Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2020 Sep;1(2):146-154.
doi: 10.1158/2643-3230.BCD-20-0020.

Defining an Optimal Dual-Targeted CAR T-cell Therapy Approach Simultaneously Targeting BCMA and GPRC5D to Prevent BCMA Escape-Driven Relapse in Multiple Myeloma

Affiliations

Defining an Optimal Dual-Targeted CAR T-cell Therapy Approach Simultaneously Targeting BCMA and GPRC5D to Prevent BCMA Escape-Driven Relapse in Multiple Myeloma

Carlos Fernández de Larrea et al. Blood Cancer Discov. 2020 Sep.

Erratum in

Abstract

CAR T-cell therapy for multiple myeloma (MM) targeting B-cell maturation antigen (TNFRSF17; BCMA) induces high overall response rates; however, relapse occurs commonly. Implicated in relapse is a reservoir of MM if cells lacking sufficient BCMA surface expression (antigen escape). We demonstrate that simultaneous targeting of an additional antigen-here, G protein-coupled receptor class-C group-5 member-D (GPRC5D)-can prevent BCMA escape-mediated relapse in a model of MM. To identify an optimal approach, we compare subtherapeutic doses of different forms of dual-targeted cellular therapy. These include (1) parallel-produced and pooled mono-targeted CAR T-cells, (2) bicistronic constructs expressing distinct CARs from a single vector, and (3) a dual-scFv "single-stalk" CAR design. When targeting BCMA-negative disease, bicistronic and pooled approaches had the highest efficacy, whereas for dual-antigen-expressing disease, the bicistronic approach was more efficacious than the pooled approach. Mechanistically, expressing two CARs on a single cell enhanced the strength of CAR T-cell/target cell interactions.

Keywords: adoptive cellular therapy; antigen escape; chimeric antigen receptor; immunotherapy; multiple myeloma.

PubMed Disclaimer

Conflict of interest statement

C. Fernández de Larrea is an employee/paid consultant for Bristol Myers Squibb, Takeda Oncology, Janssen, and Amgen, and reports receiving speaker's bureau honoraria from Bristol Myers Squibb, Janssen and Amgen. Y. Chen is an advisory board member for and reports receiving research commercial grants from Amgen, and reports receiving speaker's bureau honoraria from Bristol Myers Squibb, Janssen, and Takeda Oncology. R.J. Brentjens is an employee/paid consultant for JUNO Therapeutics, and Gracell Biotherapeutics Inc., and reports receiving research commercial grants from JUNO Therapeutics, and holds ownership interests (including patents) in JUNO Therapeutics. E.L. Smith is an employee/paid consultant for Bristol, Myers Squibb, Fate Therapeutics, and Precision Biosciences, reports receiving research commercial grants from Bristol Myers Squibb, and receiving other receiving other remuneration from Bristol Myers Squibb. No potential conflicts of interest were disclosed by the other authors.

