SLAMF7: A Potential Target for CAR T-Cell Therapy in Multiple Myeloma
Simple Summary
Abstract
1. Introduction
2. SLAMF7 Target
3. The Challenges and Limitations of CAR-T Therapy in Multiple Myeloma
4. SLAMF7-CAR-T Construct
5. SLAMF7-CAR-T Clinical Trials
5.1. Autologous Approaches
5.2. Allogeneic (‘Off-the-Shelf’) Approaches
5.3. Dual-Targeting Strategies
6. Expert Opinion
7. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Malard, F.; Neri, P.; Bahlis, N.J.; Terpos, E.; Moukalled, N.; Hungria, V.T.M.; Manier, S.; Mohty, M. Multiple myeloma. Nat. Rev. Dis. Primers 2024, 10, 45. [Google Scholar] [CrossRef]
- Sun, K.; Wu, H.; Zhu, Q.; Gu, K.; Wei, H.; Wang, S.; Li, L.; Wu, C.; Chen, R.; Pang, Y.; et al. Global landscape and trends in lifetime risks of haematologic malignancies in 185 countries: Population-based estimates from GLOBOCAN 2022. EClinicalMedicine 2025, 83, 103193. [Google Scholar] [CrossRef]
- Dimopoulos, M.A.; Terpos, E.; Boccadoro, M.; Moreau, P.; Mateos, M.V.; Zweegman, S.; Cook, G.; Engelhardt, M.; Delforge, M.; Hajek, R.; et al. EHA-EMN Evidence-Based Guidelines for diagnosis, treatment and follow-up of patients with multiple myeloma. Nat. Rev. Clin. Oncol. 2025, 22, 680–700. [Google Scholar] [CrossRef]
- van de Donk, N.W.C.J.; Moreau, P.; San-Miguel, J.F.; Mateos, M.V.; Dimopoulos, M.A.; Zweegman, S.; Gay, F.; Engelhardt, M.; Mina, R.; Zamagni, E.; et al. Optimising T-cell immunotherapy in patients with multiple myeloma: Practical considerations from the European Myeloma Network. Lancet Haematol. 2025, 12, e635–e649. [Google Scholar] [CrossRef]
- Ailawadhi, S.; Arnulf, B.; Patel, K.; Cavo, M.; Nooka, A.K.; Manier, S.; Callander, N.; Costa, L.J.; Vij, R.; Bahlis, N.J.; et al. Ide-cel vs standard regimens in triple-class-exposed relapsed and refractory multiple myeloma: Updated KarMMa-3 analyses. Blood 2024, 144, 2389–2401. [Google Scholar] [CrossRef] [PubMed]
- Sharma, P.; Lin, X.; Xu, Z.; Kanapuru, B.; Theoret, M.R.; Sokolic, R.; Fashoyin-Aje, L.A. FDA Approval Summary: Idecabtagene Vicleucel for the Treatment of Triple-Class-Exposed, Relapsed or Refractory Multiple Myeloma. Clin. Cancer Res. 2025, 31, 3362–3367. [Google Scholar] [CrossRef] [PubMed]
- Jagannath, S.; Martin, T.G.; Lin, Y.; Cohen, A.D.; Raje, N.; Htut, M.; Deol, A.; Agha, M.; Berdeja, J.G.; Lesokhin, A.M.; et al. Long-Term (≥5-Year) Remission and Survival After Treatment with Ciltacabtagene Autoleucel in CARTITUDE-1 Patients with Relapsed/Refractory Multiple Myeloma. J. Clin. Oncol. 2025, 43, 2766–2771. [Google Scholar] [CrossRef]
- Natrajan, K.; Kaushal, M.; George, B.; Kanapuru, B.; Theoret, M.R. FDA Approval Summary: Ciltacabtagene Autoleucel for Relapsed or Refractory Multiple Myeloma. Clin. Cancer Res. 2024, 30, 2865–2871. [Google Scholar] [CrossRef]
- Surveillance, Epidemiology, and End Results Program. Cancer Stat Facts: Myeloma. NIH. Available online: https://seer.cancer.gov/statfacts/html/mulmy.html (accessed on 8 April 2021).
