2017) 82–96. application, J. Hematol. Oncol. 10 (2017) 58.
[58] V. Riabov, A. Gudima, N. Wang, A. Mickley, A. Orekhov, J. Kzhyshkowska, Role of [84] D.M. Kuang, Q. Zhao, C. Peng, J. Xu, J.P. Zhang, C. Wu, L. Zheng, Activated
tumor associated macrophages in tumor angiogenesis and lymphangiogenesis, monocytes in peritumoral stroma of hepatocellular carcinoma foster immune
Front. Physiol. 5 (2014) 75. privilege and disease progression through PD-L1, J. Exp. Med. 206 (2009)
[59] E.Y. Lin, J.F. Li, L. Gnatovskiy, Y. Deng, L. Zhu, D.A. Grzesik, H. Qian, X.N. Xue, 1327–1337.
J.W. Pollard, Macrophages regulate the angiogenic switch in a mouse model of [85] T. Lu, R. Ramakrishnan, S. Altiok, J.I. Youn, P. Cheng, E. Celis, V. Pisarev,
breast cancer, Cancer Res. 66 (2006) 11238–11246. S. Sherman, M.B. Sporn, D. Gabrilovich, Tumor-infiltrating myeloid cells induce
[60] F.J. Gonzalez, L. Vicioso, M. Alvarez, I. Sevilla, E. Marques, E. Gallego, L. Alonso, tumor cell resistance to cytotoxic T cells in mice, J. Clin. Invest. 121 (2011)
A. Matilla, E. Alba, Association between VEGF expression in tumour-associated 4015–4029.
macrophages and elevated serum VEGF levels in primary colorectal cancer [86] J. Yang, X. Li, X. Liu, Y. Liu, The role of tumor-associated macrophages in breast
patients, Cancer Biomark. 3 (2007) 325–333. carcinoma invasion and metastasis, Int. J. Clin. Exp. Pathol. 8 (2015) 6656–6664.
[61] M.S. Kluger, O.R. Colegio, Lymphangiogenesis linked to VEGF-C from tumor- [87] W. Cao, J.H. Peters, D. Nieman, M. Sharma, T. Watson, J. Yu, Macrophage subtype
associated macrophages: accomplices to metastasis by cutaneous squamous cell predicts lymph node metastasis in oesophageal adenocarcinoma and promotes
carcinoma [quest], J. Invest. Dermatol. 131 (2011) 17–19. cancer cell invasion in vitro, Br. J. Cancer 113 (2015) 738–746.
[62] C. Guo, A. Buranych, D. Sarkar, P.B. Fisher, X.-Y. Wang, The role of tumor- [88] M. Falleni, F. Savi, D. Tosi, E. Agape, A. Cerri, L. Moneghini, G.P. Bulfamante, M1
associated macrophages in tumor vascularization, Vasc. Cell 5 (2013) 20. and M2 macrophages' clinicopathological significance in cutaneous melanoma,
[63] N. Linde, W. Lederle, S. Depner, N. van Rooijen, C.M. Gutschalk, M.M. Mueller, Melanoma Res. (2017). http://dx.doi.org/10.1097/CMR.0000000000000352.
Vascular endothelial growth factor-induced skin carcinogenesis depends on [89] S. Goswami, E. Sahai, J.B. Wyckoff, M. Cammer, D. Cox, F.J. Pixley, E.R. Stanley,
recruitment and alternative activation of macrophages, J. Pathol. 227 (2012) J.E. Segall, J.S. Condeelis, Macrophages promote the invasion of breast carcinoma
17–28. cells via a colony-stimulating factor-1/epidermal growth factor paracrine loop,
[64] B.L. Krock, N. Skuli, M.C. Simon, Hypoxia-induced angiogenesis: good and evil, Cancer Res. 65 (2005) 5278–5283.
Genes & Cancer 2 (2011) 1117–1133. [90] J.W. Pollard, Tumour-educated macrophages promote tumour progression and

103
Y. Singh et al. Journal of Controlled Release 254 (2017) 92–106

metastasis, Nat. Rev. Cancer 4 (2004) 71–78. J.D. Robertson, V.M. Rotello, J.M. Reid, P. Mukherjee, Modulating pharmacoki-
[91] E.Y. Lin, A.V. Nguyen, R.G. Russell, J.W. Pollard, Colony-stimulating factor 1 netics, tumor uptake and biodistribution by engineered nanoparticles, PLoS One 6
promotes progression of mammary tumors to malignancy, J. Exp. Med. 193 (2001) (2011) e24374.
727–740. [117] V.K. Pawar, J.G. Meher, Y. Singh, M. Chaurasia, B. Surendar Reddy,
[92] Y. Wu, B.P. Zhou, TNF-α/NF-κB/Snail pathway in cancer cell migration and M.K. Chourasia, Targeting of gastrointestinal tract for amended delivery of
invasion, Br. J. Cancer 102 (2010) 639–644. protein/peptide therapeutics: strategies and industrial perspectives, J. Control.
[93] T. Hagemann, J. Wilson, H. Kulbe, N.F. Li, D.A. Leinster, K. Charles, F. Klemm, Release 196 (2014) 168–183.
