CJ, RC and SF are joint first authors.
WHAT IS ALREADY KNOWN ON THIS TOPIC
B-cell maturation antigen (BCMA) chimeric antigen receptor (CAR)-T cell therapy has shown promise in treating relapsed or refractory multiple myeloma (R/RMM), but data on its long-term efficacy and safety in larger patient cohorts remain insufficient.
WHAT THIS STUDY ADDS
Prior autologous stem cell transplantation (ASCT) and extramedullary disease are adverse prognostic factors for patients with R/RMM receiving BCMA CAR-T therapy. Patients with a history of ASCT show limited peak CAR-T cell expansion.
HOW THIS STUDY MIGHT AFFECT RESEARCH, PRACTICE OR POLICY
For patients at higher risk of early relapse after ASCT, lymphocyte collection prior to ASCT is recommended to achieve more durable responses with subsequent autologous BCMA CAR-T therapy.
Introduction
Multiple myeloma (MM) is a malignant plasma cell tumor that primarily occurs in elderly individuals and is characterized by bone destruction and extramedullary involvement.1–3 Advances in treatment, including the use of immunomodulators, proteasome inhibitors, and CD38-targeting monoclonal antibodies, along with various drug combinations, have markedly improved the prognosis.4–7 For eligible patients, autologous stem cell transplantation (ASCT) following high-dose chemotherapy remains the standard consolidation therapy, effectively reducing relapse rates.8–10 Nevertheless, despite these advancements, most patients eventually experience relapsed or refractory disease, where treatment options become increasingly limited. In recent years, chimeric antigen receptor (CAR) T-cell therapy targeting B-cell maturation antigen (BCMA) has emerged as a promising therapeutic approach.11 In patients with relapsed or refractory multiple myeloma (R/RMM), BCMA CAR-T cell therapy has shown remarkable efficacy, with overall response rates (ORR) ranging from 81% to 97.9% and complete remission (CR) rates between 33% and 82.5%,12–18 confirming its significant potential in the treatment of R/RMM. With the growing application of BCMA CAR-T therapy, there is growing interest in understanding its long-term efficacy and safety in larger patient cohorts.
Relapse or disease progression was observed in approximately 20–52% of patients during a median follow-up period of 8–27.7 months.13 17 19 20 In previous studies, a deeper response was closely associated with improved progression-free survival (PFS) and overall survival (OS).21 A better understanding of the factors associated with CR rates and a durable response is crucial for improving treatment outcomes. Published data suggest that adequate CAR-T cell expansion is a fundamental prerequisite for achieving CAR-T cell efficacy.12 13 15 Sufficient CAR-T cell expansion not only exerts direct antitumor effects but also enhances endogenous T cell and natural killer cell activity, thereby contributing to tumor destruction.22 Insights from prior studies indicate that the characteristics of apheresis T cells may play an integral role, with the presence of specific T-cell subtypes, such as a high proportion of early memory cells, being associated with greater peak expansion.23 24 However, these studies were limited by small sample sizes and did not provide further analysis of infused CAR-T cells. A comprehensive evaluation of product quality, along with a well-defined strategy for optimizing treatment regimens, remains essential to advancing therapeutic efficacy.
This study comprehensively evaluated the long-term efficacy and safety of BCMA CAR-T therapy in a large cohort of patients with R/RMM. It further investigated the influence of extramedullary disease and prior ASCT on the outcomes of subsequent BCMA CAR-T therapy. Besides, the post-infusion dynamics and immunophenotypic characteristics of the administered CAR-T cells were analyzed to elucidate potential mechanisms underlying the poor prognosis observed in specific subgroups.
Materials and methods
CAR-T cell production
BCMA CAR-T cells were generated from autologous T cells obtained through leukapheresis. A lentiviral vector was employed to introduce a second-generation BCMA-targeted CAR, incorporating 4-1BB as a costimulatory domain and CD3ζ as the signaling domain (online supplemental figure S1).25
Patients
Two clinical trials were included in this study (ChiCTR1800017404 and NCT05430945), which were conducted across different time periods. Both trials employed the same BCMA CAR-T cell product and followed identical treatment protocols. Patients underwent lymphodepletion chemotherapy consisting of cyclophosphamide (500 mg/m2 administered on days −3 to −2) and fludarabine (30 mg/m2 administered on days −4 to −2), followed by the infusion of autologous BCMA CAR-T cells on day 0. All patients provided written informed consent, and all procedures adhered to the principles of the Declaration of Helsinki and its subsequent amendments.
CAR-T cell immunophenotypes and cellular kinetics
Immunophenotype analysis of the CAR-T cell products was conducted using the CytoFLEX flow cytometer (Beckman, California, USA) following the completion of the CAR-T cell manufacturing process. The analysis employed the BCMA CAR Detection Reagent Anti-(G4S) n (B02H1) mAb FITC (GS-ARFT100, Hycells) in combination with the following antibodies: CD3-APC (555335, BD), CD45RA-BV421 (562885, BD), CD62L-BV650 (563808, BD), CD8-PE-CF594 (562311, BD), and CD4-APC-CY7 (557871, BD).
The dynamic changes in the cellular kinetics of CAR-T cells post-infusion were routinely monitored using the following antibody combinations: Anti-(G4S)n (B02H1) mAb FITC (GS-ARFT100, Hycells), CD45-APC (368512, BioLegend), CD3-PECY7 (980010, BioLegend), CD4-PC5.5 (317428, BioLegend) and CD8-PE (300907, BioLegend). All data analyses were performed using FlowJo software (V.10, TreeStar, USA). Due to the low sensitivity of flow cytometry in tracking low quantities of CAR-T cells, quantitative PCR was used as a complementary method to monitor CAR transgene levels at 6 and 12 months post-CAR-T cell infusion.
