Chimeric antigen receptor T-cell therapy outcomes in T cell/histiocyte-rich large B-cell lymphoma and subsequent treatment strategies after disease progression: A GELTAMO/GETH study

IF 14.6 2区 医学 Q1 HEMATOLOGY
HemaSphere Pub Date : 2025-02-04 DOI:10.1002/hem3.70077
Mariana Bastos-Oreiro, Gloria Iacoboni, Víctor N. Garcés, Ana C. Caballero, Nuria Martínez, Javier Delgado, Aitana Balaguer, Mi Kwon, Sonia Gonzalez de Villambrosia, Rafael Hernani, Ana Jimenez-Ubieto, Rebeca Bailen, Izaskun Zaberio, Alejandro Martín García-Sancho, Pere Barba
{"title":"Chimeric antigen receptor T-cell therapy outcomes in T cell/histiocyte-rich large B-cell lymphoma and subsequent treatment strategies after disease progression: A GELTAMO/GETH study","authors":"Mariana Bastos-Oreiro,&nbsp;Gloria Iacoboni,&nbsp;Víctor N. Garcés,&nbsp;Ana C. Caballero,&nbsp;Nuria Martínez,&nbsp;Javier Delgado,&nbsp;Aitana Balaguer,&nbsp;Mi Kwon,&nbsp;Sonia Gonzalez de Villambrosia,&nbsp;Rafael Hernani,&nbsp;Ana Jimenez-Ubieto,&nbsp;Rebeca Bailen,&nbsp;Izaskun Zaberio,&nbsp;Alejandro Martín García-Sancho,&nbsp;Pere Barba","doi":"10.1002/hem3.70077","DOIUrl":null,"url":null,"abstract":"<p>Chimeric antigen receptor T-cell therapy (CAR-T) is an effective approach for patients with relapsed/refractory (R/R) large B-cell lymphoma (LBCL). However, some rare variants do not seem to respond as well to this T-cell redirecting strategy. T-cell/histiocyte-rich large B-cell lymphoma (THRLBCL) is an infrequent subtype of LBCL which typically develops in young, male patients, characterized by an aggressive clinical course and chemo-refractory disease.<span><sup>1, 2</sup></span> THRLBCL has unique biological characteristics and a inhibitory tumor immune microenvironment.<span><sup>3, 4</sup></span> It is well-known that the programmed cell death protein 1 (PD-1)/programmed cell death ligand 1 (PD-L1) pathway is a key driver of immune escape;<span><sup>5, 6</sup></span> this lymphoma subtype has been associated with PD-L1 gene alterations, such as PD-L1 copy gains and high PD-L1 expression on malignant B cells (often surrounded by abundant PD-L1–expressing macrophages and PD-1 + T cells). This distinct clinical behavior, together with its low incidence, have led to an underrepresentation of THRLBCL patients in most clinical trials, including those evaluating CAR T-cell therapy. Therefore, real-world data with this entity is highly anticipated. Taking all of this into consideration, we aimed to assess the safety and efficacy outcomes of THRLBCL after CAR T-cell therapy, outside of the clinical trial setting, as well as the efficacy of postrelapse approaches.</p><p>We carried out a retrospective, multicentre study including all adult patients with this diagnosis registered in the GELTAMO/GETH-TC database (Grupo Español de Linfomas y Trasplante Autólogo de Médula Ósea/Grupo Español de Trasplante Hematopoyético y Terapia Celular) from April 2019 to January 2024 who had received a CD19-targeted CAR T-cell infusion. The primary endpoint was overall survival (OS) and the secondary endpoints were response rate, progression-free survival (PFS), duration of response (DR), and subsequent therapy outcome. Twenty patients with R/R THRLBCL from 11 Spanish centers received CAR T-cell therapy from April 2019 to December 2023. If the patient had experienced disease progression after CAR-T, participating centers completed an additional database on subsequent treatments and their outcomes. Cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) were graded according to the ASTCT consensus criteria.<span><sup>7</sup></span> Response assessment followed the Lugano recommendations.<span><sup>8</sup></span> OS and PFS were determined from CAR-T cell infusion for CAR T-cell outcomes and since the start of the first subsequent treatment for the following approaches. These were calculated using the Kaplan–Meier method and the Cox model to obtain hazard ratios (HR) with 95% confidence intervals (CI) and <i>p</i>-values. All reported <i>p</i>-values were two-sided, and statistical significance was defined at <i>p</i> &lt; 0.05. Statistical analyses were performed using R software version 4.2.2.</p><p>In terms of baseline characteristics, median age was 50 years (interquartile range [IQR]: 40–64) and 16 patients (80%) were male. The International Prognostic Index (IPI) score at time of CAR-T was ≥3 in 55% of patients, and 52% had an Eastern Cooperative Oncology Group performance status (ECOG-PS) ≥ 1. Two patients (10%) received the CAR-T in the second line, 16 (80%) in third line, and 2 (10%) in fourth line. Considering the construct, 11 (55%) received axicabtagene ciloleucel (axi-cel), 8 (40%) tisagenlecleucel (tisa-cel), and 1 (5%) lisocabtagene maraleucel, which was administered in a clinical trial. Nineteen patients (95%) received bridging therapy (8 platinum-based regimens with Rituximab (R), 7 cyclophosphamide-R based regimens, 2 radiotherapy, 1 polatuzumab-bendamustine-R, 1 brentuximab monotherapy, 1 pembrolizumab monotherapy) (Supporting Information S1: Table 1).