Next Article in Journal
Diffusion-Weighted MRI—The Way Forward for MRI in Myeloma?
Next Article in Special Issue
Immunotherapy in Myeloma: A Theme Issue in Honor of Prof. Dr. Gösta Gahrton
Previous Article in Journal
Light Chain Stabilization: A Therapeutic Approach to Ameliorate AL Amyloidosis
Previous Article in Special Issue
Donor Lymphocyte Infusion to Enhance the Graft-versus-Myeloma Effect
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Status of CAR-T Cell Therapy in Multiple Myeloma

Department of Hematology, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), University Hospital Virgen del Rocio, Universidad de Sevilla, 41013 Sevilla, Spain
*
Author to whom correspondence should be addressed.
Submission received: 30 July 2021 / Revised: 8 October 2021 / Accepted: 18 October 2021 / Published: 21 October 2021

Abstract

:
Current data on CAR-T cell-based therapy is really promising in multiple myeloma, especially in terms of response. In heavily pretreated patients, who have already received proteasome inhibitors, immunomodulatory drugs and monoclonal antibodies, current trials report an overall response rate ranging from 81 to 97% and 45 to 67% of complete remission rates. Data are less encouraging in terms of duration of response, although most recent trials have shown significant improvements in terms of event-free survival, with medians ranging from 8 to 14 months and up to 77% progression-free survival at 12 months with an acceptable toxicity profile. These data will be consolidated in future years and will provide new evidence on the best timing for CAR-T cell therapy. Moreover, new CAR-T designs are underway and will challenge the current results.

1. Introduction

The introduction of proteasome inhibitors (PI) and immunomodulatory drugs (IMIDs) in the early 2000 has improved survival in patients with multiple myeloma (MM).
Currently, the standard treatment of MM is based on a combination of drugs with different mechanisms of action and synergistic effects, including proteasome inhibitors (bortezomib, carfilzomib, ixazomib), immunomodulatory drugs (thalidomide, lenalidomide, pomalidomide), alkylating agents (melphalan, cyclophosphamide, bendamustine), steroids and, recently, anti-CD38 monoclonal antibodies (daratumumab, isatuximab) and anti-SLAMF7 monoclonal antibody (elotuzumab). Furthermore, the addition of immunotherapy with conjugated antibodies (belantamab mafadotin) represents a therapeutic approach for refractory patients, improving survival expectations among this patient population.
Although all these drugs have improved the outcome of MM, most patients still die due to disease progression [1]. Patients who are refractory to PI, IMIDs and alkylating agents have a median overall survival of less than a year [2,3].
Therapy with genetically modified T-cells expressing a chimeric antigen receptor (CAR) represents a cutting-edge approach. Results reported in acute lymphoblastic leukaemia (ALL) [4] and non-Hodgkin lymphoma (NHL) [5,6,7] with CD19 CAR-T cells has led to the search for other targets and to expand this treatment to other diseases, such as MM. Therefore, the identification of new antigens in plasma cells which can be used as a potential target has become a priority in the development of new therapeutic approaches based on immunotherapy. Thus, extensive efforts are being put into the development of new CAR therapies to treat MM as well as novel bispecific T cell engagers/antibodies (teclistamab, talquetamab). Unlike CAR-T cell products, bispecific antibodies do not require long production times or adequate lymphocyte counts. By contrast, CAR-T cells require only one dose instead of continuous therapy with bispecific antibodies [8].
Selection of an adequate antigen is a key factor for the development of an optimal CAR-T cell product. As antigen recognition does not depend on the human leukocyte antigen (HLA) system, a tumour target should be present on the cellular surface.
One of these antigens is B-cell maturation antigen (BCMA), which is highly expressed on the surface of malignant plasma cells but not on normal tissues, except for a low expression on mature B-cells [9].
Different antigens are currently being evaluated as possible targets for CAR therapy, including CD138, CD19, kappa light chain and BCMA. Some trials using these antigens have shown promising results, mainly in terms of response rate. However, no plateau has been observed in overall survival and disease-free survival curves, which translates the lack of durable remissions. Therefore, it will be necessary to overcome potential limitations hindering the efficacy of CAR-T cells in MM, such as lack of effectiveness, off-tumour toxicities, loss of antigen or interference with soluble protein present in patients’ plasma [10].