Evaluation of long-term outcomes
Toxicity was classified based on the Common Terminology Criteria for Adverse Events V.5.0. Cytokine release syndrome (CRS) was evaluated using the modified criteria proposed by Lee et al.26 Disease responses were evaluated based on the criteria established by the International Myeloma Working Group.27 Responses to BCMA CAR-T cell therapy were categorized as CR, very good partial response (VGPR), or partial response (PR). Non-responses included progressive disease (PD), stable disease (SD), and minimal response (MR). The ORR was calculated as the sum of CR, VGPR, and PR. PFS was defined as the time from CAR-T cell infusion to the first occurrence of disease progression or death from any cause. OS was defined as the time from infusion until the patient’s death. For patients who attained CR, the cumulative incidence of relapse (CIR) was calculated from the initiation of CR to the point of relapse. Minimal residual disease (MRD) was evaluated using flow cytometry with a sensitivity threshold of 10−⁴.
Statistical analysis
Descriptive statistics encompassed medians with ranges for continuous variables and counts with corresponding percentages for categorical variables. Categorical variables were compared using the χ2 test or Fisher’s exact test, as appropriate, while continuous variables were analyzed using the Mann-Whitney U test. The Kaplan-Meier method was used to estimate PFS and OS. Multivariable Cox regression analyses were conducted to identify risk factors associated with PFS and OS. Besides, multivariable logistic regression analyses were employed to examine factors influencing CR rates. A multivariable competing risk analysis was performed to assess the CIR among patients who achieved CR. All p values were two-sided, with statistical significance set at p<0.05. Statistical analyses were conducted using SPSS V.26.0 (SPSS, Chicago, Illinois, USA) and R V.4.2.0 (R Core Team, Vienna, Austria).
Results
Baseline characteristics of patients between July 30, 2018, and September 27, 2023, a total of 141 patients with R/RMM who underwent BCMA CAR-T therapy were enrolled in the study (figure 1). As of the data cut-off date (June 5, 2024), the median follow-up duration was 20.2 months (range: 0.3–70.1 months). The baseline characteristics of the patients are summarized in table 1. The median age at the time of CAR-T cell infusion was 60 years (range: 16–79) years. Among the enrolled patients, 107 (75.9%) exhibited bone marrow (BM) plasma cell infiltration at baseline, with 27 patients (19.1%) presenting a high tumor burden (≥50% BM plasma cells). Moreover, 65 patients (46.1%) had extramedullary lesions, 122 (86.5%) were classified as having Durie-Salmon stage 3 disease, and 60 (42.6%) were categorized as International Staging System stage 3. High-risk cytogenetic abnormalities in the relapsed/refractory state were observed in 90 patients (63.8%). Regarding prior treatments, 114 patients (80.9%) had received two to four prior lines of therapy, 111 (78.7%) had been treated with a combination of proteasome inhibitors and immunomodulatory agents, 48 (34.0%) had received daratumumab, and 56 (39.7%) had undergone ASCT before CAR-T cell infusion.
Table 1The baseline characteristics of patients with R/RMM with BCMA CAR-T therapy
Characteristics | Total (N=141) |
Age | |
60 (16–79) | |
Gender (n, %) | |
80 (56.7) | |
Monoclonal type (n, %) | |
39 (27.7) | |
12 (8.5) | |
62 (44.0) | |
5 (3.5) | |
22 (15.6) | |
1 (0.7) | |
Bone marrow plasma cells (n, %) | |
27 (19.1) | |
Extramedullary disease (n, %) | 65 (46.1) |
DS stage (n, %) | |
5 (3.5) | |
14 (9.9) | |
122 (86.5) | |
ISS stage, (n, %) | |
40 (28.4) | |
41 (29.1) | |
60 (42.6) | |
Cytogenetics risk* (n, %) | |
90 (63.8) | |
27 (19.1) | |
24 (17.0) | |
Prior therapy lines (n, %) | |
114 (80.9) | |
22 (15.6) | |
5 (3.5) | |
Prior therapies, n (%) | |
Proteasome inhibitors | |
141 (100.0) | |
50 (35.5) | |
30 (21.3) | |
Immunomodulatory agents | |
56 (39.7) | |
123 (87.2) | |
49 (34.8) | |
Prior proteasome inhibitors+immunomodulatory agents | 111 (78.7) |
Monoclonal antibodies | |
48 (34.0) | |
Chemotherapy drugs exposure† (n, %) | 129 (91.5) |
Previous ASCT (n, %) | 56 (39.7) |
Diagnosis to CAR-T time (n, %) | |
68 (48.2) | |
73 (51.8) | |
Bridging therapy‡ (n, %) | 20 (14.2) |
*High-risk cytogenetics were defined as del(17p), t(4;14), t(14;16), t(14;20) or 1q21 gain or amplification by fluorescence in situ hybridization.
†Chemotherapy drugs including Cyclophosphamide, Etoposide, Epirubicin, cisplatin, Vindesine, Gemcitabine, Oxaliplatin and other similar medications.
‡Bridging therapy was administered after leukapheresis and before lymphodepletion.
ASCT, autologous stem cell transplantation; BCMA, B-cell maturation antigen ; CAR-T, chimeric antigen receptor T-cell therapy; DS, Durie-Salmon; Ig, immunoglobulin; ISS, International Staging System; R/RMM, relapsed or refractory multiple myeloma.
A total of 20 patients (14.2%) underwent bridging therapy between apheresis and lymphodepletion, following a comprehensive assessment of tumor burden and physical condition. The most frequently used agents were dexamethasone, pomalidomide, and cyclophosphamide (online supplemental table S1). Among these patients, the clinical outcomes were as follows: PD (N=1, 5%), SD (N=5, 25%), MR (N=9, 45%), and PR (N=5, 25%).