</p><p>Concerning the safety profile, any (grade ≥3) CRS and ICANS rates were 85% (5%) and 50% (15%), respectively. One patient infused with tisa-cel died in the context of severe CRS and hemophagocytic syndrome. All other deaths in the analysis were due to disease progression. Regarding efficacy, the overall and complete response rate [ORR, CRR]) was 50% and 25%, respectively (Figure 1); all cases of CR occurred in axicel-treated patients. In the univariate analysis, factors associated with achieving CR versus other response were a lower pretreatment LDH level (median 182 vs. 682, <i>p</i> = 0.03), the use of axi-cel (<i>p</i> = 0.05), a low IPI score (IPI 0-2) at time of CAR-T (<i>p</i> = 0.03), and having received a prior autologous stem cell transplantation (ASCT) (<i>p</i> = 0.03). Regarding the risk of relapse, only a low IPI (I/II) pre-CART (HR: 1.73, 95% CI: 1.08–2.7, <i>p</i> = 0.04) and a prior ASCT (HR: 0.27, 95% CI: 0.08–0.9, <i>p</i> = 0.03) were significant. With a median follow-up of 25 months (95% CI: 13–NA) from CAR-T infusion, the median PFS and OS were 3.3 (95% CI: 1.4–11) and 9.2 months (95% CI: 6.1–NA), respectively. The 12-month PFS and OS were 23% and 43%, respectively (Supporting Information S1: Figure 1A,B). Supporting Information S1: Figure 1 shows the univariate analysis for responses, PFS, OS, and response. The median DR was 3.9 months (95% CI: 2–NA). In all cases, CAR-T expansion was observed. Of the 14 cases with persistence data, only in two cases the CAR-T did not persist at 6 and 12 months; however, in all cases, there was B cell aplasia at the time of the last follow-up. Eighteen (90%) patients experienced progressive disease (PD) after CAR-T-cell therapy, with a median OS since PD of 5.1 months (3.7, NR) and a 24-month OS of 24.2% (9.6–61.1). Ten (56%) of the 18 patients with PD after CAR-T received subsequent treatment, with a median PFS and OS of 3.6 (2.9–not reached) and 6.5 (3.0–not reached) months, respectively (Supporting Information S1: Figure 2C,D). This showed a clear trend towards improved survival compared to patients who only received best supportive care after CAR-T failure. in comparison to patients who only received best supportive care after CAR-T failure (Supporting Information S1: Figures 2 and 3). The median time from CAR-T progression to next therapy was 42.5 (IQR: 38–50.5; range = 4–57). In terms of the subsequent treatment outcomes, 5/10 patients achieved a response, using regimens based on checkpoint inhibitors (pembrolizumab, atezolizumab), polatuzumab, and tafasitamab-lenalidomide (Figure 1). Two patients achieved a CR, after radiotherapy (<i>n</i> = 1) and pembrolizumab monotherapy (<i>n</i> = 1). One patient treated with atezolizumab-glofitamab (a 1-year fixed-duration treatment) achieved a PR that is still ongoing after 2.5 years. No significant immune toxicities were observed in patients treated with CPI and/or BiAb after CAR-T. Only one patient underwent an allogeneic stem cell transplantation as consolidation after achieving a PR with four cycles of Tafasitamab-Lenalidomide, and maintains the response 6 months after the transplant.</p><p>CAR T-cell therapy is the standard of care for LBCL patients with an early (&lt;12 months) relapse after frontline therapy or for patients after a second relapse, due to the high response rate, DR, and survival improvement, compared to alternative regimens.<span><sup>9-13</sup></span> Most of the pivotal trials included patients with diffuse large B-cell lymphoma, transformed follicular lymphoma, and high-grade B-cell lymphoma. However, THRLBCL was usually excluded due to its low incidence and distinct biological profile. In this study, we report one of the largest real-world cohorts of THRLBCL receiving CAR T-cells and provide a detailed analysis of subsequent treatment strategies.</p><p>Trujillo et al. published the first series of patients with THRLBCL treated with CAR-T (<i>N</i> = 9), with dismal outcomes after this T-cell redirecting strategy,<span><sup>6</sup></span> suggesting a potential refractory behavior in this patient population. They hypothesized that CAR T-cell failure in THRLBCL appeared to be related to acquired CAR T-cell dysfunction, rather than poor CAR-T cell expansion.<span><sup>14</sup></span> Recently, Pophali et al reported a 2-year PFS and OS of 29% and 42%, respectively, in 58 R/R THRLBCL patients treated with CAR T-cells;<span><sup>15</sup></span> in this study, ECOG-PS was the only factor with an impact on survival. However, beyond these intriguing data, there have been very few studies focused on the clinical characteristics and response patterns to CAR-T therapy in this infrequent disease entity.</p><p>Similar to these series, patients in our study had poor outcomes, with a median PFS and OS of 3.3 and 9.2 months, respectively. Interestingly, the patients achieving CR were those with a low LDH, early stage and low-risk IPI at time of CAR-T cell therapy. However, regardless of the response to CAR-T, 18 out of 20 patients experienced disease progression, in syntony with the previously mentioned studies. Strikingly, our long-term OS for the patients rescued after CAR-T relapse was very similar to that reported by Pophali et al., with 40% of patients alive at 2 years. Considering the high relapse rate, effective subsequent strategies seemed to be the underlying reason for these long-term outcomes. In our dataset, among patients receiving treatment after CAR-T relapse (<i>N</i> = 10), 2-year OS was 44%, even though most were early relapses (30% &lt;3 months, 70% &lt;6 months), significantly higher in comparison to other reports including outcomes of R/R LBCL patients with an early relapse post-CAR-T.