chimeric antigen receptor T-cell

嵌合抗原受体 T 细胞
  • 文章类型: English Abstract
    Objective: Exploring the efficacy and safety of bridging blinatumomab (BiTE) in combination with chimeric antigen receptor T (CAR-T) cell therapy for the treatment of adult patients with acute B-cell lymphoblastic leukemia (B-ALL) . Methods: Clinical data from 36 adult B-ALL patients treated at the First Affiliated Hospital of Suzhou University from August 2018 to May 2023 were retrospectively analyzed. A total of 36 cases were included: 18 men and 18 women. The median age was 43.5 years (21-72 years). Moreover, 21 cases of Philadelphia chromosome-positive acute lymphoblastic leukemia were reported, and 16 of these cases were relapsed or refractory. Eighteen patients underwent blinatumomab bridging followed by CAR-T cell therapy, and 18 patients received CAR-T cell therapy. This study analyzed the efficacy and safety of treatment in two groups of patients. Results: In the BiTE bridge-to-CAR-T group, 16 patients achieved complete remission (CR) after BiTE immunotherapy, with a CR rate of 88.9%. One month after bridging CAR-T therapy, bone marrow examination showed a CR rate of 100.0%, and the minimal residual disease (MRD) negativity rate was higher than the nonbridging therapy group (94.4% vs. 61.1%, Fisher, P=0.041). The incidence of cytokine release syndrome and other adverse reactions in the BiTE bridge-to-CAR-T group was lower than that in the nonbridging therapy group (11.1% vs. 50.0%, Fisher, P=0.027). The follow-up reveals that 13 patients continued to maintain MRD negativity, and five patients experienced relapse 8.40 months (2.57-10.20 months) after treatment. Two of five patients with relapse achieved CR after receiving the second CAR-T cell therapy. In the nonbridging therapy group, 10 patients maintained continuous MRD negativity, 7 experienced relapse, and 6 died. The 1 year overall survival rate in the BiTE bridge-to-CAR-T group was higher than that in the nonbridging therapy group, with a statistically significant difference at the 0.1 level (88.9%±10.5% vs. 66.7%±10.9%, P=0.091) . Conclusion: BiTE bridging CAR-T cell therapy demonstrates excellent efficacy in adult B-ALL treatment, with a low recent recurrence rate and ongoing assessment of long-term efficacy during follow-up.
    目的: 探讨贝林妥欧单抗桥接嵌合抗原受体T(CAR-T)细胞疗法治疗急性B淋巴细胞白血病(B-ALL)患者的疗效及安全性。 方法: 回顾性分析2018年8月至2023年5月在苏州大学附属第一医院住院治疗的36例成人B-ALL患者的临床资料。男18例,女18例,中位年龄为43.5(21~72)岁。其中费城染色体阳性急性淋巴细胞白血病(Ph(+)ALL)21例,复发/难治16例。18例患者接受贝林妥欧单抗桥接CAR-T细胞治疗,18例患者仅接受CAR-T细胞治疗,分析两组患者的疗效及安全性。 结果: 贝林妥欧单抗桥接CAR-T组中16例患者贝林妥欧单抗治疗后获得完全缓解(CR),CR率88.9%。桥接CAR-T治疗1个月后复查骨髓象,CR率为100.0%,微小残留病(MRD)阴性率高于未接受贝林妥欧单抗桥接治疗组(94.4%对61.1%,Fisher,P=0.041)。贝林妥欧单抗桥接CAR-T组患者细胞因子释放综合征(CRS)及其他不良反应发生率低于未接受贝林妥欧单抗桥接治疗组(11.1%对50.0%,Fisher,P=0.027)。截至随访终点,贝林妥欧单抗桥接CAR-T组中13例患者持续MRD阴性状态,5例患者(包含2例经贝林妥欧单抗治疗无效的病例)在治疗后2.57~10.20个月复发,2例复发患者后续接受第二次CAR-T细胞治疗后达CR。未接受贝林妥欧单抗桥接治疗组中10例患者持续MRD阴性状态,7例复发,6例死亡。贝林妥欧单抗桥接CAR-T组患者1年总生存率高于未接受桥接治疗组,差异在0.1水平有统计学意义[(88.9±10.5)%对(66.7±10.9)%,P=0.091]。 结论: 贝林妥欧单抗桥接CAR-T细胞治疗成人B-ALL疗效及安全性值得肯定,清除肿瘤残留效果好,近期疗效复发率低,不良反应少。.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    家庭住院代表了传统住院的替代方案,为血液学患者提供可比的临床安全性。家庭治疗可以从静脉注射抗生素到更复杂的情况,如造血干细胞移植和嵌合抗原受体T细胞治疗后早期的护理。从常规住院早期出院是可行的,并有助于减少医院资源和等待名单。多学科小组的协调努力,包括血液学家,护士,药剂师,确保患者的安全和护理的连续性。传统的家庭住院模式依赖于家访和与医生和护士的电话咨询。然而,使用电子健康技术,比如MY-Meedura,可以加强沟通和监控,从而改善患者的治疗效果,无需额外费用。临床药师积极参与家庭住院计划至关重要,不仅为了药物的适当后勤管理,而且为了确保其适当性,优化治疗,来自团队和患者的地址查询,促进坚持。总之,实施造血干细胞移植和嵌合抗原受体T细胞治疗家庭住院计划,使用电子健康工具和多学科护理模式,可以优化患者护理并提高生活质量,而不会增加医疗保健成本。