2. Results

Clinical trials of CAR-T cell therapy against MM have demonstrated promising clinical activity, providing unprecedented response rates in these heavily pretreated patients, the most commonly explored target being BCMA. There are more than 50 clinical trials ongoing using BCMA as a target. As mentioned previously, BCMA is a very specific antigen of plasma cells and mature B-cells, avoiding off-tumour toxicities following infusion [11,12].
The first clinical trial with BCMA-specific CAR was published in 2018 by Brudno et al. [13]. Sixteen patients received 9 × 106 CAR-BCMA T cells/kg. The patients had a median of 9.5 prior lines of therapy. The overall response rate was 81%, with 63% very good partial response or complete response. The median event-free survival was 31 weeks. Twelve patients (82%) developed CRS, including 6 (38%) with grade ≥ 3 CRS. Neurotoxicity was reported in 3 (19%) patients.
Idecabtagene vicleucel (ide-cel), initially known as bb2121, was developed by Bluebirdbio by transducing autologous T lymphocytes with a lentiviral vector to incorporate a second-generation CAR composed of an anti-BCMA single-variable chain domain, 41BB costimulatory domain and CD3-zeta as a signalling domain [14,15]. Lymphodepletion chemotherapy consisted of fludarabine and cyclophosphamide. In the dose escalating phase, the following doses were analysed: 50 × 106, 150 × 106, 450 × 106 and 800 × 106 CAR-positive (CAR+) T cells, with a 20% variation allowed. The expansion phase was achieved with 150 × 106 to 450 × 106 CAR+ T cells. A phase 1 trial using ide-cel included 33 patients who received multiple lines of treatment. The overall response rate was 85% with 45% of complete remission. Cytokine release syndrome (CRS) incidence was 76%, although only 2 patients developed CRS grade ≥ 3. Results of the phase 2 trial (KarMMa) have been published by Munshi et al. [16,17]. Of 140 patients enrolled, 128 received ide-cel. Patients had a median of 6 prior lines of therapy, 84% were refractory to at least one PI, one IMID and one anti-CD38. Eighty-eight percent received bridging therapy during the manufacturing process, but only 4% had some degree of response. With a median follow-up of 13.3 months, 94 of 128 (73%) patients had a response, and 42 of 128 (33%) achieved a complete remission (CR) or better. Thirty-three of 128 (26%) had CR with minimal residual disease (MRD)-negative status. Median progression-free survival (PFS) was 8.8 months and median overall survival was 19.4 months. The most common side effects among the 128 infused patients included neutropenia in 117 (91%) patients, anaemia in 89 (70%) and thrombocytopenia in 81 (63%). One hundred and seven (84%) developed CRS, including 7 (5%) with grade ≥ 3 CRS. Neurotoxicity was reported in 23 (18%) patients and were of grade 3 in 4 (3%) patients. Persistence of CAR+ T cells was documented in 59% of patients at 6 months and in 36% at 12 months following the infusion.
In addition to ide-cel, Wang B.-Y. et al. have developed a bispecific CAR with two BCMA binding sites (ciltacabtagene autoleucel or cilta-cel) [18,19]. A phase 1 study enrolled 57 patients, and lymphodepletion chemotherapy was based on single-agent cyclophosphamide. Fifty-one of 57 (90%) patients developed CRS, although only 7% had grade ≥ 3 CRS. Only one patient suffered from neurotoxicity. The overall response rate (ORR) was 88% with 47% of CR. Median PFS was 20 months. CAR+ T cells were not detectable in peripheral blood in 71% of patients at 4 months following infusion.
Similar results were reported in a phase 1b/2 study (CARTITUDE-1) performed in the United States [20]. Ninety-seven patients were enrolled; all of them had previously been exposed to PI, IMIDs and anti-CD38, and median lines of prior treatment was 6. Lymphodepletion included fludarabine and cyclophosphamide. The last update was presented at the European Hematology Association (EHA) congress in June 2021 [21]. The overall response rate was 97%, and 67% achieved CR. The median time to complete remission or better was 2 months (range, 1–15 months). Among 57 evaluable patients for MRD, 93% achieved MRD-negative status at 10−5. At 12 months, PFS was 77%, and overall survival (OS) was 89%. Median PFS has not been reached yet. The most common grade 3/4 toxicities were neutropenia in 95% of patients, anaemia in 68% and thrombocytopenia in 60%. Cytokine release syndrome was reported in 95% of the patients, 4% were grade 3/4, median time to onset was 7 days and median duration was 4 days. One patient died due to grade 5 CRS and hemophagocytic lymphohistiocytosis (HLH). Neurotoxicity occurred in 21% of the patients, and 10% were grade 3/4.
Cohen et al. [22] conducted a phase I study (NCT02546167) to evaluate autologous T cells lentivirally-transduced with a fully-human, BCMA-specific CAR containing CD3ζ and 4-1BB signalling domains (CART-BCMA). Twenty-five subjects were treated in 3 cohorts: (1) 1-5 × 108 CART-BCMA cells alone; (2) cyclophosphamide 1.5 g/m2 + 1-5 × 107 CART-BCMA cells; and (3) cyclophosphamide 1.5 g/m2 + 1-5 × 108 CART-BCMA cells. Toxicities included CRS 22/25 patients (88%) (32% g3-4) and neurotoxicity 8/25 patients (32%) (12% G3-4). The following responses were seen: 44% in cohort 1, 20% in cohort 2 and 64% in cohort 3 (including 5PR, 5 VGPR and 2CR)
Finally, the Memorial Sloan Kettering Cancer Centre group has developed a fully human anti-BCMA CAR-T cell (JCARH125, orvacabtagene-autoleucel, orva-cel) [23]. Infusion ratio CD4:CD8 is 1:1 to enhance memory T cell expansion [24]. Phase 1/2 trial (EVOLVE study) [25] still has a follow-up of only 6 months, but ORR of patients who received doses between 300 and 600 × 106 CAR+ T cells was 92% and 35% were CR. Ninety-four percent of patients were refractory to one PI, one IMID and one anti-CD38, and median number of prior regimens was 6. Incidence of CRS was 89%, only 3% developed grade ≥ 3. Neurotoxicity occurred in 13%, 3% were grade ≥ 3. There were no data on PFS in this study at the time of writing this manuscript.
These encouraging results need to be confirmed in phase 3 studies. There are two ongoing phase 3 trials (KarMMa-3 and CARTITUDE-4) comparing the efficacy and safety of BCMA CAR-T cell versus other anti-MM therapies treatments, both given in early stages of the disease.
All these studies are summarized in Table 1.
