Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

28
REVIEW ARTICLE Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma Alessandro Allegra, Giuseppa Penna, Andrea Alonci, Sabina Russo, Bruna Greve, Vanessa Innao, Viviana Minardi, Caterina Musolino Division of Haematology, University of Messina, Messina, Italy Abstract Despite recent treatments, such as bortezomib, thalidomide, and lenalidomide, therapy of multiple myeloma (MM) is limited, and MM remains an incurable disease associated with high mortality. The outcome of patients treated with cytotoxic therapy has not been satisfactory. Therefore, new therapies are needed for relapsed MM. A new anticancer strategy is the use of monoclonal antibodies (MoAbs) that represent the best available combination of tumor cytotoxicity, environmental signal privation, and immune system redirection. Clinical results in patients with relapsed/refractory MM suggest that MoAbs are likely to operate synergistically with traditional therapies (dexamethasone), immune modulators (thalidomide, lenalidomide), and other novel therapies (bortezomib); in addition, MoAbs have shown the ability to overcome resistance to these therapies. It remains to be defined how MoAb therapy can most fruitfully be incorporated into the current therapeutic paradigms that have achieved significant survival earnings in patients with MM. This will require careful consideration of the optimal sequence of treatments and their clinical position as either short-term induction therapy, frontline therapy in patients ineligible for ASCT, or long-term maintenance treatment. Key words multiple myeloma; monoclonal antibodies; therapy; microenvironmental; tumor cytotoxicity; immune system Correspondence Prof. Caterina Musolino, Division of Haematology, University of Messina, Via Consolare Valeria 98124 Messina, Italy. Tel: +39 090 221 2364; Fax: +39 090 2212355; e-mail: [email protected] Accepted for publication 13 March 2013 doi:10.1111/ejh.12107 General considerations on monoclonal antibodies and multiple myeloma Multiple myeloma (MM) is a progressive and fatal disease characterized by the malignant proliferation of plasma cells in the bone marrow and overproduction of monoclonal immunoglobulin or light-chain proteins. MM therapy remains challenging. Despite recent therapies, such as bortezomib, thalidomide, and lenalidomide, treatment is limited and palli- ative, and MM remains an incurable disease associated with a median survival of 4 yr (1). Patients refractory to both bortezomib and either lenalido- mide or thalidomide were found to have a median overall sur- vival of 9 months (2). Then, the outcome of patients with MM treated with cytotoxic therapy has not been adequate, and new treatments are needed for relapsed or refractory MM. A new anticancer strategy is the use of monoclonal anti- bodies (MoAb) that operate through completely different mechanisms of action. MoAbs directed against MM- associated markers should be taken into account in clinical practice, because they could represent the best available combination of tumor cytotoxicity, environmental signal pri- vation, and immune system redirection (3). Ideally, targets for therapeutic MoAbs should be speci- cally expressed on cancerous cells but not on normal cells. In fact, monoclonal antibodies are an interesting thera-peutic option in MM because they are specic to a tumor-associated target and have been successfully employed in the treatment of patients with other hematologic diseases (4). Moreover, given that the mechanisms of cytotoxicity by MoAb therapy are quite different from those of chemotherapeutic drugs, MoAb therapy can work synergistically with chemotherapy. In MM, MoAb can be directed against a large variety of anti- gen targets, which can be expressed either on myeloma cells or on components of the bone marrow microenvironment [bone marrow stromal cells (BMSCs) or signaling molecules] (3, 5). © 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 441 European Journal of Haematology 90 (441–468)

Transcript of Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

Page 1: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

REVIEW ARTICLE

Monoclonal antibodies: potential new therapeutic treatmentagainst multiple myelomaAlessandro Allegra, Giuseppa Penna, Andrea Alonci, Sabina Russo, Bruna Greve, Vanessa Innao,Viviana Minardi, Caterina Musolino

Division of Haematology, University of Messina, Messina, Italy

Abstract

Despite recent treatments, such as bortezomib, thalidomide, and lenalidomide, therapy of multiple

myeloma (MM) is limited, and MM remains an incurable disease associated with high mortality. The

outcome of patients treated with cytotoxic therapy has not been satisfactory. Therefore, new therapies

are needed for relapsed MM. A new anticancer strategy is the use of monoclonal antibodies (MoAbs) that

represent the best available combination of tumor cytotoxicity, environmental signal privation, and immune

system redirection. Clinical results in patients with relapsed/refractory MM suggest that MoAbs are likely

to operate synergistically with traditional therapies (dexamethasone), immune modulators (thalidomide,

lenalidomide), and other novel therapies (bortezomib); in addition, MoAbs have shown the ability to

overcome resistance to these therapies. It remains to be defined how MoAb therapy can most fruitfully

be incorporated into the current therapeutic paradigms that have achieved significant survival earnings in

patients with MM. This will require careful consideration of the optimal sequence of treatments and their

clinical position as either short-term induction therapy, frontline therapy in patients ineligible for ASCT, or

long-term maintenance treatment.

Key words multiple myeloma; monoclonal antibodies; therapy; microenvironmental; tumor cytotoxicity; immune system

Correspondence Prof. Caterina Musolino, Division of Haematology, University of Messina, Via Consolare Valeria 98124 Messina,

Italy. Tel: +39 090 221 2364; Fax: +39 090 2212355; e-mail: [email protected]

Accepted for publication 13 March 2013 doi:10.1111/ejh.12107

General considerations on monoclonal

antibodies and multiple myeloma

Multiple myeloma (MM) is a progressive and fatal diseasecharacterized by the malignant proliferation of plasma cellsin the bone marrow and overproduction of monoclonalimmunoglobulin or light-chain proteins. MM therapy remainschallenging. Despite recent therapies, such as bortezomib,thalidomide, and lenalidomide, treatment is limited and palli-ative, and MM remains an incurable disease associated witha median survival of 4 yr (1).Patients refractory to both bortezomib and either lenalido-

mide or thalidomide were found to have a median overall sur-vival of 9 months (2). Then, the outcome of patients with MMtreated with cytotoxic therapy has not been adequate, and newtreatments are needed for relapsed or refractory MM.A new anticancer strategy is the use of monoclonal anti-

bodies (MoAb) that operate through completely different

mechanisms of action. MoAbs directed against MM-associated markers should be taken into account in clinicalpractice, because they could represent the best availablecombination of tumor cytotoxicity, environmental signal pri-vation, and immune system redirection (3).Ideally, targets for therapeutic MoAbs should be specifi-

cally expressed on cancerous cells but not on normal cells. Infact, monoclonal antibodies are an interesting thera-peuticoption in MM because they are specific to a tumor-associatedtarget and have been successfully employed in the treatmentof patients with other hematologic diseases (4).Moreover, given that the mechanisms of cytotoxicity by MoAb

therapy are quite different from those of chemotherapeutic drugs,MoAb therapy can work synergistically with chemotherapy.In MM, MoAb can be directed against a large variety of anti-

gen targets, which can be expressed either on myeloma cells oron components of the bone marrow microenvironment [bonemarrow stromal cells (BMSCs) or signaling molecules] (3, 5).

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 441

European Journal of Haematology 90 (441–468)

Page 2: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

Therapeutic MoAbs use one or more mechanisms toreduce tumor burden in patients. They could directly induceapoptosis or growth arrest upon binding to cell surface anti-gen on tumor cells. Rituximab and mapatumumab couldcause growth inhibition or apoptosis signaling to blocktumor cell growth and survival. Such mechanism of actionwas employed by MoAbs conjugated with toxins, that is,maytansinoids for anti-CD56 and anti-CD138, thus directlytarget and eliminate tumor cells.Most of the approved therapeutic MoAbs belong to IgG1

subclass, which has a long half-life, and activate immuneeffector functions. After the binding of MoAbs to a specifictarget on a tumor cells, antibody-dependent cellular cytotoxicity(ADCC) is triggered by interactions between the Fc region ofan antibody bound to a tumor cell and Fc receptors,particularly FcRI and FcRIII, on immune effector cells such asnatural killer (NK) cells, macrophages, and neutrophils.MoAb-coated tumor cells are phagocytosed by macrophagesor undergo cytolysis by NK cells. In the case of complement-dependent cytotoxicity (CDC), recruitment of C1q by IgGbound to the tumor cell surface is the first step. This triggers aproteolytic cascade that induces production of the effectormolecule, C3b, and then formation of a membrane attack com-plex that kills the target cell by disrupting its cell membrane.MoAbs have been additionally designed to functionally

block both autocrine- and paracrine-secreted cytokines andgrowth factors as well as molecules mediating MM–stromalcell interaction.Although expression of MoAb targets is not always con-

fined to the myeloma cells, toxicity of the MoAbs is rela-tively mild. On the other hand, minimizing side effects ofchemotherapy can also be achieved using myeloma-specificantibody conjugates to specifically deliver cytotoxic drugs tothe tumor cells. This method will consent the administrationof lower doses of drugs and may therefore reduce toxicity ofantimyeloma treatments (6, 7).A vast assortment of antigens may be targeted in MM

treatments, including those implicated in cell survival, anti-apoptotic pathways, angiogenesis, and interactions betweenMM cells and BMSCs (8).These potential targets include mediators of adhesion, sig-

naling molecules, cell surface receptors, and plasma cellgrowth factors.

MoAbs Targeting tumor cells

Several MoAbs directed against MM cell surface are beinginvestigated as potential targets in MM patients (Table 1).

Anti-CD20 MoAb

Several works have shown that MM includes clonotypic Blineage cells at stages earlier than the compartment of malig-nant plasma cells in the bone marrow (9), and the circulating

component of the MM clone includes at least two distinctCD19+ CD20+ B-cell compartments as well as CD138+

CD20+ plasma cells. Pilarski et al. evaluated them before,during, and after treatment of patients with rituximab(anti-CD20), followed by quantifying B-cell subsets over a5-month period during and after treatment. Overall, all threetypes of circulating B lineage cells persist despite treatmentwith rituximab. The inability of rituximab to prolong sur-vival in MM may result from this failure to deplete CD20+

B and plasma cells in MM (10).Indeed, previous studies demonstrated only minimal activ-

ity of anti-CD20 rituximab and antibodies against plasmacell-specific CD38 antibodies in MM (11–13). However,

Table 1 Monoclonal antibodies (MoAbs) targeting tumor cells

Target Name

CD20 Rituximab

Tositumomab

20-C2-2b Veltuzumab

CS1 Elotuzumab

CD138 B-B4

BC/B-B4

DL-101

1 D4

1.BB.210

MI15

2Q1484

5F7

104-9

281-2

nBT062-SMCL-DM1

nBT062-SPDM4

nBT062-SPP-DM1

CD38 Doratumumab

MOR202

CD40 Lucatuzumab

Lorvotuzumab

IGF-1 AVE1642

AMG479 IMCA12

R15507

Figitumumab

Dalotuzumab

HMI.24 (CD317) AHM

Defucosylated AHM

XmAb 5592

CD48 Anti-CD48 MoAb

b2 m IgG anti-b2 m

IgM anti-b2 m

CD70 SGN-70

CD74 Milatuzumab

HLADR ID09C3

2D7-DB

CD229 Anti-CD229

GM2 ganglioside BIW-8962

CD54 (ICAM-1) BI-505

Ku 5E2

442 © 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Monoclonal antibodies and multiple myeloma Allegra et al.

Page 3: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

although the CD-2 subgroup of myeloma frequently overex-presses CD20 and few studies trying to prove otherwise(14), clinical studies of rituximab treatment in MM have forthe most part been discouraging, with few patients achievingonly minimal responses (15). Results from a clinical phase 2trial in relapsed MM showed that rituximab treatmentyielded significant reduction in circulating B cells but hadno beneficial clinical effect (16).Moreover, rituximab was tested for maintenance therapy

in MM following autologous hematopoietic stem cell trans-plantation (SCT) (12). The use of rituximab in this sort ofpatients was associated with an unexpectedly high rate ofearly relapse. The authors then hypothesized a possible rolefor rituximab in provoking an additional reduction in thenormal, residual B-cell activity.MM is generally not considered as a disease adequate for

anti-CD20 therapy due to weak and various expression ofCD20 in the preponderance of subjects. In contrast, otherstudies demonstrated that the CD20+ phenotype is associatedwith patients with t(11,14)(q13;q32) and with shorter sur-vival (17) and that sporadic responses have been achieved inpatients with CD20+ myelomatous plasma cells (18, 19).The failing of rituximab is probably attributable to the

small number of MM subjects (estimated at 13–22%) whoexpress CD20 in plasma cells, but a different mechanism thatmay render MM refractory to rituximab is the possibility thatMM cells express increased levels of complement-inhibitingproteins, such as the presence of the complement regulatorCD59 on myeloma makes complement-mediated cytotoxicityinefficacious (16, 20–22). In addition, Fc-c receptor polymor-phism may limit the efficacy of ADCC as a killing mecha-nism. Finally, the administration of rituximab in MM maycause a selective loss of CD20 expression.In spite of this, recently, a different anti-CD20 antibody

was used in MM patient. I-131 tositumomab is a radiola-beled murine anti-CD20 antibody, which is highly effectivein the treatment of low-grade B-cell NHL. It was tested in asingle arm, phase 2 study. With a median follow-up of 4 yr,none of the responding patients have progressed. Then,I-131 tositumomab is well tolerated in patients with previouslytreated MM and produces objective responses including CRs(23).

IFN-20-C2-2b MoAb

Available data indicate that progression-free survival of MMpatients is improved with IFNa, but although IFNa can havedirect cytotoxic action on plasma cells, promote both innateand adaptive immunity, and inhibit angiogenesis, its effica-ciousness as an anticancer drug has been limited due to itsshort circulating half-life and systemic toxicity (24).Rossi et al. reported the generation of the first bispecific

MoAb-IFNa, designated 20-C2-2b, which comprises twocopies of IFNa2b and a stabilized F(ab)2 of hL243 (human-

ized anti-HLA-DR; IMMU-114) site specifically linked toveltuzumab (humanized anti-CD20). In vitro, 20-C2-2binhibited eight myeloma cell lines and was more effectivethan monospecific CD20-targeted MoAb-IFNa or a mixturecomprising the parental antibodies and IFNa in all but one(HLA-DR�/CD20�) myeloma line, suggesting that 20-C2-2bshould be useful in the treatment of MM. 20-C2-2b displayedgreater cytotoxicity against KMS12-BM (CD20+/HLA-DR+

myeloma) compared with monospecific MoAb-IFNa, whichtargets only HLA-DR or CD20, indicating that all three com-ponents in 20-C2-2b could contribute to toxicity (25).

Anti-CS1 MoAb

The cell surface glycoprotein CS1 (CD2 subset 1, CRACC,SLAMF7, CD31, or 19A24), a member of the signalinglymphocyte-activating molecule–related receptor family, isselectively and constantly expressed at high levels onCD138-purified plasma cells from the MM patients (>97%),independent of the presence of metaphase cytogenetic abnor-malities or molecular subgroup. In addition, low levels ofcirculating CS1 are present in myeloma patient sera. Exceptactivated B, NK, CD8+ T cells, and mature dendritic cellswith low expression levels, it is not expressed by normal tis-sues or stem cells (26–28). Moreover, it regulates NK cellcytolytic activity via recruiting EWS-activated transcript-2and activating the PI3K/PLCg signaling pathways (29, 30).Studies propose that CS1 localizes to the uropods of polar-

ized myeloma cells suggesting a possible function for CS1 inmediating adhesion of myeloma cells to bone marrow stroma.CS1 also seems to preserve myeloma cell lines from apoptosisby lowering phosphorylation of ERK1/2, AKT, and STAT aswell as regulating pro- and anti-apoptotic pathways (31).CS1 may then contribute to MM pathogenesis by increas-

ing MM cell adhesion, clonogenic growth, and tumorigenic-ity via c-Maf-mediated interactions with BMSCs (32).The humanized anti-CS1 MoAb elotuzumab exerts antimye-

loma activity in vitro via ADCC mediated by NK cells anddoes not depend on complement-mediated cytotoxicity (33).In fact, elotuzumab caused significant ADCC against MM

cells even in the presence of BMSCs. Moreover, it provokedautologous ADCC against primary MM cells resistant totraditional or new drugs including HSP90 inhibitor and bort-ezomib and, considerably, increased HuLuc63-induced MMcell lysis when pretreated with conventional or new anti-MM therapies (34).Administration of elotuzumab causes tumor regression in

myeloma xenograft mouse models (27). Furthermore, combi-nation of elotuzumab with bortezomib significantly increasesthe in vivo therapeutic efficacy to eradicate patient-derivedmyeloma cells in a SCID-hu mouse model (34).In a phase 1, multicenter, dose-escalation study of elot-

uzumab in patients with advanced MM, patients withrelapsed/refractory MM were treated with intravenous elot-

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 443

Allegra et al. Monoclonal antibodies and multiple myeloma

Page 4: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

uzumab. The most common AEs were cough, headache,back pain, fever, and chills. Adverse events were generallymild to moderate in severity. Nearly 26.5% had stable dis-ease (35).In other studies, most AEs attributable to elotuzumab

included mild-to-moderate infusion reactions, and the imple-mentation of more aggressive premedication regimensappeared to reduce the rate of infusion-related AEs (36, 37).Elotuzumab has been associated with different drugs,

some of which have been shown to increase its ADCC activ-ity in vitro. Pretreatment with dexamethasone or new drugssuch as bortezomib, lenalidomide, MEK inhibitors, or Aktinhibitors has been shown to significantly increase elot-uzumab-induced ADCC against MM cells (26, 34, 38).Preliminary results of clinical trials of HuLuc63 in combi-

nation with bortezomib or lenalidomide or dexamethasonewere reported at the ASH meeting 2009 (39, 40), suggestingthat elotuzumab may increase the activity of bortezomib andlenalidomide in treating MM with acceptable toxicity.Rhee et al. investigated whether the activity of elot-

uzumab could be increased by bortezomib. In vitro bortezo-mib pretreatment of myeloma targets significantly increasedelotuzumab-mediated antibody-dependent cell-mediated cyto-toxicity (ADCC), both for OPM2 myeloma cells using NKor peripheral blood mononuclear cells (PBMC) from controlsand for primary myeloma cells using autologous NK effectorcells. In an OPM2 myeloma xenograft model, elotuzumab incombination with bortezomib revealed significantly increasedin vivo antitumor activity (34).A phase 1 study evaluated elotuzumab, lenalidomide,

and dexamethasone in patients with relapsed or refractoryMM. The most frequent grades 3 to 4 toxicities were neu-tropenia (36%) and thrombocytopenia (21%). Objectiveresponses were obtained in 82% of treated patients. Thecombination of elotuzumab, lenalidomide, and low-dosedexamethasone was generally well tolerated and showedpromising response rates in patients with relapsed orrefractory MM (41).In phase 1/2 studies in relapsed/refractory MM, elot-

uzumab and bortezomib were administered. The most fre-quent grades 3 to 4 adverse events were lymphopenia andfatigue. Two elotuzumab-related serious AEs (chest pain andgastroenteritis) occurred in one patient. An objectiveresponse was observed in 48% of evaluable patients and in67% of three patients refractory to bortezomib. Median timeto progression was 9.46 months (42).