    Home hospitalization represents an alternative to traditional hospitalization, providing comparable clinical safety for hematological patients. At-home therapies can range from the delivery of intravenous antibiotics to more complex scenarios, such as the care during the early period after hematopoietic stem cell transplantation and chimeric antigen receptor T-cell therapy. Early discharge from conventional hospitalization is feasible and helps reduce hospital resources and waiting lists. The coordinated efforts of multidisciplinary teams, including hematologists, nurses, and pharmacists, ensure patient safety and continuity of care. The traditional model of home hospitalization relies on home visits and telephone consultations with physicians and nurses. However, the use of eHealth technologies, such as MY-Medula, can enhance communication and monitoring, and thereby improve patient outcomes with no additional costs. The active involvement of a clinical pharmacist in home hospitalization programs is essential, not only for the proper logistical management of the medication but also to ensure its appropriateness, optimize treatment, address queries from the team and patients, and promote adherence. In conclusion, the implementation of hematopoietic stem cell transplantation and chimeric antigen receptor T-cell therapy home hospitalization programs that use both an eHealth tool and a multidisciplinary care model can optimize patient care and improve quality of life without increasing healthcare costs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    造血干细胞移植(HSCT)后B细胞急性淋巴细胞白血病(B-ALL)复发的患者的一年生存率约为30%。在同种异体HSCT后经历复发的患者在获得自体CAR-T产品时经常遇到困难。我们在我们中心进行了一项研究,涉及14名患者,他们在2019年8月至2023年5月期间接受供体衍生的CAR-T治疗HSCT后复发B-ALL。结果显示,CR/CRi率为78.6%(11/14),GVHD率为21.4%(3/14),1年总生存率(OS)为56%。CAR-T治疗后9例患者的骨髓供体细胞嵌合体减少。死亡原因主要为疾病进展和感染。进一步分析表明,GVHD(HR7.224,95%CI1.42-36.82,P=0.017)和30天血小板恢复(HR6.807,95%CI1.61-28.83,P=0.009)与CAR-T治疗后的OS显着相关。根据调查结果,我们得出的结论是,供体来源的CAR-T细胞可有效治疗HSCT后复发的B-ALL患者。此外,GVHD和血小板恢复不良影响OS,但需要用更大的样本量进一步验证。
    The one-year survival rate for patients experiencing a relapse of B-cell acute lymphocytic leukemia (B-ALL) following hematopoietic stem cell transplantation (HSCT) is approximately 30%. Patients experiencing a relapse after allogeneic HSCT frequently encounter difficulties in obtaining autologous CAR-T products. We conducted a study involving 14 patients who received donor-derived CAR-T therapy for relapsed B-ALL following HSCT between August 2019 and May 2023 in our center. The results revealed a CR/CRi rate of 78.6% (11/14), a GVHD rate of 21.4% (3/14), and a 1-year overall survival (OS) rate of 56%. Decreased bone marrow donor cell chimerism in 9 patients recovered after CAR-T therapy. The main causes of death were disease progression and infection. Further analysis showed that GVHD (HR 7.224, 95% CI 1.42-36.82, P = 0.017) and platelet recovery at 30 days (HR 6.807, 95% CI 1.61-28.83, P = 0.009) are significantly associated with OS after CAR-T therapy. Based on the findings, we conclude that donor-derived CAR-T cells are effective in treating relapsed B-ALL patients following HSCT. Additionally, GVHD and poor platelet recovery impact OS, but further verification with a larger sample size is needed.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    嵌合抗原受体T细胞(CAR-T)疗法是血液恶性肿瘤患者的范式转变疗法。