19
the bmj | BMJ 2020;370:m3176 | doi: 10.1136/bmj.m3176 1 STATE OF THE ART REVIEW Emerging immunotherapies in multiple myeloma Urvi A Shah, 1 Sham Mailankody 1 1 Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 530 East 74th Street, New York, NY 10021, USA Correspondence to: U A Shah [email protected] Cite this as: BMJ 2020;370:m3176 http://dx.doi.org/10.1136/bmj.m3176 Series explanation: State of the Art Reviews are commissioned on the basis of their relevance to academics and specialists in the US and internationally. For this reason they are written predominantly by US authors. Introduction Multiple myeloma, a malignancy of unregulated plasma cell growth, is the second most comm- on hematologic cancer among adults. 1 Multiple myeloma is characterized by progressive immune dysfunction (innate and adaptive), and over the past two decades we have seen substantial progress in treatment, including immunotherapies. 2 These newer treatments have improved outcomes with deeper responses and longer survival. Despite these advances, most patients eventually relapse, and multiple myeloma is therefore still considered an incurable disease. This review focuses on immunotherapies approved by the Food and Drug Administration, as well as several emerging treatments that have shown promising results and will likely be part of standard treatment for patients soon. Incidence and prevalence Multiple myeloma accounts for about 1% of all cancers and 10% of all hematologic malignancies. 3 The age standardized incidence rate of the disease worldwide in 2016 was 2.1 per 100 000 people, and about 160 000 patients had newly diagnosed multiple myeloma in 2018. 4 5 In the US, the age standardized incidence rate was higher than the global rate at 7.1 per 100 000 people in 2016, 6 and incident rates have gradually increased, which is consistent with the global trend. 7 The prevalence of this cancer has also increased, given that the median overall survival for patients has doubled in the past decade. 8 9 Sources and selection criteria We identified references for this review by doing a PubMed and Medline search for years 2000-19. We used keywords and combinations of keywords such as multiple myeloma, immunotherapy, CAR T cell, therapy, immune, antibody. We included articles based on the quality of study design and size, favoring randomized controlled trials, reports from large registries, and guidelines. We excluded case reports and papers in non-peer reviewed journals. We screened about 1000 articles of evidence classes I-IV. Studies were prioritized by design, and by patient numbers, quality, and publication date. However, given the breadth of this review and the lack of large studies in some aspects of immunotherapy in multiple myeloma, we included observational and preclinical studies as well as ongoing trials where preliminary results are available as meeting abstracts. We highlighted the limitations of these studies where relevant. Current immunotherapies Allogeneic stem cell transplantation One of the earliest immunotherapies used to treat hematologic cancers is allogeneic hematopoietic stem cell transplantation (alloHSCT), which has shown potential in a subset of patients with multiple myeloma (fig 1). The treatment offers the opportunity ABSTRACT Despite considerable advances in treatment approaches in the past two decades, multiple myeloma remains an incurable disease. Treatments for myeloma continue to evolve with many emerging immunotherapies. The first immunotherapy used to treat hematologic cancers, including multiple myeloma, was an allogeneic stem cell transplant. In the mid-2000s, immunomodulatory drugs thalidomide, lenalidomide, and subsequently pomalidomide were proven to be effective in multiple myeloma and substantially improved survival. The next wave of immunotherapies for multiple myeloma included the monoclonal antibodies daratumumab and elotuzumab, which were approved by the Food and Drug Administration in 2015. Subsequently, a variety of immunotherapies have been developed for multiple myeloma, including chimeric antigen receptor T cells, bispecific antibodies, antibody drug conjugates, and checkpoint inhibitors. Many of these emerging treatments target the B cell maturation antigen, which is expressed on plasma cells, although several other novel receptors are also being studied. This review summarizes the evidence of these various immunotherapies, their mechanism of action, and data from clinical trials regarding the treatments’ safety and efficacy. on 8 February 2021 by guest. Protected by copyright. http://www.bmj.com/ BMJ: first published as 10.1136/bmj.m3176 on 21 September 2020. Downloaded from

Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

the bmj | BMJ 2020;370:m3176 | doi: 10.1136/bmj.m3176 1

State of the art reVIeW

Emerging immunotherapies in multiple myelomaUrvi A Shah,1 Sham Mailankody1

1Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 530 East 74th Street, New York, NY 10021, USACorrespondence to: U A Shah [email protected] this as: BMJ 2020;370:m3176 http://dx.doi.org/10.1136/bmj.m3176

Series explanation: State of the Art Reviews are commissioned on the basis of their relevance to academics and specialists in the US and internationally. For this reason they are written predominantly by US authors.

IntroductionMultiple myeloma, a malignancy of unregulated plasma cell growth, is the second most comm­on hematologic cancer among adults.1 Multiple myeloma is characterized by progressive immune dysfunction (innate and adaptive), and over the past two decades we have seen substantial progress in treatment, including immunotherapies.2 These newer treatments have improved outcomes with deeper responses and longer survival. Despite these advances, most patients eventually relapse, and multiple myeloma is therefore still considered an incurable disease. This review focuses on immunotherapies approved by the Food and Drug Administration, as well as several emerging treatments that have shown promising results and will likely be part of standard treatment for patients soon.

Incidence and prevalenceMultiple myeloma accounts for about 1% of all cancers and 10% of all hematologic malignancies.3 The age standardized incidence rate of the disease worldwide in 2016 was 2.1 per 100 000 people, and about 160 000 patients had newly diagnosed multiple myeloma in 2018.4 5 In the US, the age standardized incidence rate was higher than the global rate at 7.1 per 100 000 people in 2016,6 and incident rates have gradually increased, which is consistent with the global trend.7 The prevalence of this cancer has also increased, given that the median

overall survival for patients has doubled in the past decade.8 9

Sources and selection criteriaWe identified references for this review by doing a PubMed and Medline search for years 2000­19. We used keywords and combinations of keywords such as multiple myeloma, immunotherapy, CAR T cell, therapy, immune, antibody. We included articles based on the quality of study design and size, favoring randomized controlled trials, reports from large registries, and guidelines. We excluded case reports and papers in non­peer reviewed journals. We screened about 1000 articles of evidence classes I­IV. Studies were prioritized by design, and by patient numbers, quality, and publication date. However, given the breadth of this review and the lack of large studies in some aspects of immunotherapy in multiple myeloma, we included observational and preclinical studies as well as ongoing trials where preliminary results are available as meeting abstracts. We highlighted the limitations of these studies where relevant.

Current immunotherapiesAllogeneic stem cell transplantation One of the earliest immunotherapies used to treat hematologic cancers is allogeneic hematopoietic stem cell transplantation (alloHSCT), which has shown potential in a subset of patients with multiple myeloma (fig 1). The treatment offers the opportunity

ABSTRACT

Despite considerable advances in treatment approaches in the past two decades, multiple myeloma remains an incurable disease. Treatments for myeloma continue to evolve with many emerging immunotherapies. The first immunotherapy used to treat hematologic cancers, including multiple myeloma, was an allogeneic stem cell transplant. In the mid-2000s, immunomodulatory drugs thalidomide, lenalidomide, and subsequently pomalidomide were proven to be effective in multiple myeloma and substantially improved survival. The next wave of immunotherapies for multiple myeloma included the monoclonal antibodies daratumumab and elotuzumab, which were approved by the Food and Drug Administration in 2015. Subsequently, a variety of immunotherapies have been developed for multiple myeloma, including chimeric antigen receptor T cells, bispecific antibodies, antibody drug conjugates, and checkpoint inhibitors. Many of these emerging treatments target the B cell maturation antigen, which is expressed on plasma cells, although several other novel receptors are also being studied. This review summarizes the evidence of these various immunotherapies, their mechanism of action, and data from clinical trials regarding the treatments’ safety and efficacy.

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 2: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

2 doi: 10.1136/bmj.m3176 | BMJ 2020;370:m3176 | the bmj

for sustained disease control with an immune mediated graft versus myeloma effect by establishing donor chimerism. AlloHSCTs are usually performed after achieving optimal cytoreduction with high dose chemotherapy or an autologous hematopoietic stem cell transplantation (autoHSCT). AlloHSCTs (or autoHSCTs and alloHSCTs in tandem) are an option for fit patients with relapsed disease that is refractory to standard options, and in rare situations such as clinical trials for young and fit patients with newly diagnosed multiple myeloma with high risk disease as a consolidation approach. This option, however, is not routinely recommended.10 11

Over the past decade, considerable improvements have been made in supportive care, typing, and use of the more tolerable reduced intensity conditioning regimens compared with the myeloablative regimens as conditioning treatment. Additional options for immunotherapy at relapse after allogeneic trans­plant include donor lymphocyte infusions, immuno­modulatory drug maintenance, and withdrawal of immune suppression.12 However, allogeneic trans­ plants do not have a defined role in multiple myeloma treatment, owing to the high mortality related to treatment and limited efficacy as well as the conflicting reports regarding long term outcomes from prospective studies.12 13 Another reason that alloHSCTs have fallen out of favor is the availability of effective novel treatments that are easier to tolerate, many of which we will discuss in this review.

Immunomodulatory imide drugs Until the early 2000s, treatment options for multiple myeloma were limited. For patients diagnosed before 2000, the relative survival at five years was less than 35%.6 The first use of thalidomide, an immunomodulatory imide drug, in the treatment of multiple myeloma in 1997 and its subsequent FDA approval in 2006 started an era of new drug approvals for myeloma including immunologic and other treatments (fig 1). A detailed review of currently available myeloma treatments is beyond the scope of this article, but we will highlight important studies informing the use of immunomodulatory imide drugs and monoclonal antibodies for treatment of multiple myeloma.

Immunomodulatory imide drugs that have been approved by the FDA in addition to thalidomide include lenalidomide in 2006 and more recently pomalidomide in 2013. These drugs exert their anti­tumor activity via multiple mechanisms. They bind to cereblon and thus increase the ubiquitination and proteasomal degradation of key transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) that are important for the development of B cells and long lived plasma cells. They also have immunomodulatory effects including enhanced natural killer cell and T cell activity and an increase in interleukin 2 and inter­feron γ.14 15 Immunomodulatory imide drugs have been studied as a single agent16 17 and in combination with proteasome inhibitors18­23 in relapsed and refractory multiple myeloma, in newly diagnosed

25%

35%

45%

55%

Timeline of drug discovery and year of multiple myeloma diagnosis (by decade)

1960s 1970s 1980s 2000s 2010s 2020s

Rela

tive

surv

ival

at fi

ve y

ears

(%),

base

d on

yea

r of d

iagn

osis

Allogeneic transplantsAutologous transplants

Alkylators

ImmunotherapyOther treatments

Chimeric antigen receptor T cellsBispecific antibodies

Antibody drug conjugates

Monoclonal antibodies

Immunomodulatory drugs

Proteasome inhibitors

Steroids

Fig 1 | Multiple myeloma treatments—timeline of drug discovery and five year relative survival (using data from the Surveillance, Epidemiology, and Ends Results program).6 Data for year of diagnosis and relative survival are: 1975, 26.5% (observed); 1980, 26.0% (observed); 1985, 27.4% (observed); 1990, 29.9% (observed); 1995, 33.5% (observed); 2000, 34.6% (observed); 2005, 47.1% (observed); 2010, 53.6% (observed); 2015, 55.3% (modelled)

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 3: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

the bmj | BMJ 2020;370:m3176 | doi: 10.1136/bmj.m3176 3

multiple myeloma,24­26 as maintenance treatment after induction and autologous transplantation,27­29 and in smoldering multiple myeloma.30 31 Iberdomide (CC­220) and CC­92480 are novel cereblon E3 ligase modulators that have shown promise in overcoming prior immunomodulatory drug resistance.32­34

Monoclonal antibodiesIn the past five years, additional immunotherapies became available with the approval of three monoclonal antibodies. Daratumumab (anti­CD38) and elotuzumab (anti­signaling lymphocytic activation molecule F7 (SLAMF7)) were approved by the FDA for the treatment of multiple myeloma in 2015, and isatuximab (anti­CD38) was most recently approved in 2020 (fig 1). These antibodies are effective via multiple processes, with some minor differences between their mechanisms. Similar mechanisms include direct tumor cell apoptosis; immune mediated mechanisms (including complement dependent cytotoxicity; antibody dependent, cell mediated cytotoxicity via natural killer cells; and macrophage mediated, antibody dependent cellular phagocytosis); and immunoregulatory actions such as decreasing regulatory T cells, regulatory B cells, and myeloid derived suppressor cells.35­37 One of the differences is that daratumumab induces apoptosis via Fc receptor mediated crosslinking of tumor bound monoclonal antibodies,38 and isatuximab induces apoptosis without crosslinking via caspase dependent and lysozyme associated pathways leading to homotypic aggregation.39

Daratumumab monotherapy has shown a favorable safety profile and encouraging efficacy in initial phase I and II trials.40­42 It was then combined with standard treatments in randomized phase III studies (that is, immunomodulatory drugs, proteasome inhibitors, and alkylators; table 1). In randomized phase III studies of relapsed and refractory multiple myeloma, daratumumab and dexamethasone were combined with bortezomib (median progression free survival 16.7 v 7.1 months),43 44 lenalidomide (44.5 v 17.5 months),45­47 carfilzomib (not reached v 15.8 months),48 showing significant improvements in overall response rates and progression free survival. Daratumumab has also been combined with pomalidomide, based on phase Ib data61 and a non­randomized, multicohort multicenter phase II study.62 With this drug’s promising results in relapsed and refractory multiple myeloma, trials evaluating daratumumab in transplant ineligible patients with newly diagnosed multiple myeloma showed significantly improved overall response rates and progression free survival. In the MAIA trial, daratumumab was combined with lenalidomide and dexamethasone (not reached v 31.9 months).51 In the ALCYONE trial, daratumumab was combined with bortezomib, melphalan, and prednisone (D­VMP) (36.4 v 19.3 months).49 51 An updated analysis also showed an overall survival benefit in the D­VMP group at a median follow­up of 40.1 months (interquartile range 37.4­43.1), and a significant benefit in overall survival in the D­VMP group (hazard ratio 0.60, 95% confidence interval 0.46 to 0.80, P<0.001).50

Table 1 | Randomized, multicenter phase III trials of monoclonal antibody treatment for multiple myeloma

TrialPatient group (type of disease, No of prior lines of treatment)

Arm (No of patients)*

Overall response rate (%)

Median progression free survival (months)

Hazard ratio (P value)

