MT: Regular Issue

  • 文章类型: Journal Article
    由于复杂的抗原呈递,抗原特异性T细胞活化不足会阻碍免疫治疗。此外,治疗性体内T细胞扩增受限于缓慢的扩增速率和有限的功能性。在这里,我们引入了一种称为抗原呈递细胞模拟融合蛋白(APC模拟物)的模型融合蛋白,旨在极大地模拟抗原呈递细胞的天然抗原呈递模式,并在体外和体内直接扩增T细胞。APC模拟物包含同源肽-人白细胞抗原(pHLA)复合物和共刺激标记CD80,它们是APC上的天然配体。在一次刺激之后,在不需要专门的抗原呈递细胞的情况下,与未处理组相比,APC模拟物导致抗原特异性T细胞的多克隆扩增增加约400倍。通过单细胞TCR测序(scTCR-seq)和单细胞RNA测序(scRNA-seq)的结合,我们在这些多克隆克隆型中鉴定出大约600倍的单克隆扩增克隆型.它还表现出在OT-1小鼠模型中证实的体内应用的适用性。此外,通过APC模拟物扩增的T细胞有效抑制过继性细胞转移(ACT)鼠模型中的肿瘤生长。这些发现为个性化治疗的多功能APC模拟平台铺平了道路,能够在体外和体内直接扩增多功能抗原特异性T细胞亚群。
    Inadequate antigen-specific T cells activation hampers immunotherapy due to complex antigen presentation. In addition, therapeutic in vivo T cell expansion is constrained by slow expansion rates and limited functionality. Herein, we introduce a model fusion protein termed antigen-presenting cell-mimic fusion protein (APC-mimic), designed to greatly mimicking the natural antigen presentation pattern of antigen-presenting cells and directly expand T cells both in vitro and in vivo. The APC-mimic comprises the cognate peptide-human leukocyte antigen (pHLA) complex and the co-stimulatory marker CD80, which are natural ligands on APCs. Following a single stimulation, APC-mimic leads to an approximately 400-fold increase in the polyclonal expansion of antigen-specific T cells compared with the untreated group in vitro without the requirement for specialized antigen-presenting cells. Through the combination of single-cell TCR sequencing (scTCR-seq) and single-cell RNA sequencing (scRNA-seq), we identify an approximately 600-fold monoclonal expansion clonotype among these polyclonal clonotypes. It also exhibits suitability for in vivo applications confirmed in the OT-1 mouse model. Furthermore, T cells expanded by APC-mimic effectively inhibits tumor growth in adoptive cell transfer (ACT) murine models. These findings pave the way for the versatile APC-mimic platform for personalized therapeutics, enabling direct expansion of polyfunctional antigen-specific T cell subsets in vitro and in vivo.
    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
    双特异性T细胞衔接剂是一类有前途的用于癌症治疗的治疗性蛋白质。它们的效力和小尺寸通常伴随着全身毒性和短半衰期,使静脉给药变得繁琐。这些限制可以通过肿瘤特异性原位表达来克服,允许高局部积累,同时降低全身浓度。然而,编码病毒或非病毒载体中的T细胞衔接者并原位表达它们消除了在重组蛋白生产期间进行的所有形式的质量控制。因此,设计具有最小域错误配对的构造至关重要,和增加的同质性的治疗产品。这里,我们报道了一种专门为载体介导的免疫治疗而设计的T细胞接合器结构.它基于设计的锚蛋白重复蛋白(DARPin)与CD3靶向单链抗体片段的融合,称为DATE(DARPin融合T细胞接合器)。DATE诱导强效T细胞介导的HER2+癌细胞杀伤,作为重组产生的治疗性蛋白和作为来自HER2+重靶向的高容量腺病毒载体(HC-AdV)的原位表达的有效载荷。我们报告了显著的肿瘤缓解,日期累积,和T细胞浸润通过在体内由HER2+重新靶向的HC-AdV介导的原位表达。我们的结果支持DATEs作为载体介导的免疫治疗的有效载荷的进一步研究和发展。
    Bispecific T cell engagers are a promising class of therapeutic proteins for cancer therapy. Their potency and small size often come with systemic toxicity and short half-life, making intravenous administration cumbersome. These limitations can be overcome by tumor-specific in situ expression, allowing high local accumulation while reducing systemic concentrations. However, encoding T cell engagers in viral or non-viral vectors and expressing them in situ ablates all forms of quality control performed during recombinant protein production. It is therefore vital to design constructs that feature minimal domain mispairing, and increased homogeneity of the therapeutic product. Here, we report a T cell engager architecture specifically designed for vector-mediated immunotherapy. It is based on a fusion of a designed ankyrin repeat protein (DARPin) to a CD3-targeting single-chain antibody fragment, termed DATE (DARPin-fused T cell Engager). The DATE induces potent T cell-mediated killing of HER2+ cancer cells, both as recombinantly produced therapeutic protein and as in situ expressed payload from a HER2+-retargeted high-capacity adenoviral vector (HC-AdV). We report remarkable tumor remission, DATE accumulation, and T cell infiltration through in situ expression mediated by a HER2+-retargeted HC-AdV in vivo. Our results support further investigations and developments of DATEs as payloads for vector-mediated immunotherapy.
