The mechanism of CAR-T cell therapy involves the binding of the CAR molecule on the surface of CAR-T cells to the corresponding antigen on the surface of tumor cells, which subsequently activates the intracellular signaling pathways in CAR-T cells, leading to the cytotoxic elimination of tumor cells. In the treatment of R/R MM, several target antigens have been identified for CAR-T cell therapy. B-cell maturation antigen (BCMA) is considered as one of the most promising targets due to its expression on mature B cells, normal and malignant plasma cells, and plasmacytoid dendritic cells, while being absent on early B cells, memory B cells, and normal hematopoietic stem cells (HSCs), which makes BCMA an ideal antigen of targeting. Additionally, G protein-coupled receptor class C group 5 member D (GPRC5D) is frequently utilized as a target antigen in MM CAR-T cell therapy that is highly expressed on primary MM cells but is restricted to the immune-privileged hair follicle regions in normal tissues. Notably, GPRC5D expression is independent of BCMA, allowing for the development of single or dual-targeted cellular therapies [
17]. Other antigens, such as CD138, CD38, and SLAMF7, are also being evaluated in ongoing clinical studies [
18]. As to June 2024, regulatory agencies in major global pharmaceutical markets have approved four CAR-T products for the treatment of R/R MM: ide-cel, cilta-cel, equecabtagene autoleucel (equ-cel), and zevorcabtagene autoleucel (zevor-cel). Additionally, numerous other candidate CAR-T therapies are undergoing preclinical and clinical evaluation.
2.1. Current Landscape of CAR-T Cell Therapy towards Multiple Myeloma
While CAR-T cell therapy has achieved remarkable success in treating R/R MM, the challenge of overcoming therapy failure remains a significant clinical hurdle. The 5-year follow-up data from the LEGEND-2 study on cilta-cel [
19] reported an overall response rate (ORR) of 87.8%, with a 5-year progression-free survival (PFS) rate of 21.0%, and the longest duration of response extending up to 6.4 years. Meanwhile, in the phase I CRB-401 trial of ide-cel, with a median follow-up of 18.1 months, ORR reached 75.8%, with a median duration of response of 10.3 months and a median PFS of 8.8 months [
20]. Similarly, the FUMANBA-1 study of equecabtagene autoleucel (equ-cel) [
21], with a median follow-up of 13.8 months, demonstrated an ORR of 96%, although the median duration of response and median PFS have yet to be determined; the 12-month PFS rate was 78.8% (95% CI: 68.6-85.97). Additionally, the 3-year follow-up from the LUMMICAR-1 study of zevorcabtagene autoleucel (zevor-cel) [
22] reported an ORR of 100%, with a median duration of response of 24.1 months and a median PFS of 25 months . Details on these outcomes of CAR-T cell therapies for R/R MM are provided in
Table 1. Although these results can be regarded as promising, it is also important to acknowledge that there is a general tendency of approximately 10-20% of the patient exhibit resistance to CAR-T therapy, and many patients who demonstrated a response might relapse. Research has identified several factors associated with less desirable PFS in the context of CAR-T therapy failure, including a history of extramedullary disease, prior BCMA-targeted therapy, elevated ferritin levels during lymphodepletion, plasma cell leukemia, and the presence of the t(4;14) translocation [
23].
2.2. Potential Factors Contributing to CAR-T Therapy Failure
The mechanisms underlying the failure of CAR-T therapy in treating R/R MM are not yet fully elucidated, with various factors potentially contributing to treatment failure. These factors include the downregulation or loss of tumor cell target antigen expression, T-cell exhaustion, and the tumor immune microenvironment. Baseline heterozygous deletion of BCMA has been reported as a possible contributor to BCMA loss following immunotherapy, which could serve as a potential mechanism for immune escape [
29]. In patients who received commercial anti-BCMA CAR-T cell therapy, the median BCMA expression decreased from 670 (interquartile range [IQR] 380-850) mol/cell prior to treatment to 390 (IQR 290-490) mol/cell post-treatment (n=15, p=0.011).