An important issue which will lead to discussion will be to define the place of new alternative approaches, such as conjugated antibodies or bispecific antibodies in the MM treatment algorithm, and whether, due to their safety profile, there will be a patient profile who will benefit more from these approaches than from CAR-T cell treatment.
Unfortunately, although most anti-BCMA CAR-T cell studies have described remarkable efficacy in terms of responses, event-free survival curves did not show a plateau, and most patients eventually relapse. Mechanisms related to CAR-T cell failure or resistance are multifactorial, including patient’s characteristics and disease biological features [26]. Loss of antigen at the time of relapse is one of the main mechanisms of resistance. In this regard, a selection of a clone with homozygous deletion of BCMA has been recently reported as the underlying mechanism of immune escape after anti-BCMA CAR-T cell therapy [27].
There are three ways to overcome this obstacle, namely CAR-T cells directed towards other antigens, dual CAR-T cells and antigen overexpression strategies [28,29].
Regarding the development of dual CAR-T cells, one potential approach is the elaboration through a bicistronic vector of two different CARs on the same T cell [30,31], another approach is the administration of two CAR-T cells produced independently and infused together or sequentially. Fernandez de Larrea et al. [30] demonstrated that expressing two CARs on a single cell enhanced the strength of CAR-T cell/target cell interactions. Also, developing a single product significantly reduces cost resources and time.
There are different ongoing clinical trials evaluating the efficacy and safety of anti-CD38 CAR-T cells alone or in combination with other CARs. The phase 1 study NCT03464916 evaluates an anti-CD38 CAR-T cell in relapse/refractory (R/R) MM patients. No results have been published yet. A phase 1/2 study, NCT03767751, is testing a dual anti-CD38 and BCMA CAR-T cells [32], and the phase 1/2 study NCT03125577 is assessing the combination of an anti-CD19 CAR-T cell plus an anti-CD38 CAR-T cell.
Regarding antigen overexpression strategies, the administration of an oral gamma secretase inhibitor to increase BCMA expression on the plasma cell surface has been assessed in a clinical trial (NCT03502577), and preliminary results in 6 patients showed an ORR of 100% [33,34,35]. In this sense, various approaches are being evaluated at the pre-clinical level, such as the case of trans retinoic acid (ATRA) (García-Guerrero et al.) [36]. It has recently been reported that BCMA expression in myeloma cells can be increased by epigenetic modulation with ATRA. After ATRA treatment, MM cells have an increased susceptibility to anti-BCMA CAR-T cell treatment in vitro and in vivo preclinical models, which can be further increased by combined treatment of ATRA and g-secretase inhibitors. Some other relevant pre-clinical data has been recently published. In this sense, GPRC5D has been reported as a novel target antigen for the immunotherapy of MM. GPRC5D is a human orphan family C G protein-coupled receptor recently described to be expressed on 98% of CD138-positive cells [37,38]. The restricted expression pattern of GPRC5D makes it an ideal target for immunotherapy. Consequently, GPRC5D CAR-T cells were generated by Smith et al. [38], showing anti-tumour efficacy against myeloma cells both in vitro and in vivo. Of note, GPRC5D CAR-T cells were also effective in eradication of myeloma cells after BCMA CAR-T cell treatment in a mouse model, which might be an option to overcome BCMA antigen escape.
Preclinical studies have also shown that CD138 is an effective target for the treatment of MM [39]. There is only one published study with an anti-CD138 CAR-T cells for R/R MM patients treated with chemotherapy and autologous stem cell transplant (ASCT). The CAR gene was detectable in peripheral blood of all patients and persisted for at least 4 weeks after the infusion. Four patients responded, but none of them achieved a CR; response lasted from 3 to 7 months. The remaining patients progressed despite having detectable CAR in marrow samples until day +90.
Although CD19 expression is uncommon on plasma cells, there is a small population of CD19+ myeloma cells which could constitute a reservoir of myeloma-initiating stem cells. The presence of CD19+ myeloma cells has been associated with a higher relapse rate and poor overall survival [40]. Therefore, targeting CD19 represents an interesting strategy to eliminate this subset of CD19+ cells. In the NCT02135406 study, ten patients with refractory MM received anti-CD19 CAR-T cells following an ASCT [41]. All patients received a previous ASCT, which resulted in a poor response with a PFS of less than one year. CD19 expression on myeloma cells was assessed by flow cytometry. As expected, the predominant myeloma population was CD19- in all patients. However, 7 out of 9 evaluable patients had subpopulations of CD19+ cells, ranging from 0.04% to 1.6%. In 10 of 11 subjects, the maximum planned dose of CTL019, 5 × 107 cells, was manufactured. In one subject, manufacturing was unsuccessful due to failure of autologous T cells to proliferate in culture. The median transduction efficiency was 10.1% (range 1.2–23.2), and the median total T cell dose was 4.4 × 108 (range 1.1 × 108 to 6.0 × 108). An ORR was achieved in 8 patients at 100 days after ASCT (including 1sCR, 4 VGPR, and 2 PR). This might be due to the fact that a significant fraction of myeloma cells expresses CD19 at molecular density, which is detectable by direct stochastic optical reconstruction microscopy (dSTORM) but not by flow cytometry [42]. Interestingly, less than 100 CD19 molecules are required for myeloma cell detection by CD19 CAR-T cells. In addition, evidence of a less differentiated MM subclone (CD19+ CD138−) with drug-resistance and disease propagating properties has emerged [40]. These results highlight antigen recognition by CAR even when it is present in very low density or not detectable by flow cytometry. Despite these encouraging findings, the use of CD19 CAR-T cells as a potential treatment for MM needs to be further explored. To determine whether CTL019 infusion improved PFS after ASCT, the authors compared each subject’s PFS after ASCT versus ASCT followed by CTL019. Two patients had significantly increased PFS after CTL019 (479 versus 181 days, 249 versus 127 days).