FDA approval

EMA approval

CASTOR43 44 Relapsed and refractory disease, at least 1 prior line

D-Vd (n=251) 83 16.7 0.31 (P<0.001) Yes YesVd (n=247) 63 7.1

POLLUX45-47 Relapsed and refractory disease, at least 1 prior line

D-Rd (n=286) 93 44.5 0.44 (P<0.001) Yes YesRd (n=283) 76 17.5

CANDOR48 Relapsed and refractory disease, 1-3 prior lines

D-Kd (n=312) 84 Not reached 0.63 (P=0.003) Yes NoKd (n=154) 75 15.8

ALCYONE49 50 Transplant ineligible, newly diagnosed disease

D-VMp (n=350) 91 36.4 0.42 (P<0.001) Yes YesVMp (n=356) 74 19.3

MAIA51 Transplant ineligible, newly diagnosed disease

D-Rd (n=368) 93 Not reached 0.56 (P<0.001) Yes YesRd (n=369) 81 31.9

CASSIOPEIA52 Transplant eligible, newly diagnosed disease

D-VTd (n=543) 93 Not reached 0.47, (P<0.001) Yes YesVTd (n=542) 90 Not reached

GRIFFIN53† Transplant eligible, newly diagnosed disease

D-RVd (n=99) 99 Not reached Not available No NoRVd (n=97) 92 Not reached

COLUMBA54 Relapsed and refractory disease, at least 3 prior lines

SC D (n=263) 41 5.6 0.99 (P=0.93) Yes YesIV D (n=259) 37 6.1

ELOQUENT-255 56 Relapsed and refractory disease, 1-3 prior lines

E-Rd (n=321) 79 19.4 0.71 (P<0.001) Yes YesRd (n=325) 66 14.9

ELOQUENT-3*57 Relapsed and refractory disease, at least 2 prior lines

E-Pd (n=60) 53 10.3 0.54 (P=0.008) Yes YesPd (n=57) 26 4.7

SWOG 121158 High risk, newly diagnosed disease

E-RVd (n=68) 83 34 0.97 (P=0.45) No NoRVd (n=66) 88 31

ICARIA-MM59 Relapsed and refractory disease, at least 2 prior lines

I-Pd (n=154) 63 11.5 0.6 (P=0.001) Yes YesPd (n=153) 32 6.5

IKEMA60 Relapsed and refractory disease, 1-3 prior lines

I-Kd (n=179) 87 Not reached 0.53 (P<0.001) No NoKd (n=123) 83 19.2

FDA=Food and Drug Administration; EMA=European Medicines Agency.*D=daratumumab; V=bortezomib; d=dexamethasone; R=lenalidomide; K=carfilzomib; M=melphalan; p=prednisone; T=thalidomide; SC=subcutaneous; IV=intravenous; E=elotuzumab; I=isatuximab; P=pomalidomide. †Randomized phase II study.

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 4: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

4 doi: 10.1136/bmj.m3176 | BMJ 2020;370:m3176 | the bmj

Daratumumab has also been studied in transplant eligible patients with newly diagnosed multiple myeloma as a quadruple drug combination with bortezomib, thalidomide, and dexamethasone (VTd).52 The patients were treated with four cycles of the VTd combination, followed by autoHSCT and then two cycles of the VTd combination. Daratumumab was added to the six cycles of the VTd combination in the experimental arm (median progression free survival, not reached).52 In a randomized phase II study in transplant eligible patients with newly diagnosed multiple myeloma,53 patients were treated with lenalidomide, bortezomib, and dexamethasone (RVd) with and without daratumumab for four cycles followed by autoHSCT followed by two additional cycles of the RVd combination. Results showed an improvement in the overall response rate, stringent complete response rate, and rate of minimal residual disease negativity (median progression free survival, not reached).53

A randomized phase III trial has shown that subcutaneous daratumumab was non­inferior to intravenous daratumumab in terms of efficacy and pharmacokinetics, and had an improved safety profile in relapsed and refractory multiple myeloma leading to its recent FDA approval in 2020 (median progression free survival 5.6 v 6.1 months).54

Although elotuzumab has limited activity on its own,63 it has shown significantly improved over­all response rates and progression free survival when combined with immunomodulatory drugs lenalidomide in a randomized phase III trial (median progression free survival 19.4 v 14.9 months)55 56 64 and pomalidomide in a randomized phase II trial (10.3 v 4.7 months).57 Only 2% of patients in the elotuzumab arm and 4% in the control arm were exposed to prior daratumumab in the phase II trial.57 Therefore, a recent randomized phase III study in high risk patients with newly diagnosed multiple myeloma compared the addition of elotuzumab to induction and maintenance of the RVd drug combination with no improvement in outcomes.58

Isatuximab is another anti­CD38 antibody that has been studied in combination with lenalidomide and dexamethasone65 as well as pomalidomide and dexamethasone66 in phase Ib studies. Isatuximab and dexamethasone with and without pomalidomide were studied in a randomized phase III study that showed a benefit in progression free survival (11.5 v 6.5 months), which led to their FDA approval in 2020 for relapsed and refractory multiple myeloma. Only one patient in the isatuximab arm had been previously treated with daratumumab.59  67 Isatuximab has been studied in combination with carfilzomib and dexamethasone in another randomized phase III trial, which showed substantial benefit in progression free survival (not reached v 19.2 months).60

Newer immunotherapiesImmunotherapies that are currently being studied in multiple myeloma are discussed in this section

under four headings: checkpoint inhibitors, chimeric antigen receptor (CAR) T cells, bispecific antibodies, and antibody drug conjugates (fig 2).

Checkpoint inhibitorsMalignant plasma cells in most patients with multiple myeloma express the checkpoint programmed death­ligand 1, which is upregulated especially when exposed to inflammatory mediators such as interferon γ. Interaction of this checkpoint molecule with programmed cell death protein 1 on T cells limits their proliferation and cytotoxic activity.68 69

The first study evaluating single agent nivolumab for relapsed multiple myeloma showed a response in only one of 27 patients.70 71 Despite a lack of single agent activity, single arm trials combining checkpoint inhibitors with immunomodulatory imide drugs and dexamethasone because of the potential synergy72 look promising.73 74

With these clinical data, three large randomized phase III trials were halted by the FDA in 2017 because of increased serious adverse events and deaths as well as decreased overall survival in the checkpoint inhibitor arm (pomalidomide and dexamethasone with and without pembrolizumab) in relapsed and refractory multiple myeloma (hazard ratio 1.61, 95% confidence interval 0.91 to 2.85),75 pomalidomide and dexamethasone with and without nivolumab in relapsed and refractory multiple myeloma (1.19, 0.64 to 2.20),76 and lenalidomide and dexamethasone with and without pembrolizumab in transplant ineligible patients with newly diagnosed multiple myeloma (2.06, 0.93 to 4.55)77).

These trials encourage caution with expedited timelines for future combination studies for drugs with limited single agent activity.78 79 Future trials in multiple myeloma will need to be based on sound preclinical and clinical rationale with other partners and be conducted in heavily treated patients (with limited standard options) initially.

Chimeric antigen receptor T cellsCAR T cells are human T cells that have been genetically modified and expanded in the laboratory before they are infused back into patients to target the tumor. The receptor on the surface of CAR T cells that targets the tumor antigens consists of several parts (fig 3): an extracellular, non­major histocompatibility complex restricted, targeting domain, usually derived from a single chain variable fragment of a monoclonal antibody; a spacer region; a transmembrane domain; an intracellular signaling domain including the CD3ζ activation domain; and a costimulatory domain (eg, CD28 or 4­1BB). The single chain variable fragment was originally derived from mice (hence the term chimeric), but many of the newer constructs are fully human.80 81

CD3 positive T cells are obtained from patients (for autologous CAR T cells) or healthy donors (for allogeneic CAR T cells) via a process called leukapheresis. These T cells are expanded manifold

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 5: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

the bmj | BMJ 2020;370:m3176 | doi: 10.1136/bmj.m3176 5

in culture and activated using beads coated with anti­CD3 or anti­CD28 monoclonal antibodies or cell based artificial antigen presenting cells.82 The T cells are then transduced with a vector (usually either lentiviral or retroviral) that carries the gene encoding a receptor to an antigen present on the surface of tumor cells. This manufacturing process takes up to four weeks at a good manufacturing practices facility, and the CAR T cells can then be stored until needed by the patient. This delay means that the disease must not be rapidly progressing, so that the patient is able to wait until the CAR T product is ready; otherwise the patient will need bridging chemotherapy. Two to seven days before CAR T cell infusion, a patient receives lymphodepleting chemotherapy to make way for the CAR T cells that are subsequently given as an intravenous infusion. Once infused into patients, the CAR T cells encounter the antigen, proliferate, and kill the tumor cells (fig 4). These cells, therefore, combine the target specificity of a monoclonal antibody with the enhanced cytotoxicity of T cells without requiring human leucocyte antigen presentation of the target antigen.83

An ideal antigen is one that is widely and exclusively expressed on cancer cells but not on normal cells to enhance efficacy and reduce toxicity.84 85 In multiple myeloma, most emerging immunotherapies (including CAR T cells) target B cell maturation antigen (BCMA), a type III trans­membrane receptor, which is a promising target antigen.86 87 BCMA is also known as tumor necrosis

factor receptor superfamily member 17 or CD269. It is expressed in nearly all plasma cells (normal and malignant) although its expression is variable.88 BCMA promotes plasma cell survival and is induced during plasma cell differentiation89 by binding to ligands (a proliferation inducing ligand (APRIL) and B cell activating factor (BAFF)) that are produced by osteoclasts.90 Increased levels of soluble BCMA are associated with high tumor burden in multiple myeloma and thus worse outcomes.91

CAR T cells targeting CD19 were approved by the FDA in 2017 for refractory large B cell lymphoma92 93 and acute lymphoblastic leukemia,94 and are being used in clinical practice. The first study on CAR T cell treatment directed by BCMA opened in 2014 at the US National Cancer Institute.87 95 96 Since then, about a dozen of different early phase clinical trials have been conducted on BCMA CAR T treatment for advanced multiple myeloma.97 98 A detailed review outlining the differences in the construct, manufacturing, and clinical efficacy of these different products has been published previously.81 86

This review focuses on four BCMA CAR T cell products that are currently being evaluated in registration (that is, for regulatory approval) phase I/II clinical trials for patients with relapsed and refractory multiple myeloma. These products include bb2121 (now known as idecabtagene vicleucel or ide­cel), JCARH125 (now known as orvacabtagene autoleucel or orva­cel), LCAR­B38M (now known as JNJ­4528), and P­Bcma­101. FDA approvals for some of these agents are anticipated in 2020­21 for

Monoclonal antibodies

Daratumumab (CD38)

Elotuzumab (SLAMF7)

Isatuximab (CD38)

TAK-079 (CD38)

Antibody drug conjugates

Belantamab mafodotin (BCMA)

Bispecific T cell engager

AMG 420 (BCMA)

CAR natural killer cells

Bispecific antibodies

CC-93269 (BCMA)

Teclistamab (BCMA)

REGN5458 (BCMA)

PF-06863135 (BCMA)

Plasma cell

T cell

MHC

PD-1PD-L1

CAR T cells

Ide-cel/Bb2121 (BCMA)

Orva-cel/JCARH125 (BCMA)

JNJ-4528 (BCMA)

P-BCMA-101 (BCMA)

TCR

Checkpoint inhibitors

Nivolumab (PD-1)

Pembrolizumab (PD-1)

Fig 2 | Recent immunotherapeutic approaches to treat multiple myeloma. CAR=chimeric antigen receptor; TCR=T cell receptor; MHC=major histocompatibility complex; BCMA=B cell maturation antigen; PD-L1=programmed death-ligand 1; PD-1=programmed cell death protein 1

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 6: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

6 doi: 10.1136/bmj.m3176 | BMJ 2020;370:m3176 | the bmj

relapsed and refractory multiple myeloma (fig 5; table 2). The high overall response rates of 60­100% seen in these trials in a highly refractory population is unprecedented, although the durability of these responses is still in question.

Ide-celThe most advanced CAR T cell treatment targeting a BCMA is ide­cel (bb2121), which uses a lentiviral vector for CAR insertion and includes a 4­1BB costimulatory domain as well as a murine single chain variable fragment.114 In a phase I non­randomized, open label, multicenter trial in relapsed and refractory multiple myeloma (≥3 prior lines of treatment) for 33 patients treated at various doses,115 researchers found an overall response rate of 85% with a median progression free survival of 11.8 months. A higher overall response rate was seen at the higher dose levels and doses of 150­450×106 CAR T cells were defined as the active dose.115 This dose is being tested currently in a multicenter, single arm, open label trial to evaluate bb2121 CAR T cells further in relapsed and refractory multiple myeloma; the trial has completed enrolment of 149 patients worldwide. Preliminary results show an overall response rate of 73% (complete response rate 33%) and median progression free survival of 8.8 months in 128 patients treated at doses of 150­450×106 cells (table 2).99 Fifty four patients treated at the highest dose level of 450×106 cells had an overall response rate of 82% and a median progression free survival of 12.1 months.99 These results have been submitted to regulatory agencies including the FDA and European Medicines Agency for treatment for advanced multi­ple myeloma.

Orva-celOrva­cel (JCARH125) is another second generation CAR product with a fully human B cell derived single chain variable fragment, a 4­1BB costimulatory domain, and optimized manufacturing (predefined CD4:CD8 ratio) that is derived from preclinical work at Memorial Sloan Kettering Cancer Center. The preliminary data for the multicenter phase I/II EVOLVE study were presented at the American Society of Clinical Oncology meeting in 2020. These patients had received a median of six prior treatments. They received escalating doses of 50­600×106 cells. The results for 62 patients treated at the 300­600×106 cells dose range showed an overall response rate of 92% (complete response rate 36%).100 101 The trial is currently enrolling at the recommended phase II dose of 600×106 cells (table 2).