T. Pukrop, C. Binder, F.R. Balkwill, Macrophages induce invasiveness of epithelial [118] J.K. Liao, U. Laufs, Pleiotropic effects of statins, Annu. Rev. Pharmacol. Toxicol. 45
cancer cells via NF-kappa B and JNK, J. Immunol. (Baltimore, Md.) 175 (2005) (2005) 89–118.
(1950) 1197–1205. [119] M.I. Yilmaz, Y. Baykal, M. Kilic, A. Sonmez, F. Bulucu, A. Aydin, A. Sayal,
[94] S.J. Lee, S.S. Park, Y.H. Cho, K. Park, E.J. Kim, K.H. Jung, S.K. Kim, W.J. Kim, I.H. Kocar, Effects of statins on oxidative stress, Biol. Trace Elem. Res. 98 (2004)
S.K. Moon, Activation of matrix metalloproteinase-9 by TNF-alpha in human 119–127.
urinary bladder cancer HT1376 cells: the role of MAP kinase signaling pathways, [120] X.F. Qi, D.H. Kim, Y.S. Yoon, S.K. Kim, D.Q. Cai, Y.C. Teng, K.Y. Shim, K.J. Lee,
Oncol. Rep. 19 (2008) 1007–1013. Involvement of oxidative stress in simvastatin-induced apoptosis of murine CT26
[95] E.C. Woenne, W. Lederle, S. Zwick, M. Palmowski, H. Krell, W. Semmler, colon carcinoma cells, Toxicol. Lett. 199 (2010) 277–287.
M.M. Mueller, F. Kiessling, MMP inhibition blocks fibroblast-dependent skin [121] M.C. Alupei, E. Licarete, L. Patras, M. Banciu, Liposomal simvastatin inhibits
cancer invasion, reduces vascularization and alters VEGF-A and PDGF-BB ex- tumor growth via targeting tumor-associated macrophages-mediated oxidative
pression, Anticancer Res. 30 (2010) 703–711. stress, Cancer Lett. 356 (2015) 946–952.
[96] D. Belotti, C. Calcagno, A. Garofalo, D. Caronia, E. Riccardi, R. Giavazzi, [122] L. Sun, B. Chen, R. Jiang, J. Li, B. Wang, Resveratrol inhibits lung cancer growth
G. Taraboletti, Vascular endothelial growth factor stimulates organ-specific host by suppressing M2-like polarization of tumor associated macrophages, Cell.
matrix metalloproteinase-9 expression and ovarian cancer invasion, Mol. Cancer Immunol. 311 (2017) 86–93.
Res. 6 (2008) 525–534. [123] Z. Huang, Z. Zhang, Y. Jiang, D. Zhang, J. Chen, L. Dong, J. Zhang, Targeted
[97] J.W. Tjiu, J.S. Chen, C.T. Shun, S.J. Lin, Y.H. Liao, C.Y. Chu, T.F. Tsai, H.C. Chiu, delivery of oligonucleotides into tumor-associated macrophages for cancer
Y.S. Dai, H. Inoue, P.C. Yang, M.L. Kuo, S.H. Jee, Tumor-associated macrophage- immunotherapy, J. Control. Release 158 (2012) 286–292.
induced invasion and angiogenesis of human basal cell carcinoma cells by [124] Y. Wang, Y.X. Lin, S.L. Qiao, H.W. An, Y. Ma, Z.Y. Qiao, R.P. Rajapaksha, H. Wang,
cyclooxygenase-2 induction, J. Invest. Dermatol. 129 (2009) 1016–1025. Polymeric nanoparticles promote macrophage reversal from M2 to M1 phenotypes
[98] J. Chen, Y. Yao, C. Gong, F. Yu, S. Su, J. Chen, B. Liu, H. Deng, F. Wang, L. Lin, in the tumor microenvironment, Biomaterials 112 (2017) 153–163.
H. Yao, F. Su, K.S. Anderson, Q. Liu, M.E. Ewen, X. Yao, E. Song, CCL18 from [125] M. Banciu, R.M. Schiffelers, G. Storm, Investigation into the role of tumor-
tumor-associated macrophages promotes breast cancer metastasis via PITPNM3, associated macrophages in the antitumor activity of Doxil, Pharm. Res. 25 (2008)
Cancer Cell 19 (2011) 541–555. 1948–1955.
[99] T. Iriki, K. Ohnishi, Y. Fujiwara, H. Horlad, Y. Saito, C. Pan, K. Ikeda, T. Mori, [126] M. Banciu, J.M. Metselaar, R.M. Schiffelers, G. Storm, Antitumor activity of
M. Suzuki, H. Ichiyasu, H. Kohrogi, M. Takeya, Y. Komohara, The cell-cell liposomal prednisolone phosphate depends on the presence of functional tumor-
interaction between tumor-associated macrophages and small cell lung cancer associated macrophages in tumor tissue, Neoplasia (New York, N.Y.) 10 (2008)
cells is involved in tumor progression via STAT3 activation, Lung Cancer 106 108–117.