然而,一些担忧仍然存在,他们可能会导致严重的心血管不良事件(AE),数据稀缺。在这项研究中,对基于梯度提升机算法的模型进行拟合,以识别截至2024年2月世界卫生组织Vigibase报告的tisagenlecleucel严重心血管不良事件的安全信号.输入数据集,由tisagenlecleucel的阳性和阴性对照组成,基于其标记信息和文献检索,是用来训练模型的.然后,我们实施了该模型,以计算由欧洲医学机构重要医学事件列表中的首选术语定义的严重心血管不良事件的预测概率.从3,280例tisagenlecleucel安全病例报告中,有467例不同的AE,其中363人(77.7%)被列为阳性对照,66(14.2%)作为阴性对照,37例(7.9%)为未知不良事件。在测试数据集应用中,预测模型的接受者工作特征曲线下面积为0.76。在未知的AE中,六个心血管不良事件被预测为安全信号:心动过缓(预测概率0.99),胸腔积液(0.98),无脉电活动(0.89),心脏毒性(0.83),心跳呼吸停止(0.69),和急性心肌梗死(0.58)。我们的发现强调了tisagenlecleucel治疗对急性心脏毒性的警惕监测。
    Chimeric antigen receptor T-cell (CAR-T) therapies are a paradigm-shifting therapeutic in patients with hematological malignancies. However, some concerns remain that they may cause serious cardiovascular adverse events (AEs), for which data are scarce. In this study, gradient boosting machine algorithm-based model was fitted to identify safety signals of serious cardiovascular AEs reported for tisagenlecleucel in the World Health Organization Vigibase up until February 2024. Input dataset, comprised of positive and negative controls of tisagenlecleucel based on its labeling information and literature search, was used to train the model. Then, we implemented the model to calculate the predicted probability of serious cardiovascular AEs defined by preferred terms included in the important medical event list from European Medicine Agency. There were 467 distinct AEs from 3,280 safety cases reports for tisagenlecleucel, of which 363 (77.7%) were classified as positive controls, 66 (14.2%) as negative controls, and 37 (7.9%) as unknown AEs. The prediction model had area under the receiver operating characteristic curve of 0.76 in the test dataset application. Of the unknown AEs, six cardiovascular AEs were predicted as the safety signals: bradycardia (predicted probability 0.99), pleural effusion (0.98), pulseless electrical activity (0.89), cardiotoxicity (0.83), cardio-respiratory arrest (0.69), and acute myocardial infarction (0.58). Our findings underscore vigilant monitoring of acute cardiotoxicities with tisagenlecleucel therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:进行成本效益分析,比较axicabtageneciloleucel(axi-cel)与标准护理(SoC;挽救性化学免疫疗法,从日本付款人的角度来看,在关键的ZUMA-7试验数据中,对复发或难治性大B细胞淋巴瘤(r/rLBCL)的成人进行二线(2L)治疗。材料和方法:使用来自寿命范围内的ZUMA-7试验数据的群体和临床输入,利用三状态分区生存模型。结果:Axi-cel与更高的增量质量调整寿命年(2.06)和更高的增量总成本(48,685.59美元/69万日元)相关,导致每个质量调整寿命年的增量成本效益比为23,590.34美元/330万日元。结论:Axi-cel是一种替代SoC的具有成本效益的治疗方法,用于2L治疗成人r/rLBCL。
    [方框:见正文]。
    Aim: To perform a cost-effectiveness analysis comparing axicabtagene ciloleucel (axi-cel) with standard of care (SoC; salvage chemoimmunotherapy, followed by high-dose therapy with autologous stem cell rescue for responders) for second-line (2L) treatment of adults with relapsed or refractory large B-cell lymphoma (r/r LBCL) in the pivotal ZUMA-7 trial data from a Japanese payer perspective. Materials & methods: A three-state partitioned survival model was utilized using population and clinical inputs from the ZUMA-7 trial data over a lifetime horizon. Results: Axi-cel was associated with greater incremental quality-adjusted life-years (2.06) and higher incremental total costs ($48,685.59/¥6.9 million) leading to an incremental cost-effectiveness ratio of $23,590.34/¥3.3 million per quality-adjusted life-years compared with SoC. Conclusion: Axi-cel is a cost-effective treatment alternative to SoC for 2L treatment of adults with r/r LBCL.
