Sei sulla pagina 1di 8

Daratumumab, Elotuzumab, and the

Development of Therapeutic Monoclonal


Antibodies in Multiple Myeloma
JP Laubach1, CE Paba Prada1, PG Richardson1 and DL Longo1

There has been substantial progress in clinical outcomes for patients with multiple myeloma (MM). This encouraging trend
derives in large part from the increasing number of effective therapeutic options and the ability because of this to achieve
higher quality responses to treatment. The approval of both daratumumab and of elotuzumab in combination with
lenalidomide and dexamethasone, in late 2015, was a notable achievement in the field, as daratumumab and elotuzumab
represent the first monoclonal antibodies available for use in MM. Given their unique mechanisms of action and favorable
side effect profiles, daratumumab and elotuzumab have considerable potential as therapeutic partners with agents in
other drug classes and in different clinical settings ranging from newly diagnosed to relapsed disease. This review
discusses the development of daratumumab and elotuzumab as well as other monoclonal antibodies currently being
evaluated for use in MM.

Multiple myeloma (MM) is a hematologic malignancy character- malignancies610 as well as the impact of mAbs in solid malignan-
ized by the presence of clonal plasma cells within the bone cies11,12 has been associated with improvement in clinical out-
marrow and clinical manifestations of lytic bone lesions, hyper- comes and favorable toxicity proles.
calcemia, renal impairment, and anemia.1,2 MM is the second This review includes discussion of the current management
most common hematologic malignancy and it is estimated that algorithm for MM, the mechanisms of action of daratumumab
in 2016 30,330 individuals in the United States were diagnosed and elotuzumab, the clinical trial development of these agents
with the disorder.3 that led to the FDA approval, as well as the manner in which
Long-term patient outcomes have continued to improve over these two drugs are optimally used within the overall MM treat-
the past three decades because of advances in MM manage- ment landscape. There is also discussion of new, emerging mAbs
ment.4,5 Nonetheless, relapsed MM in particular remains an currently being evaluated in MM in clinical trials.
important area of unmet medical need as nearly all patients with
MM ultimately relapse and require subsequent lines of therapy The current approach to multiple myeloma management
before eventually dying of the disease. Patients with newly diagnosed MM are typically treated with two
Reecting the rapid progress of MM drug development in or three-drug induction regimens incorporating an immunomo-
recent years, six regimens received approval from the US Food dulary drug (IMID) and/or proteasome inhibitor in combination
and Drug Administration (FDA) in 2015. Among these were with a glucocorticoid and, in some instances, an alkylating agent.
daratumumab monotherapy and elotuzumab in combination Clinical trial data suggest that better long-term outcomes can be
with lenalidomide and dexamethasone, which represent the rst achieved with three rather than two-drug combinations, so, when
regimens incorporating monoclonal antibodies (mAbs) approved feasible, this approach is preferred.13,14
in MM. Patients eligible for high-dose therapy and autologous stem cell
There has been strong scientic and clinical impetus behind transplantation (ASCT) generally receive three to six cycles of
the development of mAbs in MM, as advances in the laboratory induction therapy before undergoing stem cell mobilization.
have broadened the understanding of MM biology and led These patients then have the option to proceed immediately with
to identication of an array of therapeutic targets. Moreover, ASCT vs. store cells and defer ASCT to a later point in their
the use of mAbs, such as rituximab, in other hematologic management. There are data to suggest that either one or two
1
Dana-Farber Cancer Institute, Boston, Massachusetts, USA. Correspondence: JP Laubach (JacobP_Laubach@DFCI.harvard.edu)
Received 7 October 2016; accepted 31 October 2016; advance online publication 2 November 2016. doi:10.1002/cpt.550

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 101 NUMBER 1 | JANUARY 2017 81


