• First report on homozygous BCMA gene deletion and subsequent antigen loss in a patient treated with CD3xBCMA bispecific antibody.

  • Heterozygous deletions of BCMA, GPRC5D, and other immune targets in up to 15% of immunotherapy-naïve patients.

T cell–engaging immunotherapies exert unprecedented single-agent activity in multiple myeloma (MM), thereby putting a yet unexplored selective pressure on the clonal architecture. In this study, we report on homozygous BCMA (TNFRSF17) gene deletion after BCMA-targeting T cell–redirecting bispecific antibody therapy in a heavily pretreated MM patient. Loss of BCMA protein expression persisted over subsequent relapses, with no response to treatment with anti-BCMA antibody drug conjugate. In light of the multiple alternative targets that are emerging in addition to BCMA, we extended our analyses to delineate a more complete picture of genetic alterations that may have an impact on immunotherapy targets in MM. We performed whole-genome sequencing and RNA sequencing in 100 MM patients (50 were newly diagnosed; 50 were relapsed/refractory) and identified a significant proportion of patients with aberrations in genes encoding immunotherapy targets; GPRC5D ranked first with 15% heterozygous deletions, followed by CD38 (10%), SDC1 (5%), and TNFRSF17 (4%). Notably, these heterozygous deletions did not lower the expression levels of respective genes, but they may represent a first hit that drives the acquisition of homozygous deletions and subsequent antigen-loss relapse upon targeted immunotherapy. In summary, we show preexisting vulnerability in genes encoding immunotargets before and homozygous deletions after T cell–engaging immunotherapy.

Novel T cell–engaging therapies such as chimeric antigen receptor (CAR) T cells or bispecific antibodies (BsAb’s) have entered the treatment for multiple myeloma (MM) and show exceptionally high response rates in relapsed/refractory (RR) patients. Beyond BCMA, a plethora of other plasma cell targets is currently under investigation, including SLAMF7,1  GPRC5D,2  CD44v,3  and FCRL54  (also known as FcRH5), with promising preliminary results in clinical phase 1 trials. Yet patients continue to relapse, and in contrast to immunotherapy in aggressive lymphoma, there is no survival plateau after treatment with CAR T cells or BsAb’s in MM.

Recently, we described a first tumor-intrinsic resistance mechanism to TNFRSF17 (known as BCMA)-directed CAR T cell therapy. It helped explain the mystery of homozygous deletions on chromosome 16p encompassing the TNFRSF17 (BCMA) gene as the cause of antigen escape in a patient treated with the BCMA CAR T cell product ide-cel in the KarMMa trial.5  At the American Society of Hematology (ASH) 2020 meeting, other groups confirmed biallelic events (del/del and del/mut) as mechanisms of antigen escape after CAR T cell therapy.6,7  Notably , biallelic inactivation usually requires 2 independent hits, and heterozygous TNFRSF17 deletions have been described in BCMA-naïve patients8,9  at risk for a second hit. Thus, the pressure of an anti-BCMA–directed immunotherapy could lead to irreversible antigen loss and subsequent treatment failure.

Here, we report the first case of a biallelic BCMA gene deletion after BsAb therapy, highlighting that this genomic mechanism of resistance is relevant in both types of T cell treatment: CAR T cell therapy and BsAb therapy. Furthermore, by using whole-genome sequencing (WGS) data, we provide evidence that genomic alterations in genes encoding immunotherapy targets preexist in a relevant proportion of patients before immunotherapy, including heterozygous deletions, which may potentially serve as a predictive biomarker in future trials.

Patients

We report the case of a 56-year-old patient treated with a CD3xBCMA BsAb who was subject to WGS in compliance with the CARE guidelines. Furthermore, we report on WGS data derived from 100 patients with MM (50 newly diagnosed [NDMM]; 50 relapsed/refractory [RRMM]; supplemental Table 2). All patients gave their written informed consent for scientific evaluations. The study was approved by the Internal Review Board at the University of Würzburg (reference KFO216) and adhered to the tenets of the Declaration of Helsinki.