- Engelhardt, M.; Kortüm, K.M.; Goldschmidt, H.; Merz, M. Functional cure and long-term survival in multiple myeloma: How to challenge the previously impossible. Haematologica 2024, 109, 2420–2435. [Google Scholar] [CrossRef]
- Chu, E.; Wu, J.; Kang, S.S.; Kang, Y. SLAMF7 as a Promising Immunotherapeutic Target in Multiple Myeloma Treatments. Curr. Oncol. 2023, 30, 7891–7903. [Google Scholar] [CrossRef] [PubMed]
- Friend, R.; Bhutani, M.; Voorhees, P.M.; Usmani, S.Z. Clinical potential of SLAMF7 antibodies-focus on elotuzumab in multiple myeloma. Drug Des. Devel. Ther. 2017, 11, 893–900. [Google Scholar] [CrossRef] [PubMed]
- Veillette, A.; Guo, H. CS1, a SLAM family receptor involved in immune regulation, is a therapeutic target in multiple myeloma. Crit. Rev. Oncol. Hematol. 2013, 88, 168–177. [Google Scholar] [CrossRef]
- O’Neal, J.; Ritchey, J.K.; Cooper, M.L.; Niswonger, J.; González, S.L.; Street, E.; Rettig, M.P.; Gladney, S.W.; Gehrs, L.; Abboud, R.; et al. CS1 CAR-T targeting the distal domain of CS1 (SLAMF7) shows efficacy in high tumor burden myeloma model despite fratricide of CD8+CS1 expressing CAR-T cells. Leukemia 2022, 36, 1625–1634. [Google Scholar] [CrossRef]
- Luo, G.; Ni, R.; Huang, X.; Li, Y.; Luo, D. The role of signaling lymphocytic activation molecule (SLAM) family receptors in health and disease progression: Focusing on cancer and therapy. Clin. Immunol. 2025, 280, 110574. [Google Scholar] [CrossRef]
- Farhangnia, P.; Ghomi, S.M.; Mollazadehghomi, S.; Nickho, H.; Akbarpour, M.; Delbandi, A.A. SLAM-family receptors come of age as a potential molecular target in cancer immunotherapy. Front. Immunol. 2023, 14, 1174138. [Google Scholar] [CrossRef]
- Raedler, L.A. Empliciti (Elotuzumab): First SLAMF7 Antibody Therapy Approved for the Treatment of Patients with Previously Treated Multiple Myeloma. Am. Health Drug Benefits 2016, 9, 74–77. [Google Scholar] [PubMed] [PubMed Central]
- Bruno, B.; Wäsch, R.; Engelhardt, M.; Gay, F.; Giaccone, L.; D’Agostino, M.; Rodríguez-Lobato, L.G.; Danhof, S.; Gagelmann, N.; Kröger, N.; et al. European Myeloma Network perspective on CAR T-Cell therapies for multiple myeloma. Haematologica 2021, 106, 2054–2065. [Google Scholar] [CrossRef] [PubMed]
- Swan, D.; Madduri, D.; Hocking, J. CAR-T cell therapy in Multiple Myeloma: Current status and future challenges. Blood Cancer J. 2024, 14, 206. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Lonial, S.; Vij, R.; Harousseau, J.L.; Facon, T.; Moreau, P.; Mazumder, A.; Kaufman, J.L.; Leleu, X.; Tsao, L.C.; Westland, C.; et al. Elotuzumab in combination with lenalidomide and low-dose dexamethasone in relapsed or refractory multiple myeloma. J. Clin. Oncol. 2012, 30, 1953–1959. [Google Scholar] [CrossRef]
- Majzner, R.G.; Rietberg, S.P.; Sotillo, E.; Dong, R.; Vachharajani, V.T.; Labanieh, L.; Myklebust, J.H.; Kadapakkam, M.; Weber, E.W.; Tousley, A.M.; et al. Tuning the Antigen Density Requirement for CAR T-cell Activity. Cancer Discov. 2020, 10, 702–723. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Amatya, C.; Pegues, M.A.; Lam, N.; Vanasse, D.; Geldres, C.; Choi, S.; Hewitt, S.M.; Feldman, S.A.; Kochenderfer, J.N. Development of CAR T Cells Expressing a Suicide Gene Plus a Chimeric Antigen Receptor Targeting Signaling Lymphocytic-Activation Molecule F7. Mol. Ther. 2021, 29, 702–717. [Google Scholar] [CrossRef]
- Grywalska, E.; Sosnowska-Pasiarska, B.; Smok-Kalwat, J.; Pasiarski, M.; Niedźwiedzka-Rystwej, P.; Roliński, J. Paving the Way toward Successful Multiple Myeloma Treatment: Chimeric Antigen Receptor T-Cell Therapy. Cells 2020, 9, 983. [Google Scholar] [CrossRef]
- Mishra, A.K.; Gupta, A.; Dagar, G.; Das, D.; Chakraborty, A.; Haque, S.; Prasad, C.P.; Singh, A.; Bhat, A.A.; Macha, M.A.; et al. CAR-T-Cell Therapy in Multiple Myeloma: B-Cell Maturation Antigen (BCMA) and Beyond. Vaccines 2023, 11, 1721. [Google Scholar] [CrossRef] [PubMed]