    [Box: see text].
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    T细胞重定向疗法,如嵌合抗原受体T细胞和双特异性抗体,正在成为一类用于治疗复发性难治性多发性骨髓瘤(MM)的新型免疫治疗剂。它们的使用与感染性不良事件的风险增加有关,由血细胞减少症培育,低球蛋白血症和T细胞衰竭。多个正在进行的临床试验和现实世界的研究正在调查T细胞治疗的安全性,强调需要制定预防和监测感染风险的策略。推荐的风险缓解措施包括静脉注射免疫球蛋白补充,适当的预防治疗,疫苗接种和仔细评估,以便早期诊断和治疗感染。这里,我们总结了有关已批准和正在开发的用于治疗MM的T细胞重定向疗法的感染风险的现有数据.
    T-cell redirecting therapies such as chimeric antigen receptor T-cells and bispecific antibodies, are emerging as a novel class of immunotherapeutic agents for treatment of relapsed refractory multiple myeloma (MM). Their use is associated with an increased risk of infectious adverse events, fostered by cytopenias, hypogammaglobulinemia and T-cell exhaustion. Multiple ongoing clinical trials and real-world studies are investigating safety of T-cell therapy, highlighting the need for strategies to prevent and monitor the risk of infection. Recommended measures for risk mitigation include intravenous immunoglobulin supplementation, adequate prophylaxis therapy, vaccination and careful assessment for early diagnosis and treatment of infection. Here, we summarize available data on the risk of infections with approved and under development T-cell redirecting therapies for the treatment of MM.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    最近的数据突出显示驱动抗原逃逸的基因组事件是多发性骨髓瘤(MM)中嵌合抗原受体T细胞(CAR-T)和双特异性T细胞衔接剂(TCE)抗性的复发原因。然而,目前还不清楚这些事件,在进展中导致克隆优势,在治疗选择或选择预先存在的不可检测的克隆下获得的结果。随着这些免疫疗法发展到更早的治疗路线,这种区别变得越来越重要。促使需要创新的诊断测试来及早发现这些事件。通过在CART/TCE治疗前后重建系统发育树并探索11例复发难治性MM患者的化疗突变特征作为时间条形码,并使用可用的全基因组测序数据,我们证明了BCMA和GPRC5D靶向治疗的体细胞抗原逃逸机制是诊断后获得的,可能在CART/TCE治疗期间。在4名患者中使用数字PCR对这些突变进行纵向跟踪一致表明,在治疗的最初几个月中无法检测到促进抗原逃逸的基因组事件,但在治疗开始近1年后开始出现。这一发现减少了诊断小组在CART/TCE之前识别这些事件的必要性。相反,它强调了监测和识别发生这些事件风险较高的患者的重要性.
    结论:驱动抗原逃逸的基因组事件是多发性骨髓瘤对CART和T细胞衔接者耐药的复发机制。使用化疗突变特征,我们证明这些事件很可能是在治疗期间发生的.