    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
    在创新癌症治疗策略领域,溶瘤痘苗病毒(VV)已成为有前途的载体。在目前的研究中,我们将人C型凝集素结构域家族2成员A(CLEC2A)基因插入VV,创造一种复制的治疗剂,oncoVV-CLEC2A.研究结果表明,oncoVV-CLEC2A通过增强病毒繁殖能力有效抑制小鼠异种移植物和一系列人类癌细胞系的结肠直肠增殖,包括肺癌H460细胞系,结直肠癌细胞系(HCT116和SW620),和肝细胞癌HuH-7细胞系。此外,很明显,oncoVV-CLEC2A可以通过促进细胞因子的产生而不是抗病毒反应来诱导抗肿瘤免疫,增强钙网蛋白表达。进一步的研究表明,癌VV-CLEC2A可以通过激活自然杀伤细胞产生干扰素γ并诱导M1样巨噬细胞极化来增强抗肿瘤能力。这些发现揭示了癌VV-CLEC2A的抗肿瘤机制,为溶瘤治疗提供理论依据,并为修改VV的新策略奠定基础。
    In the field of innovative cancer treatment strategies, oncolytic vaccinia virus (VV)es have gained traction as promising vectors. In the current study, we inserted the human C-type lectin domain family 2 member A (CLEC2A) gene into VV, creating a replicating therapeutic, oncoVV-CLEC2A. The findings reveal that oncoVV-CLEC2A effectively suppresses colorectal proliferation of mouse xenografts and a range of human cancer cell lines by augmenting viral reproduction capabilities, including the lung cancer H460 cell line, colorectal cancer cell lines (HCT116 and SW620), and hepatocellular carcinoma HuH-7 cell line. Moreover, it is evident that oncoVV-CLEC2A can induce antitumor immunity by boosting cytokine production but not antivirus response, and enhancing calreticulin expression. Further investigation indicates that oncoVV-CLEC2A can enhance antitumor capabilities by activating natural killer cells to produce interferon-γ and induce M1-like macrophage polarization. These findings shed light on the antitumor mechanisms of oncoVV-CLEC2A, provide a theoretical basis for oncolytic therapies, and lay the groundwork for novel strategies for modifying VVs.
    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
    胰胆管癌,包括胰腺和胆道的恶性肿瘤,提出了一个可怕的临床挑战,其特征是一致的预后暗淡。其早期阶段的无症状性质通常导致延迟检测,导致不利的5年总生存率。传统的治疗方式显示出有限的疗效,强调迫切需要替代治疗方法。近年来,免疫疗法已成为对抗胰胆管癌的一个有希望的途径。诸如治疗性疫苗和肿瘤浸润性淋巴细胞的使用等策略因其引起更稳健和持久反应的潜力而引起关注。这篇综述旨在阐明新兴的免疫治疗干预措施的前景,提供从临床和研究角度的见解。通过加深对胰胆管癌的认识,探索创新的治疗方式,我们的目标是促进改善患者预后和生活质量.
