Pub Date : 2024-10-22DOI: 10.1182/bloodadvances.2024013677
Rohtesh S Mehta, Effie W Petersdorf, Tao Wang, Stephen R Spellman, Stephanie J Lee
Abstract: In 10/10 HLA-matched unrelated donor (MUD) hematopoietic cell transplantation (HCT) with calcineurin-inhibitor (CNI)-based prophylaxis, T-cell epitope DP-matched and permissive mismatched donors are associated with similar overall survival (OS) whereas donors with nonpermissive mismatches should be avoided. Younger unrelated donors are also favored over older donors. We explored outcomes associated with different combinations of DP-matching and donor age (dichotomized at 35 years) to further guide donor selection. Using a Center for International Blood and Marrow Transplant Research data set, we categorized 10 783 patients into 6 groups: DP-matched/younger donor (n = 1591), DP-matched/older donor (n = 526), permissive-mismatched/younger donor (n = 3845), permissive-mismatched/older donor (n = 1184), nonpermissive mismatched/younger donor (n = 2659), and nonpermissive mismatched/older donor (n = 978). We noted that younger donor age, rather than DP matching, was associated with better OS. Younger donors with permissive mismatches were associated with improved OS compared with older matched donors. Furthermore, younger donors with nonpermissive mismatches were associated with improved OS compared with older donors with permissive mismatches. Our study adds further information about the association of DP matching and donor age with HCT outcomes. Donor age should be prioritized over DP matching in patients undergoing 10/10 HLA-MUD with CNI prophylaxis. Among those with younger donors, permissive-mismatched or DP-matched donors are preferred over nonpermissive mismatched donors.
{"title":"Interplay between donor age and HLA-DP matching in 10/10 HLA-matched unrelated donor HCT.","authors":"Rohtesh S Mehta, Effie W Petersdorf, Tao Wang, Stephen R Spellman, Stephanie J Lee","doi":"10.1182/bloodadvances.2024013677","DOIUrl":"10.1182/bloodadvances.2024013677","url":null,"abstract":"<p><strong>Abstract: </strong>In 10/10 HLA-matched unrelated donor (MUD) hematopoietic cell transplantation (HCT) with calcineurin-inhibitor (CNI)-based prophylaxis, T-cell epitope DP-matched and permissive mismatched donors are associated with similar overall survival (OS) whereas donors with nonpermissive mismatches should be avoided. Younger unrelated donors are also favored over older donors. We explored outcomes associated with different combinations of DP-matching and donor age (dichotomized at 35 years) to further guide donor selection. Using a Center for International Blood and Marrow Transplant Research data set, we categorized 10 783 patients into 6 groups: DP-matched/younger donor (n = 1591), DP-matched/older donor (n = 526), permissive-mismatched/younger donor (n = 3845), permissive-mismatched/older donor (n = 1184), nonpermissive mismatched/younger donor (n = 2659), and nonpermissive mismatched/older donor (n = 978). We noted that younger donor age, rather than DP matching, was associated with better OS. Younger donors with permissive mismatches were associated with improved OS compared with older matched donors. Furthermore, younger donors with nonpermissive mismatches were associated with improved OS compared with older donors with permissive mismatches. Our study adds further information about the association of DP matching and donor age with HCT outcomes. Donor age should be prioritized over DP matching in patients undergoing 10/10 HLA-MUD with CNI prophylaxis. Among those with younger donors, permissive-mismatched or DP-matched donors are preferred over nonpermissive mismatched donors.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141619337","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-22DOI: 10.1182/bloodadvances.2024013508
Jean-Benoît Le Luduec, Theodota Kontopoulos, M Kazim Panjwani, Rosa Sottile, Hongtao Liu, Gesine Schäfer, Carolin Massalski, Vinzenz Lange, Katharine C Hsu
Abstract: CD94/NKG2A is a heterodimeric receptor commonly found on natural killer (NK) and T cells, and its interaction with its ligand HLA-E on adjacent cells leads to inhibitory signaling and cell suppression. We have identified several killer cell lectin-like receptor (KLR)C1 (NKG2A) single nucleotide polymorphisms (SNPs) that are associated with NKG2A expression on NK cells, CD8+ T cells, and Vγ9/Vδ2+ T cells. Additionally, due to strong linkage disequilibrium, polymorphisms in KLRC2 (NKG2C) and KLRK1 (NKG2D) are also associated with NKG2A surface density and frequency. NKG2A surface expression correlates with single-cell NK responsiveness, and NKG2A+ NK cell frequency is associated with total NK repertoire response and inhibitability, making the identification of SNPs responsible for expression and frequency important for predicting the innate immune response. Because HLA-E expression is dependent on HLA class I signal peptides, we analyzed the relationship between peptide abundance and HLA-E expression levels. Our findings revealed a strong association between peptide availability and HLA-E expression. We identified the HLA-C killer immunoglobulin-like receptor ligand epitope as a predictive marker for HLA-ABC expression, with the HLA-C1 epitope associated with high HLA-E expression and the HLA-C2 epitope associated with low HLA-E expression. The relationship between HLA-C epitopes and HLA-E expression was independent of HLA-E allotypes and HLA-B leader peptides. Although HLA-E expression showed no significant influence on NKG2A-mediated NK education, it did affect NK cell inhibition. In summary, these findings underscore the importance of NKG2A SNPs and HLA-C epitopes as predictive markers of NK cell phenotype and function and should be evaluated as prognostic markers for diseases that express high levels of HLA-E.