    Recent data highlight genomic events driving antigen escape as a recurring cause of chimeric antigen receptor T-cell (CAR-T) and bispecific T-cell engager (TCE) resistance in multiple myeloma (MM). Yet, it remains unclear if these events, leading to clonal dominance at progression, result from acquisition under treatment selection or selection of pre-existing undetectable clones. This differentiation gains importance as these immunotherapies progress to earlier lines of treatment, prompting the need for innovative diagnostic testing to detect these events early on. By reconstructing phylogenetic trees and exploring chemotherapy mutational signatures as temporal barcodes in 11 relapsed refractory MM patients with available whole genome sequencing data before and after CART/TCE treatment, we demonstrated that somatic antigen escape mechanisms for BCMA- and GPRC5D-targeting therapies are acquired post-diagnosis, likely during CART/TCE treatment. Longitudinal tracking of these mutations using digital PCR in 4 patients consistently showed that genomic events promoting antigen escape were not detectable during the initial months of therapy but began to emerge nearly 1 year post therapy initiation. This finding reduces the necessity for a diagnostic panel to identify these events before CART/TCE. Instead, it underscores the importance of surveillance and identifying patients at higher risk of acquiring these events.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    在用tisagenlecleucel(Kymriah)治疗难治性血液恶性肿瘤中看到的有希望的结果,开创性的嵌合抗原受体(CAR)T细胞免疫疗法,刺激了新型CAR-T构建体的研究和临床试验的快速增长,目标,和各代的CAR-T细胞。药剂师可能会收到有关正在进行的临床试验的询问,或者可能有助于参与这些研究的患者的护理。本简报讨论了正在进行的临床试验,探索新型CART细胞免疫疗法在血液和实体器官起源的儿科难治性恶性肿瘤中。它在帮助从业者导航快速发展的CAR-T细胞免疫疗法领域方面可能是有价值的。
    The promising results seen in the treatment of refractory hematologic malignancies with tisagenlecleucel (Kymriah), the pioneering Chimeric Antigen Receptor (CAR) T-cell immunotherapy, has stimulated a fast growth in research and clinical testing of novel CAR-T constructs, targets, and various generations of CAR T-cells. Pharmacists may receive inquiries about active clinical trials or may be contributing to the care of patients participating in these studies. This briefing discusses the on-going clinical trials that explore novel CAR T-cell immunotherapies in pediatric refractory malignancies of hematologic and solid organ origins. It can be valuable in assisting practitioners in navigating the rapidly evolving field of CAR T-cell immunotherapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:进行了成本-效果分析(CEA),以比较axicabtageneciloleucel(axi-cel)与tisagenlecleucel(tisa-cel)和lioscabtagene(liso-cel)治疗成人复发性或难治性大B细胞淋巴瘤在日本≥2行治疗后。材料和方法:使用基于ZUMA-1试验的分区生存混合物治愈模型进行成本-效果分析,并使用匹配调整的间接比较对JULIET和TRANSCEND试验进行调整。结果和结论:Axi-cel与更大的增量生命年(3.13和2.85)和增量质量调整生命年(2.65和2.24)相关,因此产生了较低的增量直接医疗费用(-976.29美元[-137,657日元]和-242.00美元[-34,122日元]),与tisa-cel和liso-cel相比。从日本付款人的角度来看,与tisa-cel和liso-cel相比,Axi-cel是具有成本效益的选择。
    Aim: Cost-effectiveness analysis (CEA) was performed to compare axicabtagene ciloleucel (axi-cel) with tisagenlecleucel (tisa-cel) and lisocabtagene (liso-cel) for treatment of relapsed or refractory large B-cell lymphoma in adult patients after ≥2 lines of therapy in Japan. Materials & methods: Cost-effectiveness analysis was conducted using the partition survival mixture cure model based on the ZUMA-1 trial and adjusted to the JULIET and TRANSCEND trials using matching-adjusted indirect comparisons. Results & conclusion: Axi-cel was associated with greater incremental life years (3.13 and 2.85) and incremental quality-adjusted life-years (2.65 and 2.24), thus generated lower incremental direct medical costs (-$976.29 [-¥137,657] and -$242.00 [-¥34,122]), compared with tisa-cel and liso-cel. Axi-cel was cost-effective option compared with tisa-cel and liso-cel from a Japanese payer\'s perspective.
    [Box: see text].