    Pancreaticobiliary cancer, encompassing malignancies of both the pancreatic and biliary tract, presents a formidable clinical challenge marked by a uniformly bleak prognosis. The asymptomatic nature of its early stages often leads to delayed detection, contributing to an unfavorable 5-year overall survival rate. Conventional treatment modalities have shown limited efficacy, underscoring the urgent need for alternative therapeutic approaches. In recent years, immunotherapy has emerged as a promising avenue in the fight against pancreaticobiliary cancer. Strategies such as therapeutic vaccines and the use of tumor-infiltrating lymphocytes have garnered attention for their potential to elicit more robust and durable responses. This review seeks to illuminate the landscape of emerging immunotherapeutic interventions, offering insights from both clinical and research perspectives. By deepening our understanding of pancreaticobiliary cancer and exploring innovative treatment modalities, we aim to catalyze improvements in patient outcomes and quality of life.
    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
    膀胱癌(BlCa)是一种广泛的异质性疾病,导致肿瘤演变情景和终身患者监测的巨大差异。强调现代的需要,微创精准医学。这里,我们探讨了BlCa中拷贝数改变(CNA)的临床意义。在15例患者来源的异种移植物(PDX)中进行了CNA分析,并在癌症基因组图谱BlCa(TCGA-BLCA;n=408)和Lindgren等人中进行了验证。(n=143)队列。CDKN2A拷贝数丢失被确定为膀胱肿瘤中最常见的CNA,与CDKN2A表达减少相关,乳头状表型的肿瘤,和延长PDX生存期。该研究的筛查队列包括243名BlCa患者,和CDKN2A拷贝数在基因组DNA和无细胞DNA(cfDNA)从217个肿瘤和189个治疗前的血清样品,分别。CDKN2A拷贝数丢失与非肌肉侵入性BlCa(NMIBC)患者的无疾病和无进展生存期相关。此外,治疗前cfDNA较高的CDKN2A指数(CDKN2A/LEP比值)与晚期肿瘤分期和分级以及短期NMIBC进展为浸润性疾病相关,虽然适用于治疗前cfDNA的CDKN2A指数的多变量模型提供了T1/高级别和EORTC高危患者的优越风险分层,增强对治疗结果的预测。CDKN2A拷贝数状态可以作为一种微创工具,以改善风险分层并支持BlCa的个性化预后。
    Bladder cancer (BlCa) is an extensively heterogeneous disease that leads to great variability in tumor evolution scenarios and lifelong patient surveillance, emphasizing the need for modern, minimally invasive precision medicine. Here, we explored the clinical significance of copy number alterations (CNAs) in BlCa. CNA profiling was performed in 15 patient-derived xenografts (PDXs) and validated in The Cancer Genome Atlas BlCa (TCGA-BLCA; n = 408) and Lindgren et al. (n = 143) cohorts. CDKN2A copy number loss was identified as the most frequent CNA in bladder tumors, associated with reduced CDKN2A expression, tumors of a papillary phenotype, and prolonged PDX survival. The study\'s screening cohort consisted of 243 BlCa patients, and CDKN2A copy number was assessed in genomic DNA and cell-free DNA (cfDNA) from 217 tumors and 189 pre-treatment serum samples, respectively. CDKN2A copy number loss was correlated with superior disease-free and progression-free survival of non-muscle-invasive BlCa (NMIBC) patients. Moreover, a higher CDKN2A index (CDKN2A/LEP ratio) in pre-treatment cfDNA was associated with advanced tumor stage and grade and short-term NMIBC progression to invasive disease, while multivariate models fitted for CDKN2A index in pre-treatment cfDNA offered superior risk stratification of T1/high-grade and EORTC high-risk patients, enhancing prediction of treatment outcome. CDKN2A copy number status could serve as a minimally invasive tool to improve risk stratification and support personalized prognosis in BlCa.