JNJ-4528The LCAR­B38M CAR construct was developed initially in China and is currently being pursued in the US and globally as JNJ­4528 (table 2). It consists of two llama derived variable­heavy chain only fragments that target two epitopes of BCMA designed to confer avidity. In a phase I/II study in China, researchers found deep durable responses with a median progression free survival of 19.9 months and a manageable safety profile in relapsed and refractory multiple myeloma, although the patients in this study were treated earlier in their disease course with a median of three prior lines of treatment and were therefore less heavily pre­treated.102­105 In the US and Europe, a multicenter phase Ib/II clinical trial of this CAR construct as JNJ­4528 in relapsed and refractory multiple myeloma (≥3

Binding domain

Transmembrane domain

Costimulatory domain

Activation domain

Receptor

CAR T cell

Plasma cell

Fig 3 | Chimeric antigen receptor (CAR) T cell structure

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 7: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

the bmj | BMJ 2020;370:m3176 | doi: 10.1136/bmj.m3176 7

prior lines of treatment) was conducted to confirm the findings of the LEGEND­2 study. Preliminary results of the phase Ib portion showed an overall response rate of 100% (complete response rate 86%) in patients with a median of five prior lines of treatment (table 2).106 107 The phase II portion is fully enrolled, and phase II and III studies have been initiated.

P-BCMA-101P­BCMA­101 is uniquely manufactured using the non­viral piggyBac gene editing system, which is less costly, produces cells with a high percentage of favorable stem cell memory phenotype T cells, and has the ability to include a safety switch. The binding molecule for this product is not a single chain variable fragment but a small fully human fibronectin domain (Centyrin) that has higher specificity and potentially less immunogenicity. In a phase I dose escalation trial, the overall response rate was 63% with a median progression free survival of 9.5 months in 19 evaluable patients108 (table 2).

ToxicityCAR T cell treatments have a unique toxicity profile where patients can develop side effects such as cytokine release syndrome and neurotoxicity that has been recently termed immune effector cell associated neurotoxicity syndrome (ICANS).116 Cytokine re­lease syndrome has been defined as a disorder characterized by fever, tachypnea, headache, tachy­cardia, hypotension, rash, or hypoxia caused by the release of cytokines from cells. The American Society for Transplantation and Cellular Therapy has developed a consensus grading system for cytokine release syndrome, which depends on the severity and presence of fever, hypotension, or hypoxia (table 3).116

ICANS has been defined as a disorder involving the central nervous system following any immuno­therapy that results in the activation or engagement of endogenous or infused T cells or other immune effector cells. Symptoms or signs can be progressive and could include aphasia, altered level of consciousness, impairment of cognitive skills, motor weakness, seizures, and cerebral edema.116

T cell collection from patient’s peripheral blood

CF

Duration: Indefinitely until needed by patient (years)

CAR T cell production CAR T cell storage

T cell activation, viral transduction,CAR T cell expansion

Leukapheresis

CAR T cell infusion Lymphodepleting chemotherapyMonitoring after CAR T cell infusion

Undertaken in good manufacturing processes facility

Duration: 2-4 weeks

Intravenous access: central line or two large bore peripherals

Duration: 2-4 hour session

No chemotherapy for specified period prior

Response assessment: 1 month after infusion

Duration: hours, days, weeks (CRS,ICANS); weeks, months (cytopenia) Duration: <60 minutes

Infusion of cells 2-7 days after chemotherapy

Duration: 3 days

Regimens: 1. Fludarabine (F) and cyclophosphamide (C); 2. Cyclophosphamide; 3. Other

Completion 2-7 days before cell infusion

CRS

ICANS

Cytopenia

Response

1 2 3

6 5 4

Fig 4 | Chimeric antigen receptor (CAR) T cell treatment for multiple myeloma—sequence of events. CRS=cytokine release syndrome; ICANS=immune effector cell associated neurotoxicity syndrome

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 8: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

8 doi: 10.1136/bmj.m3176 | BMJ 2020;370:m3176 | the bmj

It includes four grades that are determined by the ICE score (immune effector cell associated encephalopathy score, which provides objectivity to grading encephalopathy), level of consciousness, seizure, motor findings, and elevated intracranial pressure or cerebral edema (table 4).116 Management of ICANS and cytokine release syndrome is based on grading and involves supportive care, steroids, and interleukin blocking agents.117 118 Interleukin 6 blocking agents (tocilizumab and siltuximab) with or without steroids are the mainstay of management for cytokine release syndrome, whereas steroids are the mainstay for the management of neurotoxicity. Another potential agent for managing these symp­toms includes the interleukin 1 blocking agent anakinra.119

All the clinical trials on BCMA CAR T cell treatments had a high incidence of cytokine release syndrome (>80%) except for P­BCMA­101, which seemed to have a substantially lower incidence (10%). Despite this, severe cytokine release syndrome (that is, ≥grade

3) is seen in less than 10% of patients. Neurotoxicity was reported in less than 20% of patients with severe neurotoxicity (≥grade 3) in less than 7% of patients. Another common side effect is cytopenia, which has also been thought to be secondary to the lymphodepleting chemotherapy, ongoing CAR T cell activity, and disruption of hematopoiesis showing severe hypocellularity in the bone marrow, but most patients recover with time.120 121

Management of toxicityEarly recognition of cytokine release syndrome and ICANS and prompt intervention after CAR T cell treatment is vital to prevent serious consequences, although the optimal timing for intervention and benefit of prophylactic treatment is yet unknown.122 The CAR T cell therapy associated toxicity (CARTOX) working group has developed a management approach for these syndromes, based on multidisciplinary grades.123 In cytokine release syndrome, patients with grade 1 are usually

Lentivirus Lentivirus Lentivirus PiggyBac

ScFv (mouse) ScFv (human) VH only ScFv (llama) Centyrin(human)

Transmembranedomain

CD28CD8α CD8α CD8α

Costimulatory domain

4-1BB 4-1BB 4-1BB 4-1BB

Activation domain

CD3ζ CD3ζ CD3ζ CD3ζ

NCT number

026589290278651103361748036010780365112804196491

0343001103436771

03090659035482070413363604181827

0328849303741127

Bb2121(ide-cel)

JCARH 125(orva-cel)

JNJ-4528 P-BCMA-101

Transduction/transfection

Binding domain

Fig 5 | Four major constructs of chimeric antigen receptor (CAR) T cells targeting B cell maturation antigens (BCMA), currently in multicenter clinical trials investigating multiple myeloma. This figure does not include all BCMA constructs in multiple myeloma. ScFv=single chain variable fragment; VH only=variable-heavy chain only fragments

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 9: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

the bmj | BMJ 2020;370:m3176 | doi: 10.1136/bmj.m3176 9

managed with supportive care, those with grade 2 are managed with the anti­interleukin 6 receptor tocilizumab with or without steroids in addition to supportive care, and those with grade 3­4 are managed in the intensive care unit with aggressive supportive care, vasopressors, oxygen, tocilizumab, and steroids. Patients with grade 1 and 2 ICANS are managed supportively but an electroencephalogram is done to rule out electrical seizures and imaging of the brain to rule out edema. Patients with grade 3 and 4 ICANS need steroids and more aggressive supportive care.120

Bispecific antibodies including bispecific T cell engagersBispecific monoclonal antibodies direct a host’s immune system (more specifically cytotoxic T cells) against cancer cells by binding CD3 on T cells with a target protein on cancer cells (fig 6).124 A type of bispecific antibody is the bispecific T cell engager (BiTE), which differs from other bispecific antibodies by containing two different single chain variable fragments connected by a linker. BiTEs often have a short half life, requiring continuous infusion to maintain efficacy.125 The first BiTE to receive FDA approval for treatment in relapsed and refractory acute lymphoblastic leukemia is Blinatumomab, a bispecific antibody that engages T cells to CD19 positive cells.126 Because BiTEs engage and activate the patient’s own immune cells, they have a toxicity profile similar to CAR T cells including cytokine release syndrome and ICANS.116

AMG 420AMG 420 (previously named BI 836909) is a novel BiTE targeting BCMA on myeloma cells and CD3ε on T cells, which has induced multiple myeloma cell lysis in preclinical models.127 In the first­in­human phase I study of AMG 420 in patients with at least two lines of treatment, AMG 420 was given as a continuous infusion with a pump for four week infusions, six week cycles, and a maximum of 10 cycles. The maximum tolerated dose was 400 µg/day; seven (70%) of 10 patients responded to this dose. Serious adverse events were seen in 48% of patients, which were most commonly infections; and two patients had reversible grade 3 polyneuropathies. Cytokine release syndrome developed in 38% of patients, with no toxicity in the central nervous system.109 A phase Ib trial with AMG 420 is currently ongoing and although this drug looks promising, the continuous intravenous infusions present logistical challenges for patients and healthcare systems (table 2). AMG 701 is a modified version of AMG 420 (by addition of an Fc domain) with an extended half life that is suitable for dosing once a week and is being investigated in a phase I study.128

CC-93269Another BCMA bispecific antibody, CC­93269, is being studied in an ongoing phase I clinical trial. This humanized 2+1, immunoglobulin G 1 based, T cell engager binds to BCMA bivalently on myeloma cells and CD3ε monovalently on T cells. The bivalent binding could lead to improved potency, tumor

Table 2 | Summary of major multicenter clinical trials investigating multiple myeloma treatments*

Treatment agent, trial (No of participants) Treatment type

Median No of prior lines of treatment Dose range

Overall response rate (%)

Median progression free survival (months)

Cytokine release syndrome (%), any grade (grades 3-5)

Neurotoxicity (%), any grade (grades 3-5)

Bb2121 (ide-cel),99 KarMMa (n=128)

CAR T cell 6 150-450×106 cells 73 8.8 84 (6) 18 (3)

JCARH125 (orva-cel),100 101 EVOLVE (n=62)

CAR T cell 6 300-600×106 cells 92 Not reached 89 (3) 13 (3)

LCAR-B38M,102-105 LEGEND-2 (n=57)

CAR T cell 3 0.07-2.1×106 cells/kg 88 19.9 90 (7) 2

JNJ-4528,106 107 CARTITUDE-1 (n=29)

CAR T cell 5 0.52-0.89×106 cells/kg 100 Not reached 93 (7) 10 (3)

P-BCMA-101,108 PRIME (n=19)

CAR T cell 6 50-1143×106 cells 63 (100†) 9.5 10 (0) 5 (5)

AMG 420109 (n=42) Bispecific antibody (BiTE)

3.5 0.2-800 μg/day, week infusions every 6 weeks, 10 cycles

31 (70‡) Not reached 38 (2) 7 (7)

CC-93269110 (n=30) Bispecific antibody

5 0.15-10 mg weekly for 3 cycles, biweekly for 3 cycles then monthly

43 (89§) Not reached 77 (3) 0

Teclistamab111 (n=78) Bispecific antibody (DuoBody)

6 0.3-720 µg/kg weekly 31 (67¶) Not reached 56 (0) 8 (3)

Belantamab mafodotin,112 DREAMM-2 (n=196)

Antibody drug conjugate

7 2.5 mg/kg every 3 weeks 31 2.9 Not applicable Not applicable6 3.4 mg/kg every 3 weeks 34 4.9 Not applicable Not applicable

Belantamab mafodotin and bortezomib,113 DREAMM-6 (n=18)

Antibody drug conjugate

3 2.5 mg/kg every 3 weeks 78 Not reached Not applicable Not applicable

BiTE=bispecific T cell engager.*This list is not comprehensive.†P-BCMA-101 overall response rate at highest reported dose of 857×106 cells for 3 patients.‡AMG 420 overall response rate at maximum tolerated dose of 400 µg/day for 10 patients.§CC-93269 overall response rate at highest reported dose of 10 mg for 9 patients.¶Teclistamab overall response rate at highest reported dose of 270 µg/kg for 12 patients.

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 10: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

10 doi: 10.1136/bmj.m3176 | BMJ 2020;370:m3176 | the bmj

targeting, and retention.129 All doses (range 0.15­10 mg) were given intravenously over two hours weekly for the first three cycles, every two weeks for the next three cycles, and then monthly. The most common treatment emergent adverse events of grade 3 or higher included neutropenia, anemia, and infections. Cytokine release syndrome was seen in 77% of patients, with all events developing after the first dose and less common with subsequent doses. The incidence increased with higher doses, and only one patient had cytokine release syndrome of grade 3 or higher leading to their death. In 30 patients treated, the overall response rate was 43.3% and dose dependent. The overall response rate was 88.9% in nine patients in the highest dose cohort.110

TeclistamabTeclistamab (JNJ­64007957) is a humanized, immunoglobulin G­4 based, bispecific DuoBody antibody that binds to BCMA and CD3 that is being studied in a phase I clinical trial. In the dose escalation part, 78 patients received doses ranging from 0.3 µg/kg to 720 µg/kg. The drug is given intravenously every week, with one to three step­up doses given within one week before the full dose. The overall response rate was dose dependent with no responses at doses 0.3­19.2 µg/kg, 30% at 38.4­180 µg/kg, and 67% at 270 µg/kg. Cytokine release syndrome was seen in 56% of patients overall and 65% patients at doses over 38.4 µg/kg. The most common adverse events at grade 3 or higher that were related to treatment were cytopenias and infections (table 2).111

Antibody drug conjugatesAntibody drug conjugates are complex molecules composed of an antibody that targets cancer cells and are linked to a biologically active cytotoxic drug (known as the payload; fig 7).125 Belantamab mafodotin (GSK2857916) is a novel humanized and afucosylated (to improve antibody dependent cell mediated cytotoxicity) antibody drug conjugate that targets BCMA. It consists of an anti­BCMA monoclonal antibody conjugated to monomethyl auristatin F, a potent microtubule inhibitor.130 This antibody drug conjugate was shown to have selec­ tive myeloma cell killing in vitro and in vivo thus setting the stage for clinical trials.130 131

This antibody was studied in a two part phase I study. The drug was well tolerated with no dose limiting toxicities, although corneal events (such as blurry vision, dry eyes, photophobia) were seen in about 58% of patients; these events are a known toxicity of monomethyl auristatin F.132 In the dose expansion phase, 35 patients were treated, and the overall response rate was 60% with a median progression free survival 12 months.133 In a phase II, two arm study, the antibody was used in patients with relapsed and refractory multiple myeloma who had failed at least three lines of treatment. The overall response rate was 31% at the 2.5 mg/kg dose and 34% at the 3.3 mg/kg dose, which was significantly lower than the phase I study. The corneal changes or keratopathy were seen in 70% and 75% of patients, respectively. Owing to the similar response rates with the 2.5 mg/kg and 3.3 mg/kg doses and a more favorable side effect profile with the lower dose, 2.5 mg/kg will be the dose used for future studies.112 Based on these data, belantamab is the first anti­BCMA treatment to be FDA approved for relapsed and refractory multiple myeloma patients who have received four prior treatments including an anti­CD38 monoclonal antibody, a proteasome inhibitor, and an immunomodulatory agent.