(2017) 22–32. [127] J.D. Veltman, M.E.H. Lambers, M. van Nimwegen, R.W. Hendriks,
[100] Y. Singh, P. Ojha, M. Srivastava, M.K. Chourasia, Reinvestigating nanoprecipita- H.C. Hoogsteden, J.P.J.J. Hegmans, J.G.J.V. Aerts, Zoledronic acid impairs
tion via Box–Behnken design: a systematic approach, J. Microencapsul. 32 (2015) myeloid differentiation to tumour-associated macrophages in mesothelioma, Br. J.
75–85. Cancer 103 (2010) 629–641.
[101] Y. Singh, M. Singh, J.G. Meher, V.K. Pawar, M.K. Chourasia, Trichotomous gastric [128] T.L. Rogers, I. Holen, Tumour macrophages as potential targets of bisphospho-
retention of amorphous capecitabine: an attempt to overcome pharmacokinetic nates, J. Transl. Med. 9 (2011) 177.
gap, Int. J. Pharm. 478 (2015) 811–821. [129] S. Junankar, G. Shay, J. Jurczyluk, N. Ali, J. Down, N. Pocock, A. Parker,
[102] Y. Singh, S. Tomar, S. Khan, J.G. Meher, V.K. Pawar, K. Raval, K. Sharma, A. Nguyen, S. Sun, B. Kashemirov, C.E. McKenna, P.I. Croucher, A. Swarbrick,
P.K. Singh, M. Chaurasia, B. Surendar Reddy, M.K. Chourasia, Bridging small K. Weilbaecher, T.G. Phan, M.J. Rogers, Real-time intravital imaging establishes
interfering RNA with giant therapeutic outcomes using nanometric liposomes, J. tumor-associated macrophages as the extraskeletal target of bisphosphonate
Control. Release 220 (Part A) (2015) 368–387. action in cancer, Cancer Discov. 5 (2015) 35–42.
[103] V.K. Pawar, Y. Singh, J.G. Meher, S. Gupta, M.K. Chourasia, Engineered [130] M. Marra, G. Salzano, C. Leonetti, M. Porru, R. Franco, S. Zappavigna, G. Liguori,
nanocrystal technology: in-vivo fate, targeting and applications in drug delivery, J. G. Botti, P. Chieffi, M. Lamberti, G. Vitale, A. Abbruzzese, M.I. La Rotonda, G. De
Control. Release 183 (2014) 51–66. Rosa, M. Caraglia, New self-assembly nanoparticles and stealth liposomes for the
[104] X. Tang, C. Mo, Y. Wang, D. Wei, H. Xiao, Anti-tumour strategies aiming to target delivery of zoledronic acid: a comparative study, Biotechnol. Adv. 30 (2012)
tumour-associated macrophages, Immunology 138 (2013) 93–104. 302–309.
[105] K. Pienta, Targeting the m2-tumor associated macrophage for cancer therapy, in, [131] N.M. Reusser, H.J. Dalton, S. Pradeep, V. Gonzalez-Villasana, N.B. Jennings,
Google Patents, 2015. H.G. Vasquez, Y. Wen, R. Rupaimoole, A.S. Nagaraja, K. Gharpure, T. Miyake,
[106] Y. Singh, J.G. Meher, K. Raval, F.A. Khan, M. Chaurasia, N.K. Jain, J. Huang, W. Hu, G. Lopez-Berestein, A.K. Sood, Clodronate inhibits tumor
M.K. Chourasia, Nanoemulsion: concepts, development and applications in drug angiogenesis in mouse models of ovarian cancer, Cancer Biol. Ther. 15 (2014)
delivery, J. Control. Release 252 (2017) 28–49. 1061–1067.
[107] V.K. Pawar, S.B. Panchal, Y. Singh, J.G. Meher, K. Sharma, P. Singh, H.K. Bora, [132] J. Zhu, Y. Zheng, Z. Zhou, Oral adjuvant clodronate therapy could improve overall
A. Singh, D. Datta, M.K. Chourasia, Immunotherapeutic vitamin E nanoemulsion survival in early breast cancer: results from an updated systematic review and
synergies the antiproliferative activity of paclitaxel in breast cancer cells via meta-analysis, Eur. J. Cancer 49 (2013) 2086–2092.
modulating Th1 and Th2 immune response, J. Control. Release 196 (2014) [133] N.R. Miselis, Z.J. Wu, N. Van Rooijen, A.B. Kane, Targeting tumor-associated
295–306. macrophages in an orthotopic murine model of diffuse malignant mesothelioma,
[108] Y. Singh, A. Chandrashekhar, J.G. Meher, K.K. Durga Rao Viswanadham, Mol. Cancer Ther. 7 (2008) 788–799.
V.K. Pawar, K. Raval, K. Sharma, P.K. Singh, A. Kumar, M.K. Chourasia, Nanosized [134] Y.N. Kimura, K. Watari, A. Fotovati, F. Hosoi, K. Yasumoto, H. Izumi, K. Kohno,
complexation assemblies housed inside reverse micelles churn out monocytic K. Umezawa, H. Iguchi, K. Shirouzu, S. Takamori, M. Kuwano, M. Ono,
delivery cores for bendamustine hydrochloride, Eur. J. Pharm. Biopharm. 113 Inflammatory stimuli from macrophages and cancer cells synergistically promote
(2017) 198–210. tumor growth and angiogenesis, Cancer Sci. 98 (2007) 2009–2018.
