13
Translational Cancer Mechanisms and Therapy The Mechanism of Action of the Anti-CD38 Monoclonal Antibody Isatuximab in Multiple Myeloma Laura Moreno 1 , Cristina Perez 1 , Aintzane Zabaleta 1 , Irene Manrique 1 , Diego Alignani 1 , Daniel Ajona 1,2,3 , Laura Blanco 1 , Marta Lasa 1 , Patricia Maiso 1 , Idoia Rodriguez 1 , Sonia Garate 1 , Tomas Jelinek 1 , Victor Segura 1 , Cristina Moreno 1 , Juana Merino 1 , Paula Rodriguez-Otero 1 , Carlos Panizo 1 , Felipe Prosper 1 , Jesus F. San-Miguel 1 , and Bruno Paiva 1 Abstract Purpose: Knowledge about the mechanism of action (MoA) of monoclonal antibodies (mAb) is required to understand which patients with multiple myeloma (MM) benet the most from a given mAb, alone or in combination therapy. Although there is considerable research about daratumumab, knowledge about other anti-CD38 mAbs remains scarce. Experimental Design: We performed a comprehensive analysis of the MoA of isatuximab. Results: Isatuximab induces internalization of CD38 but not its signicant release from MM cell surface. In addition, we uncovered an association between levels of CD38 expression and different MoA: (i) Isatuximab was unable to induce direct apoptosis on MM cells with CD38 levels closer to those in patients with MM, (ii) isatuximab sensitized CD38 hi MM cells to bortezomib plus dexamethasone in the presence of stroma, (iii) antibody-dependent cellular cytotoxicity (ADCC) was triggered by CD38 lo and CD38 hi tumor plasma cells (PC), (iv) antibody-dependent cellular phagocytosis (ADCP) was triggered only by CD38 hi MM cells, whereas (v) complement- dependent cytotoxicity could be triggered in less than half of the patient samples (those with elevated levels of CD38). Furthermore, we showed that isatuximab depletes CD38 hi B-lymphocyte precursors and natural killer (NK) lymphocytes ex vivothe latter through activation followed by exhaustion and eventually phagocytosis. Conclusions: This study provides a framework to under- stand response determinants in patients treated with isatux- imab based on the number of MoA triggered by CD38 levels of expression, and for the design of effective combinations aimed at capitalizing disrupted tumorstroma cell protection, augmenting NK lymphocytemediated ADCC, or facilitating ADCP in CD38 lo MM patients. See related commentary by Malavasi and Faini, p. 2946 Introduction Growing knowledge about tumor and immune cell biology led to continuous development of immunotherapies against each of the four major nodes of vulnerability in the cancerimmune relationship: direct targeting of surface antigens; boosting of numbers and functioning of immune effectors; activating tumor antigen-specic immunity, and; overcoming inhibitory immune suppression (1). Multiple myeloma (MM) is no exception to this; however, among the four major nodes of cancer immunotherapy, monoclonal antibodies (mAb) are at the forefront of recent clinical development with two new drugs approved in 2015 for the treatment of relapsed/refractory disease (2). Characteristics that make antigens attractive as targets for mAb- based therapy include, amongst others, the density of expression of the target molecule by malignant and benign cells (3). Other desirable characteristics of target tumor antigens vary depending on the mAb construct and the mechanism of action (MoA) triggered by such constructs (3). Some mAbs that target antigens on the surface of malignant cells can induce apoptosis by direct transmembrane signaling (4). There is also evidence that mAbs kill target cells by complement-dependent cytotoxicity (CDC; ref. 5), or by inducing antibody-dependent cellular cytotoxicity (ADCC; ref. 6) and phagocytosis (ADCP; ref. 7). Thus, exquisite understanding about the MoA of novel mAbs is warranted, because precise identication of the (one or more) effector mechanisms triggered by these can have considerable impact on optimal selection of patients' candidates to receive a given mAb based on tumor phenotypes, the design of effective treatment combinations, and correct immune monitoring to predict treat- ment failure (i.e., precision medicine). CD38 is a type II transmembrane glycoprotein without an internal signaling domain that, although at variable levels (8), 1 Clinica Universidad de Navarra, Centro de Investigaci on M edica Aplicada (CIMA), Instituto de Investigaci on Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain. 2 Solid Tumors Program, Centro de Investigaci on M edica Aplicada (CIMA), Instituto de Investigaci on Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/ 00443, Pamplona, Spain. 3 Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain. Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). L. Moreno, C. Perez, and A. Zabaleta contributed equally to this work. Corresponding Author: Bruno Paiva, CIMA, Avda. Pio XII 55, 31008 Pamplona, Navarra. Phone: 349-4919-4700, ext. 1038; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-18-1597 Ó2019 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 25(10) May 15, 2019 3176 on April 30, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst January 28, 2019; DOI: 10.1158/1078-0432.CCR-18-1597

The Mechanism of Action of the Anti-CD38 Monoclonal ... · understanding about the MoA of novel mAbs is warranted, because precise identification of the (one or more) effector mechanisms

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: The Mechanism of Action of the Anti-CD38 Monoclonal ... · understanding about the MoA of novel mAbs is warranted, because precise identification of the (one or more) effector mechanisms

Translational Cancer Mechanisms and Therapy

The Mechanism of Action of the Anti-CD38Monoclonal Antibody Isatuximab in MultipleMyelomaLaura Moreno1, Cristina Perez1, Aintzane Zabaleta1, Irene Manrique1, Diego Alignani1,Daniel Ajona1,2,3, Laura Blanco1, Marta Lasa1, Patricia Maiso1, Idoia Rodriguez1,Sonia Garate1, Tomas Jelinek1, Victor Segura1, Cristina Moreno1, Juana Merino1,Paula Rodriguez-Otero1, Carlos Panizo1, Felipe Prosper1, Jesus F. San-Miguel1, andBruno Paiva1

Abstract

Purpose: Knowledge about the mechanism of action(MoA) of monoclonal antibodies (mAb) is required tounderstand which patients with multiple myeloma (MM)benefit the most from a given mAb, alone or in combinationtherapy. Although there is considerable research aboutdaratumumab, knowledge about other anti-CD38 mAbsremains scarce.

Experimental Design: We performed a comprehensiveanalysis of the MoA of isatuximab.

Results: Isatuximab induces internalization of CD38 butnot its significant release fromMMcell surface. In addition, weuncovered an association between levels of CD38 expressionand different MoA: (i) Isatuximab was unable to induce directapoptosis on MM cells with CD38 levels closer to those inpatients with MM, (ii) isatuximab sensitized CD38hi MM cellsto bortezomib plus dexamethasone in the presence of stroma,(iii) antibody-dependent cellular cytotoxicity (ADCC) was

triggered by CD38lo and CD38hi tumor plasma cells (PC),(iv) antibody-dependent cellular phagocytosis (ADCP) wastriggered only by CD38hi MM cells, whereas (v) complement-dependent cytotoxicity could be triggered in less than half ofthe patient samples (those with elevated levels of CD38).Furthermore, we showed that isatuximab depletes CD38hi

B-lymphocyte precursors and natural killer (NK) lymphocytesex vivo—the latter through activation followed by exhaustionand eventually phagocytosis.

Conclusions: This study provides a framework to under-stand response determinants in patients treated with isatux-imab based on the number of MoA triggered by CD38 levelsof expression, and for the design of effective combinationsaimed at capitalizing disrupted tumor–stroma cell protection,augmenting NK lymphocyte–mediated ADCC, or facilitatingADCP in CD38lo MM patients.

See related commentary by Malavasi and Faini, p. 2946

IntroductionGrowing knowledge about tumor and immune cell biology led

to continuous development of immunotherapies against each ofthe four major nodes of vulnerability in the cancer–immunerelationship: direct targeting of surface antigens; boosting ofnumbers and functioning of immune effectors; activating tumorantigen-specific immunity, and; overcoming inhibitory immunesuppression (1). Multiple myeloma (MM) is no exception to this;

however, among the fourmajor nodes of cancer immunotherapy,monoclonal antibodies (mAb) are at the forefront of recentclinical development with two new drugs approved in 2015 forthe treatment of relapsed/refractory disease (2).

Characteristics that make antigens attractive as targets formAb-based therapy include, amongst others, the density of expressionof the target molecule by malignant and benign cells (3). Otherdesirable characteristics of target tumor antigens vary dependingon the mAb construct and the mechanism of action (MoA)triggered by such constructs (3). Some mAbs that target antigenson the surface of malignant cells can induce apoptosis by directtransmembrane signaling (4). There is also evidence that mAbskill target cells by complement-dependent cytotoxicity (CDC;ref. 5), or by inducing antibody-dependent cellular cytotoxicity(ADCC; ref. 6) and phagocytosis (ADCP; ref. 7). Thus, exquisiteunderstanding about the MoA of novel mAbs is warranted,because precise identification of the (one or more) effectormechanisms triggered by these can have considerable impact onoptimal selection of patients' candidates to receive a given mAbbased on tumor phenotypes, the design of effective treatmentcombinations, and correct immune monitoring to predict treat-ment failure (i.e., precision medicine).