rounds of high-dose melphalan and ASCT following induction induces cytoxicity against SLAMF7-expressing MM cells through
therapy with lenalidomide and dexamethasone confers benet activation of SLAMF7-expressing natural killer cells.34 Of note,
with respect to progression-free and overall survival.15,16 There natural killer cell activation in this context has been shown to be
are preliminary data from the ongoing IFM/DFCI 2009 trial as dependent on the signaling molecule EAT-2, which is present in
well as the EMN02/HO95 MM trial to suggest that early trans- natural killer cells but not found in MM cells.
plant is associated with a signicant improvement in progression-
free survival but no difference in overall survival.17,18 Patients CLINICAL DEVELOPMENT OF DARATUMUMAB AND
who opt against early transplant receive additional cycles of ELOTUZUMAB
induction therapy with the aim of attaining a deep response to Daratumumab
therapy. Clinical development of daratumumab began with a phase I
Accumulating evidence suggests that, in the aftermath of dose-escalation and dose-expansion study.35 Thirty-two patients
induction therapy with or without transplant, long-term mainte- in the dose escalation portion had received a median of 5.5 lines
nance treatment with lenalidomide until progression enhances of prior therapy, and 75% were refractory to lenalidomide and
long-term outcome.1922 There is also evidence that in high-risk bortezomib. Daratumumab was administered weekly for 8 weeks
patients dened by cytogenetic ndings, such as deletion of 17p at doses ranging from 0.00524 mg/kg. Dose-limiting toxicities
or 1p, translocation t(4;14) or t(14;16), post-ASCT consolida- included an episode of grade 3 anemia at 0.1 mg/kg and an epi-
tion and maintenance with lenalidomide, bortezomib, and dexa- sode of grade 3 Aspartate Transaminase increase at 1 mg/kg. A
methasone is feasible without excessive toxicity.23 maximum tolerated dose (MTD) was not reached. In the dose
At the time of disease progression, various factors are consid- expansion portion of the study, 72 patients with a median of
ered in decisions regarding the next line of therapy.24 Combina- four prior lines of therapy received either 8 mg/kg or 16 mg/kg.
tions using the IMIDs, proteasome inhibitors, alkylating agents, The overall response rate (ORR) was higher (36% vs. 10%) and
and glucocorticoids have long been mainstays of treatment for the median progression-free survival was longer (5.6 vs. 2.4
relapsed and refractory MM. Liposomal doxorubicin in combina- months) in patients treated at 16 mg/kg than at 8 mg/kg. High-
tion with bortezomib has also been an option in this setting25 grade toxicities were infrequent and included neutropenia,
and panobinostat (a histone deacetylase inhibitor) plus bortezo- thrombocytopenia, anemia, pneumonia, and hyperglycemia. Infu-
mib and dexamethasone can now be considered as well following sion reactions were common, occurring in 71% of patients in the
the FDA approval in 2015.26 The FDA label for carlzomib was dose expansion group, most of which were grade 1 or 2. In terms
expanded in 2015 to include carlzomib plus lenalidomide and of pharmacokinetic characteristics, elimination of daratumumab
dexamethasone based on results of the ASPIRE trial,27 whereas was rapid at doses of 2 mg/kg or lower and decreased at higher
the combination of ixazomib (an orally bioavailable proteasome doses. In the dose expansion phase at the 16 mg/kg dose, the
inhibitor), lenalidomide, and dexamethasone represents the rst mean half-life of daratumumab was 9.0 days after the rst dose
all-oral IMID plus proteasome inhibitor combination.28 and 10.6 days after subsequent doses.
The approvals of daratumumab and elotuzumab in combina- In the phase II SIRIUS trial, 106 patients with a median of
tion with lenalidomide and dexamethasone have further expand- ve prior lines of therapy, all of whom were refractory to both a
ed options for relapsed and refractory MM. proteasome inhibitor and an IMID, received daratumumab
16 mg/kg once weekly for 8 weeks, every other week for 16 weeks,
Mechanism of action of daratumumab and elotuzumab in and every 4 weeks thereafter.36 The response rate among this
multiple myeloma group was 29%, with 34% of patients experiencing clinical benet
Daratumumab is a humanized immunoglobulin G1K anti-CD38 (minimal response or better). The median progression-free sur-
mAb that exerts antitumor activity through antibody-dependent vival was 3.7 months and 12-month overall survival was 64.8%.
cell-mediated cytotoxicity and complement-dependent cytotoxic- The most common grade 3/4 toxicities were anemia, thrombocy-
ity29 as well as antibody-dependent cellular phagocytosis (see topenia, and neutropenia, which occurred in 24%, 19%, and 12%
Figure 1).30 More recently, it has been shown that, in addition of patients, respectively. No other grade 3/4 toxicities occurred in
to these mechanisms, daratumumab decreases a subpopulation of more than 3% of patients. Infusion reactions were noted in 42%
regulatory T cells that express CD38 and in concert increases help- of patients, 5% of which were grade 3 and the remainder grade
er and cytotoxic T-cell populations, suggesting that the agent pos- 1 or 2.
sesses an additional immunotherapeutic effect on tumor cells.31 On the basis of data from the phase I and II clinical trials dem-
Elotuzumab is a humanized immunoglobulin G1K mAb that onstrating the safety and efcacy of daratumumab, the agent
targets signaling lymphocytic activation molecule family member received accelerated approval from the FDA in November 2015
7 (SLAMF7), a cell surface glycoprotein that is expressed at a for patients who have received at least three prior lines of therapy,
high level on both normal and tumor plasma cells, and at a lower including a proteasome inhibitor and IMID, or who are refracto-
level on other lymphocyte subsets, including natural killer, ry to both a proteasome inhibitor and an IMID.
CD81 T cells, and dendritic cells.32 Elotuzumabs antitumor Regimens incorporating daratumumab in combination with
effects derive from its ability to inhibit binding of MM cells standard agents, such as lenalidomide and bortezomib, have also
to bone marrow stromal cells and induce antibody-dependent yielded promising results. In a phase I/II study evaluating daratu-
cell-mediated cytotoxicity (Figure 2).33 In addition, elotuzumab mumab plus lenalidomide and dexamethasone in relapsed and/or

82 VOLUME 101 NUMBER 1 | JANUARY 2017 | www.wileyonlinelibrary/cpt


Figure 1 Daratumumab induces death of the myeloma cell through multiple mechanisms, including complement dependent cytotoxicity (CDC), antibody-
dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), apoptosis stimulated by antibody cross-linking, and modu-
lation of enzymatic activity. cADPR, cyclic ADP ribose; MAC, membrane attack complex; NAD, nicotinamide adenine dinucleotide.