Figures

Figure 1. Dual-targeted CAR T cells express both scFv's efficiently and specifically lyse target antigen–positive cells. A, BCMA/GPRC5D dual-targeted CAR strategies evaluated. (i, ii) simultaneous 1:1 infusion of independent CAR T cells manufactured in parallel; (iii–iv) bicistronic dual-CAR expression on T cells via a construct with a “self-cleaving” 2A peptide; (v) tandem-scFv, “single stalk” CAR design. SP, signal peptide; S, spacer; TM transmembrane domain. B, Expression of individual BCMA-targeted and GPRC5D-targeted scFvs on the surface of primary donor T cells by flow cytometry with reagents specific to either the BCMA-targeted scFv or the GPRC5D-targeted scFv. C, Total CAR expression and transduction efficiency as measured by flow cytometry with antibody to the spacer domain shared among all CARs. FMO, fluorescence-minus-one. D, Cytotoxicity measured by ATP-dependent bioluminescence after 24-hour coculture of CAR T cells with a monolayer of 3T3-aAPC ffLuc+ cells expressing the indicated antigen(s); normalized to aAPCs alone. Mean ± SD from representative biological triplicate shown.
Figure 1.
Dual-targeted CAR T cells express both scFv's efficiently and specifically lyse target antigen–positive cells. A, BCMA/GPRC5D dual-targeted CAR strategies evaluated. (i, ii) simultaneous 1:1 infusion of independent CAR T cells manufactured in parallel; (iii–iv) bicistronic dual-CAR expression on T cells via a construct with a “self-cleaving” 2A peptide; (v) tandem-scFv, “single stalk” CAR design. SP, signal peptide; S, spacer; TM transmembrane domain. B, Expression of individual BCMA-targeted and GPRC5D-targeted scFvs on the surface of primary donor T cells by flow cytometry with reagents specific to either the BCMA-targeted scFv or the GPRC5D-targeted scFv. C, Total CAR expression and transduction efficiency as measured by flow cytometry with antibody to the spacer domain shared among all CARs. FMO, fluorescence-minus-one. D, Cytotoxicity measured by ATP-dependent bioluminescence after 24-hour coculture of CAR T cells with a monolayer of 3T3-aAPC ffLuc+ cells expressing the indicated antigen(s); normalized to aAPCs alone. Mean ± SD from representative biological triplicate shown.
Figure 2. Upfront treatment with dual-targeted CAR T cells prevents BCMA escape–mediated relapse in a rechallenge model. A, Experimental scheme, 2 × 106 cells of the human bone marrow tropic myeloma cell line OPM2-WT were injected via tail vein into NSG mice and allowed to engraft and expand for 14 days. Mice were randomized to treatment with donor T cells (3 × 106) gene-modified as indicated. Mice in the treatment arms that showed signs of xenogeneic GvHD were euthanized, and on day 105 the remaining long-term surviving mice were challenged with OPM2-BCMA KO cells (2 × 106). B, Kaplan–Meier curves of overall survival after OPM2-WT injection. P values are indicative of each arm compared with BCMA-Δ control. C, Bioluminescent imaging of tumors over time. D, Kaplan–Meier curves after OPM2-BCMA KO injection. P values are indicative of each arm compared with BCMA-41BBz arm.
Figure 2.
Upfront treatment with dual-targeted CAR T cells prevents BCMA escape–mediated relapse in a rechallenge model. A, Experimental scheme, 2 × 106 cells of the human bone marrow tropic myeloma cell line OPM2-WT were injected via tail vein into NSG mice and allowed to engraft and expand for 14 days. Mice were randomized to treatment with donor T cells (3 × 106) gene-modified as indicated. Mice in the treatment arms that showed signs of xenogeneic GvHD were euthanized, and on day 105 the remaining long-term surviving mice were challenged with OPM2-BCMA KO cells (2 × 106). B, Kaplan–Meier curves of overall survival after OPM2-WT injection. P values are indicative of each arm compared with BCMA-Δ control. C, Bioluminescent imaging of tumors over time. D, Kaplan–Meier curves after OPM2-BCMA KO injection. P values are indicative of each arm compared with BCMA-41BBz arm.