CD38 is a type II transmembrane glycoprotein without aninternal signaling domain that, although at variable levels (8),

1Clinica Universidad de Navarra, Centro de Investigaci�on M�edica Aplicada(CIMA), Instituto de Investigaci�on Sanitaria de Navarra (IDISNA), CIBER-ONCnumber CB16/12/00369 and CB16/12/00489, Pamplona, Spain. 2Solid TumorsProgram, Centro de Investigaci�on M�edica Aplicada (CIMA), Instituto deInvestigaci�on Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00443, Pamplona, Spain. 3Department of Biochemistry and Genetics, Schoolof Sciences, University of Navarra, Pamplona, Spain.

Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

L. Moreno, C. Perez, and A. Zabaleta contributed equally to this work.

Corresponding Author: Bruno Paiva, CIMA, Avda. Pio XII 55, 31008 Pamplona,Navarra. Phone: 349-4919-4700, ext. 1038; E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-18-1597

�2019 American Association for Cancer Research.

ClinicalCancerResearch

Clin Cancer Res; 25(10) May 15, 20193176

on April 30, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 28, 2019; DOI: 10.1158/1078-0432.CCR-18-1597

Page 2: The Mechanism of Action of the Anti-CD38 Monoclonal ... · understanding about the MoA of novel mAbs is warranted, because precise identification of the (one or more) effector mechanisms

is expressed onmalignant plasma cells (PC) from all patients withMM(9).CD38mAbs currently available orbeingdeveloped for thetreatment of MM include daratumumab, isatuximab, MOR202,and Ab19. Except the latter, all other threemAbs are under clinicaldevelopment but, so far, only daratumumab has been approvedfor the treatment of patients with MM. Coincidently, the MoA ofdaratumumab has been considerably investigated and severaleffector mechanisms were identified: tumor cell apoptosis uponFcgR cross-linking (10), CDC (11), ADCP (12), and ADCC (13).More recently, Krejcik and colleagues (14, 15) have shown thatdaratumumab depletes CD38þve immune regulatory cells, whichpromoted the expansion of TCR-related T lymphocytes. Althoughthese findings are surprising because they suggest that daratumu-mab is capable of depleting cells with considerable lower levels ofCD38, they also indicate that drugs able to enhance the immunesystem may be preferred partners to combine with anti-CD38mAbs (16). Accordingly, both preclinical (11, 13, 17, 18) andclinical data have confirmed the efficacy of daratumumab incombination with lenalidomide (19, 20) or pomalidomide (21).Isatuximab has also demonstrated single-agent and combinationactivity in heavily pretreated patients with relapsed/refractoryMM (22); however, preclinical data to support the clinical devel-opment of isatuximab are scarce (23, 24).

Here, we aimed to gain an understanding of isatuximab MoA.Our results allowed us to identify thresholds of CD38 expressionrequired for triggering different MoA, which can be used foroptimal patient selection and to design effective treatment com-binations. In addition, we unveil potential mechanisms by whichnatural killer (NK) lymphocytes decrease after treatment withanti-CD38 mAbs.

Materials and MethodsA complete description of methods and techniques used in thisstudy is available in the Supplementary Methods

Briefly, ex vivo studies were performed in bonemarrow samplesfrom 13 patients with MM (six newly diagnosed and seven with

relapse/refractory disease). All samples were collected afterinformed consent was given by each patient, according to thelocal ethics committee and the Helsinki Declaration. We used theRPMI-8226, MM1S, OPM2, and H929 MM cell lines for studyinginternalization or release of CD38 after isatuximab binding. Theeffect of isatuximab on proliferation and apoptosis wasmeasuredusing diphenyltetrazolium bromide (MTT) and flow cytometryassays, whereas the ability of isatuximab to trigger CDC, ADCPand ADCC was determined using various flow cytometry meth-ods. In parallel, we evaluated the immune modulatory effects ofisatuximab in the series of patients described above, and purifiedNK lymphocytes from healthy individuals to understand themechanismsbehind their activation anddepletion after treatmentwith isatuximab, using fluorescence-activated cell sorting (FACS)and gene expression profiling (GEP). We also evaluated isatux-imab MoA in NOD/scid/gc�/� (NSG) xenotransplant models,which were inoculated with firefly luciferase-expressing MM1Scells in the presence or absence of immune effector cells [i.e.,peripheral blood mononucleated cells (PBMC) obtained fromhealthy donors]. We examined the potential of an anti-CD137mAb, lenalidomide and bortezomib to enhance isatuximab-mediated activation of NK lymphocytes and prevent their exhaus-tion, using flow cytometry methods.

The Wilcoxon signed rank test was used to evaluate the statis-tical significance of the differences observed between CD16meanfluorescence intensity (MFI) levels of NK lymphocytes untreatedversus treated with isatuximab, whereas the Mann–Whitney Uand the Kruskal–Wallis tests were used to estimate the statisticalsignificance of differences observed between two or more groups,respectively. Correlation studies were performed using the Pear-son test. Survival curves were plotted according to the Kaplan–Meier method and compared using the log-rank test. The SPSSsoftware (version 20.0; SPSS Inc.) was used for all statistical tests.

ResultsIsatuximab induces internalization of CD38 but not itssignificant release from MM cell surface

Recent evidence indicates that daratumumab induces rapid(i.e., less than 4 hours) redistribution of CD38 molecules andformation of polar aggregates leading to the release of CD38 inmicrovesicles (25). Thus, we sought to investigate whether isa-tuximab has a similar effect onCD38 density in the surface ofMMcells. First, we started by selecting the H929, MM1S, OPM2, andRPMI-8226 cell lines to investigate MM cells with a well-definedrange of specific isatuximab antibody-binding capacity (SABC)and CD38 antigen density. Thus, when compared with MMpatients (n ¼ 13; median SABC of 120.486), anti-CD38 SABCfound in H929, MM1S and OPM2 cell lines was considerablylower (7.079, 10.421, and 21.886, respectively) and equivalent toanti-CD38 SABC values found in only one patient (Fig. 1A). Bycontrast, RPMI-8226MM cells displayed SABC levels comparableto the median value of patients with MM (137.759 vs. 120.486,respectively). Therefore, H929, MM1S, and OPM2 cell lines arerepresentative of patients whose tumor PCs have low CD38expression (CD38lo), whereas RPMI-8226 cells are representativeof patients with high CD38 levels (CD38hi). To measure CD38occupancy and changes in CD38 surface expression upon treat-ment with isatuximab, we used monoclonal (targeting a singleepitope that competeswith isatuximab) andpolyclonal (targetingmultiple epitopes) anti-CD38–FITC-conjugated antibodies, to

Translational Relevance

Greater knowledge about the immune effector mechanismsof monoclonal antibodies is a prerequisite for better predic-tion of patients' response and optimal monitoring of treat-ment effects. Here, we performed a comprehensive analysisabout the mechanism of action of isatuximab in multiplemyeloma. Our results underline similarities and differencesbetween isatuximab and other anti-CD38 monoclonal anti-bodies and unveiled a direct association between the levels ofCD38 expression and the mechanisms triggered by isatuxi-mab; accordingly, antibody-dependent cellular cytotoxicityemerges as the most prevalent effector mechanism by whichisatuximab eliminates tumor cells. They also provide informa-tion that could explain, at least in part, why patients with lowerexpression of CD38 may respond poorly to these drugs thanpatients with higher CD38 expression. We also provide newinsight about the paradoxical depletion of natural killer (NK)lymphocytes following treatment with anti-CD38monoclonalantibodies; further development of therapeutic strategies toovercome this phenomenon should become a priority.

Mechanism of Action of Isatuximab in Myeloma

www.aacrjournals.org Clin Cancer Res; 25(10) May 15, 2019 3177

on April 30, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 28, 2019; DOI: 10.1158/1078-0432.CCR-18-1597

Page 3: The Mechanism of Action of the Anti-CD38 Monoclonal ... · understanding about the MoA of novel mAbs is warranted, because precise identification of the (one or more) effector mechanisms

Figure 1.

