Chimeric antigen receptor T cells

嵌合抗原受体 T 细胞
  • 文章类型: Journal Article
    目的:构建靶向上皮细胞粘附分子(EpCAM)抗原的嵌合抗原受体(CAR)-T细胞。
    方法:第三代CAR-T细胞构建体使用了源自抗人EpCAM的单克隆抗体的单链可变片段。从志愿者中提取外周血单核细胞。使用流式细胞术测量分化8阳性(CD8+)和CD4+T细胞簇的比例。Westernblot检测EpCAM-CAR的表达。使用MTT测定法和transwell测定法检测杀灭效率,用ELISA法检测肿瘤坏死因子-α(TNF-α)和干扰素-γ(IFN-γ)的分泌。使用异种移植物检测EpCAM-CAR-T对结直肠癌的体内抑制作用。
    结果:发现T细胞大量扩增,和CD3+的比例,CD8+和CD4+T细胞比例均在60%以上。此外,EpCAM-CAR-T细胞表达阳性组肿瘤抑制率高于阴性组(P<0.05)。EpCAM表达阳性细胞组杀伤细胞因子TNF-α和IFN-γ的分泌高于阴性细胞组(P<0.05)。在用EpCAM-CAR-T细胞治疗的实验组中,裸鼠成活率较高(P<0.05),肿瘤小于空白组和对照组(P<0.05)。EpCAM-CAR-T细胞治疗组荷瘤裸鼠血清杀伤细胞因子TNF-α和IFN-γ的分泌高于空白组和对照组(P<0.05)。
    结论:这项研究成功构建了EpCAM-CAR细胞,并发现它们可以靶向和识别EpCAM阳性肿瘤细胞,分泌杀伤细胞因子TNF-α和IFN-γ,并在体外和体内比未修饰的T细胞更好地抑制结直肠癌的生长和转移。
    OBJECTIVE: To construct chimeric antigen receptor (CAR)-T cells targeting epithelial cell adhesion molecule (EpCAM) antigen (anti-EpCAM-CAR-T).
    METHODS: A third-generation CAR-T cell construct used a single-chain variable fragment derived from monoclonal antibody against human EpCAM. Peripheral blood mononuclear cells were extracted from volunteers. The proportion of cluster of differentiation 8 positive (CD8+) and CD4 + T cells was measured using flow cytometry. Western blot was used to detect the expression of EpCAM-CAR. The killing efficiency was detected using the MTT assay and transwell assay, and the secretion of killer cytokines tumour necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) was detected using the ELISA. The inhibitory effect of EpCAM-CAR-T on colorectal cancer in vivo was detected using xenografts.
    RESULTS: It was found that T cells expanded greatly, and the proportion of CD3+, CD8 + and CD4 + T cells was more than 60%. Furthermore, EpCAM-CAR-T cells had a higher tumour inhibition rate in the EpCAM expression positive group than in the negative group (P < 0.05). The secretion of killer cytokines TNF-α and IFN-γ in the EpCAM expression positive cell group was higher than that in the negative group (P < 0.05). In the experimental group treated with EpCAM-CAR-T cells, the survival rate of nude mice was higher (P < 0.05), and the tumour was smaller than that in the blank and control groups (P < 0.05). The secretion of serum killer cytokines TNF-α and IFN-γ in tumour-bearing nude mice in the experimental group treated with EpCAM-CAR-T cells was higher than that in the blank and control groups (P < 0.05).
    CONCLUSIONS: This study successfully constructed EpCAM-CAR cells and found that they can target and recognise EpCAM-positive tumour cells, secrete killer cytokines TNF-α and IFN-γ and better inhibit the growth and metastasis of colorectal cancer in vitro and in vivo than unmodified T cells.