    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
    现成的(OTS)过继性T细胞疗法具有许多好处,例如立即可用,改善访问和降低成本,但是面对移植物抗宿主病(GVHD)和移植物排斥的主要挑战,由移植物和宿主中存在的同种反应性T细胞介导,分别。我们通过使用表达靶向CD30的嵌合抗原受体(CAR)的爱泼斯坦-巴尔病毒(EBV)特异性T细胞(EBVST)开发了OTST细胞治疗平台。同种异体EBVST在几项临床试验中没有引起GVHD,而CD30。汽车,对淋巴瘤的治疗有效,还可以靶向在激活时上调CD30的同种异体反应性T细胞。尽管EBVST表达高水平的CD30,但它们在顺式中受到保护,CD30汽车。因此,它们可以通过其天然EBV特异性T细胞受体和CD30广泛增殖并维持功能。汽车。CD30CAR使EBVST能够与幼稚和引发的同种异体反应性T细胞在共培养中持续存在,并消除也可能具有同种反应性的活化自然杀伤细胞。总之,我们显示CD30。CAREBVST有潜力成为一种有效的OTS治疗CD30+肿瘤,如果成功,然后可以用作靶向其他肿瘤抗原的平台。
    Off-the-shelf (OTS) adoptive T cell therapies have many benefits such as immediate availability, improved access and reduced cost, but face the major challenges of graft-vs-host disease (GVHD) and graft rejection, mediated by alloreactive T cells present in the graft and host, respectively. We have developed a platform for OTS T cell therapies by using Epstein-Bar virus (EBV)-specific T cells (EBVSTs) expressing a chimeric antigen receptor (CAR) targeting CD30. Allogeneic EBVSTs have not caused GVHD in several clinical trials, while the CD30.CAR, that is effective for the treatment of lymphoma, can also target alloreactive T cells that upregulate CD30 on activation. Although EBVSTs express high levels of CD30, they were protected from fratricide in cis, by the CD30.CAR. Hence, they could proliferate extensively and maintained function both through their native EBV-specific T cell receptor and the CD30.CAR. The CD30.CAR enabled EBVSTs to persist in co-cultures with naive and primed alloreactive T cells and eliminate activated natural killer cells that can also be alloreactive. In conclusion, we show that CD30.CAR EBVSTs have the potential to be an effective OTS therapy against CD30+ tumors and, if successful, could then be used as a platform to target other tumor antigens.
    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
    poly(A)尾的存在对于癌症中基因表达的转录后调节是必不可少的。转录物的这种动态和可修改的特征在各种细胞核和细胞质蛋白的控制下。这项研究旨在开发一种新的细胞质poly(A)相关的标记来预测预后,临床属性,肿瘤免疫微环境(TIME),和肝细胞癌(HCC)的治疗反应。利用来自癌症基因组图谱(TCGA)的RNA测序(RNA-seq)数据,非负矩阵分解(NMF),和主成分分析(PCA)用于将HCC患者分为三个集群,从而证明了细胞质poly(A)尾调节因子的关键预后作用。此外,机器学习算法,如最小绝对收缩和选择算子(LASSO),生存分析,和Cox比例风险模型能够区分不同的细胞质poly(A)亚型。因此,使用国际癌症基因组联盟(ICGC)HCC数据集开发并验证了来自TCGA的5个基因签名。这种基于细胞质poly(A)调节因子的新分类有可能改善预后预测并为化疗提供指导。免疫疗法,和肝动脉化疗栓塞(TACE)在肝癌。
    The presence of a poly(A) tail is indispensable for the post-transcriptional regulation of gene expression in cancer. This dynamic and modifiable feature of transcripts is under the control of various nuclear and cytoplasmic proteins. This study aimed to develop a novel cytoplasmic poly(A)-related signature for predicting prognosis, clinical attributes, tumor immune microenvironment (TIME), and treatment response in hepatocellular carcinoma (HCC). Utilizing RNA sequencing (RNA-seq) data from The Cancer Genome Atlas (TCGA), non-negative matrix factorization (NMF), and principal-component analysis (PCA) were employed to categorize HCC patients into three clusters, thus demonstrating the pivotal prognostic role of cytoplasmic poly(A) tail regulators. Furthermore, machine learning algorithms such as least absolute shrinkage and selection operator (LASSO), survival analysis, and Cox proportional hazards modeling were able to distinguish distinct cytoplasmic poly(A) subtypes. As a result, a 5-gene signature derived from TCGA was developed and validated using International Cancer Genome Consortium (ICGC) HCC datasets. This novel classification based on cytoplasmic poly(A) regulators has the potential to improve prognostic predictions and provide guidance for chemotherapy, immunotherapy, and transarterial chemoembolization (TACE) in HCC.