Isatuximab induces internalization of CD38 but not its significant release fromMM cell surface. A, SABC and CD38 antigen density in RPMI-8226, OPM2, MM1S,and H929 MM cell lines, as well as on primary tumor PCs (n¼ 13 patients). B and C,MM cell lines were cultured in triplicates and in the presence of isatuximab (10mg/mL) for 24, 48, 72, and 96 hours (h). After 96 h, cells were collected for flow cytometry immunophenotyping; half were labeled with a mAb, whereas the otherhalf were labeled with a polyclonal antibody to measure the MFI levels of the two CD38-FITC–conjugated antibodies. Baseline values of each cell line werenormalized, and the variability of CD38 MFI levels is shown every 24 h.D,MM1S and RPMI-8226 MM cell lines were treated with A647-labeled isatuximab for 0.5, 1,2, 4, 24, and 48 hours (h). After treatment, cells were washed with PBS, and flow cytometry analysis showed that, over time, there was an increase in MFI of theCD38/isatuximab-A647 signal (measured intracellularly and in the surface membrane) with respect to the control sample. E, Surface membrane CD38-isatuximab complexes were measured with a secondary antibody FITC–anti-human IgG Fab after treatment with unlabeled isatuximab for 0.5, 1, 2, 4, 24, and 48hours (h). No significant differences on CD38-isatuximab surface levels were noted with respect to the Control-T0 [MM cells incubated with A647-labeledisatuximab or unlabeled isatuximab (10 mg/mL) at 4�C for 15 minutes]. The MFI levels measured in each time point were normalized with respect to the Control-T0. All experiments were performed in triplicate.

Moreno et al.

Clin Cancer Res; 25(10) May 15, 2019 Clinical Cancer Research3178

on April 30, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 28, 2019; DOI: 10.1158/1078-0432.CCR-18-1597

Page 4: The Mechanism of Action of the Anti-CD38 Monoclonal ... · understanding about the MoA of novel mAbs is warranted, because precise identification of the (one or more) effector mechanisms

label the fourMMcell lines after treatment with isatuximab for 24up to 96 hours. Although the MFI of CD38 measured with themonoclonal antibody was significantly lower after the first 24hours compared with baseline and remained low thereafter(median decrease of 73% in all cell lines, P¼ 0.04; Fig. 1B), therewas no significant variation in the MFI of CD38 when using thepolyclonal antibody (median variationof 12% in all cell lines; P¼0.36; Fig. 1C). Similarly, RPMI-8226 cells treated during 24 hoursand cultured in RPMI medium with 10% normal human serum(NHS) or heat-inactivated NHS (HI-NHS), in presence or absenceof PBMCs and stained with the polyclonal anti-CD38–FITC-conjugated antibody, displayed continuous intracellular and sur-face expression of CD38 despite the formation of CD38 polaraggregates (Supplementary Fig. S1).

To further investigate whether CD38 persisted in MM cells afterbinding to isatuximab, RPMI-8226 and MM1S MM cells wereculturedup to48hours at 4�Cor 37�C in the presence of unlabeledversus A647-labeled isatuximab. Flow cytometry analysis of MMcells cultured at 37�C in the presence of isatuximab-A647 revealeda significant increase inMFI levels after 24 and 48 hours (Fig. 1D).By contrast, MFI levels remained unaltered when MM cells werecultured with isatuximab-A647at 4�C (Supplementary Fig. S1D).To confirm the presence of surface CD38-isatuximab complexesover time, we cultured RPMI-8226 and MM1S MM cells withunlabeled isatuximab and stained them with a secondary FITCgoat anti-human IgG Fab antibody to measure the amount ofisatuximab bound to CD38 in the surface of MM cells. No signif-icant differences were observed in the MFI measured in the FITCchannel during 48 hours (Fig. 1E). Thus, our results indicate thatisatuximab effectively binds CD38 andmay induce internalizationbut not significant release of CD38 from MM cells.

Isatuximab sensitizes CD38hi MM cells to bortezomib plusdexamethasone but does not induce apoptosis by directtransmembrane signaling on MM cells with CD38 levels closerto those in MM patients

After demonstrating that there was no significant release ofCD38 after binding to isatuximab, we then investigated whetherthe drug had a direct antiproliferative or pro-apoptotic effect onMM cells. Upon treating CD38lo and CD38hi MM cells withincreasing concentrations of isatuximab, we have not observeda significant (P > 0.05) effect on viability, proliferation or survivalof MM cells (Supplementary Fig. S2A–S2C). To understandwhether, in the absence of direct apoptosis, isatuximab wasmodulating MM cells at the molecular level, we analyzed thetranscriptome of CD38hi RPMI-8226 MM cells after treatmentwith isatuximab. No significant gene deregulation (using a Bstatistic cutoff B > 0 and log FC > 1) induced by treatment withisatuximab was observed when compared with untreated RPMI-8226 MM cells (Supplementary Fig. S2D).

Because CD38 may synergize with the CXCR4 pathway andcooperate in CXCL12-mediated homing (26), we investigatedwhether CD38 blocking by isatuximab could increase the efficacyof anti-myeloma drugs known to have reduced effect in thepresenceof stroma(e.g., bortezomib anddexamethasone; ref. 27).Interestingly, we observed that in the presence of stroma, isatux-imab significantly increased the percentage of dying CD38hi

RPMI-8226 cells after treatment with bortezomib plus dexameth-asone (mean increment, 12%; P ¼ 0.006; Supplementary Fig.S2E). However, adhesion of RPMI-8226 and MM1S MM cells tothe HS5 stromal cell line or patient-derived bonemarrow stromal

cells was not reduced by treatment with isatuximab (Supplemen-tary Fig. S2F and S2G). Thus, our results suggest that isatuximabhas no direct effect on MM cell lines with CD38lo and CD38hi

levels but may sensitize CD38hi MM cells to combined bortezo-mib and dexamethasone through mechanisms other than dis-rupting tumor–stromal cell adhesion.

Isatuximab activates complement in MM cells with elevatedlevels of CD38

In the absence of a direct effect on MM cells with CD38 levelscomparable to that of most MM patients, we then exploredwhich immune effector mechanisms mediate the anti-myelomaactivity of isatuximab. To determine whether isatuximab trig-gered activation of complement, we measured the deposition ofC3-related fragments in the four MM cells lines as well as inprimary tumor PCs (n ¼ 9 patients). Although no deposition ofC3-related fragments was noted in CD38lo and CD38hi MM celllines (Fig. 2A), there was a non-significant increment ofC3-related fragments deposition in primary tumor PCs (meanC3 MFI levels of 1.8 and 7.0 in the absence versus presence ofisatuximab; P ¼ 0.13). More detailed analyses showed a sig-nificant correlation between anti-CD38 SABC and depositionof C3-related fragments in primary tumor PCs (r ¼ 0.86; P ¼0.003; Fig. 2B), as well as a trend for a correlation betweentumor PC depletion and deposition of C3-related fragments(r ¼ 0.61; P ¼ 0.08; Fig. 2C). Thus, our results suggest thatalthough isatuximab may induce CDC in patients with elevatedlevels of CD38, immune effector mechanisms other than CDCmust mediate the activity of isatuximab against MM cells withlower levels of CD38 expression.

Isatuximab induces ADCC in MM cells with a broad range ofCD38 expression and selective ADCP in CD38hi MM cells

To determine whether isatuximab triggered ADCC, we startedby incubating CD38lo and CD38hi MM cells in absence or pres-ence of the drug, with or without donor NK lymphocytes. WhenMM cells were incubated only with isatuximab, survival wasnearly 100%, thus reproducing our previous findings that indi-cated lack of direct apoptosis. By contrast, significant cell deathwas observed when both isatuximab and NK lymphocytes werecultured with MM1S, OPM2 and RPMI-8226 but not with H929MM cells (Fig. 3A), which had the lowest isatuximab anti-CD38SABC (Fig. 1A). We also evaluated the potential of isatuximabto trigger ADCC against primary tumor PCs (n ¼ 13 patients)and observed significant tumor PC depletion (median 51%;P ¼ 0.003); importantly, we noted a significant correlationbetween depletion of tumor PCs and their anti-CD38 SABC(r ¼ 0.58; P ¼ 0.04; Fig. 3B).

Because, in addition to NK lymphocytes, part of T lympho-cytes also express FcRIIIA (CD16A), we sought to determine therelative contribution of each subset to the ADCC triggered byisatuximab. Accordingly, we used FACS to culture CD38hi

RPMI-8226 MM cells either with donor PBMCs, or with PBMCswithout NK lymphocytes (W/o NK), or with PBMCs withoutCD16þve T lymphocytes (W/o T), in the presence or absence ofisatuximab. Our results show that the percentage of tumor PCsremained stable in the absence of NK lymphocytes, whereassignificant tumor depletion was noted in the absence ofCD16þve T lymphocytes (Fig. 3C), thus indicating that NKlymphocytes are the critical mediators of ADCC triggered byisatuximab.