    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
    脊索瘤是一种罕见的骨肿瘤,手术后经常复发,目前的治疗方法预后较差。这项研究旨在通过识别临床样品中的靶蛋白以及肿瘤微环境因素来增强疗效,从而确定脊索瘤潜在的新型免疫治疗靶标。通过单细胞RNA测序分析了14个脊索瘤样本,B7-H3和IL-7被确定为潜在的靶标和增效剂,分别。合成B7-H3靶向的嵌合抗原受体T(CAR-T)细胞和表达IL-7的B7-H3CAR-T细胞,并在体外评估其抗肿瘤活性。包括原发性脊索瘤类器官模型。B7-H3CAR-T/IL-7疗法显示出增强的细胞毒性和延长的抗肿瘤细胞作用持续时间。此外,IL-7调节培养的CAR-T细胞的有利亚群,T细胞表面免疫检查点表达减少,和增强的T细胞功能。将IL-7分子掺入B7-H3CAR结构增强了CAR-T细胞功能并改善了CAR-T细胞功效,从而为脊索瘤的治疗提供了一种新的双重治疗策略。
    Chordoma is a rare bone tumor that frequently recurs after surgery, and the prognosis is poor with current treatments. This study aimed to identify potential novel immunotherapeutic targets for chordomas by identifying target proteins in clinical samples as well as tumor microenvironmental factors to enhance efficacy. Fourteen chordoma samples were analyzed by single-cell RNA sequencing, and B7-H3 and IL-7 were identified as potential targets and potentiators, respectively. B7-H3-targeted chimeric antigen receptor T (CAR-T) cells and B7-H3 CAR-T cells expressing IL-7 were synthesized and their anti-tumor activity evaluated in vitro, including in primary chordoma organoid models. The B7-H3 CAR-T/IL-7 therapy showed enhanced cytotoxicity and prolonged duration of action against tumor cells. Additionally, IL-7 modulated favorable subpopulations of cultured CAR-T cells, diminished immune checkpoint expression on T-cell surfaces, and enhanced T-cell functionality. The incorporation of IL-7 molecules into the B7-H3 CAR structure augmented CAR-T-cell function and improved CAR-T-cell efficacy, thus providing a novel dual therapeutic strategy for chordoma treatment.
    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
    自2017年FDA批准嵌合抗原受体(CAR)T细胞以来,嵌合抗原受体构建体的设计和CART细胞疗法的制造取得了显著改善,导致体内CART细胞持久性增加,并改善了某些血液恶性肿瘤的临床结果。尽管在一些患者中看到了显着的临床反应,在实现持久的长期无瘤生存方面仍然存在挑战,减少治疗相关的恶性肿瘤和毒性,并扩大可以用这种治疗方式治疗的癌症类型。仔细分析与次优CART细胞反应区分有效的生物因素对于解决这些缺点至关重要。随着实验方法工具箱的不断扩大,单细胞技术,和计算资源,有发现新的方法来简化新的CART细胞产品的开发和验证的兴趣。可以开发更好,更准确的预后和预测模型,以通过将这些方法纳入转化和临床工作流程来帮助指导和告知临床决策。在这次审查中,我们简要概述了CART细胞制造的最新进展,并描述了用于选择性扩增特定表型亚群的策略.此外,我们回顾了评估CART细胞功能的实验方法,并总结了目前有可能改善CART细胞制造和预测临床结局的计算机模拟方法.
    Since the FDA\'s approval of chimeric antigen receptor (CAR) T cells in 2017, significant improvements have been made in the design of chimeric antigen receptor constructs and in the manufacturing of CAR T cell therapies resulting in increased in vivo CAR T cell persistence and improved clinical outcome in certain hematological malignancies. Despite the remarkable clinical response seen in some patients, challenges remain in achieving durable long-term tumor-free survival, reducing therapy associated malignancies and toxicities, and expanding on the types of cancers that can be treated with this therapeutic modality. Careful analysis of the biological factors demarcating efficacious from suboptimal CAR T cell responses will be of paramount importance to address these shortcomings. With the ever-expanding toolbox of experimental approaches, single-cell technologies, and computational resources, there is renowned interest in discovering new ways to streamline the development and validation of new CAR T cell products. Better and more accurate prognostic and predictive models can be developed to help guide and inform clinical decision making by incorporating these approaches into translational and clinical workflows. In this review, we provide a brief overview of recent advancements in CAR T cell manufacturing and describe the strategies used to selectively expand specific phenotypic subsets. Additionally, we review experimental approaches to assess CAR T cell functionality and summarize current in silico methods which have the potential to improve CAR T cell manufacturing and predict clinical outcomes.