CD94/NKG2A是一种常见于NK细胞和T细胞上的异源二聚体受体,它与邻近细胞上的配体HLA-E相互作用,导致抑制性信号传导和细胞抑制。我们发现了几个与 NK 细胞、CD8+ T 细胞和 V9/V2+ T 细胞上 NKG2A 表达相关的 KLRC1(NKG2A)单核苷酸多态性(SNPs)。此外,由于强连锁不平衡,KLRC2(NKG2C)和 KLRK1(NKG2D)的多态性也与 NKG2A 的表面密度和频率有关。NKG2A 表面表达与单细胞 NK 反应性相关,而 NKG2A+ NK 细胞频率与总 NK 复合物反应和抑制性相关,因此鉴定导致表达和频率的 SNPs 对于预测先天性免疫反应非常重要。由于 HLA-E 的表达依赖于 HLA I 类信号肽,我们分析了肽丰度与 HLA-E 表达水平之间的关系。我们的研究结果表明,肽的可用性与 HLA-E 的表达之间存在密切联系。我们发现 HLA-C KIR 配体表位是 HLA-ABC 表达的预测标志物,HLA-C1 表位与 HLA-E 高表达相关,而 HLA-C2 表位与 HLA-E 低表达相关。HLA-C 表位与 HLA-E 表达之间的关系与 HLA-E 异型和 HLA-B 头肽无关。虽然 HLA-E 表达对 NKG2A 介导的 NK 教育没有显著影响,但它确实会影响 NK 细胞的抑制作用。总之,这些发现强调了 NKG2A SNPs 和 HLA-C 表位作为 NK 细胞表型和功能预测标志物的重要性,并应作为高水平 HLA-E 表达疾病的预后标志物进行评估。
{"title":"Polygenic polymorphism is associated with NKG2A repertoire and influences lymphocyte phenotype and function.","authors":"Jean-Benoît Le Luduec, Theodota Kontopoulos, M Kazim Panjwani, Rosa Sottile, Hongtao Liu, Gesine Schäfer, Carolin Massalski, Vinzenz Lange, Katharine C Hsu","doi":"10.1182/bloodadvances.2024013508","DOIUrl":"10.1182/bloodadvances.2024013508","url":null,"abstract":"<p><strong>Abstract: </strong>CD94/NKG2A is a heterodimeric receptor commonly found on natural killer (NK) and T cells, and its interaction with its ligand HLA-E on adjacent cells leads to inhibitory signaling and cell suppression. We have identified several killer cell lectin-like receptor (KLR)C1 (NKG2A) single nucleotide polymorphisms (SNPs) that are associated with NKG2A expression on NK cells, CD8+ T cells, and Vγ9/Vδ2+ T cells. Additionally, due to strong linkage disequilibrium, polymorphisms in KLRC2 (NKG2C) and KLRK1 (NKG2D) are also associated with NKG2A surface density and frequency. NKG2A surface expression correlates with single-cell NK responsiveness, and NKG2A+ NK cell frequency is associated with total NK repertoire response and inhibitability, making the identification of SNPs responsible for expression and frequency important for predicting the innate immune response. Because HLA-E expression is dependent on HLA class I signal peptides, we analyzed the relationship between peptide abundance and HLA-E expression levels. Our findings revealed a strong association between peptide availability and HLA-E expression. We identified the HLA-C killer immunoglobulin-like receptor ligand epitope as a predictive marker for HLA-ABC expression, with the HLA-C1 epitope associated with high HLA-E expression and the HLA-C2 epitope associated with low HLA-E expression. The relationship between HLA-C epitopes and HLA-E expression was independent of HLA-E allotypes and HLA-B leader peptides. Although HLA-E expression showed no significant influence on NKG2A-mediated NK education, it did affect NK cell inhibition. In summary, these findings underscore the importance of NKG2A SNPs and HLA-C epitopes as predictive markers of NK cell phenotype and function and should be evaluated as prognostic markers for diseases that express high levels of HLA-E.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141999421","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-22DOI: 10.1182/bloodadvances.2024013205
Nardjas Bettazova, Jana Senavova, Kristyna Kupcova, Dana Sovilj, Anezka Rajmonova, Ladislav Andera, Karla Svobodova, Adela Berkova, Zuzana Zemanova, Lenka Daumova, Vaclav Herman, Alexandra Dolníkova, R Eric Davis, Marek Trneny, Pavel Klener, Ondrej Havranek
Abstract: Besides many other mutations in known cancer driver genes, mantle cell lymphoma (MCL) is characterized by recurrent genetic alterations of important regulators of the phosphoinositol-3-kinase (PI3K) cascade including PIK3CA gains and PTEN losses. To evaluate the biological and functional consequences of these aberrations in MCL, we have introduced transgenic expression of PIK3CA (PIK3CA UP) and performed knockout/knockdown of PTEN gene (PTEN KO/KD) in 5 MCL cell lines. The modified cell lines were tested for associated phenotypes including dependence on upstream B-cell receptor (BCR) signaling (by an additional BCR knockout). PIK3CA overexpression decreased the dependence of the tested MCL on prosurvival signaling from BCR, decreased levels of oxidative phosphorylation, and increased resistance to 2-deoxy-glucose, a glycolysis inhibitor. Unchanged protein kinase B (AKT) phosphorylation status and unchanged sensitivity to a battery of PI3K inhibitors suggested that PIK3CA gain might affect MCL cells in AKT-independent manner. PTEN KO was associated with a more distinct phenotype: AKT hyperphosphorylation and overactivation, increased resistance to multiple inhibitors (most of the tested PI3K inhibitors, Bruton tyrosine kinase inhibitor ibrutinib, and BCL2 inhibitor venetoclax), increased glycolytic rates with resistance to 2-deoxy-glucose, and significantly decreased dependence on prosurvival BCR signaling. Our results suggest that the frequent aberrations of the PI3K pathway may rewire associated signaling with lower dependence on BCR signaling, better metabolic and hypoxic adaptation, and targeted therapy resistance in MCL.