Anti-CD138 MoAb

CD138 (syndecan-1) is a heparan sulfate proteoglycan thatserves as a receptor for epidermal growth factor (EGF)ligands. Binding of EGF ligands stimulates cell growth (43).Moreover, the large extracellular domain of CD138 binds

via its heparin sulfate chains to other soluble extracellular

molecules, including the fibroblast growth factor and hepato-cyte growth factor, and to insoluble extracellular molecules,such as collagen and fibronectin. CD138 also mediates cell–cell adhesion through interactions with heparin-bindingmolecules. Studies of plasma cell differentiation show thatCD138 is a differentiation antigen and a coreceptor for MMgrowth factors (44).Immunohistochemical and flow cytometric analysis of

patient MM cells has shown that CD138 is expressed in alarge majority of cases. Within the hematopoietic compart-ment, CD138 expression is confined to normal plasma cells,with no expression on hematopoietic stem cells (HSC),while expression of CD138 on MM cells is significantlyhigher than on normal plasma cells (45).Almost all MM cells, even after exposure to multiple ther-

apies, express the antigen, making it a useful target at anystage of the disease. The extracellular components of CD138and the heparan sulfate side chains may be shed from thecell surface. Soluble syndecan-1 in serum serves as a prog-nostic indicator in MM (46).Different monoclonal antibodies (i.e., B-B4, BC/B-B4,

B-B2, DL-101, 1 D4, MI15, 1.BB.210, 2Q1484, 5F7, 104-9,281-2) specific to CD138 have been reported.Ikeda et al. showed the antitumor efficacy of three new

anti-CD138 antibody–maytansinoid conjugates, nBT062-SMCC-DM1, nBT062-SPDB-DM4, and nBT062-SPP-DM1,which vary in the linkage between the maytansinoid moietyand MoAb. The nBT062-SMCC-DM1 linkage contains athioether bond, which is not cleavable by disulfide exchange,whereas the nBT062-SPDB-DM4 and nBT062-SPP-DM1conjugates contain disulfide linkages, which can be cleavedby disulfide exchange, resulting in liberation of active may-tansinoid agent. The anti-CD138 antibody nBT062 is a mur-ine/human chimeric form of B-B4. The observed preclinicalantitumor activity of the nBT062–maytansinoid conjugatesprovides the framework for clinical development of theseagents to improve patient outcome in MM (47).Anti-CD138 immunoconjugates significantly inhibited

growth of MM cell lines and primary tumor cells from MMpatients without cytotoxicity against PBMC from healthycontrols. In MM cells, they induced G2–M cell cycle arrest,followed by apoptosis associated with cleavage of caspase-3,caspase-8, caspase-9, and poly(ADP-ribose) polymerase.Non-conjugated nBT062 completely blocked cytotoxicityinduced by nBT062–maytansinoid conjugate, confirming thatspecific binding is required for inducing cytotoxicity. More-over, nBT062–maytansinoid conjugates blocked adhesion ofMM cells to BMSC. The coculture of MM cells with BMSCprotects against dexamethasone-induced death but had noeffect on the cytotoxicity of immunoconjugates. Importantly,nBT062-SPDB-DM4 and nBT062-SPP-DM1 significantlyinhibited MM tumor growth in vivo and prolonged hostsurvival in both the xenograft mouse models of humanMM and SCID-hu mouse model (47).

444 © 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Monoclonal antibodies and multiple myeloma Allegra et al.

Page 5: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

Tassone et al. first reported the potential of tumor target-ing with an anti-CD138 antibody–maytansinoid conjugateusing the murine parent of the antibody (B-B4) found inBT062. Treatment of CD138-positive cells with B-B4-DM1significantly reduced cell survival in a dose-dependent man-ner, while B-B4 antibody alone had little efficacy (48).Antitumor activity of B-B4-DM1 was also assessed in MM

xenograft studies in mice. Marked tumor regressions wereobserved upon treatment with B-B4-DM1 in subcutaneousMM models as measured by tumor volume or by fluorescentimaging of green fluorescent protein–expressing tumors. Theantitumor activity of B-B4-DM1 was proved in a SCID-humodel of human MM, where patient MM cells proliferate ina human bone chip microenvironment (49).Recently, Rousseau et al. reported preliminary biodistribu-

tion and dosimetry results obtained in refractory MMpatients in a phase 1/2 RAIT study using iodine-131-labeledanti-CD138 (B-B4) monoclonal antibody (MoAb). Fourpatients with progressive disease were enrolled after threelines of therapy. Grade 3 thrombocytopenia was documentedin two cases, and no grade 4 hematologic toxicity wasobserved. One patient experienced partial response, with60% reduction in M-spike on serum electrophoresis, andtotal alleviation of pain, lasting for 1 yr (50).

Anti-CD38 MoAb

CD38 is a 46-kDa type II transmembrane glycoprotein with ashort 20-aa N-terminal cytoplasmic tail and a long 256-aaextracellular domain. Functions ascribed to CD38 includereceptor-mediated adhesion and signaling events, as well asimportant activities that contribute to intracellular calciummobilization. Under normal conditions, CD38 is expressed atrelatively low levels on lymphoid and myeloid cells and insome tissues of non-hematopoietic origin. The relatively highexpression of CD38 on all malignant cells in MM in combina-tion with its role in cell signaling suggests CD38 as a potentialtherapeutic Ab target for the treatment of MM (51–56).Previous studies using anti-CD38 MoAb with or without

an immunotoxin (ricin) have not led to useful clinical appli-cations (57). Nevertheless, recently, a human anti-CD38IgG1 HuMax-CD38 (daratumumab) was raised after immu-nizing transgenic mice (HuMax-Mouse) possessing human,but not mouse, Ig genes. Preclinical works evidenced thatHuMax-CD38 was able in killing primary CD38+ CD138+

patient MM cells and a range of MM/lymphoid cell lines byboth ADCC and CDC (58).De Weers et al. reported the cytotoxic mechanisms of

action of daratumumab. Daratumumab induced potentADCC in CD38-expressing MM-derived cell lines as well asin patient MM cells. Daratumumab stood out from otherCD38 MoAbs in its strong ability to induce CDC in patientMM cells. Importantly, daratumumab-induced ADCC andCDC were not affected by the presence of BMSC, suggest-

ing that daratumumab can effectively kill MM tumor cells ina tumor-preserving bone marrow microenvironment. In vivo,daratumumab was highly active and interrupted xenografttumor growth at low dosing (59).Van der Veer et al. investigated the action of lenalidomide

combined with daratumumab. Daratumumab-dependent cell-mediated cytotoxicity of purified primary MM cells, as wellas of the UM-9 cell line, was significantly enhanced by lena-lidomide pretreatment of the effector cells derived fromPBMC from healthy individuals. More importantly, theydemonstrated a synergy between lenalidomide- anddaratumumab-induced antibody-dependent cell-mediatedcytotoxicity directly in the bone marrow mononuclear cells ofMM patients, indicating that lenalidomide can also potentiatethe daratumumab-dependent lysis of myeloma cells by acti-vating the autologous effector cells within the natural environ-ment of malignant cells. Finally, daratumumab-dependentcell-mediated cytotoxicity was significantly up-regulated inPBMC derived from three MM patients during lenalidomidetreatment.The lenalidomide–daratumumab combination appears to be

a highly attractive choice because these results demonstratethat lenalidomide significantly synergizes with daratumumabto improve the MM cell lysis (60).In a dose-escalation study of daratumumab in patients

with MM, the safety profile has been acceptable. In patientswith relapsed or refractory MM treated with daratumumab, amarked reduction in paraprotein and bone marrow plasmacells was observed in the higher dose cohorts. This has notpreviously been demonstrated with a single-agent monoclo-nal antibody in MM. No unexpected buildup of dar-atumumab was seen, and in patients treated with 4 mg/kgand upwards, the observed plasma concentrations were closeto those predicted. The MTD was not yet established, andthe toxicity was manageable (61).Finally, MOR202 (MorphoSysAG), a fully human anti-

CD38 IgG1 MoAb produced by a human combinatorialantibody library (HuCAL) platform, also efficiently triggersADCC against CD38+ MM cell lines and patient MM cellsin vitro as well as in vivo in a xenograft mouse model(62).

Anti-CD40 MoAb

CD40 is a transmembrane glycoprotein of the tumor necro-sis factor (TNF) receptor superfamily that is involved inB-cell activation and the formation of germinal centers (63).It is highly expressed in B-cell malignancies, such as MM,chronic lymphocytic leukemia, and NHL (64–69).CD40 activation by its ligand (CD40L) seems to partici-

pate in B-cell tumorigenesis via NFjB, ERK, p38 MAPK,and PI3-kinase signaling (70). CD40L-CD40 signaling alsoparticipates in protective tumor immunity. Thus, inhibitionof CD40L-CD40 signaling reduces tumor cell proliferation

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 445

Allegra et al. Monoclonal antibodies and multiple myeloma

Page 6: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

and survival and may disrupt the protective immune stateand stimulate immune-mediated antitumor activity in MM(71, 72).Lucatumumab (HCD122, formerly CHIR-12.12) is a fully

human, recombinant IgG1 isotype monoclonal antibody thattargets CD40 and inhibits the growth and survival of B-cellmalignancies. In contrast to other anti-CD40 monoclonalantibodies that demonstrate agonist activity, lucatumumab isa full antagonist of the CD40L-binding site of CD40.Lucatumumab, in fact, binds CD40 with high affinity and

a slow off-rate and efficiently competes with CD40L toreduce B-cell differentiation. Lucatumumab also mediates anantitumor immune response by binding effector cells andinducing cell lysis via antibody-dependent cell-mediatedcytotoxicity (ADCC) in B-cell malignancies, includingCD40-positive MM cells (73–75).In an open-label, multicenter, phase 1 study lucatumumab

was evaluated in patients with relapsed/refractory MM.Common lucatumumab-related adverse events were mild-to-moderate infusion reactions. Severe adverse events wereanemia, chills, hypercalcaemia, and pyrexia (7% each). 43%of patients had stable disease, and one patient (4%) main-tained a partial response for 8 months. These findings indi-cate that single-agent lucatumumab was well tolerated up to4.5 mg/kg with modest clinical activity in relapsed/refractoryMM (76).Dacetuzumab is a different humanized anti-CD40 mono-

clonal antibody with multiple mechanisms of action. Dac-etuzumab kills tumor cells via ADCC and phagocytosis andinduction of apoptosis through direct signal transduction(77–79).Previous works have evaluated the pharmacokinetics (PK),

safety, and efficacy of dacetuzumab monotherapy in patientswith relapsed/refractory MM (80). Phase 1 data suggest it iswell tolerated with no immunogenicity (81).Treatment was generally well tolerated. The most common

adverse events were fatigue, headache, nausea, and anemia.Although transient decreases in serum and 24-h urineM-protein levels were observed for some patients, no objec-tive responses were reported.While single-agent dacetuzumab was not highly active in

this study, the observed safety profile suggested that testingdacetuzumab in combination with other MM therapies wouldbe possible. Preliminary in vitro results report that combin-ing dacetuzumab with lenalidomide is synergistic, and thiscombination may produce better response rates. Two trialsare underway in patients with relapsed MM to evaluatedacetuzumab in combination with lenalidomide or bortezo-mib (82, 83).Preclinical data have indicated that lenalidomide augments

the anti-MM efficacy of dacetuzumab, and the combinationregimen is undergoing clinical evaluation. A phase 1b studyof combination dacetuzumab plus lenalidomide and dexa-methasone in subjects with relapsed or refractory MM has

produced promising clinical response rates. The regimen wasgenerally well tolerated; fatigue, neutropenia, and thrombo-cytopenia were the most common adverse events (84).

Anti-CD56 MoAb

CD56 (neuronal cell adhesion molecule) is a membraneglycoprotein from the immunoglobulin superfamily (85),expressed on muscle cells and neurons. It appears to mediatecell adhesion, migration, invasion, and anti-apoptosis (86–88). CD 56 is also expressed on 70–90% of MM cells (89–91). Moreover, expression of CD56/neural cell adhesionmolecule correlates with the presence of lytic bone lesionsin MM and distinguishes myeloma from MGUS andlymphomas with plasmacytoid differentiation (92).Several studies have brought to the formation of anti-56

antibodies conjugated to cytotoxic moieties that combine thespecificity of antimyeloma-targeting antibodies with highlyactive antitumor compounds. One such immunoconjugatecurrently in clinical development is composed of antibodiesthat target cell surface proteins found on MM cells and arecoupled to cytotoxic maytansinoids (49).Lorvotuzumab mertansine (IMGN901) is an immunoconju-

gate composed of a humanized MoAb to CD56 (huN901)conjugated to the maytansinoid N2′-deacetyl-N2′-(3-mercapto-1-oxopropyl)-maytansine (DM1), a potent antimicrotubularcytotoxic agent. HuN901-DM1 has significant in vitro and invivo anti-MM activity at low doses. Target-dependent cyto-toxicity was shown in cocultures of CD56+ and CD56� cells.Importantly, adhesion of CD56+ MM cell lines and patientMM cells to BMSCs, which is known to protect MM cellsfrom drug-induced cytotoxicity, did not protect against thespecific cytotoxicity of IMGN901. Treatment with IMGN901in a human MM tumor xenograft model in immune-compro-mised mice showed that the immunoconjugate was effectivein both a minimal and bulky disease setting (85).The phase 1 clinical study of huN901-DM1 (BB-10901)

in MM patients demonstrated an overall favorable safetyprofile (93, 94). There were no hypersensitivity reactions,and no patients developed antibodies to huN901 or DM1.Three patients achieved MR, with reductions in serum Mcomponent and urine M component. Eight patients achieveddurable stable disease, and two patients remained on treat-ment for 42 wk. Finally, additive to synergistic activity hasbeen observed in combinations of IMGN901 with lenalido-mide, bortezomib or melphalan (95, 96).

Anti-IGF-1 MoAb

The insulin-like growth factor (IGF) signaling system is com-prised of the IGF ligands (IGF-1 and IGF-2), the cell surfacereceptors that mediate the biological effects of the IGFs (IGF-1 receptor (IGF-1R), IGF-2 receptor, and the insulin receptor),as well as a family of circulating IGF-binding proteins.

446 © 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Monoclonal antibodies and multiple myeloma Allegra et al.

Page 7: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

IGF-1R (CD221) is expressed on MM cells in about 75%of the cases and is associated with disease outcome (97).IGF-1 produced by the microenvironment induces the activa-tion of several pathways such as phosphatidylinositol-3kinase (PI-3K)/Akt, Janus kinase (JAK)/signal transducerand activator of transduction 3 (STAT3), mitogen-activatedprotein kinase (MAPK)/extracellular signal-regulated kinase(ERK), and nuclear factor-kappa B (NF-jB).In vivo and in vitro, IGF-1 decreases drug sensitivity of

MM cells and up-regulates several anti-apoptotic proteinssuch as A1/Bfl-1, XIAP, and Bcl-2 and causes a reductionin pro-apoptotic proteins (such as caspase 3, caspase 8, cas-pase 9) and plays a role in drug resistance (dexamethasone,rapamycin) (98–103).Monoclonal antibodies targeting the IGF-1R constitute a

very attractive approach to block the IGF-1/IGF-1R interac-tion, subsequently blocking the stimulation of signalizationpathways, especially PI-3K/Akt and NF-jB (104).Tagoug et al. analyzed the effect of the combination of

the IKK2 inhibitor AS602868 (an anilinopyrimidine deriva-tive and adenosine triphosphatase competitor selected for itsinhibitory effect on IKK2 in vitro) and a monoclonal anti-body directed against IGF-1R on MM cell lines. They foundthat anti-IGF-1R potentiated the apoptotic effect ofAS602868 in LP1 and RPMI8226 MM cell lines, whichexpress high levels of IGF-1R. Anti-IGF-1R enhanced theinhibitory effect of AS602868 on NF-jB pathway signalingand potentiated the disruption of mitochondrial membranepotential caused by AS602868 (105, 106).AVE1642 is a humanized version of the murine monoclo-

nal antibody, EM164, raised against the human IGF-1R. Ithas been shown to bind specifically, with a high affinity, tohuman IGF-1R, to inhibit IGF-1 binding and receptor activa-tion, and to down-regulate the receptor by internalizationand degradation. AVE1642 has been able to delay growthand survival of some cancer cells in vitro, human tumorxenografts in nude mice, and to inhibit proliferation andsurvival of most MM cells (107, 108).Moreau et al. reported the results of a phase 1, multicen-

ter, open-label study, made to evaluate the effects ofAVE1642 alone and in combination with bortezomib, inpatients with relapsed MM. Despite the good toxicity profileof the antibody, the response rates for patients treated withAVE1642 in this study, as a single agent or in combinationwith bortezomib, were considered insufficient (109, 110).Other IGF-IR MoAbs in clinical trials in patients with

advanced solid tumors include AMG 479, IMCA12 (111,112), and R1507 (113, 114). All these antibodies vary withregard to their IgG subclass and pharmacokinetic properties,but do share some similarities. Generally, they have anadvantageous toxicity profile, and no dose-limiting toxicitieshave been described (115).The first-in-human study of figitumumab (CP-751,871)

was conducted in patients with refractory myeloma. No

dose-limiting toxicities (DLTs) were observed, althoughindividual cases of grade 3 adverse events of hyperglycemiaand anemia were seen. Of the 27 patients who received bothfigitumumab and dexamethasone, nine responded (116).Finally, dalotuzumab (MK-0646; h7C10), is a recombinant

humanized IgG1 MoAb against the IGFR1. Preliminary datafrom phase I clinical trials suggest that dalotuzumab is safe,well tolerated, and significantly inhibits tumor proliferation.Several clinical trials evaluating dalotuzumab, alone and incombination with other anticancer agents, were ongoing inpatients with MM (117).

Anti-HM1.24 (anti-317) MoAb

The HM1.24 antigen (HM1.24) is a transmembrane proteinthat has unique topology with two membrane anchordomains: an NH2-terminal transmembrane domain and a gly-cosylphosphatidylinositol attached to the COOH terminus(118). HM1.24 (CD317) was originally identified as a cellsurface protein differentially overexpressed on MM cells andlater was found to be identical to bone stromal cell antigen2 (BST-2) (119, 120).A role of HM1.24 in trafficking and signaling between the

intracellular and cell surface of MM cells was suggestedbecause it is one of the important activators of NF-jB path-way.A humanized MoAb specific to HM1.24 antigen has been

developed. Injection of the MoAb significantly reduces M-protein levels in sera and tumor cell numbers in BM andprolongs survival of myeloma-bearing mice in myelomaxenograft mouse models (121). However, its antimyelomaactivity is diminished when the mice are pretreated withanti-Fcc receptor III/II antibodies, indicating that anti-HM1.24 MoAb kills myeloma cells via ADCC and/or CDC(121–123).Single intravenous injection of AHM significantly inhib-

ited tumor growth in both orthotopic and ectopic humanMM xenograft models. A phase 1/2 clinical study showedthat a humanized anti-HM1.24 MoAb did not cause any seri-ous toxicity when administered to patients with relapsed orrefractory MM (123).Ishiguro et al. produced defucosylated AHM and evalu-

ated it by performing autologous ADCC assays against pri-mary MM cells from patients. Defucosylated AHM showedsignificant ADCC activity against three of six primary MMcells in the presence of autologous PBMC, whereas conven-tional AHM did not. The results indicate that the potency ofAHM to induce ADCC against primary MM cells was insuf-ficient, but was significantly augmented by defucosylation(124).Tai et al. investigated in vitro and in vivo anti-MM activi-

ties of XmAb5592, a humanized anti-HM1.24 MoAb withFc domain engineered to significantly enhance FccR bindingand associated immune effector functions. XmAb5592

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 447

Allegra et al. Monoclonal antibodies and multiple myeloma

Page 8: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

increased ADCC several folds relative to the anti-HM1.24IgG1 analog against both MM cell lines and primary patientmyeloma cells. XmAb5592 also augmented antibody-dependent cellular phagocytosis (ADCP) by macrophages.NK cells became more activated by XmAb5592 than theIgG1 analog, evidenced by increased cell surface expressionof granzyme B-dependent CD107a and MM cell lysis, evenin the presence of BMSCs. XmAb5592 potently inhibitedtumor growth in mice bearing human MM xenografts viaFccR-dependent mechanisms and was significantly moreeffective than the IgG1 analog. Lenalidomide synergisticallyenhanced in vitro ADCC against MM cells and in vivotumor inhibition induced by XmAb5592. A single dose of20 mg/kg XmAb5592 effectively depleted both blood andbone marrow plasma cells in cynomolgus monkeys (125,126).Indeed, it is conceivable that HM1.24 may be a promising

target antigen for a cytotoxic antibody in the treatment ofMM (127).