In vitro studies have shown that as antigen levels decline, the tumor-lytic and cytokine production functions of CAR-T cells are compromised, and when antigen levels fall below 50 mol/cell, CAR-T cell functionality is lost [
30]. Beyond BCMA, a reduction or loss of GPRC5D expression has also been observed in patients who experienced disease progression after achieving remission with anti-GPRC5D CAR-T therapy [
31,
32].
The expansion and persistence of CAR-T cells are closely linked to the efficacy of CAR-T therapy. Studies indicate that circulating CAR-T cell expansion exceeding 180/mm
3 following ide-cel infusion is associated with prolonged PFS [
33]. In patients with sustained best responses after cilta-cel infusion, CAR-T cell persistence was notably longer (median duration of 535.3 days to 261.6 days, p=0.01) [
19]. However, the human anti-mouse antibody (HAMA) response, induced by murine-derived scFv, has been shown to diminish CAR-T cell persistence [
34]. The tumor microenvironment in MM further influences T-cell functionality, with CD8+ T cells expressing several molecules associated with T-cell exhaustion (PD-1, CTLA-4, 2B4, CD160) and T-cell senescence (CD57, loss of CD28), a phenotype correlated with reduced cell proliferation and impaired function [
35]. Moreover, high CAR expression levels are linked to increased signaling and an exhaustion phenotype, characterized by the expression of multiple inhibitory receptors, including PD-1, CTLA-4, LAG3, TIM3, and TIGIT, which is associated with diminished responses and poorer long-term PFS outcomes [
36].
2.4. Recommended Strategies
The approval and commercialization of Ide-cel have ushered in a new era of immune cell therapy towards R/R MM, with the subsequent approval of Cilta-cel for second-line treatment further emphasizing the pivotal role of CAR-T cell therapy in this context. However, this advancement brings forth critical research challenges, such as the need to extend progression-free survival (PFS) and overall survival (OS) associated with CAR-T therapy, as well as determining the most effective salvage treatment strategies following disease relapse or progression. Based on the findings from relevant clinical studies, we propose the following recommendations.
2.4.1. moving CAR-T therapy to earlier lines of treatment
The potential therapeutic mechanism of CAR-T cell therapy in MM might include
a) At the time of disease onset or with fewer treatment lines, the patient's autologous T cells have not been significantly weakened by other therapies, hence the quality of the prepared CAR-T cells is higher, and the efficacy as an early-line treatment is also better [
44,59];
b) The invasiveness of MM in the early stages of the disease is lower, with slow disease progression or low tumor burden, therefore, the use of CAR-T cell therapy can yield more benefits [60]. Studies have shown that the T cell population at the time of MM diagnosis has a higher proportion of naive T cells compared to T cells after treatment with daratumumab or after relapse, with lower proportions of exhaustion markers PD1 and LAG3, and the prepared CAR-T cells exhibit better cell proliferation and cytotoxic activity [61] Patients with fewer ( < 4) prior treatment lines before receiving anti-BCMA CAR-T cell therapy have a lower proportion of extramedullary disease (EMD), triple-drug refractory, and penta-drug refractory, and their response to subsequent salvage therapy is significantly better than that of patients with more ( ≥ 4) prior treatment lines [
39]. In a phase 1, single-arm, multicenter study of ide-cel for the treatment of R/R MM, the number of prior treatment lines was 6 (range, 3-18), with an overall response rate (ORR) of 75.8%, a median progression-free survival (PFS) of 8.8 months, and a median overall survival (OS) of 34.2 months [
20]. In a comparative study between ide-cel and standard therapy, the number of prior treatment lines in the ide-cel group was 3 (range, 2-4), with an ORR of 71%, a median PFS of 13.3 months, and the median OS has not been reached, while the risk of disease progression or death was reduced by 51% [62]. Further analysis indicates that patients with PFS ≥18 months had more primitive and less exhausted T cells at the time of apheresis, with a better functional T cell phenotype in the CAR-T cell preparation [
20]. Therefore, the results of related studies support the advancement of CAR-T cell therapy line numbers, which can bring greater benefits to patients [63].