    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
    最近,双特异性T细胞衔接剂(BiTE)和嵌合抗原受体修饰的T细胞(CAR-Ts)已显示在血液肿瘤中具有高治疗功效。CD87在实体瘤中高表达,具有致癌功能。为了评估它们对侵袭性无功能垂体腺瘤(iNFPA)的细胞毒性作用,我们起首检测了CD87的表达及其对iNFPA细胞代谢的影响。我们产生了CD87特异性BiTE和CAR/IL-12T细胞,并确定了它们对iNFPAs细胞和小鼠模型的细胞毒性作用。CD87在iNFPA组织和细胞样品中高表达,但在非癌性脑样品中未检测到。CD87×CD3BiTE和CD87CAR/IL-12T细胞表现出抗原特异性,发挥了满意的细胞毒作用。降低体外肿瘤细胞增殖和减少实验小鼠中现有的肿瘤。总的来说,上述研究结果表明,CD87是使用抗CD87BiTE和CD87特异性CAR/IL-12T细胞对iNFPAs进行免疫治疗的有前景的靶标.
    Recently, bispecific T-cell engagers (BiTEs) and chimeric antigen receptor-modified T cells (CAR-Ts) have been shown to have high therapeutic efficacy in hematological tumors. CD87 is highly expressed in solid tumors with an oncogenic function. To assess their cytotoxic effects on invasive nonfunctioning pituitary adenomas (iNFPAs), we first examined CD87 expression and its effects on the metabolism of iNFPA cells. We generated CD87-specific BiTE and CAR/IL-12 T cells, and their cytotoxic effects on iNFPAs cells and in mouse models were determined. CD87 had high expression in iNFPA tissue and cell samples but was undetected in noncancerous brain samples. CD87×CD3 BiTE and CD87 CAR/IL-12 T-cells showed antigenic specificity and exerted satisfactory cytotoxic effects, decreasing tumor cell proliferation in vitro and reducing existing tumors in experimental mice. Overall, the above findings suggest that CD87 is a promising target for the immunotherapeutic management of iNFPAs using anti-CD87 BiTE and CD87-specific CAR/IL-12 T cells.
    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:由于T细胞持久性差,靶向单一抗原的嵌合抗原受体T(CAR-T)细胞对实体瘤的活性有限,低效率渗透,与异质性肿瘤相关抗原(TAA)表达一起耗尽。在高风险神经母细胞瘤(HRNB)中也是如此,一种致命的小儿颅外恶性肿瘤.为了克服这些障碍,开发了一种使用GD2特异性和GPC2特异性CAR-T细胞的组合策略,以提高免疫治疗效果.
    方法:我们单独开发了GD2特异性和GPC2特异性CAR,其中包含选择性结构域(sCAR),该选择性结构域是源自人核自身抗原La/SS-B的10个氨基酸的肽。这些构建体使我们能够通过经由选择性结构域特异性单克隆抗体(SmAb)刺激sCAR移植的T细胞来产生具有增强的生物活性的两种不同的HRNB抗原特异性CAR-T细胞。测量了SmAb对GD2-和GPC2特异性sCAR的结合亲和力和刺激,在表达不同TAA水平的神经母细胞瘤细胞系中定量GD2sCAR-T和GPC2sCAR-T细胞的瞬时和持续抗肿瘤细胞毒性。在体外和体内评估了由单个或组合sCAR-T细胞介导的抗肿瘤药物作用和细胞机制。
    结果:GD2-和GPC2特异性sCAR具有与其亲本对应物相似的抗原特异性结合亲和力,并被SmAb识别。SmAb介导的刺激选择性激活最终产物中的sCAR-T增殖并增加中枢记忆T细胞。SmAb刺激的sCAR-T细胞具有增强的瞬时细胞溶解活性,联合治疗通过TNF-α和IL-15的释放延长了体外长期抗肿瘤活性。刺激的sCAR-T细胞克服了HRNB中的异质性抗原表达,多TAA靶向策略在体内特别有效,通过caspase-3/PARP途径诱导细胞凋亡并抑制几种促肿瘤细胞因子的释放。
    结论:这些数据表明,多个TAA的联合靶向是克服实体肿瘤中异质性抗原表达并延长CAR-T细胞持久性的有希望的策略用于HRNB免疫治疗。
    OBJECTIVE: Chimeric antigen receptor T (CAR-T) cells targeting single antigens show limited activity against solid tumors due to poor T cell persistence, low efficiency infiltration, and exhaustion together with heterogeneous tumor-associated antigen (TAA) expression. This is also true in high-risk neuroblastoma (HRNB), a lethal pediatric extracranial malignancy. To overcome these obstacles, a combinational strategy using GD2-specific and GPC2-specific CAR-T cells was developed to improve immunotherapeutic efficacy.