Mechanism of Action of Isatuximab in Myeloma

www.aacrjournals.org Clin Cancer Res; 25(10) May 15, 2019 3179

on April 30, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 28, 2019; DOI: 10.1158/1078-0432.CCR-18-1597

Page 5: The Mechanism of Action of the Anti-CD38 Monoclonal ... · understanding about the MoA of novel mAbs is warranted, because precise identification of the (one or more) effector mechanisms

Afterwards, we investigated if isatuximab also triggered ADCPby labeling MM cells with violet proliferation dye (VPD) priorculturing them with unlabeled M2-like macrophages. Thus, theintensity of VPD measured in CD11bþve macrophages, afterculture with VPDþve MM cells and in the presence vs absence ofisatuximab, would be a surrogate for ADCP. Our results shownon-significant ADCP against CD38lo MM cells, but significantADCP against CD38hi RPMI-8226 MM cells (median of 43%,P ¼ 0.005; Fig. 3D).

To evaluate isatuximab MoA in vivo, we inoculated NOD/scid/gc�/� (NSG) mice (which are deficient in NK cells but retainphagocytic cells) with firefly luciferase-expressing MM1S cellsand treatedwith isatuximab in the presence or absence of immuneeffector cells (i.e., PBMCs from healthy donors). Our resultsshow that treatment with isatuximab significantly prolongedsurvival of mice in the absence and presence of immune effector

cells, indicating that ADCP and ADCC were triggered in vivo byisatuximab (Fig. 3E and F).

Effect of isatuximab on other immune cellsImmune modulation triggered by daratumumab due to deple-

tion of CD38þve immune regulatory cells has been recentlydescribed (14, 15). Thus, we investigated whether isatuximabalso depleted specific immune cell subsets by treating primarybone marrow samples from 13 patients with MM with isatux-imab. As a control for the effect of isatuximab, we previouslydemonstrated that the drug significantly depleted tumor PCs(median of 51%; P ¼ 0.003; Fig. 3B). Interestingly, we observedsignificant depletionofCD38hi B-lymphocyte precursors (medianof 54%; P¼ 0.009), basophils (median of 26.5%; P¼ 0.006) andNK lymphocytes (median of 18%; P¼ 0.002) after treatmentwithisatuximab. By contrast, no differences were noted in the relative

Figure 2.

Isatuximab may induce complement activation in MM cells with elevated levels of CD38. A, Deposition of C3-derived fragments in CD38lo (H929,MM1S, and OPM2) and CD38hi (RPMI-8226) MM cells after complement activation in the presence of 10 mg/mL of isatuximab for 24 hours (treated) orsaline (untreated). B, Deposition of C3-derived fragments in tumor PCs from primary bone marrow samples (n ¼ 9 patients) untreated and treatedwith 10 mg/mL of isatuximab for 24 hours was measured, and correlated with the levels of SABC and CD38 antigen density per tumor PC. C,Correlation between C3-fragment deposition and depletion of tumor PCs from primary bone marrow samples (n ¼ 9 patients) untreated and treatedwith 10 mg/mL of isatuximab for 24 hours. Data are expressed as the ratio of the MFI between cells incubated with 10% serum and cells incubatedwith its respective complement inactivated serum. Bar graphs represent the median � the 95% confidence intervals (from three independentexperiments). The statistical significance was evaluated using the Mann–Whitney U test.

Moreno et al.

Clin Cancer Res; 25(10) May 15, 2019 Clinical Cancer Research3180

on April 30, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 28, 2019; DOI: 10.1158/1078-0432.CCR-18-1597

Page 6: The Mechanism of Action of the Anti-CD38 Monoclonal ... · understanding about the MoA of novel mAbs is warranted, because precise identification of the (one or more) effector mechanisms

Figure 3.

Isatuximab induces ADCC in MM cells with a broad range of CD38 levels and selective ADCP in CD38hi MM cells. A, ADCCwas measured according to thepercentage of Annexin V–positive CD38lo (H929, MM1S, and OPM2) and CD38hi (RPMI-8226) MM cells cocultured with donor NK lymphocytes, and aftertreatment with 10 mg/mL of isatuximab for 4 hours. B, Tumor PC depletion was determined in bone marrow samples from 13 patients with MM after treatmentwith 10 mg/mL of isatuximab for 24 hours, and correlated with the SABC and CD38 antigen density per tumor PC. C, To determine the relative contribution of NKand CD16þve T lymphocytes to the ADCC triggered by isatuximab against CD38hi RPMI-8226 MM cells, we used FACS to coculture CD38hi RPMI-8226 MM cellseither with donor PBMCs, with PBMCsW/o NK, or with PBMCswithout CD16þve W/o T, in the presence versus absence of 10 mg/mL of isatuximab for 24 hours. D,CD38lo (H929, MM1S, and OPM2) and CD38hi (RPMI-8226) MM cells were labeled with VPD andwere preincubated with 10 mg/mL of isatuximab before addingmonocyte-derived M2-like macrophages obtained from healthy donors in a 1:4�:MM cell ratio. VPDwas measured on CD11bþve macrophages after 2 hours ofcoculture. All experiments were performed in triplicate. In A, C, andD, bars represent median values and vertical lines the upper bound of the 95% confidenceintervals; the statistical significance was evaluated using the Kruskal–Wallis (A and C) and the Mann–Whitney U (D) tests. E,Mice (n¼ 7 per group) wereinoculated with 5 x 106 MM1S-GFP-Luc cells on day 0, and either were left untreated or were treated with isatuximab (20mg/kg) on days 7 and 14. F, In anothertwo groups of mice (n¼ 7 per group), 10 x 106 PBMCs were administered on day 8. Survival was monitored twice weekly.

Mechanism of Action of Isatuximab in Myeloma

www.aacrjournals.org Clin Cancer Res; 25(10) May 15, 2019 3181

on April 30, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 28, 2019; DOI: 10.1158/1078-0432.CCR-18-1597

Page 7: The Mechanism of Action of the Anti-CD38 Monoclonal ... · understanding about the MoA of novel mAbs is warranted, because precise identification of the (one or more) effector mechanisms

percentage of regulatory T cells (Treg), and T lymphocytes were notimmune modulated after exposure to isatuximab (SupplementaryFig. S3). Other hematopoietic cells (including CD34þveCD19�ve

hematopoietic progenitors, neutrophils or monocytes) were notdepleted by isatuximab (Supplementary Fig. S3E and S3F).

Activation and depletion of NK lymphocytes upon treatmentwith isatuximab

Because NK-mediated ADCC was the only MoA developed byisatuximab to kill CD38lo and CD38hi MM cells, but at the sametime isatuximab was depleting (CD38hi) NK lymphocytes, wesought to gain further insight into the mechanism behind thedirect effect of isatuximab onNK-lymphocyte depletion. Thus, wecompared the transcriptomeofNK lymphocytes FACSorted after a24-hour culture with CD38hi RPMI-8226 MM cells, in the pres-ence or absence of isatuximab. Interestingly, NK lymphocytesshowed deregulated expression of 70 genes in the presence ofisatuximab (Fig. 4A), for which Ingenuity analysis attributedbiological processes such as cell chemotaxis, cytolysis and defenseresponse. One such gene was TNFRSF9 (i.e., 4-1BB or CD137),

whose upregulation was also observed at the protein (antigen)level (Fig. 4D). These results suggest that isatuximab induces NK-lymphocyte activation beyond tumor–NK cell cross-talk. Becausein previous experiments with primary patient samples (n ¼ 13),we observed significant downregulation of CD16 MFI levels onNK lymphocytes after treatment with isatuximab (62% down-regulation, P < 0.001; Fig. 4B), we thus investigated whetherisatuximab was activating NK lymphocytes through Fc binding,or if activation was also being triggered after binding to CD38 onCD38hi NK lymphocytes. We observed that in the presence ofincreasing concentrations of an Fc blocker containing specializedhuman IgG, NK lymphocytes cultured with RPMI-8226 showedincreased expression of the activation markers CD69 and CD137in a dose-dependent manner. Of note, the extent of activationinducedby the Fc blockerwas consistently lower than the extent ofactivation observed in the presence of isatuximab alone. Thus, thecombination of Fc blocker with isatuximab induced a significantincrement in the expression of the activation markers CD69 andCD137 comparedwithNK lymphocytes treatedwith Fc blocker inthe absence of isatuximab (Fig. 4C and D). Altogether, these

Figure 4.