{"title":"Impact of PIK3CA gain and PTEN loss on mantle cell lymphoma biology and sensitivity to targeted therapies.","authors":"Nardjas Bettazova, Jana Senavova, Kristyna Kupcova, Dana Sovilj, Anezka Rajmonova, Ladislav Andera, Karla Svobodova, Adela Berkova, Zuzana Zemanova, Lenka Daumova, Vaclav Herman, Alexandra Dolníkova, R Eric Davis, Marek Trneny, Pavel Klener, Ondrej Havranek","doi":"10.1182/bloodadvances.2024013205","DOIUrl":"10.1182/bloodadvances.2024013205","url":null,"abstract":"<p><strong>Abstract: </strong>Besides many other mutations in known cancer driver genes, mantle cell lymphoma (MCL) is characterized by recurrent genetic alterations of important regulators of the phosphoinositol-3-kinase (PI3K) cascade including PIK3CA gains and PTEN losses. To evaluate the biological and functional consequences of these aberrations in MCL, we have introduced transgenic expression of PIK3CA (PIK3CA UP) and performed knockout/knockdown of PTEN gene (PTEN KO/KD) in 5 MCL cell lines. The modified cell lines were tested for associated phenotypes including dependence on upstream B-cell receptor (BCR) signaling (by an additional BCR knockout). PIK3CA overexpression decreased the dependence of the tested MCL on prosurvival signaling from BCR, decreased levels of oxidative phosphorylation, and increased resistance to 2-deoxy-glucose, a glycolysis inhibitor. Unchanged protein kinase B (AKT) phosphorylation status and unchanged sensitivity to a battery of PI3K inhibitors suggested that PIK3CA gain might affect MCL cells in AKT-independent manner. PTEN KO was associated with a more distinct phenotype: AKT hyperphosphorylation and overactivation, increased resistance to multiple inhibitors (most of the tested PI3K inhibitors, Bruton tyrosine kinase inhibitor ibrutinib, and BCL2 inhibitor venetoclax), increased glycolytic rates with resistance to 2-deoxy-glucose, and significantly decreased dependence on prosurvival BCR signaling. Our results suggest that the frequent aberrations of the PI3K pathway may rewire associated signaling with lower dependence on BCR signaling, better metabolic and hypoxic adaptation, and targeted therapy resistance in MCL.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11497468/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141999419","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-22DOI: 10.1182/bloodadvances.2023012549
Nausheen Ahmed, William Wesson, Forat Lutfi, David L Porter, Veronika Bachanova, Loretta J Nastoupil, Miguel-Angel Perales, Richard T Maziarz, Jamie Brower, Gunjan L Shah, Andy I Chen, Olalekan O Oluwole, Stephen J Schuster, Michael R Bishop, Joseph P McGuirk, Peter A Riedell
Abstract: CD19-directed chimeric antigen receptor T-cell (CAR T) therapies, including axicabtagene ciloleucel (axi-cel), tisagenlecleucel (tisa-cel), and lisocabtagene maraleucel (liso-cel), have transformed the treatment landscape for B-cell non-Hodgkin lymphoma, showcasing significant efficacy but also highlighting toxicity risks such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). The US Food and Drug Administration has mandated patients remain close to the treatment center for 4 weeks as part of a Risk Evaluation and Mitigation Strategy to monitor and manage these toxicities, which, although cautious, may add to cost of care, be burdensome for patients and their families, and present challenges related to patient access and socioeconomic disparities. This retrospective study across 9 centers involving 475 patients infused with axi-cel, tisa-cel, and liso-cel from 2018 to 2023 aimed to assess CRS and ICANS onset and duration, as well as causes of nonrelapse mortality (NRM) in real-world CAR T recipients. Although differences were noted in the incidence and duration of CRS and ICANS between CAR T products, new-onset CRS and ICANS are exceedingly rare after 2 weeks after infusion (0% and 0.7% of patients, respectively). No new cases of CRS occurred after 2 weeks and a single case of new-onset ICANS occurred in the third week after infusion. NRM is driven by ICANS in the early follow-up period (1.1% until day 28) and then by infection through 3 months after infusion (1.2%). This study provides valuable insights into optimizing CAR T therapy monitoring, and our findings may provide a framework to reduce physical and financial constraints for patients.
CD19定向嵌合抗原受体T细胞(CAR T)疗法,包括axicabtagene ciloleucel(axi-cel)、tisagenlecleucel(tisa-cel)和lisocabtagene maraleucel(liso-cel)、改变了 B 细胞非霍奇金淋巴瘤(NHL)的治疗格局,展示了显著疗效,但也凸显了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)等毒性风险。作为风险评估和缓解策略的一部分,美国食品药品管理局规定患者必须在治疗中心就近治疗四周,以监测和管理这些毒性反应,尽管这很谨慎,但可能会增加治疗成本,给患者及其家庭带来负担,并带来与患者就诊和社会经济差异相关的挑战。这项横跨 9 个中心的回顾性研究涉及 2018 年至 2023 年期间输注 axi-cel、tisa-cel 和 liso-cel 的 475 例患者,旨在评估真实世界 CAR T 受者中 CRS 和 ICANS 的发病情况和持续时间,以及非复发死亡率(NRM)的原因。虽然CAR T产品之间的CRS和ICANS发病率和持续时间存在差异,但输注两周后新发CRS和ICANS的情况极为罕见(分别为0%和0.7%的患者)。输注两周后没有出现新的 CRS 病例,输注后第三周出现了一例新发 ICANS 病例。NRM在早期随访期间由ICANS引起(第28天前为1.1%),然后在输注后三个月内由感染引起(1.2%)。这项研究为优化 CAR T 疗法的监测提供了宝贵的见解,我们的研究结果可能为减少患者的身体和经济限制提供了一个框架。
{"title":"Optimizing the post-CAR T monitoring period in recipients of axicabtagene ciloleucel, tisagenlecleucel, and lisocabtagene maraleucel.","authors":"Nausheen Ahmed, William Wesson, Forat Lutfi, David L Porter, Veronika Bachanova, Loretta J Nastoupil, Miguel-Angel Perales, Richard T Maziarz, Jamie Brower, Gunjan L Shah, Andy I Chen, Olalekan O Oluwole, Stephen J Schuster, Michael R Bishop, Joseph P McGuirk, Peter A Riedell","doi":"10.1182/bloodadvances.2023012549","DOIUrl":"10.1182/bloodadvances.2023012549","url":null,"abstract":"<p><strong>Abstract: </strong>CD19-directed chimeric antigen receptor T-cell (CAR T) therapies, including axicabtagene ciloleucel (axi-cel), tisagenlecleucel (tisa-cel), and lisocabtagene maraleucel (liso-cel), have transformed the treatment landscape for B-cell non-Hodgkin lymphoma, showcasing significant efficacy but also highlighting toxicity risks such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). The US Food and Drug Administration has mandated patients remain close to the treatment center for 4 weeks as part of a Risk Evaluation and Mitigation Strategy to monitor and manage these toxicities, which, although cautious, may add to cost of care, be burdensome for patients and their families, and present challenges related to patient access and socioeconomic disparities. This retrospective study across 9 centers involving 475 patients infused with axi-cel, tisa-cel, and liso-cel from 2018 to 2023 aimed to assess CRS and ICANS onset and duration, as well as causes of nonrelapse mortality (NRM) in real-world CAR T recipients. Although differences were noted in the incidence and duration of CRS and ICANS between CAR T products, new-onset CRS and ICANS are exceedingly rare after 2 weeks after infusion (0% and 0.7% of patients, respectively). No new cases of CRS occurred after 2 weeks and a single case of new-onset ICANS occurred in the third week after infusion. NRM is driven by ICANS in the early follow-up period (1.1% until day 28) and then by infection through 3 months after infusion (1.2%). This study provides valuable insights into optimizing CAR T therapy monitoring, and our findings may provide a framework to reduce physical and financial constraints for patients.