2.4.2. Exploring CAR-T Cells with Different Targets and Origins
For R/R MM patients after CAR-T therapy, it is recommended to initially assess the expression of antigens such as BCMA and GPRC5D on the surface of MM cells and select CAR-T cell therapy targeting the corresponding antigens. However, some studies have indicated that the response rate is not related to baseline BCMA expression [
4,64]. Currently, CAR-T cells targeting BCMA have been approved for marketing, while those targeting other antigens such as GPRC5D are undergoing clinical trials. Treatments based on anti-GPRC5D CAR-T cells [
31,
32,
42] , anti-BCMA CAR-T [
39] or dual-target CAR-T cells [
43] have been shown to be effective for patients who have failed previous anti-BCMA CAR-T cell therapy, and we recommend using CAR-T cells with different targets from those previously used as thefirst line of therapy. Additionally, considering that different sources and constructs of scFv can affect the efficacy of CAR-T cell therapy [64,65],it is also possible to choose humanized or fully human CAR-T as salvage therapy following the progression of CAR-T treatment.
2.4.3. Rapid-Manufactured CAR-T
During the conventional autologous CAR-T cell manufacturing process, patients with a high tumor burden often require bridging therapy to manage disease progression. However, the rapid advancement of the disease in some patients can preclude the administration of CAR-T cell therapy, potentially leading to mortality before the therapy can be administered [66]. Moreover, bridging therapy can also affect subsequent efficacy, such as longer median hospital stay, shorter median progression-free survival (PFS) (8.1 months to 11.5 months, p = 0.03), and shorter median overall survival (OS) (13.8 months to not reached, p = 0.002) [67]. Rapid CAR-T cell manufacturing processes, by shortening production time, yield T cells with lower differentiation, requiring fewer cell doses, reducing production costs, and enhancing in vivo expansion of CAR-T cells, thereby potentially offering significant advantages in the treatment of R/R MM. In addition, the advantages of rapid manufacturing also include reducing bridging therapy and mitigating adverse events following bridging therapy. Therefore, rapidly manufactured CAR-T cells are a very promising type of CAR-T cell. The results of a phase I clinical study conducted by CARsgen Therapeutics, targeting GPRC5D with rapidly manufactured CAR-T cells (CT071) for the treatment of R/R MM, were presented at the EHA Annual Meeting (Poster P941). Ten R/R MM patients were treated with CT071, achieving an ORR of 90%. Among them, two patients who had previously received BCMA/CD19 dual-target CAR-T cell therapy achieved sCR and PR, respectively, with MRD testing negative at a 10
-6 threshold. Novartis
TM has developed an anti-BCMA CAR-T cell product, PHE885, using its T-charge platform for rapid manufacturing (less than 2 days), which achieved an ORR of 98% in R/R MM patients who had undergone a median of four prior lines of therapy [68]. The incidence of CRS was 96%, with 11% experiencing grade 3 CRS, while ICANS occurred in 22%, with 7% experiencing grade 3 ICANS. Additionally, other companies such as Gracell Biotechnologies [
28], BMS [69], and IASO Bio [70] are also developing rapidly manufactured CAR-T cells for the treatment of R/R MM, featuring a desirable landscape of the application of rapid-manufactured CAR-T towards the failure of post CAR-T therapies.