Preliminary results for another study with 18 patients treated on the belantamab, bortezomib, and dexamethasone arm was presented recently, with an overall response rate of 78%; however, all 18 patients developed grade 1­3 keratopathy.113 This visual toxicity is a unique but potentially serious side effect to this drug that needs close monitoring with an ophthalmologist. Another antibody drug conjugate, DFRF4539A, is an anti­FcRH5 (also known as FcRL5) antibody conjugated to monomethyl auristatin and has shown limited activity and high incidence of toxicity in a phase I study; therefore, it was unsuccessful for this disease (table 2).134 135

Future directionsAlthough most immunotherapies are relatively new, this field is rapidly changing and many other immunotherapies are emerging, as are new indications and improved versions of current immunotherapies (some of which we have discussed here). Although having many drugs potentially available for patients with multiple myeloma is promising, one critical unanswered question is the

Table 3 | American Society for Transplantation and Cellular Therapy consensus grading for cytokine release syndrome (CRS)116

CRS parameter Grade 1 Grade 2 Grade 3 Grade 4Fever* Temperature ≥38°C Temperature ≥38°C Temperature ≥38°C Temperature ≥38°CWith hypotension† None Not requiring

vasopressorsRequiring a vasopressor with or without vasopressin

Requiring multiple vasopressors (excluding vasopressin)

And/or hypoxia† None Requiring low flow nasal cannula‡ or blow-by

Requiring high flow nasal cannula‡, facemask, non-rebreather mask, or Venturi mask

Requiring positive pressure (eg, CPAP, BiPAP, intubation, and mechanical ventilation)

CPAP=continuous positive airway pressure; BiPAP=bilevel positive airway pressure.*Fever defined as temperature ≥38°C not attributable to any other cause. In patients who have CRS then receive anti-pyretic or anti-cytokine treatment such as tocilizumab or steroids, fever is no longer required to grade subsequent CRS severity. In this case, CRS grading is driven by hypotension or hypoxia.†CRS grade is determined by the more severe event: hypotension or hypoxia not attributable to any other cause. For example, a patient with temperature of 39.5°C, hypotension requiring one vasopressor, and hypoxia requiring low flow nasal cannula would be classified as grade 3 CRS.‡Low flow nasal cannula is defined as oxygen delivered at up to 6 L/min; low flow also includes blow-by oxygen delivery, sometimes used in pediatrics. High flow nasal cannula is defined as oxygen delivered at more than 6 L/min.

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 11: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

the bmj | BMJ 2020;370:m3176 | doi: 10.1136/bmj.m3176 11

pricing for these newer drugs.136 Historically, cost of multiple myeloma treatments has been a major barrier leading to disparities in cancer care.137 These treatments should be priced fairly so that they can be universally available to patients with the disease.

Monoclonal antibodiesMore recent studies are now including the mono­clonal antibodies daratumumab, elotuzumab, and isatuximab in the upfront setting as part of a quadruplet regimen that includes a monoclonal

antibody with standard triplet combinations to impro ve depth of response and thus outcomes.52 138­144

Two other newer monoclonal antibodies targeting CD38 include TAK­079, which only needs to be given in small volumes subcutaneously and is being studied as a single agent145; and MOR202,146 which is being studied in combination with dexa­methasone as well as immunomodulatory drugs. Furthermore, a phase I clinical trial looking at the first monoclonal antibody targeting BCMA (SEA­BCMA) is recruiting.147

Table 4 | American Society for Transplantation and Cellular Therapy consensus grading for immune effector cell associated neurotoxicity syndrome (ICANS) in adults116

Neurotoxicity domain Grade 1 Grade 2 Grade 3 Grade 4ICE score* 7-9 3-6 0-2 0 (patient is unarousable and

unable to perform ICE)Depressed level of consciousness†

Awakens spontaneously Awakens to voice Awakens only to tactile stimulus Patient is unarousable or needs vigorous or repetitive tactile stimuli to arouse; stupor or coma

Seizure Not applicable Not applicable Any clinical seizure (focal or generalized) that resolves rapidly or non-convulsive seizures on EEG that resolve with intervention

Life threatening prolonged seizure (>5 min); or repetitive clinical or electrical seizures without return to baseline in between

Motor findings‡ Not applicable Not applicable Not applicable Deep focal motor weakness such as hemiparesis or paraparesis

Elevated ICP/cerebral edema

Not applicable Not applicable Focal or local edema on neuroimaging§ Diffuse cerebral edema on neuroimaging, decerebrate or decorticate posturing, cranial nerve VI palsy, papilledema; or Cushing’s triad

EEG=electroencephalogram. ICANS grade is determined by the most severe event: ICE score, level of consciousness, seizure, motor findings, or raised intercranial pressure or cerebral edema not attributable to any other cause. For example, a patient with an ICE score of 3 who has a generalized seizure is classified as grade 3 ICANS. *Immune effector cell associated encephalopathy (ICE) score. Score is based on orientation (orientation to year, month, city, hospital=4 points); naming (ability to name three objects (eg, point to clock, pen, button)=3 points); following commands (ability to follow simple commands (eg, “show me two fingers” or “close your eyes and stick out your tongue”)=1 point); writing (ability to write a standard sentence (eg, “Our national bird is the bald eagle”)=1 point); attention (ability to count backwards from 100 by 10)=1 point). A patient with an ICE score of 0 might be classified as grade 3 ICANS if they are awake with global aphasia, but a patient with an ICE score of 0 might be classified as grade 4 ICANS if unarousable.†Depressed level of consciousness should be attributable to no other cause (eg, no sedating medication).‡Tremors and myoclonus associated with immune effector cell treatments might be graded according to Common Terminology Criteria for Adverse Events version 5.0, but they do not influence ICANS grading.§Intracranial hemorrhage with or without associated edema is not considered a neurotoxicity feature and is excluded from ICANS grading. It might be graded according to Common Terminology Criteria for Adverse Events version 5.0.

CD3

BiTEs have no Fc domain

Fab regionVariable domain

Constant domain

T cell

CD3 binding site

Tumor specific antigen binding site

Receptor

Plasma cell

Fc domain

Fig 6 | Structure of a bispecific antibody. BiTEs=bispecific T cell engagers

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 12: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

12 doi: 10.1136/bmj.m3176 | BMJ 2020;370:m3176 | the bmj

Chimeric antigen receptor T cellsAlthough a high proportion of patients with relap­sed and refractory multiple myeloma have deep responses to BCMA CAR T cell treatment, relapses are common and the median progression free survival in early studies ranges from 8.8 months to 19.9 months. A key area of future research focuses on building on these early results to improve durability of responses. Some of the approaches being evaluated include the identification of additional targets for cellular treatment, improved manufacturing processes, use of CAR T cell treatment in earlier stages of multiple myeloma, combinations with other drugs, and off­the­shelf approaches including treatment with allogeneic CAR T cells and CAR natural killer cells.

Novel targetsCAR T cells directed towards targets other than BCMA in multiple myeloma are being developed. These targets include CD38,148 149 GPRC5D,150 and SLAMF7,151 and clinical trials are ongoing. Another method to potentially decrease the risk of relapse and maximize treatment effect is the use of a combination of two CAR T products, or the development of CAR T cells that target two antigens (BCMA combined with CD19, GPRC5D, CS1, or TACI), and early results from phase I trials demonstrate feasibility and safety of this approach.81 152­156

Improved manufacturingNewer transduction systems (including non­viral methods such as the piggyBac system) and alter­natives to single chain variable fragments (such as Centyrin in P­BCMA­101) are being evaluated. With promising preliminary results from the phase I

study, a phase II trial of P­BCMA­101 CAR T cells in patients with at least three prior lines of treatment has been initiated with an estimated enrollment of 100 patients.81 108 157 The ratio of CD4:CD8 T cells is undefined in most CAR T cell products and is determined by the ratio in the sample collected during leukapheresis; however, some CAR T products (eg, orva­cel) have predefined ratios of CD4 and CD8 T cells to evaluate whether this would improve their efficacy.100 158 159 Other products such as bb21217 have been modified to improve persistence, given that patients have limited persistence of CAR T cells at the time of relapse. Bb21217 includes T cells that have been cocultured with a phosphoinositide 3 kinase (PI3K) inhibitor bb007 to generate a product with enhanced memory­like phenotype. A trial with bb21217 is currently ongoing with an estimated enrolment of 74 patients.81 160 161

Earlier lines of treatmentNewer, large multicenter clinical trials with products ide­cel and JNJ­4528 are opening to treat patients with CAR T cell products in combination with standard myeloma regimens in earlier lines of treatment. These studies include the KarMMa­2, KarMMa­3, and KarMMa­4 studies with ide­cel and the CARTITUDE­2 and CARTITUDE­4 studies with JNJ­4528. KarMMa­2 is a multicenter, multicohort phase II study in high risk relapsed myeloma. KarMMa­3 is a randomized phase III study comparing bb2121 with standard triplet regimens such as daratumumab, pomalidomide, and dexamethasone; or daratumumab, bortezomib, and dexamethasone; or ixazomib, lenalidomide, and dexamethasone in patients with two to four prior lines of treatment.

Fig 7 | Structure of an antibody drug conjugate

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 13: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

the bmj | BMJ 2020;370:m3176 | doi: 10.1136/bmj.m3176 13

KarMMa­4 is a phase I study to evaluate bb2121 in high risk patients with newly diagnosed multiple myeloma after four cycles of induction treatment. CARTITUDE­2 is a phase II multicohort study looking at JNJ­4528 in relapsed and refractory multiple myeloma including cohorts with high risk disease as well. CARTITUDE­4 is a randomized phase III trial of 400 patients with relapsed and refractory multiple myeloma who will be treated with JNJ­4528 or standard triplet regimens (pomalidomide, bortezomib, and dexamethasone; or daratumumab, pomalidomide, and dexamethasone) in patients with one to three prior lines of treatment.

Combination with other drugs CAR T cells are being tested in combination with other drugs such as the immunomodulatory drugs (lenalidomide or iberdomide),162 163 checkpoint inhibitors (pembrolizumab)164 or a gamma secre­tase inhibitor (JSMD194) that increases BCMA expression on tumor cells. As patients could have downregulation of BCMA expression at the time of relapse, JSMD194 is a unique approach to relapses after CAR T cell treatment.165 166

Armored chimeric antigen receptor T cellsSo­called armored CAR T cells have been engineered to secrete proinflammatory cytokines such as interleukin 12 or ligands designed to further improve CAR T cell efficacy. These cells enhance anti­tumor efficacy and have the potential to overcome the hostile tumor microenvironment via different mechanisms.167 This approach has shown promise in CD19 positive cancers.168 Similar approaches will likely be tested in the future in multiple myeloma, particularly for patients with extramedullary disease.

Other CAR approachesAllogeneic (off-the-shelf) chimeric antigen receptor T cellsAllogeneic CAR T cells could resolve some of the challenges with autologous CAR T cell treatment, including the delays (and costs) associated with individualized manufacturing of cells, the T cells from patients with multiple myeloma might be more exhausted, and the limited ability to give multiple doses. However, the challenges include graft versus host disease (that is, allogeneic CAR T cells attacking healthy organs such as skin), and host versus graft response (that is, the recipient immune system rejecting the allogenic CAR T cells).169 Multiple groups have worked to overcome these challenges, and allogeneic CAR T cells targeting BCMA (P­BCMA­ALLO1170 and ALLO­715171) and targeting CS1/SLAMF7 (UCARTCS181 172) are currently in clinical trials.

Chimeric antigen receptor, natural killer cellsAn alternate off­the­shelf approach would be to use natural killer cells for manufacturing CAR. Unlike T cells, natural killer cells are not restricted by human leukocyte antigen and can therefore be

produced from natural killer cell lines or cord blood, leading to inherent advantages such as off­the­shelf products and reduced toxicity and cost. Preliminary results from a first­in­human trial of anti­CD19 CAR natural killer cells for B cell cancers showed promising efficacy, with eight (73%) of the first 13 patients responding and no evidence of cytokine release syndrome, neurotoxicity, or graft versus host disease.173 Preclinical data for CAR natural killer cells in multiple myeloma targeting CS1, BCMA, CD138, and NKG2D ligands are promising and clinical data are awaited.174­176 A table comparing CAR T cells to CAR natural killer cells has been published elsewhere.81

Bispecific antibodiesGiven the logistical difficulty with continuous infusions of AMG 420, ongoing phase I trials are evaluating the efficacy of intermittent dosing of AMG 420 and evaluating the half life extended BCMA BiTE molecule (AMG 701). AMG 701 is a modified, newer generation version of AMG 420; it contains an Fc domain leading to a longer half life. Other bispecific antibodies targeting BCMA in phase I/II trials include REGN5458,177 JNJ­64007957, TNB­383B,178 and PF­06863135.179

Although BCMA has been the main target of interest, researchers have looked at bispecific antibodies targeting other molecules such as CD38. CD38 is universally expressed and has also been targeted with other effective drugs such as daratumumab for multiple myeloma.40 AMG 424 is an anti­CD38/CD3 bispecific antibody with the ability to kill cancer cells with high and low levels of CD38 expression in vitro and in animal models without triggering excessive cytokine release.180 181 With these findings, AMG 424 is currently being studied in a first­in­human phase I, two part study in patients with relapsed and refractory multiple myeloma. Clinical trials of a bispecific antibody targeting other molecules such as GPRC5D (JNJ­64407564) and FcRH5 (BFCR4350A) are also under way.

Antibody drug conjugatesFurther clinical trials exploring belantamab mafodotin as a single agent or in combinations are under way such as the DREAMM­3 study, which is a randomized phase III trial comparing single agent belantamab mafodotin with pomalidomide and low dexamethasone. Other trials combining this drug with pembrolizumab (DREAMM­4), other cancer drugs (DREAMM­5), and with lenalidomide dexamethasone or bortezomib dexamethasone (DREAMM­6) in relapsed and refractory multiple myeloma; or with bortezomib, lenalidomide, and dexamethasone in transplant ineligible pati­ents with newly diagnosed multiple myeloma (DREAMM­9) are also ongoing. Other antibody drug conjugates targeting BCMA in clinical trials include MEDI2228182  183 and CC­99712. Ongoing trials are also targeting proteins other than BCMA and CD38 such as CD48, CD46, CD56, and CD74.125

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 14: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

14 doi: 10.1136/bmj.m3176 | BMJ 2020;370:m3176 | the bmj

Other approaches—vaccinationCancer vaccines have been shown to generate potent immune responses that can translate into clinically meaningful improvements in outcomes for patients.184 In multiple myeloma, many such approaches have been tested including an idiotype DNA vaccine,185 peptide derived from Bcl­2 family vaccine,186 gene transduced tumor cell vaccine (GVAX),187 188 idiotype loaded antigen presenting cells,189 and whole cell dendritic cell/myeloma fusion vaccine.184 190 191 Most recently, multipeptide vaccine PVX­410 with or without lenalidomide was tested in 22 patients with smoldering multiple myeloma, and results suggested that the vaccine was safe and immunogenic.192 BMT CTN 1401 is a randomized phase II multicenter trial that has enrolled 203 patients, results for which are awaited. The trial randomizes patients to vaccination with (1) dendritic cell/myeloma fusion vaccine with granulocyte macrophage colony stimulating factor (GM­CSF) adjuvant plus lenalidomide maintenance treatment versus (2) maintenance treatment alone or with GM­CSF after autologous transplant as part of an upfront treatment of multiple myeloma.