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141751122","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-22DOI: 10.1182/bloodadvances.2024014227
Innocent G Asiimwe, Marc Blockman, Larisa H Cavallari, Karen Cohen, Clint Cupido, Collet Dandara, Brittney H Davis, Barry Jacobson, Julie A Johnson, Mohammed Lamorde, Nita A Limdi, Jennie Morgan, Johannes P Mouton, Sarudzai Muyambo, Doreen Nakagaayi, Arinao Ndadza, Emmy Okello, Minoli A Perera, Elise Schapkaitz, Christine Sekaggya-Wiltshire, Jerome R Semakula, Gayle Tatz, Catriona Waitt, Guang Yang, Eunice J Zhang, Andrea L Jorgensen, Munir Pirmohamed
Abstract: Warfarin dose requirements are highly variable because of clinical and genetic factors. Although genetic variants influencing warfarin dose have been identified in European and East Asian populations, more work is needed to identify African-specific genetic variants to help optimize warfarin dosing. We performed genome-wide association studies (GWASs) in 4 African cohorts from Uganda, South Africa, and Zimbabwe, totaling 989 warfarin-treated participants who reached stable dose and had international normalized ratios within therapeutic ranges. We also included 2 African American cohorts recruited by the International Warfarin Pharmacogenetics Consortium (n = 316) and the University of Alabama at Birmingham (n = 199). After the GWAS, we performed standard error-weighted meta-analyses and then conducted stepwise conditional analyses to account for known loci in chromosomes 10 and 16. The genome-wide significance threshold was set at P < 5 × 10-8. The meta-analysis, comprising 1504 participants, identified 242 significant SNPs across 3 genomic loci, with 99.6% of these located within known loci on chromosomes 10 (top SNP: rs58800757, P = 4.27 × 10-13) and 16 (top SNP: rs9925964, P = 9.97 × 10-16). Adjustment for the VKORC1 SNP -1639G>A revealed an additional locus on chromosome 2 (top SNPs rs116057875/rs115254730/rs115240773, P = 3.64 × 10-8), implicating the MALL gene, that could indirectly influence warfarin response through interactions with caveolin-1. In conclusion, we reaffirmed the importance of CYP2C9 and VKORC1 in influencing warfarin dose requirements, and identified a new locus (MALL), that still requires direct evidence of biological plausibility.
{"title":"Meta-analysis of genome-wide association studies of stable warfarin dose in patients of African ancestry.","authors":"Innocent G Asiimwe, Marc Blockman, Larisa H Cavallari, Karen Cohen, Clint Cupido, Collet Dandara, Brittney H Davis, Barry Jacobson, Julie A Johnson, Mohammed Lamorde, Nita A Limdi, Jennie Morgan, Johannes P Mouton, Sarudzai Muyambo, Doreen Nakagaayi, Arinao Ndadza, Emmy Okello, Minoli A Perera, Elise Schapkaitz, Christine Sekaggya-Wiltshire, Jerome R Semakula, Gayle Tatz, Catriona Waitt, Guang Yang, Eunice J Zhang, Andrea L Jorgensen, Munir Pirmohamed","doi":"10.1182/bloodadvances.2024014227","DOIUrl":"10.1182/bloodadvances.2024014227","url":null,"abstract":"<p><strong>Abstract: </strong>Warfarin dose requirements are highly variable because of clinical and genetic factors. Although genetic variants influencing warfarin dose have been identified in European and East Asian populations, more work is needed to identify African-specific genetic variants to help optimize warfarin dosing. We performed genome-wide association studies (GWASs) in 4 African cohorts from Uganda, South Africa, and Zimbabwe, totaling 989 warfarin-treated participants who reached stable dose and had international normalized ratios within therapeutic ranges. We also included 2 African American cohorts recruited by the International Warfarin Pharmacogenetics Consortium (n = 316) and the University of Alabama at Birmingham (n = 199). After the GWAS, we performed standard error-weighted meta-analyses and then conducted stepwise conditional analyses to account for known loci in chromosomes 10 and 16. The genome-wide significance threshold was set at P < 5 × 10-8. The meta-analysis, comprising 1504 participants, identified 242 significant SNPs across 3 genomic loci, with 99.6% of these located within known loci on chromosomes 10 (top SNP: rs58800757, P = 4.27 × 10-13) and 16 (top SNP: rs9925964, P = 9.97 × 10-16). Adjustment for the VKORC1 SNP -1639G>A revealed an additional locus on chromosome 2 (top SNPs rs116057875/rs115254730/rs115240773, P = 3.64 × 10-8), implicating the MALL gene, that could indirectly influence warfarin response through interactions with caveolin-1. In conclusion, we reaffirmed the importance of CYP2C9 and VKORC1 in influencing warfarin dose requirements, and identified a new locus (MALL), that still requires direct evidence of biological plausibility.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11493193/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142008336","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-22DOI: 10.1182/bloodadvances.2024013170
Anders Lindholm Sørensen, Vibe Skov, Lasse Kjær, Mads Emil Bjørn, Christina Schjellerup Eickhardt-Dalbøge, Morten Kranker Larsen, Claus H Nielsen, Carsten Thomsen, Lise Mette Rahbek Gjerdrum, Trine Alma Knudsen, Christina Ellervik, Ulrik Malthe Overgaard, Christen Lykkegaard Andersen, Hans Hasselbalch