Activation of NK lymphocytes upon treatment with isatuximab. A, Heat map of genes with significantly different expression (using a B statistic cutoff B > 0 andlog FC > 1) from NK lymphocytes cocultured with CD38hi RPMI-8226 MM cells, left untreated versus treated with 10 mg/mL of isatuximab for 24 hours. B, CD16MFI levels were measured on NK lymphocytes from bone marrow samples of 13 patients with MM treated with 10 mg/mL of isatuximab for 24 hours. C and D,The percentage of CD69 (C) and CD137 (D) expression on NK lymphocytes cocultured with RPMI-8226 MM cells, left untreated or treated with increasingconcentrations of an Fc blocker (0.5, 1, and 2.5 mg/mL), in the absence versus presence of 10 mg/mL of isatuximab. Experiments inA, C, and Dwere performed intriplicate. In C and D, bars represent median values and vertical lines the upper bound of the 95% confidence intervals; the statistical significance was evaluatedusing theWilcoxon (B) and Kruskal–Wallis (C and D) tests.

Moreno et al.

Clin Cancer Res; 25(10) May 15, 2019 Clinical Cancer Research3182

on April 30, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 28, 2019; DOI: 10.1158/1078-0432.CCR-18-1597

Page 8: The Mechanism of Action of the Anti-CD38 Monoclonal ... · understanding about the MoA of novel mAbs is warranted, because precise identification of the (one or more) effector mechanisms

results suggest that in the presence of target cells, isatuximabactivates NK lymphocytes by Fc binding and CD38 transmem-brane signaling.

Afterwards, we investigated whether the depletion of CD38hi

NK lymphocytes occurred due to coating and activation byisatuximab (i.e., fratricide). Accordingly, VPD-labeled NK lym-phocytes (VPD-NK) preincubated with isatuximab were culturedwith VPD-unlabeled NK lymphocytes (NK) from the same sub-ject. Our results show that in the absence of isatuximab, there wasno NK-lymphocyte activation or depletion; however, exposure toisatuximab induced significant activation (mean 17% incrementin CD69 expression, P ¼ 0.001) and death of VPD-labeled NKlymphocytes (mean 27% increment, P ¼ 0.027), without anydifferences in activation and death rates when VPD-NK lympho-cytes were cultured with VPD-unlabeled NK lymphocytes(Fig. 5A). These results suggest that rather than fratricide, activa-tion followed by exhaustionwould be responsible, at least in part,for depletion of NK lymphocytes after treatment with isatuximab.Driven by the upregulation of CD137, we subsequently evaluatedif an anti-CD137 agonist mAb could prolong isatuximab-medi-ated activation of NK lymphocytes and prevent their exhaustion.However, both viability and activation of NK lymphocytes wasidentical after treatment with isatuximab plus anti-CD137 as

compared with isatuximab alone (Fig. 5B); accordingly, no dif-ferences in tumor cell depletion were noted by adding the anti-CD137 mAb (Supplementary Fig. S4A). Equivalent results werefound using similar experimental conditions in primary patientbone marrow samples (Supplementary Fig. S4B). We also inves-tigated whether lenalidomide could prolong isatuximab-mediat-ed activation of NK lymphocytes and prevent their exhaustion, bypretreating PBMCs with increasing doses of lenalidomide beforeculture with RPMI-8226, MM1S and OPM2 MM cells, in thepresence or absence of isatuximab. However, no significant dif-ferences were noted (Supplementary Fig. S5A and S5B). Asexpected, immune suppression of NK lymphocytes induced byproteasome inhibition after pretreatment with bortezomibresulted in lower NK cell viability (Supplementary Fig. S5C).

Because recent findings suggesting that SLAMF7 could becritical for phagocytosis of hematopoietic cells (7) and, coinci-dently, someNK lymphocytes co-express SLAMF7 andCD38withlevels above that of RPMI-8226MM cells (Fig. 6), we investigatedwhether phagocytosis could also contribute to the depletion ofsome NK lymphocytes. Accordingly, we labeled increasing num-bers ofNK lymphocyteswithVPDand cultured themwithM2-likemacrophages, in the presence or absence of isatuximab; thepercentage of VPDþvemacrophageswould thus indicate the extent

Figure 5.

Depletion of NK lymphocytes upon treatment with isatuximab. A, Activation of VPD-labeled NK lymphocytes (NK-VPD) was measured according to thepercentage of CD69 expression, in the presence or absence of VPD-unlabeled NK lymphocytes (NK) from the same subject, and after treatment with 10 mg/mL ofisatuximab for 24 hours. Cell death was determined according to the percentage of Annexin V–positive cells in the same experimental conditions. B, Activation ofNK lymphocytes was measured according to the percentage of CD69 expression, in the presence of isatuximab (10 mg/mL) and anti-CD137 (mouse IgG1, clone4B4-1, k isotype, Biolegend) alone or in combination. Cell survival was determined according to the percentage of Annexin V–negative cells in the sameexperimental conditions. C, NK lymphocytes were labeled with VPD and were incubated with 10 mg/mL of isatuximab for 24 hours before coculturing themwithmonocyte-derived M2-like macrophages obtained from healthy donors in 1:1, 1:2, and 1:4�:NK cell ratios. VPD was measured on CD11bþve macrophages after 2hours of coculture. All experiments were performed in triplicate. Bars represent median values and vertical lines the upper bound of the 95% confidence intervals;the statistical significance was evaluated using the Mann–Whitney U tests.

Mechanism of Action of Isatuximab in Myeloma

www.aacrjournals.org Clin Cancer Res; 25(10) May 15, 2019 3183

on April 30, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 28, 2019; DOI: 10.1158/1078-0432.CCR-18-1597

Page 9: The Mechanism of Action of the Anti-CD38 Monoclonal ... · understanding about the MoA of novel mAbs is warranted, because precise identification of the (one or more) effector mechanisms

ofNK-lymphocyte phagocytosis. Interestingly, our results showeda non-significant increment of NK-lymphocyte phagocytosiswith isatuximab (P ¼ 0.16; Fig. 5C), suggesting that some NKlymphocytes (possibly those with co-expression of SLAMF7 andCD38hi) could be phagocyted by M2-like macrophages in thepresence of isatuximab.

DiscussionSeveral anti-CD38mAbshavebeendeveloped for the treatment

of MM (28). Thus, in-depth understanding of their MoA is ofutmost importance to select patients who, according to theirtumor and immune phenotypes, are potential candidates tobenefit the most from a given mAb, alone or within rationaltreatment combinations. Although there is considerableresearch about the MoA of daratumumab (10, 11, 12–15, 17,25, 29, 30, 31), preclinical data about other anti-CD38 mAbsin clinical development remain scarce (23, 24). Here, we per-formed a comprehensive analysis on the MoA of isatuximab inMM, and uncovered a direct association between the levels ofCD38 expression and the mechanisms triggered by isatuximab;accordingly, ADCC emerges as the most prevalent effector mech-anism by which isatuximab eliminates tumor PCs.

An antigen that can be effectively targeted by a cancer-specificmAb needs to be found on the surface of tumor cells in numbershigh enough to trigger one ormore effectormechanisms.Here, weshowed that continuous exposure to effective concentrations ofisatuximab does not result in a decrease of CD38 numbers on thesurface of MM cells, which contrasts with daratumumab-inducedpolar aggregates and release of CD38 in microvesicles almostimmediately after antigen–antibody binding in vitro (25). Thesefindings suggest that sequential analyses of antigen saturation andturnover could be valuable to design cost-effective anti-CD38treatment schedules.

Some mAbs can induce tumor cell death in the absence ofimmune effector mechanisms; the strength of this effect variesconsiderably depending on the mAb, the target antigen and thetarget cell (32, 33). Direct killing by daratumumab (upon cross-linking) and isatuximab (without cross-linking) has been previ-ously reported, which is in contrast with the data shown here.However, such results were obtained in Burkitt's lymphoma orCD38-overexpressing MM cell lines (i.e., following lentiviraltransduction; refs. 10, 23, 24), and it should be noted that, ascompared with normal PCs, CD38 density is downregulated inclonal PCs from nearly half of patients withMM (8), to levels thatare similar to that of parental (non-transduced) MM cell lines

Figure 6.

Schematic representation of the different mechanisms of action possibly triggered by isatuximab according to the levels of CD38 expression in normal and tumorcells. Wemeasured, in normal bone marrow samples from healthy individuals (n¼ 3), the levels of CD38 expression in T-, NK- and B-lymphocyte subsets, normalPCs, distinct myeloid cells and nucleated red blood cells. Using the Infinicyt software (Cytognos; Salamanca, Spain), we merged the FCS files from normal bonemarrow samples with those from H929, MM1S, OPM2, and RPMI-8226 cell lines, as well as from bone marrow samples from 13 patients with MM. The levels ofCD38 expression (measured with the clone HB7 conjugated with APCH7; BD Biosciences) are represented by population-band histograms and ordered from thelowest to highest expression detected among normal bone marrow cells, MM cell lines, and primary tumor PCs. Of note, patient MM-01 was studied at relapseafter being treated with daratumumab. Levels of CD38 expression required to trigger different MoA induced by isatuximab are represented by dashed lines as aschematic representation in accordance with the results reported in this study.