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:嵌合抗原受体(CAR)-T细胞在约30%至40%的复发/难治性(r/r)B细胞非霍奇金淋巴瘤(B-NHL)中获得了长期持久性。CAR-T后的维持治疗是必要的,PD1抑制剂是重要的维持治疗选择之一。
    方法:从2019年3月至2022年7月,在CD19/22CAR-T治疗单独或联合自体造血干细胞移植(ASCT)后,总共173例接受PD1抑制剂维持治疗的r/rB-NHL患者被评估为两项试验的资格。有81例患者接受PD1抑制剂维持治疗。
    结果:在CD19/22CAR-T治疗试验中,PD1抑制剂维持组的客观缓解率(ORR)(82.9%vs60%;P=0.04)和2年无进展生存期(PFS)(59.8%vs21.3%;P=0.001)均优于非维持组.两组的估计2年总生存率(OS)相当(60.1%vs45.1%;P=0.112)。两组之间的CD19CAR-T和CD22CAR-T的峰值扩张水平没有观察到差异。PD1抑制剂维持组的CD19和CD22CAR-T的持续时间长于非维持组。在CD19/22CAR-T疗法联合ASCT试验中,ORR无显著差异(81.4%vs84.8%;P=0.67),2年PFS(72.3%vs74.9%;P=0.73),在非维持治疗组和PD1抑制剂维持治疗组之间观察到2年OS(84.1%vs80.7%;P=0.79)。CD19和CD22CAR-T的峰值扩张水平和持续时间在两组之间没有统计学差异。在PD1抑制剂维持治疗期间,所有不良事件均可控.在多变量分析中,类型和R3m是影响CAR-T治疗后使用PD1抑制剂维持的r/rB-NHLOS的独立预测因素。
    结论:CD19/22CAR-T治疗后维持PD1抑制剂在r/rB-NHL中获得了较好的反应和生存率,但在CD19/22CAR-T细胞疗法联合ASCT的试验中没有。
    BACKGROUND: Chimeric antigen receptor (CAR)-T cells obtained long-term durability in about 30% to 40% of relapsed/refractory (r/r) B-cell non-Hodgkin lymphoma (B-NHL). Maintenance therapy after CAR-T is necessary, and PD1 inhibitor is one of the important maintenance therapy options.
    METHODS: A total of 173 r/r B-NHL patients treated with PD1 inhibitor maintenance following CD19/22 CAR-T therapy alone or combined with autologous hematopoietic stem cell transplantation (ASCT) from March 2019 to July 2022 were assessed for eligibility for two trials. There were 81 patients on PD1 inhibitor maintenance therapy.
    RESULTS: In the CD19/22 CAR-T therapy trial, the PD1 inhibitor maintenance group indicated superior objective response rate (ORR) (82.9% vs 60%; P = 0.04) and 2-year progression-free survival (PFS) (59.8% vs 21.3%; P = 0.001) than the non-maintenance group. The estimated 2-year overall survival (OS) was comparable in the two groups (60.1% vs 45.1%; P = 0.112). No difference was observed in the peak expansion levels of CD19 CAR-T and CD22 CAR-T between the two groups. The persistence time of CD19 and CD22 CAR-T in the PD1 inhibitor maintenance group was longer than that in the non-maintenance group. In the CD19/22 CAR-T therapy combined with ASCT trial, no significant differences in ORR (81.4% vs 84.8%; P = 0.67), 2-year PFS (72.3% vs 74.9%; P = 0.73), and 2-year OS (84.1% vs 80.7%; P = 0.79) were observed between non-maintenance and PD1 inhibitor maintenance therapy groups. The peak expansion levels and duration of CD19 and CD22 CAR-T were not statistically different between the two groups. During maintenance treatment with PD1 inhibitor, all adverse events were manageable. In the multivariable analyses, type and R3m were independent predictive factors influencing the OS of r/r B-NHL with PD1 inhibitor maintenance after CAR-T therapy.
    CONCLUSIONS: PD1 inhibitor maintenance following CD19/22 CAR-T therapy obtained superior response and survival in r/r B-NHL, but not in the trial of CD19/22 CAR-T cell therapy combined with ASCT.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号