Yan L et al., a cooperative group from China, have published a phase 1 trial with 10 patients treated with sequential infusions of an anti-CD19 CAR-T cell followed by an anti-BCMA CAR-T cell [43,44]. Patients received lymphodepletion chemotherapy with fludarabine and cyclophosphamide on days -5, -4 and -3. Patients were infused on day 0 with a fixed dose of 1 × 107/kg antiCD19 CAR-T, on day 1 with 40% of anti-BCMA-CART and on day 2 with the remaining dose. Three dose levels were assessed for anti-BCMA CAR-T (3 × 107/kg, 5 × 107/kg and 6.5 × 107/kg). Median follow-up was 20 months. Ninety percent of patients developed CRS grade 1-2. Overall response rate was 90% with 40% of strict CR. Three out of 4 patients in strict RC maintained PFS at 2 years of follow-up.
A host immune response against a murine CAR is another potential limitation to CAR T cell persistence. Thus, developing a fully human CAR construct is an area of active research for several groups.
Jie J et al. developed the first fully human anti-BCMA CAR-T cell called CT053 [45]. Twenty-four patients with a median age of 60.1 years were included in the phase 1 trial. The subjects had a median of 4.5 prior regimens of therapy. They enrolled a high-risk population with extramedullary involvement (45.8%), ECOG score 2–3 (33.3%) and ISS grade 3 (37.5%). Overall response rate was 87.5% with 79.2% of CR. Among 20 subjects who underwent the evaluation of minimal residual disease (MRD) status, 17 achieved MRD-negative status. Median duration of response was 21.8 months. They demonstrated a good safety profile. The most common grade 3 or higher toxicities were neutropenia (66.7%), decreased lymphocyte count (79.2%) and thrombocytopenia (25%). In view of these results, a phase 1b/2 study (LUMMICAR-2) with CT053 is ongoing [46]. Patients received fludarabine and cyclophosphamide on days -5, -4 and -3. CT053 dose was 1.5–3.0 × 108, and it was administered in a single infusion. Median age was 59 years, and median number of prior lines of treatment was 6. Sixty-four percent of patients were refractory to 5 lines of treatment, and all received bridging therapy. Results published so far included 10 evaluable patients with a median follow-up of 4.5 months. Overall response rate was 100%, and 40% achieved at least a CR. Responses have been independent of BCMA expression in bone marrow. Peak CAR-T cell expansion was observed between 7 and 14 days after infusion. No grade 3 or higher CRS or neurotoxicity was observed.
Also, at the American Society of Hematology (ASH) meeting in 2020, the Kochenderfer group reported the results of a phase 1 trial with a fully human CAR-T cell which has a BCMA heavy chain single binding domain (FHVH-CD8BBZ) [47]. The FHVH33 binding domain lacks the light chain, artificial linker sequence and 2 associated junctions of a scFv, which can be immunogenic leading to CAR rejection. FHVH33-CD8BBZ was encoded by a γ-retroviral vector and incorporated FHVH33, CD8α hinge and transmembrane domains, a 4-1BB costimulatory domain and a CD3ζ domain. Twenty-one patients were enrolled, median number of prior lines of treatment was 6 and median age was 64 years. Lymphodepletion consisted of fludarabine and cyclophosphamide on days -5, -4 and -3. The maximum tolerated dose was 6 × 10 [6] CAR+ T cells /kg. The overall response rate was 90%. At the last cut-off, 10 patients maintained the response with a range of 0–80 weeks of follow-up. Ten patients discontinued the study, 9 due to disease progression and 1 due to death because of virus influenzae infection. Cytokine release syndrome occurred in 95% of patients, 20% were grade 3 and there were no grade 4 CRS. Thirty-eight percent developed neurotoxicity, but only 9% were grade 3.
Tumour microenvironment plays a crucial role in CAR-T cell resistance through immunological escape [48,49,50,51]. Some studies have shown that a high number of immunosuppressant cells, regulatory T cells, helper-2 T cells, cancer associated fibroblasts or osteoclasts contribute to decrease effector T cell activation and impair their function [51]. So, developing CAR-T cells against programmed death 1 and programmed death-ligand 1 (PD1/PDL1) might decrease the relapse risk related to the effect of microenvironment [52,53], but off-target toxicities might also increase.
Finally, and probably the most promising long-term strategy to overcome current limitations is the development of allogeneic CAR-T cells. There are already several phase 1 clinical trials assessing allogeneic CAR-T cells in R/R MM patients (UNIVERSAL trial, NCT04093596; MELANI-01 trial, NCT04142619; ALLO-605-201, NCT05000450; BCMA-UCART, NCT03752541; CTX120, NCT04244656; CYAD-211, NCT04613557). The reduction in time to infusion may be critical for life expectancy in a MM patient with refractory disease. Products from patients with fewer prior lines of treatment have a higher proportion of memory T cells and better ratio of CD4 T cell/CD8 T cells, which might improve the duration and depth of response 53. This statement must be confirmed in further studies since Yan et al. [44] describe 3 patients infused with alloCAR products who had early relapses. In this sense, Shah et al. designed a clinical trial with a next-generation CAR-T cell (bb21217) [54]. bb21217 is an anti-BCMA CAR-T cell therapy that uses the same CAR molecule as idecabtagene vicleucel (bb2121) but adds the PI3K inhibitor bb007 during ex vivo culture to enrich the cell product for memory-like T cells, thereby reducing the proportion of highly differentiated or senescent T cells. In the update presented at the American Society of Hematology Annual Meeting 2020, response was assessed per investigator for 44 patients with ≥2 months of follow up or PD/death within 2 months. Twenty-four (55%) patients had confirmed response per IMWG criteria, including 8 (18%) with ≥CR and 13 (30%) with VGPR. CRS occurred in 67% of patients and neurotoxicity in 22% [55]. In the context of allogeneic CAR-T cells, to decrease the risk of graft-versus-host disease (GvHD) several bioengineering methods have been planned to regulate the expression of T cell receptor (TCR) and major histocompatibility complex (MHC) [56,57].
Another field under development is the use of CARs in natural killer cells (NK) as NK cells reduce the risk of GvHD and CRS [58,59]. There is an ongoing phase 1/2 study with anti-BCMA CAR NK cells (NCT03940833).