Abstract: We report the 2-year end-of-study results from the phase 2 COMBI II clinical trial investigating the combination treatment of ruxolitinib and low-dose pegylated interferon alfa-2a in patients with newly diagnosed polycythemia vera (PV). The primary outcome was safety and key secondary endpoints were efficacy, based on hematologic parameters, quality-of-life measurements, and JAK2V617F variant allele frequency (VAF). We used the 2013 European LeukemiaNet and International Working Group-Myeloproliferative Neoplasms Research remission criteria. The remission criteria included remissions in symptoms, splenomegaly, peripheral blood counts, and bone marrow. We included 25 patients with PV with a median age of 70 years; 5 of those had prior thromboembolic events and 3 had computed tomography-verified splenomegaly. Two patients stopped both study drugs; 1 of these due to progression to post-PV myelofibrosis, the only one with a grade 3 infection. No events of herpes zoster infections were observed. None of the patients discontinued treatment due to psychiatric symptoms. The peripheral blood cell count remission rate was 92% at 24 months. Using the 2013 European LeukemiaNet and International Working Group-Myeloproliferative Neoplasms Research remission criteria, 14 (56%) achieved remission at 24 months; 3 (12%) achieved complete remission and 11 (44%) achieved partial remission. The following items from the Myeloproliferative Neoplasm Symptom Total Symptom Score were significantly reduced: abdominal discomfort, night sweats, itching, and bone pain. The median JAK2V617F VAF decreased from 47% (95% confidence interval [CI], 35-59) to 7% (95% CI, 3-15), and 60% of patients achieved molecular remission. In conclusion, combination treatment improved cell counts; bone marrow cellularity, and fibrosis; and decreased JAK2V617F VAF; with acceptable toxicity in patients with PV. The trial was registered at www.clinicaltrialsregister.eu as #EudraCT2018-004150-13.
{"title":"Combination therapy with ruxolitinib and pegylated interferon alfa-2a in newly diagnosed patients with polycythemia vera.","authors":"Anders Lindholm Sørensen, Vibe Skov, Lasse Kjær, Mads Emil Bjørn, Christina Schjellerup Eickhardt-Dalbøge, Morten Kranker Larsen, Claus H Nielsen, Carsten Thomsen, Lise Mette Rahbek Gjerdrum, Trine Alma Knudsen, Christina Ellervik, Ulrik Malthe Overgaard, Christen Lykkegaard Andersen, Hans Hasselbalch","doi":"10.1182/bloodadvances.2024013170","DOIUrl":"10.1182/bloodadvances.2024013170","url":null,"abstract":"<p><strong>Abstract: </strong>We report the 2-year end-of-study results from the phase 2 COMBI II clinical trial investigating the combination treatment of ruxolitinib and low-dose pegylated interferon alfa-2a in patients with newly diagnosed polycythemia vera (PV). The primary outcome was safety and key secondary endpoints were efficacy, based on hematologic parameters, quality-of-life measurements, and JAK2V617F variant allele frequency (VAF). We used the 2013 European LeukemiaNet and International Working Group-Myeloproliferative Neoplasms Research remission criteria. The remission criteria included remissions in symptoms, splenomegaly, peripheral blood counts, and bone marrow. We included 25 patients with PV with a median age of 70 years; 5 of those had prior thromboembolic events and 3 had computed tomography-verified splenomegaly. Two patients stopped both study drugs; 1 of these due to progression to post-PV myelofibrosis, the only one with a grade 3 infection. No events of herpes zoster infections were observed. None of the patients discontinued treatment due to psychiatric symptoms. The peripheral blood cell count remission rate was 92% at 24 months. Using the 2013 European LeukemiaNet and International Working Group-Myeloproliferative Neoplasms Research remission criteria, 14 (56%) achieved remission at 24 months; 3 (12%) achieved complete remission and 11 (44%) achieved partial remission. The following items from the Myeloproliferative Neoplasm Symptom Total Symptom Score were significantly reduced: abdominal discomfort, night sweats, itching, and bone pain. The median JAK2V617F VAF decreased from 47% (95% confidence interval [CI], 35-59) to 7% (95% CI, 3-15), and 60% of patients achieved molecular remission. In conclusion, combination treatment improved cell counts; bone marrow cellularity, and fibrosis; and decreased JAK2V617F VAF; with acceptable toxicity in patients with PV. The trial was registered at www.clinicaltrialsregister.eu as #EudraCT2018-004150-13.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11526083/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142008368","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-22DOI: 10.1182/bloodadvances.2023011540
Maja Jankovic, William W L Poon, Cristobal Gonzales-Losada, Gabriela Galicia Vazquez, Bahram Sharif-Askari, Yi Ding, Constance Craplet-Desombre, Alexandru Ilie, Jiantao Shi, Yongjie Wang, Ashok Kumar Jayavelu, Alexandre Orthwein, François Émile Mercier
Abstract: For several decades, induction therapy with nucleoside analogs, in particular cytarabine (Ara-C) and, to a lesser extent, fludarabine, has been the standard of care for patients diagnosed with acute myeloid leukemia (AML). However, the antitumor efficacy of nucleoside analogs is often limited by intrinsic and acquired drug resistance, thereby leading to poor therapeutic response and suboptimal clinical outcomes. In this study, we used genome-wide CRISPR-based pharmacogenomic screening to map the genetic factors that modulate the response to nucleoside analogs in AML and identified the E3 ubiquitin ligase, Herc1, as a key modulator of Ara-C response in mouse AML models driven by the KMT2A/MLLT3 fusion or by the constitutive coexpression of Hoxa9 and Meis1, both in vitro and in vivo. Loss of HERC1 enhanced nucleoside analog-induced cell death in both murine and human AML cell lines by compromising cell cycle progression. In-depth proteomic analysis and subsequent validation identified deoxycytidine kinase as a novel target of Herc1 in both mouse AML models. We observed that HERC1 is overexpressed in AML when compared with other cancer types and that higher HERC1 expression was associated with shorter overall survival in patients with AML in the The Cancer Gene Atlas program (TCGA) and BEAT-AML cohorts. Collectively, this study highlights the importance of HERC1 in the response of AML cells to nucleoside analogs, thereby establishing this E3 ubiquitin ligase as a novel predictive biomarker and potential therapeutic target for the treatment of AML.