    METHODS: We individually developed GD2-specific and GPC2-specific CARs containing a selective domain (sCAR) which was a peptide of 10 amino acids derived from human nuclear autoantigen La/SS-B. These constructs allowed us to generate two different HRNB antigen-specific CAR-T cells with enhanced biological activity through stimulating sCAR-engrafted T cells via a selective domain-specific monoclonal antibody (SmAb). Binding affinity and stimulation of GD2- and GPC2-specific sCARs by SmAb were measured, and transient and persistent anti-tumor cytotoxicity of GD2sCAR-T and GPC2sCAR-T cells were quantified in neuroblastoma cell lines expressing different TAA levels. The anti-tumor pharmaceutical effects and cellular mechanisms mediated by single or combinational sCAR-T cells were evaluated in vitro and in vivo.
    RESULTS: GD2- and GPC2-specific sCARs had antigen-specific binding affinity similar to their parental counterparts and were recognized by SmAb. SmAb-mediated stimulation selectively activated sCAR-T proliferation and increased central memory T cells in the final products. SmAb-stimulated sCAR-T cells had enhanced transient cytolytic activity, and combination therapy extended long-term anti-tumor activity in vitro through TNF-α and IL-15 release. Stimulated sCAR-T cells overcame heterogeneous antigen expression in HRNB, and the multi-TAA-targeting strategy was especially efficacious in vivo, inducing apoptosis through the caspase-3/PARP pathway and inhibiting the release of several tumor-promoting cytokines.
    CONCLUSIONS: These data suggest that combined targeting of multiple TAAs is a promising strategy to overcome heterogenous antigen expression in solid tumors and extend CAR-T cell persistence for HRNB immunotherapy.
    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细胞表达外源肿瘤靶向受体如嵌合抗原受体(CAR)或T细胞受体(TCR)的有效工程化是癌症的有效过继细胞疗法的关键要求。基因组编辑技术,例如CRISPR/Cas9可以通过敲除感兴趣的基因同时敲入可以识别癌细胞的合成受体来进一步改变治疗性T细胞的功能特征。通过精确的基因组编辑进行多轮基因转移,称为多路复用,仍然是一个关键的挑战,特别是对于非病毒递送平台。这里,我们证明了结合三个临床相关基因(B2M,TRAC,和PD1)以及靶向二唾液酸神经节苷脂GD2的CAR的非病毒转染。多重敲除导致所有三个基因的高目标缺失,低脱靶编辑和染色体改变。掺入非病毒递送以敲入GD2-CAR导致TRAC-B2M-PD1缺陷的GD2CART细胞产物,其具有中央记忆细胞表型和针对表达GD2的神经母细胞瘤靶细胞的高细胞毒性。通过CRISPR/Cas9与非病毒递送的多重基因编辑是可行和安全的,具有快速高效制造高效同种异体CAR-T细胞产品的巨大潜力。
    Efficient engineering of T cells to express exogenous tumor-targeting receptors such as chimeric antigen receptors (CARs) or T-cell receptors (TCRs) is a key requirement of effective adoptive cell therapy for cancer. Genome editing technologies, such as CRISPR/Cas9, can further alter the functional characteristics of therapeutic T cells through the knockout of genes of interest while knocking in synthetic receptors that can recognize cancer cells. Performing multiple rounds of gene transfer with precise genome editing, termed multiplexing, remains a key challenge, especially for non-viral delivery platforms. Here, we demonstrate the efficient production of primary human T cells incorporating the knockout of three clinically relevant genes (B2M, TRAC, and PD1) along with the non-viral transfection of a CAR targeting disialoganglioside GD2. Multiplexed knockout results in high on-target deletion for all three genes, with low off-target editing and chromosome alterations. Incorporating non-viral delivery to knock in a GD2-CAR resulted in a TRAC-B2M-PD1-deficient GD2 CAR T-cell product with a central memory cell phenotype and high cytotoxicity against GD2-expressing neuroblastoma target cells. Multiplexed gene-editing with non-viral delivery by CRISPR/Cas9 is feasible and safe, with a high potential for rapid and efficient manufacturing of highly potent allogeneic CAR T-cell products.