In this issue of Blood, Ali et al provide the first clinical data that describe safety and efficacy of adoptive therapy with patient-derived chimeric antigen receptor (CAR) T cells targeting B-cell maturation antigen (BCMA) in multiple myeloma.1 

The clinical development of cancer immunotherapy with CAR T cells is in a global race to obtain clinical proof of concept in entities other than B-cell leukemia and lymphoma. Multiple myeloma remains a largely incurable hematologic malignancy. Thus, the development of novel, targeted immunotherapies has been pursued aggressively. About 3 years ago, Carpenter et al reported their work on a novel CAR targeting BCMA in multiple myeloma and demonstrated efficacy against myeloma cell lines and primary myeloma cells in preclinical models.2  The group has now accomplished clinical translation and reports the first clinical trial with CAR T cells targeting a unique myeloma antigen.1  The study reported 4 dose levels from 0.3 × 106 to 9 × 106 CAR T cells/kg body weight, and clinical responses were seen at the 2 higher dose levels with 2 very good partial responses and 1 complete response lasting 17 weeks. Although this initial trial run was successful, highlighting the therapeutic potential of myeloma-reactive CAR T cells, it clearly leaves opportunity for improvement. The finish line of long-term disease control and cure is still distant in this CAR T-cell race against multiple myeloma.

BCMA has been implicated in myeloma pathogenesis. Recently, attention has shifted to BCMA as a target for immunotherapy because it is uniformly expressed on myeloma cells. BCMA has no known expression on normal solid tissue and only very restricted expression on normal hematopoietic cells, that is, on normal plasma cells, some memory B cells, and plasmacytoid dendritic cells.2-4  This favorable expression pattern has allowed the development of a remarkable armamentarium of BCMA-specific monoclonal antibodies (mAbs), bispecific antibodies, antibody-drug conjugates, and CAR T cells. Several additional clinical trials of CAR therapy are ongoing and employ alternative BCMA-CARs that incorporate humanized binding domains to reduce immunogenicity, alternative CAR spacer designs and costimulatory domains to enhance reactivity, and alternative gene transfer vectors. All of these variables have been shown to affect the potency and potentially safety of CAR T-cell products.5  The present clinical trial provides a first proof of concept and benchmark for subsequent trials.

The toxicity observed with BCMA-CARs in this trial included cytokine release syndrome with fever and hypotension as well as hematologic toxicity. The latter was quite significant in some patients, but within the range that can be explained by the lymphodepleting conditioning regimen and inflammation mediated by BCMA-CAR T cells in the bone marrow. B-cell depletion is not anticipated when targeting BCMA, as the majority of normal B cells are BCMA negative. The BCMA-CAR T-cell doses required for efficacy in this trial were somewhat higher than reported with CD19-CARs and responses were not as durable.6,7  The authors suggest that low-antigen density of BCMA on myeloma cells is a potential factor, and in some patients, antigen density was certainly lower compared with CD19 on lymphoma, but still far above the threshold presumed to be required for complete CAR activation. Another, potentially underestimated, variable is soluble BCMA protein that is present in serum of all myeloma patients and is used in this and previous studies as a biomarker for disease burden.2,8  The influence of soluble BCMA protein on CAR binding and CAR T-cell function has been studied in vitro with no negative impact.2  Still, it is logical to assume that soluble BCMA protein that contains the epitope recognized by the CAR and is present in molar excess in serum and bone marrow milieu has an impact. The persistence of BCMA-CAR T cells after administration to patients in this trial was relatively short (<3 months), reminiscent of what has been observed with similarly designed CD19-CARs and retroviral gene transfer by the same group.6  The persistence and depth of engraftment may be augmented with optimizing CAR design, attention to the proportions of CD8+ killer and CD4+ helper T cells in the infused product, and optimizing the lymphodepleting conditioning regimen.5,7  The immunogenicity of the BCMA-CAR that contains a murine-binding domain has unfortunately not been investigated.

The “gold medal” outcome for hematologists caring for myeloma patients is to achieve long-term disease control and eventually cure. Many novel substances are currently being employed in treatment algorithms, including new proteasome inhibitors, histone deacetylase inhibitors, immune modulatory drugs, and mAbs. The conceptual advantage of CAR T cells is that they provide a new and currently uncovered spectrum of effector mechanisms against myeloma that can be complemented in combination therapies. There is a fully loaded pipeline of novel CARs targeting alternative myeloma antigens, including CD38 and CD44v6,9,10  to name just a few, as well as SLAMF7-CARs that are being developed at our own institution. An important side note is that the clinical efficacy of BCMA-CAR therapy correlated with and was predicted by preclinical studies in murine xenograft models that will also aid in shaping the next wave of CAR T-cell therapy in myeloma.

CAR T cells have made their debut in multiple myeloma! It is an encouraging start. Keep on rolling, myeloma CARs!

Conflict-of-interest disclosure: The authors declare no competing financial interests.

1
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
, vol. 
128
 
13
(pg. 
1688
-
1700
)
2
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
, vol. 
19
 
8
(pg. 
2048
-
2060
)
3
Darce
 
JR
Arendt
 
BK
Wu
 
X
Jelinek
 
DF
Regulated expression of BAFF-binding receptors during human B cell differentiation.
J Immunol
2007
, vol. 
179
 
11
(pg. 
7276
-
7286
)
4
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
, vol. 
123
 
20
(pg. 
3128
-
3138
)
5
Hudecek
 
M
Lupo-Stanghellini
 
MT
Kosasih
 
PL
, et al. 
Receptor affinity and extracellular domain modifications affect tumor recognition by ROR1-specific chimeric antigen receptor T cells.
Clin Cancer Res
2013
, vol. 
19
 
12
(pg. 
3153
-
3164
)
6
Kochenderfer
 
JN
Dudley
 
ME
Kassim
 
SH
, et al. 
Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor.
J Clin Oncol
2015
, vol. 
33
 
6
(pg. 
540
-
549
)
7
Turtle
 
CJ
Hanafi
 
LA
Berger
 
C
, et al. 
CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients.
J Clin Invest
2016
, vol. 
126
 
6
(pg. 
2123
-
2138
)
8
Sanchez
 
E
Li
 
M
Kitto
 
A
, et al. 
Serum B-cell maturation antigen is elevated in multiple myeloma and correlates with disease status and survival.
Br J Haematol
2012
, vol. 
158
 
6
(pg. 
727
-
738
)
9
Casucci
 
M
Nicolis di Robilant
 
B
Falcone
 
L
, et al. 
CD44v6-targeted T cells mediate potent antitumor effects against acute myeloid leukemia and multiple myeloma.
Blood
2013
, vol. 
122
 
20
(pg. 
3461
-
3472
)
10
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
, vol. 
101
 
5
(pg. 
616
-
625
)
Sign in via your Institution