{"title":"The E3 ubiquitin ligase Herc1 modulates the response to nucleoside analogs in acute myeloid leukemia.","authors":"Maja Jankovic, William W L Poon, Cristobal Gonzales-Losada, Gabriela Galicia Vazquez, Bahram Sharif-Askari, Yi Ding, Constance Craplet-Desombre, Alexandru Ilie, Jiantao Shi, Yongjie Wang, Ashok Kumar Jayavelu, Alexandre Orthwein, François Émile Mercier","doi":"10.1182/bloodadvances.2023011540","DOIUrl":"10.1182/bloodadvances.2023011540","url":null,"abstract":"<p><strong>Abstract: </strong>For several decades, induction therapy with nucleoside analogs, in particular cytarabine (Ara-C) and, to a lesser extent, fludarabine, has been the standard of care for patients diagnosed with acute myeloid leukemia (AML). However, the antitumor efficacy of nucleoside analogs is often limited by intrinsic and acquired drug resistance, thereby leading to poor therapeutic response and suboptimal clinical outcomes. In this study, we used genome-wide CRISPR-based pharmacogenomic screening to map the genetic factors that modulate the response to nucleoside analogs in AML and identified the E3 ubiquitin ligase, Herc1, as a key modulator of Ara-C response in mouse AML models driven by the KMT2A/MLLT3 fusion or by the constitutive coexpression of Hoxa9 and Meis1, both in vitro and in vivo. Loss of HERC1 enhanced nucleoside analog-induced cell death in both murine and human AML cell lines by compromising cell cycle progression. In-depth proteomic analysis and subsequent validation identified deoxycytidine kinase as a novel target of Herc1 in both mouse AML models. We observed that HERC1 is overexpressed in AML when compared with other cancer types and that higher HERC1 expression was associated with shorter overall survival in patients with AML in the The Cancer Gene Atlas program (TCGA) and BEAT-AML cohorts. Collectively, this study highlights the importance of HERC1 in the response of AML cells to nucleoside analogs, thereby establishing this E3 ubiquitin ligase as a novel predictive biomarker and potential therapeutic target for the treatment of AML.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11497402/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141878391","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-22DOI: 10.1182/bloodadvances.2024013685
Fieke W Hoff, William G Blum, Ying Huang, Rina Li Welkie, Ronan T Swords, Elie Traer, Eytan M Stein, Tara L Lin, Kellie J Archer, Prapti A Patel, Robert H Collins, Maria R Baer, Vu H Duong, Martha L Arellano, Wendy Stock, Olatoyosi Odenike, Robert L Redner, Tibor Kovacsovics, Michael W Deininger, Joshua F Zeidner, Rebecca L Olin, Catherine C Smith, James M Foran, Gary J Schiller, Emily K Curran, Kristin L Koenig, Nyla A Heerema, Timothy Chen, Molly Martycz, Mona Stefanos, Sonja G Marcus, Leonard Rosenberg, Brian J Druker, Ross L Levine, Amy Burd, Ashley O Yocum, Uma M Borate, Alice S Mims, John C Byrd, Yazan F Madanat
Abstract: Although the 2022 European LeukemiaNet (ELN) acute myeloid leukemia (AML) risk classification reliably predicts outcomes in younger patients treated with intensive chemotherapy, it is unclear whether it applies to adults ≥60 years treated with lower-intensity treatment (LIT). We aimed to test the prognostic impact of ELN risk in patients with newly diagnosed (ND) AML aged ≥60 years given LIT and to further refine risk stratification for these patients. A total of 595 patients were included: 11% had favorable-, 11% intermediate-, and 78% had adverse-risk AML. ELN risk was prognostic for overall survival (OS) (P < .001) but did not stratify favorable- from intermediate-risk (P = .71). Within adverse-risk AML, the impact of additional molecular abnormalities was further evaluated. Multivariable analysis was performed on a training set (n = 316) and identified IDH2 mutation as an independent favorable prognostic factor, and KRAS, MLL2, and TP53 mutations as unfavorable (P < .05). A "mutation score" was calculated for each combination of these mutations, assigning adverse-risk patients to 2 risk groups: -1 to 0 points ("Beat-AML intermediate") vs 1+ points ("Beat-AML adverse"). In the final refined risk classification, ELN favorable- and intermediate-risk were combined into a newly defined "Beat-AML favorable-risk" group, in addition to mutation scoring within the ELN adverse-risk group. This approach redefines risk for older patients with ND AML and proposes refined Beat-AML risk groups with improved discrimination for OS (2-year OS, 48% vs 33% vs 11%, respectively; P < .001), providing patients and providers additional information for treatment decision-making.