- Zhou, X.; Rasche, L.; Kortüm, K.M.; Mersi, J.; Einsele, H. BCMA loss in the epoch of novel immunotherapy for multiple myeloma: From biology to clinical practice. Haematologica 2023, 108, 958–968. [Google Scholar] [CrossRef] [PubMed]
- Mishra, A.; Maiti, R.; Mohan, P.; Gupta, P. Antigen loss following CAR-T cell therapy: Mechanisms, implications, and potential solutions. Eur. J. Haematol. 2024, 112, 211–222. [Google Scholar] [CrossRef]
- Rejeski, K.; Jain, M.D.; Smith, E.L. Mechanisms of Resistance and Treatment of Relapse after CAR T-cell Therapy for Large B-cell Lymphoma and Multiple Myeloma. Transpl. Cell. Ther. 2023, 29, 418–428. [Google Scholar] [CrossRef]
- Tedder, B.; Bhutani, M. Resistance Mechanisms to BCMA Targeting Bispecific Antibodies and CAR T-Cell Therapies in Multiple Myeloma. Cells 2025, 14, 1077. [Google Scholar] [CrossRef] [PubMed]
- Shen, C.; Zhang, Z.; Zhang, Y. Chimeric Antigen Receptor T Cell Exhaustion during Treatment for Hematological Malignancies. Biomed. Res. Int. 2020, 2020, 8765028. [Google Scholar] [CrossRef]
- Liu, Z.; Lei, W.; Wang, H.; Liu, X.; Fu, R. Challenges and strategies associated with CAR-T cell therapy in blood malignancies. Exp. Hematol. Oncol. 2024, 13, 22. [Google Scholar] [CrossRef]
- Nussinov, R.; Tsai, C.J.; Jang, H. Anticancer drug resistance: An update and perspective. Drug Resist. Updates 2021, 59, 100796. [Google Scholar] [CrossRef]
- Valtis, Y.K.; Shouval, R. The fire or the forest: Untangling the role of inflammation and bone marrow health in CAR-T outcomes in acute lymphoblastic leukemia. Transplant. Cell. Ther. 2025, 31, 474–476. [Google Scholar] [CrossRef]
- Liu, Z.; Zhou, Z.; Dang, Q.; Xu, H.; Lv, J.; Li, H.; Han, X. Immunosuppression in tumor immune microenvironment and its optimization from CAR-T cell therapy. Theranostics 2022, 12, 6273–6290. [Google Scholar] [CrossRef]
- Sheykhhasan, M.; Ahmadieh-Yazdi, A.; Vicidomini, R.; Poondla, N.; Tanzadehpanah, H.; Dirbaziyan, A.; Mahaki, H.; Manoochehri, H.; Kalhor, N.; Dama, P. CAR T therapies in multiple myeloma: Unleashing the future. Cancer Gene Ther. 2024, 31, 667–686. [Google Scholar] [CrossRef]
- Cho, S.F.; Xing, L.; Anderson, K.C.; Tai, Y.T. Promising Antigens for the New Frontier of Targeted Immunotherapy in Multiple Myeloma. Cancers 2021, 13, 6136. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Vural, E.; Beksaç, M. Current Anti-Myeloma Chimeric Antigen Receptor-T Cells: Novel Targets and Methods. Balkan Med. J. 2025, 42, 301–310. [Google Scholar] [CrossRef] [PubMed]
- Schmidts, A.; Ormhøj, M.; Choi, B.D.; Taylor, A.O.; Bouffard, A.A.; Scarfò, I.; Larson, R.C.; Frigault, M.J.; Gallagher, K.; Castano, A.P.; et al. Rational design of a trimeric APRIL-based CAR-binding domain enables efficient targeting of multiple myeloma. Blood Adv. 2019, 3, 3248–3260. [Google Scholar] [CrossRef]
- Larson, R.C.; Kann, M.C.; Graham, C.; Mount, C.W.; Castano, A.P.; Lee, W.H.; Bouffard, A.A.; Takei, H.N.; Almazan, A.J.; Scarfó, I.; et al. Anti-TACI single and dual-targeting CAR T cells overcome BCMA antigen loss in multiple myeloma. Nat. Commun. 2023, 14, 7509. [Google Scholar] [CrossRef] [PubMed]
- Zhou, R.; Wu, S.T.; Yazdanifar, M.; Williams, C.; Sanders, A.; Brouwer, C.; Maher, J.; Mukherjee, P. Mucin-1-Targeted Chimeric Antigen Receptor T Cells Are Effective and Safe in Controlling Solid Tumors in Immunocompetent Host. J. Immunother. 2024, 47, 77–88. [Google Scholar] [CrossRef]
- Leivas, A.; Valeri, A.; Córdoba, L.; García-Ortiz, A.; Ortiz, A.; Sánchez-Vega, L.; Graña-Castro, O.; Fernández, L.; Carreño-Tarragona, G.; Pérez, M.; et al. NKG2D-CAR-transduced natural killer cells efficiently target multiple myeloma. Blood Cancer J. 2021, 11, 146. [Google Scholar] [CrossRef] [PubMed]
- Yan, S.; Ming, X.; Zheng, R.; Zhu, X.; Xiao, Y. Application of GPRC5D Targeting Therapy in Relapsed Refractory Multiple Myeloma. Cancer Med. 2025, 14, e70764. [Google Scholar] [CrossRef]