<span><sup>16</sup></span> Patients receiving PD1-blockers and bispecific antibodies presented better outcomes, supporting the hypothesis that THRLBCL could be responsive to PD-1 blockade therapy in light of the exceptionally high numbers of PD-L1-expressing tumor-associated macrophages and PD-1 + T cells that surround the malignant B cells.<span><sup>5</sup></span></p><p>Concerning toxicity, incidence and severity of short-term adverse events, such as CRS and ICANS, were similar to the pivotal trials and real-world DLBCL data.<span><sup>17, 18</sup></span></p><p>The main limitation of our study is the small sample size, as well as the lack of centralized pathology review. However, despite this drawback, we report long follow-up after CAR-T infusion and provide insight into subsequent treatment strategies, which remain an important knowledge gap in this particular setting.</p><p>In conclusion, the efficacy of CAR T-cell therapy was significantly lower for THRLBCL patients in comparison with LBCL patients. However, response rates to subsequent treatment were encouraging. In light of the results with checkpoint inhibitors or bispecific antibodies in the post-CART scenario, the development of clinical trials focused on this patient population exploring combination strategies with CAR-T is highly anticipated.</p><p>Mariana Bastos-Oreiro and Gloria Iacoboni designed research, performed research, collected, analyzed and interpreted data, and wrote the paper. Víctor N. Garcés performed the statistical analysis. Ana C. Caballero, Javier Delgado, Aitana Balaguer, Mi Kwon, Sonia Gonzalez de Villambrosia, Ana Jimenez-Ubieto, Rebeca Bailen, and Alejandro Martín García-Sancho collected data and performed research. Pere Barba designed research, conceptualized, and supervised the study.</p><p>Mariana Bastos-Oreiro: Honoraria and/or travel support from Abbvie, Bristol-Myers Squibb, Kite/Gilead, and Novartis. Gloria Iacoboni: Honoraria and/or travel support from Abbvie, AstraZeneca, Autolus, Bristol-Myers Squibb, Kite/Gilead, Miltenyi, and Novartis. Víctor N. Garcés: No disclosures. Ana C. Caballero: No disclosures. Javier Delgado: No disclosures. Aitana Balaguer: No disclosures. Mi Kwon: Honoraria and/or travel support from Kite/Gilead, Novartis. Sonia Gonzalez de Villambrosia: Honoraria and/or travel support from Abbvie, Roche, Incyte, Bristol-Myers Squibb, Kite/Gilead, Miltenyi, and Novartis. Ana Jimenez-Ubieto: Honoraria and/or travel support from Abbvie, Roche, Incyte, Kite/Gilead, Miltenyi, and Novartis. Rebeca Bailen: Honoraria and/or travel support from Incyte, Bristol-Myers Squibb, Kite/Gilead, Miltenyi, and Novartis. Alejandro Martín García-Sancho: Honoraria and/or consulting fees: Roche, BMS, Takeda, Janssen, Kyowa Kirin, Gilead/Kite, Incyte, Lilly, Miltenyi, Ideogen, Genmab, Abbvie, Sobi, Astra-Zeneca, GSK, Regeneron. Pere Barba: Honoraria and/or travel support from Abbvie, AstraZeneca, Autolus, Bristol-Myers Squibb, Kite/Gilead, Miltenyi, and Novartis.</p><p>This research received no funding.</p>","PeriodicalId":12982,"journal":{"name":"HemaSphere","volume":"9 2","pages":""},"PeriodicalIF":14.6000,"publicationDate":"2025-02-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hem3.70077","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"HemaSphere","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/hem3.70077","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"HEMATOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Chimeric antigen receptor T-cell therapy (CAR-T) is an effective approach for patients with relapsed/refractory (R/R) large B-cell lymphoma (LBCL). However, some rare variants do not seem to respond as well to this T-cell redirecting strategy. T-cell/histiocyte-rich large B-cell lymphoma (THRLBCL) is an infrequent subtype of LBCL which typically develops in young, male patients, characterized by an aggressive clinical course and chemo-refractory disease.1, 2 THRLBCL has unique biological characteristics and a inhibitory tumor immune microenvironment.3, 4 It is well-known that the programmed cell death protein 1 (PD-1)/programmed cell death ligand 1 (PD-L1) pathway is a key driver of immune escape;5, 6 this lymphoma subtype has been associated with PD-L1 gene alterations, such as PD-L1 copy gains and high PD-L1 expression on malignant B cells (often surrounded by abundant PD-L1–expressing macrophages and PD-1 + T cells). This distinct clinical behavior, together with its low incidence, have led to an underrepresentation of THRLBCL patients in most clinical trials, including those evaluating CAR T-cell therapy. Therefore, real-world data with this entity is highly anticipated. Taking all of this into consideration, we aimed to assess the safety and efficacy outcomes of THRLBCL after CAR T-cell therapy, outside of the clinical trial setting, as well as the efficacy of postrelapse approaches.