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    嵌合抗原受体T(CAR-T)细胞疗法是复发性和难治性(R/R)B细胞急性淋巴细胞白血病(B-ALL)免疫治疗的里程碑成就。然而,一些用CAR-T细胞治疗的患者没有达到完全缓解,其机制尚未阐明。在本研究中,我们报道了1例9岁的难治性B-ALL患儿在第二次复发后接受了自体CD19CAR-T细胞三联输注治疗.CAR-T细胞在外周血和骨髓中扩增。然而,患者没有达到完全缓解,表明对CAR-T细胞疗法缺乏反应。病因分析显示,外周血中CD4和CD8双阴性T(DNT)细胞数量明显上调,骨髓,和自体CAR-T细胞产品。在结论中,这些研究结果表明,在患有R/RB-ALL的儿科患者中,DNT细胞介导了对CAR-T细胞治疗的抵抗.
    Chimeric antigen receptor T (CAR-T) cells therapy is a milestone achievement in the immunotherapy of relapsed and refractory (R/R) B cell acute lymphoblastic leukemia (B-ALL). However, some patients treated with CAR-T cells do not achieve complete remission, the mechanisms of which have not been elucidated. In the present study, we report a 9-year-old pediatric patient with refractory B-ALL received a triple infusion of autologous CD19 CAR-T cells therapy after the second relapse. CAR-T cells expanded in the peripheral blood and bone marrow. However, the patient did not achieve complete remission, indicating a lack of response to CAR-T cells therapy. Analysis of etiological factors revealed that the number of CD4 and CD8 double-negative T (DNT) cells was significantly upregulated in the peripheral blood, bone marrow, and autologous CAR-T cells products. In conclusiont, these findings indicate that DNT cells mediated resistance to CAR-T cells therapy in this pediatric patient with R/R B-ALL.
    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
    急性髓性白血病(AML)是一种致命且难治性的血液癌症,主要影响成人。它干扰骨髓细胞增殖。尽管有多种治疗方法,患者的5年生存率仍低于30%。包括化疗,异基因造血干细胞移植(Allo-HSCT),和受体拮抗剂药物。Allo-HSCT是急性髓性白血病治疗的主要手段。虽然它确实有效,有严重的副作用,例如移植物抗宿主病(GVHD)。近年来,嵌合抗原受体(CAR)-T细胞疗法在癌症治疗中取得了重大进展。这些工程化的T细胞可以在体内定位和识别肿瘤细胞,并通过免疫作用释放大量效应子,从而有效地杀死肿瘤细胞。由于这种特性,CAR-T细胞是最有效的癌症治疗方法之一。CAR-T细胞在急性髓系白血病的治疗中表现出积极的治疗效果。根据许多临床调查。本文综述了AML免疫治疗新靶点的最新进展。和局限性,和CAR-T治疗AML的困难。
    Acute myeloid leukaemia (AML) is a fatal and refractory haematologic cancer that primarily affects adults. It interferes with bone marrow cell proliferation. Patients have a 5 years survival rate of less than 30% despite the availability of several treatments, including chemotherapy, allogeneic haematopoietic stem cell transplantation (Allo-HSCT), and receptor antagonist drugs. Allo-HSCT is the mainstay of acute myeloid leukaemia treatment. Although it does work, there are severe side effects, such as graft-versus-host disease (GVHD). In recent years, chimeric antigen receptor (CAR)-T cell therapies have made significant progress in the treatment of cancer. These engineered T cells can locate and recognize tumour cells in vivo and release a large number of effectors through immune action to effectively kill tumour cells. CAR-T cells are among the most effective cancer treatments because of this property. CAR-T cells have demonstrated positive therapeutic results in the treatment of acute myeloid leukaemia, according to numerous clinical investigations. This review highlights recent progress in new targets for AML immunotherapy, and the limitations, and difficulties of CAR-T therapy for AML.