Long-term efficacy
Seven patients experienced early mortality within the first month following the infusion of BCMA CAR-T cells. In the overall cohort, the ORR was 90.1% (127/141), with a CR rate of 48.2% (68/141). Among the 134 evaluable patients, the ORR was 94.8% (127/134), with 50.7% (68/134) achieving CR. Moreover, 16.4% (22/134) achieved VGPR, and 27.6% (37/134) reached PR (figure 2F). The median PFS was 15.2 months (95% CI, 7.6 to 22.8) (figure 2A). The 4-year PFS and OS rates were 37.4% (95% CI, 29.1% to 48.1%) and 63.2% (95% CI, 54.8% to 72.8%), respectively (figure 2A, B). Notably, 13 of the 60 patients with ongoing responses had responses lasting over 4 years, with the longest remission duration being 5.6 years. A total of 81 patients experienced PD and/or death. Of these, 32 PD patients had ongoing survival after receiving subsequent treatments. There were 49 deaths, primarily due to disease progression (N=38). Three patients died from intracerebral hemorrhage caused by tumor lysis and severe CRS, which led to coagulopathy and thrombocytopenia. Two of these patients had pre-existing grade 3 or higher thrombocytopenia prior to lymphodepletion. Seven patients died due to infections, including two from COVID-19. Moreover, one patient succumbed to liver failure resulting from hepatitis B reactivation.
Figure 2. PFS (2A), OS (2B) for all 141 patients, and response rates (2F) for 134 evaluable patients. PFS (2C) and OS (2D) compared between CR and non-CR patients, and CIR among CR patients (2E) in patients with R/RMM receiving BCMA CAR-T therapy. CIR, cumulative incidence of relapse; CR, complete remission; NR, no response; OS, overall survival; PFS, progression-free survival; R/RMM, relapsed/refractory multiple myeloma; VGPR, very good partial response.
Among the 68 patients who achieved CR, 66 (97.1%) attained MRD negativity at the 10−4 threshold. The survival outcomes for CR patients were notably improved, with a median PFS of 42.37 months (range: 18.5 months to not reached), compared with 6.03 months (range: 3.2–13.27 months) in non-CR patients (p<0.001) (figure 2C). This significant benefit was also observed in OS (p=0.013), where the median OS was not reached (figure 2D). Among the CR patients, 33 (48.5%) experienced disease progression, with a 4-year CIR of 54.5% (95% CI, 40.0% to 69.1%) (figure 2E).
Safety
CRS occurred in 128 patients (90.8%), with 46 patients (32.6%) experiencing grade 3 CRS and 5 patients (3.5%) experiencing grade 4. Immune effector cell-associated neurotoxicity syndrome (ICANS) was reported in 11 patients (7.8%), all of whom had grade 1 or 2. The use of steroids, tocilizumab, or both for the management of CRS and ICANS was observed in 57 patients (40.4%) and 52 patients (36.9%), respectively (table 2).
Table 2Main toxicities and management approach
Toxicity | Total (N=141) |
CRS grade, n (%) | |
13 (9.2) | |
27 (19.1) | |
50 (35.5) | |
46 (32.6) | |
5 (3.5) | |
ICANS grade, n (%) | |
130 (92.2) | |
10 (7.1) | |
1 (0.7) | |
0 (0) | |
0 (0) | |
Treatment for CRS/ICANS, n (%) | |
57 (40.4) | |
52 (36.9) | |
Hematologic toxicity up to 1 month after infusion, n (%) | Total (N=134) |
Anemia | |
74 (55.2) | |
53 (39.6) | |
Thrombocytopenia | |
87 (64.9) | |
70 (52.2) | |
Neutropenia | |
92 (68.7) | |
58 (43.3) | |
Supportive therapy for cytopenia, n (%) | |
66 (49.3) | |
22 (16.4) | |
2 (1.5) | |
31 (23.1) | |
36 (26.9) |
pRBC packed red blood cells. EPO, erythropoietin; G-CSF, granulocyte colony-stimulating factor; ICANS, immune effector cell-associated neurotoxicity syndrome; TPO, thrombopoietin analogus.
Regarding the prolonged cytopenias observed 1 month after infusion, grade ≥3 anemia persisted in 53 patients (39.6%), while 58 patients (43.3%) experienced grade ≥3 neutropenia, and 70 patients (52.2%) had grade ≥3 thrombocytopenia. Moreover, 66 patients (49.3%) continued to receive granulocyte colony-stimulating factor, 31 patients (23.1%) required packed red blood cell transfusions, and 36 patients (26.9%) received pooled platelet transfusions more than 1 month after BCMA CAR-T cell infusion (table 2).
Secondary primary malignancies were reported in one case. The patient was diagnosed with acute myeloid leukemia 5.7 months after BCMA CAR-T cell infusion and subsequently received chemotherapy. The patient remains alive at the time of reporting.
Predictive factors associated with CR, CIR, PFS and OS
Multivariable logistic regression analyses revealed that prior ASCT was associated with a lower CR rate (relative risk (RR), 0.320 (95% CI, 0.132 to 0.778); p=0.012) (figure 3A). To further explore the risk factors for relapse among CR patients, multivariable competing risk models indicated that patients with extramedullary disease (RR, 2.191 (95% CI, 1.027 to 4.678); p=0.043) and those with prior ASCT (RR, 2.903 (95% CI, 1.120 to 7.524); p=0.028) had a higher CIR (figure 3A).
Figure 3. Forest plot depicting the results of characteristics in patients with R/RMM receiving BCMA CAR-T therapy associated with CR rate (N=134), CIR among CR patients (N=68) (3A), PFS (N=141) and OS (N=141) (3B). ASCT, autologous stem cell transplantation; CAR-T, chimeric antigen receptor T cell therapy; CD38 Ab exposure, CD38 monoclonal antibodies exposure; chemo exposure, chemotherapy drugs exposure; CIR, cumulative incidence of relapse; CR, complete remission; DS, Durie-Salmon; ISS, International Staging System; PFS, progression-free survival; OS, overall survival; PIs+IMIDs, proteasome inhibitors combined with immunomodulatory agents; R/RMM, relapsed/refractory multiple myeloma.