GuidelinesEarly clinical trials of CAR T cell treatment uncovered greater toxicities related to generalized immune system activation (cytokine release syndrome and ICANS) than those seen with other cellular treatments. Investigators followed the National Cancer Institute’s Common Terminology Criteria for Adverse Events to assess severity and struggled with the terminology because it did not directly apply to CAR T cell treatments. As a result, many institutions treating patients with these cell treatments develo­ped their own grading systems, making comparisons difficult between trials and centers.123 193­195 The American Society for Transplantation and Cellular Therapy therefore developed consensus grading systems incorporating data from these prior indivi­dualized grading criteria. These guidelines for grading cytokine release syndrome (table 3) and ICANS (table 4) have now been adopted widely in the management of toxicities associated with CAR T cell treatment.116

ConclusionSubstantial progress has been made in the care of patients with multiple myeloma over the past two decades resulting in a longer overall survival. Despite this, an unmet need still exists for newer treatments as patients eventually relapse. The landscape for treatment options in multiple myeloma is rapidly changing with many drugs in the pipeline, most of them being immunotherapies. The monoclonal antibodies have shown remarkable efficacy with minimal toxicity and have thus been widely adopted to treat multiple myeloma in relapsed patients initially. Trials looking at monoclonal antibodies in newly diagnosed multiple myeloma are promising and additional large randomized trials are ongoing.

Although this field of immunotherapy is promising, not all treatments are successful in multiple myeloma and some might also be harmful, as seen by the lack of response to single agent checkpoint inhibitors and the increased rate of serious adverse events and deaths in clinical trials combining checkpoint inhibitors with immunomodulatory drugs.

More recently, several large multicenter trials of BCMA targeted CAR T cell treatment have shown unprecedented overall response rates between 60­100% in patients with highly relapsed and refractory multiple myeloma; however, their progression free survival is relatively short (8­20 months). Therefore, much research is still needed into improving duration of response as well as developing products that can be off the shelf (such as CAR natural killer cells and allogeneic CAR T cells). Other BCMA treatments with promise include the bispecific antibodies AMG 420, teclistamab, and CC­93269 and the antibody drug conjugate belantamab mafodotin, among many others. Ultimately, which BCMA directed treatment will become prominent in multiple myeloma will depend on several factors, such as reduced toxicity, higher efficacy, lower cost, and ease of use, and whether different BCMA treatments given sequentially will continue to be effective.

Contributors: Both authors were involved in the conceptualization, writing, and editing of the manuscript. UAS is the guarantor.Funding: Funding support for this publication was provided by the Memorial Sloan Kettering Core Grant (P30 CA008748) and Parker Institute for Cancer Immunotherapy.Competing interests: We have read and understood BMJ policy on declaration of interests and declare the following competing interests: UAS has received research funding from Celgene/Bristol Myers Squibb (BMS) and Parker Institute for Cancer Immunotherapy to her institution and honorariums for continuing medical education activity from the Physicians Education Resource. SM has received research funding from the National Cancer Institute R21CA235154, Janssen, Juno/Celgene/BMS, Takeda, Allogene to his institution and honorariums for continuing medical education activity from the Physicians Education Resource.Provenance and peer review: Commissioned; externally peer reviewed.Patient involvement: No patients were asked for input in the creation of this article.

QuESTIONS FOR FuTuRE RESEARCH•Despite high overall response rates, many of these

treatments still have only a modest progression free survival. What are the mechanisms that lead to relapse with these immunotherapies and how can they be overcome?

•Will using these drugs in earlier lines of treatments improve outcomes further?

•Although these novel drugs are effective, they are cost prohibitive and questions arise as to the affordability for the broader myeloma population worldwide

•The field is dominated by multiple BCMA directed treatments in the pipeline, but how these drugs can be sequenced, and can they retain their efficacy after another prior BCMA directed treatment?

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 15: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

the bmj | BMJ 2020;370:m3176 | doi: 10.1136/bmj.m3176 15

1  Rajkumar SV, Dimopoulos MA, Palumbo A, et al. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol 2014;15:e538-48. doi:10.1016/S1470-2045(14)70442-5 

2  Cohen AD, Raje N, Fowler JA, Mezzi K, Scott EC, Dhodapkar MV. How to train your T cells: overcoming immune dysfunction in multiple myeloma. Clin Cancer Res 2020;26:1541-54. doi:10.1158/1078-0432.CCR-19-2111 

3  Kazandjian D. Multiple myeloma epidemiology and survival: A unique malignancy. Semin Oncol 2016;43:676-81. doi:10.1053/j.seminoncol.2016.11.004 

4  Cowan AJ, Allen C, Barac A, et al. Global burden of multiple myeloma: a systematic analysis for the Global Burden of Disease Study 2016. JAMA Oncol 2018;4:1221-7. doi:10.1001/jamaoncol.2018.2128 

5  Ganguly S, Mailankody S, Ailawadhi S. Many shades of disparities in myeloma care. 2019;519-29.

6  Howlader NA, Noone AM, Krapcho M, et al, (eds). SEER cancer statistics review, 1975-2017, National Cancer Institute. Bethesda, MD. 2020. https://seer.cancer.gov/csr/1975_2017/.

7  Jane Henley S, Ward EM, Scott S, et al. Annual report to the nation on the status of cancer, part I: national cancer statistics. Cancer 2020;126:2225-49. doi:10.1002/cncr.32802.

8  Kumar SK, Dispenzieri A, Lacy MQ, et al. Continued improvement in survival in multiple myeloma: changes in early mortality and outcomes in older patients. Leukemia 2014;28:1122-8. doi:10.1038/leu.2013.313 

9  Kumar SK, Rajkumar SV, Dispenzieri A, et al. Improved survival in multiple myeloma and the impact of novel therapies. Blood 2008;111:2516-20. doi:10.1182/blood-2007-10-116129 

10  Giralt S, Costa LJ, Maloney D, et al. Tandem autologous-autologous versus autologous-allogeneic hematopoietic stem cell transplant for patients with multiple myeloma: long-term follow-up results from the Blood and Marrow Transplant Clinical Trials Network 0102 Trial. Biol Blood Marrow Transplant 2020;26:798-804. doi:10.1016/j.bbmt.2019.11.018 

11  Greil C, Engelhardt M, Ihorst G, et al. Allogeneic transplantation of multiple myeloma patients may allow long-term survival in carefully selected patients with acceptable toxicity and preserved quality of life. Haematologica 2019;104:370-9. doi:10.3324/haematol.2018.200881 

12  Dhakal B, Vesole DH, Hari PN. Allogeneic stem cell transplantation for multiple myeloma: is there a future?Bone Marrow Transplant 2016;51:492-500. doi:10.1038/bmt.2015.325 

13  Shah N, Callander N, Ganguly S, et al, American Society for Blood and Marrow Transplantation. Hematopoietic stem cell transplantation for multiple myeloma: guidelines from the American Society for Blood and Marrow Transplantation. Biol Blood Marrow Transplant 2015;21:1155-66. doi:10.1016/j.bbmt.2015.03.002 

14  Holstein SA, McCarthy PL. Immunomodulatory drugs in multiple myeloma: mechanisms of action and clinical experience. Drugs 2017;77:505-20. doi:10.1007/s40265-017-0689-1 

15  Holstein SA, Suman VJ, McCarthy PL. Update on the role of lenalidomide in patients with multiple myeloma. Ther Adv Hematol 2018;9:175-90. doi:10.1177/2040620718775629 

16  Weber DM, Chen C, Niesvizky R, et al, Multiple Myeloma (009) Study Investigators. Lenalidomide plus dexamethasone for relapsed multiple myeloma in North America. N Engl J Med 2007;357:2133-42. doi:10.1056/NEJMoa070596 

17  Dimopoulos M, Spencer A, Attal M, et al, Multiple Myeloma (010) Study Investigators. Lenalidomide plus dexamethasone for relapsed or refractory multiple myeloma. N Engl J Med 2007;357:2123-32. doi:10.1056/NEJMoa070594 

18  Moreau P, Masszi T, Grzasko N, et al, TOURMALINE-MM1 Study Group. Oral ixazomib, lenalidomide, and dexamethasone for multiple myeloma. N Engl J Med 2016;374:1621-34. doi:10.1056/NEJMoa1516282 

19  Stewart AK, Rajkumar SV, Dimopoulos MA, et al, ASPIRE Investigators. Carfilzomib, lenalidomide, and dexamethasone for relapsed multiple myeloma. N Engl J Med 2015;372:142-52. doi:10.1056/NEJMoa1411321 

20  Dimopoulos MA, Stewart AK, Masszi T, et al. Carfilzomib, lenalidomide, and dexamethasone in patients with relapsed multiple myeloma categorised by age: secondary analysis from the phase 3 ASPIRE study. Br J Haematol 2017;177:404-13. doi:10.1111/bjh.14549 

21  Dimopoulos MA, Stewart AK, Masszi T, et al. Carfilzomib-lenalidomide-dexamethasone vs lenalidomide-dexamethasone in relapsed multiple myeloma by previous treatment. Blood Cancer J 2017;7:e554. doi:10.1038/bcj.2017.31 

22  Dimopoulos M, Wang M, Maisnar V, et al. Response and progression-free survival according to planned treatment duration in patients with relapsed multiple myeloma treated with carfilzomib, lenalidomide, and dexamethasone (KRd) versus lenalidomide and dexamethasone (Rd) in the phase III ASPIRE study. J Hematol Oncol 2018;11:49. doi:10.1186/s13045-018-0583-7 

23  Richardson PG, Oriol A, Beksac M, et al, OPTIMISMM trial investigators. Pomalidomide, bortezomib, and dexamethasone for patients with relapsed or refractory multiple myeloma previously treated with lenalidomide (OPTIMISMM): a randomised, open-label, phase 3 trial. Lancet Oncol 2019;20:781-94. doi:10.1016/S1470-2045(19)30152-4 

24  Benboubker L, Dimopoulos MA, Dispenzieri A, et al, FIRST Trial Team. Lenalidomide and dexamethasone in transplant-ineligible patients with myeloma. N Engl J Med 2014;371:906-17. doi:10.1056/NEJMoa1402551 

25  Attal M, Lauwers-Cances V, Hulin C, et al, IFM 2009 Study. Lenalidomide, bortezomib, and dexamethasone with transplantation for myeloma. N Engl J Med 2017;376:1311-20. doi:10.1056/NEJMoa1611750 

26  Durie BGM, Hoering A, Abidi MH, et al. Bortezomib with lenalidomide and dexamethasone versus lenalidomide and dexamethasone alone in patients with newly diagnosed myeloma without intent for immediate autologous stem-cell transplant (SWOG S0777): a randomised, open-label, phase 3 trial. Lancet 2017;389:519-27. doi:10.1016/S0140-6736(16)31594-X 

27  Palumbo A, Cavallo F, Gay F, et al. Autologous transplantation and maintenance therapy in multiple myeloma. N Engl J Med 2014;371:895-905. doi:10.1056/NEJMoa1402888 

28  Attal M, Lauwers-Cances V, Marit G, et al, IFM Investigators. Lenalidomide maintenance after stem-cell transplantation for multiple myeloma. N Engl J Med 2012;366:1782-91. doi:10.1056/NEJMoa1114138 

29  McCarthy PL, Owzar K, Hofmeister CC, et al. Lenalidomide after stem-cell transplantation for multiple myeloma. N Engl J Med 2012;366:1770-81. doi:10.1056/NEJMoa1114083 

30  Lonial S, Jacobus S, Fonseca R, et al. Randomized trial of lenalidomide versus observation in smoldering multiple myeloma. J Clin Oncol 2020;38:1126-37. doi:10.1200/JCO.19.01740 

31  Mateos MV, Hernández MT, Giraldo P, et al. Lenalidomide plus dexamethasone for high-risk smoldering multiple myeloma. N Engl J Med 2013;369:438-47. doi:10.1056/NEJMoa1300439 

32  Bjorklund CC, Kang J, Amatangelo M, et al. Iberdomide (CC-220) is a potent cereblon E3 ligase modulator with antitumor and immunostimulatory activities in lenalidomide- and pomalidomide-resistant multiple myeloma cells with dysregulated CRBN. Leukemia 2020;34:1197-201. doi:10.1038/s41375-019-0620-8 

33  Lonial S, Van de Donk NWCJ, Popat R, et al. First clinical (phase 1b/2a) study of iberdomide (CC-220; IBER), a CELMoD, in combination with dexamethasone (DEX) in patients (pts) with relapsed/refractory multiple myeloma (RRMM). J Clin Oncol 2019;37(15_suppl):8006.

34  Richardson PG, Vangsted AJ, Ramasamy K, et al. First-in-human phase I study of the novel CELMoD agent CC-92480 combined with dexamethasone (DEX) in patients (pts) with relapsed/refractory multiple myeloma (RRMM). J Clin Oncol 2020:38(15_suppl):8500.

35  Jain A, Ramasamy K. Evolving role of daratumumab: from backbencher to frontline agent. Clin Lymphoma Myeloma Leuk 2020;S2152-2650(20)30143-9.