- Robat-Jazi, B.; Mahalleh, M.; Dashti, M.; Nejati, N.; Ahmadpour, M.; Alinejad, E.; Mohammadi, S.; Lorestani, P.; Hamidieh, A.A.; Habibi, M.A.; et al. A Systematic Review and Meta-analysis on the Safety and Efficacy of CAR T Cell Therapy Targeting GPRC5D in Patients with Multiple Myeloma: A New Insight in Cancer Immunotherapy. Anticancer Agents Med. Chem. 2025, 25, 1017–1028. [Google Scholar] [CrossRef]
- Moares, N.; Gonzalez-Garcia, P.; Yi-He, W.; Muñoz-Miranda, J.P.; Gabucio, A.; Luna-Espejo, R.; Ocaña-Cuesta, J.; Fernandez-Cisnal, R.; Fernandez-Ponce, C.M.; Garcia-Cozar, F. Dual targeting of BCMA and SLAMF7 with the CARtein system: Chimeric antigen receptors with intein-mediated splicing elicit specific T cell activation against multiple myeloma. Front. Immunol. 2025, 16, 1613222. [Google Scholar] [CrossRef]
- Zhao, J.; Zheng, M.; Ma, L.; Guan, T.; Su, L. From spear to trident: Upgrading arsenal of CAR-T cells in the treatment of multiple myeloma. Heliyon 2024, 10, e29997. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Larson, R.C.; Maus, M.V. Recent advances and discoveries in the mechanisms and functions of CAR T cells. Nat. Rev. Cancer 2021, 21, 145–161. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Ghorai, S.K.; Pearson, A.N. Current Strategies to Improve Chimeric Antigen Receptor T (CAR-T) Cell Persistence. Cureus 2024, 16, e65291. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Morris, E.C.; Neelapu, S.S.; Giavridis, T.; Sadelain, M. Cytokine release syndrome and associated neurotoxicity in cancer immunotherapy. Nat. Rev. Immunol. 2022, 22, 85–96. [Google Scholar] [CrossRef] [PubMed]
- Jain, M.D.; Smith, M.; Shah, N.N. How I treat refractory CRS and ICANS after CAR T-cell therapy. Blood 2023, 141, 2430–2442. [Google Scholar] [CrossRef] [PubMed]
- Bishop, M.R. Late complications and long-term care of adult CAR T-cell patients. Hematol. Am. Soc. Hematol. Educ. Program 2024, 2024, 109–115. [Google Scholar] [CrossRef]
- Dulobdas, V.; Kirkwood, A.A.; Serpenti, F.; Gautama, B.; Panopoulou, A.; Mathew, A.; Gabriel, S.; Malladi, R.; Pealing, J.; Bonney, D.; et al. Risk factors for CAR T-cell manufacturing failure and patient outcomes in large B-cell lymphoma: A report from the UK National CAR T Panel. Blood Cancer J. 2025, 15, 30. [Google Scholar] [CrossRef]
- Ayala Ceja, M.; Khericha, M.; Harris, C.M.; Puig-Saus, C.; Chen, Y.Y. CAR-T cell manufacturing: Major process parameters and nex Tea Gogishvili, t-generation strategies. J. Exp. Med. 2024, 221, e20230903. [Google Scholar] [CrossRef]
- Gonçalves, E. CAR-T cell therapies: Patient access and affordability solutions. Future Sci. OA 2025, 11, 2483613. [Google Scholar] [CrossRef] [PubMed]
- Gogishvili, T.; Danhof, S.; Prommersberger, S.; Rydzek, J.; Schreder, M.; Brede, C.; Einsele, H.; Hudecek, M. SLAMF7-CAR T cells eliminate myeloma and confer selective fratricide of SLAMF7+ normal lymphocytes. Blood 2017, 130, 2838–2847. [Google Scholar] [CrossRef]
- Diaconu, I.; Ballard, B.; Zhang, M.; Chen, Y.; West, J.; Dotti, G.; Savoldo, B. Inducible Caspase-9 Selectively Modulates the Toxicities of CD19-Specific Chimeric Antigen Receptor-Modified T Cells. Mol. Ther. 2017, 25, 580–592. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Available online: https://clinicaltrials.gov/study/NCT03958656 (accessed on 8 April 2021).
- Kandra, P.; Nandigama, R.; Eul, B.; Huber, M.; Kobold, S.; Seeger, W.; Grimminger, F.; Savai, R. Utility and Drawbacks of Chimeric Antigen Receptor T Cell (CAR-T) Therapy in Lung Cancer. Front. Immunol. 2022, 13, 903562. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Study Details | A Phase I/IIa Clinical Trial to Assess Feasibility, Safety and Antitumor Activity of Autologous SLAMF7 CAR-T Cells in Multiple Myeloma. Available online: https://clinicaltrials.gov/study/NCT04499339 (accessed on 8 April 2021).