Anti-CD48 MoAb

CD48 is a 47-kD glycophosphatidylinositol-linked glycopro-tein that is expressed on mature lymphocytes and mono-cytes, but not on non-hematopoietic tissues (128).Hosen et al. found that CD48 was highly expressed on

MM plasma cells. In 22 of 24 MM patients, CD48 wasexpressed on more than 90% of MM plasma cells at signifi-cantly higher levels than it was on normal lymphocytes andmonocytes. CD48 was only weakly expressed on someCD34+ hematopoietic stem/progenitor cells and notexpressed on erythrocytes or platelets.Administration of the anti-CD48 MoAb significantly

inhibited tumor growth in severe combined immunodeficientmice inoculated subcutaneously with MM cells. Further-more, anti-CD48 MoAb treatment inhibited growth of MMcells transplanted directly into murine bone marrow. Finally,they demonstrated that the anti-CD48 MoAb did not damagenormal CD34+ hematopoietic stem/progenitor cells (129).However, a major concern regarding CD48 as a therapeutic

target is its broad expression on normal lymphocytes andmonocytes, which may induce severe cytopenia and immuno-suppression when anti-CD48 MoAb is used as a therapeuticdrug. The potential hematologic toxicity of anti-CD48 MoAbshould therefore be very carefully tested, and anti-CD48MoAb may not be suitable for long-term maintenance ther-apy because of hematologic toxicities. For induction therapy,it could be useful for the eradication of MM plasma cells.

Anti-b2 microglobulin MoAb

b2-microglobulin (b2M) is an 11.6-kDa non-glycosylatedpolypeptide composed of 100 amino acids. It is part of theMHC class I molecule on the cell surface of nucleated cells.

Its best characterized function is to interact with and stabi-lize the tertiary structure of the MHC class I a-chain (130).High levels of serum b2M are present in hematologic

malignancies, including MM, and correlate with a poor out-come regardless of a patient’s renal function (131, 132).Anti-b2M MoAbs have been developed recently (133).

Several studies demonstrated that anti-b2M MoAbs haveremarkably strong tumoricidal activities to kill all examinedmyeloma, including tumor cell lines and primary CD138+

malignant plasma cells isolated from subjects with MM, andb2M

+/HLA-ABC+ hematologic malignant cells. Furthermore,they provoke a direct induction of tumor cell death withoutthe need for exogenous immunologic effector cells and/ormolecules, and they have capacity to kill chemotherapy-refractory myeloma. They demonstrated therapeutic efficacyin vivo in xenograft mouse models of myeloma and killmyeloma cells in the presence of BMSCs. Although themechanisms of its action in myeloma cell death need to befurther investigated, evidence has shown that anti-b2MMoAbs directly induce tumor cell apoptosis without immu-nologic effector mechanisms (133).b2M/MHC class I complex has been shown to operate as

an important signal-transducing molecule, which is involvedin responses ranging from anergy and apoptosis to cell pro-liferation and IL-2 production (134, 135). Earlier studiesshowed that cross-linking of b2M/MHC class I induces arise of intracellular free calcium concentration and activationof STAT3 and/or JNK through phosphorylation of a signal-ing motif at tyrosine 320 residue in the cytoplasmic domainof MHC class I a-chain (136, 137). It was evidenced thatbinding of anti-b2M MoAbs to myeloma cells results ininternalization and down-modulation of surface b2M/MHCclass I molecules and induction of myeloma cell apoptosis(133). Knockdown of surface b2M by specific small interfer-ence RNAs (siRNAs) significantly abrogates the MoAb-induced tumor cell apoptosis. As a result, cross-linking ofb2M/MHC class I molecules by the MoAbs activates JNKand inhibits PI3K/Akt and ERK, leading to compromisedmitochondrial integrity, cytochrome c release, and activationof the caspase-9 cascade (133). However, the mechanismsunderlying the MoAb-mediated binding to and cross-linkingof surface b2M/MHC class I molecules, and transduction ofapoptotic signals to cells needs further investigation (138).Nevertheless, IgG anti-b2M MoAbs induced apoptosis in

up to 90% of cells in a 48-h culture in all tested humanmyeloma cell lines (HMCLs) and primary myeloma cellsfrom patients. Anti-b2M MoAb-induced apoptosis in mye-loma cells was not blocked by soluble b2M, IL-6, or othermyeloma growth factors and was stronger than apoptosisobserved with chemotherapy drugs currently used to treatMM (e.g., dexamethasone) (139).Finally, Cao et al. hypothesized that IgM anti-b2M

MoAbs might have stronger apoptotic effects because oftheir pentameric structure. Compared with IgG MoAbs, IgM

448 © 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Monoclonal antibodies and multiple myeloma Allegra et al.

Page 9: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

anti-b2M MoAbs exhibited stronger tumoricidal activity invitro against myeloma cells and in vivo in a human-likexenografted myeloma mouse model without damaging normaltissues. IgM MoAb-induced apoptosis is dependent on thepentameric structure of the MoAbs. Disrupting pentamericIgM into monomeric IgM significantly reduced their abilityto induce cell apoptosis. Monomeric IgM MoAbs were lessefficient at recruiting MHC class I molecules into and exclu-sion of cytokine receptors from lipid rafts, and at activatingthe intrinsic apoptosis cascade (140).

Anti-CD70 MoAb

CD70 is a member of TNF family. Interaction of CD70 andits ligand CD27 modulates the expansion and differentiationof effector and memory T-cell populations (141) and pro-motes B-cell expansion and plasma cell differentiation (142).CD70 is only transiently expressed on activated B cells, Tcells, mature dendritic cells, and thymic medulla stromalcells, but it is not expressed in other normal, non-hematopoi-etic tissues (1). However, aberrant CD70 expression intumor cells has been reported in NHL, B-cell lymphocyticleukemias, Waldenstr€om macroglobulinemia, and MM (143).SGN-70, a humanized MoAb specific to CD70, has been

developed (144). SGN-70 exhibits potent antimyeloma activ-ity in vitro and significantly prolonged the survival of tumor-bearing mice in vivo. This MoAb induces Fc-mediated effectorfunctions, such as ADCC, complement fixation, and CDC.

Anti-CD74 MoAb

CD74 is an integral membrane protein that was described in1970 as an invariant chain (Ii) of the major histocompatibil-ity complex II (MHC II) in both humans and mice (145,146). It may interact with macrophage migration inhibitoryfactors that are critical mediators of the host defense and isinvolved in both acute and chronic response (147). Recentstudies have shown that CD74 is frequently expressed inMM (148). Malignant plasma cells from 80% of MMpatients and from majority of myeloma cell lines expressCD74 mRNA and protein.Moreover, CD74 protein is expressed in more than 85%

of NHL and CLL, while it shows limited expression innormal human tissues (148).Milatuzumab is a humanized anti-CD74 MoAb that

shows selective binding and rapid internalization into CD74-positive cancer cells. Milatuzumab causes growth inhibitionand induction of apoptosis in CD74-expressing MM cell lineswhen cross-linked with an anti-human immunoglobulin G sec-ondary antibody (149). Moreover, milatuzumab demonstratedpromising therapeutical activity in a CAG-SCID mouse modelofMM when used alone or in combination with doxorubicin,dexamethasone, bortezomib, or lenalidomide. Treatment ofCAG and KMS-11 MM tumors with milatuzumab, bortezomib,

and combination of both agents showed a significantly greaterefficacy of milatuzumab than that of bortezomib for treatmentof these tumors. Additionally, the combination of both agentswas even more efficacious (150, 151).In a phase I trial, milatuzumab showed no severe adverse

effects in patients with relapsed/refractory MM, and it stabi-lized the disease in some patients for up to 12 wk.A multicenter trial evaluated milatuzumab for treatment of

relapsed or refractory MM in patients who had received twoprevious standard therapies. Milatuzumab was administeredtwice weekly for 4 wk. There was a reported dose-limitingtoxicity infusion reaction and severe adverse effects (SAEs),which included a case of bacterial meningitis, fever, unex-plained hemoglobin drop, cord compression confusion,hypercalcemia, and thrombocytopenia (152).Antagonistic targeting of CD74 by milatuzumab could

remove autocrine prosurvival signals and reinstate Fas-medi-ated apoptotic signaling.

Anti-HLA-DR MoAb

Additional MoAbs are directed against a variety of furtherMM cell targets including HLA-DR by 1D09C3 (153) andHLA class I by 2D7-DB (154).NK cells play a critical role in ADCC to lyse tumor target

cells via therapeutic monoclonal antibodies, inhibitory-cellkiller immunoglobulin-like receptors (KIRs) negatively regu-late NK cell-mediated killing of HLA class I–expressingtumors, and MoAbs targeting KIR might prevent their inhib-itory signaling leading to enhanced ADCC. A new fullyhuman anti-KIR blocking MoAb, 1-7F9 (or IPH 2101),antagonizes inhibitory KIR signaling, activates NK cells,and augments NK-mediated killing of tumor cells (155,156). Importantly, 1-7F9 increases patient NK cell cytotoxic-ity against autologous MM tumor cells in vitro and appearssafe in an ongoing phase I clinical trial (157). A multicenter,open-label phase IIa clinical trial has started to evaluate IPH2101 as a single agent in patients with stable measurableMM after induction therapy. Another phase II clinical trialto assess the potential of lenalidomide combined with 1-7F9will be initiated in patients with MM (157).KIRs are receptors expressed on NK cells and a subset of

T cells and function as key regulators of NK cell activity(158). Several studies are currently underway in smolderingand first relapse MM (NCT01222286, NCT01217203,NCT00999830, NCT01248455), and safety and tolerabilityresults are expected for a phase 1 study in relapsed or refrac-tory MM (NCT00552396).Kimura et al. demonstrated that HLA class I molecules

are overexpressed in MM cells and that a recombinantsingle-chain Fv diabody against this molecule efficientlycross-links HLA class I and induces rho-mediated actinaggregation, which leads to MM cell death without effectorfunction (154, 159).

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 449

Allegra et al. Monoclonal antibodies and multiple myeloma

Page 10: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

Anti-CD229 MoAb

The family of signaling lymphocytic activation molecules(SLAM) consists of nine leukocyte cell surface molecules[CD229, CD48, CD48-H1, CD84, CD150 (SLAM), CD244,BLAME, CS1, and NTB-A], which are members of theimmunoglobulin superfamily and are involved in lymphocyteactivation (160, 161).The SLAM family member CD229, which has been

shown to interact with its intracellular adapter protein Grb2in a phosphorylation-dependent manner, showed the strong-est overexpression/phosphorylation in all myeloma cell lines(162).Atanackovic et al. identified CD229 as the most strongly

over-expressed/phosphorylated immunoreceptor in myelomacell lines. Overexpression was further demonstrated in theCD138-negative population, which has been suggested torepresent myeloma precursors, as well as on primary tumorcells from myeloma patients. Accordingly, CD229 stainingof patients’ bone marrow samples enabled the identificationof myeloma cells by flow cytometry and immunohistochem-istry. Down-regulation of CD229 led to a reduced numberof viable myeloma cells and clonal myeloma colonies andincreased the antitumor activity of conventional chemothera-peutics. Targeting CD229 with a monoclonal antibodyresulted in complement- and cell-mediated lysis of myelomacells (163).

Anti-GM-2 ganglioside MoAb

GM-2 is a ganglioside expressed on MM cells. A human-ized anti-GM-2 MoAb, BIW-8962, has demonstrated in vitrokilling of MM cell lines and in vivo effectiveness in mousexenograft models, with ADCC and CDC the most prominentcytotoxic mechanisms. BIW-8962 is being evaluated asmonotherapy in a phase 1/2 study for patients with relapsed/refractory MM (164).

Anti-CD200 MoAb

CD200 is a highly conserved transmembrane glycoproteinexpressed on a wide range of cell types; however, expres-sion of the receptor for CD200 (CD200R1) is apparentlyconfined to antigen-presenting cells of myeloid lineage andcertain T-cell populations and is thought to deliver inhibitorysignals. Several studies have shown that CD200 imparts animmunoregulatory signal through CD200R, leading to thesuppression of T-cell-mediated immune responses. CD200-deficient mice have a compromised capacity to down-regu-late the activation of antigen-presenting cells (165).Moreaux et al. identified that CD200 was expressed in

malignant plasma cells in 78% of newly diagnosed MMpatients. The expression of the CD200 gene by MM cellshas been found to be a predictor of poor prognosis inpatients with MM (166).

ALXN6000 is a humanized anti-CD200 MoAb that is cur-rently being evaluated in a phase 1/2 study in patients withMM or B-cell CLL (NCT00648739), with results expectedin the near future.

Anti-CD 54 (ICAM-1) MoAb

Intercellular adhesion molecule-1 (ICAM-1) mediates adhe-sion of myeloma cells to BMSCs and thereby participates tocell adhesion–mediated drug resistance. ICAM-1 is highlyexpressed on the plasma cell surface in the majority of MMsubjects, but is also expressed on normal cell types, includingepithelial cells, endothelial cells (ECs), fibroblasts, and sev-eral types of leukocytes (167). Expression of ICAM-1 onmyeloma cells increases after chemotherapy, and high levelsof ICAM-1 predict poor response to therapy in chemo-naivepatients. Moreover, macrophages preserve myeloma cellsfrom melphalan or dexamethasone-induced apoptosis in vitro.Zheng et al. found that macrophage-mediated myeloma drugresistance was also seen with purified macrophages frommyeloma patients’ bone marrow in vitro and was confirmedin vivo using the human myeloma-SCID mouse model. Theyshowed that ICAM-1/CD18 played an important role in mac-rophage-mediated myeloma cell drug resistance, as blockingantibodies against these molecules or genetic knockdown ofICAM-1 in myeloma cells repressed macrophages’ ability toprotect myeloma cells.BI-505 is a fully human immunoglobulin G1 antibody

specific to ICAM-1 that kills myeloma cells in vitro throughinduction of ADCC and apoptosis. In addition, BI-505 inhib-its myeloma tumor growth in mice. On the basis of thesedata, a phase 1 dose-escalation study is currently recruitingrelapsed/refractory myeloma patients to evaluate efficacy andtoxicity of single-agent BI-505 (168).

Anti-Ku MoAb

The Ku heterodimer is made up of two subunits of approxi-mately 86 and 70 kDa in higher eukaryotes, which binds toDNA. Identified in 1981 from Japanese patients with sclero-derma–polymyositis overlap syndrome, Ku has now beenfound to play major roles in many cellular processes (169).First of all, Ku86 in association with Ku70 and DNA proteinkinase C plays a critical role in DNA repair especially innon-homologous end joining where the lack of functionalKu proteins in the cell typically results in genomic instabil-ity and hypersensitivity to DNA damage as well as anincreased likelihood of tumor development and immunodefi-ciency (170–173). Moreover, Ku86 has been discovered totranslocate to the cell surface of MM cells upon CD40Ltreatment and also mediate the binding of MM cells to fibro-nectin and BMSC. Most importantly, the MoAb 5E2 direc-ted against Ku86 were seen to induced apoptosis of MMcells (174–176).

450 © 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Monoclonal antibodies and multiple myeloma Allegra et al.

Page 11: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

Liew et al. (177) found good-affinity antibodies againstKu86, and they can be used for further studies on MM andform the basis for further development as anticancer thera-peutic agents.

MoAbs Targeting components of bone marrow

milieu

Anti-IL6 MoAb

Autocrine and paracrine IL-6 have been seen to play a cru-cial role in growth and survival of MM cells within theBM milieu (178). In BM microenvironment, IL-6 is pre-dominantly produced by BMSCs, mediating MM cellgrowth and preventing apoptotic cell death. IL-6 stimulatesat least three different signaling pathways; Ras/MEK/ERKcascade, JAK2/signal transducer and activator of transcrip-tion (STAT-3) cascade, and PI3K/Akt cascade. Importantly,IL-6 protects against apoptotic cell death induced by a vari-ety of agents. In addition, IL-6 also controls expression ofvarious other key growth in MM subjects. For example, IL-6 plays an important role in the transcriptional regulation ofmyeloid cell leukemia (Mcl)-1, an anti-apoptotic B-celllymphoma-2 family member, a critical mediator of MM cellsurvival, and tightly regulated by the proteasome (179).Therefore, down-regulation of IL-6 signaling would alsosensitize MM cells to proteasome inhibitor-mediated apop-tosis by interfering with the induction of the HSP responseand Mcl-1(180). IL-6 is also a potent osteoclast activatingfactor (OAF) for human osteoclast precursors (181–186),while clinical studies have revealed that increased serumIL-6 concentrations in patients are associated with advancedstages of MM and short survival in patients. Therefore,blocking IL-6 signaling is a potential therapeutic strategyfor cancer (187).Several clinical approaches using MoAb directed at IL-6

and IL-6R have been reported (Table 2). In the past, anti-IL-6-neutralizing MoAbs have been reported to exert notable invitro anti-MM activity. However, their in vivo and clinicaleffectiveness remains ambiguous. For example, in a phase Istudy, using a mouse–human chimeric monoclonal anti-IL6antibody, none of the MM patients achieved a response(188).However, although the clinical activity of single-agent

anti-IL-6/IL-6R in MM patients has been limited, any clini-cal studies of the anti-IL-6 monoclonal antibody CNTO 328have shown evidence of activity (189, 190).Siltuximab, formerly CNTO 328, a chimeric human–

mouse monoclonal IL-6-neutralizing antibody, has, in fact,demonstrated promising antimyeloma activity (191–193).Moreover, two recent clinical studies on CNTO 328, onein combination with dexamethasone and another with bort-ezomib, have shown evidence of encouraging activity(180).

Voorhees et al. evaluated whether CNTO 328 couldenhance the apoptotic activity of dexamethasone (dex) in pre-clinical models of myeloma. CNTO 328 potently increasedthe cytotoxicity of dex in IL-6-dependent and IL-6-indepen-dent HMCLs, including a bortezomib-resistant HMCL. Isobo-logram analysis revealed that the CNTO 328/dex combinationwas highly synergistic. Addition of bortezomib to CNTO 328/dex further enhanced the cytotoxicity of the combination.Although CNTO 328 alone induced minimal cell death, itpotentiated dex-mediated apoptosis, as evidenced by increasedactivation of caspase-8, caspase-9, and caspase-3, annexin Vstaining, and DNA fragmentation. The ability of CNTO 328to sensitize HMCLs to dex-mediated apoptosis was preservedin the presence of human BMSC (192).Hunsucker et al. reported that the combination of siltux-

imab and melphalan attenuated cell proliferation in an addi-tive to synergistic manner and enhanced apoptosis inHMCLs. This increased cell death correlated with enhancedBak activation, and siltuximab also inhibited IL-6 activationof the prosurvival PI3-K/Akt signaling pathway. Importantly,the siltuximab/melphalan combination was also effective inpatient-derived myeloma samples and partially overcamemelphalan resistance (194).The anti-interleukin-6 receptor monoclonal antibody toc-

ilizumab (TCZ) was reported to inhibit in vitro proliferationof cloned and freshly isolated myeloma cells from patientswith advanced MM (195). In addition, Nishimoto et al.(196) reported the first 2 cases of refractory MM treatedwith TCZ, noting that it improved fever and systemic edemaand also stabilized the monoclonal protein levels.Matsuyama et al. reported a patient with rheumatoid

arthritis (RA) and smoldering MM, in whom TCZ improvedRA symptoms and also stabilized the serum levels of mono-clonal IgA (197).Tocilizumab treatment is generally well tolerated and safe.

It is now evaluated in open-label phase I (USA) and II(France) trials to assess its safety and efficacy as monothera-py in MM patients who are not candidates for, or who haverelapsed after SCT (197).In addition, NRI, another receptor inhibitor of IL-6 geneti-

cally engineered from tocilizumab, is under preclinical evalua-

Table 2 Monoclonal antibodies targeting components of bone

marrow milieu

Target Name

IL-6 Siltuximab (CNTO328)

Tocilizumab

NRI

Elsilimomab (B-E8)

Azintrel (OP-RR003-1)

SANT-7

VEGF Bevacizumab

EGFR Cetuximab

FGFR-3 MFGR1877A

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 451

Allegra et al. Monoclonal antibodies and multiple myeloma

Page 12: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

tion (198). An adenovirus vector encoding NRI was adminis-tered to mice intraperitoneally (i.p.) and monitored for theserum NRI level and growth reduction property on the xeno-grafted IL-6-dependent MM cell line S6B45.Elsilimomab is a murine monoclonal antibody, also known

as B-E8, which has been studied in hematologic malignancies(199–201). Initial studies of BE-8 demonstrated a transienttumor cytostasis and reduction in toxicities from IL-6 (202).The potential of combination therapy, including BE-8

(250 mg), Dex (49 mg/d), and high-dose melphalan[220 mg/m2 (HDM220)], followed by autologous SCT wasdemonstrated. 81.3% of patients exhibited a response, with acomplete response seen in 37.5% of patients without anytoxic or allergic reactions.A high-affinity fully human version of BE-8, OP-R003-1

(or 1339, Azintrel), was studied. It enhanced cytotoxicityinduced by dexamethasone, as well as bortezomib, lenalido-mide, and perifosine, in a synergistic fashion. Importantly,Azintrel also blocked bone turnover in SCID-hu mousemodel of MM, providing an additional rationale for its usein MM (203).Finally, IL-6R antagonist SANT-7, in combination with

Dex and all-trans retinoic acid (ATRA) or zoledronic acid,strongly inhibited growth and induced apoptosis in MM cells(204–206).