[109] V.K. Pawar, S. Gupta, Y. Singh, J.G. Meher, K. Sharma, P. Singh, A. Gupta, [135] S.M. Zeisberger, B. Odermatt, C. Marty, A.H. Zehnder-Fjallman, K. Ballmer-Hofer,
H.K. Bora, M. Chaurasia, M.K. Chourasia, Pluronic F-127 stabilised docetaxel R.A. Schwendener, Clodronate-liposome-mediated depletion of tumour-associated
nanocrystals improve apoptosis by mitochondrial depolarization in breast cancer macrophages: a new and highly effective antiangiogenic therapy approach, Br. J.
cells: pharmacokinetics and toxicity assessment, J. Biomed. Nanotechnol. 11 Cancer 95 (2006) 272–281.
(2015) 1747–1763. [136] J.M. Fritz, M.A. Tennis, D.J. Orlicky, H. Lin, C. Ju, E.F. Redente, K.S. Choo,
[110] P. Verma, J.G. Meher, S. Asthana, V.K. Pawar, M. Chaurasia, M.K. Chourasia, T.A. Staab, R.J. Bouchard, D.T. Merrick, A.M. Malkinson, L.D. Dwyer-Nield,
Perspectives of nanoemulsion assisted oral delivery of docetaxel for improved Depletion of tumor-associated macrophages slows the growth of chemically
chemotherapy of cancer, Drug Deliv. 23 (2016) 479–488. induced mouse lung adenocarcinomas, Front. Immunol. 5 (2014) 587.
[111] A.Z. Wang, R. Langer, O.C. Farokhzad, Nanoparticle delivery of cancer drugs, [137] R. Pal, B. Chakraborty, A. Nath, L.M. Singh, M. Ali, D.S. Rahman, S.K. Ghosh,
Annu. Rev. Med. 63 (2012) 185–198. A. Basu, S. Bhattacharya, R. Baral, M. Sengupta, Noble metal nanoparticle-induced
[112] W. Poon, X. Zhang, J. Nadeau, Nanoparticle drug formulations for cancer oxidative stress modulates tumor associated macrophages (TAMs) from an M2 to
diagnosis and treatment, Crit. Rev. Oncog. 19 (2014) 223–245. M1 phenotype: an in vitro approach, Int. Immunopharmacol. 38 (2016) 332–341.
[113] M.E. Davis, Z. Chen, D.M. Shin, Nanoparticle therapeutics: an emerging treatment [138] X. Wu, B.C. Schulte, Y. Zhou, D. Haribhai, A.C. Mackinnon, J.A. Plaza,
modality for cancer, Nat. Rev. Drug Discov. 7 (2008) 771–782. C.B. Williams, S.T. Hwang, Depletion of M2-like tumor-associated macrophages
[114] A. Schroeder, D.A. Heller, M.M. Winslow, J.E. Dahlman, G.W. Pratt, R. Langer, delays cutaneous T-cell lymphoma development in vivo, J. Invest. Dermatol. 134
T. Jacks, D.G. Anderson, Treating metastatic cancer with nanotechnology, Nat. (2014) 2814–2822.
Rev. Cancer 12 (2012) 39–50. [139] M. Niu, Y.W. Naguib, A.M. Aldayel, Y.C. Shi, S.D. Hursting, M.A. Hersh, Z. Cui,
[115] F. Jia, X. Liu, L. Li, S. Mallapragada, B. Narasimhan, Q. Wang, Multifunctional Biodistribution and in vivo activities of tumor-associated macrophage-targeting
nanoparticles for targeted delivery of immune activating and cancer therapeutic nanoparticles incorporated with doxorubicin, Mol. Pharm. 11 (2014) 4425–4436.
agents, J. Control. Release 172 (2013) 1020–1034. [140] F. Piaggio, V. Kondylis, F. Pastorino, D. Di Paolo, P. Perri, I. Cossu, F. Schorn,
[116] R.R. Arvizo, O.R. Miranda, D.F. Moyano, C.A. Walden, K. Giri, R. Bhattacharya, C. Marinaccio, D. Murgia, A. Daga, F. Raggi, M. Loi, L. Emionite, E. Ognio,

104
Y. Singh et al. Journal of Controlled Release 254 (2017) 92–106

M. Pasparakis, D. Ribatti, M. Ponzoni, C. Brignole, A novel liposomal Clodronate globulin mobilization and cysteine proteinases in embryonic axes and cotyledons
depletes tumor-associated macrophages in primary and metastatic melanoma: during germination and seedling growth of vetch (Vicia sativa L.), J. Exp. Bot. 51
anti-angiogenic and anti-tumor effects, J. Control. Release 223 (2016) 165–177. (2000) 1423–1433.