WGS and RNA-seq

WGS and bulk RNA sequencing (RNA-seq) were performed as previously described.10  Briefly, WGS libraries were prepared from 1 µg DNA from CD138 purified cells with the TruSeq polymerase chain reaction free library prep kit, and 2 × 150-bp paired-end sequences were generated on a NovaSeq 6000 instrument with a median coverage of 106×. For bulk RNA-seq, 250 ng of total RNA per sample was used to produce stranded RNA libraries (TruSeq Total Stranded RNA, Illumina). Altogether, 2 × 100-bp paired-end reads were sequenced on the NovaSeq 6000 with a median of 64 million reads per sample (bioinformatic approach is provided in the supplemental Data).

Immunohistochemistry

BCMA protein expression was determined by immunohistochemistry using a polyclonal goat anti-BCMA antibody (dilution 1:10; target retrieval pH 6.1) (AF193, R&D Systems) on paraffin-embedded bone marrow sections according to standard procedures.

We detected a clonal biallelic deletion at 16p13.13 encompassing the BCMA gene in a 56-year-old patient with penta-refractory MM who was receiving daratumumab, pomalidomide, and dexamethasone in his eighth line of therapy. WGS showed a hypodiploid karyotype with monosomy 16 and a 586-kb homozygous deletion involving BCMA (Figure 1; supplemental Figure 1; supplemental Table 1). The patient was treated previously on the AMG420 trial, which evaluated a CD3xBCMA bispecific T cell–engager construct at the sixth line of therapy; he achieved a complete response but relapsed after 6 months of treatment. We retrospectively assessed the BCMA status using immunohistochemistry on bio-banked trephine biopsies that showed strong and consistent BCMA expression before the patient participated in the AMG420 trial but loss of BCMA expression at relapse. Notably, BCMA loss was persistent at a subsequent relapse from carfilzomib, lenalidomide, dexamethasone (KRD) salvage therapy 20 months later. Without knowing about BCMA loss at relapse at that time, the anti-BCMA antibody-drug conjugate belantamab mafodotin was administered at the ninth line of therapy, to which the patient was primary refractory (supplemental Figure 2). This is the first case study that shows homozygous BCMA gene deletion to be a mechanism of resistance to BCMA-directed BsAb’s. In this patient, antigen loss was irreversible, and subsequent BCMA therapy failed, suggesting that targeted immunotherapies, in theory, are an evolutionary bottleneck in the progression of MM. It is still unclear whether the BCMA deletion was a rare preexisting event or whether it developed during anti-BCMA therapy. Highly sensitive single-cell approaches and accounting for spatial heterogeneity will be required to address this question.

Figure 1.

WGS, read coverage track, and immunohistochemistry. (A) Circos plot with copy number variations, structural variations, and single-nucleotide variants based on WGS data at progression after therapy with CD3xBCMA BsAb. Outer track runs clockwise from chromosome 1 to Y. Inner track shows gains >1 Mb in blue and losses >1 Mb in red. Red lines inside the circle represent interchromosomal translocations with variant allele frequency (VAF) >0.1. Genes with mutations (CRBN) are depicted in red. (B) Read coverage visualized with Integrative Genomics Viewer illustrates copy number variations in the short arm of chromosome 16 comprising heterozygous deleted regions caused by monosomy 16 and a homozygous deletion of 586 kb from 12.036.001 to 12.622.000 (hg19 reference genome) including TNFRSF17 (BCMA ). (C) BCMA protein expression was determined by immunohistochemistry using a polyclonal goat anti-BCMA antibody on paraffin-embedded bone marrow sections. Lambda light chain staining (immunohistochemistry) served as positive control (magnification, x200).

Figure 1.