Anti-VEGF MoAb

One key compartment of the MM microenvironment isthe vascular niche. The role of angiogenesis in hematolog-ic malignancies is now well established. VEGF within theMM BM microenvironment induces growth, survival aswell as migration of MM cells in an autocrine manner viaVEGFR-1 and triggers angiogenesis via VEGF-2 in ECs.Recent works suggest the existence of MM-specific ECs(MMECs), which produce growth and invasive factors forplasma cells, including VEGF, FGF-2, MMP-2, as well asMMP-9. Compared with healthy human umbilical vein EC(HUVEC), MMECs secrete higher amounts of the CXCchemokines (e.g., IL8, SDF1-a, MCP-1), which act in aparacrine manner to mediate plasma cell proliferation andchemotaxis. Moreover, MM cells and BBMSC prolongsurvival of ECs both by increased secretion of EC survivalfactors, such as VEGF, and by decreased secretion ofanti-angiogenic factors (183, 207–210). Research on angio-genesis has led to the clinical approval of several anti-angiogenic agents in MM.Bevacizumab blocks VEGF and VEGF’s binding to its

receptor on the vascular endothelium (211). Anti-VEGF Abswere active alone and in combination with radiation in earlierpreclinical studies (212). It is currently being studied clinicallyin many diseases as systemic amyloidosis and MM (213).NCI’s Cancer Therapy Evaluation Program is sponsoring aphase II study of bevacizumab plus thalidomide in MM (214).

Attar-Schneider et al. explored the efficacy of anti-VEGFtreatment with bevacizumab directly on MM cells. Theyshowed that blocking VEGF is detrimental to the MM cellsand provokes cytostasis. This was evidenced in MM celllines, as well as in primary BM samples (BM MM). Utiliz-ing a constitutively Akt-expressing MM model, they showedthat the effect of bevacizumab on viability and eIF4E statusis Akt dependent (215).Somlo et al. tested the efficacy of bevacizumab alone and

in combination with thalidomide in MM patients with aphase II prospective randomized/stratified trial. Toxicitieswere mild. Among those who received bevacizumab alone,one patient – with the greatest expression of VEGF on MMcells – achieved stable disease for 238 d, but the medianEFS for the cohort was only 49 d (216).

Anti-EGFR MoAb

The epidermal growth factor receptor (EGFR) is a member ofthe ErbB family of transmembrane tyrosine kinase receptorsand contributes to malignant cell survival and proliferation(217).Upon ligand binding, the EGFR dimerizes in hetero- or

homodimers, which results in the activation of an intrinsictyrosine kinase and the initiation of activating signaling cas-cades: the Ras and MAPK pathways, and the PI-3K and pro-tein kinase B (Akt) pathways. In addition to activating thepathways that stimulate cell proliferation and cell survival,recent evidence shows that the EGFR might directly act as atranscription factor upon activation (218).Cetuximab is a chimeric human–murine monoclonal anti-

body that binds competitively and with high affinity to theEGFR. Binding of the antibody to the EGFR prevents stimu-lation of the receptor by endogenous ligands and results ininhibition of cell proliferation and angiogenesis andenhanced apoptosis. Binding of cetuximab to the receptoralso results in internalization of the antibody–receptor com-plex, which leads to an overall down-regulation of EGFRexpression (219).Cetuximab is approved for the treatment of metastatic colo-

rectal cancer and relapsed/metastatic head-and-neck-cancerand other epithelial malignancies (220, 221). Recent data indi-cate that the EGFR is expressed on the malignant plasma cellsof MM and on cells of the MM microenvironment. Moreover,MM cells coexpress the EGFR ligands amphiregulin and hep-arin-binding EGF-like growth factor (HBEGF), and inhibitionof EGFR ligand signaling induces MM cell apoptosis (222,223).B€oll et al. showed an activity of cetuximab as single agent

in a patient with relapsed MM. Cetuximab was administeredintravenously. After the initiation of cetuximab treatment,they measured disease stabilization as measured by IgG andserum-free kappa light chains in serum and urine. After atotal of 44 wk, they stopped cetuximab treatment, and they

452 © 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Monoclonal antibodies and multiple myeloma Allegra et al.

Page 13: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

measured an increase in both IgG and kappa light chains inserum and urine (224).

Anti-FGFR3 MoAb

At the cytogenetic level, the MM genome is recognized asbeing complex. The study of chromosomal translocationsgenerated by aberrant class switch recombination shows thatseveral oncogenes, including fibroblast growth factor recep-tor 3 (FGFR3), are placed under the control of the strongenhancers of the heavy chain Ig (IGH) loci, leading to theirderegulation (225).Overexpression of FGFR3 has been implicated in the

specific changes in gene expression observed in plasma cellsas a result of the t(4;14) translocation. Microarray analysis ofpatient samples with the t(4;14) translocation identified a spe-cific genetic signature characterized by perturbations in theexpression of several other genes. A small proportion ofpatients with the t(4;14) translocation also have FGFR3-acti-vating mutations, including A1157G, A1987G, A761G, andG1138A; however, overall these mutations are rare, with afrequency of approximately 5% among t(4;14) patients (226).As a part of the translational effort, new drugs that inhibit

oncogenic proteins overexpressed in defined molecular sub-groups of the disease, such as FGFR3 and MMSET in t(4;14) MM, are currently being developed (227).MFGR1877A is a human IgG1 monoclonal antibody

that binds to FGFR3 and is being investigated as a poten-tial therapy for relapsed/refractory FGFR3+ MM. Kamathet al. characterized the PK of MFGR1877A in mouse, rat,and monkey. PK of MFGR1877A was determined inathymic nude mice, Sprague–Dawley rats, and cynomolgusmonkeys after administration of single intravenous doses.The antitumor efficacy in mice bearing human tumorxenografts was used in conjunction with inhibitory activityin cell proliferation assays. Doses ranging from 2 to 3 mg/kg weekly to 6–10 mg/kg every 4 wk were predicted toachieve the target exposure in � 90% of MM patients(228).

MoAbs Targeting Tumor–BMSC Interaction

MM cells are highly dependent on the BM microenviron-ment for growth and survival through interactions particu-larly with BMSCs and osteoclasts, which secrete importantMM growth factors. Thus, MoAbs designed to blockthe binding of MM cell growth and survival factors to theircognate receptors have been under intensive development(Table 3).Moreover, in addition to therapy directed at MM cells and

tumor-promoting interactions, some efforts have beendevoted to MoAb therapy directed against the developmentof complications; to date, these efforts have been restrictedto the suppression of myeloma-related bone disease.

Anti-RANKL MoAb

Denosumab is an investigational fully human MoAb withhigh affinity and specificity for RANKL that mimics thenatural bone-protecting actions of OPG (229). Although de-nosumab was recently approved to treat osteoporosis andprevent the skeletal-related events in patients with bonemetastases from solid tumors (230, 231) in the United Statesand Europe, it is still undergoing phase III clinical trials ofits efficacy in treating MM-induced bone disease.A phase 1 clinical trial in patients with MM or breast can-

cer with bone metastases showed that following a single s.c.dose of denosumab, levels of urinary and serum N-telopep-tide decreased within 1 d, and this reduction lasted through84 d at the higher denosumab doses (232).The most commonly reported adverse events after denosu-

mab administration in patients with MM were fatigue, ane-mia, upper respiratory tract infection, and headache (233). Inaddition, a case of ONJ in a patient who had received deno-sumab was reported (234).Larger trials are ongoing to investigate the effect of deno-

sumab for the treatment of cancer-induced bone disease(235), and a recent in silico investigation supports the ideathat denosumab represents a convenient alternative topamidronate in the treatment of MM-induced bone disease(236).

Anti-Dickkopf-1 MoAb

Dickkopf-1 (DKK1), a soluble inhibitor of Wnt/b–cateninsignaling required for embryonic head development, regu-lates Wnt signaling by binding to the Wnt coreceptor lipo-protein-related protein-5 (LRP5)/Arrow. LRP5 mutationscausing high bone mass syndromes disrupt DKK1-mediatedregulation of LRP5. Forced overexpression of Dkk1 inosteoblasts causes osteopenia, disruption of the HSC niche,and defects in HSC function. Dkk1 also inhibits fracturerepair. Studies suggest that DKK1 activation in osteoblastsis the underlying cause of glucocorticoid- and estrogendeficiency–mediated osteoporosis and at least partiallyunderlies the teratogenic effects of thalidomide on limbdevelopment.

Table 3 Monoclonal antibodies targeting tumor–bone marrow stromal

cell interaction

Target Name

RANKL Denosumab

Dickkopf Anti-DKK1

BrlQ880

Activin RAP-011

ACE-011

BAFF Atacicept

SG1

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 453

Allegra et al. Monoclonal antibodies and multiple myeloma

Page 14: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

DKK1 has been implicated in the osteolytic phenotypes ofMM. Preclinical studies have shown that neutralizingDKK1/Dkk1 and/or enhancing Wnt/b–catenin signaling mayprove effective in treating bone pathologies (237).The effect of anti-DKK1 MoAb on bone metabolism and

tumor growth in a SCID-rab system has been evaluated. Theimplants of control animals showed signs of MM-inducedresorption, whereas mice treated with anti-DKK1 antibodiesblunted resorption and improved the bone mineral density ofthe implants. Histologic examination revealed that myeloma-tous bones of anti-DKK1-treated mice had increased num-bers of osteocalcin-expressing osteoblasts and reducednumber of multinucleated TRAP-expressing osteoclasts. Thebone anabolic effect of anti-DKK1 was associated withreduced MM burden (238).A different anti-DKK1 agent is BHQ880 (239, 240).

Although BHQ880 had no direct effect on MM cell growth,BHQ880 increased osteoblast differentiation, neutralized thenegative effect of MM cells on osteoblastogenesis, andreduced IL-6 secretion. Furthermore, in a SCID-hu murinemodel of human MM, BHQ880 treatment led to a significantincrease in osteoblast number, serum human osteocalcinlevel, and trabecular bone. A preliminary result from a phaseI/II trial in MM where BHQ880 was given was well toler-ated when given in combination with zoledronic acid.Finally, Dickkopf-1 (DKK1), broadly expressed in mye-

loma cells but highly restricted in normal tissues, togetherwith its functional roles as an osteoblast formation inhibitor,may be an ideal target for immunotherapy in myeloma.Quian et al. examined whether DKK1 can be used as a

tumor vaccine to elicit DKK1-specific immunity that cancontrol myeloma growth or even eradicate established mye-loma in vivo. They used DKK1-DNA vaccine in the murineMOPC-21 myeloma model, and the results showed thatactive vaccination using the DKK1 vaccine not only wasable to protect mice from developing myeloma, but also wastherapeutic against established myeloma. Mechanistic studiesrevealed that DKK1 vaccine elicited a strong DKK1- andtumor-specific CD4+ and CD8+ immune responses, andtreatment with B7H1 or OX40 Abs significantly reduced thenumbers of IL-10-expressing and Foxp3+ regulatory T cellsin vaccinated mice (241).

Anti-activin A MoAb

Activin A is a TGF-b superfamily member most commonlyassociated with embryogenesis and gonadal hormone signal-ing (242). In addition, activin A is involved in bone remod-eling with growth stimulatory effects on osteoclasts (Ocs)(243). Activin A inhibits osteoblasts (OB) differentiation bystimulating SMAD2 activity and inhibiting distal-lesshomeobox (DLX)-5 expression. More importantly, inhibitionof activin A signaling rescued MM-induced OB impairmentin vitro and in vivo while reducing MM burden in a human-

ized myeloma model. Malignant plasma cells disrupt thenormal regulatory pathway of bone homeostasis by inducingBMSC secretion of activin A via JNK pathway (244).Activin can be targeted by a chimeric antibody RAP-011,

derived from the fusion of the extracellular domain of acti-vin receptor IIA and the constant domain of the murineIgG2a. ACE-011, a novel bone anabolic agent currently in aphase 2 clinical trial in MM, is a protein therapeutic basedon the activin receptor IIA. In MM, an ongoing multicenterphase 2 trial is conducted in patients who are treated withmelphalan, prednisone, and thalidomide. Preliminary resultsshow clinical significant increases in biomarkers of bone for-mation, improvement in skeletal metastases, and decreases inbone pain as well as antitumor activity. Moreover, ACE-011has potential as a novel therapy for chemotherapy-inducedanemia and may be an effective alternative to erythropoietin(EPO)-based treatments (245).

Anti-BAFF MoAb-

B-cell-activating factor (BAFF) is a TNF superfamily mem-ber and produced in the bone marrow microenvironment bymonocytes, osteoclasts, and neutrophils. Recently, BAFFwas recognized as new survival factors for MM (246). Inaddition to BMSCs, osteoclasts produce these factors tosupport MM cells in the BM microenvironment (247).Binding of BAFF to the three different TNF-R-related

receptors, TACI, BCMA, and BAFF-R, triggers activation ofNF-jB, PI3K, and MAPK pathways, resulting in survivaland dexamethasone resistance of myeloma cells (248, 249).BAFF also enhances adhesion of myeloma cells to BMSC

(250). In a mouse model, an anti-BAFF-neutralizing anti-body had antimyeloma activity and inhibited osteoclastogen-esis.Atacicept acts as a decoy receptor by binding to and neutral-

izing soluble BAFF and APRIL and preventing these ligandsfrom binding to their cognate receptors on B-cell tumors,thereby enhancing cytotoxicity. An open-label, dose-escalation phase I/II study enrolled patients with refractory orrelapsed MM or active, progressive Waldenstr€om macroglob-ulinemia (251, 252). Atacicept was well tolerated and showedclinical and biological activity consistent with its mechanismof action. TACI was expressed heterogeneously among patientMM cells, which may explain promising results for the treat-ment of TACIhigh MM cells in a trial for atacicept (253).On the basis of these results, a phase 1 study is currently

enrolling relapsed/refractory myeloma patients to evaluatethe combination of the neutralizing anti-BAFF antibody,LY2127399, combined with bortezomib.Finally, B-cell maturation antigen (BCMA) is expressed

on normal and malignant plasma cells and represents apotential target for therapeutic intervention. BCMA binds totwo ligands that promote tumor cell survival, a proliferationinducing ligand (APRIL) and B-cell activating factor. To

454 © 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Monoclonal antibodies and multiple myeloma Allegra et al.

Page 15: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

selectively target BCMA for plasma cell malignancies, Ryanet al. developed antibodies with ligand-blocking activity thatcould promote cytotoxicity of MM cell lines as naked anti-bodies or as antibody–drug conjugates. They showed thatSG1, an inhibitory BCMA antibody, blocks APRIL-dependent activation of NF-jB in a dose-dependent mannerin vitro. Cytotoxicity of SG1 was assessed as a naked anti-body after chimerization with and without Fc mutations thatenhance FcgammaRIIIA binding. The Fc mutationsincreased the antibody-dependent cell-mediated cytotoxicitypotency of BCMA antibodies against MM lines by approxi-mately 100-fold with a � 2-fold increase in maximal lysis.As an alternative therapeutic strategy, anti-BCMA antibodieswere endowed with direct cytotoxic activity by conjugationto the cytotoxic drug, monomethyl auristatin F. The mostpotent BCMA antibody–drug conjugate displayed IC(50)values of � 130 pmol/L for three different MM lines (254).

Other potential targets

Anti-TRAIL-R1 R2 MoAb

The TNF family members Fas ligand and TRAIL induceapoptosis upon binding to the death receptors Fas, andTRAIL receptor 1(TRAIL-R1; DR4), or TRAIL receptor 2(TRAIL-R2; DR5), respectively. TRAIL also binds to thedecoy receptors DcR1 and DcR2 and to the soluble decoyreceptor osteoprotegerin (OPG), which is produced by osteo-blasts and stromal MoAbs activating death receptors (255–258) (Table 4).TRAIL-R1 and TRAIL-R2 antimyeloma activity was

observed for the fully human agonistic antibody directedagainst TRAIL-R1 (mapatumumab, HGS-ETR1) and to alesser extent for the anti-TRAIL-R2 antibody (lexatumumab,HGS-ETR2). The two human agonistic MoAbs killed 68%and 45% of MM cell lines, respectively. Only 18% of MMcell lines are resistant to either antibody (259).Importantly, the antimyeloma action of anti-DR4 and anti-

DR5 antibodies was not affected by the presence of cells ofthe bone marrow microenvironment, whereas these cellsprotected myeloma cells against TRAIL-induced apoptosis(260). This protective effect was at least partly mediated byOPG.Bortezomib treatment is associated with the up-regulation

of TRAIL and its receptors in B-CLL and lymphoma cells(261). Furthermore, blockage of TRAIL-R1 and TRAIL-R2expression using RNA interference, which prevents TRAILapoptotic signaling, inhibited proteasome inhibitor-inducedapoptosis.Based on enhanced cytotoxicity when combining map-

atumumab with bortezomib in preclinical experiments (262),a randomized phase II study was recently started comparingTRM-1 plus bortezomib versus bortezomib alone in patientswith relapsed or refractory MM (263).

Indeed, combined bortezomib plus mapatumumab treat-ment resulted in enhanced myeloma cell killing when com-pared to either agent alone, in both wild-type andbortezomib-resistant cells. In contrast to the preclinical data,a randomized phase 2 study in relapsed/refractory myelomashowed that addition of mapatumumab to bortezomib treat-ment did not increase response rate or progression-free sur-vival compared with bortezomib alone (264).

Anti-PD-L1 MoAb

PD-L1 (also known as B7-H1 or CD 274) is a B7 familymember and is the ligand for PD-1 (programmed death-1),a member of the B28 family. PD-L1 interacts with PD-1and an unknown receptor on T cells and can inhibit T-cellactivation and cytotoxic T-lymphocytes-mediated lysis(265).PD-L1 overexpression appears as a possible mechanism

for tumors to avoid the host’ immune response, and a wayof improving antitumor activity of NK cells against mye-loma cells is by modulating the PD-1/PD-L1 axis, whichdown-regulates the immune response. PD-1 is not present onNK cells from healthy donors, but can be up-regulated byexogenous IL-2. In contrast, PD-1 is present on NK cellsfrom patients, and interaction with PD-L1 on myeloma cellsinhibits NK cell function. PD-L1 is expressed on plasmacells from the majority of myeloma patients, but only rarelyon plasma cells from MGUS patients and not on normalplasma cells (266). CT-011 is a humanized anti-PD-1MoAb, which enhanced NK cell function against myelomatumor cells through increased NK cell trafficking, improvedimmune complex formation between myeloma cells and NKcells, and increased NK cell cytotoxicity.Lenalidomide not only activates NK cells, but also down-

regulates PD-L1 expression on myeloma cells and synergizedwith CT-011 in the activation of NK cells and subsequentkilling of myeloma cells. Apart from effects on NK cells,blockade of the PD-1/PD-L1 interaction with anti-PD-L1MoAbs enhanced myeloma-specific T-cell immunity in vitroand in vivo (267).Furthermore, CT-011 also enhanced T-cell responses to

autologous dendritic cell/myeloma fusion vaccines (268).This may be explained by augmented expression of PD-1 onT cells from myeloma patients when compared to healthycontrols.