3. Conclusions

Exciting times are ahead of us, with this wide variety of options for improvement. Soon, the CARs we will be administering will differ greatly from the ones we have available now, including those not approved yet in Europe for commercial use. Furthermore, defining the profile of patients who will benefit from these treatments in an early stage of the disease remains an unsolved challenge.

Author Contributions

J.L.R.-O. wrote and revised the manuscript and references and supervised the table. E.G.-G. wrote the manuscript and table, assisted in the elaboration of the references list. J.A.P.-S. supervised the manuscript, figures and references. All authors have read and agreed to the published version of the manuscript.

Funding

The authors would like to thank the CIBERONC (CB16/12/00480) and Red TerCel, and ISCIII (RD16/0011/0015, RD16/0011/0035).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

Jose A Perez Simon participated in advisory boards and/or educational sessions and/or research projects from Novartis, BMS/Celgene, Kyte, JANSSEN. All other authors declare no conflict of interest.

References

  1. Gandhi, U.H.; Cornell, R.F.; Lakshman, A.; Gahvari, Z.J.; McGehee, E.; Jagosky, M.H.; Gupta, R.; Varnado, W.; Fiala, M.A.; Chhabra, S.; et al. Outcomes of patients with multiple myeloma refractory to CD38-targeted monoclonal antibody therapy. Leukemia 2019, 33, 2266–2275. [Google Scholar] [CrossRef]
  2. Schinke, C.; Hoering, A.; Wang, H.; Carlton, V.; Thanandrarajan, S.; Deshpande, S.; Patel, P.; Molnar, G.; Susanibar, S.; Mohan, M.; et al. The prognostic value of the depth of response in multiple myeloma depends on the time of assessment, risk status and molecular subtype. Haematologica 2017, 102, e313–e316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Kumar, S.; Paiva, B.; Anderson, K.C.; Durie, B.; Landgren, O.; Moreau, P.; Munshi, N.; Lonial, S.; Bladé, J.; Mateos, M.-V.; et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol. 2016, 17, e328–e346. [Google Scholar] [CrossRef]
  4. Maude, S.L.; Laetsch, T.W.; Buechner, J.; Rives, S.; Boyer, M.; Bittencourt, H.; Bader, P.; Verneris, M.R.; Stefanski, H.E.; Myers, G.D.; et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N. Engl. J. Med. 2018, 378, 439–448. [Google Scholar] [CrossRef]
  5. Schuster, S.J.; Bishop, M.R.; Tam, C.S.; Waller, E.K.; Borchmann, P.; McGuirk, J.P.; Jäger, U.; Jaglowski, S.; Andreadis, C.; Westin, J.R.; et al. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2019, 380, 45–56. [Google Scholar] [CrossRef]
  6. Neelapu, S.S.; Locke, F.L.; Bartlett, N.L.; Lekakis, L.J.; Miklos, D.B.; Jacobson, C.A.; Braunschweig, I.; Oluwole, O.O.; Siddiqi, T.; Lin, Y.; et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 2531–2544. [Google Scholar] [CrossRef] [PubMed]
  7. Abramson, J.S.; Palomba, M.L.; Gordon, L.I.; Lunning, M.A.; Wang, M.; Arnason, J.; Mehta, A.; Purev, E.; Maloney, D.G.; Andreadis, C.; et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): A multicentre seamless design study. Lancet 2020, 396, 839–852. [Google Scholar] [CrossRef]
  8. Barilà, G.; Rizzi, R.; Zambello, R.; Musto, P. Drug Conjugated and Bispecific Antibodies for Multiple Myeloma: Improving Immunotherapies off the Shelf. Pharmaceuticals 2021, 14, 40. [Google Scholar] [CrossRef]
  9. Carpenter, R.O.; Evbuomwan, M.O.; Pittaluga, S.; Rose, J.J.; Raffeld, M.; Yang, S.; Gress, R.E.; Hakim, F.T.; Kochenderfer, J.N. B-cell maturation antigen is a promising target for adoptive T-cell therapy of multiple myeloma. Clin. Cancer Res. 2013, 19, 2048–2060. [Google Scholar] [CrossRef] [Green Version]
  10. Kravets, V.G.; Zhang, Y.; Sun, H. Chimeric-Antigen-Receptor (CAR) T Cells and the Factors Influencing their Therapeutic Efficacy. J. Immunol. Res. Ther. 2017, 2, 100–113. [Google Scholar] [PubMed]
  11. Green, D.J.; Pont, M.; Sather, B.D.; Cowan, A.J.; Turtle, C.J.; Till, B.G.; Nagengast, A.M.; Libby, E.N., III; Becker, P.S.; Coffey, D.G.; 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, 1011. [Google Scholar] [CrossRef]
  12. Timmers, M.; Roex, G.; Wang, Y.; Campillo-Davo, D.; Van Tendeloo, V.F.I.; Chu, Y.; Berneman, Z.; Luo, F.; Van Acker, H.H.; Anguille, S. Chimeric Antigen Receptor-Modified T Cell Therapy in Multiple Myeloma: Beyond B Cell Maturation Antigen. Front. Immunol. 2019, 10, 1613. [Google Scholar] [CrossRef] [PubMed]
  13. Brudno, J.N.; Maric, I.; Hartman, S.D.; Rose, J.J.; Wang, M.; Lam, N.; Stetler-Stevenson, M.; Salem, D.; Yuan, C.; Pavletic, S.; 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, 2267–2280. [Google Scholar] [CrossRef]
  14. Raje, N.; Berdeja, J.; Lin, Y.; Siegel, D.; Jagannath, S.; Madduri, D.; Liedtke, M.; Rosenblatt, J.; Maus, M.V.; Turka, A.; et al. Anti-BCMA CAR T-Cell Therapy bb2121 in Relapsed or Refractory Multiple Myeloma. N. Engl. J. Med. 2019, 380, 1726–1737. [Google Scholar] [CrossRef]
  15. Berdeja, J.G.; Alsina, M.; Shah, N.D.; Siegel, D.S.; Jagannath, S.; Madduri, D.; Kaufman, J.L.; Munshi, N.C.; Rosenblatt, J.; Jasielec, J.K.; et al. Updated Results from an Ongoing Phase 1 Clinical Study of bb21217 Anti-Bcma CAR T Cell Therapy. Blood 2019, 134, 927. [Google Scholar] [CrossRef]