WGS, read coverage track, and immunohistochemistry. (A) Circos plot with copy number variations, structural variations, and single-nucleotide variants based on WGS data at progression after therapy with CD3xBCMA BsAb. Outer track runs clockwise from chromosome 1 to Y. Inner track shows gains >1 Mb in blue and losses >1 Mb in red. Red lines inside the circle represent interchromosomal translocations with variant allele frequency (VAF) >0.1. Genes with mutations (CRBN) are depicted in red. (B) Read coverage visualized with Integrative Genomics Viewer illustrates copy number variations in the short arm of chromosome 16 comprising heterozygous deleted regions caused by monosomy 16 and a homozygous deletion of 586 kb from 12.036.001 to 12.622.000 (hg19 reference genome) including TNFRSF17 (BCMA ). (C) BCMA protein expression was determined by immunohistochemistry using a polyclonal goat anti-BCMA antibody on paraffin-embedded bone marrow sections. Lambda light chain staining (immunohistochemistry) served as positive control (magnification, x200).

Close modal

To explore the relevance of this resistance mechanism in MM, we screened for genomic alterations in BCMA and 20 alternative immunotherapy targets (supplemental Table 2) by using WGS in a group of 100 T cell immunotherapy-naïve patients (supplemental Tables 3 and 4 ). Indeed, we found heterozygous deletions in GPRC5D (15%), CD38 (10%), SDC1 (5%), TNFRSF17 (4%), and NCAM1 (3%). Overall, heterozygous deletions occurred in 30% of patients, and some patients showed deletions in up to 3 different immunotarget genes. Considering the emerging dual-antigen targeting immunotherapies,11,12  it is worth mentioning that 1 patient presented with both heterozygous deletion of BCMA and heterozygous deletion of GPRC5D. Of note, 15 of 21 targets were encoded on chromosomes involved in hyperdiploid karyotypes or on 1q and were consequently amplified in about half the patients. Single-nucleotide variants (SNVs) in genes encoding immune targets were found at low frequency (Figure 2). With the exception of 3 frameshift mutations in GPRC5D, no mutation was nonsense or frameshift, and the functional consequences of gene mutations remain elusive. Nevertheless, we observed 1 biallelic event (del/mut) for GPRC5D.

Figure 2.

Overview on WGS and expression data for immunotherapy targets. (A) Gains, single-nucleotide variants (SNV), and deletions of 21 immunotherapy targets in 50 NDMM and 50 RRMM patients. (B) Deletions of target genes . (C) Expression of immunotherapy target genes in 44 NDMM and 28 RRMM patients. Box-and-whisker plot shows the minimum, the maximum, the sample median, and the interquartile range between the lower and upper quartiles. Depicted are gains of at least 3 patients and losses, SNVs, as well as copy number variation (CNV) (gains) accompanied by SNVs of at least 2 patients. CPM, counts per million.

Figure 2.

Overview on WGS and expression data for immunotherapy targets. (A) Gains, single-nucleotide variants (SNV), and deletions of 21 immunotherapy targets in 50 NDMM and 50 RRMM patients. (B) Deletions of target genes . (C) Expression of immunotherapy target genes in 44 NDMM and 28 RRMM patients. Box-and-whisker plot shows the minimum, the maximum, the sample median, and the interquartile range between the lower and upper quartiles. Depicted are gains of at least 3 patients and losses, SNVs, as well as copy number variation (CNV) (gains) accompanied by SNVs of at least 2 patients. CPM, counts per million.

Close modal

The genomic makeup of MM changes from baseline to relapse,9  and 1q gain or amplification is more common at relapse (supplemental Figure 3). Consequently, in our cohort, gain of FCRL5 (19 vs 30 patients; P = .045) and SLAMF7 (19 vs 31 patients; P = .027, Fisher’s exact test) were significantly enriched in RRMM. Furthermore, as expected, we observed a clear trend for more deletions in pretreated patients (Figure 2) because karyotypes become more complex at later stages of the disease.13  This trend was confirmed when comparing our data to the data set from the CoMMpass trial, which included NDMM patients (supplemental Table 5).