refractory MM, for example, no dose-limiting toxicities were not- This study was followed by the phase III POLLUX study eval-
ed at doses of daratumumab ranging from 216 mg/kg and the uating lenalidomide and dexamethasone with or without daratu-
16 mg/kg dose was selected for further study in combination mumab in patients with MM who received a least one prior line
with standard lenalidomide and dexamethasone.37 In the second of therapy.38 In this study, 569 patients who had received one or
phase of the study, 32 patients with a median of two prior lines more prior lines of therapy received lenalidomide plus dexameth-
of therapy had an ORR of 81%, including a stringent complete asone with or without daratumumab. The incorporation of
response of 25%, complete response rate of 9%, and a very good daratumumab was associated with improvement in response rate
partial response rate of 28%. (93% vs. 76%; P < 0.0001) and rate of complete response or

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 101 NUMBER 1 | JANUARY 2017 83


clearance. Moreover, saturation of SLAMF7 increased with higher
doses of elotuzumab, reaching a maximum level of saturation at
the 10 mg/kg dose. The degree of target saturation observed at
the 10 and 20 mg/kg doses was equivalent. The mean half-life of
elotuzumab at the 10 mg/kg dose was 4.6 days.
Despite the lack of response to elotuzumab monotherapy
observed in the phase I study, it was anticipated based on the
agents mechanism of action it could serve as an effective thera-
peutic partner with standard therapies in MM and further studies
were undertaken. This hypothesis was rst evaluated in two phase
I trials in relapsed and/or refractory MM, one of elotuzumab
plus lenalidomide and dexamethasone,41 and the other of elotu-
zumab plus bortezomib and dexamethasone.42 No dose-limiting
toxicities were noted in either trial up to the maximum planned
elotuzumab dose of 20 mg/kg and the MTD was not reached.
The response rate with elotuzumab plus lenalidomide and
Figure 2 Elotuzumab induces death of the myeloma cell through inhibi-
tion tumor cell binding to bone marrow stromal cells, antibody dependent dexamethasone was 82% among patients with median of 3 prior
cell-mediated cytotoxicity, and activation of natural killer cells expressing lines of therapy, and 48% with elotuzumab plus bortezomib and
signaling lymphocytic activation molecule family member 7 (SLAMF7). dexamethasone among patients with 2 prior lines of therapy.
A subsequent randomized phase II trial evaluating 10 and
20 mg/kg doses of elotuzumab in combination with lenalidomide
better (43% vs. 19%; P < 0.0001). The rate of minimal residual and dexamethasone showed a promising response rate of 84%
disease negativity (1 tumor cell per 105 white cells) was signicant- and median progression-free survival of 32.5 months (10 mg/kg)
ly higher among patients treated with daratumumab (22.4% vs. and 25 months (20 mg/kg).43 This was followed by a random-
4.6%; P < 0.001). The progression-free survival rate at 12 months ized, phase III ELOQUENT 2 trial of lenalidomide and dexa-
was superior in the daratumumab arm (83.2% vs. 60.1%) and there methasone with or without elotuzumab in relapsed and
was a 63% reduction in risk of disease progression or death. refractory MM.44 The incorporation of elotuzumab was associat-
Daratumumab has also proven to be an effective therapeutic ed with improvement in response rate (79% vs. 66%; P < 0.001)
partner with bortezomib, as evidenced by results from the phase as well as median progression-free survival (19.4 months vs 14.9
III CASTOR trial.39 In this study, 498 patients who had received months; hazard ratio 5 0.70; P < 0.001). Of note, fewer com-
at least one prior line of therapy received bortezomib plus dexa- plete responses were observed in patients treated with elotuzu-
methasone with or without daratumumab. The incorporation of mab, which may have been due to interference by the therapeutic
daratumumab was associated with improvement in 12-month mAb with results of electrophoresis. The rate of adverse events
progression-free survival (60.7% vs. 26.9% with median not was similar in the two groups, although fatigue, pyrexia, diarrhea,
reached vs. 7.2 months; hazard ratio 5 0.39); response rate constipation, and cough were more common in the elotuzumab
(82.9% vs. 63.2%; P < 0.001), rate of very good partial response group. There was a higher rate of serious adverse events in the
or better (59.2% vs. 29.1%; P < 0.001), and rate of complete elotuzumab group. Infusion reactions occurred in 10% of
response or better (19.2% vs. 9.0%; P 5 0.001). Grade 3/4 events patients treated with elotuzumab, most of which were grades 1/2.
were uncommon overall and similar across the two treatment Extended, 3-year follow-up of ELOQUENT 2 demonstrated
groups except for hypertension, which occurred in 6.6% of those that the clinical benet associated with the addition of elotuzu-
in the daratumumab group vs. 0.8 in the control group. All-grade mab to lenalidomide plus dexamethasone was durable, with a
diarrhea, upper respiratory infection, cough, dyspnea, and periph- 27% reduction in the risk of disease progression or death (hazard
eral edema were more common in the daratumumab group. ratio 5 0.73; P 5 0.0014). There was also a strong trend toward
benet in overall survival associated with the three-drug regimen
Elotuzumab (hazard ratio 5 0.77; P 5 0.0257).45
Clinical development of elotuzumab began with a phase I dose- Based on results from the phase III study, the FDA approved
escalation study involving 35 patients with relapsed and refractory elotuzumab plus lenalidomide and dexamethasone for patients
MM with a median of 4.5 lines of prior therapy who received elo- with relapsed and refractory MM who have received one to three
tuzumab at doses ranging from 0.5 to 20 mg/kg every other prior lines of therapy.
week.40 Elotuzumab was well tolerated overall and the MTD was
not reached up to the maximum planned dose of 20 mg/kg. There
were no responses to elotuzumab as a single agent; 26.5% of Specific clinical scenarios of importance
patients experienced stable disease. With respect to pharmacoki- There are a number of clinical scenarios that are of specic
netic characteristics, mean clearance decreased with higher doses importance to the optimal use of daratumumab and elotuzumab
of elotuzumab, consistent with saturation of target-mediated in practice.