Table 4 Other potential targets of monoclonal antibodies

Target Name

TRAIL-R1 Mapatuzumab

TRAIL-R2 Lexatuzumab

PD-L1 CD-011

VLA-4 Natalizumab

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 455

Allegra et al. Monoclonal antibodies and multiple myeloma

Page 16: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

On the basis of these data, a phase 1 clinical trial wasstarted to study the action of CT-011 in patients withadvanced hematologic malignancies including myelomapatients. CT-011 was demonstrated to be well tolerated withevidence of antitumor activity. Treatment with CT-011 wasaccompanied with an elevated percentage of peripheral bloodCD4-positive T cells.The combination of CT-011 with an IMiD may further

enhance host antitumor immunity and warrants further inves-tigation. Alternatively, the immune response to myelomamay be enhanced by anti-PD-L1 antibodies such as MDX-1105.

Anti-VLA-4 MoAb

BMSCs as well as extra cellular matrix (ECM) proteinslaminin, microfibrillar collagen type IV, and fibronectin arestrong adhesive components for MM cells via interaction ofa variety of integrins including integrin-a4 (also CD49d, asubunit of CD49d/CD29 or very-late antigen-4; VLA-4)(269).A marked increase in BM angiogenesis in MM was found

to correlate with VLA-4 expression in plasma cells isolatedfrom patients with active MM. The ability of bortezomib toovercome cell adhesion–mediated drug resistance (CAM-DR) (vincristine and dexamethasone) is, at least in part, dueto down-regulation of VLA-4 expression (270).Directly targeting VLA-4 and its subunit integrin-a4 in

particular is therefore of high therapeutic interest in MM.Previous studies using a murine anti-integrin-a4 antibodyreduced MM growth, IgG2b production, and associatedosteoclastic osteolysis in 5TGM1 murine MM cells both invitro and in vivo (271).Podar et al. evaluated the therapeutic potential of the new-

in-class-molecule selective-adhesion molecule (SAM) inhibi-tor natalizumab, a recombinant humanized IgG4 monoclonalantibody, which binds integrin-a4, iMM (272).Natalizumab, but not a control antibody, inhibited adhe-

sion of MM cells to non-cellular and cellular components ofthe microenvironment as well as disrupted the binding ofalready adherent MM cells. Consequently, natalizumabblocked both the proliferative effect of MM–BMSC interac-tion on tumor cells and vascular endothelial growth factor(VEGF)-induced angiogenesis in the BM milieu. Moreover,natalizumab also blocked VEGF- and IGF-1-inducedsignaling sequelae triggering MM cell migration. Moreover,natalizumab inhibited tumor growth, VEGF secretion, andangiogenesis in a human severe combined immunodeficiencymurine model of human MM in the human BM microenvi-ronment.Importantly, natalizumab not only blocked tumor

cell adhesion, but also chemo-sensitized MM cells to bort-ezomib, in an in vitro therapeutically representative humanMM–stroma cell coculture system model (273).

Anti-CD11C1 (antikininogen) MoAb

Sainz et al. used two cell lines (B38 and C11C1) derivedfrom P3X63Ag8 myeloma cells. The new cell lines wereimplanted subcutaneously in the strain of origin (Balb/cmice) and used as a model to monitor the effects of C11C1MoAb to kininogen (HK). They assessed their behavior byintraperitoneal and subcutaneous implantation, by implantingthem together and by treating B38-MM with purified MoAbC11C1. They found that MoAb C11C1 inhibits its owntumor growth in vivo and slows down B38-MM growth rateboth when MM is implanted together and when MoAbC11C1 is injected intraperitoneally. MAb C11C1-treatedMM showed decreased MVD and HK binding in vivo with-out FGF-2, B1R, or B2R expression changes (274).

Polyclonal antibody (rATG)

Polyclonal antibody preparations may have several advanta-ges over monoclonal therapeutic agents, including the abilityto target multiple surface proteins and simultaneously triggerseveral parallel or additive pathways for cell death. This maybe a distinct advantage when attempting to eradicate mye-loma cells, which emerge from a common less differentiatedprecursor (275, 276), and may be responsive to coordinateactivation of several cell death pathways (277, 278).Zand et al. measured complement-independent cell death

measured after addition of polyclonal rabbit antithymocyteglobulin (rATG) to cultures of myeloma cell lines or primaryCD138+ isolates from patient bone marrow aspirates. rATGinduced significant levels of apoptosis in myeloma cells asassayed by caspase induction, annexin V binding, subdiploidDNA fragmentation, plasma membrane permeability, andloss of mitochondrial membrane potential. Three pathwaysof cell death were identified involving caspase activation,cathepsin D, and the genistein sensitive tyrosine kinase path-way. Fab′2 fragments of rATG had reduced pro-apoptoticactivity, which was restored by co-incubation with Fc frag-ments and anti-CD32 or anti-CD64 antibodies (279).

Future perspectives for monoclonal antibodies

in multiple myeloma: limits and challenges

Therapeutic monoclonal antibodies have revolutionized treat-ment options for many cancers; however, MoAbs targetingmyeloma cells have not yet been included as part of stan-dard myeloma therapy. Despite several efforts, the benefit ofMoAb-based therapy directed at different targets in MMremains incompletely articulated. MAbs, when employed asmonotherapy in MM, have generally not produced impres-sive levels of response with respect to either response ratesor extent of response in individual patients. However, pre-clinical results in MM cell lines and murine explant modelsand preliminary clinical results in patients with relapsed/refractory MM suggest that MAbs are likely to act synergis-

456 © 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Monoclonal antibodies and multiple myeloma Allegra et al.

Page 17: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

tically with traditional therapies (dexamethasone), immunemodulators (thalidomide, lenalidomide), and other noveltherapies (such as the first-in-class proteasome inhibitor bort-ezomib); in addition, MAbs have shown the ability to over-come resistance to these therapies (280).Moreover, along with efforts to develop functional anti-

bodies that could provide benefit to MM patients, substantialefforts are underway to develop therapies using antibodiesconjugated to potent cytotoxic agents. A variety of highlycytotoxic compounds are being evaluated for antibody-baseddelivery, including calicheamicin, doxorubicin, taxanes,maytansinoids, dolastatins, and CC-1065 analogs (281, 282).The near future will see a novel interest in developing

novel targets for antibody-based therapies for MM. BMangiogenesis has an important role in the initiation and pro-gression of MM. Berardi et al. looked at novel mechanismsof vessel formation in patients with MM through a compara-tive proteomic analysis between BM ECs of patients withactive MM (MMECs) and ECs of patients with monoclonalgammopathy of undetermined significance (MGECs) and ofsubjects with benign anemia (normal ECs). Four proteinswere found overexpressed in MMECs: filamin A, vimentin,a-crystallin B, and 14-3-3f/d protein. Berardi et al. investi-gated the differences in MMEC versus MGEC proteome toidentify new targets for MM anti-angiogenic management.They found that FLNA, VIM, CRYAB, and YWHAZ areconstantly overexpressed in MMECs and enhanced byVEGF, FGF2, HGF, and MM plasma cell CM. These pro-teins are critically involved in MMEC overangiogenic phe-notype, and indeed, their silencing is anti-angiogenic (283).However, one of the major problems with MoAb therapy is

its immunogenicity, specifically defined as human anti-mouseantibody (HAMA). To overcome this limitation, murinemolecules are engineered to remove immunogenic murinecontent, which has been initially achieved by generation ofmouse–human antibodies. Chimeric antibodies are composedof murine variable regions fused with human constant regions.Although with reduced immunogenicity and prolonged half-life, chimeric MoAbs still contain a significant proportion(approximately 35%) of antigenic mouse determinants, sug-gesting a possibility to generate HAMA.Finally, although the ability to create essentially human

antibody structures has reduced the likelihood of host-protective immune responses that otherwise limit the utilityof therapy, majority of MM patients are immunosuppressive.The immediate goal would be testing next generations ofgenetically Fc-engineered MoAbs that not only bind to targetMM antigens with high affinity but also have superior inter-action with host immune effectors.

Conclusions

Results of experimental agents targeting a number of potentialcandidate molecules expressed on the surface of MM tumor

cells have been reported as have those of MoAbs targetingother proteins involved in the MM immunologic microenvi-ronment. The introduction of novel antimyeloma agents willresult in a more individualized targeted therapy (284).It remains to be defined how MAb therapy can most

productively be incorporated into the current therapeutic para-digms that have achieved significant survival gains in patientswith MM. This will require careful consideration of the opti-mal sequence of therapies and their clinical placement aseither short-term induction therapy, frontline treatment inpatients ineligible for ASCT, or long-term maintenance ther-apy (285).

References

1. Eshaghian S, Berenson JR. Multiple myeloma: improvedoutcomes with new therapeutic approaches. Curr OpinSupport Palliat Care 2012;6:330–6.

2. Kumar SK, Lee JH, Lahuerta JJ, et al. Risk of progressionand survival in multiple myeloma relapsing after therapywith IMiDs and bortezomib: a multicenter internationalmyeloma working group study. Leukemia 2012;26:149–57.

3. Di Bernardo A, Macor P, Guarnotta C, Franco G, FlorenaAM, Tedesco F, Tripodo C. Humoral immunotherapy ofmultiple myeloma: perspectives and perplexities. ExpertOpin Biol Ther 2010;10:863–73.

4. Lozanski G, Heerema NA, Flinn IW, Smith L, Harbison J,Webb J, Moran M, Lucas M, Lin T, Hackbarth ML. Ale-mtuzumab is an effective therapy for chronic lymphocyticleukemia with p53 mutations and deletions. Blood2004;103:3278–81.

5. Blad�e J, de Larrea CF, Rosi~nol L. Incorporating monoclonalantibodies into the therapy of multiple myeloma. J ClinOncol 2012;30:1904–6.

6. van de Donk NW, Kamps S, Mutis T, Lokhorst HM. Mono-clonal antibody-based therapy as a new treatment strategy inmultiple myeloma. Leukemia 2012;26:199–213.

7. Weiner LM, Surana R, Wang S. Monoclonal antibodies:versatile platforms for cancer immunotherapy. Nat RevImmunol 2010;10:317–27.

8. Kastritis E, Charidimou A, Varkaris A, Dimopoulos MA.Targeted therapies in multiple myeloma. Target Oncol2009;4:23–36.

9. Pilarski LM, Giannakopoulos NV, Szczepek AJ, MasellisAM, Mant MJ, Belch AR. In multiple myeloma, circulatinghyper-diploid B cells have clonotypic immunoglobulinheavy chain rearrangements and may mediate spread ofdisease. Clin Cancer Res 2000;6:585–96.

10. Pilarski LM, Baigorri E, Mant MJ, Pilarski PM, AdamsonP, Zola H, Belch AR. Multiple myeloma includes phenotyp-ically defined subsets of clonotypic CD20+ B cells thatpersist during treatment with rituximab. Clin Med Oncol2008;2:275–87.

11. Treon SP, Shima Y, Grossbard ML, Preffer FI, Belch AR,Pilarski LM, Anderson KC. Treatment of multiple myeloma

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 457

Allegra et al. Monoclonal antibodies and multiple myeloma

Page 18: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

by antibody mediated immunotherapy and induction ofmyeloma selective antigens. Ann Oncol 2000;11:S107–11.

12. Gemmel C, Cremer FW, Weis M, Witzens M, MoldenhauerG, Koniczek KH, Imbach U, Ho AD, Moos M,Goldschmidt H. Anti-CD20 antibody as consolidationtherapy in a patient with primary plasma cell leukemia afterhigh-dose therapy and autologous stem cell transplantation.Ann Hematol 2002;81:119–23.

13. Musto P, Carella AM Jr, Greco MM, Falcone A, SanpaoloG, Bodenizza C, Cascavilla N, Melillo L, Carella AM. Shortprogression-free survival in myeloma patients receivingrituximab as maintenance therapy after autologoustransplantation. Br J Haematol 2003;123:746–7.

14. Ohno H. Long-term response to maintenance treatment withrituximab in CD20(+) multiple myeloma. Leuk Lymphoma2010;51:2144–6.

15. Kapoor P, Greipp PT, Morice WG, Rajkumar SV, WitzigTE, Greipp PR. Anti-CD20 monoclonal antibody therapy inmultiple myeloma. Br J Haematol 2008;141:135–48.

16. Zojer N, Kirchbacher K, Vesely M, H€ubl W, Ludwig H.Rituximab treatment provides no clinical benefit in patientswith pretreated advanced multiple myeloma. Leuk Lym-phoma 2006;47:1103–9.

17. Robillard N, Avet-Loiseau H, Garand R, Moreau P, PineauD, Rapp MJ, Harousseau JL, Bataille R. CD20 is associatedwith a small mature plasma cell morphology and t(11;14) inmultiple myeloma. Blood 2003;102:1070–1.

18. Gozzetti A, Fabbri A, Lazzi S, Bocchia M, Lauria F. Replyto Rituximab activity in CD20 positive multiple myeloma.Leukemia 2007;21:1842–3.

19. Hofer S, Hunziker S, Dirnhofer S, Ludwig C. Rituximabeffective in a patient with refractory autoimmune haemolyticanaemia and CD20-negative multiple myeloma. Br JHaematol 2003;122:690–1.

20. Moreau P, Voillat L, Benboukher L, et al. Rituximab inCD20 positive multiple myeloma. Leukemia 2007;21:835–6.

21. Treon SP, Pilarski LM, Belch AR, et al. CD20-directedserotherapy in patients with multiple myeloma: biologicconsiderations and therapeutic applications. J Immunother2002;25:72–81.

22. Almeida J, Orfao A, Ocqueteau M, Mateo G, Corral M,Caballero MD, Blade J, Moro MJ, Hernandez J, San MiguelJF. High-sensitive immunophenotyping and DNA ploidystudies for the investigation of minimal residual disease inmultiple myeloma. Br J Haematol 1999;107:121–31.

23. Lebovic D, Kaminski MS, Anderson TB, Detweiler-ShortK, Griffith KA, Jobkar TL, Kandarpa M, Jakubowiak A. Aphase II study of consolidation treatment with iodione-131tositumomab (BexxarTM) in multiple myeloma (MM). Blood(ASH Annual Meeting Abstracts) 2012;120:1854.

24. Gisslinger H, Kees M. Therapy strategies for multiple mye-loma: current status.Wien Klin Wochenschr 2003;115:451–61.

25. Rossi EA, Rossi DL, Stein R, Goldenberg DM, Chang CH.A bispecific antibody-IFNa2b immunocytokine targetingCD20 and HLA-DR is highly toxic to human lymphomaand multiple myeloma cells. Cancer Res 2010;70:7600–9.

26. Tai YT, Dillon M, Song W, et al. Anti-CSl humanizedmonoclonal antibody HuLuc63 inhibits myeloma cell adhe-sion and induces antibody-dependent cellular cytotoxicity inthe bone marrow milieu. Blood 2008;112:1329–37.

27. Hsi ED, Steinle R, Balasa B, et al. CS1, a potential newtherapeutic antibody target for the treatment of multiplemyeloma. Clin Cancer Res 2008;14:2775–84.

28. Boles KS, Stepp SE, Bennett M, Kumar V, Mathew PA.2B4 (CD244) and CS1: novel members of the CD2 subsetof the immunoglobulin superfamily molecules expressed onnatural killer cells and other leukocytes. Immunol Rev2001;181:234–49.

29. Tassi I, Colonna M. The cytotoxicity receptor CRACC (CS-1) recruits EAT-2 and activates the PI3K and phospholipaseCgamma signaling pathways in human NK cells. J Immunol2005;175:7996–8002.

30. Tassi I, Presti R, Kim S, Yokoyama WM, Gilfillan S,Colonna M. Phospholipase C-gamma 2 is a critical signalingmediator for murine NK cell activating receptors. J Immunol2005;175:749–54.

31. Tai YT, Tonon G, Leiba M. CS1, a new surface target onmultiple myeloma (MM) cells, protects myeloma cells fromapoptosis via regulation of ERK1/2, AKT and STAT3 sig-naling cascades. Blood (ASH Annual Meeting Abstracts)2007;110:40.

32. Tai YT, Soydan E, Song W, et al. CS1 promotes multiplemyeloma cell adhesion, clonogenic growth, and tumorige-nicity via c-maf-mediated interactions with bone marrowstromal cells. Blood 2009;113:4309–18.

33. Bensinger W, Zonder J, Singhal S. Phase I trial of HuLuc63in multiple myeloma. Blood (ASH Annual MeetingAbstracts) 2007;110:358.

34. van Rhee F, Szmania SM, Dillon M, et al. Combinatorialefficacy of anti-CS1 monoclonal antibody elotuzumab(HuLuc63) and bortezomib against multiple myeloma. MolCancer Ther 2009;8:2616–24.

35. Zonder JA, Mohrbacher AF, Singhal S, van Rhee F,Bensinger WI, Ding H, Fry J, Afar DE, Singhal AK. Aphase 1, multicenter, open-label, dose escalation study ofelotuzumab in patients with advanced multiple myeloma.Blood 2012;120:552–9.

36. Zonder JA, Singhal S, Bensinger W, Mohrbacher A,Hussein MA, Munshi NC, Caras I, Singhal A, van Rhee F.Phase I study of elotuzumab (HuLuc63) in relapsed/refrac-tory multiple myeloma. Blood (ASH Annual MeetingAbstracts) 2008;112:2773.

37. Richardson PG, Moreau P, Jakubowiak AJ, et al. Elot-uzumab in combination with lenalidomide and dexametha-sone in patients with relapsed multiple myeloma: interimresults of a phase 2 study. Blood (ASH Annual MeetingAbstracts) 2010;116:986.

38. Rice AG, Dillon MB, Van Abbema AM. HuLuc63 in com-bination regimens with conventional and targeted therapieshas additive and synergistic anti-tumor activity in pre-clini-cal models of myeloma. Blood (ASH Annual MeetingAbstracts) 2007;110:2517.

458 © 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Monoclonal antibodies and multiple myeloma Allegra et al.

Page 19: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

39. Jakubowiak AJ, Bensinger W, Siegel D. Phase 1/2 study ofelotuzumab in combination with bortezomib in patients withmultiple myeloma with one to three prior therapies: interimresults. Blood (ASH Annual Meeting Abstracts)2009;114:3876.

40. Lonial S, Vij R, Harousseau J-L. Phase 1/2 study of elot-uzumab in combination with lenalidomide and low dosedexamethasone in relapsed or refractory multiple myeloma:interim results. Blood (ASH Annual Meeting Abstracts)2009;114:432.

41. Lonial S, Vij R, Harousseau JL, et al. Elotuzumab in com-bination with lenalidomide and low-dose dexamethasone inrelapsed or refractory multiple myeloma. J Clin Oncol2012;30:1953–9.

42. Jakubowiak AJ, Benson DM, Bensinger W, Siegel DS,Zimmerman TM, Mohrbacher A, Richardson PG, Afar DE,Singhal AK, Anderson KC. Phase I trial of anti-CS1 mono-clonal antibody elotuzumab in combination with bortezomibin the treatment of relapsed/refractory multiple myeloma.J Clin Oncol 2012;30:1960–5.

43. Sanderson RD, Lalor P, Bernfield M. B lymphocytesexpress and lose syndecan at specific stages of differentia-tion. Cell Regul 1989;1:27–35.

44. Yang Y, Macleod V, Miao HQ, et al. Heparanase enhancessyndecan-1 shedding: a novel mechanism for stimulation oftumor growth and metastasis. J Biol Chem 2007;282:13326–33.

45. Wijdenes J, Vooijs WC, Clement C, Post J, Morard F, VitaN, Laurent P, Sun RX, Klein B, Dore JM. A plasmocyteselective monoclonal antibody (B-B4) recognizes syndecan-1. Br J Haematol 1996;94:318–23.

46. Seidel C, Sundan A, Hjorth M, Turesson I, Dahl IM, Abil-dgaard N, Waage A, Borset M. Serum syndecan-1: a newindependent prognostic marker in multiple myeloma. Blood2000;95:388–92.

47. Ikeda H, Hideshima T, Fulciniti M, et al. The monoclonalantibody nBT062 conjugated to cytotoxic Maytansinoidshas selective cytotoxicity against CD138-positive multiplemyeloma cells in vitro and in vivo. Clin Cancer Res2009;15:4028–37.

48. Tassone P, Goldmacher VS, Neri P, et al. Cytotoxic activ-ity of the maytansinoid immunoconjugate B-B4-DM1against CD138+ multiple myeloma cells. Blood 2004;104:3688–96.