36  Moreno L, Perez C, Zabaleta A, et al. The mechanism of action of the anti-CD38 monoclonal antibody isatuximab in multiple myeloma. Clin Cancer Res 2019;25:3176-87. doi:10.1158/1078-0432.CCR-18-1597 

37  Ritchie D, Colonna M. Mechanisms of action and clinical development of elotuzumab. Clin Transl Sci 2018;11:261-6. doi:10.1111/cts.12532 

38  Overdijk MB, Jansen JH, Nederend M, et al. The therapeutic CD38 monoclonal antibody daratumumab induces programmed cell death via Fcγ receptor-mediated cross-linking. J Immunol 2016;197:807-13. doi:10.4049/jimmunol.1501351 

39  Jiang H, Acharya C, An G, et al. SAR650984 directly induces multiple myeloma cell death via lysosomal-associated and apoptotic pathways, which is further enhanced by pomalidomide. Leukemia 2016;30:399-408. doi:10.1038/leu.2015.240 

40  Lokhorst HM, Plesner T, Laubach JP, et al. Targeting CD38 with daratumumab monotherapy in multiple myeloma. N Engl J Med 2015;373:1207-19. doi:10.1056/NEJMoa1506348 

41  Lonial S, Weiss BM, Usmani SZ, et al. Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial. Lancet 2016;387:1551-60. doi:10.1016/S0140-6736(15)01120-4 

42  Usmani SZ, Weiss BM, Plesner T, et al. Clinical efficacy of daratumumab monotherapy in patients with heavily pretreated relapsed or refractory multiple myeloma. Blood 2016;128:37-44. doi:10.1182/blood-2016-03-705210 

43  Palumbo A, Chanan-Khan A, Weisel K, et al, CASTOR Investigators. Daratumumab, bortezomib, and dexamethasone for multiple myeloma. N Engl J Med 2016;375:754-66. doi:10.1056/NEJMoa1606038 

44  Spencer A, Lentzsch S, Weisel K, et al. Daratumumab plus bortezomib and dexamethasone versus bortezomib and dexamethasone

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 16: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

16 doi: 10.1136/bmj.m3176 | BMJ 2020;370:m3176 | the bmj

in relapsed or refractory multiple myeloma: updated analysis of CASTOR. Haematologica 2018;103:2079-87. doi:10.3324/haematol.2018.194118 

45  Dimopoulos MA, Oriol A, Nahi H, et al, POLLUX Investigators. Daratumumab, lenalidomide, and dexamethasone for multiple myeloma. N Engl J Med 2016;375:1319-31. doi:10.1056/NEJMoa1607751 

46  Dimopoulos MA, San-Miguel J, Belch A, et al. Daratumumab plus lenalidomide and dexamethasone versus lenalidomide and dexamethasone in relapsed or refractory multiple myeloma: updated analysis of POLLUX. Haematologica 2018;103:2088-96. doi:10.3324/haematol.2018.194282 

47  Bahlis NJ, Dimopoulos MA, White DJ, et al. Daratumumab plus lenalidomide and dexamethasone in relapsed/refractory multiple myeloma: extended follow-up of POLLUX, a randomized, open-label, phase 3 study. Leukemia 2020;34:1875-84. doi:10.1038/s41375-020-0711-6 

48  Dimopoulos M, Quach H, Mateos MV, et al. Carfilzomib, dexamethasone, and daratumumab versus carfilzomib and dexamethasone for patients with relapsed or refractory multiple myeloma (CANDOR): results from a randomised, multicentre, open-label, phase 3 study. Lancet 2020;396:186-97. doi:10.1016/S0140-6736(20)30734-0

49  Mateos MV, Dimopoulos MA, Cavo M, et al, ALCYONE Trial Investigators. Daratumumab plus bortezomib, melphalan, and prednisone for untreated myeloma. N Engl J Med 2018;378:518-28. doi:10.1056/NEJMoa1714678 

50  Mateos MV, Cavo M, Blade J, et al. Overall survival with daratumumab, bortezomib, melphalan, and prednisone in newly diagnosed multiple myeloma (ALCYONE): a randomised, open-label, phase 3 trial. Lancet 2020;395:132-41. doi:10.1016/S0140-6736(19)32956-3 

51  Facon T, Kumar S, Plesner T, et al, MAIA Trial Investigators. Daratumumab plus lenalidomide and dexamethasone for untreated myeloma. N Engl J Med 2019;380:2104-15. doi:10.1056/NEJMoa1817249 

52  Moreau P, Attal M, Hulin C, et al. Bortezomib, thalidomide, and dexamethasone with or without daratumumab before and after autologous stem-cell transplantation for newly diagnosed multiple myeloma (CASSIOPEIA): a randomised, open-label, phase 3 study. Lancet 2019;394:29-38. doi:10.1016/S0140-6736(19)31240-1 

53  Voorhees PM, Kaufman JL, Laubach J, et al. Daratumumab, lenalidomide, bortezomib, and dexamethasone for transplant-eligible newly diagnosed multiple myeloma: the GRIFFIN trial. Blood 2020;136:936-45.

54  Mateos MV, Nahi H, Legiec W, et al. Subcutaneous versus intravenous daratumumab in patients with relapsed or refractory multiple myeloma (COLUMBA): a multicentre, open-label, non-inferiority, randomised, phase 3 trial. Lancet Haematol 2020;7:e370-80. doi:10.1016/S2352-3026(20)30070-3 

55  Lonial S, Dimopoulos M, Palumbo A, et al, ELOQUENT-2 Investigators. Elotuzumab therapy for relapsed or refractory multiple myeloma. N Engl J Med 2015;373:621-31. doi:10.1056/NEJMoa1505654 

56  Dimopoulos MA, Lonial S, Betts KA, et al. Elotuzumab plus lenalidomide and dexamethasone in relapsed/refractory multiple myeloma: Extended 4-year follow-up and analysis of relative progression-free survival from the randomized ELOQUENT-2 trial. Cancer 2018;124:4032-43. doi:10.1002/cncr.31680 

57  Dimopoulos MA, Dytfeld D, Grosicki S, et al. Elotuzumab plus pomalidomide and dexamethasone for multiple myeloma. N Engl J Med 2018;379:1811-22. doi:10.1056/NEJMoa1805762 

58  Usmani SZ, Ailawadhi S, Sexton R, et al. Primary analysis of the randomized phase II trial of bortezomib, lenalidomide, dexamthasone with/without elotuzumab for newly diagnosed, high-risk multiple myeloma (SWOG-1211). J Clin Oncol 2020;38(15_suppl):8507.

59  Attal M, Richardson PG, Rajkumar SV, et al, ICARIA-MM study group. Isatuximab plus pomalidomide and low-dose dexamethasone versus pomalidomide and low-dose dexamethasone in patients with relapsed and refractory multiple myeloma (ICARIA-MM): a randomised, multicentre, open-label, phase 3 study. Lancet 2019;394:2096-107. doi:10.1016/S0140-6736(19)32556-5 

60  Moreau P, Dimopoulos MA, Mikhael J, et al. Isatuximab plus carfilzomib and dexamethasone vs carfilzomib and dexamethasone in relapsed/refractory multiple myeloma (Ikema): interim analysis of a phase 3, randomized, open-label study. European Hematology Association 2020.

61  Chari A, Suvannasankha A, Fay JW, et al. Daratumumab plus pomalidomide and dexamethasone in relapsed and/or refractory multiple myeloma. Blood 2017;130:974-81. doi:10.1182/blood-2017-05-785246 

62  Siegel DS, Schiller GJ, Samaras C, et al. Pomalidomide, dexamethasone, and daratumumab in relapsed refractory multiple myeloma after lenalidomide treatment. Leukemia 2020. doi:10.1038/s41375-020-0813-1 

63  Zonder JA, Mohrbacher AF, Singhal S, et al. A phase 1, multicenter, open-label, dose escalation study of elotuzumab in patients with advanced multiple myeloma. Blood 2012;120:552-9. doi:10.1182/blood-2011-06-360552 

64  Dimopoulos MA, Lonial S, White D, et al. Elotuzumab plus lenalidomide/dexamethasone for relapsed or refractory multiple myeloma: ELOQUENT-2 follow-up and post-hoc analyses on progression-free survival and tumour growth. Br J Haematol 2017;178:896-905. doi:10.1111/bjh.14787 

65  Martin T, Baz R, Benson DM, et al. A phase 1b study of isatuximab plus lenalidomide and dexamethasone for relapsed/refractory multiple myeloma. Blood 2017;129:3294-303. doi:10.1182/blood-2016-09-740787 

66  Mikhael J, Richardson P, Usmani SZ, et al. A phase 1b study of isatuximab plus pomalidomide/dexamethasone in relapsed/refractory multiple myeloma. Blood 2019;134:123-33. doi:10.1182/blood-2019-02-895193 

67  Richardson PG, Attal M, Campana F, et al. Isatuximab plus pomalidomide/dexamethasone versus pomalidomide/dexamethasone in relapsed/refractory multiple myeloma: ICARIA Phase III study design. Future Oncol 2018;14:1035-47. doi:10.2217/fon-2017-0616 

68  Liu J, Hamrouni A, Wolowiec D, et al. Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-gamma and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood 2007;110:296-304. doi:10.1182/blood-2006-10-051482 

69  Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012;12:252-64. doi:10.1038/nrc3239 

70  Lesokhin AM, Ansell SM, Armand P, et al. Nivolumab in patients with relapsed or refractory hematologic malignancy: preliminary results of a phase Ib study. J Clin Oncol 2016;34:2698-704. doi:10.1200/JCO.2015.65.9789 

71  Pianko MJ, Funt SA, Page DB, et al. Efficacy and toxicity of therapy immediately after treatment with nivolumab in relapsed multiple myeloma. Leuk Lymphoma 2018;59:221-4. doi:10.1080/10428194.2017.1320713 

72  Görgün G, Samur MK, Cowens KB, et al. Lenalidomide enhances immune checkpoint blockade-induced immune response in multiple myeloma. Clin Cancer Res 2015;21:4607-18. doi:10.1158/1078-0432.CCR-15-0200 

73  Badros A, Hyjek E, Ma N, et al. Pembrolizumab, pomalidomide, and low-dose dexamethasone for relapsed/refractory multiple myeloma. Blood 2017;130:1189-97. doi:10.1182/blood-2017-03-775122 

74  Mateos MV, Orlowski RZ, Ocio EM, et al. Pembrolizumab combined with lenalidomide and low-dose dexamethasone for relapsed or refractory multiple myeloma: phase I KEYNOTE-023 study. Br J Haematol 2019;186:e117-21. doi:10.1111/bjh.15946 

75  Mateos MV, Blacklock H, Schjesvold F, et al, KEYNOTE-183 Investigators. Pembrolizumab plus pomalidomide and dexamethasone for patients with relapsed or refractory multiple myeloma (KEYNOTE-183): a randomised, open-label, phase 3 trial. Lancet Haematol 2019;6:e459-69. doi:10.1016/S2352-3026(19)30110-3 

76  Lonial SRP, Reece D, Mohamed H, Shelat S, San Miguel J. CheckMate 602: a phase 3, open-label, randomized trial of combinations of nivolumab, elotuzumab, pomalidomide, and dexamethasone in relapsed or relapsed and refractory multiple myeloma. Clin Lymphoma Myeloma Leuk 2017. doi:10.1016/j.clml.2017.07.142

77  Usmani SZ, Schjesvold F, Oriol A, et al, KEYNOTE-185 Investigators. Pembrolizumab plus lenalidomide and dexamethasone for patients with treatment-naive multiple myeloma (KEYNOTE-185): a randomised, open-label, phase 3 trial. Lancet Haematol 2019;6:e448-58. doi:10.1016/S2352-3026(19)30109-7 

78  Gormley NJ, Pazdur R. Immunotherapy combinations in multiple myeloma - known unknowns. N Engl J Med 2018;379:1791-5. doi:10.1056/NEJMp1803602 

79  Gyawali B, Prasad V. Drugs that lack single-agent activity: are they worth pursuing in combination?Nat Rev Clin Oncol 2017;14:193-4. doi:10.1038/nrclinonc.2017.27 

80  Ghosh A, Mailankody S, Giralt SA, Landgren CO, Smith EL, Brentjens RJ. CAR T cell therapy for multiple myeloma: where are we now and where are we headed?Leuk Lymphoma 2018;59:2056-67. doi:10.1080/10428194.2017.1393668 

81  Shah UA, Mailankody S. CAR T and CAR NK cells in multiple myeloma: expanding the targets. Best Pract Res Clin Haematol 2020;33:101141. doi:10.1016/j.beha.2020.101141 

82  Levine BL, Miskin J, Wonnacott K, Keir C. Global manufacturing of CAR T cell therapy. Mol Ther Methods Clin Dev 2016;4:92-101. doi:10.1016/j.omtm.2016.12.006 

83  Ma T, Shi J, Liu H. Chimeric antigen receptor T cell targeting B cell maturation antigen immunotherapy is promising for multiple myeloma. Ann Hematol 2019;98:813-22. doi:10.1007/s00277-018-03592-9 

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 17: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

the bmj | BMJ 2020;370:m3176 | doi: 10.1136/bmj.m3176 17

84  Curran KJ, Pegram HJ, Brentjens RJ. Chimeric antigen receptors for T cell immunotherapy: current understanding and future directions. J Gene Med 2012;14:405-15. doi:10.1002/jgm.2604 

85  Bonifant CL, Jackson HJ, Brentjens RJ, Curran KJ. Toxicity and management in CAR T-cell therapy. Mol Ther Oncolytics 2016;3:16011. doi:10.1038/mto.2016.11 

86  Shah UA, Smith EL. Multiple myeloma, targeting B-cell maturation antigen with chimeric antigen receptor T-cells. Cancer J 2019;25:208-16. doi:10.1097/PPO.0000000000000379 

87  Carpenter RO, Evbuomwan MO, Pittaluga S, et al. B-cell maturation antigen is a promising target for adoptive T-cell therapy of multiple myeloma. Clin Cancer Res 2013;19:2048-60. doi:10.1158/1078-0432.CCR-12-2422 

88  O’Connor BP, Raman VS, Erickson LD, et al. BCMA is essential for the survival of long-lived bone marrow plasma cells. J Exp Med 2004;199:91-8. doi:10.1084/jem.20031330 

89  Avery DT, Kalled SL, Ellyard JI, et al. BAFF selectively enhances the survival of plasmablasts generated from human memory B cells. J Clin Invest 2003;112:286-97. doi:10.1172/JCI18025 

90  Hengeveld PJ, Kersten MJ. B-cell activating factor in the pathophysiology of multiple myeloma: a target for therapy?Blood Cancer J 2015;5:e282. doi:10.1038/bcj.2015.3 

91  Cho SF, Anderson KC, Tai YT. Targeting B cell maturation antigen (BCMA) in multiple myeloma: potential uses of BCMA-based immunotherapy. Front Immunol 2018;9:1821. doi:10.3389/fimmu.2018.01821 

92  Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med 2017;377:2531-44. doi:10.1056/NEJMoa1707447 

93  Schuster SJ, Bishop MR, Tam CS, et al, JULIET Investigators. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med 2019;380:45-56. doi:10.1056/NEJMoa1804980 

94  Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med 2018;378:439-48. doi:10.1056/NEJMoa1709866 

95  Ali SA, Shi V, Maric I, et al. T cells expressing an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma. Blood 2016;128:1688-700. doi:10.1182/blood-2016-04-711903 

96  Brudno JN, Maric I, Hartman SD, et al. T cells genetically modified to express an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of poor-prognosis relapsed multiple myeloma. J Clin Oncol 2018;36:2267-80. doi:10.1200/JCO.2018.77.8084 

97  Cohen AD, Garfall AL, Stadtmauer EA, et al. B cell maturation antigen-specific CAR T cells are clinically active in multiple myeloma. J Clin Invest 2019;129:2210-21. doi:10.1172/JCI126397 

98  Mailankody S, Ghosh A, Staehr M, et al. Clinical responses and pharmacokinetics of MCARH171, a human-derived BCMA targeted CAR T cell therapy in relapsed/refractory multiple myeloma: final results of a phase I clinical trial. Blood 2018;132(Suppl 1):959. doi:10.1182/blood-2018-99-119717

99  Munshi NC, Anderson LDJr, Shah N, et al. on behalf of the KarMMa Study Investigators. Idecabtagene vicleucel (ide-cel; bb2121), a BCMA-targeted CAR T-cell therapy, in patients with relapsed and refractory multiple myeloma (RRMM): Initial KarMMa results. J Clin Oncol 2020;38(15_suppl):8503.