84 VOLUME 101 NUMBER 1 | JANUARY 2017 | www.wileyonlinelibrary/cpt


Infusion reactions patients receiving daratumumab, and patients should undergo
As with other therapeutic mAbs, daratumumab and elotuzumab typing and screening before the rst dose of daratumumab.
are associated with infusion reactions, which manifest in various
ways, including headache, hypertension, hypotension, rhinitis, Renal dysfunction
wheezing, cough, dyspnea, nausea, vomiting, and rash. They are Renal dysfunction is observed in approximately 2040% of
estimated to occur in up to 50% of patients receiving daratumu- patients with newly diagnosed MM and a signicantly higher per-
mab and 10% of those who receive elotuzumab. Infusion reac- centage of patients experience decreased renal function through-
tions typically occur with the rst or second dose of the drug and out the course of the disease.51 The etiology is typically
are most often grade 1/2 in severity but higher-grade reactions multifactorial and related to paraprotein-mediated kidney injury,
can occur. Thus, it is important to adhere to the recommended hypercalcemia, dehydration, and drug effect. The ability to safely
schedule of premedications to lessen the frequency and severity administer chemotherapeutic agents in the setting of renal
of these reactions. impairment is therefore a priority in the management of MM.
In the case of daratumumab, premedications should include a Importantly, therapy-related renal toxicity has not been
glucocorticoid, such as methylprednisolone, antihistamine, such observed in phase III trials evaluating elotuzumab44 or
as diphenhydramine, and Tylenol. Our group also routinely daratumumab.39
administers montelukast before daratumumab during the initial In a phase Ib study evaluating elotuzumab plus lenalidomide
cycles of treatment due to the relatively high rate of respiratory and dexamethasone in 26 patients with relapsed MM with a
symptoms encountered during or following the infusion, particu- range of renal function ranging from normal to severe to end-
larly among patients with a history of asthma or chronic obstruc- stage, there were no signicant differences in mean and maxi-
tive pulmonary disease. During the initial cycles of treatment, an mum elotuzumab serum concentrations or area under the curve
oral glucocorticoid is also recommended for 2 days following dar- as a function of renal impairment.52 The rate of high-grade
atumumab infusion as well to mitigate the risk of delayed infu- toxicities was equivalent among patients with normal vs. impaired
sion reaction affecting respiratory function. renal function, and the response rate was similar across
In the case of elotuzumab, oral dexamethasone 28 mg is the groups. These ndings indicate elotuzumab can be utilized in
administered within 24 hours of the infusion and a dose of intra- this patient population with appropriate monitoring for
toxicities.
venous dexamethasone 8 mg is given immediately before treat-
ment. In addition, patients receive H1 and H2 blocking Other monoclonal antibodies currently in development
antihistamines as well as Tylenol as premedications. As shown in Table 1, numerous novel mAbs are currently in
development in MM.
Laboratory assay interference
As humanized immunoglobulin GK mAbs, both daratumumab Additional anti-CD38 monoclonal antibodies. In addition to daratu-
and elotuzumab are detected on standard serum protein electro- mumab, two other CD38-targeting mAbs are currently being
phoresis and immunoxation, and, thus, can affect interpretation developed, including SAR650984 and MOR202. SAR650984
of this assay. This phenomenon of interference has potential demonstrated signicant single agent activity in a phase I trial
implications with respect to accurate classication of disease sta- involving 35 patients with relapsed and/or refractory MM who
tus according to the International Myeloma Working Group cri- had received a median of 6 prior lines of therapy.53 The MTD
teria.46 Thus, it is important to notify the laboratory performing was not reached, and the agent resulted in a response rate of
the serum protein electrophoresis procedures that a particular 24%, including 6% complete and 18% partial responses. At doses
patient is receiving mAb therapy. of >10 mg/kg, the rates of overall, complete, and partial response
A daratumumab-specic immunoxation electrophoresis reex were 33%, 11%, and 22%, respectively. Grades 3/4 pneumonia
assay has been developed to allow for differentiation between dar- occurred in three patients.
atumumab and M-protein, which may be available for commer- In a phase Ib study involving heavily pretreated patients with
cial use in the future.47 There is no such interference assay for relapsed and/or refractory MM, SAR650984 at doses of 10 or
elotuzumab at present. 20 mg/kg in combination with standard dose/schedule lenalido-
It is important to note that in contrast to the effects on serum mide, the MTD once again was not reached and the combination
protein electrophoresis/immunoxation, daratumumab does not yielded a response rate of 50%.54 Twenty-ve percent of patients
interfere with results of the serum-free light chain assay.48 treated at 10 mg/kg and 20% of those treated at 20 mg/kg
achieved a very good partial response, whereas 25% of patients
Blood typing interference by daratumumab treated at 10 mg/kg and 30% of those treated at 20 mg/kg
CD38 is expressed on human red blood cells.49 Daratumumab achieved partial response.
interferes with blood typing by binding to CD38 on the surface There is an ongoing study of SAR650984 in combination with
of reagent blood cells and leads to a positive indirect Coombs pomalidomide and dexamethasone as well (NCT02283775).
test.50 Treatment of reagent red cells with dithiothreitol, which MOR202 has been evaluated as monotherapy and in combi-
denatures cell surface CD38, represents one method to potential- nation with pomalidomide or lenalidomide in an ongoing
ly circumvent this issue. Blood blanks should be notied about phase I/IIa study (NCT01421186).55 Preliminary data indicate