In the multivariable Cox regression analysis, extramedullary disease (HR, 2.161 (95% CI, 1.340 to 3.484); p=0.002) and prior ASCT (HR, 2.375 (95% CI, 1.359 to 4.152); p=0.002) were identified as independent risk factors for PFS (figure 3B). Only extramedullary disease (HR, 1.887 (95% CI, 1.003 to 3.552); p=0.049) was significantly associated with shortened OS (figure 3B). At disease progression, 33.3% (47 out of 141) of patients developed extramedullary lesions, including 42 patients who had pre-existing extramedullary disease at baseline.
The peak expansion of CAR-T cells correlated with the response
The infused CAR-T cell dose in the overall cohort was 2.36×10⁶/kg (range: 0.58–7.1×10⁶/kg), with a CD4/CD8 ratio of 3.06 (range: 0.09–56.05). The median time to peak CAR-T cell expansion occurred at 14 days post-infusion (range: 5–33 days), with the peak CAR-T cell proportion reaching 83.4% of T cells (range: 0.91–98.8%). CAR-T cells were detected in 34 of 61 (55.7%) patients who did not experience disease progression and had evaluable samples at 6 months, and in 17 of 53 (32.1%) patients at 12 months.
A higher CAR-T cell expansion peak was observed in responding patients (84.0%) compared with non-responding patients (14.7%) (p<0.001) (online supplemental figure S2A), as well as in CR patients (87.7%) compared with non-CR patients (75.2%) (p<0.001) (online supplemental figure S2B). Although the infused CAR-T cell dose in the CR group (2.73×106/kg) tended to be higher than that in the non-CR group (2.18×106/kg) (p=0.034) (online supplemental figure S2C), the infused dose showed no correlation with the in vivo expansion capacity of CAR-T cells following infusion (online supplemental figure S2B).
Given that CAR-T cell expansion is a critical initial step in achieving CAR-T efficacy, the cellular dynamics were analyzed in subgroups that exhibited poor efficacy following CAR-T therapy. No significant decrease in peak expansion was observed in patients with extramedullary disease (p=0.814). Meanwhile, a detailed analysis of the ASCT subgroup was conducted. The baseline characteristics of the ASCT and non-ASCT groups are summarized in online supplemental table S2. Infused CAR-T cell doses were also comparable between the two groups (p=0.812) (online supplemental figure S3A). The median number of peak CAR-T cell expansion in the ASCT group was significantly lower than that in the non-ASCT group (74.8% vs 89.8%, p<0.001) (online supplemental figure S3B). However, the CD4/CD8 ratio of CAR-T cells showed no significant difference between the two groups during the peak expansion period (p=0.812) (online supplemental figure S3C).
Recent ASCT adversely affected the fitness of infused CAR-T cells, potentially impacting expansion dynamics
An investigation was conducted to assess whether the intrinsic characteristics of infused CAR-T cells contributed to the observed variations in CAR-T cell expansion dynamics. The immunophenotypes of available infused CAR-T cell products from 55 patients in the ASCT group and 83 patients in the non-ASCT group were analyzed using flow cytometry. The CD4/CD8 ratio of the infused CAR-T cells showed no significant difference between the two groups (p=0.236). Furthermore, within the CD4 and CD8 CAR-T cell subsets, no notable differences were observed in the distribution of naive CAR-T cells (CARTN), central memory CAR-T cells (CARTCM), effector memory CAR-T cells (CARTEM), or effector CAR-T cells (CARTEFF) (figure 4A,B).
Figure 4. Distribution of infused CAR-T cells at various time intervals from ASCT to BCMA CAR-T therapy: the overall ASCT group (N=55) versus the non-ASCT group (N=83) (4A, 4B); the group over 1-year post-ASCT (N=43) versus the non-ASCT group (N=83) (4C, 4D); the group within 1 year post-ASCT (N=12) versus the non-ASCT group (N=83) (4E, 4F); the group over 1-year post-ASCT (N=43) versus the group within 1-year post-ASCT (N=12) (4G, 4 F). CAR-T cell subsets were assessed by flow cytometry as follows: naive CAR-T cell (CART N ) as CD62L+CD45RA+, central memory CAR-T cell (CART CM ) as CD62L+CD45RA-, effector memory CAR-T cell (CART EM ) as CD62L-CD45RA- and effector CAR-T cell (CART EFF ) as CD62L- CD45RA+. 3 of the 141 patients lacked infused CAR-T cell phenotype data. ASCT, autologous stem cell transplantation; BCMA, B-cell maturation antigen; CAR-T, chimeric antigen receptor T cells.
To evaluate the impact of varying time intervals between ASCT and CAR-T cell therapy, a detailed analysis was conducted on infused CAR-T cells derived from T cells collected more than 1 year post-ASCT. No significant differences were found in the CD4/CD8 ratio (p=0.148) or in the distribution of CARTN, CARTCM, CARTEM, and CARTEFF cells between patients who received CAR-T therapy more than 1-year post-ASCT and those who did not undergo ASCT (figure 4C,D).
Notably, the proportion of CARTN significantly decreased in infused CAR-T cells in patients who received ASCT in the past year compared with those who had not (p=0.006) in the CD4 subset (figure 4E). The CD8 subset exhibited a significant decrease in CARTN (p=0.005) and a marked increase in CARTEM (p=0.006) in patients who received ASCT within the previous year compared with those who had not (figure 4F). The CD4/CD8 ratio remained comparable between the two groups (p=0.920).
Compared with the group more than 1-year post-ASCT, no significant difference was observed in the CD4/CD8 ratio (p=0.349). However, in the group within 1-year post-ASCT, there was a significant decrease in CARTN (p=0.004, 0.021) and a marked increase in CARTEM (p=0.03, 0.01) for both the CD4 and CD8 subsets (figure 4G,H).
Salvage therapies after progression
Among patients with disease progression, seven received only palliative care, and data regarding salvage therapy were unavailable for six patients. Of the 57 patients eligible for salvage therapy, the median PFS2 was 4.1 months (95% CI, 2.3 to 5.9 months). Detailed information on salvage therapy is provided in online supplemental table S3. A total of 18 patients (31.6%) were treated with doublet, triplet, and quadruplet combinations of approved agents, yielding an ORR of 44.4% (8/18). Additionally, 16 patients (28.1%) who received chemotherapy had an ORR of 43.8% (7/16). Besides, 19 patients (33.3%) underwent CAR-T cell therapy or bispecific antibody therapy, achieving an ORR of 63.2% (12/19). Notably, one patient achieved CR following salvage allogeneic hematopoietic stem cell transplantation, and another patient with relapsed solitary plasmacytoma achieved CR after radiotherapy. Given the limited number of patients, these results warrant cautious interpretation.