100  Mailankody S, Htut M, Lee KP, et al. JCARH125, anti-BCMA CAR T-cell therapy for relapsed/refractory multiple myeloma: initial proof of concept results from a phase 1/2 multicenter study (EVOLVE). Blood 2018;132(Suppl 1):957. doi:10.1182/blood-2018-99-113548

101  Mailankody S, Jakubowiak AJ, Htut M, et al. Orvacabtagene autoleucel (orva-cel), a B-cell maturation antigen (BCMA)-directed CAR T cell therapy for patients (pts) with relapsed/refractory multiple myeloma (RRMM): update of the phase 1/2 EVOLVE study (NCT03430011). J Clin Oncol 2020;38(15_suppl):8504.

102  Xu J, Chen LJ, Yang SS, et al. Exploratory trial of a biepitopic CAR T-targeting B cell maturation antigen in relapsed/refractory multiple myeloma. Proc Natl Acad Sci U S A 2019;116:9543-51. doi:10.1073/pnas.1819745116 

103  Zhao WH, Liu J, Wang BY, et al. A phase 1, open-label study of LCAR-B38M, a chimeric antigen receptor T cell therapy directed against B cell maturation antigen, in patients with relapsed or refractory multiple myeloma. J Hematol Oncol 2018;11:141. doi:10.1186/s13045-018-0681-6 

104  Zhao W-H, Liu J, Wang B-Y, et al. Updated analysis of a phase 1, open-label study of LCAR-B38M, a chimeric antigen receptor T cell therapy directed against B-cell maturation antigen, in patients with relapsed/refractory multiple myeloma. Blood 2018;132(Suppl 1):955. doi:10.1182/blood-2018-99-110548

105  Wang B-Y, Zhao W-H, Liu J, et al. Long-term follow-up of a phase 1, first-in-human open-label study of LCAR-B38M, a structurally differentiated chimeric antigen receptor T (CAR-T) cell therapy targeting B-Cell maturation antigen (BCMA), in

patients (pts) with relapsed/refractory multiple myeloma (RRMM). Blood 2019;134(Suppl_1):579. doi:10.1182/blood-2019-124953

106  Madduri D, Usmani SZ, Jagannath S, et al. Results from CARTITUDE-1: a phase 1b/2 study of JNJ-4528, a CAR-T cell therapy directed against B-cell maturation antigen (BCMA), in patients with relapsed and/or refractory multiple myeloma (R/R MM). Blood 2019;134(Suppl_1):577. doi:10.1182/blood-2019-121731

107  Berdeja JG, Madduri D, Usmani SZ, et al. Update of CARTITUDE-1: A phase Ib/II study of JNJ-4528, a B-cell maturation antigen (BCMA)-directed CAR-T-cell therapy, in relapsed/refractory multiple myeloma. J Clin Oncol 2020;38(15_suppl):8505.

108  Gregory T, Cohen AD, Costello CL, et al. Efficacy and safety of P-Bcma-101 CAR-T cells in patients with relapsed/refractory (r/r) multiple myeloma (MM). Blood 2018;132(Suppl 1):1012. doi:10.1182/blood-2018-99-111419

109  Topp MS, Duell J, Zugmaier G, et al. Anti-B-cell maturation antigen BiTE Molecule AMG 420 induces responses in multiple myeloma. J Clin Oncol 2020;38:775-83. doi:10.1200/JCO.19.02657 

110  Costa LJ, Wong SW, Bermúdez A, et al, First clinical study of the B-cell maturation antigen (BCMA) 2+1 T cell engager (TCE) CC-93269 in patients (Pts) with relapsed/refractory multiple myeloma (RRMM): interim results of a phase 1 multicenter trial. Blood 2019;134(Suppl_1):143. doi:10.1182/blood-2019-122895

111  Usmani SZ, Mateos MV, Nahi H, et al. Phase I study of teclistamab, a humanized B-cell maturation antigen (BCMA) x CD3 bispecific antibody, in relapsed/refractory multiple myeloma (R/R MM). J Clin Oncol 2020;38(15_suppl_):100.

112  Lonial S, Lee HC, Badros A, et al. Belantamab mafodotin for relapsed or refractory multiple myeloma (DREAMM-2): a two-arm, randomised, open-label, phase 2 study. Lancet Oncol 2020;21:207-21. doi:10.1016/S1470-2045(19)30788-0 

113  Nooka AK, Stockerl-Goldstein K, Quach H, et al. DREAMM-6: Safety and tolerability of belantamab mafodotin in combination with bortezomib/dexamethasone in relapsed/refractory multiple myeloma (RRMM). J Clin Oncol 2020;38(15_suppl):8502.

114  Friedman KM, Garrett TE, Evans JW, et al. Effective targeting of multiple B-cell maturation antigen-expressing hematological malignances by anti-B-cell maturation antigen chimeric antigen receptor T cells. Hum Gene Ther 2018;29:585-601. doi:10.1089/hum.2018.001 

115  Raje N, Berdeja J, Lin Y, et al. Anti-BCMA CAR T-cell therapy bb2121 in relapsed or refractory multiple myeloma. N Engl J Med 2019;380:1726-37. doi:10.1056/NEJMoa1817226 

116  Lee DW, Santomasso BD, Locke FL, et al. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biol Blood Marrow Transplant 2019;25:625-38. doi:10.1016/j.bbmt.2018.12.758 

117  Riegler LL, Jones GP, Lee DW. Current approaches in the grading and management of cytokine release syndrome after chimeric antigen receptor T-cell therapy. Ther Clin Risk Manag 2019;15:323-35. doi:10.2147/TCRM.S150524 

118  Brudno JN, Kochenderfer JN. Recent advances in CAR T-cell toxicity: mechanisms, manifestations and management. Blood Rev 2019;34:45-55. doi:10.1016/j.blre.2018.11.002 

119  Norelli M, Camisa B, Barbiera G, et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med 2018;24:739-48. doi:10.1038/s41591-018-0036-4 

120  Neelapu SS. Managing the toxicities of CAR T-cell therapy. Hematol Oncol 2019;37(Suppl 1):48-52. doi:10.1002/hon.2595 

121  Jain T, Knezevic A, Pennisi M, et al. Hematopoietic recovery in patients receiving chimeric antigen receptor T-cell therapy for hematologic malignancies. Blood Adv 2020;4:3776-87. doi:10.1182/bloodadvances.2020002509

122  Frey N, Porter D. Cytokine release syndrome with chimeric antigen receptor T cell therapy. Biol Blood Marrow Transplant 2019;25:e123-7. doi:10.1016/j.bbmt.2018.12.756 

123  Neelapu SS, Tummala S, Kebriaei P, et al. Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat Rev Clin Oncol 2018;15:47-62. doi:10.1038/nrclinonc.2017.148 

124  Velasquez MP, Bonifant CL, Gottschalk S. Redirecting T cells to hematological malignancies with bispecific antibodies. Blood 2018;131:30-8. doi:10.1182/blood-2017-06-741058 

125  Cohen AD. Myeloma: next generation immunotherapy. Hematology Am Soc Hematol Educ Program 2019;2019:266-72. doi:10.1182/hematology.2019000068 

126  Kantarjian H, Stein A, Gökbuget N, et al. Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N Engl J Med 2017;376:836-47. doi:10.1056/NEJMoa1609783 

127  Hipp S, Tai YT, Blanset D, et al. A novel BCMA/CD3 bispecific T-cell engager for the treatment of multiple myeloma induces selective lysis in vitro and in vivo [correction in: Leukemia 2017;31:2278]. Leukemia 2017;31:1743-51. doi:10.1038/leu.2016.388 

128  Cho S-F, Lin L, Xing L, et al. AMG 701 potently induces anti-multiple myeloma (MM) functions of T Cells and IMiDs

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 18: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

State of the art reVIeW

18 doi: 10.1136/bmj.m3176 | BMJ 2020;370:m3176 | the bmj

further enhance its efficacy to prevent MM relapse in vivo. Blood 2019;134(Suppl_1):135. doi:10.1182/blood-2019-128528

129  Seckinger A, Delgado JA, Moser S, et al. Target expression, generation, preclinical activity, and pharmacokinetics of the BCMA-T cell bispecific antibody EM801 for multiple myeloma treatment. Cancer Cell 2017;31:396-410. doi:10.1016/j.ccell.2017.02.002 

130  Tai YT, Mayes PA, Acharya C, et al. Novel anti-B-cell maturation antigen antibody-drug conjugate (GSK2857916) selectively induces killing of multiple myeloma. Blood 2014;123:3128-38. doi:10.1182/blood-2013-10-535088 

131  Lee L, Bounds D, Paterson J, et al. Evaluation of B cell maturation antigen as a target for antibody drug conjugate mediated cytotoxicity in multiple myeloma. Br J Haematol 2016;174:911-22. doi:10.1111/bjh.14145 

132  Trudel S, Lendvai N, Popat R, et al. Targeting B-cell maturation antigen with GSK2857916 antibody-drug conjugate in relapsed or refractory multiple myeloma (BMA117159): a dose escalation and expansion phase 1 trial. Lancet Oncol 2018;19:1641-53. doi:10.1016/S1470-2045(18)30576-X 

133  Trudel S, Lendvai N, Popat R, et al. Antibody-drug conjugate, GSK2857916, in relapsed/refractory multiple myeloma: an update on safety and efficacy from dose expansion phase I study. Blood Cancer J 2019;9:37. doi:10.1038/s41408-019-0196-6 

134  Stewart AK, Krishnan AY, Singhal S, et al. Phase I study of the anti-FcRH5 antibody-drug conjugate DFRF4539A in relapsed or refractory multiple myeloma. Blood Cancer J 2019;9:17. doi:10.1038/s41408-019-0178-8 

135  Elkins K, Zheng B, Go M, et al. FcRL5 as a target of antibody-drug conjugates for the treatment of multiple myeloma. Mol Cancer Ther 2012;11:2222-32. doi:10.1158/1535-7163.MCT-12-0087 

136  Prasad V, Mailankody S. Research and development spending to bring a single cancer drug to market and revenues after approval. JAMA Intern Med 2017;177:1569-75. doi:10.1001/jamainternmed.2017.3601 

137  Rajkumar SV. Value and cost of myeloma therapy. 2018;(38):662-6. doi:10.1200/EDBK_200867

138  Salwender H, Bertsch U, Weisel K, et al. Rationale and design of the German-speaking myeloma multicenter group (GMMG) trial HD6: a randomized phase III trial on the effect of elotuzumab in VRD induction/consolidation and lenalidomide maintenance in patients with newly diagnosed myeloma. BMC Cancer 2019;19:504. doi:10.1186/s12885-019-5600-x 

139  Voorhees PM, Kaufman JL, Laubach JP, et al. Depth of response to daratumumab (DARA), lenalidomide, bortezomib, and dexamethasone (RVd) improves over time in patients (pts) with transplant-eligible newly diagnosed multiple myeloma (NDMM): Griffin study update. Blood 2019;134(Suppl_1):691. doi:10.1182/blood-2019-123465

140  Orlowski RZ, Goldschmidt H, Cavo M, et al. Phase III (IMROZ) study design: Isatuximab plus bortezomib (V), lenalidomide (R), and dexamethasone (d) vs VRd in transplant-ineligible patients (pts) with newly diagnosed multiple myeloma (NDMM). J Clin Oncol 2018;36(15_suppl):TPS8055. doi:10.1200/JCO.2018.36.15_suppl.TPS8055

141  Landgren O, Hultcrantz M, Lesokhin AM, et al. Weekly carfilzomib, lenalidomide, dexamethasone and daratumumab (wKRd-D) combination therapy provides unprecedented MRD negativity rates in newly diagnosed multiple myeloma: a clinical and correlative phase 2 study. Blood 2019;134(Suppl_1):862. doi:10.1182/blood-2019-126378

142  Chari A, Usmani SZ, Krishnan A, et al. Daratumumab (DARA) in combination with carfilzomib, lenalidomide, and dexamethasone (KRd) in patients with newly diagnosed multiple myeloma (MMY1001): updated results from an open-label, phase 1b study. Blood 2017;130(Suppl 1):3110.