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 101 NUMBER 1 | JANUARY 2017 85


Table 1 Antibodies being evaluated in myeloma
Antibody Target Phase
Isatuximab (SAR650984) Anti-CD38 III
MOR202 Anti-CD38 I/IIa
Milatuzumab Anti-CD74 I/II
Indatuximab ravtansine (drug conjugate) Anti-CD138 I/II
Tabalumab Anti-BAFF (B-cell activating factor) II
Siltuximab Anti-IL6 II
Lucatumumab Anti-CD40 I
Dacetumumab Anti-CD40 I
BHQ880 Anti-DKK1 II
Sotatercept (RAP-011) Activin receptor ligand trap IIa
huN901-DM1 (drug conjugate) Anti-CD56 I
Pembrolizumab Anti-PD1 II/III
Nivolumab Anti-PD1 II/III
Atezolizumab Anti-CD274 (PD-L1) I
IL6, interleukin 6; PD1, programmed cell death 1; PD-L1, programmed cell death ligand 1.

that MOR202 monotherapy and in combination with either Indatuximab ravtansine. Indatuximab ravtansine is an mAb con-
pomalidomide or lenalidomide is safe and generally well tolerat- jugated to the maytansinoid DM4 toxin that targets syndecan 1
ed. An MTD has not been reached in the study to date. The (CD138) expressed on plasma cells, and in preclinical studies
infusion duration for MOR202 at 2 h is relatively brief in com- exerted antimyeloma activity by inhibiting tublin polymerization
parison to daratumumab and the rate of infusion reactions is and inducing cell cycle arrest.60 The compound has minimal
low. Responses were observed in 33% of patients. activity as a single agent61 yet there is interest in assessing its
potential in combination therapy. In an ongoing phase I/II study
Siltuximab. Interleukin-6 is an attractive therapeutic target for in relapsed and/or refractory MM, the agent is being adminis-
MM as it is known to be involved in the pathogenesis of tered in combination with lenalidomide and dexamethasone and
MM.56,57 The anti-interleukin-6 mAb siltuximab was evaluated in a preliminary analysis produced an ORR of 78%.62 There were
in a randomized phase II study of bortezomib plus melphalan two dose-limiting toxicities at a dose of 120 mg/m2 mucosal
and prednisone with or without the antibody.58 The addition inammation and anemia.
of siltuximab led to a trend toward improvement in overall
response (88% vs. 80%) and a signicant improvement in Monoclonal antibodies targeting programmed cell death
the rate of very good partial response or better (71% vs. 51%; 1/programmed cell death ligand 1
P 5 0.0382). However, there was no difference in progression- There is considerable interest in therapeutic strategies targeting
free survival across the two arms of the study. Rates of high- the programmed death 1 (PD-1) receptor and its ligand pro-
grade adverse events were similar overall, although patients grammed cell death ligand 1 in MM. In vitro studies have shown
treated with siltuximab had a higher incidence of neutropenia that blockade of PD-1/programmed cell death ligand 1 inhibits
and thrombocytopenia. growth of tumor cells, providing strong rationale for this
Siltuximab was partnered with lenalidiomide, bortezomib, approach and numerous clinical trials evaluating this hypothesis
and dexamethasone in a phase I trial in patients with newly are underway.63,64 Examples of this include a phase I trial evaluat-
diagnosed MM.59 The MTD was reached at the rst dose level ing the combination of lenalidomide and dexamethasone plus the
of siltuximab at 8.3 mg/kg after one patient experienced grade anti-PD-1 mAb pembrolizumab in relapsed MM, in which the
3 pneumonia and another patient had grade 4 thrombocytope- combination led to an ORR of 76%, including 23% who
nia. The most common high-grade adverse events were hema- achieved a very good partial response.65 Of note, 41% of patients
tologic, including neutropenia, thrombocytopenia, and were IMID-refractory and 18% were refractory to both IMID
lymphopenia. The ORR was 91%, whereas the rates of com- and proteasome inhibitor.
plete response and very good partial response were 9% and Furthermore, in an ongoing phase II study, 24 patients with
45%, respectively. relapsed MM, 75% of whom were refractory to both IMID and
A study of siltuximab in high-risk smoldering MM is ongoing proteasome inhibitor, received the anti-PD-1 mAb pembrolizu-
(NCT01484275). mab in combination with pomalidomide and dexamethasone.66