[141] T. Luo, J. Sun, S. Zhu, J. He, L. Hao, L. Xiao, Y. Zhu, Q. Wang, X. Pan, Z. Wang, [164] J.M. Chen, P.M. Dando, N.D. Rawlings, M.A. Brown, N.E. Young, R.A. Stevens,
S. Chang, Ultrasound-mediated destruction of oxygen and paclitaxel loaded dual- E. Hewitt, C. Watts, A.J. Barrett, Cloning, isolation, and characterization of
targeting microbubbles for intraperitoneal treatment of ovarian cancer xenografts, mammalian legumain, an asparaginyl endopeptidase, J. Biol. Chem. 272 (1997)
Cancer Lett. 391 (2017) 1–11. 8090–8098.
[142] S.W. Jones, R.A. Roberts, G.R. Robbins, J.L. Perry, M.P. Kai, K. Chen, T. Bo, [165] R.V. Murthy, G. Arbman, J. Gao, G.D. Roodman, X.F. Sun, Legumain expression in
M.E. Napier, J.P. Ting, J.M. Desimone, J.E. Bear, Nanoparticle clearance is relation to clinicopathologic and biological variables in colorectal cancer, Clin.
governed by Th1/Th2 immunity and strain background, J. Clin. Invest. 123 (2013) Cancer Res. 11 (2005) 2293–2299.
3061–3073. [166] C. Liu, C. Sun, H. Huang, K. Janda, T. Edgington, Overexpression of legumain in
[143] J. Hoppstadter, M. Seif, A. Dembek, C. Cavelius, H. Huwer, A. Kraegeloh, tumors is significant for invasion/metastasis and a candidate enzymatic target for
A.K. Kiemer, M2 polarization enhances silica nanoparticle uptake by macro- prodrug therapy, Cancer Res. 63 (2003) 2957–2964.
phages, Front. Pharmacol. 6 (2015) 55. [167] N. Li, Q. Liu, Q. Su, C. Wei, B. Lan, J. Wang, G. Bao, F. Yan, Y. Yu, B. Peng, J. Qiu,
[144] D. Wang, N. Phan, C. Isely, L. Bruene, K.M. Bratlie, Effect of surface modification X. Yan, S. Zhang, F. Guo, Effects of legumain as a potential prognostic factor on
and macrophage phenotype on particle internalization, Biomacromolecules 15 gastric cancers, Medical Oncol. (Northwood, London, England) 30 (2013) 621.
(2014) 4102–4110. [168] Y. Luo, H. Zhou, J. Krueger, C. Kaplan, S.H. Lee, C. Dolman, D. Markowitz, W. Wu,
[145] A.K. Fuchs, T. Syrovets, K.A. Haas, C. Loos, A. Musyanovych, V. Mailander, C. Liu, R.A. Reisfeld, R. Xiang, Targeting tumor-associated macrophages as a novel
K. Landfester, T. Simmet, Carboxyl- and amino-functionalized polystyrene nano- strategy against breast cancer, J. Clin. Invest. 116 (2006) 2132–2141.
particles differentially affect the polarization profile of M1 and M2 macrophage [169] Y. Lin, C. Wei, Y. Liu, Y. Qiu, C. Liu, F. Guo, Selective ablation of tumor-associated
subsets, Biomaterials 85 (2016) 78–87. macrophages suppresses metastasis and angiogenesis, Cancer Sci. 104 (2013)
[146] R.A. Ortega, W.J. Barham, B. Kumar, O. Tikhomirov, I.D. McFadden, F.E. Yull, 1217–1225.
T.D. Giorgio, Biocompatible mannosylated endosomal-escape nanoparticles en- [170] Z. Liu, M. Xiong, J. Gong, Y. Zhang, N. Bai, Y. Luo, L. Li, Y. Wei, Y. Liu, X. Tan,
hance selective delivery of short nucleotide sequences to tumor associated R. Xiang, Legumain protease-activated TAT-liposome cargo for targeting tumours
macrophages, Nano 7 (2015) 500–510. and their microenvironment, Nat. Commun. 5 (2014) 4280.
[147] S.S. Yu, C.M. Lau, W.J. Barham, H.M. Onishko, C.E. Nelson, H. Li, C.A. Smith, [171] X. Zhang, W. Tian, X. Cai, X. Wang, W. Dang, H. Tang, H. Cao, L. Wang, T. Chen,
F.E. Yull, C.L. Duvall, T.D. Giorgio, Macrophage-specific RNA interference Hydrazinocurcumin Encapsuled nanoparticles "re-educate" tumor-associated
targeting via “click”, mannosylated polymeric micelles, Mol. Pharm. 10 (2013) macrophages and exhibit anti-tumor effects on breast cancer following STAT3
975–987. suppression, PLoS One 8 (2013) e65896.