Moreno et al.

Clin Cancer Res; 25(10) May 15, 2019 Clinical Cancer Research3184

on April 30, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 28, 2019; DOI: 10.1158/1078-0432.CCR-18-1597

Page 10: The Mechanism of Action of the Anti-CD38 Monoclonal ... · understanding about the MoA of novel mAbs is warranted, because precise identification of the (one or more) effector mechanisms

(Fig. 6).Here,we showed that isatuximabhadnoeffect onviability,proliferation and survival of four different MM cell lines withvariable levels of CD38 expression. It was also surprising to seethat isatuximab does not induce gene expression changes in MMcells with normal CD38 expression. Functions ascribed to CD38include receptor-mediated adhesion, signaling events, or bifunc-tional ectoenzymatic activities that contribute to intracellular cal-cium mobilization (34), although none of these functions havebeen actually demonstrated on MM cells. Furthermore, given theend-stage long-lived and quiescent phenotype of bone marrowPCs, it could be hypothesized that rather than signaling, CD38 actsas a marker of adhesion to the bone marrow stroma (35, 36).Interestingly, we demonstrated that tumor cells became moresensitive to the combined administration of bortezomib anddexamethasone when CD38 was blocked by isatuximab; however,those findings appear to be unrelated to a disruption of tumor-stroma cell adhesion. Thus, other functions ascribed to CD38 suchas its ectoenzymatic activity should be considered. CD38 togetherwithCD203a (PC-1) andCD73mayflank theCD39/CD73 canon-ical pathway in the production of adenosine (ADO), which couldfavor tumor survival (37, 38). Accordingly, Morandi and collea-gues (39) unveiled thatMMPCs express high levels of ectoenzymescontributing tohigher catabolismofADO, andproductionofADOhas been demonstrated in cultures of MM and stromal celllines (25). Notwithstanding, our results suggest that direct apo-ptosis triggered by isatuximab may only occur in selected patients(i.e., those in which CD38 is expressed at elevated levels).

The ability of a given mAb to fix complement and to induceCDC is partly dependent on antigen concentration, the orienta-tion of the antigen in the membrane, and if the antigen is presentin the surface as amonomer or polymer. CDC can also depend onthemAb isotype and the characteristics of the target cell, includingwhether the malignant cell expresses complement-regulatoryproteins (3). Here, we showed that isatuximab triggered thedeposition of C3-related fragments (i.e., a pre-requisite for CDC)in less than half of the MM patient samples included in thisstudy. Although CDC has been previously ascribed as a killingmechanism to daratumumab and isatuximab, this has beendemonstrated in Burkitt's lymphoma cell lines (11, 23) and inprimary patient samples without depletion of NK lymphocytes(23, 29, 30); thus, the contribution of ADCC to the killing of MMcells in the later experiments cannot be ruled out, as depositionof C3 was not measured. Furthermore, surface expression ofcomplement inhibitory proteins such as CD46, CD55, and CD59was not associated with clinical response to daratumumab in theGEN501 and SIRIUS studies (29). Although the results obtainedwith MM cell lines should be interpreted with caution, ourfindings suggest that CDC may occur only in selected patients(i.e., those in which CD38 is expressed at elevated levels).

mAbs can induce cytotoxicity by binding to FcRs, which areexpressed by a variety of immune effector cells, includingNK lymphocytes as well as macrophages (i.e., ADCC and ADCP,respectively; ref. 3). ADCP triggered by daratumumab and isatux-imab has also only been demonstrated against Burkitt's lympho-ma cell lines (12, 23) and in primary patient samples withoutdepletion of NK lymphocytes (12). Here, we showed that isatux-imab only induced significant ADCP in CD38hi MM cells. Bycontrast, the extent of ADCC was significant in all but H929 cells(the onewith the lowest anti-CD38 SABC), and detectable againsttumor PCs in most patient samples (11 out of 13, Fig. 4B).Interestingly, the extent of ADCC triggered by isatuximab against

primary tumor PCs (median of 51%) was similar to that reportedfor daratumumab in other preclinical studies (11, 13, 17, 30),suggesting that in the event of different efficacy betweenboth anti-CD38 mAbs, such difference would be related to mechanismsother than ADCC.

It has been reported that daratumumab depletes CD38þve

immune regulatory cells which have substantially lower MFIlevels of CD38 as compared with normal and tumor PCs as wellas MM cell lines (Fig. 6; ref. 14). These observations are surprisingbecause daratumumab (refs. 40, 41; or isatuximab; ref. 22) hasnot shown significant hematologic toxicity, suggesting that mostmechanisms triggered by these drugs require high CD38 densityand spare all other cells with considerable lower MFI levelsof CD38 (14). Here, we demonstrated that, although isatuxi-mab significantly killed primary tumor PCs, it also depletedB-lymphocyte precursors and basophils. The density of CD38 inB-lymphocyte precursors and basophils are the second and thirdhighest (after PCs) among all bonemarrow cells (Fig. 6), andover-laps with that found in immature/transitional B lymphocytes(42, 43), which are also depleted by daratumumab (14).Altogether, these data suggest that anti-CD38 mAbs can targetB lymphocytes with elevated levels of CD38, but although thismay be valuable if these cells are immune suppressive (14), itcould also delay the regeneration of mature B lymphocytes(including normal PCs andnormal immunoglobulin production;ref. 44). By contrast, and as expected on the basis of the observa-tion that isatuximab had no effect on H929 MM cells (Fig. 6),other hematopoietic cells with CD38MFI levels below the formerwere not depleted by the drug.Namely, Tregswere not depleted byisatuximab in our experimental conditions, which supports thefindings of Krejcik and colleagues (14) that only rareCD38þve andnot CD38�/lo Tregs were eliminated during treatment with dar-atumumab. Similarly, there were no significant differences in thepercentage of neutrophils and monocytes after exposure to isatux-imab, consistent with the low levels of CD38 expression found inthese cell types (Fig. 6). Because daratumumab induces release ofCD38 from the surface of MM and other CD38þve hematopoieticcells (15, 25, 29), it is uncertain if the drug depletes CD38þve

immune regulatory cells, or if these cells becomeundetectable afterrelease of CD38 from their surface (in this case, monitoring ofCD38 with another mAb such as HuMax-003 would render false-negative results). Accordingly, the EuroFlow uses a multiepitopeanti-CD38 antibody and simultaneous surface plus intracellularstaining tomeasure CD38 expression duringMRD assessment (9).That notwithstanding, it cannot be excluded that other mechan-isms beyond cell depletion induced by anti-CD38 mAbs canlead to decreased levels of immune suppressive cells as well asenhanced adaptive immunity (e.g., immune modulation afterrobust lymphocyte activation). Altogether, these findings urge forin-depth and longitudinal monitoring of patients with MM (i.e.,from baseline to MRD and eventually disease progression) to fullyunderstand both tumor and immune cell mechanisms behind theresponse and resistance to anti-CD38 mAbs.

Another interesting finding of this study was that, whereas NK-lymphocytes emerged as the most relevant immune subset forthe efficacy of isatuximab, anti-CD38 mAbs may induce theirdepletion through exhaustion and, eventually, CD38/SLAMF7-mediated phagocytosis (7). These results, if appliedmore broadly,could help to explain the rapid decrease in absolute counts ofNK lymphocytes after the first infusions of daratumumab inpatients enrolled into the GEN501, GEN503 and SIRIUS

Mechanism of Action of Isatuximab in Myeloma

www.aacrjournals.org Clin Cancer Res; 25(10) May 15, 2019 3185

on April 30, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 28, 2019; DOI: 10.1158/1078-0432.CCR-18-1597

Page 11: The Mechanism of Action of the Anti-CD38 Monoclonal ... · understanding about the MoA of novel mAbs is warranted, because precise identification of the (one or more) effector mechanisms

studies (14, 15). Accordingly, drugs able to enhance or prolongthe functionality of NK lymphocytes (e.g., lenalidomide; refs. 20,22; or pomalidomide; ref. 19) emerge as attractive partners tocombine with anti-CD38mAbs. On the basis of the upregulationof CD137 expression on NK lymphocytes following CD16 liga-tion to isatuximab, we explored whether the combination of anti-CD38 plus anti-CD137 mAbs could increase activation andprolong the lifespan of NK lymphocytes; however, our resultswere not comparable to those reported in lymphoma withCD137 stimulation and anti-CD20 mAbs (45). Similarly, wefound that pretreatment with lenalidomide did not increaseisatuximab-mediated activation of NK lymphocytes or prevent-ed their exhaustion and death. Thus, further investigations arewarranted to assess if immunotherapies other than immunomodu-latory drugs can potentiate ADCC, as well as to define if there is acorrelation between the absolute counts of NK lymphocytes andpatients' response to anti-CD38 mAb therapy.