    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
    推进嵌合抗原受体(CAR)工程化T细胞用于治疗实体瘤是细胞免疫疗法领域的主要焦点。几个障碍阻碍了实体肿瘤中类似的CART细胞临床反应,如血液恶性肿瘤中所见。这些挑战包括靶点外肿瘤毒性,这激发了优化CAR以改善肿瘤抗原选择性和整体安全性的努力。我们最近开发了针对前列腺癌和胰腺癌的针对前列腺干细胞抗原(PSCA)的CART细胞疗法,通过优化细胞内共刺激结构域,显示出改善的临床前抗肿瘤活性和T细胞持久性。进行了类似的研究以优化HER2定向的CART细胞,并对细胞内共刺激结构域进行修饰,以选择性靶向乳腺癌脑转移。在本研究中,我们评估了这些CAR中的各种非信号细胞外间隔区,以进一步提高肿瘤抗原选择性.我们的发现表明,细胞外间隔区的长度和结构可以决定CAR选择性靶向具有高抗原密度的肿瘤细胞的能力。同时保留低抗原密度的细胞。这项研究有助于CAR构建体设计考虑因素,并扩展了我们调整实体肿瘤CART细胞疗法以提高安全性和有效性的知识。
    Advancing chimeric antigen receptor (CAR)-engineered T cells for the treatment of solid tumors is a major focus in the field of cellular immunotherapy. Several hurdles have hindered similar CAR T cell clinical responses in solid tumors as seen in hematological malignancies. These challenges include on-target off-tumor toxicities, which have inspired efforts to optimize CARs for improved tumor antigen selectivity and overall safety. We recently developed a CAR T cell therapy targeting prostate stem cell antigen (PSCA) for prostate and pancreatic cancers, showing improved preclinical antitumor activity and T cell persistence by optimizing the intracellular co-stimulatory domain. Similar studies were undertaken to optimize HER2-directed CAR T cells with modifications to the intracellular co-stimulatory domain for selective targeting of breast cancer brain metastasis. In the present study, we evaluate various nonsignaling extracellular spacers in these CARs to further improve tumor antigen selectivity. Our findings suggest that length and structure of the extracellular spacer can dictate the ability of CARs to selectively target tumor cells with high antigen density, while sparing cells with low antigen density. This study contributes to CAR construct design considerations and expands our knowledge of tuning solid tumor CAR T cell therapies for improved safety and efficacy.
    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
    患有复发性和/或难治性神经母细胞瘤(NB)的儿童的预后令人沮丧。受体酪氨酸激酶样孤儿受体1(ROR1),在NB细胞表面高度表达,为新型免疫治疗提供了潜在的靶标。抗ROR1嵌合抗原受体工程化的离体扩增的外周血自然杀伤(抗ROR1CARexPBNK)细胞代表了这种方法。N-803是具有增强的生物活性的IL-15超激动剂。在这项研究中,我们研究了有或没有N-803的抗ROR1CARexPBNK细胞对ROR1+NB模型的体外和体内抗肿瘤作用。与模拟exPBNK细胞相比,抗ROR1CARexPBNK细胞对ROR1+NB细胞的细胞毒性显着增强,和N-803进一步增加细胞毒性。高维分析显示,N-803增强了有或没有NB细胞的exPBNK和抗ROR1CARexPBNK细胞中的Stat5磷酸化和Ki67水平。在体内,与单独的抗ROR1CARexPBNK相比,抗ROR1CARexPBNK加N-803显着(p<0.05)提高了人ROR1NB异种移植NSG小鼠的存活率。我们的结果为进一步开发抗ROR1CARexPBNK细胞加N-803作为复发性和/或难治性ROR1NB患者的新型联合免疫治疗提供了理论基础。