143  Sonneveld P, Broijl A, Gay F, et al. Bortezomib, lenalidomide, and dexamethasone (VRd) ± daratumumab (DARA) in patients (pts) with transplant-eligible (TE) newly diagnosed multiple myeloma (NDMM): a multicenter, randomized, phase III study (PERSEUS). J Clin Oncol 2019;37(15_suppl):TPS8055. doi:10.1200/JCO.2019.37.15_suppl.TPS8055

144  Zweegman S, Usmani SZ, Chastain K, et al. Bortezomib, lenalidomide, and dexamethasone (VRd) ± daratumumab (DARA) in patients (pts) with newly diagnosed multiple myeloma (NDMM) for whom transplant is not planned as initial therapy: a multicenter, randomized, phase III study (CEPHEUS). J Clin Oncol 2019;37(15_suppl):TPS8056. doi:10.1200/JCO.2019.37.15_suppl.TPS8056

145  Krishnan AY, Patel KK, Hari P, et al. Preliminary results from a phase 1b study of TAK-079, an investigational anti-CD38 monoclonal antibody (mAb) in patients with relapsed/ refractory multiple myeloma (RRMM). Blood 2019;134(Suppl_1):140. doi:10.1182/blood-2019-128007

146  Raab MS, Chatterjee M, Goldschmidt H, et al. MOR202 with low-dose dexamethasone (Dex) or pomalidomide/Dex or lenalidomide/Dex in relapsed or refractory multiple myeloma (RRMM): primary

analysis of a phase I/IIa, multicenter, dose-escalation study. Blood 2018;132(Suppl 1):153. doi:10.1182/blood-2018-153

147  Benson DMJr, Holmes H, Hari P, et al. A phase 1 study of two investigational agents, ACTR087, an autologous T cell product expressing an antibody-coupled T cell receptor, in combination with SEA-BCMA, a novel non-fucosylated monoclonal antibody, in subjects with relapsed or refractory multiple myeloma. Blood 2018;132(Suppl 1):1997. doi:10.1182/blood-2018-99-111729

148  Drent E, Groen RW, Noort WA, et al. Pre-clinical evaluation of CD38 chimeric antigen receptor engineered T cells for the treatment of multiple myeloma. Haematologica 2016;101:616-25. doi:10.3324/haematol.2015.137620 

149  Drent E, Themeli M, Poels R, et al. A rational strategy for reducing on-target off-tumor effects of CD38-chimeric antigen receptors by affinity optimization. Mol Ther 2017;25:1946-58. doi:10.1016/j.ymthe.2017.04.024 

150  Smith EL, Harrington K, Staehr M, et al. GPRC5D is a target for the immunotherapy of multiple myeloma with rationally designed CAR T cells. 2019;11:eaau7746. doi:10.1126/scitranslmed.aau7746 

151  Gogishvili T, Danhof S, Prommersberger S, et al. SLAMF7-CAR T cells eliminate myeloma and confer selective fratricide of SLAMF7+ normal lymphocytes. Blood 2017;130:2838-47. doi:10.1182/blood-2017-04-778423 

152  Lee L, Draper B, Chaplin N, et al. An APRIL-based chimeric antigen receptor for dual targeting of BCMA and TACI in multiple myeloma. Blood 2018;131:746-58. doi:10.1182/blood-2017-05-781351 

153  Fernandez de Larrea C, Staehr M, Lopez A, et al. Optimal dual-targeted CAR construct simultaneously targeting Bcma and GPRC5D prevents bcma-escape driven relapse in multiple myeloma.Blood 2019;134(Suppl_1):136. doi:10.1182/blood-2019-126145

154  Popat R, Zweegman S, Cavet J, et al. Phase 1 first-in-human study of AUTO2, the first chimeric antigen receptor (CAR) T cell targeting APRIL for patients with relapsed/refractory multiple myeloma (RRMM). Blood 2019;134(Suppl_1):3112. doi:10.1182/blood-2019-126689

155  Chen KH, Wada M, Pinz KG, et al. A compound chimeric antigen receptor strategy for targeting multiple myeloma. Leukemia 2018;32:402-12. doi:10.1038/leu.2017.302 

156  Yan Z, Cao J, Cheng H, et al. A combination of humanised anti-CD19 and anti-BCMA CAR T cells in patients with relapsed or refractory multiple myeloma: a single-arm, phase 2 trial. Lancet Haematol 2019;6:e521-9. doi:10.1016/S2352-3026(19)30115-2 

157  Costello CL, Gregory TK, Ali SA, et al. Phase 2 study of the response and safety of P-Bcma-101 CAR-T cells in patients with relapsed/refractory (r/r) multiple myeloma (MM) (PRIME). Blood 2019;134(Suppl_1):3184. doi:10.1182/blood-2019-129562

158  Lee DH, Cervantes-Contreras F, Lee SY, Green DJ, Till BG. Improved expansion and function of CAR T cell products from cultures initiated at defined CD4:CD8 ratios. Blood 2018;132(Suppl 1):3334. doi:10.1182/blood-2018-99-111576

159  Green DJ, Pont M, Sather BD, et al. Fully human Bcma targeted chimeric antigen receptor T Cells administered in a defined composition demonstrate potency at low doses in advanced stage high risk multiple myeloma. Blood 2018;132(Suppl 1):1011. doi:10.1182/blood-2018-99-117729

160  Klebanoff CA, Crompton JG, Leonardi AJ, et al. Inhibition of AKT signaling uncouples T cell differentiation from expansion for receptor-engineered adoptive immunotherapy. JCI Insight 2017;2:95103. doi:10.1172/jci.insight.95103 

161  Shah NAM, Siegel DS, Jagannath S, et al. Initial results from a phase 1 clinical study of bb21217, a next-generation anti Bcma CAR T therapy. Blood 2018. doi:10.1182/blood-2018-99-116953

162  Wang X, Walter M, Urak R, et al. Lenalidomide enhances the function of CS1 chimeric antigen receptor-redirected T Cells against multiple myeloma. Clin Cancer Res 2018;24:106-19. doi:10.1158/1078-0432.CCR-17-0344 

163  Lonial S, Amatangelo M, Popat R, et al. Translational and clinical evidence of a differentiated profile for the novel CELMoD, iberdomide (CC-220). Blood 2019;134(Suppl_1):3119. doi:10.1182/blood-2019-124298

164  Bernabei L, Garfall AL, Melenhorst JJ, et al. PD-1 inhibitor combinations as salvage therapy for relapsed/refractory multiple myeloma (MM) patients progressing after bcma-directed CAR T cells. Blood 2018;132(Suppl 1):1973. doi:10.1182/blood-2018-99-119514

165  Pont MJ, Hill T, Cole GO, et al. γ-Secretase inhibition increases efficacy of BCMA-specific chimeric antigen receptor T cells in multiple myeloma. Blood 2019;134:1585-97. doi:10.1182/blood.2019000050 

166  Cowan AJ, Pont M, Sather BD, et al. Efficacy and safety of fully human Bcma CAR T cells in combination with a gamma secretase inhibitor to increase Bcma surface expression in patients with relapsed or refractory multiple myeloma. Blood 2019;134(Suppl_1):204. doi:10.1182/blood-2019-129405

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from

Page 19: Emerging immunotherapies in multiple myeloma...2020/09/21  · multiple myeloma,24 26 as maintenance treatment after induction and autologous transplantation,27 29 and in smoldering

No commercial reuse: See rights and reprints http://www.bmj.com/permissions Subscribe: http://www.bmj.com/subscribe

State of the art reVIeW

167  Yeku OO, Brentjens RJ. Armored CAR T-cells: utilizing cytokines and pro-inflammatory ligands to enhance CAR T-cell anti-tumour efficacy. Biochem Soc Trans 2016;44:412-8. doi:10.1042/BST20150291 

168  Park JH, Palomba ML, Batlevi CL, et al. A phase I first-in-human clinical trial of CD19-targeted 19-28z/4-1BBL “armored” CAR T cells in patients with relapsed or refractory NHL and CLL including Richter’s transformation. Blood 2018;132(Suppl 1):224. doi:10.1182/blood-2018-99-117737

169  Depil S, Duchateau P, Grupp SA, Mufti G, Poirot L. ‘Off-the-shelf’ allogeneic CAR T cells: development and challenges. Nat Rev Drug Discov 2020;19:185-99. doi:10.1038/s41573-019-0051-2 

170  Cranert SA, Richter M, Tong M, et al. Manufacture of an allogeneic CAR-T stem cell memory product candidate for multiple myeloma, P-Bcma-ALLO1, is robust, reproducible and highly scalable. Blood 2019;134(suppl_1):4445. doi:10.1182/blood-2019-131839

171  Sommer C, Boldajipour B, Valton J, et al. ALLO-715, an allogeneic BCMA CAR T therapy possessing an off-switch for the treatment of multiple myeloma. Blood 2018;132(Suppl 1):591. doi:10.1182/blood-2018-99-119227

172  Mathur R, Zhang Z, He J, et al. Universal SLAMF7-specific CAR T-cells as treatment for multiple myeloma. Blood 2017;130(Suppl 1):502.

173  Liu E, Marin D, Banerjee P, et al. Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors. N Engl J Med 2020;382:545-53. doi:10.1056/NEJMoa1910607 

174  Jiang H, Zhang W, Shang P, et al. Transfection of chimeric anti-CD138 gene enhances natural killer cell activation and killing of multiple myeloma cells. Mol Oncol 2014;8:297-310. doi:10.1016/j.molonc.2013.12.001 

175  Chu J, Deng Y, Benson DM, et al. CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma. Leukemia 2014;28:917-27. doi:10.1038/leu.2013.279 

176  Maroto-Martín E, Encinas J, García-Ortiz A, et al. NKG2D and BCMA-CAR NK cells efficiently eliminate multiple myeloma cells. A comprehensive comparison between two clinically relevant CARs. European Hematology Association Library, 2019. doi:10.1097/01.HS9.0000563120.66927.63

177  Cooper D, Madduri D, Lentzsch S, et al. Safety and preliminary clinical activity of REGN5458, an anti-Bcma x anti-CD3 bispecific antibody, in patients with relapsed/refractory multiple myeloma. Blood 2019;134(Suppl_1):3176. doi:10.1182/blood-2019-126818

178  Buelow B, D’Souza A, Rodriguez C, et al. TNB383B.0001: a multicenter, phase 1, open-label, dose-escalation andexpansion study of TNB-383B, a bispecific antibody targeting BCMA in subjects with relapsed or refractory multiple myeloma. Blood 2019;134(Suppl_1):1874. doi:10.1182/blood-2019-123220

179  Raje NS, Jakubowiak A, Gasparetto C, et al. Safety, clinical activity, pharmacokinetics, and pharmacodynamics from a phase I study of PF-06863135, a B-cell maturation antigen (BCMA)-CD3 bispecific antibody, in patients with relapsed/refractory multiple myeloma (RRMM). Blood 2019;134(Suppl_1):1869. doi:10.1182/blood-2019-121805

180  Zuch de Zafra CL, Fajardo F, Zhong W, et al. Targeting multiple myeloma with AMG 424, a novel anti-CD38/CD3 bispecific T-cell-recruiting antibody optimized for cytotoxicity and cytokine release. Clin Cancer Res 2019;25:3921-33. doi:10.1158/1078-0432.CCR-18-2752 

181  de Zafra C, Balazs M, Fajardo F, et al. Preclinical characterization of AMG 424, a novel humanized T cell-recruiting bispecific anti-CD3/CD38 antibody. Blood 2017;130(Suppl 1):500.

182  Xing L, Li Y, Lin L, et al. MEDI2228, a novel Bcma antibody-PBD conjugate, sensitizes human multiple myeloma cells to NK cell-mediated cytotoxicity and upregulates CD38 expression in MM cells. Blood 2019;134(Suppl_1):3096. doi:10.1182/blood-2019-127135

183  Xing L, Lin L, Yu T, et al. Anti-Bcma PBD MEDI2228 combats drug resistance and synergizes with bortezomib and inhibitors to DNA damage response in multiple myeloma. Blood 2019;134(Suppl_1):1817. doi:10.1182/blood-2019-127163

184  Avigan D, Rosenblatt J. Vaccine therapy in hematologic malignancies. Blood 2018;131:2640-50. doi:10.1182/blood-2017-11-785873 

185  McCann KJ, Godeseth R, Chudley L, et al. Idiotypic DNA vaccination for the treatment of multiple myeloma: safety and immunogenicity in a phase I clinical study. Cancer Immunol Immunother 2015;64:1021-32. doi:10.1007/s00262-015-1703-7 

186  Jørgensen NG, Ahmad SM, Abildgaard N, et al. Peptide vaccination against multiple myeloma using peptides derived from anti-apoptotic proteins: a phase I trial. Stem Cell Investig 2016;3:95. doi:10.21037/sci.2016.11.09 

187  Borrello IM, Noonan K, Huff CA, et al. Allogeneic myeloma GVAX with lenalidomide enhances progression free survival through the generation of tumor specific immunity in patients in near complete remission. Blood 2015;126:4238. doi:10.1182/blood.V126.23.4238.4238

188  Borrello I, Noonan K, Ferguson AK, et al. Allogeneic myeloma GVAX with lenalidomide in near complete remission to enhance progression free survival. 2015;33(15_suppl):3082.

189  Lacy MQ, Mandrekar S, Dispenzieri A, et al. Idiotype-pulsed antigen-presenting cells following autologous transplantation for multiple myeloma may be associated with prolonged survival. Am J Hematol 2009;84:799-802. doi:10.1002/ajh.21560 

190  Rosenblatt J, Vasir B, Uhl L, et al. Vaccination with dendritic cell/tumor fusion cells results in cellular and humoral antitumor immune responses in patients with multiple myeloma. Blood 2011;117:393-402. doi:10.1182/blood-2010-04-277137 

191  Rosenblatt J, Avivi I, Vasir B, et al. Vaccination with dendritic cell/tumor fusions following autologous stem cell transplant induces immunologic and clinical responses in multiple myeloma patients. Clin Cancer Res 2013;19:3640-8. doi:10.1158/1078-0432.CCR-13-0282 

192  Nooka AK, Wang ML, Yee AJ, et al. Assessment of safety and immunogenicity of PVX-410 vaccine with or without lenalidomide in patients with smoldering multiple myeloma: a nonrandomized clinical trial. JAMA Oncol 2018;4:e183267. doi:10.1001/jamaoncol.2018.3267 

193  Lee DW, Gardner R, Porter DL, et al. Current concepts in the diagnosis and management of cytokine release syndrome [correction in: Blood 2015;126:1048]. Blood 2014;124:188-95. doi:10.1182/blood-2014-05-552729 

194  Park JH, Rivière I, Gonen M, et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med 2018;378:449-59. doi:10.1056/NEJMoa1709919 

195  Porter D, Frey N, Wood PA, Weng Y, Grupp SA. Grading of cytokine release syndrome associated with the CAR T cell therapy tisagenlecleucel [correction in: J Hematol Oncol 2018;11:81]. J Hematol Oncol 2018;11:35. doi:10.1186/s13045-018-0571-y 

on 8 February 2021 by guest. P

rotected by copyright.http://w

ww

.bmj.com

/B

MJ: first published as 10.1136/bm

j.m3176 on 21 S

eptember 2020. D

ownloaded from