Discussion
This study presents updated efficacy and safety results from 141 patients with R/RMM who underwent BCMA CAR-T cell therapy at our center. The findings are consistent with previously published clinical trial data.13 17 19 20 The median OS for these patients with advanced R/RMM has not yet been reached and is projected to exceed 60 months. Notably, among the 60 patients who demonstrated ongoing responses, 13 have maintained long-term responses for over 4 years. For these individuals, CAR-T cell therapy has proven to be profoundly impactful, enabling most to resume normal life and work without the need for long-term medication. This represents a transformative shift from traditional treatment paradigms.
Despite the promising efficacy of BCMA CAR-T cell therapy in R/RMM, a subset of patients fails to achieve CR or experiences relapse after an initial response.13 17 19 20 The depth of response has been identified as a critical factor linked to durable remission,21 with survival outcomes significantly improved in patients who achieve CR compared with those who do not. The exceptionally high rate of MRD negativity in BM, observed in 97.1% of CR patients, is promising. The clearance of BM plasma cells was rapid, typically occurring within 1 month, consistent with findings from prior studies.13 However, the accuracy of MRD detection may be limited at a sensitivity threshold of 10−4 nucleated cells. Despite the high rate of MRD negativity observed, this did not invariably lead to sustained remission for all patients, possibly due to the advanced, refractory nature of the disease, as well as the presence of extramedullary disease in a proportion of patients at baseline.
Extramedullary disease has been identified as an independent predictor of PFS, OS and relapse following CR in patients with R/RMM undergoing BCMA CAR-T cell therapy. Recently published prospective studies and real-world evidence have consistently supported these findings.28–33 Notably, prior extramedullary disease remains the predominant site of relapse following CAR-T cell therapy, indicating that extramedullary disease may act as an immunological sanctuary, predisposing it to tumor recurrence.34 This could be attributed to the high heterogeneity of extramedullary tumors, which may harbor clones inclined to evade CAR-T cell therapy. Furthermore, the unique microenvironment of extramedullary disease appears to impede CAR-T cell infiltration and sustained activity, thereby limiting therapeutic efficacy.35 36 Despite being less effective in patients with extramedullary disease compared with those without, it still yields meaningful clinical responses. In the absence of more effective treatments or other therapeutic options, CAR-T cell therapy remains a viable treatment option. Strategies for optimization should be considered, such as administering chemotherapy or radiation before the CAR-T cell infusion, to reduce tumor burden and enhance the clinical responses of CAR-T cell therapy.
ASCT is currently the predominant standard of care for eligible patients; however, progression and relapse remain inevitable.37 38 Several small-scale studies indicated that prior ASCT adversely affected the outcomes of subsequent BCMA CAR T-cell therapy, including PFS and ORR.30 39 Notably, our observations revealed a significant reduction in the peak expansion levels of CAR-T cells in vivo among patients who had previously undergone ASCT. Peak CAR-T cell expansion has been identified as a critical determinant of response.12 13 15 The insufficient expansion of CAR-T cells may be multifactorial. For autologous CAR-T cells, the functional capacity of the patient’s endogenous T cells potentially plays an integral role.23 24 Variations in the composition of T-cell subpopulations among patients with MM were notable and may be influenced by factors such as exposure to various treatment medications.40 Notably, MM is typically diagnosed in elderly patients,2 a population in which the number and functional diversity of naïve T cells are significantly diminished due to age-related thymic involution.41–43 After high-dose chemotherapy followed by ASCT, lymphocyte, the recovery of lymphocytes primarily depends on the peripheral expansion of memory cells, as the diversity of the T-cell repertoire available for lymphocyte expansion is limited.44 45 Despite the fact that patients in the ASCT group were younger than those in the non-ASCT group in our study, a reduced proportion of naive cells was observed in the infused CAR-T cells of patients who had undergone ASCT within the past year. Previous studies have indicated that a higher proportion of early memory and naïve cells in infused BCMA CAR-T cells is associated with superior in vivo expansion of CAR-T cells.23 24 However, due to the limited selection of makers, no significant differences were detected between the overall ASCT and non-ASCT groups, nor between patients more than 1-year post-ASCT and the non-ASCT group. Further research is needed to explore differences in CAR-T cell senescence and exhaustion among these patients. Given the significant impact of lymphocyte quality on the efficacy of CAR-T therapy, the timing of lymphocyte collection becomes crucial for enhancing clinical outcomes. Previous studies have demonstrated that patients with extramedullary disease, high-risk cytogenetic abnormalities, or those who did not achieve ≥VGPR before ASCT experienced significantly shorter PFS following ASCT.46–48 For these high-risk patients, lymphocyte collection should be considered before proceeding with ASCT.
Patients who experience disease progression following BCMA CAR-T therapy present significant management challenges, as there is currently no established standard of care for this patient population. This study found that salvage therapy with CAR T-cell therapy or bispecific antibody therapy following CAR-T therapy is feasible and may lead to relatively higher ORR compared with other conventional treatment options. Notably, GPRC5D CAR-T cell therapy showed the highest ORR (66.7%) and CR rate (33.3%) within our limited data set. GPRC5D, which is predominantly expressed on the surface of myeloma cells independently of BCMA, emerges as a promising alternative therapeutic target.49 Further studies with larger cohorts are needed to evaluate the efficacy of GPRC5D CAR-T cell therapy in treating patients with R/RMM with progressive disease after BCMA CAR-T cell therapy.