Greater knowledge about the immune effector mechanisms ofanti-CD38mAbs is a prerequisite for better prediction of patients'response and optimal monitoring of treatment effects. Here, weperformed a comprehensive analysis about the MoA of isatux-imab in MM; our results underline similarities and differencesbetween isatuximab and other anti-CD38 mAbs. They also pro-vide information that could explain, at least in part, why patientswith lower expression of CD38 may respond poorly to thesedrugs compared with patients with higher CD38 expression.We also provide new insight about the paradoxical depletion ofNK lymphocytes following treatment with anti-CD38 mAbs;further development of therapeutic strategies to overcome thisphenomenon should become a priority.

Disclosure of Potential Conflicts of InterestP. Rodriguez-Otero reports receiving speakers bureau honoraria from Cel-

gene, Janssen, and Bristol-Myers Squibb, and is a consultant/advisory boardmember for Celgene, Janssen, and Takeda. C. Panizo reports receiving speakersbureau honoraria from Roche Pharma, Janssen, and Bristol-Myers Squibb, andis a consultant/advisory board member for Takeda and Janssen. J.F. San-Miguelis a consultant/advisory board member for Takeda, Janssen, Sanofi, Bristol-Myers Squibb, Amgen,Novartis, andCelgene. B. Paiva reports receiving speakersbureau honoraria from Celgene, is a consultant/advisory board member forAmgen, Celgene, Janssen, Karyopharm, Takeda, and Sanofi, and reports receiv-ing commercial research support from Sanofi, Celgene, and Takeda. No poten-tial conflicts of interest were disclosed by the other authors.

Authors' ContributionsConception and design: C. Perez, A. Zabaleta, J.F. San-Miguel, B. PaivaDevelopment of methodology: L. Moreno, C. Perez, A. Zabaleta, I. Manrique,M. Lasa, P. Maiso, S. Garate, B. PaivaAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): C. Perez, A. Zabaleta, I. Manrique, D. Alignani,D. Ajona, L. Blanco, M. Lasa, I. Rodriguez, S. Garate, P. Rodriguez-Otero,F. Prosper, B. PaivaAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): L. Moreno, C. Perez, A. Zabaleta, I. Manrique,D. Ajona, M. Lasa, S. Garate, V. Segura, C. Moreno, J. Merino, J.F. San-Miguel,B. PaivaWriting, review, and/or revision of the manuscript: L. Moreno, C. Perez,A. Zabaleta, I. Manrique, D. Ajona, M. Lasa, T. Jelinek, P. Rodriguez-Otero,C. Panizo, F. Prosper, J.F. San-Miguel, B. PaivaAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): D. Alignani, B. PaivaStudy supervision: B. Paiva

AcknowledgmentsThis studywas supported by theCentro de Investigaci�onBiom�edica enRed—

�Area de Oncología—del Instituto de Salud Carlos III (CIBERONC; CB16/12/00369 CB16/12/00489 and CB16/12/00443), formerly named as CooperativeResearch Thematic Network (grant numbers RD12/0036/0058 and RD12/0036/0068) of the Red de Cancer (Cancer Network of Excellence); Institutode Salud Carlos III/Subdirecci�on General de Investigaci�on Sanitaria (FIS num-ber PI17/01243 and FIS number PI17/00411); Asociaci�on Espa~nola Contra elC�ancer (GCB120981SAN and Accelerator); and Fundaci�on Areces. The authorsalso acknowledge theNavarra Blood and Tissue Bank, NavarrabiomedBiobank,Navarra Health Department for the samples from healthy donors. This studywas supported internationally by the Black Swan Research Initiative of theInternational Myeloma Foundation, the European Research Council (ERC)2015 Starting Grant (MYELOMANEXT), the Multiple Myeloma Research Foun-dation (MMRF) Immunotherapy Networks of Excellence, and the 2017 Euro-pean Hematology Association (EHA) non-clinical advanced research grant(3680644). This workwas also supported by an independent grant from Sanofi,and the authors would like to acknowledge the intellectual input from FranciscoAldrian and Zhili Song.

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received May 22, 2018; revised November 26, 2018; accepted January 14,2019; published first January 28, 2019.

References1. Bachireddy P, Burkhardt UE, Rajasagi M, Wu CJ. Haematological malig-

nancies: at the forefront of immunotherapeutic innovation.Nat RevCancer2015;15:201–15.

2. Rodriguez-Otero P, Paiva B, Engelhardt M, Prosper F, San Miguel JF. Isimmunotherapy here to stay in multiple myeloma? Haematologica 2017;102:423–432.

3. Weiner GJ. Building better monoclonal antibody-based therapeutics.Nat Rev Cancer 2015;15:361–70.

4. Tutt AL, French RR, Illidge TM, Honeychurch J, McBride HM, Penfold CA,et al. Monoclonal antibody therapy of B cell lymphoma: signaling activityon tumor cells appears more important than recruitment of effectors.J Immunol 1998;161:3176–85.

5. Taylor RP. Of mice and mechanisms: identifying the role of complement inmonoclonal antibody-based immunotherapy.Haematologica 2006;91:146a.

6. Clynes R, Takechi Y, Moroi Y, Houghton A, Ravetch J V. Fc receptors arerequired in passive and active immunity to melanoma. Proc Natl Acad SciU S A 1998;95:652–6.

7. Chen J, Zhong M-C, Guo H, Davidson D, Mishel S, Lu Y, et al. SLAMF7 iscritical for phagocytosis of haematopoietic tumour cells viaMac-1 integrin.Nature 2017;544:493–7.

8. Arana P, Paiva B, Cedena M-T, Puig N, Cordon L, Vidriales M-B, et al.Prognostic value of antigen expression inmultiple myeloma: a PETHEMA/GEM study on 1265 patients enrolled in four consecutive clinical trials.Leukemia 2018;32:971–8.

9. Flores-Montero J, Flores LS, Paiva B, Puig N, Garcia-Sanchez O, Bottcher S,et al. Next generation flow (NGF) for highly sensitive and standardizeddetection of minimal residual disease in multiple myeloma. Leukemia2017;31:2094–103.

10. OverdijkMB, Jansen JH, NederendM, Lammerts van Bueren JJ, Groen RW,Parren PW, et al. The Therapeutic CD38 monoclonal antibody daratumu-mab induces programmed cell death via fcgamma receptor-mediatedcross-linking. J Immunol 2016;197:807–13.

11. deWeers M, Tai Y-T, van der VeerMS, Bakker JM, Vink T, Jacobs DCH, et al.Daratumumab, a novel therapeutic human CD38 monoclonal antibody,

Moreno et al.

Clin Cancer Res; 25(10) May 15, 2019 Clinical Cancer Research3186

on April 30, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 28, 2019; DOI: 10.1158/1078-0432.CCR-18-1597

Page 12: The Mechanism of Action of the Anti-CD38 Monoclonal ... · understanding about the MoA of novel mAbs is warranted, because precise identification of the (one or more) effector mechanisms

induces killing of multiple myeloma and other hematological tumors.J Immunol 2011;186:1840–8.

12. Overdijk MB, Verploegen S, Bogels M, van Egmond M, Lammerts vanBueren JJ, Mutis T, et al. Antibody-mediated phagocytosis contributes tothe anti-tumor activity of the therapeutic antibody daratumumab inlymphoma and multiple myeloma. MAbs 2015;7:311–21.

13. Nijhof IS, Groen RW, Noort WA, van Kessel B, de Jong-Korlaar R, Bakker J,et al. Preclinical evidence for the therapeutic potential of CD38-targetedimmuno-chemotherapy in multiple myeloma patients refractory to lena-lidomide and bortezomib. Clin Cancer Res 2015;21:2802–10.

14. Krejcik J, Casneuf T, Nijhof IS, Verbist B, Bald J, Plesner T, et al. Daratu-mumab depletes CD38þ immune regulatory cells, promotes T-cell expan-sion, and skews T-cell repertoire in multiple myeloma. Blood 2016;128:384–94.

15. Krejcik J, Frerichs KA, Nijhof IS, van Kessel B, van Velzen J, Bloem AC, et al.Monocytes and granulocytes reduce CD38 expression levels on myelomacells in patients treated with daratumumab. Clin Cancer Res 2017;23:7498–511.