- Prommersberger, S.; Reiser, M.; Beckmann, J.; Danhof, S.; Amberger, M.; Quade-Lyssy, P.; Einsele, H.; Hudecek, M.; Bonig, H.; Ivics, Z. CARAMBA: A first-in-human clinical trial with SLAMF7 CAR-T cells prepared by virus-free Sleeping Beauty gene transfer to treat multiple myeloma. Gene Ther. 2021, 28, 560–571. [Google Scholar] [CrossRef]
- Ivics, Z.; Hackett, P.B.; Plasterk, R.H.; Izsvak, Z. Molecular reconstruction of Sleeping Beauty, a Tc1-like transposon from fish, and its transposition in human cells. Cell 1997, 91, 501–510. [Google Scholar] [CrossRef]
- Hackett, P.B.; Largaespada, D.A.; Cooper, L.J.N. A transposon and transposase system for human application. Mol. Ther. 2010, 18, 674–683. [Google Scholar] [CrossRef]
- Mathur, R.; Zhang, Z.; He, J.; Galetto, R.; Gouble, A.; Chion-Sotinel, I.; Filipe, S.; Gariboldi, A.; Veeramachaneni, T.; Manasanch, E.E.; et al. Universal SLAMF7-specific CAR T-cells as treatment for multiple myeloma. Blood 2017, 130, 502. [Google Scholar]
- Korst, C.L.B.M.; O’Neill, C.; Bruins, W.S.C.; Cosovic, M.; Twickler, I.; Verkleij, C.P.M.; Le Clerre, D.; Themeli, M.; Chion-Sotinel, I.; Zweegman, S.; et al. Preclinical activity of allogeneic SLAMF7-specific CAR T-cells (UCARTCS1) in multiple myeloma. J. Immunother. Cancer 2024, 12, e008769. [Google Scholar] [CrossRef]
- Allogeneic CAR T-Cell Therapy UCARTCS1A Shows Promise for Multiple Myeloma. Available online: https://www.cgtlive.com/view/allogeneic-car-t-cell-therapy-ucartcs1a-shows-promise-for-multiple-myeloma (accessed on 8 April 2021).
- FDA Lifts Clinical Hold on Study Evaluating UCARTCS1 for Myeloma. ASH Clinical News. American Society of Hematology. Available online: https://ashpublications.org/ashclinicalnews/news/5433/FDA-Lifts-Clinical-Hold-on-Study-Evaluating (accessed on 8 April 2021).
- Lonez, C.; Breman, E. Allogeneic CAR-T Therapy Technologies: Has the Promise Been Met? Cells 2024, 13, 146. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Li, C.; Xu, J.; Luo, W.; Liao, D.; Xie, W.; Wei, Q.; Zhang, Y.; Wang, X.; Wu, Z.; Kang, Y.; et al. Bispecific CS1-BCMA CAR-T cells are clinically active in relapsed or refractory multiple myeloma. Leukemia 2024, 38, 149–159. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Zah, E.; Nam, E.; Bhuvan, V.; Tran, U.; Ji, B.Y.; Gosliner, S.B.; Wang, X.; Brown, C.E.; Chen, Y.Y. Systematically optimized BCMA/CS1 bispecific CAR-T cells robustly control heterogeneous multiple myeloma. Nat. Commun. 2020, 11, 2283. [Google Scholar] [CrossRef]
- Chen, K.H.; Wada, M.; Pinz, K.G.; Liu, H.; Shuai, X.; Chen, X.; Wang, X.; Brown, C.E.; Chen, Y.Y. A compound chimeric antigen receptor strategy for targeting multiple myeloma. Leukemia 2018, 32, 402–412. [Google Scholar] [CrossRef] [PubMed]
- Geis, M.; Nowotny, B.; Bohn, M.D.; Kouhestani, D.; Einsele, H.; Bumm, T.; Stuhler, G. Combinatorial targeting of multiple myeloma by complementing T cell engaging antibody fragments. Commun. Biol. 2021, 4, 44. [Google Scholar] [CrossRef]
- Ali, S.A.; Shi, V.; Maric, I.; Wang, M.; Stroncek, D.F.; Rose, J.J.; Brudno, J.N.; Stetler-Stevenson, M.; Feldman, S.A.; Hansen, B.G.; et al. T cells expressing an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma. Blood 2016, 128, 1688–1700. [Google Scholar] [CrossRef] [PubMed]
- Kolanu, N.D. CRISPR-Cas9 Gene Editing: Curing Genetic Diseases by Inherited Epigenetic Modifications. Glob. Med. Genet. 2024, 11, 113–122. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Lei, T.; Wang, Y.; Zhang, Y.; Yang, Y.; Cao, J.; Huang, J.; Chen, J.; Chen, H.; Zhang, J.; Wang, L.; et al. Leveraging CRISPR gene editing technology to optimize the efficacy, safety and accessibility of CAR T-cell therapy. Leukemia 2024, 38, 2517–2543. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Seijas, A.; Cora, D.; Novo, M.; Al-Soufi, W.; Sánchez, L.; Arana, Á.J. CRISPR/Cas9 Delivery Systems to Enhance Gene Editing Efficiency. Int. J. Mol. Sci. 2025, 26, 4420. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]

| PRO | CON | Potential Solutions | ||
|---|---|---|---|---|
| High expression in MM cells | Expressed in >95% of MM cells, including in R/R patients. Consistent and stable expression over time | Expression on normal immune cells | It is also expressed on NK cells, CD8+ T cells, B cells, and some dendritic cells. CAR-Ts targeting SLAMF7 may kill endogenous immune cells, leading to immune suppression, T-cell fratricide, or T-cell exhaustion during manufacture. | SLAMF7 knockout in the CAR-T product (most direct solution to fratricide). Careful monitoring of NK cell counts and immune function. Combination approaches with other MM targets to reduce reliance on SLAMF7 alone. Patient selection based on disease burden and immune status |
| Clinically validated target | Well-studied biologically in MM, reducing unknowns compared to novel antigens. It is the target of elotuzumab, which has a safety profile, | Fratricide and manufacturing complexity: | It is expressed on T cells themselves, which leads to fratricide (CAR-Ts killing each other). This requires gene editing (e.g., CRISPR/Cas9 or shRNA) to knock out SLAMF7 in the CAR-T product, adding complexity, cost, and potential risks. |