We carried out a retrospective, multicentre study including all adult patients with this diagnosis registered in the GELTAMO/GETH-TC database (Grupo Español de Linfomas y Trasplante Autólogo de Médula Ósea/Grupo Español de Trasplante Hematopoyético y Terapia Celular) from April 2019 to January 2024 who had received a CD19-targeted CAR T-cell infusion. The primary endpoint was overall survival (OS) and the secondary endpoints were response rate, progression-free survival (PFS), duration of response (DR), and subsequent therapy outcome. Twenty patients with R/R THRLBCL from 11 Spanish centers received CAR T-cell therapy from April 2019 to December 2023. If the patient had experienced disease progression after CAR-T, participating centers completed an additional database on subsequent treatments and their outcomes. Cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) were graded according to the ASTCT consensus criteria.7 Response assessment followed the Lugano recommendations.8 OS and PFS were determined from CAR-T cell infusion for CAR T-cell outcomes and since the start of the first subsequent treatment for the following approaches. These were calculated using the Kaplan–Meier method and the Cox model to obtain hazard ratios (HR) with 95% confidence intervals (CI) and p-values. All reported p-values were two-sided, and statistical significance was defined at p < 0.05. Statistical analyses were performed using R software version 4.2.2.

In terms of baseline characteristics, median age was 50 years (interquartile range [IQR]: 40–64) and 16 patients (80%) were male. The International Prognostic Index (IPI) score at time of CAR-T was ≥3 in 55% of patients, and 52% had an Eastern Cooperative Oncology Group performance status (ECOG-PS) ≥ 1. Two patients (10%) received the CAR-T in the second line, 16 (80%) in third line, and 2 (10%) in fourth line. Considering the construct, 11 (55%) received axicabtagene ciloleucel (axi-cel), 8 (40%) tisagenlecleucel (tisa-cel), and 1 (5%) lisocabtagene maraleucel, which was administered in a clinical trial. Nineteen patients (95%) received bridging therapy (8 platinum-based regimens with Rituximab (R), 7 cyclophosphamide-R based regimens, 2 radiotherapy, 1 polatuzumab-bendamustine-R, 1 brentuximab monotherapy, 1 pembrolizumab monotherapy) (Supporting Information S1: Table 1).

Concerning the safety profile, any (grade ≥3) CRS and ICANS rates were 85% (5%) and 50% (15%), respectively. One patient infused with tisa-cel died in the context of severe CRS and hemophagocytic syndrome. All other deaths in the analysis were due to disease progression. Regarding efficacy, the overall and complete response rate [ORR, CRR]) was 50% and 25%, respectively (Figure 1); all cases of CR occurred in axicel-treated patients. In the univariate analysis, factors associated with achieving CR versus other response were a lower pretreatment LDH level (median 182 vs. 682, p = 0.03), the use of axi-cel (p = 0.05), a low IPI score (IPI 0-2) at time of CAR-T (p = 0.03), and having received a prior autologous stem cell transplantation (ASCT) (p = 0.03). Regarding the risk of relapse, only a low IPI (I/II) pre-CART (HR: 1.73, 95% CI: 1.08–2.7, p = 0.04) and a prior ASCT (HR: 0.27, 95% CI: 0.08–0.9, p = 0.03) were significant. With a median follow-up of 25 months (95% CI: 13–NA) from CAR-T infusion, the median PFS and OS were 3.3 (95% CI: 1.4–11) and 9.2 months (95% CI: 6.1–NA), respectively. The 12-month PFS and OS were 23% and 43%, respectively (Supporting Information S1: Figure 1A,B). Supporting Information S1: Figure 1 shows the univariate analysis for responses, PFS, OS, and response. The median DR was 3.9 months (95% CI: 2–NA). In all cases, CAR-T expansion was observed. Of the 14 cases with persistence data, only in two cases the CAR-T did not persist at 6 and 12 months; however, in all cases, there was B cell aplasia at the time of the last follow-up. Eighteen (90%) patients experienced progressive disease (PD) after CAR-T-cell therapy, with a median OS since PD of 5.1 months (3.7, NR) and a 24-month OS of 24.2% (9.6–61.1). Ten (56%) of the 18 patients with PD after CAR-T received subsequent treatment, with a median PFS and OS of 3.6 (2.9–not reached) and 6.5 (3.0–not reached) months, respectively (Supporting Information S1: Figure 2C,D). This showed a clear trend towards improved survival compared to patients who only received best supportive care after CAR-T failure. in comparison to patients who only received best supportive care after CAR-T failure (Supporting Information S1: Figures 2 and 3). The median time from CAR-T progression to next therapy was 42.5 (IQR: 38–50.5; range = 4–57). In terms of the subsequent treatment outcomes, 5/10 patients achieved a response, using regimens based on checkpoint inhibitors (pembrolizumab, atezolizumab), polatuzumab, and tafasitamab-lenalidomide (Figure 1). Two patients achieved a CR, after radiotherapy (n = 1) and pembrolizumab monotherapy (n = 1). One patient treated with atezolizumab-glofitamab (a 1-year fixed-duration treatment) achieved a PR that is still ongoing after 2.5 years. No significant immune toxicities were observed in patients treated with CPI and/or BiAb after CAR-T. Only one patient underwent an allogeneic stem cell transplantation as consolidation after achieving a PR with four cycles of Tafasitamab-Lenalidomide, and maintains the response 6 months after the transplant.