    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
    轻链(AL)淀粉样变性是一种罕见的浆细胞疾病,其特征是错误折叠的免疫球蛋白轻链在靶器官中沉积。导致多器官功能障碍。治疗方法历来反映但落后于多发性骨髓瘤(MM)的治疗方法。MM免疫治疗的最新进展正在逐渐被评估和采用在AL淀粉样变性中。这篇综述探讨了AL淀粉样变性的免疫治疗策略的现状,包括单克隆抗体,抗体-药物缀合物,双特异性抗体,和嵌合抗原受体T细胞疗法。我们讨论了这些疗法在AL淀粉样变性中的独特挑战和前景,包括脆弱的AL淀粉样变性患者暴露于免疫介导的毒性,如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),以及它们在促进快速和深层血液学反应方面的功效。此外,我们强调需要采取国际举措和富有同情心的计划,以提供获得这些有希望的治疗方法,并解决AL淀粉样变性管理中未满足的关键需求。最后,我们讨论未来的方向,包括优化治疗顺序和减轻毒性,改善AL淀粉样变性患者的预后。
    Light-chain (AL) amyloidosis is a rare plasma cell disorder characterized by the deposition of misfolded immunoglobulin light chains in target organs, leading to multi-organ dysfunction. Treatment approaches have historically mirrored but lagged behind those of multiple myeloma (MM). Recent advancements in MM immunotherapy are gradually being evaluated and adopted in AL amyloidosis. This review explores the current state of immunotherapeutic strategies in AL amyloidosis, including monoclonal antibodies, antibody-drug conjugates, bispecific antibodies, and chimeric antigen receptor T-cell therapy. We discuss the unique challenges and prospects of these therapies in AL amyloidosis, including the exposure of frail AL amyloidosis patients to immune-mediated toxicities such as cytokine release syndrome (CRS) and immune effector-cell-associated neurotoxicity syndrome (ICANS), as well as their efficacy in promoting rapid and deep hematologic responses. Furthermore, we highlight the need for international initiatives and compassionate programs to provide access to these promising therapies and address critical unmet needs in AL amyloidosis management. Finally, we discuss future directions, including optimizing treatment sequencing and mitigating toxicities, to improve outcomes for AL amyloidosis patients.
    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
    免疫治疗策略,特别是T细胞重定向疗法,在多发性骨髓瘤(MM)的背景下具有变革性。随着两种嵌合抗原受体T细胞(CAR-T)药物产品和三种双特异性抗体/T细胞衔接剂(bsAbs/TCEs)在复发/难治性MM(RRMM)中的批准,第20届IMS年会专门讨论这些疗法的实际问题.这里,我们强调在本届会议期间的讨论,包括CAR-T和bsAb疗法在一线MM治疗中的作用,急性毒性的管理,预防和管理感染,最后是T细胞重定向疗法的治疗测序。
    Immunotherapeutic strategies, specifically T-cell-redirected therapies, have been transformative in the context of multiple myeloma (MM). With the approval of two chimeric antigen receptor T-cell (CAR-T) drug products and three bispecific antibodies/T-cell engagers (bsAbs/TCEs) in relapsed/refractory MM (RRMM), the 20th annual IMS meeting dedicated a session to the practical aspects of these therapies. Here, we highlight the discussion during this session, including the role of CAR-T and bsAb therapies in frontline MM treatment, management of acute toxicities, prevention and management of infections, and finally treatment sequencing of T-cell redirected therapies.
    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