Virus-Free Manufacturing (Sleeping Beauty System)
Gene Editing to Knock Out SLAMF7 Natural Phenotype Acquisition |
| Lack of expression on HSCs | Unlike CD38 or CD138, SLAMF7 is not expressed on HSCs, thereby preserving hematopoietic reconstitution. | Modest clinical data: | SLAMF7 CAR-Ts have shown some preclinical promise, but clinical trials are limited and mostly in early phases. Some studies paused or terminated due to manufacturing or safety challenges. | Waiting for phase II trials |
| Possible use in dual-targeting CARs | SLAMF7 can be part of bispecific CAR-T strategies (e.g., SLAMF7 + BCMA), which may reduce escape and enhance durability. | Functional concerns | SLAMF7 may have co-stimulatory roles in T and NK cells. Eliminating SLAMF7+ immune cells could dampen immune surveillance and anti-MM immunity over time. | The success of dual-targeting CARs depends on carefully balancing specificity, activation strength, and T-cell persistence through rational design and iterative optimization based on functional assays. |
| Target | Characteristics | PRO | CON | Clinical Status/ Considerations |
|---|---|---|---|---|
| CD138 | also known as syndecan-1, is a cell surface protein that plays a significant role in MM | High expression on mature plasma cells and most MM cell—good tumor coverage in many patients. Well-characterized antigen used diagnostically in MM and not expressed on T cells or most hematopoietic stem/progenitor cells—no fratricide and low risk of long-term bone marrow aplasia from target expression on HSCs. Targeting CD138 will deplete malignant plasma cells directly (and normal plasma cells), which is therapeutically beneficial for disease control. | On-target/off-tumor toxicity: also expressed on some normal epithelial cells (e.g., specific mucosal/skin/epithelial compartments). Soluble/shed antigen: Syndecan-1 is shed into the circulation in MM (sCD138). Soluble CD138 can act as a decoy, lowering effective CAR binding, impairing tumor engagement, or causing inappropriate activation/exhaustion/tonic signaling. CD138 expression can be heterogeneous between clones and can be downregulated | Limited clinical experience; potential for poor CAR T persistence/efficacy against marrow niches. |
| CD19 | cell surface glycoprotein traditionally expressed on B cells and their precursors. | Large clinical experience in B-ALL and NHL. Potential to eradicate tumor-initiating clones. Useful in combination/sequential regimens: CD19 CARs can be administered after cytoreduction (e.g., post-ASCT) or combined with BCMA/GPRC5D CARs or bispecific strategies to reduce antigen-escape risk. Predictable, manageable toxicities. | Limited antigen expression on bulk disease: Most myeloma plasma cells lack CD19, so single-agent CD19 CAR T usually has limited direct anti-myeloma activity for the bulk tumor. The benefit is restricted to a minority of patients with measurable disease. | Clinical experience is limited to case reports, small pilot cohorts, and combination/sequential approaches. |
| CD38 | A prominent cell surface glycoprotein that plays a critical role in MM therapy. | High and frequent expression on MM plasma cells—good target coverage in many patients. Well-validated antigen biologically and clinically (therapeutic target of daratumumab/isatuximab)—a known safety/biology profile helps with risk assessment. Not restricted to late B-cell stages only, so can target a broad tumor population, including some CD19-clones. Off-the-shelf approaches feasible (CAR NK, transient mRNA CARs) have been developed to reduce long-term toxicity. | On-target/off-tumor toxicity | Most data are from early-phase studies (preclinical, first-in-human/phase I) with small cohorts. Reports show feasibility and some anti-myeloma activity, but safety concerns and durability remain an open issue |
| CD56 | also known as NCAM (Neural Cell Adhesion Molecule), is a cell surface protein with specific characteristics in MM | Expressed on a large fraction of MM.CD56 is a cell-surface molecule that serves as a biologically accessible antigen for CAR binding.CD56 can be used in patients who relapse after BCMA-directed therapies or in dual-target approaches (BCMA + CD56) to reduce antigen-escape risk. | Widely expressed in the nervous system and some neuroendocrine cells—risk of severe neurotoxicity, peripheral neuropathy, or autonomic dysfunction. NK cells and some T/NKT subsets express CD56; CAR T may deplete NK cells, causing impaired innate immunity and increased infection risk. T cells can upregulate CD56 during activation—CAR T cells may kill each other during expansion, reducing yield and function. Heterogeneous/absent expression on some myeloma clones—antigen escape Not all myeloma cells express CD56 or express it uniformly, which increases the risk of relapse from CD56—negative subclones. Variable antigen density/low target density | Minimal clinical data. Most work was preclinical or at the IND/early-phase planning stage due to serious safety concerns. |