CAR T-cell therapy is the standard of care for LBCL patients with an early (<12 months) relapse after frontline therapy or for patients after a second relapse, due to the high response rate, DR, and survival improvement, compared to alternative regimens.9-13 Most of the pivotal trials included patients with diffuse large B-cell lymphoma, transformed follicular lymphoma, and high-grade B-cell lymphoma. However, THRLBCL was usually excluded due to its low incidence and distinct biological profile. In this study, we report one of the largest real-world cohorts of THRLBCL receiving CAR T-cells and provide a detailed analysis of subsequent treatment strategies.

Trujillo et al. published the first series of patients with THRLBCL treated with CAR-T (N = 9), with dismal outcomes after this T-cell redirecting strategy,6 suggesting a potential refractory behavior in this patient population. They hypothesized that CAR T-cell failure in THRLBCL appeared to be related to acquired CAR T-cell dysfunction, rather than poor CAR-T cell expansion.14 Recently, Pophali et al reported a 2-year PFS and OS of 29% and 42%, respectively, in 58 R/R THRLBCL patients treated with CAR T-cells;15 in this study, ECOG-PS was the only factor with an impact on survival. However, beyond these intriguing data, there have been very few studies focused on the clinical characteristics and response patterns to CAR-T therapy in this infrequent disease entity.

Similar to these series, patients in our study had poor outcomes, with a median PFS and OS of 3.3 and 9.2 months, respectively. Interestingly, the patients achieving CR were those with a low LDH, early stage and low-risk IPI at time of CAR-T cell therapy. However, regardless of the response to CAR-T, 18 out of 20 patients experienced disease progression, in syntony with the previously mentioned studies. Strikingly, our long-term OS for the patients rescued after CAR-T relapse was very similar to that reported by Pophali et al., with 40% of patients alive at 2 years. Considering the high relapse rate, effective subsequent strategies seemed to be the underlying reason for these long-term outcomes. In our dataset, among patients receiving treatment after CAR-T relapse (N = 10), 2-year OS was 44%, even though most were early relapses (30% <3 months, 70% <6 months), significantly higher in comparison to other reports including outcomes of R/R LBCL patients with an early relapse post-CAR-T.16 Patients receiving PD1-blockers and bispecific antibodies presented better outcomes, supporting the hypothesis that THRLBCL could be responsive to PD-1 blockade therapy in light of the exceptionally high numbers of PD-L1-expressing tumor-associated macrophages and PD-1 + T cells that surround the malignant B cells.5

Concerning toxicity, incidence and severity of short-term adverse events, such as CRS and ICANS, were similar to the pivotal trials and real-world DLBCL data.17, 18

The main limitation of our study is the small sample size, as well as the lack of centralized pathology review. However, despite this drawback, we report long follow-up after CAR-T infusion and provide insight into subsequent treatment strategies, which remain an important knowledge gap in this particular setting.

In conclusion, the efficacy of CAR T-cell therapy was significantly lower for THRLBCL patients in comparison with LBCL patients. However, response rates to subsequent treatment were encouraging. In light of the results with checkpoint inhibitors or bispecific antibodies in the post-CART scenario, the development of clinical trials focused on this patient population exploring combination strategies with CAR-T is highly anticipated.

Mariana Bastos-Oreiro and Gloria Iacoboni designed research, performed research, collected, analyzed and interpreted data, and wrote the paper. Víctor N. Garcés performed the statistical analysis. Ana C. Caballero, Javier Delgado, Aitana Balaguer, Mi Kwon, Sonia Gonzalez de Villambrosia, Ana Jimenez-Ubieto, Rebeca Bailen, and Alejandro Martín García-Sancho collected data and performed research. Pere Barba designed research, conceptualized, and supervised the study.