Overall, the long-term outcomes of this study highlight the safety and efficacy of BCMA CAR-T cell therapy in patients with R/RMM. Prior ASCT and extramedullary disease are recognized as adverse prognostic factors. The correlation between these adverse prognostic factors and the immunophenotype of the infused CAR-T cells, as well as post-infusion cell dynamics, has been identified, offering critical insights to further advance autologous CAR-T cell therapy. The ultimate goal is to refine treatment strategies to achieve the most durable and sustained responses in patients undergoing these therapies.
We express our gratitude to the entire team at Shanghai YaKe Biotechnology for their technical support and to the nurses at the Bone Marrow Transplantation Center, First Affiliated Hospital, Zhejiang University School of Medicine.
We also thank the editorial team of Home for Researchers (www.home-for-researchers.com) for their professional language editing services.
Data availability statement
Data are available upon reasonable request.
Ethics statements
Patient consent for publication
Not applicable.
Ethics approval
The study received approval from the Ethics Committee of the First Affiliated Hospital of Zhejiang University School of Medicine (IIT20240442A).
Contributors YH and HH conceived the project, supervised the research and revised the paper. CJ, RC and SF collected the data, designed and conducted the majority of the experiments, and wrote and revised the paper. MZ, YT, TY, FS, YK, TC and ZC reviewed and revised the article. JF, RH and GW provided CAR-T cell treatment and care to patients. JC and SH assisted with part of experiments and data analysis. HX, YZ and AHC helped the manufacture of CAR-T cells. YH is the guarantor of the study.
Funding This work was supported by National Natural Science Foundation of China (82270234, 82130003 and 82425002), the Key Project of Science and Technology Department of Zhejiang Province (2021C03010 and 2023C03060), and the Key Research and Development Program of Zhejiang Province (2024SSYS0023, 2024SSYS0024, and 2024SSYS0025).
Competing interests Authors YZ and AHC are employed by Shanghai YaKe Biotechnology. The remaining authors declare no competing interests.
Provenance and peer review Not commissioned; externally peer reviewed.
Supplemental material This content has been supplied by the author(s). It has not been vetted by BMJ Publishing Group Limited (BMJ) and may not have been peer-reviewed. Any opinions or recommendations discussed are solely those of the author(s) and are not endorsed by BMJ. BMJ disclaims all liability and responsibility arising from any reliance placed on the content. Where the content includes any translated material, BMJ does not warrant the accuracy and reliability of the translations (including but not limited to local regulations, clinical guidelines, terminology, drug names and drug dosages), and is not responsible for any error and/or omissions arising from translation and adaptation or otherwise.
1 Röllig C, Knop S, Bornhäuser M. Multiple myeloma. Lancet 2015; 385: 2197–208. doi:10.1016/S0140-6736(14)60493-1
2 Turesson I, Velez R, Kristinsson SY, et al. Patterns of multiple myeloma during the past 5 decades: stable incidence rates for all age groups in the population but rapidly changing age distribution in the clinic. Mayo Clin Proc 2010; 85: 225–30. doi:10.4065/mcp.2009.0426
3 Cowan AJ, Allen C, Barac A, et al. Global Burden of Multiple Myeloma: A Systematic Analysis for the Global Burden of Disease Study 2016. JAMA Oncol 2018; 4: 1221–7. doi:10.1001/jamaoncol.2018.2128
4 Chari A, Suvannasankha A, Fay JW, et al. Daratumumab plus pomalidomide and dexamethasone in relapsed and/or refractory multiple myeloma. Blood 2017; 130: 974–81. doi:10.1182/blood-2017-05-785246
5 Dimopoulos MA, Dytfeld D, Grosicki S, et al. Elotuzumab plus Pomalidomide and Dexamethasone for Multiple Myeloma. N Engl J Med 2018; 379: 1811–22. doi:10.1056/NEJMoa1805762
6 Moreau P, Masszi T, Grzasko N, et al. Oral Ixazomib, Lenalidomide, and Dexamethasone for Multiple Myeloma. N Engl J Med 2016; 374: 1621–34. doi:10.1056/NEJMoa1516282
7 Dimopoulos MA, Moreau P, Palumbo A, et al. Carfilzomib and dexamethasone versus bortezomib and dexamethasone for patients with relapsed or refractory multiple myeloma (ENDEAVOR): a randomised, phase 3, open-label, multicentre study. Lancet Oncol 2016; 17: 27–38. doi:10.1016/S1470-2045(15)00464-7
8 Attal M, Lauwers-Cances V, Hulin C, et al. Lenalidomide, Bortezomib, and Dexamethasone with Transplantation for Myeloma. N Engl J Med 2017; 376: 1311–20. doi:10.1056/NEJMoa1611750
9 Dhakal B, Szabo A, Chhabra S, et al. Autologous Transplantation for Newly Diagnosed Multiple Myeloma in the Era of Novel Agent Induction: A Systematic Review and Meta-analysis. JAMA Oncol 2018; 4: 343–50. doi:10.1001/jamaoncol.2017.4600
10 Gay F, Oliva S, Petrucci MT, et al. Chemotherapy plus lenalidomide versus autologous transplantation, followed by lenalidomide plus prednisone versus lenalidomide maintenance, in patients with multiple myeloma: a randomised, multicentre, phase 3 trial. Lancet Oncol 2015; 16: 1617–29. doi:10.1016/S1470-2045(15)00389-7
11 Carpenter RO, Evbuomwan MO, Pittaluga S, et al. B-cell maturation antigen is a promising target for adoptive T-cell therapy of multiple myeloma. Clin Cancer Res 2013; 19: 2048–60. doi:10.1158/1078-0432.CCR-12-2422