86 VOLUME 101 NUMBER 1 | JANUARY 2017 | www.wileyonlinelibrary/cpt


The ORR was 50%. The most common high-grade toxicities 12. Slamon, D.J. et al. Use of chemotherapy plus a monoclonal antibody
against HER2 for metastatic breast cancer that overexpresses HER2.
were hematologic, including neutropenia, lymphopenia, and N. Engl. J. Med. 344, 783792 (2001).
thrombocytopenia. Several patients experienced autoimmune- 13. Cavo, M. et al. Bortezomib with thalidomide plus dexamethasone
related toxicities, such as hypothyroidism, transaminitis, and compared with thalidomide plus dexamethasone as induction therapy
before, and consolidation therapy after, double autologous stem-cell
pneumonitis. transplantation in newly diagnosed multiple myeloma: a randomised
phase 3 study. Lancet 376, 20752085 (2010).
CONCLUSION 14. Durie, B. et al. Bortezomib, lenalidomide and dexamethasone vs.
The approvals of daratumumab and elotuzumab in combination lenalidomide and dexamethasone in patients (pts) with previously
untreated multiple myeloma without an intent for immediate
with lenalidomide and dexamethasone in relapsed MM in late autologous stem cell transplant (ASCT): results of the randomized
2015 represented a major advance in MM therapeutics. The phase III trial SWOG S0777. Blood 126, 25 (2015).
availability of these novel regimens further increases the number 15. Palumbo, A. et al. Autologous transplantation and maintenance
therapy in multiple myeloma. N. Engl. J. Med. 371, 895905 (2014).
of treatment options available for relapsed disease. Moreover, the 16. Gay, F. et al. Chemotherapy plus lenalidomide versus autologous
unique antitumor mechanisms as well as the favorable toxicity transplantation, followed by lenalidomide plus prednisone versus
prole of both agents make them ideal partners in multi-agent lenalidomide maintenance, in patients with multiple myeloma: a
randomised, multicentre, phase 3 trial. Lancet Oncol. 16, 1617
chemotherapy regimens that have the potential to produce deeper 1629 (2015).
and more durable responses for patients with both newly diag- 17. Attal, M. et al. Autologous transplantation for multiple myeloma in the
era of new drugs: a phase III study of the Intergroupe Francophone Du
nosed and relapsed disease. Ongoing and future studies of these Myelome (IFM/DFCI 2009 Trial). Blood 126, 391 (2015).
agents will dene which combinations are most effective at differ- 18. Cavo, M. et al. Upfront autologous stem cell transplantation (ASCT)
ent stages of the disease, as well as the potential role for daratu- versus novel agent-based therapy for multiple myeloma (MM): a
randomized phase 3 study of the European Myeloma Network
mumab and elotuzumab in the context of maintenance therapy. (EMN02/HO95 MM trial). J. Clin. Oncol. 34(suppl.), abstract 8000
Other mAbs, particularly those targeting the immune check- (2016).
points PD-1 and programmed cell death ligand 1 are likewise 19. Attal, M. et al. Lenalidomide maintenance after stem-cell
transplantation for multiple myeloma. N. Engl. J. Med. 366, 1782
associated with considerable promise and results of ongoing stud- 1791 (2012).
ies evaluating these agents are awaited with interest. 20. McCarthy, P.L. et al. Lenalidomide after stem-cell transplantation for
multiple myeloma. N. Engl. J. Med. 366, 17701781 (2012).
21. Palumbo, A. et al. Continuous lenalidomide treatment for newly
diagnosed multiple myeloma. N. Engl. J. Med. 366, 17591769
ACKNOWLEDGMENTS (2012).
The authors thank Megan Hiserodt and Michelle Maglio for their editorial 22. Benboubker, L. et al. Lenalidomide and dexamethasone in transplant-
assistance with the manuscript. ineligible patients with myeloma. N. Engl. J. Med. 371, 906917