2.4.4. Sequential Selection of T-Cell Redirecting Therapies
T-cell redirecting therapies currently encompass CAR-T cells, ADCs, and BsAbs, with CAR-T cells and BsAbs being the most frequently utilized salvage treatments after CAR-T therapy failure in R/R MM [
16]. Both CAR-T cells and BsAbs are forms of immunotherapy that mediate non-MHC-restricted cytotoxicity; however, BsAbs depend on the patient’s own T cells, which raises concerns about potential T-cell dysfunction [71]. Due to differences in toxicity profiles, expected response rates, and administration methods, CAR-T cell therapy is generally more suitable for younger patients, those with aggressive or relapsed disease, and those with a high tumor burden [72].
At present, there is no established consensus on the optimal sequencing of CAR-T cells, ADCs, and BsAbs. Research indicates that the effectiveness of CAR-T therapy is somewhat diminished when administered following ADC or BsAb treatment. In a real-world study of Ide-cel treatment in R/R MM patients who were ineligible for the KarMMa-1 trial, those with prior BCMA-targeted therapy (16 treated with ADCs, 1 with a trispecific T-cell activator, and 1 with an experimental CAR-T therapy) exhibited a lower median PFS compared to patients who had not received BCMA-targeted therapy (6.2 months to 9.0 months, p = 0.11) [73]. Similarly, in the phase II CARTITUDE-2 study evaluating Cilta-cel in R/R MM, cohort C focused on patients previously treated with BCMA-targeted therapies (13 with belantamab mafodotin, 7 with BsAbs, including 1 who had received both in the belantamab mafodotin group) [74]. The results showed an ORR of 60.0% and a median PFS of only 9.1 months, significantly lower than the outcomes in BCMA-targeted therapy-naive patients in the CARTITUDE-1 study. In contrast, BsAb therapy following CAR-T cell treatment has demonstrated promising efficacy. Jakubowiak et al. [
12] examined the safety and effectiveness of talquetamab in 70 R/R MM patients who had undergone prior T-cell redirecting therapies, including 50 treated with CAR-T, 25 with BsAbs, and 5 with both CAR-T and BsAbs. The ORR was 72.9% in patients who had received prior BCMA CAR-T therapy, compared to 56.5% in those with prior BsAb therapy. Further analysis indicated that whether CAR-T was used as the last or a previous line of therapy had no significant impact on BsAb efficacy (ORR, 71.4% to 75.9%), whereas BsAb efficacy was significantly reduced when BsAbs were used as the last line of therapy comparing to the previous line (ORR, 28.6% to 66.7%). Given that CAR-T cell therapy can induce profound and durable responses with extended treatment-free intervals, thereby reducing the risk of T-cell exhaustion and the selective pressure from continuous therapy [
16],it is recommended that CAR-T cell therapy be prioritized in the treatment course of MM, with BsAb therapy considered upon disease progression after CAR-T treatment.
2.4.5. Combination Therapy
Combining CAR-T therapy with other drugs can enhance the persistence of CAR-T cells in vivo, improve BCMA expression, and reduce T-cell exhaustion. Such drugs include immunomodulatory agents, γ-secretase inhibitors, and CRBN E3 ligase modulators.
Lenalidomide is a commonly used immunomodulatory drug (IMiD) that was approved by the FDA for the treatment of MM in June 2006. Preclinical studies have shown that lenalidomide can provide a co-stimulatory effect on CS1 CAR-T cells, enhancing their antitumor activity and persistence by improving CS1 CAR-T cell cytotoxicity, memory maintenance, Th1 cytokine production, and immunological synapse formation [75]. Lenalidomide can regulate the ICOSL/ICOS pathway, Th1/Th2 pathway, increase CAR-T cell secretion of cytokines such as IFN-γ, IL-2, and TNF-α, and enhance CAR-T cell cytotoxicity [76]. Researchers reported a case of an R/R MM patient who relapsed after multiple chemotherapies, autologous hematopoietic stem cell transplantation, murine CAR-T cell therapy, and human CAR-T cell therapy. After lymphodepletion with fludarabine and cyclophosphamide, the patient was given lenalidomide on day -1, followed by human CAR-T cell infusion on day 0, achieving VGPR within 14 days and maintaining it for over 8 months [77].