[148] L.W. Locke, M.W. Mayo, A.D. Yoo, M.B. Williams, S.S. Berr, PET imaging of tumor [172] M. Cieslewicz, J. Tang, J.L. Yu, H. Cao, M. Zavaljevski, K. Motoyama, A. Lieber,
associated macrophages using mannose coated 64Cu liposomes, Biomaterials 33 E.W. Raines, S.H. Pun, Targeted delivery of proapoptotic peptides to tumor-
(2012) 7785–7793. associated macrophages improves survival, Proc. Natl. Acad. Sci. 110 (2013)
[149] S. Zhu, M. Niu, H. O'Mary, Z. Cui, Targeting of tumor-associated macrophages 15919–15924.
made possible by PEG-sheddable, mannose-modified nanoparticles, Mol. Pharm. [173] J. Conde, C. Bao, Y. Tan, D. Cui, E.R. Edelman, H.S. Azevedo, H.J. Byrne, N. Artzi,
10 (2013) 3525–3530. F. Tian, Dual targeted immunotherapy via in vivo delivery of Biohybrid RNAi-
[150] X. Zhan, L. Jia, Y. Niu, H. Qi, X. Chen, Q. Zhang, J. Zhang, Y. Wang, L. Dong, peptide nanoparticles to tumor-associated macrophages and cancer cells, Adv.
C. Wang, Targeted depletion of tumour-associated macrophages by an alendro- Funct. Mater. 25 (2015) 4183–4194.
nate–glucomannan conjugate for cancer immunotherapy, Biomaterials 35 (2014) [174] Y. Lu, G. Yunxiang, P. Ryan, D. Liming, K. Julian, Z. Mei, Development of Y-shaped
10046–10057. peptide for constructing nanoparticle systems targeting tumor-associated macro-
[151] S. Zhou, T. Zhang, B. Peng, X. Luo, X. Liu, L. Hu, Y. Liu, D. Di, Y. Song, Y. Deng, phages in vitro and in vivo, Mater. Res. Express 1 (2014) 025007.
Targeted delivery of epirubicin to tumor-associated macrophages by sialic acid- [175] V. Fogal, L. Zhang, S. Krajewski, E. Ruoslahti, Mitochondrial/cell-surface protein
cholesterol conjugate modified liposomes with improved antitumor activity, Int. J. p32/gC1qR as a molecular target in tumor cells and tumor stroma, Cancer Res. 68
Pharm. (2017). (2008) 7210–7218.
[152] R. Weissleder, K. Kelly, E.Y. Sun, T. Shtatland, L. Josephson, Cell-specific targeting [176] P. Laakkonen, M.E. Åkerman, H. Biliran, M. Yang, F. Ferrer, T. Karpanen,
of nanoparticles by multivalent attachment of small molecules, Nat. Biotechnol. 23 R.M. Hoffman, E. Ruoslahti, Antitumor activity of a homing peptide that targets
(2005) 1418–1423. tumor lymphatics and tumor cells, Proc. Natl. Acad. Sci. U. S. A. 101 (2004)
[153] A. Leimgruber, C. Berger, V. Cortez-Retamozo, M. Etzrodt, A.P. Newton, 9381–9386.
P. Waterman, J.L. Figueiredo, R.H. Kohler, N. Elpek, T.R. Mempel, F.K. Swirski, [177] Z. Yan, F. Wang, Z. Wen, C. Zhan, L. Feng, Y. Liu, X. Wei, C. Xie, W. Lu, LyP-1-
M. Nahrendorf, R. Weissleder, M.J. Pittet, Behavior of endogenous tumor- conjugated PEGylated liposomes: a carrier system for targeted therapy of
associated macrophages assessed in vivo using a functionalized nanoparticle, lymphatic metastatic tumor, J. Control. Release 157 (2012) 118–125.
Neoplasia (New York, N.Y.) 11 (2009) 459–468. 452 p following 468. [178] L. Zou, A.M. Sustaita, T. Thomas, L. Do, J. Rodriguez, H. Dou, Size effects of
[154] H.E. Daldrup-Link, D. Golovko, B. Ruffell, D.G. Denardo, R. Castaneda, C. Ansari, Nanocomplex on tumor associated macrophages targeted delivery for glioma, J.
J. Rao, G.A. Tikhomirov, M.F. Wendland, C. Corot, L.M. Coussens, MRI of tumor- Nanomed. Nanotechnol. 6 (2015) 1.
associated macrophages with clinically applicable iron oxide nanoparticles, Clin. [179] L. Zou, Y. Tao, G. Payne, L. Do, T. Thomas, J. Rodriguez, H. Dou, Targeted delivery
Cancer Res. 17 (2011) 5695–5704. of nano-PTX to the brain tumor-associated macrophages, Oncotarget 8 (2017)
[155] G. Raes, L. Brys, B.K. Dahal, J. Brandt, J. Grooten, F. Brombacher, G. Vanham, 6564–6578.
W. Noel, P. Bogaert, T. Boonefaes, A. Kindt, R. Van den Bergh, P.J. Leenen, P. De [180] K. Mizutani, S. Sud, N.A. McGregor, G. Martinovski, B.T. Rice, M.J. Craig,
Baetselier, G.H. Ghassabeh, Macrophage galactose-type C-type lectins as novel Z.S. Varsos, H. Roca, K.J. Pienta, The chemokine CCL2 increases prostate tumor
markers for alternatively activated macrophages elicited by parasitic infections growth and bone metastasis through macrophage and osteoclast recruitment,
and allergic airway inflammation, J. Leukoc. Biol. 77 (2005) 321–327. Neoplasia (New York, N.Y.) 11 (2009) 1235–1242.