In addition to using WGS, we performed transcriptome analysis for 72 of the 100 patients for whom genetic material was available. Strikingly, gains, heterozygous deletions, and SNVs did not result in significant changes in the gene expression of the immunotherapy targets (Figure 2), suggesting that expression of these genes is not solely regulated by gene copy number; with respect to loss of expression, both alleles have to be dysfunctional. In addition, we found tumor suppressor genes with implications in hematologic malignancies within common deleted regions such as CDKN1B14  and RERG15  on chromosome 12 deleted in 100% and 93% of patients with del(12p), respectively, or LITAF16  deleted in all patients with del(16p); thus, one might speculate whether loss of them may provide a growth advantage to the affected clones.

The current treatment landscape of MM is rapidly changing, and we expect therapy to move toward the use of T cell–based immunotherapies upfront. Our study supports the use of immunotherapies earlier in the disease process, because the frequency of deletions and mutations in genes encoding immunotherapy targets was lower in patients with NDMM vs those with RRMM. Our early data suggest that biallelic BCMA gene deletion is a key immune evasion mechanism for BCMA-targeting T cell therapies. It also argues for BCMA expression testing before BCMA therapy, especially in patients pretreated with anti-BCMA. Similarly, this mechanism of resistance to T cell–based therapies will affect alternative antigens, and we detected genomic vulnerability in their encoding genes in a significant number of treatment-naïve patients. Combination of different immunotargets (eg, multispecific CAR T cells) might be a promising approach for overcoming drug resistance caused by loss of a single antigen.11,17 

The authors thank Sabine Roth and Rabea Konietschke for their excellent technical support.

This work was supported by German Cancer Aid via the Mildred Scheel Early Career Center (MSNZ Würzburg) Programme (L.R. and K.M.K.), by the patient advocacy group ‘Forschung hilft’-Stiftung zur Förderung der Krebsforschung an der Universität Würzburg and the Nationales Centrum für Tumorerkrankungen (H.E., K.M.K., and L.R.), and by Sonderforschungsbereich (SFB)/Transregio TR221 (projects A03 and A04) (H.E.).

Contribution: M.S. Truger, L.R., and K.M.K. conceived of and designed the study; J.D., X.Z., L.H., A. Ruckdeschel, S.H., L.H., J.P., M.S. Topp, A. Riedel, L.R., K.M.K., and H.E. provided study material or patients; W.W., M.M., and M.S. Truger analyzed bioinformatic and statistical data; L.R., M.S. Truger, K.M.K., J.D., X.Z., A. Ruckdeschel, M.J., A. Riedel, H.E., C.H., and N.W. interpreted data; M.C.D.V. and N.B. analyzed additional data sets; L.R., M.S. Truger, and K.M.K. wrote the paper; and all authors reviewed and approved the paper.

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

Correspondence: Leo Rasche, Department of Internal Medicine 2, University Hospital Würzburg, Oberdürrbacherstr. 6, 97080 Würzburg, Germany; e-mail: rasche_l@ukw.de.