BCMA is cleaved from the surface of MM cells by γ-secretase. Preclinical studies have indicated that inhibiting γ-secretase can increase BCMA expression on MM cells, thereby enhancing the efficacy of CAR-T cell therapy [78]. A phase I clinical study evaluated the safety of crenigacestat, a γ-secretase inhibitor, in combination with anti-BCMA CAR-T cells for the treatment of R/R MM patients. The results showed that the γ-secretase inhibitor could block BCMA shedding, overcoming antigen downregulation and thereby improving CAR-T cell efficacy [79].
In vitro studies on the CRBN E3 ligase modulator iberdomide have demonstrated that iberdomide can significantly reduce the transcription factors Ikaros and Aiolos in CAR-T cells, promote the persistence and proliferation of IL-2-starved cells, significantly increase the expression of Ki67 in CAR-T cells, enhance cell viability, upregulate activation markers such as HLADR and CD69, downregulate TIGIT expression, and increase the expression of IL-2, IL-17a, and TNF-α. Therefore, iberdomide shows promise in assisting CAR-T cell therapy by overcoming CAR-T cell resistance and enhancing its efficacy [80].
Based on these findings, combining CAR-T cell therapy with related drugs may improve the efficacy of CAR-T treatment from multiple aspects, potentially extending PFS and OS in patients.
2.4.6. Maintenance Therapy
A survey conducted by the International Myeloma Society revealed that 30% of the clinicians surveyed administer maintenance therapy after CAR-T cell treatment, primarily to sustain the level of remission achieved or to deepen the response further [81]. However, a survey by the American Society for Transplantation and Cellular Therapy (ASTCT) Practice Guidelines Committee found that 86% of clinicians would not consider maintenance therapy in the absence of biochemical or clinical evidence of myeloma, with only 18% considering it for patients who achieved a partial response (PR) after CAR-T cell therapy but did not convert to a complete response (CR) after several months of follow-up [82]. Therefore, it is recommended to identify risk factors before initiating CAR-T cell therapy, assess which patients might benefit from maintenance therapy, and determine the appropriate regimens. Continuous monitoring of disease status post-CAR-T cell therapy is crucial, with maintenance therapy being initiated at the optimal time to prolong progression-free survival (PFS) and overall survival (OS).
In a study by Li et al.[
27], the use of bispecific CS1-BCMA CAR-T cells in treating R/R MM patients resulted in an overall response rate (ORR) of 81% (13/16). Within the study, three patients (Patients 10, 11, and 15) received lenalidomide as maintenance therapy (10 mg/day, 21 days of a 28-day cycle) following CAR-T cell treatment. Patients 10 and 11 achieved deeper responses; however, Patients 10 and 15 eventually experienced relapse or progression, while Patient 11 continued to maintain a stringent complete response (sCR). Another case involved an R/R MM patient who developed secondary plasma cell leukemia and was treated with anti-BCMA CAR-T cells, achieving a sustained sCR for 9 months. This patient then received intermittent venetoclax treatment at 10 mg/day, maintaining the response for an additional 7 months [83]. In the phase II CARTITUDE-2 clinical study [84] evaluating Cilta-cel for newly diagnosed MM patients with suboptimal response to first-line autologous stem cell transplantation, 17 patients were assessed. Among all the patients, five received Cilta-cel alone, while 12 began continuous lenalidomide maintenance therapy (for up to 2 years) 21 days after Cilta-cel infusion. The results showed a general ORR of 94%, with both the 18-month PFS rate (as assessed by investigators) and OS rate at 94%, suggesting that a single infusion of Cilta-cel combined with lenalidomide maintenance therapy can produce a durable, deep response. These findings indicate that incorporating lenalidomide as maintenance therapy after CAR-T cell treatment may be an effective strategy to address CAR-T cell therapy failure.