12 Munshi NC, Anderson LD, Shah N, et al. Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. N Engl J Med 2021; 384: 705–16. doi:10.1056/NEJMoa2024850
13 Raje N, Berdeja J, Lin Y, et al. Anti-BCMA CAR T-Cell Therapy bb2121 in Relapsed or Refractory Multiple Myeloma. N Engl J Med 2019; 380: 1726–37. doi:10.1056/NEJMoa1817226
14 Berdeja JG, Madduri D, Usmani SZ, et al. Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): a phase 1b/2 open-label study. Lancet 2021; 398: 314–24. doi:10.1016/S0140-6736(21)00933-8
15 Brudno JN, Maric I, Hartman SD, 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: 2267–80. doi:10.1200/JCO.2018.77.8084
16 San-Miguel J, Dhakal B, Yong K, et al. Cilta-cel or Standard Care in Lenalidomide-Refractory Multiple Myeloma. N Engl J Med 2023; 389: 335–47. doi:10.1056/NEJMoa2303379
17 Martin T, Usmani SZ, Berdeja JG, et al. Ciltacabtagene Autoleucel, an Anti-B-cell Maturation Antigen Chimeric Antigen Receptor T-Cell Therapy, for Relapsed/Refractory Multiple Myeloma: CARTITUDE-1 2-Year Follow-Up. J Clin Oncol 2023; 41: 1265–74. doi:10.1200/JCO.22.00842
18 Rodriguez-Otero P, Ailawadhi S, Arnulf B, et al. Ide-cel or Standard Regimens in Relapsed and Refractory Multiple Myeloma. N Engl J Med 2023; 388: 1002–14. doi:10.1056/NEJMoa2213614
19 Lin Y, Raje NS, Berdeja JG, et al. Idecabtagene vicleucel for relapsed and refractory multiple myeloma: post hoc 18-month follow-up of a phase 1 trial. Nat Med 2023; 29: 2286–94. doi:10.1038/s41591-023-02496-0
20 Zhao WH, Liu J, Wang BY, 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. doi:10.1186/s13045-018-0681-6
21 Xu J, Wang BY, Yu SH, et al. Long-term remission and survival in patients with relapsed or refractory multiple myeloma after treatment with LCAR-B38M CAR T cells: 5-year follow-up of the LEGEND-2 trial. J Hematol Oncol 2024; 17: 23. doi:10.1186/s13045-024-01530-z
22 Boulch M, Cazaux M, Loe-Mie Y, et al. A cross-talk between CAR T cell subsets and the tumor microenvironment is essential for sustained cytotoxic activity. Sci Immunol 2021; 6: eabd4344. doi:10.1126/sciimmunol.abd4344
23 Cohen AD, Garfall AL, Stadtmauer EA, et al. B cell maturation antigen-specific CAR T cells are clinically active in multiple myeloma. J Clin Invest 2019; 129: 2210–21: 126397. doi:10.1172/JCI126397
24 Finney OC, Yeri A, Mao P, et al. Molecular and Phenotypic Profiling of Drug Product and Post-Infusion Samples from CRB-402, an Ongoing: Phase I Clinical Study of bb21217 a BCMA-Directed CAR T Cell Therapy. Blood 2020; 136: 3–4. doi:10.1182/blood-2020-142426
25 Zhang M, Zhou L, Zhao H, et al. Risk Factors Associated with Durable Progression-Free Survival in Patients with Relapsed or Refractory Multiple Myeloma Treated with Anti-BCMA CAR T-cell Therapy. Clin Cancer Res 2021; 27: 6384–92. doi:10.1158/1078-0432.CCR-21-2031
26 Lee DW, Gardner R, Porter DL, et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014; 124: 188–95. doi:10.1182/blood-2014-05-552729
27 Kumar S, Paiva B, Anderson KC, et al. International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. Lancet Oncol 2016; 17: e328–46. doi:10.1016/S1470-2045(16)30206-6
28 Zhao WH, Wang BY, Chen LJ, et al. Four-year follow-up of LCAR-B38M in relapsed or refractory multiple myeloma: a phase 1, single-arm, open-label, multicenter study in China (LEGEND-2). J Hematol Oncol 2022; 15: 86. doi:10.1186/s13045-022-01301-8
29 Dima D, Abdallah A-O, Davis JA, et al. Impact of Extraosseous Extramedullary Disease on Outcomes of Patients with Relapsed-Refractory Multiple Myeloma receiving Standard-of-Care Chimeric Antigen Receptor T-Cell Therapy. Blood Cancer J 2024; 14: 90. doi:10.1038/s41408-024-01068-w
30 Wang Y, Cao J, Gu W, et al. Long-Term Follow-Up of Combination of B-Cell Maturation Antigen and CD19 Chimeric Antigen Receptor T Cells in Multiple Myeloma. JCO 2022; 40: 2246–56. doi:10.1200/JCO.21.01676
31 Deng H, Liu M, Yuan T, et al. Efficacy of Humanized Anti-BCMA CAR T Cell Therapy in Relapsed/Refractory Multiple Myeloma Patients With and Without Extramedullary Disease. Front Immunol 2021; 12: 720571. doi:10.3389/fimmu.2021.720571
32 Hashmi H, Hansen DK, Peres LC, et al. Factors associated with refractoriness or early progression after idecabtagene vicleucel in patients with relapsed/ refractory multiple myeloma: US Myeloma Immunotherapy Consortium real world experience. Haematologica 2024; 109: 1514–24. doi:10.3324/haematol.2023.283888
33 Gagelmann N, Dima D, Merz M, et al. Development and Validation of a Prediction Model of Outcome After B-Cell Maturation Antigen-Directed Chimeric Antigen Receptor T-Cell Therapy in Relapsed/Refractory Multiple Myeloma. J Clin Oncol 2024; 42: 1665–75. doi:10.1200/JCO.23.02232
34 Richard S, Lancman G, Rossi A, et al. Extramedullary Relapse Post CAR-T. Blood 2022; 140: 4301–2. doi:10.1182/blood-2022-170119
35 Bhutani M, Foureau DM, Atrash S, et al. Extramedullary multiple myeloma. Leukemia 2020; 34: 1–20. doi:10.1038/s41375-019-0660-0
36 Ryu D, Kim SJ, Hong Y, et al. Alterations in the Transcriptional Programs of Myeloma Cells and the Microenvironment during Extramedullary Progression Affect Proliferation and Immune Evasion. Clin Cancer Res 2020; 26: 935–44. doi:10.1158/1078-0432.CCR-19-0694
37 Cowan AJ, Green DJ, Kwok M, et al. Diagnosis and Management of Multiple Myeloma: A Review. JAMA 2022; 327: 464–77. doi:10.1001/jama.2022.0003
38 Rajkumar SV. Multiple myeloma: 2024 update on diagnosis, risk-stratification, and management. Am J Hematol 2024; 99: 1802–24. doi:10.1002/ajh.27422