{"title":"Beat-AML 2024 ELN-refined risk stratification for older adults with newly diagnosed AML given lower-intensity therapy.","authors":"Fieke W Hoff, William G Blum, Ying Huang, Rina Li Welkie, Ronan T Swords, Elie Traer, Eytan M Stein, Tara L Lin, Kellie J Archer, Prapti A Patel, Robert H Collins, Maria R Baer, Vu H Duong, Martha L Arellano, Wendy Stock, Olatoyosi Odenike, Robert L Redner, Tibor Kovacsovics, Michael W Deininger, Joshua F Zeidner, Rebecca L Olin, Catherine C Smith, James M Foran, Gary J Schiller, Emily K Curran, Kristin L Koenig, Nyla A Heerema, Timothy Chen, Molly Martycz, Mona Stefanos, Sonja G Marcus, Leonard Rosenberg, Brian J Druker, Ross L Levine, Amy Burd, Ashley O Yocum, Uma M Borate, Alice S Mims, John C Byrd, Yazan F Madanat","doi":"10.1182/bloodadvances.2024013685","DOIUrl":"10.1182/bloodadvances.2024013685","url":null,"abstract":"<p><strong>Abstract: </strong>Although the 2022 European LeukemiaNet (ELN) acute myeloid leukemia (AML) risk classification reliably predicts outcomes in younger patients treated with intensive chemotherapy, it is unclear whether it applies to adults ≥60 years treated with lower-intensity treatment (LIT). We aimed to test the prognostic impact of ELN risk in patients with newly diagnosed (ND) AML aged ≥60 years given LIT and to further refine risk stratification for these patients. A total of 595 patients were included: 11% had favorable-, 11% intermediate-, and 78% had adverse-risk AML. ELN risk was prognostic for overall survival (OS) (P < .001) but did not stratify favorable- from intermediate-risk (P = .71). Within adverse-risk AML, the impact of additional molecular abnormalities was further evaluated. Multivariable analysis was performed on a training set (n = 316) and identified IDH2 mutation as an independent favorable prognostic factor, and KRAS, MLL2, and TP53 mutations as unfavorable (P < .05). A \"mutation score\" was calculated for each combination of these mutations, assigning adverse-risk patients to 2 risk groups: -1 to 0 points (\"Beat-AML intermediate\") vs 1+ points (\"Beat-AML adverse\"). In the final refined risk classification, ELN favorable- and intermediate-risk were combined into a newly defined \"Beat-AML favorable-risk\" group, in addition to mutation scoring within the ELN adverse-risk group. This approach redefines risk for older patients with ND AML and proposes refined Beat-AML risk groups with improved discrimination for OS (2-year OS, 48% vs 33% vs 11%, respectively; P < .001), providing patients and providers additional information for treatment decision-making.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11497398/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141900970","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-22DOI: 10.1182/bloodadvances.2024013917
Warren B Fingrut, Andromachi Scaradavou, Juliet N Barker
{"title":"Accurate analysis of race/ethnicity and socioeconomic status.","authors":"Warren B Fingrut, Andromachi Scaradavou, Juliet N Barker","doi":"10.1182/bloodadvances.2024013917","DOIUrl":"10.1182/bloodadvances.2024013917","url":null,"abstract":"","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141909754","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-22DOI: 10.1182/bloodadvances.2024013726
Vincent Camus, Roch Houot, Gabriel Brisou, Benoit Tessoulin, Sébastien Bailly, Pierre Sesques, Justine Decroocq, Daphné Krzisch, Lucie Oberic, François Lemonnier, Krimo Bouabdallah, Arnaud Campidelli, Ledraa Tounes, Julie Abraham, Charles Herbaux, Franck Morschhauser, Gandhi Laurent Damaj, Stéphanie Guidez, Sylvain Carras, Luc-Matthieu Fornecker, Sylvain Choquet, Olivier Hermine, Jérome Paillassa, Adrien Chauchet, Olivier Casasnovas, Laurianne Drieu La Rochelle, Cristina Castilla-Llorente, Magalie Joris, Vivien Dupont, Alexandra Marquet, Steven Le Gouill, Fabrice Jardin
Abstract: Tafasitamab plus lenalidomide (TAFA-LEN) treatment relevance pre- or post-anti-CD19 chimeric antigen receptor (CAR) T-cell therapy is debated. We analyzed patients with large B-cell lymphoma in the DESCAR-T registry treated with axi[1]cel or tisa-cel in ≥3rd line and TAFA-LEN before (n = 15, "TL-pre-CAR-T" set) or directly after (n = 52, "TL-post-CAR-T" set) CAR T-cell therapy. We compared TAFA-LEN v. other treatments using inverse probability weighting in the TL-post-CAR[1]T set. In the TL-post-CAR-T set, the median progression-free survival (mPFS), overall survival (mOS), and duration of response (mDOR) since the first treatment for progression (mPFS2/mOS2/mDOR2) were 3, 4.7, and 8.1 months, respectively. The best overall response rate (bORR) and best complete response rate (bCRR) after TAFA-LEN were 13.5% and 7.7%, respectively. Outcomes were better for patients who relapsed >6 months after CAR T-cell therapy (mPFS2: 5.6 vs 2 months, P = .0138; mOS2: not reached vs 3.8 months, P = .0034). The bORR and bCRR between TAFA-LEN and other treatments were 20.6% vs 24.9% and 11.6% vs 15.6%, respectively. Outcomes were similar between TAFA-LEN and other treatments (mPFS2: 2.9 vs 2.4 months, P = .91; mOS2: 3.3 vs 5.5 months, P = .06). In an exploratory analysis of the TL-pre-CAR-T set, the median TAFA-LEN treatment duration before CAR-T was 3.7 months with no patient becoming CD19 negative. The bORR, bCRR, 6- month PFS, and OS rates after CAR T-cell infusion were 45.5%, 36.4%, 20.1%, and 58.2%, respectively. Neither TAFA-LEN nor comparative salvage treatment improved outcomes for patients relapsing after CAR T-cell therapy.