Mariana Bastos-Oreiro: Honoraria and/or travel support from Abbvie, Bristol-Myers Squibb, Kite/Gilead, and Novartis. Gloria Iacoboni: Honoraria and/or travel support from Abbvie, AstraZeneca, Autolus, Bristol-Myers Squibb, Kite/Gilead, Miltenyi, and Novartis. Víctor N. Garcés: No disclosures. Ana C. Caballero: No disclosures. Javier Delgado: No disclosures. Aitana Balaguer: No disclosures. Mi Kwon: Honoraria and/or travel support from Kite/Gilead, Novartis. Sonia Gonzalez de Villambrosia: Honoraria and/or travel support from Abbvie, Roche, Incyte, Bristol-Myers Squibb, Kite/Gilead, Miltenyi, and Novartis. Ana Jimenez-Ubieto: Honoraria and/or travel support from Abbvie, Roche, Incyte, Kite/Gilead, Miltenyi, and Novartis. Rebeca Bailen: Honoraria and/or travel support from Incyte, Bristol-Myers Squibb, Kite/Gilead, Miltenyi, and Novartis. Alejandro Martín García-Sancho: Honoraria and/or consulting fees: Roche, BMS, Takeda, Janssen, Kyowa Kirin, Gilead/Kite, Incyte, Lilly, Miltenyi, Ideogen, Genmab, Abbvie, Sobi, Astra-Zeneca, GSK, Regeneron. Pere Barba: Honoraria and/or travel support from Abbvie, AstraZeneca, Autolus, Bristol-Myers Squibb, Kite/Gilead, Miltenyi, and Novartis.

This research received no funding.