Figure 3. Pooled and bicistronic CAR approaches demonstrate enhanced efficacy over the single-stalk approach at subtherapeutic doses in an established BCMA-escape model. A, Experimental scheme, NSG mice were injected intravenously with 2 × 106 OPM2 tumor cells, including a subpopulation (5%–10%) of BCMA-KO cells. Each OPM2 population had been modified to express a distinct luciferase with a nonoverlapping substrate (OPM2-BCMA KO, firefly luciferase; OPM2-WT, membrane-tethered Cypridina luciferase) for in vivo imaging from the same animals over time. Day 14, mice were randomized for treatment with 2.5 × 105 gene-modified T cells. B, Kaplan–Meier curves for mice treated with the indicated CAR T cells. C, Tumor burden by bioluminescent imaging over time of OPM2-WT membrane-tethered Cypridina luciferase+ (vargulin substrate) and OPM2-BCMA KO ffLuc+ (d-luciferin substrate).
Figure 3.
Pooled and bicistronic CAR approaches demonstrate enhanced efficacy over the single-stalk approach at subtherapeutic doses in an established BCMA-escape model. A, Experimental scheme, NSG mice were injected intravenously with 2 × 106 OPM2 tumor cells, including a subpopulation (5%–10%) of BCMA-KO cells. Each OPM2 population had been modified to express a distinct luciferase with a nonoverlapping substrate (OPM2-BCMA KO, firefly luciferase; OPM2-WT, membrane-tethered Cypridina luciferase) for in vivo imaging from the same animals over time. Day 14, mice were randomized for treatment with 2.5 × 105 gene-modified T cells. B, Kaplan–Meier curves for mice treated with the indicated CAR T cells. C, Tumor burden by bioluminescent imaging over time of OPM2-WT membrane-tethered Cypridina luciferase+ (vargulin substrate) and OPM2-BCMA KO ffLuc+ (d-luciferin substrate).
Figure 4. Bicistronic CAR approach demonstrates enhanced efficacy over the pooled approach at subtherapeutic doses in a model when target cells exclusively express both antigens. A, Experimental scheme: NSG mice were injected intravenously with a pure population of 2 × 106 OPM2-WT cells (endogenously expressing BCMA and GPRC5D). After a 14-day engraftment/expansion period, mice were randomized to the 4 treatment groups shown in B, each receiving a single injection of 2.5 × 105 gene-modified T cells. B, Tumor burden imaged at the time of treatment and day 14 posttreatment with gene-modified CAR T cells. C, Kaplan–Meier curves of overall survival. D, Summary of results. In the presence of BCMA-escape (GPRC5D-only expressing) target cells, the single-stalk CAR approach was less efficacious than pooled or bicistronic strategies, which were similarly efficacious to each other. In contrast, when targeting BCMA/GPRC5D dual-expressing target cells, the bicistronic strategy showed enhanced efficacy compared with the pooled approach.
Figure 4.
Bicistronic CAR approach demonstrates enhanced efficacy over the pooled approach at subtherapeutic doses in a model when target cells exclusively express both antigens. A, Experimental scheme: NSG mice were injected intravenously with a pure population of 2 × 106 OPM2-WT cells (endogenously expressing BCMA and GPRC5D). After a 14-day engraftment/expansion period, mice were randomized to the 4 treatment groups shown in B, each receiving a single injection of 2.5 × 105 gene-modified T cells. B, Tumor burden imaged at the time of treatment and day 14 posttreatment with gene-modified CAR T cells. C, Kaplan–Meier curves of overall survival. D, Summary of results. In the presence of BCMA-escape (GPRC5D-only expressing) target cells, the single-stalk CAR approach was less efficacious than pooled or bicistronic strategies, which were similarly efficacious to each other. In contrast, when targeting BCMA/GPRC5D dual-expressing target cells, the bicistronic strategy showed enhanced efficacy compared with the pooled approach.