[156] S.J. van Vliet, E. Saeland, Y. van Kooyk, Sweet preferences of MGL: carbohydrate [181] S. Svensson, A. Abrahamsson, G.V. Rodriguez, A.K. Olsson, L. Jensen, Y. Cao,
specificity and function, Trends Immunol. 29 (2008) 83–90. C. Dabrosin, CCL2 and CCL5 are novel therapeutic targets for estrogen-dependent
[157] S.K. Biswas, L. Gangi, S. Paul, T. Schioppa, A. Saccani, M. Sironi, B. Bottazzi, breast cancer, Clin. Cancer Res. 21 (2015) 3794–3805.
A. Doni, B. Vincenzo, F. Pasqualini, L. Vago, M. Nebuloni, A. Mantovani, A. Sica, A [182] S.K. Sandhu, K. Papadopoulos, P.C. Fong, A. Patnaik, C. Messiou, D. Olmos,
distinct and unique transcriptional program expressed by tumor-associated G. Wang, B.J. Tromp, T.A. Puchalski, F. Balkwill, B. Berns, S. Seetharam, J.S. de
macrophages (defective NF-kappaB and enhanced IRF-3/STAT1 activation), Blood Bono, A.W. Tolcher, A first-in-human, first-in-class, phase I study of carlumab
107 (2006) 2112–2122. (CNTO 888), a human monoclonal antibody against CC-chemokine ligand 2 in
[158] X. Sun, D. Gao, L. Gao, C. Zhang, X. Yu, B. Jia, F. Wang, Z. Liu, Molecular imaging patients with solid tumors, Cancer Chemother. Pharmacol. 71 (2013) 1041–1050.
of tumor-infiltrating macrophages in a preclinical mouse model of breast cancer, [183] S. Aharinejad, P. Paulus, M. Sioud, M. Hofmann, K. Zins, R. Schafer, E.R. Stanley,
Theranostics 5 (2015) 597–608. D. Abraham, Colony-stimulating factor-1 blockade by antisense oligonucleotides
[159] K. Movahedi, S. Schoonooghe, D. Laoui, I. Houbracken, W. Waelput, K. Breckpot, and small interfering RNAs suppresses growth of human mammary tumor
L. Bouwens, T. Lahoutte, P. De Baetselier, G. Raes, N. Devoogdt, J.A. Van xenografts in mice, Cancer Res. 64 (2004) 5378–5384.
Ginderachter, Nanobody-based targeting of the macrophage mannose receptor for [184] S. Aharinejad, D. Abraham, P. Paulus, H. Abri, M. Hofmann, K. Grossschmidt,
effective in vivo imaging of tumor-associated macrophages, Cancer Res. 72 (2012) R. Schafer, E.R. Stanley, R. Hofbauer, Colony-stimulating factor-1 antisense
4165–4177. treatment suppresses growth of human tumor xenografts in mice, Cancer Res. 62
[160] Y. Komohara, D. Niino, Y. Saito, K. Ohnishi, H. Horlad, K. Ohshima, M. Takeya, (2002) 5317–5324.
Clinical significance of CD163(+) tumor-associated macrophages in patients with [185] C.H. Ries, M.A. Cannarile, S. Hoves, J. Benz, K. Wartha, V. Runza, F. Rey-Giraud,
adult T-cell leukemia/lymphoma, Cancer Sci. 104 (2013) 945–951. L.P. Pradel, F. Feuerhake, I. Klaman, T. Jones, U. Jucknischke, S. Scheiblich,
[161] A. Etzerodt, M.B. Maniecki, J.H. Graversen, H.J. Møller, V.P. Torchilin, K. Kaluza, I.H. Gorr, A. Walz, K. Abiraj, P.A. Cassier, A. Sica, C. Gomez-Roca,
S.K. Moestrup, Efficient intracellular drug-targeting of macrophages using stealth K.E. de Visser, A. Italiano, C. Le Tourneau, J.P. Delord, H. Levitsky, J.Y. Blay,
liposomes directed to the hemoglobin scavenger receptor CD163, J. Control. D. Ruttinger, Targeting tumor-associated macrophages with anti-CSF-1R antibody
Release 160 (2012) 72–80. reveals a strategy for cancer therapy, Cancer Cell 25 (2014) 846–859.
[162] A.A. Kembhavi, D.J. Buttle, C.G. Knight, A.J. Barrett, The two cysteine endopep- [186] C.L. Manthey, D.L. Johnson, C.R. Illig, R.W. Tuman, Z. Zhou, J.F. Baker,
tidases of legume seeds: purification and characterization by use of specific M.A. Chaikin, R.R. Donatelli, C.F. Franks, L. Zeng, C. Crysler, Y. Chen,
fluorometric assays, Arch. Biochem. Biophys. 303 (1993) 208–213. E.J. Yurkow, L. Boczon, S.K. Meegalla, K.J. Wilson, M.J. Wall, J. Chen,
[163] A. Schlereth, C. Becker, C. Horstmann, J. Tiedemann, K. Muntz, Comparison of S.K. Ballentine, H. Ott, C. Baumann, D. Lawrence, B.E. Tomczuk, C.J. Molloy, JNJ-

105
Y. Singh et al. Journal of Controlled Release 254 (2017) 92–106

28312141, a novel orally active colony-stimulating factor-1 receptor/FMS-related R. Sorrentino, M. Zonfrillo, P. Pierimarchi, P. Sinibaldi-Vallebona, Thymosin α1
receptor tyrosine kinase-3 receptor tyrosine kinase inhibitor with potential utility and cancer: action on immune effector and tumor target cells, Ann. N. Y. Acad. Sci.
in solid tumors, bone metastases, and acute myeloid leukemia, Mol. Cancer Ther. 8 1269 (2012) 26–33.