(2014).
CONFLICT OF INTEREST 23. Nooka, A.K. et al. Consolidation and maintenance therapy with
The authors declared no conflict of interest. lenalidomide, bortezomib and dexamethasone (RVD) in high-risk
myeloma patients. Leukemia 28, 690693 (2014).
24. Laubach, J. et al. Management of relapsed multiple myeloma:
C 2016 American Society for Clinical Pharmacology and Therapeutics
V recommendations of the International Myeloma Working Group.
Leukemia 30, 10051017 (2016).
25. Orlowski, R.Z. et al. Randomized phase III study of pegylated
1. Kyle, R.A. & Rajkumar, S.V. Multiple myeloma. Blood 111, 2962 liposomal doxorubicin plus bortezomib compared with bortezomib
2972 (2008). alone in relapsed or refractory multiple myeloma: combination
2. Palumbo, A. & Anderson, K. Multiple myeloma. N. Engl. J. Med. 364, therapy improves time to progression. J. Clin. Oncol. 25, 38923901
10461060 (2011). (2007).
3. Siegel, R.L., Miller, K.D. & Jemal, A. Cancer statistics, 2016. CA 26. San-Miguel, J.F. et al. Panobinostat plus bortezomib and
Cancer J. Clin. 66, 730 (2016). dexamethasone versus placebo plus bortezomib and dexamethasone
4. Kumar, S.K. et al. Improved survival in multiple myeloma and the in patients with relapsed or relapsed and refractory multiple
impact of novel therapies. Blood 111, 25162520 (2008). myeloma: a multicentre, randomised, double-blind phase 3 trial.
5. Robinson, D., Kaura, S., Kiely, D., Hussein, M.A., Nersesyan, K. & Lancet Oncol. 15, 11951206 (2014).
Durie, B.G.M. Impact of novel treatments on multiple myeloma 27. Stewart, A.K. et al. Carfilzomib, lenalidomide, and dexamethasone for
survival. Blood 124, 5676 (2014). relapsed multiple myeloma. N. Engl. J. Med. 372, 142152 (2015).
6. Maloney, D.G. et al. IDEC-C2B8 (Rituximab) anti-CD20 monoclonal 28. Moreau, P. et al. Oral ixazomib, lenalidomide, and dexamethasone for
antibody therapy in patients with relapsed low-grade non-Hodgkins multiple myeloma. N. Engl. J. Med. 374, 16211634 (2016).
lymphoma. Blood 90, 21882195 (1997). 29. de Weers, M. et al. Daratumumab, a novel therapeutic human CD38
7. McLaughlin, P. et al. Rituximab chimeric anti-CD20 monoclonal antibody monoclonal antibody, induces killing of multiple myeloma and other
therapy for relapsed indolent lymphoma: half of patients respond to a hematological tumors. J. Immunol. 186, 18401848 (2011).
four-dose treatment program. J. Clin. Oncol. 16, 28252833 (1998). 30. Overdijk, M.B. et al. Crosstalk between human IgG isotypes and
8. Coiffier, B. et al. CHOP chemotherapy plus rituximab compared with murine effector cells. J. Immunol. 189, 34303438 (2012).
CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N. 31. Krejcik, J. et al. Daratumumab depletes CD381 immune regulatory
Engl. J. Med. 346, 235242 (2002). cells, promotes T-cell expansion, and skews T-cell repertoire in
9. Habermann, T.M. et al. Rituximab-CHOP versus CHOP alone or with multiple myeloma. Blood 128, 384394 (2016).
maintenance rituximab in older patients with diffuse large B-cell 32. Hsi, E.D. et al. CS1, a potential new therapeutic antibody target for
lymphoma. J. Clin. Oncol. 24, 31213127 (2006). the treatment of multiple myeloma. Clin. Cancer Res. 14, 27752784
10. Maury, S. et al. Rituximab in B-lineage adult acute lymphoblastic (2008).
leukemia. N. Engl. J. Med. 375, 10441053 (2016). 33. Tai, Y.T. et al. Anti-CS1 humanized monoclonal antibody HuLuc63
11. Brahmer, J.R. et al. Safety and activity of anti-PD-L1 antibody in inhibits myeloma cell adhesion and induces antibody-dependent
patients with advanced cancer. N. Engl. J. Med. 366, 24552465 cellular cytotoxicity in the bone marrow milieu. Blood 112, 1329
(2012). 1337 (2008).

CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 101 NUMBER 1 | JANUARY 2017 87


34. Collins, S.M. et al. Elotuzumab directly enhances NK cell cytotoxicity 52. Berdeja, J. et al. Pharmacokinetics and safety of elotuzumab
against myeloma via CS1 ligation: evidence for augmented NK cell combined with lenalidomide and dexamethasone in patients with
function complementing ADCC. Cancer Immunol. Immunother. 62, multiple myeloma and various levels of renal impairment: results of a
18411849 (2013). phase Ib study. Clin. Lymphoma Myeloma Leuk. 16, 129138
35. Lokhorst, H.M. et al. Targeting CD38 with daratumumab monotherapy (2016).
in multiple myeloma. N. Engl. J. Med. 373, 12071219 (2015). 53. Martin, T.G., Hsu, K., Strickland, S.A., Glenn, M.J., Mikhael, J. &
36. Lonial, S. et al. Daratumumab monotherapy in patients with Charpentier, E. A phase I trial of SAR650984, a CD38 monoclonal
treatment-refractory multiple myeloma (SIRIUS): an open-label, antibody, in relapsed or refractory multiple myeloma. J. Clin. Oncol. 32
randomised, phase 2 trial. Lancet 387, 15511560 (2016). (suppl.), abstract 8532 (2014).
37. Plesner, T. et al. Phase 1/2 study of daratumumab, lenalidomide, 54. Martin, T.G. III et al. A phase Ib dose-escalation trial of SAR650984
and dexamethasone for relapsed multiple myeloma. Blood 128, (anti-CD-38 mAb) in combination with lenalidomide and
18211828 (2016). dexamethasone in relapsed/refractory multiple myeloma. J. Clin.
38. Dimopoulos, M.A. et al. Daratumumab, lenalidomide, and Oncol. 32 Suppl, abstract 8512 (2014).
dexamethasone for multiple myeloma. N. Engl. J. Med. 375, 1319 55. Raab, M.S. et al. MOR202 alone and in combination with
1331 (2016). pomalidomide or lenalidomide in relapsed or refractory multiple
39. Palumbo, A. et al. Daratumumab, bortezomib, and dexamethasone myeloma: data from clinically relevant cohorts from a phase I/IIa
for multiple myeloma. N. Engl. J. Med. 375, 754766 (2016). study. J. Clin. Oncol. 34 (suppl.), abstract 8012 (2016).
40. Zonder, J.A. et al. A phase 1, multicenter, open-label, dose escalation 56. Chauhan, D. et al. Multiple myeloma cell adhesion-induced
study of elotuzumab in patients with advanced multiple myeloma. interleukin-6 expression in bone marrow stromal cells involves
Blood 120, 552559 (2012). activation of NF-kappa B. Blood 87, 11041112 (1996).
41. Lonial, S. et al. Elotuzumab in combination with lenalidomide and low- 57. Rawstron, A.C. et al. The interleukin-6 receptor alpha-chain (CD126)
dose dexamethasone in relapsed or refractory multiple myeloma. J. is expressed by neoplastic but not normal plasma cells. Blood 96,
Clin. Oncol. 30, 19531959 (2012). 38803886 (2000).
42. Jakubowiak, A.J. et al. Phase I trial of anti-CS1 monoclonal antibody 58. San-Miguel, J. et al. Phase 2 randomized study of bortezomib-
elotuzumab in combination with bortezomib in the treatment of melphalan-prednisone with or without siltuximab (anti-IL-6) in multiple
relapsed/refractory multiple myeloma. J. Clin. Oncol. 30, 19601965 myeloma. Blood 123, 41364142 (2014).
(2012). 59. Shah, J.J. et al. Siltuximab (CNTO 328) with lenalidomide, bortezomib
43. Richardson, P.G. et al. Elotuzumab in combination with lenalidomide and dexamethasone in newly-diagnosed, previously untreated
and dexamethasone in patients with relapsed multiple myeloma: final multiple myeloma: an open-label phase I trial. Blood Cancer J. 6,
phase 2 results from the randomised, open-label, phase 1b-2 dose- e396 (2016).
escalation study. Lancet Haematol. 2, e516e527 (2015). 60. Ikeda, H. et al. The monoclonal antibody nBT062 conjugated to
44. Lonial, S. et al. Elotuzumab therapy for relapsed or refractory multiple cytotoxic maytansinoids has selective cytotoxicity against CD138-
myeloma. N. Engl. J. Med. 373, 621631 (2015). positive multiple myeloma cells in vitro and in vivo. Clin. Cancer Res.
45. Dimopoulos, M.A. et al. Eloquent-2 update: a phase 3, randomized, 15, 40284037 (2009).
open-label study of elotuzumab in combination with lenalidomide/ 61. Heffner, L.T. et al. BT062, an antibody-drug conjugate directed
dexamethasone in patients with relapsed/refractory multiple against CD138, given weekly for 3 weeks in each 4 week cycle:
myeloma 3-year safety and efficacy follow-up. Blood 126, abstract safety and further evidence of clinical activity. Blood 120, 4042
28 (2015). (2012).
46. van de Donk, N.W. et al. Interference of daratumumab in monitoring 62. Kelly, K.R. et al. Indatuximab ravtansine (BT062) in combination with
multiple myeloma patients using serum immunofixation lenalidomide and low-dose dexamethasone in patients with relapsed
electrophoresis can be abrogated using the daratumumab IFE reflex and/or refractory multiple myeloma: clinical activity in patients
assay (DIRA). Clin. Chem. Lab. Med. 54, 11051109 (2016). already exposed to lenalidomide and bortezomib. Blood 124, 4736
47. McCudden, C. et al. Monitoring multiple myeloma patients treated (2014).
with daratumumab: teasing out monoclonal antibody interference. 63. Gorgun, G. et al. Lenalidomide enhances immune checkpoint
Clin. Chem. Lab. Med. 54, 10951104 (2016). blockade-induced immune response in multiple myeloma. Clin.
48. Rosenberg, A.S., Bainbridge, S., Pahwa, R. & Jialal, I. Investigation Cancer Res. 21, 46074618 (2015).
into the interference of the monoclonal antibody daratumumab on the 64. Ray, A. et al. Targeting PD1-PDL1 immune checkpoint in
free light chain assay. Clin. Biochem. 49, 12021204 (2016). plasmacytoid dendritic cell interactions with T cells, natural killer
49. Zocchi, E. et al. A single protein immunologically identified as CD38 cells and multiple myeloma cells. Leukemia 29, 14411444 (2015).
displays NAD1 glycohydrolase, ADP-ribosyl cyclase and cyclic ADP- 65. Mateos, M-V. et al. Pembrolizumab in combination with lenalidomide
ribose hydrolase activities at the outer surface of human erythrocytes. and low-dose dexamethasone for relapsed/refractory multiple
Biochem. Biophys. Res. Commun. 196, 14591465 (1993). myeloma (RRMM): final efficacy and safety analysis. J. Clin. Oncol. 34
50. Chapuy, C.I. et al. Resolving the daratumumab interference with blood (suppl.), abstract 8010 (2016).
compatibility testing. Transfusion 55(6 Pt 2), 15451554 (2015). 66. Badros, A.Z. et al. A phase II study of anti PD-1 antibody
51. Gonsalves, W.I. et al. Improvement in renal function and its impact on pembrolizumab, pomalidomide and dexamethasone in patients with
survival in patients with newly diagnosed multiple myeloma. Blood relapsed/refractory multiple myeloma (RRMM). Blood 126, 506
Cancer J. 5, e296 (2015). (2015).

88 VOLUME 101 NUMBER 1 | JANUARY 2017 | www.wileyonlinelibrary/cpt

Potrebbero piacerti anche