  16. Munshi, N.C.; Anderson, J.L.D., Jr.; Shah, N.; Jagannath, S.; Berdeja, J.G.; Lonial, S.; Raje, N.S.; Siegel, D.S.D.; Lin, Y.; Oriol, A.; et al. 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, 8503. [Google Scholar] [CrossRef]
  17. Oriol, A.; Abril, L.; Torrent, A.; Ibarra, G.; Ribera, J.-M. The role of idecabtagene vicleucel in patients with heavily pretreated refractory multiple myeloma. Ther. Adv. Hematol. 2021, 12. [Google Scholar] [CrossRef]
  18. Wang, B.-Y.; Zhao, W.-H.; Liu, J.; Chen, Y.-X.; Cao, X.-M.; Yang, Y.; Zhang, Y.-L.; Wang, F.-X.; Zhang, P.-Y.; Lei, B.; 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, 579. [Google Scholar] [CrossRef]
  19. Zhao, W.-H.; Liu, J.; Wang, B.-Y.; Chen, Y.-X.; Cao, X.-M.; Yang, Y.; Zhang, Y.-L.; Wang, F.-X.; Zhang, P.-Y.; Lei, B.; 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. [Google Scholar] [CrossRef]
  20. Madduri, D.; Usmani, S.Z.; Jagannath, S.; Singh, I.; Zudaire, E.; Yeh, T.-M.; Allred, A.J.; Banerjee, A.; Goldberg, J.D.; Schecter, J.M.; 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, 577. [Google Scholar] [CrossRef]
  21. Usmani, S.Z.; Berdeja, J.G.; Madduri, D.; Jakubowiak, A.J.; Agha, M.E.; Cohen, A.D.; Hari, P.; Yeh, T.-M.; Olyslager, Y.; Banerjee, A.; et al. Ciltacabtagene autoleucel, a B-cell maturation antigen (BCMA)-directed chimeric antigen receptor T-cell (CAR-T) therapy, in relapsed/refractory multiple myeloma (R/R MM): Updated results from CARTITUDE-1. J. Clin. Oncol. 2021, 39, 8005. [Google Scholar] [CrossRef]
  22. Cohen, A.D.; Garfall, A.L.; Stadtmauer, E.A.; Melenhorst, J.J.; Lacey, S.F.; Lancaster, E.; Vogl, D.T.; Weiss, B.M.; Dengel, K.; Nelson, A.; et al. B cell maturation antigen–specific CAR T cells are clinically active in multiple myeloma. J. Clin. Investig. 2019, 129, 2210–2221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Mailankody, S.; Htut, M.; Lee, K.P.; Bensinger, W.; Devries, T.; Piasecki, J.; Ziyad, S.; Blake, M.; Byon, J.; Jakubowiak, A. 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, 957. [Google Scholar] [CrossRef]
  24. Sommermeyer, D.; Hudecek, M.; Kosasih, P.L.; Gogishvili, T.; Maloney, D.G.; Turtle, C.J.; Riddell, S.R. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia 2016, 30, 492–500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Mailankody, S.; Jakubowiak, A.J.; Htut, M.; Costa, L.J.; Lee, K.; Ganguly, S.; Kaufman, J.L.; Siegel, D.S.D.; Bensinger, W.; Cota, M. Orvacabtagene autoleucel (orva-cel), a B- cellmaturation 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, 8504. [Google Scholar] [CrossRef]
  26. Fraietta, J.A.; Lacey, S.F.; Orlando, E.J.; Pruteanu-Malinici, I.; Gohil, M.; Lundh, S.; Boesteanu, A.C.; Wang, Y.; O’Connor, R.S.; Hwang, W.-T.; et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat. Med. 2018, 24, 563–571. [Google Scholar] [CrossRef]
  27. Da Vià, M.C.; Dietrich, O.; Truger, M.; Arampatzi, P.; Duell, J.; Heidemeier, A.; Zhou, X.; Danhof, S.; Kraus, S.; Chatterjee, 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, 616–619. [Google Scholar] [CrossRef]
  28. Works, M.; Soni, N.; Hauskins, C.; Sierra, C.; Baturevych, A.; Jones, J.C.; Curtis, W.; Carlson, P.; Johnstone, T.G.; Kugler, D.; et al. Anti-B-cell maturation antigen chimeric antigen receptor T cell function against multiple myeloma is enhanced in the presence of lenalidomide. Mol. Cancer Ther. 2019, 18, 2246–2257. [Google Scholar] [CrossRef] [Green Version]
  29. Wang, X.; Walter, M.; Urak, R.; Weng, L.; Huynh, C.; Lim, L.; Wong, C.W.; Chang, W.-C.; Thomas, S.; Sanchez, J.F.; et al. Lenalidomide Enhances the Function of CS1 Chimeric Antigen Receptor–Redirected T Cells Against Multiple Myeloma. Clin. Cancer Res. 2018, 24, 106–119. [Google Scholar] [CrossRef] [Green Version]
  30. de Larrea, C.F.; Staehr, M.; Lopez, A.V.; Ng, K.Y.; Chen, Y.; Godfrey, W.D.; Purdon, T.J.; Ponomarev, V.; Wendel, H.-G.; Brentjens, R.J.; et al. Defining an optimal dualtargeted CAR T- cell therapy approach simultaneously targeting BCMA and GPRC5D to prevent BCMA escape– driven relapse in multiple myeloma. Blood Cancer Discov. 2020, 1, 146–154. [Google Scholar] [CrossRef]
  31. 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] [PubMed]
  32. Li, C.; Mei, H.; Hu, Y.; Guo, T.; Liu, L.; Jiang, H.; Tang, L.; Wu, Y.; Ai, L.; Deng, J. A bispecific CAR-T cell therapy targeting BCMA and CD38 for relapsed/refractory multiple myeloma: Updated results from a phase 1 dose-climbing trial. Blood 2019, 134, 930. [Google Scholar] [CrossRef]
  33. Pont, M.J.; Hill, T.; Cole, G.O.; Abbott, J.J.; Kelliher, J.; Salter, A.I.; Hudecek, M.; Comstock, M.L.; Rajan, A.; Patel, B.K.R.; et al. γ-Secretase inhibition increases efficacy of BCMA-specific chimeric antigen receptor T cells in multiple myeloma. Blood 2019, 134, 1585–1597. [Google Scholar] [CrossRef] [PubMed]