| NKG2D Ligands | It is a crucial activating receptor in the immune system, primarily found on natural killer (NK) cells and specific T cell subsets | Upregulated in tumor and infected cells, not healthy ones. Broad expression. It can recognize multiple ligands via one receptor with a broad coverage. | It can be upregulated in normal tissues during inflammation with potential off-tumor effects. Soluble MICA/B can act as decoys, suppressing CAR-T function. MM cells can downregulate NKG2D ligands to escape immune detection. | Limited clinical validation; unclear clinical safety profile |
| MUC1 | Mucin1 or MUC1 (also known as CD227, EMA, MCD, MAM6, PEM, PUM, KL-6, CA 27.29/CA 15-3) is a highly glycosylated transmembrane mucin family member protein that is overexpressed in various solid or hematological cancers. | Overexpressed in MM, e specially in advanced or high-risk disease. Tumor-associated isoform (MUC1-C): this isoform is more selectively expressed in cancer cells and can be specifically targeted. Linked to drug resistance and stemness: targeting MUC1 could eliminate more aggressive or resistant clones. | Expression on normal epithelial cells. Glycosylation heterogeneity | Limited clinical data. Mostly preclinical or early-phase trials. |
| GPRC5D | Class C orphan G protein-coupled receptor is predominantly expressed in MM cells and hard keratinized tissues, with low expression in normal human tissues. | Uniformly and highly expressed in MM, even in patients who relapse after BCMA CAR-T. Low expression in normal tissues. It shows vigorous activity, including post-BCMA relapse. | On-target off-tumor toxicity: Nail, skin, taste-related side effects (e.g., dysgeusia, rash, nail changes), although generally manageable. Bispecifics competition. | The most clinically mature target. Demonstrates strong efficacy, even in patients with BCMA-refractory disease. |
| CD229 | Also known as Ly9, it is a cell surface receptor belonging to the SLAM family. | Broad expression: Many studies (preclinical and translational) report high and relatively uniform CD229 expression on malignant plasma cells, including some clones with low BCMA. This can increase tumor coverage and reduce the risk of antigen escape when used alone or in combination. Potential to target progenitor/subclone population | On-target/off-tumor toxicity: CD229 is expressed on normal lymphoid cells (B cells, subsets of T cells, and possibly NK/other hematopoietic cells). Fratricide and manufacturing difficulty. Immune dysfunction and dysregulation. Prolonged immune suppression. Antigen heterogeneity/escape. Soluble forms or modulation of CD229 are not well characterized; the potential impact on CAR function is uncertain. | Most data are preclinical or early translational; clinical toxicity spectrum, durability, and real-world feasibility are not well established. |
| APRIL | A Proliferation-Inducing Ligand, member of the TNF family | High specificity for myeloma cells: APRIL binds to BCMA, TACI, and BAFF-R, which are frequently overexpressed on MM plasma cells, providing potentially broad coverage. Dual receptor targeting Potential: By using APRIL-based ligands or diminished affinity CARs, the approach can target multiple receptors | APRIL interacts with receptors on normal immune cells, including B cells and other lymphocytes, thereby raising the risk of on-target/off-tumor toxicity (e.g., B cell aplasia, immune dysregulation). Variability in receptor expression levels may result in antigen escape or reduced efficacy. | APRIL-based CAR T cells have shown effective cytotoxicity against myeloma cells in preclinical models. |
| κ Light Chain | Components of immunoglobulins. MM cells express either κ or λ light chains, but not both. | MM express only one type of light chain (κ or λ), targeting κ allows selective killing of malignant cells while sparing normal B cells expressing the other type. It can be customized per patient based on the expressed light chain. Reduces the risk of total B-cell aplasia | Only ~60% of myeloma cases are κ-restricted. Therapy is not suitable for λ+ myeloma. Antigen escape can occur via downregulation or mutation. Some normal B cells expressing κ will still be targeted, leading to partial B-cell aplasia. | Useful for κ+ MM patients with minimal B-cell aplasia |
| CCR10 | Chemokine receptor involved in plasma cell homing to bone marrow and mucosal tissues, and overexpressed in some MM subtypes. | Targeting CCR10 may disrupt tumor localization or retention in the bone marrow niche, potentially leading to tumor-selective targeting. | Not universally or uniformly expressed in all MM patients. Some expression in mucosal tissues (e.g., skin, gut), raising concerns about off-tumor effects. | Limited clinical validation |
| CD44v6 | It is a cell surface glycoprotein involved in cell adhesion and migration. The v6 isoform is selectively expressed in several cancers, including MM. | More specific to malignant plasma cells than CD44 (ubiquitous). CD44v6 is associated with poor prognosis and disease aggressiveness. |
Expression in normal keratinocytes (related to cutaneous side effects).