49. Lutz RJ, Whiteman KR. Antibody-maytansinoid conjugatesfor the treatment of myeloma. MAbs 2009;1:548–51.

50. Rousseau C, Ferrer L, Supiot S, et al. Dosimetry resultssuggest feasibility of radioimmunotherapy using anti-CD138(B-B4) antibody in multiple myeloma patients. Tumour Biol2012;33:679–88.

51. Lin P, Owens R, Tricot G, Wilson CS. Flow cytometricimmunophenotypic analysis of 306 cases of multiplemyeloma. Am J Clin Pathol 2004;121:482–8.

52. Malavasi FA, Funaro S, Roggero A, Horenstein L, MehtaCK. Human CD38: a glycoprotein in search of a function.Immunol Today 1994;15:95–7.

53. Deaglio S, Mehta K, Malavasi F. Human CD38: a (r)evolu-tionary story of enzymes and receptors. Leuk Res2001;25:1–12.

54. Gallay N, Anani L, Lopez A, Colombat P, Binet C,Domenech J, Weksler BB, Malavasi F, Herault O. The roleof platelet/endothelial cell adhesion molecule 1 (CD31) andCD38 antigens in marrow microenvironmental retention ofacute myelogenous leukemia cells. Cancer Res2007;67:8624–32.

55. Partida-Sanchez S, Cockayne DA, Monard S, et al. CyclicADP-ribose production by CD38 regulates intracellular cal-cium release, extracellular calcium influx and chemotaxis inneutrophils and is required for bacterial clearance in vivo.Nat Med 2001;7:1209–16.

56. Santonocito AM, Consoli U, Bagnato S, Milone G,Palumbo GA, Di Raimondo F, Stagno F, Guglielmo P, Giu-stolisi R. Flow cytometric detection of aneuploid CD38(++)plasmacells and CD19(+) B-lymphocytes in bone marrow,peripheral blood and PBSC harvest in multiple myelomapatients. Leuk Res 2004;28:469–77.

57. Goldmacher VS, Bourret LA, Levine BA, Rasmussen RA,Pourshadi M, Lambert JM, Anderson KC. Anti-CD38-blocked ricin: an immunotoxin for the treatment of multiplemyeloma. Blood 1994;84:3017–25.

58. Tai YT, de Weers M, Li X. Daratumumab, a novel potenthuman anti-CD38 monoclonal antibody, induces significantkilling of human multiple myeloma cells. Therapeutic Impli-cation. Blood (ASH Annual Meeting Abstracts)2009;114:608.

59. de Weers M, Tai YT, van der Veer MS, et al. Dar-atumumab, a novel therapeutic human CD38 monoclonalantibody, induces killing of multiple myeloma and otherhematological tumors. J Immunol 2011;186:1840–8.

60. van der Veer MS, de Weers M, van Kessel B, Bakker JM,Wittebol S, Parren PWHI, Lokhorst HM, Mutis T. Towardseffective immunotherapy of myeloma: enhanced eliminationof myeloma cells by combination of lenalidomide with thehuman CD38 monoclonal antibody daratumumab. Haemato-logica 2011;96:284–90.

61. Plesner T, Lokhorst H, Gimsing P, Nahi H, Lisby S,Richardson PG. Daratumumab, a CD38 monoclonal anti-body in patients with multiple myeloma – data from a dose-escalation phase I/II study. Blood (ASH Annual MeetingAbstracts) 2012;143:654.

62. Tesar M. Fully human antibody MOR202 against CD38 forthe treatment of multiple myeloma and other blood–bornemalignancies. J Clin Oncol 2007; 25:article 8106.

63. Lee BO, Moyron-Quiroz J, Rangel-Moreno J, Kusser KL,Hartson L, Sprague F, Lund FE, Randall TD. CD40, butnot CD154, expression on B cells is necessary for optimalprimary B cell responses. J Immunol 2003;171:5707–17.

64. Pellat-Deceunynck C, Bataille R, Robillard N, HarousseauJL, Rapp MJ, Juge-Morineau N, Wijdenes J, Amiot M.Expression of CD28 and CD40 in human myeloma cells: acomparative study with normal plasma cells. Blood1994;84:2597–603.

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 459

Allegra et al. Monoclonal antibodies and multiple myeloma

Page 20: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

65. Rodrigues-Lima F, Josephs M, Katan M, Cassinat B.Sequence analysis identifies TTRAP, a protein that associ-ates with CD40 and TNF receptor-associated factors, as amember of a superfamily of divalent cation-dependentphosphodiesterases. Biochem Biophys Res Commun2001;285:1274–9.

66. Morel Y, Truneh A, Sweet RW, Olive D, Costello RT. TheTNF superfamily members LIGHT and CD154 (CD40ligand) costimulate induction of dendritic cell maturationand elicit specific CTL activity. J Immunol 2001;167:2479–86.

67. Mizuno T, Rothstein TL. B cell receptor (BCR) cross-talk:CD40 engagement creates an alternate pathway for BCRsignaling that activates I kappa B kinase/I kappa B alpha/NF-kappa B without the need for PI3K and phospholipaseC gamma. J Immunol 2005;174:6062–70.

68. Solanilla A, Dechanet J, El Andaloussi A, et al. CD40-ligand stimulates myelopoiesis by regulating flt3-ligand andthrombopoietin production in bone marrow stromal cells.Blood 2000;95:3758–64.

69. Futagami S, Hiratsuka T, Shindo T, et al. COX-2 andCCR2 induced by CD40 ligand and MCP-1 are linked toVEGF production in endothelial cells. Prostaglandins Leu-kot Essent Fatty Acids 2008;78:137–46.

70. Dadgostar H, Zarnegar B, Hoffmann A, Qin XF, Truong U,Rao G, Baltimore D, Cheng G. Cooperation of multiple sig-naling pathways in CD40-regulated gene expression in Blymphocytes. Proc Natl Acad Sci USA 2002;99:1497–502.

71. Younes A, Snell V, Consoli U, Clodi K, Zhao S, PalmerJL, Thomas EK, Armitage RJ, Andreeff M. Elevated levelsof biologically active soluble CD40 ligand in the serum ofpatients with chronic lymphocytic leukaemia. Br J Haematol1998;100:135–41.

72. Advani R, Forero-Torres A, Furman RR, Rosenblatt JD,Younes A, Ren H, Harrop K, Whiting N, Drachman JG.Phase I study of the humanized anti-CD40 monoclonal anti-body dacetuzumab in refractory or recurrent non-Hodgkin’slymphoma. J Clin Oncol 2009;27:4371–7.

73. Tai Y, Li X, Tong X, et al. Human anti-CD40 antagonistantibody triggers significant antitumor activity againsthuman multiple myeloma. Cancer Res 2005;65:5898–906.

74. Long L, Tong X, Patawaran M, Aukerman SL, Jallal B,Luqman M. Antagonist anti-CD40 antibody, CHIR-12.12,induces ADCC, inhibits tumor growth, and prolongs sur-vival in a human multiple myeloma xenograft model. Blood(ASH Annual Meeting Abstracts) 2005;106:3470.

75. Luqman M, Klabunde S, Lin K, et al. The antileukemiaactivity of a human anti-CD40 antagonist antibody,HCD122, on human chronic lymphocytic leukemia cells.Blood 2008;112:711–20.

76. Bensinger W, Maziarz RT, Jagannath S, et al. A phase 1study of lucatumumab, a fully human anti-CD40 antagonistmonoclonal antibody administered intravenously to patientswith relapsed or refractory multiple myeloma. Br J Haema-tol 2012;159:58–66.

77. Law CL, Gordon KA, Collier J, et al. Preclinical antilym-phoma activity of a humanized anti-CD40 monoclonalantibody, SGN-40. Cancer Res 2005;65:8331–8.

78. Lewis TS, McCormick RS, Kissler K, Stone IJ, Jonas M,Sutherland MSK. The humanized anti-CD40 monoclonalantibody, SGN-40, potentiates chemotherapy regimens inNHL Xenograft models via pro-apoptotic signaling. Blood2007;110:2342.

79. Tai YT, Catley LP, Mitsiades CS, et al. Mechanisms bywhich SGN-40, a humanized anti-CD40 antibody, inducescytotoxicity in human multiple myeloma cells: clinicalimplications. Cancer Res 2004;64:2846–52.

80. Bensinger W, Jagannath S, Becker PS, et al. A phase 1dose escalation study of a fully human, antagonist anti-CD40 antibody, HCD 122 (formerly CHIR-12.12), inpatients with relapsed and refractory multiple myeloma.Blood (ASH Annual Meeting Abstracts) 2006;108:3675.

81. Khubchandani S, Czuczman MS, Hernandez-Ilizaliturri FJ.Dacetuzumab, a humanized mAb against CD40 for the treat-ment of hematological malignancies. Curr Opin InvestDrugs 2009;10:579–87.

82. Tai YT, Li XF, Catley L, et al. Immunomodulatory druglenalidomide (CC-5013, IMiD3) augments anti-CD40SGN-40-induced cytotoxicity in human multiple myeloma:clinical implications. Cancer Res 2005;65:11712–20.

83. Hussein M, Berenson JR, Niesvizky R, Munshi N, MatousJ, Sobecks R, Harrop K, Drachman JG, Whiting N. A phaseI multidose study of dacetuzumab (SGN-40; humanizedanti-CD40 monoclonal antibody) in patients with multiplemyeloma. Haematologica 2010;95:845–8.

84. Agura E, Niesvizky R, Matous J. Dacetuzumab (SGN-40),lenalidomide, and weekly dexamethasone in relapsed orrefractory multiple myeloma: multiple responses observed ina phase 1b study. Blood (ASH Annual Meeting Abstracts)2009;114:2870.

85. Tassone P, Gozzini A, Goldmacher V, et al. In vitro and invivo activity of the maytansinoid immunoconjugatehuN901-N2′-deacetyl-N2′-(3-mercapto-1-oxopropyl)-maytan-sine against CD56+ multiple myeloma cells. Cancer Res2004;64:4629–36.

86. Mechtersheimer G, Staudter M, M€oller P. Expression of thenatural killer (NK) cell-associated antigen CD56(Leu-19),which is identical to the 140-kDa isoform of N-CAM, inneural and skeletal muscle cells and tumors derivedtherefrom. Ann N Y Acad Sci 1992;650:311–6.

87. Griffin JD, Hercend T, Beveridge R, Schlossman SF.Characterization of an antigen expressed by human naturalkiller cells. J Immunol 1983;130:2947–51.

88. Lanier LL, Le AM, Civin CI, Loken MR, Phillips JH. Therelationship of CD16 (Leu-11) and Leu-19 (NKH-1) antigenexpression on human peripheral blood NK cells andcytotoxic T lymphocytes. J Immunol 1986;136:4480–6.

89. Mart�õn P, Sant�on A, Bellas C. Neural cell adhesion mole-cule expression in plasma cells in bone marrow biopsiesand aspirates allows discrimination between multiple mye-loma, monoclonal gammopathy of uncertain significance

460 © 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Monoclonal antibodies and multiple myeloma Allegra et al.

Page 21: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

and polyclonal plasmacytosis. Histopathology 2004;44:375–80.

90. Harada H, Kawano MM, Huang N, Harada Y, Iwato K,Tanabe O, Tanaka H, Sakai A, Asaoku H, Kuramoto A.Phenotypic difference of normal plasma cells from maturemyeloma cells. Blood 1993;81:2658–63.

91. Damgaard T, Knudsen LM, Dahl IM, Gimsing P, LodahlM, Rasmussen T. Regulation of the CD56 promoter and itsassociation with proliferation, anti-apoptosis and clinical fac-tors in multiple myeloma. Leuk Lymphoma 2009;50:236–46.

92. Ely SA, Knowles DM. Expression of CD56/neural celladhesion molecule correlates with the presence of lytic bonelesions in multiple myeloma and distinguishes myelomafrom monoclonal gammopathy of undetermined significanceand lymphomas with plasmacytoid differentiation. Am JPathol 2002;160:1293–9.

93. Chanan-Khan AA, Gharibo M, Jagannath S, Munshi NC,Anderson KC, DePaolo D. Phase I study of IMGN901 inpatients with relapsed and relapsed/refractory CD56-positivemultiple myeloma. Blood (ASH Annual Meeting Abstracts)2008;112:3689.

94. Chanan-Khan A, Wolf J, Gharibo M. Phase I study ofIMGN901, used as monotherapy, in patients with heavilypre-treated CD56-positive multiple myeloma—a preliminarysafety and efficacy analysis. Blood (ASH Annual MeetingAbstracts) 2009;114:2883.

95. Lutz R, Ab O, Foley K, Goldmacher V, Whiteman K, XieH. Efficacy of the huN901-DM1 conjugate in combinationwith antineoplastic agents against multiple myeloma cells inpreclinical studies. AACR Meet Abstr 2007;2007:5577.

96. Whiteman K, Ab O, Bartle L, Foley K, Goldmacher V, LutzR. Efficacy of IMGN901 (huN901-DM1) in combinationwith bortezomib and lenalidomide against multiple myelomacells in preclinical studies. AACR Meet Abstr2008;2008:2146.

97. Georgii-Hemming P, Wiklund HJ, Ljunggren O, Nilsson K.Insulin-like growth factor I is a growth and survival factorin human multiple myeloma cell lines. Blood1996;88:2250–8.

98. Jourdan M, De Vos J, Mechti N, Klein B. Regulation ofBcl-2-family proteins in myeloma cells by three myelomasurvival factors: interleukin-6, interferon-alpha and insulin-like growth factor 1. Cell Death Differ 2000;7:1244–52.

99. Jelinek DF, Witzig TE, Arendt BK. A role for insulin-likegrowth factor in the regulation of IL-6-responsive humanmyeloma cell line growth. J Immunol 1997;159:487–96.

100. Heron-Milhavet L, LeRoith D. Insulin-like growth factor Iinduces MDM2- dependent degradation of p53 via the p38MAPK pathway in response to DNA damage. J Biol Chem2002;277:15600.

101. Ogata A, Chauhan D, Teoh G, Treon SP, Urashima M,Schlossman RL, Anderson KC. IL-6 triggers cell growth viathe Ras-dependent mitogen-activated protein kinase cascade.J Immunol 1997;159:2212–21.

102. Pene F, Claessens YE, Muller O, Viguie F, Mayeux P,Lacombe C, Bouscary D. Role of the phosphatidylinositol

3-kinase/Akt and mTOR/P70S6-kinase pathways in the pro-liferation and apoptosis in multiple myeloma. Oncogene2002;21:6587–97.

103. Mitsiades CS, Mitsiades N, Poulaki V, et al. Activation ofNF-jB and upregulation of intracellular anti-apoptoticproteins via the IGF 1/Akt signalling in human multiplemyeloma cells: therapeutic implications. Oncogene2002;21:5673–83.

104. Cohen BD, Baker DA, Soderstrom C, et al. Combinationtherapy enhances the inhibition of tumor growth with thefully human anti-type 1 insulin-like growth factor receptormonoclonal antibody CP-751,871. Clin Cancer Res2005;11:2063–73.

105. Tagoug I, Sauty De Chalon A, Dumonte C. Inhibition ofIGF-1 signalling enhances the apoptotic effect ofAS602868, an IKK2 inhibitor, in multiple myeloma celllines. PLoS ONE 2011;6:e22641.

106. Descamps G, Gomez-Bougie P, Venot C, Moreau P,Bataille R, Amiot M. A humanised anti-IGF-1R monoclonalantibody (AVE1642) enhances bortezomib-induced apopto-sis in myeloma cells lacking CD45. Br J Cancer2009;100:366–9.

107. Moreau P, Cavallo F, Leleu X, Hulin C, Amiot M,Descamps G, Facon T, Boccadoro M, Mignard D,Harousseau JL. Phase I study of the anti insulin-likegrowth factor 1 receptor (IGF-1R) monoclonal antibody,AVE1642, as single agent and in combination with bort-ezomib in patients with relapsed multiple myeloma. Leu-kemia 2011;25:872–4.

108. Tolcher AW, Patnaik A, Till E, Daud AI, Patnaik A,Papadopoulos K, Takimoto C, Bartels P, Keating A, Anto-nia S. A phase I study of AVE1642, a humanized monoclo-nal antibody IGF-1R (insulin like growth factor1 receptor)antagonist, in patients (pts) with advanced solid tumor(ST).J Clin Oncol 2008;26:3582.

109. Rothenberg ML, Poplin E, LoRusso P. Pharmacokinetic(PK) and pharmacodynamic (PD) results of Phase I studiesof IMC-A12, a fully human insulin like growth factor-Ireceptor IgG1 monoclonal antibody, in patients withadvanced solid malignancies. In: 20th EORTC-NCI-AACRSymposium on Molecular Targets and Cancer Therapeutics,21–24 October. Geneva, Switzerland: Pergamon-ElsevierScience Ltd, 2008:174–5.

110. Higano C, LoRusso P, Gordon M. A phase I study of therecombinant human IgG1 anti-IGF-IR monoclonal antibody(Mab) IMC-A12, administered on a weekly basis to patientswith advanced solid tumors: interim analysis. Mol CancerTher 2007;6:3439S.

111. Leong S, Gore L, Benjamin R. A phase I study of R1507, ahuman monoclonal antibody lGF1R (insulin-like growthfactor receptor) antagonist given weekly in patients withadvanced solid tumors. Mol Cancer Ther 2007;6:3360S.

112. Rodon J, Patnaik A, Stein M. A phase I study of q3WR1507, a human monoclonal antibody IGF-1R (insulin-likegrowth factor receptor) antagonist in patients with advancedsolid tumors. Mol Cancer Ther 2007;6:3360S.

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 461

Allegra et al. Monoclonal antibodies and multiple myeloma

Page 22: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

113. Yap TA, Olmos D, Molife LR, de Bono JS. Targeting theinsulin-like growth factor signaling pathway: figitumumaband other novel anticancer strategies. Expert Opin InvestigDrugs 2011;20:1293–304.

114. Lacy MQ, Alsina M, Fonseca R, et al. Phase I, pharmacoki-netic and pharmacodynamic study of the anti-insulinlikegrowth factor type 1 Receptor monoclonal antibodyCP-751,871 in patients with multiple myeloma. J ClinOncol 2008;26:3196–203.

115. Hidalgo M, Tirado Gomez M, Lewis N. A phase I study ofMK-0646, a humanized monoclonal antibody against theinsulin-like growth factor receptor type 1 (IGF1R) inadvanced solid tumor patients in a q2 wk schedule. ASCOMeet Abstr 2008;26:3520.

116. Atzori F, Tabernero J, Cervantes A. A phase I, pharma-cokinetic (PK) and pharmacodynamic (PD) study ofweekly (qW) MK-0646, an insulin-like growth factor-1receptor (IGF1R) monoclonal antibody (MAb) in patients(pts) with advanced solid tumors. J Clin Oncol2008;26:3519.

117. Scartozzi M, Bianconi M, Maccaroni E, Giampieri R,Berardi R, Cascinu S. Dalotuzumab, a recombinant humanizedmAb targeted against IGFR1 for the treatment of cancer.Curr Opin Mol Ther 2010;12:361–71.

118. Ohtomo T, Sugamata Y, Ozaki Y, et al. Molecular cloningand characterization of a surface antigen preferentially over-expressed on multiple myeloma cells. Biochem Biophys ResCommun 1999;258:583–91.

119. Ozaki S, Kosaka M, Wakatsuki S, Abe M, Koishihara Y,Matsumoto T. Immunotherapy of multiple myeloma with amonoclonal antibody directed against a plasma cell-specificantigen, HM1.24. Blood 1997;90:3179–86.

120. Wang W, Nishioka Y, Ozaki S, Jalili A, Abe S, KakiuchiS, Kishuku M, Minakuchi K, Matsumoto T, Sone S.HM1.24 (CD317) is a novel target against lung cancer forimmunotherapy using anti-HM1.24 antibody. CancerImmunol Immunother 2009;58:967–76.

121. Kawai S, Yoshimura Y, Iida S, Kinoshita Y, Koishihara Y,Ozaki S, Matsumoto T, Kosaka M, Yamada-Okabe H. Anti-tumor activity of humanized monoclonal antibody againstHM1.24 antigen in human myeloma xenograft models.Oncol Rep 2006;15:361–7.