  34. Laurent, S.A.; Hoffmann, F.S.; Kuhn, P.-H.; Cheng, Q.; Chu, Y.; Schmidt-Supprian, M.; Hauck, S.; Schuh, E.; Krumbholz, M.; Rübsamen, H.; et al. γ-secretase directly sheds the survival receptor BCMA from plasma cells. Nat. Commun. 2015, 6, 7333. [Google Scholar] [CrossRef]
  35. Cowan, A.J.; Pont, M.; Sather, B.D.; Turtle, M.C.J.; Till, B.G.; Nagengast, R.A.M.; Libby, I.E.N.; Becker, P.S.; Coffey, D.G.; Tuazon, S.A.; 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, 204. [Google Scholar] [CrossRef]
  36. Garcia-Guerrero, G.; Rodríguez-Lobato, L.G.; Danhof, S.; Sierro-Martínez, B.; Goetz, R.; Sauer, M.; Perez-Simon, J.A.; Einsele, H.; Hudecek, M.; Prommersbe, S. ATRA Augments BCMA Expression on Myeloma Cells and Enhances Recognition By BCMA-CAR T-Cells. Blood 2020, 136, 13–14. [Google Scholar] [CrossRef]
  37. Bräuner-Osborne, H.; Jensen, A.A.; Sheppard, P.O.; Brodin, B.; Krogsgaard-Larsen, P.; O’Hara, P. Cloning and characterization of a human orphan family C G-protein coupled receptor GPRC5D. Biochim. Et Biophys. Acta (BBA)-Gene Struct. Expr. 2001, 1518, 237–248. [Google Scholar] [CrossRef]
  38. Smith, E.L.; Harrington, K.; Staehr, M.; Masakayan, R.; Jones, J.; Long, T.J.; Ng, K.Y.; Ghoddusi, M.; Purdon, T.J.; Wang, X.; et al. GPRC5D is a target for the immunotherapy of multiple myeloma with rationally designed CAR T cells. Sci. Transl. Med. 2019, 11, eaau7746. [Google Scholar] [CrossRef] [PubMed]
  39. Sun, C.; Mahendravada, A.; Ballard, B.; Kale, B.; Ramos, C.; West, J.; Maguire, T.; McKay, K.; Lichtman, E.; Tuchman, S.; et al. Safety and efficacy of targeting CD138 with a chimeric antigen receptor for the treatment of multiple myeloma. Oncotarget 2019, 10, 2369–2383. [Google Scholar] [CrossRef] [Green Version]
  40. Matsui, W.; Wang, Q.; Barber, J.P.; Brennan, S.; Smith, B.D.; Borrello, I.; McNiece, I.; Lin, L.; Ambinder, R.F.; Peacock, C.; et al. Clonogenic Multiple Myeloma Progenitors, Stem Cell Properties, and Drug Resistance. Cancer Res. 2008, 68, 190–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Garfall, A.; Stadtmauer, E.; Hwang, W.; Lacey, S.F.; Melenhorst, J.J.; Krevvata, M.; Carroll, M.P.; Matsui, M.H.; Wang, Q.; Dhodapkar, M.V.; et al. Anti-CD19 CAR T cells with high-dose melphalan and autologous stem cell transplantation for refractory multiple myeloma. JCI Insight 2018, 3, e120505. [Google Scholar] [CrossRef] [PubMed]
  42. Nerreter, T.; Letschert, S.; Götz, R.; Doose, S.; Danhof, S.; Einsele, H.; Sauer, M.; Hudecek, M. Super-resolution microscopy reveals ultra-low CD19 expression on myeloma cells that triggers elimination by CD19 CAR-T. Nat. Commun. 2019, 10, 3137. [Google Scholar] [CrossRef] [PubMed]
  43. Yan, Z.; Cao, J.; Cheng, H.; Qiao, J.; Zhang, H.; Wang, Y.; Shi, M.; Lan, J.; Fei, X.; Jin, L.; 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–e529. [Google Scholar] [CrossRef]
  44. Yan, L.; Qu, S.; Shang, J.; Shi, X.; Kang, L.; Xu, N.; Zhu, M.; Zhou, J.; Jin, S.; Yao, W.; et al. Sequential CD19 and BCMA-specific CAR T-cell treatment elicits sustained remission of relapsed and/or refractory myeloma. Cancer Med. 2021, 10, 563–574. [Google Scholar] [CrossRef] [PubMed]
  45. Jie, J.; Hao, S.; Jiang, S.; Li, Z.; Yang, M.; Zhang, W.; Yu, K.; Xiao, J.; Meng, H.; Ma, L.; et al. Phase 1 Trial of the Safety and Efficacy of Fully Human Anti-Bcma CAR T Cells in Relapsed/Refractory Multiple Myeloma. Blood 2019, 134, 4435. [Google Scholar] [CrossRef]
  46. Kumar, S.; Baz, R.; Orlowski, R.; Larry, D.A., Jr.; Ma, H.; Shrewsbury, A.; Croghan, K.A.; Bilgi, M.; Kansagra, A.; Kapoor, P.; et al. Results from Lummicar-2: A Phase 1b/2 Study of Fully Human B-Cell Maturation Antigen-Specific CAR T Cells (CT053) in Patients with Relapsed and/or Refractory Multiple Myeloma. Blood 2020, 136, 28–29. [Google Scholar] [CrossRef]
  47. Mikkilineni, L.; Manasanch, E.; Vanasse, D.; Brudno, J.N.; Mann, J.; Sherry, R.; Goff, S.L.; Yang, J.C.; Lam, N.; Maric, I.; et al. Deep and Durable Remissions of Relapsed Multiple Myeloma on a First-in-Humans Clinical Trial of T Cells Expressing an Anti-B-Cell Maturation Antigen (BCMA) Chimeric Antigen Receptor (CAR) with a Fully-Human Heavy-Chain-Only Antigen Recognition Domain. Blood 2020, 136, 50–51. [Google Scholar] [CrossRef]
  48. Kawano, Y.; Moschetta, M.; Manier, S.; Glavey, S.; Görgün, G.T.; Roccaro, A.M.; Anderson, K.C.; Ghobrial, I.M. Targeting the bone marrow microenvironment in multiple myeloma. Immunol. Rev. 2015, 263, 160–172. [Google Scholar] [CrossRef] [PubMed]
  49. Yeku, O.O.; Purdon, T.J.; Koneru, M.; Spriggs, D.; Brentjens, R.J. Armored CAR T cells enhance antitumor efficacy and overcome the tumor microenvironment. Sci. Rep. 2017, 7, 10541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Zhang, C.; Peng, Y.; Hublitz, P.; Zhang, H.; Dong, T. Genetic abrogation of immune checkpoints in antigen-specific cytotoxic T-lymphocyte as a potential alternative to blockade immunotherapy. Sci. Rep. 2018, 8, 5549. [Google Scholar] [CrossRef] [PubMed]
  51. Sakemura, R.; Cox, M.J.; Hansen, M.J.; Hefazi, M.; Roman, C.M.; Schick, K.J.; Tapper, E.E.; Moreno, P.R.; Ruff, M.W.; Walters, D.K.; et al. Targeting Cancer Associated Fibroblasts in the Bone Marrow Prevents Resistance to Chimeric Antigen Receptor T Cell Therapy in Multiple Myeloma. Blood 2019, 134, 865. [Google Scholar] [CrossRef]