Abstract Image

嵌合抗原受体T细胞治疗T细胞/富含组织细胞的大b细胞淋巴瘤的结果和疾病进展后的后续治疗策略:一项GELTAMO/GETH研究
嵌合抗原受体t细胞疗法(CAR-T)是治疗复发/难治性(R/R)大b细胞淋巴瘤(LBCL)的有效方法。然而,一些罕见的变异似乎对这种t细胞重定向策略没有反应。t细胞/组织细胞丰富的大b细胞淋巴瘤(THRLBCL)是一种罕见的LBCL亚型,通常发生在年轻男性患者中,其特点是临床病程侵袭性和化疗难治性。1,2 THRLBCL具有独特的生物学特性和抑制性肿瘤免疫微环境。3,4众所周知,程序性细胞死亡蛋白1 (PD-1)/程序性细胞死亡配体1 (PD-L1)通路是免疫逃逸的关键驱动因素;5,6这种淋巴瘤亚型与PD-L1基因改变有关,如PD-L1拷贝增加和PD-L1在恶性B细胞(通常被大量表达PD-L1的巨噬细胞和PD-1 + T细胞包围)上的高表达。这种独特的临床行为,加上其低发病率,导致THRLBCL患者在大多数临床试验中代表性不足,包括那些评估CAR - t细胞治疗的临床试验。因此,这个实体的真实数据备受期待。考虑到所有这些因素,我们的目的是评估CAR - t细胞治疗后THRLBCL的安全性和有效性结果,在临床试验环境之外,以及复发后方法的有效性。我们进行了一项回顾性的多中心研究,包括2019年4月至2024年1月接受cd19靶向CAR -t细胞输注的GELTAMO/GETH-TC数据库(Grupo Español de linfoomas y Trasplante Autólogo de membrodula Ósea/Grupo Español de Trasplante hematopoyterapia cell)中登记的所有患有这种诊断的成年患者。主要终点是总生存期(OS),次要终点是缓解率、无进展生存期(PFS)、缓解持续时间(DR)和后续治疗结果。2019年4月至2023年12月,来自西班牙11个中心的20名R/R THRLBCL患者接受了CAR - t细胞治疗。如果患者在CAR-T后经历了疾病进展,参与中心完成了后续治疗及其结果的额外数据库。细胞因子释放综合征(CRS)和免疫效应细胞相关神经毒性综合征(ICANS)根据ASTCT共识标准进行分级响应评估遵循卢加诺的建议OS和PFS是通过CAR- t细胞输注来确定CAR- t细胞结果的,并且自以下方法的第一次后续治疗开始以来。使用Kaplan-Meier方法和Cox模型计算这些数据,以获得95%置信区间(CI)和p值的风险比(HR)。所有报告的p值均为双侧,p &lt; 0.05定义为统计学显著性。采用R软件4.2.2进行统计分析。基线特征方面,中位年龄为50岁(四分位间距[IQR]: 40-64), 16例(80%)为男性。CAR-T治疗时,55%的患者国际预后指数(IPI)评分≥3,52%的患者东部肿瘤合作组表现状态(ECOG-PS)≥1。2例(10%)患者在二线接受CAR-T治疗,16例(80%)患者在三线接受CAR-T治疗,2例(10%)患者在四线接受CAR-T治疗。考虑到结构,11人(55%)接受了axicabtagene ciloleucel(轴细胞),8人(40%)接受了tisagenlecleucel(组织细胞),1人(5%)接受了lisocabtagene maraleucel,这是在临床试验中给予的。19例(95%)患者接受了桥接治疗(8例铂基方案联合利妥昔单抗(R), 7例环磷酰胺-R方案,2例放疗,1例polatuzumab-苯达莫司汀-R, 1例brentuximab单药治疗,1例派姆单抗单药治疗)(支持信息S1:表1)。关于安全性,任何(分级≥3)的CRS和ICANS率分别为85%(5%)和50%(15%)。1例输注组织细胞的患者死于严重的CRS和噬血细胞综合征。分析中所有其他死亡都是由于疾病进展。在疗效方面,总缓解率和完全缓解率[ORR, CRR]]分别为50%和25%(图1);所有CR病例均发生在阿克塞尔治疗的患者中。在单变量分析中,实现CR与其他应答相关的因素是预处理LDH水平较低(中位数182对682,p = 0.03),使用轴细胞(p = 0.05), CAR-T时IPI评分较低(IPI 0-2) (p = 0.03),以及先前接受过自体干细胞移植(ASCT) (p = 0.03)。关于复发风险,只有cart前的低IPI (I/II) (HR: 1.73, 95% CI: 1.08-2.7, p = 0.04)和先前的ASCT (HR: 0.27, 95% CI: 0.08-0.9, p = 0.03)具有显著性。CAR-T输注后的中位随访为25个月(95% CI: 13-NA),中位PFS和OS分别为3.3个月(95% CI: 1.4-11)和9.2个月(95% CI: 6.1-NA)。 12个月PFS和OS分别为23%和43%(支持信息S1:图1A,B)。支持信息S1:图1显示了响应、PFS、OS和响应的单变量分析。中位DR为3.9个月(95% CI: 2-NA)。所有病例均观察到CAR-T扩增。在14例有持续数据的病例中,只有2例CAR-T在6个月和12个月时没有持续;然而,在所有病例中,在最后一次随访时都有B细胞发育不全。18例(90%)患者在car - t细胞治疗后出现了进展性疾病(PD), PD后的中位生存期为5.1个月(3.7个月,NR), 24个月的生存期为24.2%(9.6-61.1)。18例CAR-T后PD患者中有10例(56%)接受了后续治疗,中位PFS和OS分别为3.6(2.9 -未达到)和6.5(3.0 -未达到)个月(支持信息S1:图2C,D)。与CAR-T失败后仅接受最佳支持治疗的患者相比,这显示出明显的生存率提高趋势。与CAR-T失败后仅接受最佳支持治疗的患者相比(支持信息S1:图2和3)。从CAR-T进展到下一次治疗的中位时间为42.5 (IQR: 38-50.5;范围= 4-57)。在随后的治疗结果方面,5/10患者使用基于检查点抑制剂(派姆单抗、阿特唑单抗)、polatuzumab和他法西他马-来那度胺的方案获得了缓解(图1)。2例患者在放疗(n = 1)和派姆单抗单药治疗(n = 1)后获得了CR。一名患者接受阿特唑单抗-格非他单抗(1年固定疗程治疗)治疗后达到了PR,在2.5年后仍在持续。CAR-T后接受CPI和/或BiAb治疗的患者未观察到明显的免疫毒性。只有1例患者在接受4个周期的他法西他麦-来那度胺治疗后,接受了同种异体干细胞移植作为巩固,并在移植后6个月保持了疗效。CAR - t细胞治疗是一线治疗后早期(12个月)复发的LBCL患者或第二次复发患者的标准治疗,因为与其他治疗方案相比,CAR - t细胞治疗的有效率、DR和生存率都很高。大多数关键性试验包括弥漫性大b细胞淋巴瘤、转化滤泡性淋巴瘤和高级别b细胞淋巴瘤患者。然而,THRLBCL通常因其低发病率和独特的生物学特征而被排除在外。在这项研究中,我们报告了一个最大的现实世界THRLBCL接受CAR - t细胞治疗的队列,并提供了后续治疗策略的详细分析。Trujillo等人发表了第一批接受CAR-T治疗的THRLBCL患者(N = 9),采用这种t细胞重定向策略后的结果令人沮丧,6表明该患者群体中存在潜在的难治性行为。他们假设THRLBCL的CAR- t细胞衰竭似乎与获得性CAR- t细胞功能障碍有关,而不是CAR- t细胞增殖不良最近,Pophali等人报道了58例CAR - t细胞治疗的R/R THRLBCL患者的2年PFS和OS分别为29%和42%;在本研究中,ECOG-PS是唯一影响生存的因素。然而,除了这些有趣的数据之外,很少有研究关注这种罕见疾病的临床特征和CAR-T治疗的反应模式。与这些系列相似,我们研究中的患者预后较差,中位PFS和OS分别为3.3和9.2个月。