122. Ono K, Ohtomo T, Yoshida K, Yoshimura Y, Kawai S,Koishihara Y, Ozaki S, Kosaka M, Tsuchiya M. Thehumanized anti-HM1.24 antibody effectively kills multiplemyeloma cells by human effector cell-mediated cytotoxicity.Mol Immunol 1999;36:387–95.

123. Ozaki S, Kosaka M, Wakahara Y, Ozaki Y, Tsuchiya M,Koishihara Y, Goto T, Matsumoto T. Humanized anti-HM1.24 antibody mediates myeloma cell cytotoxicity thatis enhanced by cytokine stimulation of effector cells. Blood1999;93:3922–30.

124. Ishiguro T, Kawai S, Habu K, Sugimoto M, Shiraiwa H,Iijima S, Ozaki S, Matsumoto T, Yamada-Okabe H. Adefucosylated anti-CD317 antibody exhibited enhanced anti-body-dependent cellular cytotoxicity against primary

myeloma cells in the presence of effectors from patients.Cancer Sci 2010;101:2227–33.

125. Tai YT, Muchhal U, Li X, et al. XmAb�5592Fc-engineered humanized anti-HM1.24 monoclonal antibodyhas potent in vitro and in vivo efficacy against multiplemyeloma. Blood (ASH Annual Meeting Abstracts)2009;114:609.

126. Tai YT, Horton HM, Kong SY, et al. Potent in vitro and invivo activity of an Fc-engineered humanized anti-HM1.24antibody against multiple myeloma via augmented effectorfunction. Blood 2012;119:2074–82.

127. Amano J, Masuyama N, Hirota Y, Tanaka Y, Igawa Y,Shiokawa R, Okutani T, Miyayama T, Nanami M, IshigaiM. Antigen-dependent internalization is related to rapidelimination from plasma of humanized anti-HM1.24 mono-clonal antibody. Drug Metab Dispos 2010;38:2339–46.

128. Vaughan HA, Thompson CH, Sparrow RL, McKenzie IF.Hu Ly-M3-a human leukocyte antigen. Transplantation1983;36:446–50.

129. Hosen N, Ichihara H, Mugitani A, et al. CD48 as a novelmolecular target for antibody therapy in multiple myeloma.Br J Haematol 2012;156:213–24.

130. Bjorkman PJ, Burmeister WP. Structures of two classes ofMHC molecules elucidated: crucial differences and similari-ties. Curr Opin Struct Biol 1994;4:852–6.

131. Bataille R, Durie BG, Grenier J. Serum beta2 microglobulinand survival duration in multiple myeloma: a simple reliablemarker for staging. Br J Haematol 1983;55:439–47.

132. Alexanian R, Barlogie B, Fritsche H. Beta 2 microglobulinin multiple myeloma. Am J Hematol 1985;20:345–51.

133. Yang J, Qian J, Wezeman M, Wang S, Lin P, Wang M,Yaccoby S, Kwak LW, Barlogie B, Yi Q. Targeting beta2-microglobulin for induction of tumor apoptosis in humanhematological malignancies. Cancer Cell 2006;10:295–307.

134. Skov S, Nielsen M, Bregenholt S, Odum N, Claesson MH.Activation of Stat-3 is involved in the induction of apopto-sis after ligation of major histocompatibility complex class Imolecules on human JurkatT cells. Blood 1998;91:3566–73.

135. Pedersen AE, Skov S, Bregenholt S, Ruhwald M, ClaessonMH. Signal transduction by the major histocompatibilitycomplex class I molecule. APMIS 1999;107:887–95.

136. Gur H, Geppert TD, Lipsky PE. Structural analysis of classI MHC molecules: the cytoplasmic domain is not requiredfor cytoskeletal association, aggregation and internalization.Mol Immunol 1997;34:125–32.

137. Gur H, Geppert TD, Wacholtz MC, Lipsky PE. The cyto-plasmic and the transmembrane domains are not sufficientfor class I MHC signal transduction. Cell Immunol1999;191:105–16.

138. Yang J, Yi Q. Therapeutic monoclonal antibodies for multi-ple myeloma: an update and future perspectives. Am JBlood Res 2011;1:22–33.

139. Yi Q. Novel immunotherapies. Cancer J 2009;15:502–10.140. Cao Y, Lan Y, Qian J, et al. Targeting cell surface

b2-microglobulin by pentameric IgM antibodies.Br J Haematol 2011;154:111–21.

462 © 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Monoclonal antibodies and multiple myeloma Allegra et al.

Page 23: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

141. van Gisbergen KP, van Olffen RW, van Beek J, van derSluijs KF, Arens R, Nolte MA, van Lier RA. ProtectiveCD8 T cell memory is impaired during chronic CD70-driven costimulation. J Immunol 2009;182:5352–62.

142. Arens R, Tesselaar K, Baars PA, van Schijndel GM,Hendriks J, Pals ST, Krimpenfort P, Borst J, van Oers MH,van Lier RA. Constitutive CD27/CD70 interaction inducesexpansion of effector-type T cells and results in IFNgamma-mediated B cell depletion. Immunity 2001;15:801–12.

143. McEarchern JA, Oflazoglu E, Francisco L, et al. Engineeredanti-CD70 antibody with multiple effector functions exhibitsin vitro and in vivo antitumor activities. Blood2007;109:1185–92.

144. McEarchern JA, Smith LM, McDonagh CF, et al. Preclin-ical characterization of SGN-70, a humanized antibodydirected against CD70. Clin Cancer Res 2008;14:7763–72.

145. Long EO, Strubin M, Wake CT, Gross N, Carrel S, Good-fellow P, Accolla RS, Mach B. Isolation of cDNA clonesfor the p33 invariant chain associated with HLA-DRantigens. Proc Natl Acad Sci USA 1983;80:5714–8.

146. Faure-Andre G, Vargas P, Yuseff MI, et al. Regulation ofdendritic cell migration by CD74, the MHC class II-associ-ated invariant chain. Science 2008;322:1705–10.

147. Veillat V, Carli C, Metz CN, Al-Abed Y, Naccache PH,Akoum A. Macrophage migration inhibitory factor elicits anangiogenic phenotype in human ectopic endometrial cellsand triggers the production of major angiogenic factors viaCD44, CD74, and MAPK signaling pathways. J Clin Endo-crinol Metab 2010;95:E403–12.

148. Burton JD, Ely S, Reddy PK, Stein R, Gold DV, CardilloTM, Goldenberg DM. CD74 is expressed by multiple mye-loma and is a promising target for therapy. Clin Cancer Res2004;10:6606–11.

149. Stein R, Qu Z, Cardillo TM, Chen S, Rosario A, Horak ID,Hansen HJ, Goldenberg DM. Antiproliferative activity of ahumanized anti-CD74 monoclonal antibody, hLL1, onB-cell malignancies. Blood 2004;104:3705–11.

150. Stein R, Smith MR, Chen S, Zalath M, Goldenberg DM.Combining milatuzumab with bortezomib, doxorubicin, ordexamethasone improves responses in multiple myelomacell lines. Clin Cancer Res 2009;15:2808–17.

151. Berkova Z, Tao RH, Samaniego F. Milatuzumab-a promis-ing new immunotherapeutic agent. Expert Opin InvestigDrugs 2010;19:141–9.

152. Kaufman JNR, Stadtmauer E, Chanan-Khan A. First trial ofhumanized anti-CD-74 monoclonal antibody (Mab), mila-tuzumab in multiple myeloma. Blood 2008;112:1266–7.

153. Carlo-Stella C, Guidetti A, Di Nicola M, et al. IFN-cenhances the antimyeloma activity of the fully human anti--human leukocyte antigen-DR monoclonal antibody1D09C3. Cancer Res 2007;67:3269–75.

154. Sekimoto E, Ozaki S, Ohshima T, et al. A single-chain Fvdiabody against human leukocyte antigen—a molecules spe-cifically induces myeloma cell death in the bone marrowenvironment. Cancer Res 2007;67:1184–92.

155. Johansson SE, Hejdeman B, Hinkula J, Johansson MH,Romagn�e F, Wahren B, Wagtmann NR, K€arre K, Berg L. NKcell activation by KIR-binding antibody 1-7F9 and responseto HIV-infected autologous cells in viremic and controllerHIV-infected patients. Clin Immunol 2010;134:158–68.

156. Romagne F, Andre P, Spee P, et al. Preclinical characteriza-tion of 1-7F9, a novel human anti-KIR receptor therapeuticantibody that augments natural killer-mediated killing oftumor cells. Blood 2009;114:2667–77.

157. Benson DM, Romagne F, Squiban P. Novel monoclonalantibody that enhances natural killer (NK) cell cytotoxicityagainst multiple myeloma (MM): preclinical data andinterim phase I clinical trial results. J Clin Oncol2009;27:3032.

158. Campbell KS, Purdy AK. Structure/function of human killercell immunoglobulin-like receptors: lessons frompolymorphisms, evolution, crystal structures and mutations.Immunology 2011;132:315–25.

159. Kimura N, Kawai S, Kinoshita Y, et al. 2D7 diabody boundto the alpha2 domain of HLA class I efficiently induces cas-pase-independent cell death against malignant and activatedlymphoid cells. Biochem Biophys Res Commun2004;325:1201–9.

160. Sandrin MS, Henning MM, Lo MF, Baker E, SutherlandGR, McKenzie IF. Isolation and characterization of cDNAclones for Humly9: the human homologue of mouse Ly9.Immunogenetics 1996;43:13–9.

161. Tangye SG, Phillips JH, Lanier LL. The CD2-subset of theIg superfamily of cell surface molecules: receptor-ligandpairs expressed by NK cells and other immune cells. SeminImmunol 2000;12:149–57.

162. Martin M, Del Valle JM, Saborit I, Engel P. Identificationof Grb2 as a novel binding partner of the signaling lympho-cytic activation molecule-associated protein binding receptorCD229. J Immunol 2005;174:5977–86.

163. Atanackovic D, Panse J, Hildebrandt Y, et al. Surfacemolecule CD229 as a novel target for the diagnosis andtreatment of multiple myeloma. Haematologica 2011;96:1512–20.

164. Ishii T, Chanan-Khan A, Jafferjee J, Ersing N, TakahashiH, Mizutani M, Shiotsu Y, Hanai N. A humanized anti-gan-glioside GM2 antibody, BIW-8962, exhibits ADCC/CDCactivity against multiple myeloma cells and potent anti-tumor activity in mouse xenograft models. Blood (ASHAnnual Meeting Abstracts) 2008;112:1718.

165. Gorczynski RM, Cohen Z, Fu XM, Lei J. Anti-rat OX-2blocks increased small intestinal transplant survival afterportal vein immunization. Transplant Proc 1999;31:577–8.

166. Moreaux J, Hose D, Reme T, et al. CD200 is a new prog-nostic factor in multiple myeloma. Blood 2006;108:4194–7.

167. Zheng Y, Yang J, Qian J, et al. PSGL-1/selectin andICAM-1/CD18 interactions are involved in macrophage-induced drug resistance in myeloma. Leukemia. 2013;27:702–10.

168. Schmidmaier R, Morsdorf K, Baumann P, Emmerich B,Meinhardt G. Evidence for cell adhesion-mediated drug

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 463

Allegra et al. Monoclonal antibodies and multiple myeloma

Page 24: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

resistance of multiple myeloma cells in vivo. Int J BiolMarkers 2006;21:218–22.

169. Downs JA, Jackson SP. A means to a DNA end: the manyroles of Ku. Nat Rev Mol Cell Biol 2004;5:367–77.

170. Difilippantonio MJ, Zhu J, Chen HT, Meffre E,Nussenzweig MC, Max EE, Ried T, Nussenzweig A.DNA repair protein Ku80 suppresses chromosomal aberra-tions and malignant transformation. Nature 2000;404:510–4.

171. Gao Y, Ferguson DO, Xie W, Manis JP, Sekiguchi J, FrankKM, Chaudhuri J, Horner J, DePinho RA, Alt FW. Inter-play of p53 and DNA-repair protein XRCC4 in tumorigene-sis, genomic stability and development. Nature2000;404:897–900.

172. Khanna KK, Jackson SP. DNA double-strand breaks:signaling, repair and the cancer connection. Nat Genet2001;27:247–54.

173. Gullo C, Au M, Feng G, Teoh G. The biology of Ku andits potential oncogenic role in cancer. Biochim Biophys Acta2006;1765:223–34.

174. Tuteja R, Tuteja N. Ku autoantigen: a multifunctionalDNA-binding protein. Crit Rev Biochem Mol Biol2000;35:1–33.

175. Tai YT, Podar K, Kraeft SK, Wang F, Young G, Lin B,Gupta D, Chen LB, Anderson KC. Translocation of Ku86/Ku70 to the multiple myeloma cell membrane: functionalimplications. Exp Hematol 2002;30:212–20.

176. Teoh G, Urashima M, Greenfield EA, Nguyen KA, Lee JF,Chauhan D, Ogata A, Treon SP, Anderson KC. The 86-kDsubunit of Ku autoantigen mediates homotypic and hetero-typic adhesion of multiple myeloma cells. J Clin Invest1998;101:1379–88.

177. Liew PX, Ge F, Gullo C, Teoh GKH, Hwang WYK. Useof phage display to isolate specific human monoclonal anti-body fragments against a potential target for multiple mye-loma. Ann Acad Med Singapore 2009;38:621–9.

178. Klein B, Zhang XG, Jourdan M, Content J, Houssiau F,Aarden L, Piechaczyk M, Bataille R. Paracrine rather thanautocrine regulation of myeloma-cell growth and differentia-tion by interleukin-6. Blood 1989;73:517–26.

179. Podar K, Gouill SL, Zhang J, et al. A pivotal role for Mcl-1 in Bortezomib-induced apoptosis. Oncogene2008;27:721–31.

180. Voorhees PM, Chen Q, Kuhn DJ, Small GW, HunsuckerSA, Strader JS, Corringham RE, Zaki MH, Nemeth JA,Orlowski RZ. Inhibition of interleukin-6 signaling withCNTO 328 enhances the activity of bortezomib in preclini-cal models of multiple myeloma. Clin Cancer Res2007;13:6469–78.

181. Mundy GR, Raisz LG, Cooper RA, Schechter GP, SalmonSE. Evidence for the secretion of an osteoclast stimulatingfactor in myeloma. N Engl J Med 1974;291:1041–6.

182. Kurihara N, Bertolini D, Suda T, Akiyama Y, RoodmanGD. IL-6 stimulates osteoclast-like multinucleated cell for-mation in long term human marrow cultures by inducingIL-1 release. J Immunol 1990;144:4226–30.

183. Hideshima T, Mitsiades C, Tonon G, Richardson PG,Anderson KC. Understanding multiple myeloma pathogene-sis in the bone marrow to identify new therapeutic targets.Nat Rev Cancer 2007;7:585–98.

184. Greipp PR, Leong T, Bennett JM, Gaillard JP, Klein B,Stewart JA, Oken MM, Kay NE, Van Ness B, Kyle RA.Plasmablastic morphology – an independent prognostic fac-tor with clinical and laboratory correlates: Eastern Coopera-tive Oncology Group (ECOG) myeloma trial E9486 reportby the ECOG Myeloma Laboratory Group. Blood1998;91:2501–7.

185. Stasi R, Brunetti M, Parma A, Di Giulio C, Terzoli E,Pagano A. The prognostic value of soluble interleukin-6receptor in patients with multiple myeloma. Cancer1998;82:1860–6.

186. Wierzbowska A, Urbanska-Rys H, Robak T. CirculatingIL-6-type cytokines and sIL-6R in patients with multiplemyeloma. Br J Haematol 1999;105:412–9.

187. Guo Y, Xu F, Lu T, Duan Z, Zhang Z. Interleukin-6 signal-ing pathway in targeted therapy for cancer. Cancer TreatRev 2012;38:904–10.

188. van Zaanen HC, Lokhorst HM, Aarden LA, Rensink HJ,Warnaar SO, van der Lelie J, van Oers MH. Chimaericanti-interleukin 6 monoclonal antibodies in the treatment ofadvanced multiple myeloma: a phase I dose-escalatingstudy. Br J Haematol 1998;102:783–90.

189. Trikha M, Corringham R, Klein B, Rossi JF. Targeted anti-interleukin-6 monoclonal antibody therapy for cancer: areview of the rationale and clinical evidence. Clin CancerRes 2003;9:4653–65.

190. Gu ZJ, De Vos J, Rebouissou C, Jourdan M, Zhang XG,Rossi JF, Wijdenes J, Klein B. Agonist anti-gp130 trans-ducer monoclonal antibodies are human myeloma cellsurvival and growth factors. Leukemia 2000;14:188–97.

191. Rossi JF, Manges RF, Sutherland HJ, et al. Preliminaryresults of CNTO 328, an anti-interleukin-6 monoclonal anti-body, in combination with bortezomib in the treatment ofrelapsed or refractory multiple myeloma. Blood (ASHAnnual Meeting Abstracts) 2008;112:867.

192. Voorhees PM, Chen Q, Small GW, Kuhn DJ, HunsuckerSA, Nemeth JA, Orlowski RZ. Targeted inhibition of inter-leukin-6 with CNTO 328 sensitizes pre-clinical models ofmultiple myeloma to dexamethasone-mediated cell death. BrJ Haematol 2009;145:481–90.

193. van Zaanen HCT, Lokhorst HM, Aarden LA, RensinkHJAM, Warnaar SO, van Oers MHJ. Blocking interleukin-6activity with chimeric anti-IL6 monoclonal antibodies inmultiple myeloma: effects on soluble IL6 receptor and solu-ble gp130. Leuk Lymphoma 1998;3:551–8.

194. Hunsucker SA, Magarotto V, Kuhn DJ, et al. Blockade ofinterleukin-6 signalling with siltuximab enhances melphalancytotoxicity in preclinical models of multiple myeloma. BrJ Haematol 2011;152:579–92.

195. Hirata T, Shimazaki C, Sumikuma T, Ashihara E, Goto H,Inaba T, Koishihara Y, Nakagawa M. Humanized anti-inter-leukin-6 receptor monoclonal antibody induced apoptosis of

464 © 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Monoclonal antibodies and multiple myeloma Allegra et al.

Page 25: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

fresh and cloned human myeloma cells in vitro. Leuk Res2003;27:343–9.

196. Nishimoto N, Shima Y, Sasai M, Danno N, Yoshizaki K.Clinical application of interleukin-6 receptor antibody.Nihon Rinsho Meneki Gakkai Kaishi 1997;20:87–94. (InJapanese, Abstract in English.)

197. Matsuyama Y, Nagashima T, Honne K, Kamata Y, Iwamot-o M, Okazaki H, Sato K, Ozawa K, Minota S. Successfultreatment of a patient with rheumatoid arthritis and IgA-kappa multiple myeloma with tocilizumab. Intern Med2011;50:639–42.

198. Yoshio-Hoshino N, Adachi Y, Aoki C, Pereboev A, CurielDT, Nishimoto N. Establishment of a new interleukin-6 (IL-6) receptor inhibitor applicable to the gene therapy for IL-6-dependent tumor. Cancer Res 2007;67:871–5.

199. Bataille R, Barlogie B, Lu ZY, Rossi JF, Lavabre-BertrandT, Beck T, Wijdenes J, Brochier J, Klein B. Biologic effectsof anti-interleukin-6 murine monoclonal antibody inadvanced multiple myeloma. Blood 1995;86:685–91.

200. Emilie D, Wijdenes J, Gisselbrecht C, Jarrousse B, BillaudE, Blay JY, Gabarre J, Gaillard JP, Brochier J, Raphael M.Administration of an anti-interleukin-6 monoclonal antibodyto patients with acquired immunodeficiency syndrome andlymphoma: effect on lymphoma growth and on B clinicalsymptoms. Blood 1994;84:2472–9.

201. Rossi JF, Fegueux N, Lu ZY, et al. Optimizing the use ofanti-interleukin-6 monoclonal antibody with dexamethasoneand 140 mg/m2 of melphalan in multiple myeloma: resultsof a pilot study including biological aspects. Bone MarrowTransplant 2005;36:771–9.