  52. Rafiq, S.; Yeku, O.O.; Jackson, H.J.; Purdon, T.J.; Van Leeuwen, D.G.; Drakes, D.J.; Song, M.; Miele, M.M.; Li, Z.; Wang, P.; et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat. Biotechnol. 2018, 36, 847–856. [Google Scholar] [CrossRef]
  53. Suarez, E.; Chang, D.-K.; Sun, J.; Sui, J.; Freeman, G.J.; Signoretti, S.; Zhu, Q.; Marasco, W.A. Chimeric antigen receptor T cells secreting anti-PD-L1 antibodies more effectively regress renal cell carcinoma in a humanized mouse model. Oncotarget 2016, 7, 34341–34355. [Google Scholar] [CrossRef] [Green Version]
  54. Shah, N.; Alsina, M.; Siegel, D.S.; Jagannath, S.; Madduri, D.; Kaufman, J.L.; Turka, A.; Lam, L.P.; Massaro, M.M.; Hege, K.; et al. Initial Results from a Phase 1 Clinical Study of bb21217, a Next-Generation Anti Bcma CAR T Therapy. Blood 2018, 132, 488. [Google Scholar] [CrossRef]
  55. Alsina, M.; Shah, N.; Raje, N.S.; Jagannath, S.; Madduri, D.; Kaufman, J.L.; Siegel, D.S.; Munshi, N.C.; Rosenblatt, J.; Lin, Y.; et al. Updated Results from the Phase I CRB-402 Study of Anti-Bcma CAR-T Cell Therapy bb21217 in Patients with Relapsed and Refractory Multiple Myeloma: Correlation of Expansion and Duration of Response with T Cell Phenotypes. Blood 2020, 136, 25–26. [Google Scholar] [CrossRef]
  56. Garfall, A.L.; Dancy, E.K.; Cohen, A.D.; Hwang, W.-T.; Fraietta, J.A.; Davis, M.M.; Levine, B.L.; Siegel, D.L.; Stadtmauer, E.A.; Vogl, D.T.; et al. T-cell phenotypes associated with effective CAR T-cell therapy in postinduction vs relapsed multiple myeloma. Blood Adv. 2019, 3, 2812–2815. [Google Scholar] [CrossRef] [Green Version]
  57. Sadelain, M.; Rivière, M.S.I.; Riddell, S. Therapeutic T cell engineering. Nat. Cell Biol. 2017, 545, 423–431. [Google Scholar] [CrossRef] [Green Version]
  58. Themeli, M.; Kloss, C.C.; Ciriello, G.; Fedorov, V.D.; Perna, F.; Gonen, M.; Sadelain, M. Generation of tumor-targeted human T lymphocytes from induced pluripotent stem cells for cancer therapy. Nat. Biotechnol. 2013, 31, 928–933. [Google Scholar] [CrossRef] [PubMed]
  59. Bjordahl, R.; Gaidarova, S.; Goodridge, J.P.; Mahmood, S.; Bonello, G.; Robinson, M.; Ruller, C.; Pribadi, M.; Lee, T.; Abujarour, R.; et al. FT576: A novel multiplexed engineered off-the- shelf natural killer cell immunotherapy for the dualtargeting of CD38 and Bcma for the treatment of multiple myeloma. Blood 2019, 134, 3214. [Google Scholar] [CrossRef]
Table 1. Summary of clinical trials on MM using CAR-T.
Table 1. Summary of clinical trials on MM using CAR-T.
StudynPhaseVectorProductCostimulatory DomainLD ChemoTherapyCAR_T Cell DosePrevious Lines MedianCRS ≥ Grade 3ICANS ≥ Grade 3ORR %CR %MDR neg %Median PFS (Months)Median OS (Months)
CRB-401 1331LentiIde-Cel (bb2121)
Ide-Cel (bb2121)
4-1BBFluCy50/150/450/800 × 106 cells76385459411.8 NA
KArMMA 2,31282LentiIde-Cel (bb2121)4-1BBFluCy150/300/450
× 106 cells
6637353338.819.4
LEGEND-2 357/741LentiCiltacabtagene Autoleucel
LCAR-B38M (JNJ68284528)
4-1BBCy0.5 × 106 cells / kg37089746819.936.1
CARTITUDE-1 4,5971b/2LentiCiltacabtagene Autoleucel
LCAR-B38M (JNJ68284528)
4-1BBFlu/Cy0.75 × 106 cells / kg6410976793Not reachedNA
EVOLVE 6441LentiOrvacabtagene autocel (JCARH125) 4-1BBFlu/Cy50/150/450 × 106 cells797822767NANA
EVOLVE 7621LentiOrvacabtagene autocel (JCARH125) 4-1BBFlu/Cy300/450/600 × 106 cells633923596NANA
NCI 8161RetroNACD28Flu/Cy9 × 106 cells/kg93819816310031 wksNA
UPENN 9251LentiNA4-1BBNone or Cy10/50/100/500 × 106 cells7321263283365-125 d502 d
CT053 10241RetroCT0534-1BBFlu/Cy150 × 106 cells4.504888385NANA
Dual CD19-BCMA 11101LentiSequential CART-CD19/CART-BCMACD28Flu/CyCD19: 1 x107 cells BCMA: 3/5/6.5 ×107 cells41009040305NA
FHVH-BCMA-T 12211RetroFHVH-BCMA-T4-1BBFlu/Cy0.75/1.5/3/6/12 × 106 cells6191090NANANANA
Adapted from Wudhikarn ASH 2020: 1. Raje NEJM 2019; 2. Munshi 2020; 3 Berdeja Blood 2019; 34 Wang Blood 2019, 134 (supl 1): 579; 4. Maduri Blood 2019, 134 (supl 1): 577; 5 Berdeja JCO 2020, 38 (supl 15): 8505; 6. Stadtmauer Science 2020; 7. Mailankody JCO 2020, 38 (supl 15): 8504; 8. Brudno JCO 2018; 9. Cohen JCO 2019; 10. Jie Blood 2019, 134 (supl 1): 4435; 11. Yan Cancer Medicine 2021. 12. Mikkilineni Blood 2020, 136 (supl 1): 50–51.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Reguera-Ortega, J.L.; García-Guerrero, E.; Pérez-Simón, J.A. Current Status of CAR-T Cell Therapy in Multiple Myeloma. Hemato 2021, 2, 660-671. https://0-doi-org.brum.beds.ac.uk/10.3390/hemato2040043

AMA Style

Reguera-Ortega JL, García-Guerrero E, Pérez-Simón JA. Current Status of CAR-T Cell Therapy in Multiple Myeloma. Hemato. 2021; 2(4):660-671. https://0-doi-org.brum.beds.ac.uk/10.3390/hemato2040043

Chicago/Turabian Style

Reguera-Ortega, Juan Luis, Estefanía García-Guerrero, and Jose Antonio Pérez-Simón. 2021. "Current Status of CAR-T Cell Therapy in Multiple Myeloma" Hemato 2, no. 4: 660-671. https://0-doi-org.brum.beds.ac.uk/10.3390/hemato2040043

Article Metrics

Back to TopTop