39 Li C, Cao W, Que Y, et al. A phase I study of anti-BCMA CAR T cell therapy in relapsed/refractory multiple myeloma and plasma cell leukemia. Clin Transl Med 2021; 11: e346. doi:10.1002/ctm2.346
40 Rytlewski J, Madduri D, Fuller J, et al. Effects of Prior Alkylating Therapies on Preinfusion Patient Characteristics and Starting Material for CAR T Cell Product Manufacturing in Late-Line Multiple Myeloma. Blood 2020; 136: 7–8. doi:10.1182/blood-2020-134369
41 Nikolich-Žugich J. Aging of the T cell compartment in mice and humans: from no naive expectations to foggy memories. J Immunol 2014; 193: 2622–9. doi:10.4049/jimmunol.1401174
42 Herndler-Brandstetter D, Ishigame H, Flavell RA. How to define biomarkers of human T cell aging and immunocompetence? Front Immunol 2013; 4: 136. doi:10.3389/fimmu.2013.00136
43 Mittelbrunn M, Kroemer G. Hallmarks of T cell aging. Nat Immunol 2021; 22: 687–98. doi:10.1038/s41590-021-00927-z
44 Sarzotti M, Patel DD, Li X, et al. T cell repertoire development in humans with SCID after nonablative allogeneic marrow transplantation. J Immunol 2003; 170: 2711–8. doi:10.4049/jimmunol.170.5.2711
45 Dumont-Girard F, Roux E, van Lier RA, et al. Reconstitution of the T-cell compartment after bone marrow transplantation: restoration of the repertoire by thymic emigrants. Blood 1998; 92: 4464–71.
46 Kazmi SM, Nusrat M, Gunaydin H, et al. Outcomes Among High-Risk and Standard-Risk Multiple Myeloma Patients Treated With High-Dose Chemotherapy and Autologous Hematopoietic Stem-Cell Transplantation. Clin Lymphoma Myeloma Leuk 2015; 15: 687–93. doi:10.1016/j.clml.2015.07.641
47 Gagelmann N, Eikema D-J, Iacobelli S, et al. Impact of extramedullary disease in patients with newly diagnosed multiple myeloma undergoing autologous stem cell transplantation: a study from the Chronic Malignancies Working Party of the EBMT. Haematologica 2018; 103: 890–7. doi:10.3324/haematol.2017.178434
48 Hsu TL, Tsai CK, Liu CY, et al. Risk factors of early disease progression and decreased survival for multiple myeloma patients after upfront autologous stem cell transplantation. Ann Hematol 2024; 103: 2893–904. doi:10.1007/s00277-024-05641-y
49 Smith EL, Harrington K, Staehr M, et al. GPRC5D is a target for the immunotherapy of multiple myeloma with rationally designed CAR T cells. Sci Transl Med 2019; 11: eaau7746. doi:10.1126/scitranslmed.aau7746
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2025 Author(s) (or their employer(s)) 2025. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ Group. http://creativecommons.org/licenses/by-nc/4.0/ This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See http://creativecommons.org/licenses/by-nc/4.0/ . Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Background
B-cell maturation antigen (BCMA)-targeting chimeric antigen receptor (CAR) T-cell immunotherapy has shown promising results in the treatment of relapsed or refractory multiple myeloma (R/RMM). This study presents the updated long-term outcomes from our center.
Methods
Between July 30, 2018, and September 27, 2023, 141 patients with R/RMM who received BCMA CAR-T therapy were enrolled. Patients underwent conditioning chemotherapy with cyclophosphamide and fludarabine, followed by BCMA CAR-T cell infusion at a median dose of 2.36×106 cells/kg. The study evaluated overall response rates, long-term efficacy, safety profiles, and their associations with clinical and disease characteristics.
Results
At a median follow-up of 20.2 months, the safety profile of the therapy was manageable. Grade 3/4 cytokine release syndrome occurred in 36.2% of patients, with no cases of severe neurotoxicity reported. 1-month post-infusion, grade ≥3 anemia persisted in 39.6% of patients, while neutropenia (43.3%) and thrombocytopenia (52.2%) were observed. The objective response rate (ORR) among evaluable patients was 94.8%, with 50.7% achieving a complete response (CR). The 4-year progression-free survival and overall survival rates were 37.4% (95% CI, 29.1% to 48.1%) and 63.2% (95% CI, 54.8% to 72.8%), respectively, with survival curves showing gradual flattening over time. Patients with a history of autologous stem cell transplantation (ASCT) and those with extramedullary disease demonstrated significantly inferior efficacy and survival outcomes. Peak CAR-T cell expansion was positively correlated with ORR (p<0.001) and CR (p<0.001). Notably, patients with prior ASCT exhibited significantly lower CAR-T cell expansion compared with those without prior ASCT (p<0.001). Immunophenotypic analysis of infused CAR-T cells demonstrated impaired fitness in patients who received ASCT in the past year.
Conclusions
BCMA CAR-T therapy in patients with R/RMM results in significant and sustained responses, with a manageable safety profile on a large scale. Prior ASCT and extramedullary disease represent adverse prognostic factors. Patients with a history of ASCT demonstrate limited peak CAR-T cell expansion.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details




1 Bone Marrow Transplantation Center of The First Affiliated Hospital and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
2 Shanghai YaKe Biotechnology Ltd, Shanghai, China
3 Division of Haematology, Medical Oncology and HSCT, Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, Hong Kong
4 Shanghai YaKe Biotechnology Ltd, Shanghai, China; Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, Shanghai, China