202. Moreau P, Harousseau JL, Wijdenes J, Morineau N, MilpiedN, Bataille R. A combination of anti-interleukin 6 murinemonoclonal antibody with dexamethasone and high-dosemelphalan induces high complete response rates in advancedmultiple myeloma. Br J Haematol 2000;109:661–4.

203. Fulciniti M, Hideshima T, Vermot-Desroches C, et al. Ahigh-affinity fully human anti-IL-6 mAb (OP-R003-1, 1339)for the treatment of Multiple Myeloma. Clin Cancer Res2009;15:7144–52.

204. Honemann D, Chatterjee M, Savino R, Bommert K, BurgerR, Gramatzki M, D€orken B, Bargou RC. The IL-6 receptorantagonist SANT-7 overcomes bone marrow stromal cell-mediated drug resistance of multiple myeloma cells. Int JCancer 2001;93:674–80.

205. Tassone P, Forciniti S, Galea E, Savino R, Turco MC,Iacopino P, Tagliaferri P, Morrone G, Ciliberto G, VenutaS. Synergistic induction of growth arrest and apoptosis ofhuman myeloma cells by the IL-6 super-antagonist Sant7and Dexamethasone. Cell Death Differ 2000;7:327–8.

206. Tassone P, Galea E, Forciniti S, Tagliaferri P, Venuta S.The IL-6 receptor super-antagonist Sant7 enhances antipro-liferative and apoptotic effects induced by dexamethasoneand zoledronic acid on multiple myeloma cells. Int J Oncol2002;2:867–73.

207. Ribatti D, Vacca A. The role of microenvironment in tumorangiogenesis. Genes Nutr 2008;3:29–34.

208. Podar K, Hideshima T, Chauhan D, Anderson KC. Target-ing signalling pathways for the treatment of multiplemyeloma. Expert Opin Ther Targets 2005;9:359–81.

209. Jakob C, Sterz J, Zavrski I, Heider U, Kleeberg L, FleissnerC, Kaiser M, Sezer O. Angiogenesis in multiple myeloma.Eur J Cancer 2006;42:1581–90.

210. Fan F, Schimming A, Jaeger D, Podar K. Targeting thetumor microenvironment: focus on angiogenesis. J Oncol2012;2012:281261.

211. Ferrara N, Hillan KJ, Novotny W. Bevacizumab (Avastin),a humanized anti-VEGF monoclonal antibody for cancertherapy. Biochem Biophys Res Commun 2005;333:328–35.

212. Gorski DH, Beckett MA, Jaskowiak NT, et al. Blockade ofthe vascular endothelial growth factor stress responseincreases the antitumor effects of ionizing radiation. CancerRes 1999;59:3374–8.

213. Hoyer RJ, Leung N, Witzig TE, Lacy MQ. Treatment ofdiuretic refractory pleural effusions with bevacizumab infour patients with primary systemic amyloidosis. Am JHematol 2007;82:409–13.

214. Goldman B. For investigational targeted drugs, combinationtrials pose challenges. J Natl Cancer Inst 2003;95:1744–6.

215. Attar-Schneider O, Drucker L, Zismanov V, Tartakover-Matalon S, Rashid G, Lishner M. Bevacizumab attenuatesmajor signaling cascades and eIF4E translation initiationfactor in multiple myeloma cells. Lab Invest 2012;92:178–90.

216. Somlo G, Lashkari A, Bellamy W, et al. Phase II random-ized trial of bevacizumab versus bevacizumab andthalidomide for relapsed/refractory multiple myeloma: aCalifornia Cancer Consortium trial. Br J Haematol2011;154:533–5.

217. Martinelli E, De Palma R, Orditura M, De Vita F, Ciardi-ello F. Anti-epidermal growth factor receptor monoclonalantibodies in cancer therapy. Clin Exp Immunol2009;158:1–9.

218. Mendelsohn J, Baselga J. Epidermal growth factor receptortargeting in cancer. Semin Oncol 2006;33:369–85.

219. Harding J, Burtness B. Cetuximab: an epidermal growthfactor receptor chimeric human-murine monoclonalantibody. Drugs Today 2005;41:107–27.

220. Bonner JA, Harari PM, Giralt J, et al. Radiotherapy pluscetuximab for squamous-cell carcinoma of the head andneck. N Engl J Med 2006;354:567–78.

221. VanCutsem E, Kohne CH, Hitre E, et al. Cetuximab andchemotherapy as initial treatment for metastatic colorectalcancer. N Engl J Med 2009;360:1408–17.

222. Mahtouk K, Hose D, Reme T, et al. Expression of EGF-family receptors and amphiregulin in multiple myeloma.Amphiregulin is a growth factor for myeloma cells.Oncogene 2005;24:3512–24.

223. Mahtouk K, Jourdan M, De Vos J, Hertogh C, Fiol G,Jourdan E, Rossi JF, Klein B. An inhibitor of the EGFreceptor family blocks myeloma cell growth factor activityof HB-EGF and potentiates dexamethasone or anti- IL-6antibody-induced apoptosis. Blood 2004;103:1829–37.

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 465

Allegra et al. Monoclonal antibodies and multiple myeloma

Page 26: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

224. B€oll B, Eichenauer DA, Von Tresckow B, Peine D, HallekM, Engert A, H€ubel K. Activity of cetuximab as singleagent in a patient with relapsed multiple myeloma. LeukLymphoma 2010;51:562–4.

225. Chapman MA, Lawrence MS, Keats JJ, et al. Initial gen-ome sequencing and analysis of multiple myeloma. Nature2011;471:467–72.

226. Brito JL, Walker B, Jenner M, et al. MMSET deregulationaffects cell cycle progression and adhesion regulons in t(4;14) myeloma plasma cells. Haematologica 2009;94:78–86.

227. Morgan GJ, Kaiser MF. How to use new biology to guidetherapy in multiple myeloma. Hematology Am Soc HematolEduc Program 2012;2012:342–9.

228. Kamath AV, Lu D, Gupta P, et al. Preclinical pharmacoki-netics of MFGR1877A, a human monoclonal antibody toFGFR3, and prediction of its efficacious clinical dose forthe treatment of t(4;14)-positive multiple myeloma. CancerChemother Pharmacol 2012;69:1071–8.

229. Burkiewicz JS, Scarpace SL, Bruce SP. Denosumab in oste-oporosis and oncology. Ann Pharmacother 2009;43:1445–55.

230. Smith MR, Saad F, Coleman R, et al. Denosumab andbone-metastasis-free survival in men with castration-resistantprostate cancer: results of a phase 3, randomised, placebo-controlled trial. Lancet 2012;379:39–46.

231. Fizazi K, Carducci M, Smith M, et al. Denosumab versuszoledronic acid for treatment of bone metastases in menwith castration-resistant prostate cancer: a randomised, dou-ble-blind study. Lancet 2011;3779768:813–22.

232. Body JJ, Facon T, Coleman RE, Lipton A, Geurs F, FanM, Holloway D, Peterson MC, Bekker PJ. A study of thebiological receptor activator of nuclear factor-jB ligandinhibitor, denosumab, in patients with multiple myeloma orbone metastases from breast cancer. Clin Cancer Res2006;12:1221–8.

233. Vij R, Horvath N, Spencer A, Taylor K, Vadhan-Raj S,Vescio R, Smith J, Qian Y, Yeh H, Jun S. An open-label,phase 2 trial of denosumab in the treatment of relapsed orplateau-phase multiple myeloma. Am J Hematol2009;8410:650–6.

234. Aghaloo TL, Felsenfeld AL, Tetradis S. Osteonecrosis ofthe jaw in a patient on Denosumab. J Oral Maxillofac Surg2010;685:959–63.

235. Santini D, Fratto ME, Vincenzi B, Napoli N, Galluzzo S,Tantardini M, Abbruzzese A, Caraglia M, Tonini G. Deno-sumab: the era of targeted therapies in bone metastaticdiseases. Curr Cancer Drug Targets 2009;9:834–42.

236. Wang Y, Lin B. In silico investigations of the anti-cataboliceffects of pamidronate and denosumab on multiplemyeloma-induced bone disease. PLoS ONE 2012;7:e44868.

237. Pinzone JJ, Hall BM, Thudi NK, Vonau M, Qiang YW,Rosol TJ, Shaughnessy JD Jr. The role of Dickkopf-1 inbone development, homeostasis, and disease. Blood2009;113:517–25.

238. Yaccoby S, Ling W, Zhan F, Walker R, Barlogie B,Shaughnessy JD. Antibody-based inhibition of DKK1 sup-presses tumor-induced bone resorption and multiple mye-loma growth in vivo. Blood 2007;109:2106–11.

239. Heath DJ, Chantry AD, Buckle CH, Coulton L,Shaughnessy JD Jr, Evans HR, Snowden JA, Stover DR,Vanderkerken K, Croucher PI. Inhibiting dickkopf-1 (Dkkl)removes suppression of bone formation and prevents thedevelopment of osteolytic bone disease in multiplemyeloma. J Bone Miner Res 2009;24:425–36.

240. Fulciniti M, Tassone P, Hideshima T, et al. Anti-DKK1mAb (BHQ880) as a potential therapeutic agent for multiplemyeloma. Blood 2009;114:371–9.

241. Qian J, Zheng Y, Zheng C, et al. Active vaccination withDickkopf-1 induces protective and therapeutic antitumorimmunity in murine multiple myeloma. Blood2012;119:161–9.

242. Woodruff TK. Regulation of cellular and system functionby activin. Biochem Pharmacol 1998;55:953–63.

243. Fuller K, Bayley KE, Chambers TJ. Activin A is an essen-tial cofactor for osteoclast induction. Biochem Biophys ResCommun 2000;268:2–7.

244. Vallet S, Mukherjee S, Vaghela N, et al. Activin A pro-motes multiple myeloma-induced osteolysis and is a promis-ing target for myeloma bone disease. Proc Natl Acad SciUSA 2010;107:5124–9.

245. Abdulkadyrov KM, Salogub GN, Khuazheva NK. ACE-011, a soluble activin receptor type Iia IgG-Fc fusion pro-tein, increases hemoglobin (Hb) and improves bone lesionsin multiple myeloma patients receiving myelosuppressivechemotherapy: preliminary analysis. Blood (ASH AnnualMeeting Abstracts) 2009;114:749.

246. Novak AJ, Darce JR, Arendt BK, Harder B, Henderson K,Kindsvogel W, Gross JA, Greipp PR, Jelinek DF. Expres-sion of BCMA, TACI, and BAFF-R in multiple myeloma: amechanism for growth and survival. Blood 2004;103:689–94.

247. Moreaux J, Cremer FW, Reme T, et al. The level of TACIgene expression in myeloma cells is associated with a signa-ture of microenvironment dependence versus a plasmablas-tic signature. Blood 2005;106:1021–30.

248. Neri P, Kumar S, Fulciniti MT, et al. Neutralizing B-cellactivating factor antibody improves survival and inhibitsosteoclastogenesis in a severe combined immunodeficienthuman multiple myeloma model. Clin Cancer Res2007;13:5903–9.

249. Moreaux J, Legouffe E, Jourdan E, Quittet P, Reme T,Lugagne C, Moine P, Rossi JF, Klein B, Tarte K. BAFFand APRIL protect myeloma cells from apoptosis inducedby interleukin 6 deprivation and dexamethasone. Blood2004;103:3148–57.

250. Tai YT, Li XF, Breitkreutz I, et al. Role of B-cell-activatingfactor in adhesion and growth of human multiple myelomacells in the bone marrow microenvironment. Cancer Res2006;66:6675–82.

466 © 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Monoclonal antibodies and multiple myeloma Allegra et al.

Page 27: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

251. Rossi JF, Moreaux J, Hose D, et al. Atacicept in relapsed/refractory multiple myeloma or active Waldenstrom’s mac-roglobulinemia: a phase I study. Br J Cancer2009;101:1051–8.

252. Rossi JF, Phase I. study of atacicept in relapsed/refractorymultiple myeloma (MM) and Waldenstr€om’s macroglobulin-emia. Clin Lymphoma Myeloma Leuk 2011;11:136–8.

253. Yaccoby S, Pennisi A, Li X, Dillon SR, Zhan F, BarlogieB, Shaughnessy JD Jr. Atacicept (TACI-Ig) inhibits growthof TACI primary myeloma cells in SCID-hu mice and incoculture with osteoclasts. Leukemia 2008;22:406–13.

254. Ryan MC, Hering M, Peckham D, McDonagh CF, BrownL, Kim KM, Meyer DL, Zabinski RF, Grewal IS, Carter PJ.Antibody targeting of B-cell maturation antigen on malig-nant plasma cells. Mol Cancer Ther 2007;6:3009–18.

255. Kimberley FC, Screaton GR. Following a TRAIL: updateon a ligand and its five receptors. Cell Res 2004;14:359–72.

256. Bouralexis S, Findlay DM, Evdokiou A. Death to the badguys: targeting cancer via APO2L/TRAIL. Apoptosis2005;10:35–51.

257. Van Geelen CM, de Vries EG, de Jong S. Lessons fromTRAIL-resistance mechanisms in colorectal cancer cells:paving the road to patient-tailored therapy. Drug ResistUpdat 2004;7:345–58.

258. Pukac L, Kanakaraj P, Humphreys R, et al. HGS-ETR1, afully human TRAIL-receptor 1 monoclonal antibody,induces cell death in multiple tumour types in vitro and invivo. Br J Cancer 2005;92:1430–41.

259. Menoret E, Gomez-Bougie P, Geffroy-Luseau A, Daniels S,Moreau P, Le Gouill S, Harousseau JL, Bataille R, AmiotM, Pellat-Deceunynck C. Mcl-1L cleavage is involved inTRAIL-R1- and TRAIL-R2-mediated apoptosis induced byHGS-ETR1 and HGS-ETR2 human mAbs in myelomacells. Blood 2006;108:1346–52.

260. Locklin RM, Croucher PI, Russell RG, Edwards CM.Agonists of TRAIL death receptors induce myeloma cellapoptosis that is not prevented by cells of the bone marrowmicroenvironment. Leukemia 2007;21:805–12.

261. Kabore AF, Sun J, Hu X, McCrea K, Johnston JB, GibsonSB. The TRAIL apoptotic pathway mediates proteasomeinhibitor induced apoptosis in primary chronic lymphocyticleukemia cells. Apoptosis 2006;11:1175–93.

262. Smith MR, Jin F, Joshi I. Bortezomib sensitizes non-Hodgkin’s lymphoma cells to apoptosis induced by antibod-ies to tumor necrosis factor-related apoptosis-inducingligand (TRAIL) receptors TRAIL-R1 and TRAIL-R2. ClinCancer Res 2007;13:5528s–34s.

263. Ghoshal P, Chitta K, Vujcic S, Gaddy J, Miles KM, SteinL. Mapatumumab, a TRAIL receptor 1 agonist antibody,induces apoptosis in bortezomib resistant multiple myeloma.Blood (ASH Annual Meeting Abstracts) 2009;114:2832.

264. Belch A, Sharma A, Spencer A, Tarantolo S, Bahlis NJ,Doval D. A multicenter randomized phase II trial of map-atumumab, a TRAIL-R1 agonist monoclonal antibody, incombination with bortezomib in patients with relapsed/

refractory multiple myeloma (MM). Blood (ASH AnnualMeeting Abstracts) 2010;116:2321.

265. Greenwald RJ, Freeman GJ, Sharpe AH. The B7 familyrevised. Annu Rev Immunol 2005;23:515–48.

266. Liu J, Hamrouni A, Wolowiec D, Coiteux V, KuliczkowskiK, Hetuin D, Saudemont A, Quesnel B. Plasma cells frommultiple myeloma patients express B7–H1 (PD-L1) andincrease expression after stimulation with IFN-{gamma}and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood 2007;110:296–304.

267. Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, MinatoN. Involvement of PD-L1 on tumor cells in the escape fromhost immune system and tumor immunotherapy by PD-L1blockade. Proc Natl Acad Sci USA 2002;99:12293–7.

268. Rosenblatt J, Glotzbecker B, Mills H, et al. CT-011, anti-PD-1 antibody, enhances ex-vivo T cell responses to autolo-gous dendritic/myeloma fusion vaccine developed for thetreatment of multiple myeloma. Blood (ASH Annual Meet-ing Abstracts) 2009;114:781.

269. Uchiyama H, Barut BA, Chauhan D, Cannistra SA,Anderson KC. Characterization of adhesion molecules onhuman myeloma cell lines. Blood 1992;80:2306–14.

270. Noborio-Hatano K, Kikuchi J, Takatoku M, et al. Bortezo-mib overcomes cell-adhesion-mediated drug resistancethrough down-regulation of VLA-4 expression in multiplemyeloma. Oncogene 2009;28:231–42.

271. Mori Y, Shimizu N, Dallas M, Niewolna M, Story B, WilliamsPJ, Mundy GR, Yoneda T. Anti-alpha4 integrin antibodysuppresses the development of multiple myeloma and associ-ated osteoclastic osteolysis. Blood 2004;104:2149–54.

272. Olson DL, Burkly LC, Leone DR, Dolinski BM, Lobb RR.Anti-alpha4 integrin monoclonal antibody inhibits multiplemyeloma growth in a murine model. Mol Cancer Ther2005;4:91–9.

273. Podar K, Zimmerhackl A, Fulciniti M, et al. The selectiveadhesion molecule inhibitor Natalizumab decreases multiplemyeloma cell growth in the bone marrow microenvironment:therapeutic implications. Br J Haematol 2011;155:438–48.

274. Sainz IM, Isordia-Salas I, Espinola RG, Long WK, PixleyRA, Colman RW. Multiple myeloma in a murine syngeneicmodel: modulation of growth and angiogenesis by a mono-clonal antibody to kininogen. Cancer Immunol Immunother2006;55:797–807.

275. Matsui W, Huff CA, Wang Q, Malehorn MT, Barber J,Tanhehco Y, Smith BD, Civin CI, Jones RJ. Characteriza-tion of clonogenic multiple myeloma cells. Blood2004;103:2332–6.

276. Treon SP, Mitsiades C, Mitsiades N, Young G, Doss D, Sch-lossman R, Anderson KC. Tumor cell expression of CD59 isassociated with resistance to CD20 serotherapy in patientswith B-cell malignancies. J Immunother 2001;24:263–71.

277. Rozenfeld-Granot G, Toren A, Amariglio N, Brok-SimoniF, Rechavi G. Mutation analysis of the FAS and TNFRapoptotic cascade genes in hematological malignancies. ExpHematol 2001;29:228–33.

© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd 467

Allegra et al. Monoclonal antibodies and multiple myeloma

Page 28: Monoclonal antibodies: potential new therapeutic treatment against multiple myeloma

278. Matsui W, Huff CA, Vala M, Barber J, Smith BD, JonesRJ. Anti-tumour activity of interferon-alpha in multiplemyeloma: role of interleukin 6 and tumor celldifferentiation. Br J Haematol 2003;121:251–8.

279. Zand MS, Vo T, Pellegrin T, Felgar R, Liesveld JL,Ifthikharuddin JJ, Abboud CN, Sanz I, Huggins J. Apopto-sis and complement-mediated lysis of myeloma cells bypolyclonal rabbit antithymocyte globulin. Blood2006;107:2895–903.

280. Richardson PG, Lonial S, Jakubowiak AJ, Harousseau JL,Anderson KC. Monoclonal antibodies in the treatment ofmultiple myeloma. Br J Haematol 2011; July 21, doi:10.1111/j.1365-2141.2011.08790.x.

281. Lambert JM. Drug-conjugated monoclonal antibodies forthe treatment of cancer. Curr Opin Pharmacol 2005;5:543–9.

282. Senter PD. Potent antibody drug conjugates for cancer ther-apy. Curr Opin Chem Biol 2009;13:235–44.

283. Berardi S, Caivano A, Ria R, et al. Four proteins governingoverangiogenic endothelial cell phenotype in patients withmultiple myeloma are plausible therapeutic targets.Oncogene 2012;31:2258–69.

284. Tai YT, Anderson KC. Antibody-based therapies in multiplemyeloma. Bone Marrow Res 2011;2011:924058.

285. Morgan G. Future drug developments in multiple myeloma:an overview of novel lenalidomide-based combination thera-pies. Blood Rev 2010;24:27–32.

468 © 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Monoclonal antibodies and multiple myeloma Allegra et al.