(2009) 3151–3161. [195] G.C. Chan, W.K. Chan, D.M. Sze, The effects of beta-glucan on human immune and
[187] S. Mok, R.C. Koya, C. Tsui, J. Xu, L. Robert, L. Wu, T.G. Graeber, B.L. West, cancer cells, J. Hematol. Oncol. 2 (2009) 25.
G. Bollag, A. Ribas, Inhibition of CSF-1 receptor improves the antitumor efficacy of [196] G. Germano, R. Frapolli, C. Belgiovine, A. Anselmo, S. Pesce, M. Liguori, E. Erba,
adoptive cell transfer immunotherapy, Cancer Res. 74 (2014) 153–161. S. Uboldi, M. Zucchetti, F. Pasqualini, M. Nebuloni, N. van Rooijen, R. Mortarini,
[188] S.K. Chambers, Role of CSF-1 in progression of epithelial ovarian cancer, Future L. Beltrame, S. Marchini, I. Fuso Nerini, R. Sanfilippo, P.G. Casali, S. Pilotti,
Oncol. (London, England) 5 (2009) 1429–1440. C.M. Galmarini, A. Anichini, A. Mantovani, M. D'Incalci, P. Allavena, Role of
[189] D. Duluc, M. Corvaisier, S. Blanchard, L. Catala, P. Descamps, E. Gamelin, macrophage targeting in the antitumor activity of trabectedin, Cancer Cell 23
S. Ponsoda, Y. Delneste, M. Hebbar, P. Jeannin, Interferon-gamma reverses the (2013) 249–262.
immunosuppressive and protumoral properties and prevents the generation of [197] M. Jiao, K.J. Nan, Activation of PI3 kinase/Akt/HIF-1alpha pathway contributes to
human tumor-associated macrophages, Int. J. Cancer 125 (2009) 367–373. hypoxia-induced epithelial-mesenchymal transition and chemoresistance in he-
[190] L.S. Chang, C.H. Leng, Y.C. Yeh, C.C. Wu, H.W. Chen, H.M. Huang, S.J. Liu, Toll- patocellular carcinoma, Int. J. Oncol. 40 (2012) 461–468.
like receptor 9 agonist enhances anti-tumor immunity and inhibits tumor- [198] Y. Cao, X. Liu, W. Lu, Y. Chen, X. Wu, M. Li, X.A. Wang, F. Zhang, L. Jiang,
associated immunosuppressive cells numbers in a mouse cervical cancer model Y. Zhang, Y. Hu, S. Xiang, Y. Shu, R. Bao, H. Li, W. Wu, H. Weng, Y. Yen, Y. Liu,
following recombinant lipoprotein therapy, Mol. Cancer 13 (2014) 60. Fibronectin promotes cell proliferation and invasion through mTOR signaling
[191] S. Hallam, M. Escorcio-Correia, R. Soper, A. Schultheiss, T. Hagemann, Activated pathway activation in gallbladder cancer, Cancer Lett. 360 (2015) 141–150.
macrophages in the tumour microenvironment – dancing to the tune of TLR and [199] M. Hockel, P. Vaupel, Tumor hypoxia: definitions and current clinical, biologic,
NF-κB, J. Pathol. 219 (2009) 143–152. and molecular aspects, J. Natl. Cancer Inst. 93 (2001) 266–276.
[192] A.L. Rakhmilevich, K.L. Alderson, P.M. Sondel, T-cell-independent antitumor [200] A.M. Shannon, D.J. Bouchier-Hayes, C.M. Condron, D. Toomey, Tumour hypoxia,
effects of CD40 ligation, Int. Rev. Immunol. 31 (2012) 267–278. chemotherapeutic resistance and hypoxia-related therapies, Cancer Treat. Rev. 29
[193] S.F. Hussain, L.Y. Kong, J. Jordan, C. Conrad, T. Madden, I. Fokt, W. Priebe, (2003) 297–307.
A.B. Heimberger, A novel small molecule inhibitor of signal transducers and [201] J.M. Brown, W.R. Wilson, Exploiting tumour hypoxia in cancer treatment, Nat.
activators of transcription 3 reverses immune tolerance in malignant glioma Rev. Cancer 4 (2004) 437–447.
patients, Cancer Res. 67 (2007) 9630–9636. [202] W.R. Wilson, M.P. Hay, Targeting hypoxia in cancer therapy, Nat. Rev. Cancer 11
[194] E. Garaci, F. Pica, A. Serafino, E. Balestrieri, C. Matteucci, G. Moroni, (2011) 393–410.

106

Potrebbero piacerti anche