16. Kastritis E, Dimopoulos MA. Daratumumab combinations: what can welearn? Blood 2017;130:957–8.

17. Nijhof IS, Lammerts van Bueren JJ, vanKessel B, Andre P,Morel Y, LokhorstHM, et al. Daratumumab-mediated lysis of primary multiple myelomacells is enhanced in combination with the human anti-KIR antibodyIPH2102 and lenalidomide. Haematologica 2015;100:263–8.

18. Chiu C, Casneuf T, Axel A, Lysaght A, Bald J, Khokhar NZ, et al. Daratu-mumab in combination with lenalidomide plus dexamethasone inducesclonality increase and T-cell expansion: results from a phase 3 randomizedstudy (POLLUX). Blood 2016;128:4531.

19. Plesner T, ArkenauH-T,Gimsing P, Krejcik J, LemechC,MinnemaMC, et al.Phase 1/2 study of daratumumab, lenalidomide, and dexamethasone forrelapsed multiple myeloma. Blood 2016;128:1821–8.

20. Dimopoulos MA, Oriol A, Nahi H, San-Miguel J, Bahlis NJ, Usmani SZ,et al. Daratumumab, lenalidomide, and dexamethasone for multiplemyeloma. N Engl J Med 2016;375:1319–31.

21. Chari A, SuvannasankhaA, Fay JW, Arnulf B, Kaufman JL, Ifthikharuddin JJ,et al. Daratumumab plus pomalidomide and dexamethasone in relapsedand/or refractory multiple myeloma. Blood 2017;130:974–81.

22. Martin T, Baz R, BensonDM, Lendvai N,Wolf J,Munster P, et al. A phase 1bstudy of isatuximab plus lenalidomide and dexamethasone for relapsed/refractory multiple myeloma. Blood 2017;129:3294–303.

23. Deckert J, Wetzel MC, Bartle LM, Skaletskaya A, Goldmacher VS, ValleeF, et al. SAR650984, a novel humanized CD38-targeting antibody,demonstrates potent antitumor activity in models of multiple myelomaand other CD38þ hematologic malignancies. Clin Cancer Res 2014;20:4574–83.

24. Jiang H, Acharya C, An G, Zhong M, Feng X, Wang L, et al. SAR650984directly induces multiple myeloma cell death via lysosomal-associatedand apoptotic pathways, which is further enhanced by pomalidomide.Leukemia 2016;30:399–408.

25. Horenstein AL, Chillemi A, Quarona V, Zito A, Roato I, Morandi F, et al.NAD(þ)-Metabolizing ectoenzymes in remodeling tumor-host interac-tions: the human myeloma model. Cells 2015;4:520–37.

26. Waldschmidt JM, Simon A, Wider D, Muller SJ, Follo M, Ihorst G, et al.CXCL12 and CXCR7 are relevant targets to reverse cell adhesion-mediateddrug resistance in multiple myeloma. Br J Haematol 2017;179:36–49.

27. McMillin DW, Delmore J, Weisberg E, Negri JM, Geer DC, Klippel S, et al.Tumor cell-specific bioluminescence platform to identify stroma-inducedchanges to anticancer drug activity. Nat Med 2010;16:483–9.

28. van de Donk NWCJ, Richardson PG, Malavasi F. CD38 antibodies inmultiple myeloma: back to the future. Blood 2018;131:13–29.

29. Nijhof IS, Casneuf T, van Velzen J, van Kessel B, Axel AE, Syed K, et al. CD38expression and complement inhibitors affect response and resistance todaratumumab therapy in myeloma. Blood 2016;128:959–70.

30. Nijhof IS, Groen RWJ, Lokhorst HM, van Kessel B, Bloem AC, van Velzen J,et al. Upregulation of CD38 expression on multiple myeloma cells by all-trans retinoic acid improves the efficacy of daratumumab. Leukemia 2015;29:2039–49.

31. van der Veer MS, de Weers M, van Kessel B, Bakker JM, Wittebol S, ParrenPWHI, et al. Towards effective immunotherapy of myeloma: enhancedelimination of myeloma cells by combination of lenalidomide with thehuman CD38monoclonal antibody daratumumab. Haematologica 2011;96:284–90.

32. Beers SA, Chan CHT, James S, French RR, Attfield KE, Brennan CM, et al.Type II (tositumomab) anti-CD20 monoclonal antibody out performstype I (rituximab-like) reagents in B-cell depletion regardless of comple-ment activation. Blood 2008;112:4170–7.

33. Shan D, Ledbetter JA, Press OW. Apoptosis of malignant human B cells byligation of CD20 with monoclonal antibodies. Blood 1998;91:1644–52.

34. Deaglio S, Mehta K, Malavasi F. Human CD38: a (r)evolutionary story ofenzymes and receptors. Leuk Res 2001;25:1–12.

35. Deaglio S, Aydin S, Vaisitti T, Bergui L, Malavasi F. CD38 at the junctionbetween prognostic marker and therapeutic target. Trends Mol Med 2008;14:210–8.

36. Deaglio S, Morra M, Mallone R, Ausiello CM, Prager E, Garbarino G, et al.Human CD38 (ADP-ribosyl cyclase) is a counter-receptor of CD31, an Igsuperfamily member. J Immunol 1998;160:395–402.

37. Vijayan D, Young A, Teng MWL, Smyth MJ. Targeting immunosuppressiveadenosine in cancer. Nat Rev Cancer 2017;17:709–24.

38. Chen L, Diao L, Yang Y, Yi X, Rodriguez BL, Li Y, et al. CD38-mediatedimmunosuppression as a mechanism of tumor cell escape from PD-1/PD-L1 blockade. Cancer Discov 2018;8:1156–75.

39. Morandi F, Marimpietri D, Horenstein AL, Bolzoni M, Toscani D, Costa F,et al. Microvesicles released from multiple myeloma cells are equippedwith ectoenzymes belonging to canonical and non-canonical adeno-sinergic pathways and produce adenosine from ATP and NAD.Oncoimmunology 2018;7:e1458809.

40. Lonial S, Weiss BM, Usmani SZ, Singhal S, Chari A, Bahlis NJ, et al.Daratumumab monotherapy in patients with treatment-refractory multi-ple myeloma (SIRIUS): an open-label, randomised, phase 2 trial. Lancet2016;387:1551–60.

41. Lokhorst HM, Plesner T, Laubach JP, Nahi H, Gimsing P, HanssonM, et al.Targeting CD38 with daratumumab monotherapy in multiple myeloma.N Engl J Med 2015;373:1207–19.

42. Perez-Andres M, Paiva B, Nieto WG, Caraux A, Schmitz A, Almeida J, et al.Human peripheral blood B-cell compartments: a crossroad in B-cell traffic.Cytometry B Clin Cytom 2010;78:S47–60.

43. Caraux A, Klein B, Paiva B, Bret C, Schmitz A, Fuhler GM, et al. Circulatinghuman B and plasma cells. Age-associated changes in counts and detailedcharacterization of circulating normal CD138- and CD138þ plasma cells.Haematologica 2010;95:1016–20.

44. Paiva B, Cedena MT, Puig N, Arana P, Vidriales MB, Cordon L, et al.Minimal residual disease monitoring and immune profiling in multiplemyeloma in elderly patients. Blood 2016;127:3165–74.

45. Kohrt HE, Houot R, Goldstein MJ, Weiskopf K, Alizadeh AA, Brody J, et al.CD137 stimulation enhances the antilymphoma activity of anti-CD20antibodies. Blood 2011;117:2423–32.

www.aacrjournals.org Clin Cancer Res; 25(10) May 15, 2019 3187

Mechanism of Action of Isatuximab in Myeloma

on April 30, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 28, 2019; DOI: 10.1158/1078-0432.CCR-18-1597

Page 13: The Mechanism of Action of the Anti-CD38 Monoclonal ... · understanding about the MoA of novel mAbs is warranted, because precise identification of the (one or more) effector mechanisms

2019;25:3176-3187. Published OnlineFirst January 28, 2019.Clin Cancer Res   Laura Moreno, Cristina Perez, Aintzane Zabaleta, et al.   Isatuximab in Multiple MyelomaThe Mechanism of Action of the Anti-CD38 Monoclonal Antibody

  Updated version

  10.1158/1078-0432.CCR-18-1597doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2019/02/08/1078-0432.CCR-18-1597.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/25/10/3176.full#ref-list-1

This article cites 45 articles, 24 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/25/10/3176.full#related-urls

This article has been cited by 1 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/25/10/3176To request permission to re-use all or part of this article, use this link

on April 30, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 28, 2019; DOI: 10.1158/1078-0432.CCR-18-1597