有趣的是,达到CR的患者是那些在CAR-T细胞治疗时具有低LDH,早期和低风险IPI的患者。然而,不管对CAR-T的反应如何,20名患者中有18名出现了疾病进展,与之前提到的研究一致。引人注目的是,CAR-T复发后获救的患者的长期生存期与Pophali等人报道的非常相似,40%的患者在2年存活。考虑到高复发率,有效的后续策略似乎是这些长期结果的潜在原因。在我们的数据集中,在CAR-T复发后接受治疗的患者(N = 10)中,2年OS为44%,尽管大多数是早期复发(30% &lt;3个月,70% &lt;6个月),与CAR-T - 16后早期复发的R/R LBCL患者的结果相比,显着高于其他报告接受PD-1阻滞剂和双特异性抗体的患者表现出更好的结果,这支持了THRLBCL可能对PD-1阻断治疗有反应的假设,因为在恶性B细胞周围有异常高数量的表达pd - l1的肿瘤相关巨噬细胞和PD-1 + T细胞。5关于毒性、短期不良事件的发生率和严重程度,如CRS和ICANS,与关键试验和真实DLBCL数据相似。 17,18本研究的主要局限性是样本量小,以及缺乏集中的病理检查。然而,尽管存在这一缺陷,我们报告了CAR-T输注后的长期随访,并提供了后续治疗策略的见解,这在这种特殊情况下仍然是一个重要的知识缺口。总之,CAR - t细胞治疗THRLBCL患者的疗效明显低于LBCL患者。然而,对后续治疗的反应率令人鼓舞。鉴于检查点抑制剂或双特异性抗体在cart后情况下的结果,人们高度期待针对这类患者群体探索CAR-T联合策略的临床试验的发展。Mariana Bastos-Oreiro和Gloria Iacoboni设计研究,进行研究,收集,分析和解释数据,并撰写论文。Víctor N. garcsams进行了统计分析。Ana C. Caballero, Javier Delgado, Aitana Balaguer, Mi Kwon, Sonia Gonzalez de Villambrosia, Ana Jimenez-Ubieto, Rebeca Bailen和Alejandro Martín García-Sancho收集数据并进行研究。Pere Barba设计研究,构思并监督研究。Mariana Bastos-Oreiro:来自艾伯维(Abbvie)、百时美施贵宝(Bristol-Myers Squibb)、Kite/Gilead和诺华(Novartis)的酬金和/或旅行支持。Gloria Iacoboni:来自艾伯维(Abbvie)、阿斯利康(AstraZeneca)、Autolus、百时美施贵宝(Bristol-Myers Squibb)、Kite/Gilead、Miltenyi和诺华的酬金和/或旅行支持。Víctor N. garcsams:没有披露。Ana C. Caballero:不透露。哈维尔·德尔加多:没有披露。Aitana Balaguer:没有披露。米权:来自Kite/Gilead、Novartis的酬金和/或旅行支持。Sonia Gonzalez de Villambrosia:艾伯维(Abbvie)、罗氏(Roche)、Incyte、百时美施贵宝(Bristol-Myers Squibb)、Kite/Gilead、Miltenyi和诺华(Novartis)的酬金和/或旅行支持。Ana Jimenez-Ubieto:来自艾伯维(Abbvie)、罗氏(Roche)、英赛(Incyte)、Kite/吉利德(Gilead)、密天尼(Miltenyi)和诺华(Novartis)的酬金和/或差旅支持。Rebeca Bailen: Incyte、百时美施贵宝(Bristol-Myers Squibb)、Kite/Gilead、Miltenyi和诺华(Novartis)的酬金和/或差旅支持。Alejandro Martín García-Sancho:酬金和/或咨询费:罗氏、BMS、武田、杨森、Kyowa Kirin、Gilead/Kite、Incyte、Lilly、Miltenyi、Ideogen、Genmab、Abbvie、Sobi、Astra-Zeneca、GSK、Regeneron。Pere Barba:来自艾伯维(Abbvie)、阿斯利康(AstraZeneca)、Autolus、百时美施贵宝(Bristol-Myers Squibb)、Kite/Gilead、Miltenyi和诺华(Novartis)的酬金和/或差旅支持。这项研究没有得到资助。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
HemaSphere
HemaSphere Medicine-Hematology
CiteScore
6.10
自引率
4.50%
发文量
2776
审稿时长
7 weeks
期刊介绍: HemaSphere, as a publication, is dedicated to disseminating the outcomes of profoundly pertinent basic, translational, and clinical research endeavors within the field of hematology. The journal actively seeks robust studies that unveil novel discoveries with significant ramifications for hematology. In addition to original research, HemaSphere features review articles and guideline articles that furnish lucid synopses and discussions of emerging developments, along with recommendations for patient care. Positioned as the foremost resource in hematology, HemaSphere augments its offerings with specialized sections like HemaTopics and HemaPolicy. These segments engender insightful dialogues covering a spectrum of hematology-related topics, including digestible summaries of pivotal articles, updates on new therapies, deliberations on European policy matters, and other noteworthy news items within the field. Steering the course of HemaSphere are Editor in Chief Jan Cools and Deputy Editor in Chief Claire Harrison, alongside the guidance of an esteemed Editorial Board comprising international luminaries in both research and clinical realms, each representing diverse areas of hematologic expertise.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信