1.
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
(
26
):
2838
-
2847
.
2.
Smith
EL
,
Harrington
K
,
Staehr
M
, et al
.
GPRC5D is a target for the immunotherapy of multiple myeloma with rationally designed CAR T cells
.
Sci Transl Med.
2019
;
11
(
485
):
eaau7746
.
3.
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
;
122
(
20
):
3461
-
3472
.
4.
Li
J
,
Stagg
NJ
,
Johnston
J
, et al
.
Membrane-proximal epitope facilitates efficient T cell synapse formation by anti-FcRH5/CD3 and is a requirement for myeloma cell killing
.
Cancer Cell
.
2017
;
31
(
3
):
383
-
395
.
5.
Da Vià
MC
,
Dietrich
O
,
Truger
M
, et al
.
Homozygous BCMA gene deletion in response to anti-BCMA CAR T cells in a patient with multiple myeloma
.
Nat Med.
2021
;
27
(
4
):
616
-
619
.
6.
Leblay
N
,
Maity
R
,
Barakat
E
, et al
.
Cite-seq profiling of T cells in multiple myeloma patients undergoing BCMA targeting CAR-T or Bites immunotherapy [abstract]
.
Blood
.
2020
;
136
(
suppl 1
). Abstract 719.
7.
Samur
MK
,
Fulciniti
M
,
Aktas-Samur
A
, et al
.
Biallelic loss of BCMA triggers resistance to anti-BCMA CAR T cell therapy in multiple myeloma [abstract]
.
Blood
.
2020
;
136
(
suppl 1
). Abstract 721.
8.
Sawyer
JR
,
Tian
E
,
Shaughnessy
JD
Jr
, et al
.
Hyperhaploidy is a novel high-risk cytogenetic subgroup in multiple myeloma
.
Leukemia
.
2017
;
31
(
3
):
637
-
644
.
9.
Weinhold
N
,
Ashby
C
,
Rasche
L
, et al
.
Clonal selection and double-hit events involving tumor suppressor genes underlie relapse in myeloma
.
Blood
.
2016
;
128
(
13
):
1735
-
1744
.
10.
Höllein
A
,
Twardziok
SO
,
Walter
W
, et al
.
The combination of WGS and RNA-Seq is superior to conventional diagnostic tests in multiple myeloma: Ready for prime time?
Cancer Genet.
2020
;
242
:
15
-
24
.
11.
de Larrea
CF
,
Staehr
M
,
Lopez
AV
, et al
.
Defining an optimal dual-targeted CAR T-cell therapy approach simultaneously targeting BCMA and GPRC5D to prevent BCMA escape-driven relapse in multiple myeloma
.
Blood Cancer Discov.
2020
;
1
(
2
):
146
-
154
.
12.
Garfall
AL
,
June
CH
.
Trispecific antibodies offer a third way forward for anticancer immunotherapy
.
Nature
.
2019
;
575
(
7783
):
450
-
451
.
13.
Sawyer
JR
,
Tian
E
,
Heuck
CJ
, et al
.
Jumping translocations of 1q12 in multiple myeloma: a novel mechanism for deletion of 17p in cytogenetically defined high-risk disease
.
Blood
.
2014
;
123
(
16
):
2504
-
2512
.
14.
Delfau-Larue
MH
,
Klapper
W
,
Berger
F
, et al
.
European Mantle Cell Lymphoma Network
.
High-dose cytarabine does not overcome the adverse prognostic value of CDKN2A and TP53 deletions in mantle cell lymphoma
.
Blood
.
2015
;
126
(
5
):
604
-
611
.
15.
Finlin
BS
,
Gau
CL
,
Murphy
GA
, et al
.
RERG is a novel ras-related, estrogen-regulated and growth-inhibitory gene in breast cancer
.
J Biol Chem.
2001
;
276
(
45
):
42259
-
42267
.
16.
Mestre-Escorihuela
C
,
Rubio-Moscardo
F
,
Richter
JA
, et al
.
Homozygous deletions localize novel tumor suppressor genes in B-cell lymphomas
.
Blood
.
2007
;
109
(
1
):
271
-
280
.
17.
Rasche
L
,
Hudecek
M
,
Einsele
H
.
What is the future of immunotherapy in multiple myeloma?
Blood
.
2020
;
136
(
22
):
2491
-
2497
.

Author notes

*

K.M.K and L.R. contributed equally to this work.

All requests for raw and analyzed data and materials will be promptly reviewed by the University Hospital Würzburg and the MLL Munich Leukemia Laboratory to determine whether the request is subject to any confidentiality or data protection obligations. Any data and materials that can be shared will be released via a Material Transfer Agreement. Adherence to the Datenschutz-Grundverordnung (https://dsgvo-gesetz.de/ and https://data.consilium.europa.eu/doc/document/ST-5419-2016-REV-1/en/pdf) is mandatory for sharing WGS data. Thus, a complete whole genome raw data set for a single patient cannot be shared according to European law. Processed data from which the identification of a patient is not possible can be made available. For requests, contact Leo Rasche via e-mail at rasche_l@ukw.de.

The full-text version of this article contains a data supplement.

Supplemental data