塔法司他单抗加来那度胺(TAFA-LEN)治疗与抗CD-19 CAR T细胞治疗前或治疗后的相关性目前还存在争议。我们分析了 DESCAR-T 登记的大 B 细胞淋巴瘤患者,这些患者在接受 CAR T 细胞治疗前(n=15,"TL-pre-CAR-T "组)或直接接受 CAR T 细胞治疗后(n=52,"TL-post-CAR-T "组)接受了axi-cel 或 tisa-cel ≥3线(L3+)和 TAFA-LEN 治疗。在TL-post-CAR-T组中,我们使用反概率加权法比较了TAFA-LEN与其他治疗方法。在 "TL-post-CAR-T "组中,中位随访时间(mFUD)为 7 个月,中位无进展生存期、总生存期和自首次治疗进展以来的反应持续时间(mPFS2/mOS2/mDOR2)分别为 3 个月、4.7 个月和 8.1 个月。TAFA-LEN治疗后的最佳总反应率(bORR)和最佳完全反应率(bCRR)分别为13.5%和7.7%。CAR T细胞治疗后6个月以上复发的患者疗效更好(mPFS2:5.6个月 vs. 2个月,p=0.0138;mOS2:未达到 vs. 3.8个月,p=0.0034)。TAFA-LEN和其他治疗方法的bORR和bCRR分别为20.6% vs. 24.9%和11.6% vs. 15.6%。TAFA-LEN与其他疗法的疗效相似(mPFS2:2.9 [2-2.6] 个月 vs. 2.4 [1.5-4.2] 个月,p=0.91;mOS2:3.3 [1.8-6.4] 个月 vs. 5.5 [4.4-6.3] 个月,p=0.06)。在对TL-pre-CAR-T组(CAR-T细胞后mFUD:2.8个月)的探索性分析中,CAR-T前的中位TAFA-LEN治疗持续时间为3.7个月,没有患者报告CD19阴性。CAR T 细胞输注后的 bORR、bCRR、6 个月 PFS 和 OS 率分别为 45.5%、36.4%、20.1% 和 58.2%。TAFA-LEN和其他挽救治疗都没有改善CAR T细胞治疗后复发患者的预后。
{"title":"Outcome of patients with large B-cell lymphoma treated with tafasitamab plus lenalidomide either before or after CAR T-cell therapy.","authors":"Vincent Camus, Roch Houot, Gabriel Brisou, Benoit Tessoulin, Sébastien Bailly, Pierre Sesques, Justine Decroocq, Daphné Krzisch, Lucie Oberic, François Lemonnier, Krimo Bouabdallah, Arnaud Campidelli, Ledraa Tounes, Julie Abraham, Charles Herbaux, Franck Morschhauser, Gandhi Laurent Damaj, Stéphanie Guidez, Sylvain Carras, Luc-Matthieu Fornecker, Sylvain Choquet, Olivier Hermine, Jérome Paillassa, Adrien Chauchet, Olivier Casasnovas, Laurianne Drieu La Rochelle, Cristina Castilla-Llorente, Magalie Joris, Vivien Dupont, Alexandra Marquet, Steven Le Gouill, Fabrice Jardin","doi":"10.1182/bloodadvances.2024013726","DOIUrl":"10.1182/bloodadvances.2024013726","url":null,"abstract":"<p><strong>Abstract: </strong>Tafasitamab plus lenalidomide (TAFA-LEN) treatment relevance pre- or post-anti-CD19 chimeric antigen receptor (CAR) T-cell therapy is debated. We analyzed patients with large B-cell lymphoma in the DESCAR-T registry treated with axi[1]cel or tisa-cel in ≥3rd line and TAFA-LEN before (n = 15, \"TL-pre-CAR-T\" set) or directly after (n = 52, \"TL-post-CAR-T\" set) CAR T-cell therapy. We compared TAFA-LEN v. other treatments using inverse probability weighting in the TL-post-CAR[1]T set. In the TL-post-CAR-T set, the median progression-free survival (mPFS), overall survival (mOS), and duration of response (mDOR) since the first treatment for progression (mPFS2/mOS2/mDOR2) were 3, 4.7, and 8.1 months, respectively. The best overall response rate (bORR) and best complete response rate (bCRR) after TAFA-LEN were 13.5% and 7.7%, respectively. Outcomes were better for patients who relapsed >6 months after CAR T-cell therapy (mPFS2: 5.6 vs 2 months, P = .0138; mOS2: not reached vs 3.8 months, P = .0034). The bORR and bCRR between TAFA-LEN and other treatments were 20.6% vs 24.9% and 11.6% vs 15.6%, respectively. Outcomes were similar between TAFA-LEN and other treatments (mPFS2: 2.9 vs 2.4 months, P = .91; mOS2: 3.3 vs 5.5 months, P = .06). In an exploratory analysis of the TL-pre-CAR-T set, the median TAFA-LEN treatment duration before CAR-T was 3.7 months with no patient becoming CD19 negative. The bORR, bCRR, 6- month PFS, and OS rates after CAR T-cell infusion were 45.5%, 36.4%, 20.1%, and 58.2%, respectively. Neither TAFA-LEN nor comparative salvage treatment improved outcomes for patients relapsing after CAR T-cell therapy.</p>","PeriodicalId":9228,"journal":{"name":"Blood advances","volume":null,"pages":null},"PeriodicalIF":7.4,"publicationDate":"2024-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142008337","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}