    The prognosis for children with recurrent and/or refractory neuroblastoma (NB) is dismal. The receptor tyrosine kinase-like orphan receptor 1 (ROR1), which is highly expressed on the surface of NB cells, provides a potential target for novel immunotherapeutics. Anti-ROR1 chimeric antigen receptor engineered ex vivo expanded peripheral blood natural killer (anti-ROR1 CAR exPBNK) cells represent this approach. N-803 is an IL-15 superagonist with enhanced biological activity. In this study, we investigated the in vitro and in vivo anti-tumor effects of anti-ROR1 CAR exPBNK cells with or without N-803 against ROR1+ NB models. Compared to mock exPBNK cells, anti-ROR1 CAR exPBNK cells had significantly enhanced cytotoxicity against ROR1+ NB cells, and N-803 further increased cytotoxicity. High-dimensional analysis revealed that N-803 enhanced Stat5 phosphorylation and Ki67 levels in both exPBNK and anti-ROR1 CAR exPBNK cells with or without NB cells. In vivo, anti-ROR1 CAR exPBNK plus N-803 significantly (p < 0.05) enhanced survival in human ROR1+ NB xenografted NSG mice compared to anti-ROR1 CAR exPBNK alone. Our results provide the rationale for further development of anti-ROR1 CAR exPBNK cells plus N-803 as a novel combination immunotherapeutic for patients with recurrent and/or refractory ROR1+ NB.
    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
    嵌合抗原受体(CAR)T细胞疗法已证明对血液恶性肿瘤的强大功效,但是在治疗实体肿瘤方面仍然存在一些挑战,包括肿瘤异质性,抗原逃逸,和免疫抑制微环境。这里,我们发现SNU398,一种肝细胞癌(HCC)细胞系,高表达成纤维细胞活化蛋白(FAP)和磷脂酰肌醇蛋白聚糖3(GPC3),与患者预后呈负相关。HepG2HCC细胞系高表达GPC3,而SNU387细胞系高表达FAP。因此,我们开发了双特异性CAR-T细胞以同时靶向FAP和GPC3来解决HCC中的肿瘤异质性。抗FAP-GPC3双特异性CAR-T细胞可以识别并被肿瘤细胞表达的FAP或GPC3激活。与抗FAPCAR-T细胞或抗GPC3CAR-T细胞相比,双特异性CAR-T细胞在体外对表达FAP和GPC3的肿瘤细胞实现了更强的活性。抗FAP-GPC3双特异性CAR-T细胞还表现出优异的抗肿瘤功效,并且在体内与单靶标CAR-T细胞相比显著延长小鼠的存活。总的来说,使用抗FAP-GPC3双特异性CAR-T细胞是减少由肿瘤抗原异质性引起的肿瘤复发的有前景的治疗方法.
    Chimeric antigen receptor (CAR) T cell therapy has demonstrated robust efficacy against hematological malignancies, but there are still some challenges regarding treating solid tumors, including tumor heterogeneity, antigen escape, and an immunosuppressive microenvironment. Here, we found that SNU398, a hepatocellular carcinoma (HCC) cell line, exhibited high expression levels of fibroblast activation protein (FAP) and Glypican 3 (GPC3), which were negatively correlated with patient prognosis. The HepG2 HCC cell line highly expressed GPC3, while the SNU387 cell line exhibited high expression of FAP. Thus, we developed bispecific CAR-T cells to simultaneously target FAP and GPC3 to address tumor heterogeneity in HCC. The anti-FAP-GPC3 bispecific CAR-T cells could recognize and be activated by FAP or GPC3 expressed by tumor cells. Compared with anti-FAP CAR-T cells or anti-GPC3 CAR-T cells, bispecific CAR-T cells achieved more robust activity against tumor cells expressing FAP and GPC3 in vitro. The anti-FAP-GPC3 bispecific CAR-T cells also exhibited superior antitumor efficacy and significantly prolonged the survival of mice compared with single-target CAR-T cells in vivo. Overall, the use of anti-FAP-GPC3 bispecific CAR-T cells is a promising treatment approach to reduce tumor recurrence caused by tumor antigen heterogeneity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号