Comment in

Similar articles

  • GPRC5D is a target for the immunotherapy of multiple myeloma with rationally designed CAR T cells.
    Smith EL, Harrington K, Staehr M, Masakayan R, Jones J, Long TJ, Ng KY, Ghoddusi M, Purdon TJ, Wang X, Do T, Pham MT, Brown JM, De Larrea CF, Olson E, Peguero E, Wang P, Liu H, Xu Y, Garrett-Thomson SC, Almo SC, Wendel HG, Riviere I, Liu C, Sather B, Brentjens RJ. Smith EL, et al. Sci Transl Med. 2019 Mar 27;11(485):eaau7746. doi: 10.1126/scitranslmed.aau7746. Sci Transl Med. 2019. PMID: 30918115 Free PMC article.
  • Efficacy and safety of chimeric antigen receptor T cells targeting BCMA and GPRC5D in relapsed or refractory multiple myeloma.
    Yang X, Wang F, Yuan X, Yang B, Chen J, Cheng J, Liu G, Tang D, Xu X, Wang S, He Z, Liu Y, Li Y. Yang X, et al. Front Immunol. 2024 Dec 23;15:1466443. doi: 10.3389/fimmu.2024.1466443. eCollection 2024. Front Immunol. 2024. PMID: 39763668 Free PMC article.
  • Mechanisms of antigen escape from BCMA- or GPRC5D-targeted immunotherapies in multiple myeloma.
    Lee H, Ahn S, Maity R, Leblay N, Ziccheddu B, Truger M, Chojnacka M, Cirrincione A, Durante M, Tilmont R, Barakat E, Poorebrahim M, Sinha S, McIntyre J, M Y Chan A, Wilson H, Kyman S, Krishnan A, Landgren O, Walter W, Meggendorfer M, Haferlach C, Haferlach T, Einsele H, Kortüm MK, Knop S, Alberge JB, Rosenwald A, Keats JJ, Rasche L, Maura F, Neri P, Bahlis NJ. Lee H, et al. Nat Med. 2023 Sep;29(9):2295-2306. doi: 10.1038/s41591-023-02491-5. Epub 2023 Aug 31. Nat Med. 2023. PMID: 37653344 Free PMC article.
  • GPRC5D-Targeted CAR T Cells for Myeloma.
    Mailankody S, Devlin SM, Landa J, Nath K, Diamonte C, Carstens EJ, Russo D, Auclair R, Fitzgerald L, Cadzin B, Wang X, Sikder D, Senechal B, Bermudez VP, Purdon TJ, Hosszu K, McAvoy DP, Farzana T, Mead E, Wilcox JA, Santomasso BD, Shah GL, Shah UA, Korde N, Lesokhin A, Tan CR, Hultcrantz M, Hassoun H, Roshal M, Sen F, Dogan A, Landgren O, Giralt SA, Park JH, Usmani SZ, Rivière I, Brentjens RJ, Smith EL. Mailankody S, et al. N Engl J Med. 2022 Sep 29;387(13):1196-1206. doi: 10.1056/NEJMoa2209900. N Engl J Med. 2022. PMID: 36170501 Free PMC article. Clinical Trial.
  • Beyond BCMA, why GPRC5D could be the right way: treatment strategies with immunotherapy at relapse after anti-BCMA agents.
    Del Giudice ML, Galimberti S, Buda G. Del Giudice ML, et al. Cancer Immunol Immunother. 2023 Dec;72(12):3931-3937. doi: 10.1007/s00262-023-03559-4. Epub 2023 Nov 4. Cancer Immunol Immunother. 2023. PMID: 37924369 Free PMC article. Review.

Cited by

References

    1. Lahuerta JJ, Paiva B, Vidriales MB, Cordón L, Cedena MT, Puig Net al. . Depth of response in multiple myeloma: a pooled analysis of three PETHEMA/GEM clinical trials. J Clin Oncol 2017;35:2900–10. - PMC - PubMed
    1. Bergsagel PL. Where we were, where we are, where we are going: progress in multiple myeloma. Am Soc Clin Oncol Educ Book 2014;34:199–203. - PubMed
    1. Bladé J, Rosiñol L, Fernández de Larrea C. How I treat relapsed myeloma. Blood 2015;125:1532–40. - PubMed
    1. Sonneveld P, Avet-Loiseau H, Lonial S, Usmani S, Siegel D, Anderson KCet al. . Treatment of multiple myeloma with high-risk cytogenetics: a consensus of the International Myeloma Working Group. Blood 2016;127:2955–62. - PMC - PubMed
    1. Ghobrial I, Cruz CH, Garfall A, Shah N, Munshi N, Kaufman Jet al. . Immunotherapy in multiple myeloma: accelerating on the path to the patient. Clin Lymphoma Myeloma Leuk 2019;19:332–44. - PubMed

Publication types

MeSH terms

Substances