Not all MM cells express CD44v6, which may lead to immune escape. Variable expression due to alternative splicing may affect durability and efficacy. | Encouraging results in MM and AML. Novel mechanisms, such as targeting the microenvironment or aggressive subtypes |
| FcRH5 | A B-lineage-specific protein expressed on normal and malignant plasma cells, including MM | High expression in MM. Limited to the B-cell lineage, reducing off-target effects. | Low expression on some normal B cells. Myeloma cells may reduce FcRH5 expression under selective conditions. | Under clinical evaluation. Relatively safe and promising target |
| TACI | Transmembrane Activator and CAML Interactor, also known as TNFRSF13B, is a receptor expressed on specific B cells and plasma cells. | Frequently overexpressed on MM plasma cells, providing a relevant target for cell therapy. It may help target BCMA-negative or low-expressing clones. TACI is found on some memory B cells, which may support immune reconstitution after therapy. | It is present on normal B cells and some immune regulatory cells, raising concerns about on-target/off-tumor toxicity, including B cell aplasia and immune deficiency. Safety concerns: Off-tumor toxicity could result in Potential for antigen escape: Tumor cells may downregulate TACI, leading to relapse. | TACI-targeted CAR T therapies are primarily in preclinical or early-stage trials, resulting in limited safety, efficacy, and durability data. |
| Integrin β7 | Integrin β7 is a cell adhesion molecule involved in the regulation of immune cell migration and adhesion. In the context of MM, it plays a role in the homing and retention of plasma cells within the bone marrow microenvironment. |
Selective expression in MM. Plays a role in MM homing and interaction with the bone marrow, and targeting may disrupt microenvironmental support.
Expression is limited primarily to lymphoid tissue, with potentially less systemic toxicity. | Integrin β7 is involved in lymphocyte homing, allowing for potential off-tumor effects. The long-term expression and resistance dynamics are not well understood. | Clinical data in preclinical or early-phase |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Alati, C.; Pitea, M.; Porto, G.; Bilardi, E.; Greve, M.B.; Vincelli, I.D.; Rizzuto, A.; Policastro, G.; Alvaro, M.E.; Utano, G.; et al. SLAMF7: A Potential Target for CAR T-Cell Therapy in Multiple Myeloma. Cancers 2025, 17, 3471. https://doi.org/10.3390/cancers17213471
Alati C, Pitea M, Porto G, Bilardi E, Greve MB, Vincelli ID, Rizzuto A, Policastro G, Alvaro ME, Utano G, et al. SLAMF7: A Potential Target for CAR T-Cell Therapy in Multiple Myeloma. Cancers. 2025; 17(21):3471. https://doi.org/10.3390/cancers17213471
Chicago/Turabian StyleAlati, Caterina, Martina Pitea, Gaetana Porto, Erica Bilardi, Maria Bruna Greve, Iolanda Donatella Vincelli, Andrea Rizzuto, Giorgia Policastro, Maria Eugenia Alvaro, Giovanna Utano, and et al. 2025. "SLAMF7: A Potential Target for CAR T-Cell Therapy in Multiple Myeloma" Cancers 17, no. 21: 3471. https://doi.org/10.3390/cancers17213471
APA StyleAlati, C., Pitea, M., Porto, G., Bilardi, E., Greve, M. B., Vincelli, I. D., Rizzuto, A., Policastro, G., Alvaro, M. E., Utano, G., Gerace, D., Allegra, A., Piro, E., Rossi, M., & Martino, M. (2025). SLAMF7: A Potential Target for CAR T-Cell Therapy in Multiple Myeloma. Cancers, 17(21), 3471. https://doi.org/10.3390/cancers17213471

