CAR-T-cell therapy

  • 文章类型: Journal Article
    在用于治疗复发性或难治性B细胞血液系统恶性肿瘤的嵌合抗原受体T(CAR-T)细胞疗法中已经取得了重大进展。然而,CAR-T细胞疗法在治疗侵袭性T细胞恶性肿瘤方面尚未取得可比的成功。本文综述了CAR-T细胞疗法在治疗T细胞恶性肿瘤方面的挑战,并总结了该领域的临床前和临床研究进展。我们对CAR-T细胞疗法的临床试验进行了分析,用于治疗按靶抗原分类分组的T细胞恶性肿瘤。此外,这篇综述集中在CAR-T细胞疗法遇到的主要挑战,包括制备过程中由于T细胞靶抗原共享和细胞产物污染而导致的非特异性杀伤。这篇综述讨论了克服这些挑战的策略,提出了新的治疗方法,可以提高CAR-T细胞疗法在T细胞恶性肿瘤治疗中的有效性和适用性。这些思路和策略为以后的研究提供了重要信息,促进CAR-T细胞治疗在该领域的进一步发展和应用。
    Significant advances have been made in chimeric antigen receptor T (CAR-T)-cell therapy for the treatment of recurrent or refractory B-cell hematologic malignancies. However, CAR-T-cell therapy has not yet achieved comparable success in the management of aggressive T-cell malignancies. This article reviews the challenges of CAR-T-cell therapy in treating T-cell malignancies and summarizes the progress of preclinical and clinical studies in this area. We present an analysis of clinical trials of CAR-T-cell therapies for the treatment of T-cell malignancies grouped by target antigen classification. Moreover, this review focuses on the major challenges encountered by CAR-T-cell therapies, including the nonspecific killing due to T-cell target antigen sharing and contamination with cell products during preparation. This review discusses strategies to overcome these challenges, presenting novel therapeutic approaches that could enhance the efficacy and applicability of CAR-T-cell therapy in the treatment of T-cell malignancies. These ideas and strategies provide important information for future studies to promote the further development and application of CAR-T-cell therapy in this field.
    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
    基于Car-T细胞疗法在血液系统恶性肿瘤治疗中取得的令人印象深刻的成功,实体瘤的广泛应用似乎也很有希望。然而,一些重要的障碍需要克服。其中之一当然是鉴定癌细胞上的特异性靶抗原。低甲基化是许多肿瘤实体中的特征性表观遗传畸变。对肿瘤中一致的DNA低甲基化的全基因组筛选能够鉴定异常上调的转录本,这可能导致细胞表面蛋白。因此,这种方法为发现几乎所有肿瘤实体的潜在新型Car-T细胞靶抗原提供了新的视角.首先,我们将这种方法作为前列腺癌的可能治疗方法。
    Based on the impressive success of Car-T-cell therapy in the treatment of hematological malignancies, a broad application for solid tumors also appears promising. However, some important hurdles need to be overcome. One of these is certainly the identification of specific target antigens on cancer cells. Hypomethylation is a characteristic epigenetic aberration in many tumor entities. Genome-wide screenings for consistent DNA hypomethylations in tumors enable the identification of aberrantly upregulated transcripts, which might result in cell surface proteins. Thus, this approach provides a new perspective for the discovery of potential new Car-T-cell target antigens for almost every tumor entity. First, we focus on this approach as a possible treatment for prostate cancer.
    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
    胶质母细胞瘤(GBM)是最常见的原发性恶性脑肿瘤,在手术后联合放化疗的标准治疗下,中位总生存期不到2年,死亡率接近100%。因此,迫切需要新的治疗策略。嵌合抗原受体(CAR)T细胞在血液癌症中的成功促进了对GBM的CAR-T细胞治疗的临床前和临床研究。然而,最近的试验没有显示出任何重大的成功。这里,我们描述了阻碍CAR-T细胞治疗GBM有效性的现有挑战,包含寒冷(免疫抑制)微环境,肿瘤异质性,T细胞耗尽,局部和全身免疫抑制,和中枢神经系统(CNS)实质固有的免疫特权。此外,我们研究了在开发下一代CAR-T细胞和新的创新方法方面取得的进展,如低强度脉冲聚焦超声,旨在克服GBMCAR-T细胞疗法的当前障碍。
    Glioblastoma (GBM) is the most common primary malignant brain tumor, with a median overall survival of less than 2 years and a nearly 100% mortality rate under standard therapy that consists of surgery followed by combined radiochemotherapy. Therefore, new therapeutic strategies are urgently needed. The success of chimeric antigen receptor (CAR) T cells in hematological cancers has prompted preclinical and clinical investigations into CAR-T-cell treatment for GBM. However, recent trials have not demonstrated any major success. Here, we delineate existing challenges impeding the effectiveness of CAR-T-cell therapy for GBM, encompassing the cold (immunosuppressive) microenvironment, tumor heterogeneity, T-cell exhaustion, local and systemic immunosuppression, and the immune privilege inherent to the central nervous system (CNS) parenchyma. Additionally, we deliberate on the progress made in developing next-generation CAR-T cells and novel innovative approaches, such as low-intensity pulsed focused ultrasound, aimed at surmounting current roadblocks in GBM CAR-T-cell 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: Case Reports
    尽管嵌合抗原受体T细胞(CAR-T)疗法显著改善了复发/难治性B细胞恶性肿瘤的预后,受者患有严重的体液免疫缺陷。此外,2019年冠状病毒病(COVID-19)患者预后不良,正如在输注前接受COVID-19治疗的几例病例报告中指出的那样。我们报道了一例70岁的女性,她在接受CAR-T治疗高级别B细胞淋巴瘤之前立即患上了COVID-19。她接受了Tixagevimab-Cilgavimab化疗和放疗,但从未缓解。她被转到我们医院接受CAR-T治疗,但开发了COVID-19。她的症状很轻,接受了长期的molnupiravir治疗。感染后第28天,在聚合酶链反应(PCR)检测阴性后,患者重新开始淋巴消耗性化疗.患者未出现COVID-19症状或严重细胞因子释放综合征的复发。根据以往报告与本案的分析比较,我们认为CAR-T治疗应该推迟到PCR检测阴性后进行.此外,Tixagevimab-Cilgavimab和长期直接抗病毒药物治疗可有效预防重症COVID-19并缩短感染持续时间。
    Although chimeric antigen receptor T-cell (CAR-T) therapies have dramatically improved the outcomes of relapsed/refractory B-cell malignancies, recipients suffer from severe humoral immunodeficiencies. Furthermore, patients with coronavirus disease 2019 (COVID-19) have a poor prognosis, as noted in several case reports of recipients who had COVID-19 before the infusion. We report the case of a 70-year-old woman who developed COVID-19 immediately before CAR-T therapy for high-grade B-cell lymphoma. She received Tixagevimab-Cilgavimab chemotherapy and radiation therapy but never achieved remission. She was transferred to our hospital for CAR-T therapy, but developed COVID-19. Her symptoms were mild and she was treated with long-term molnupiravir. On day 28 post-infection, lymphodepleting chemotherapy was restarted after a negative polymerase chain reaction (PCR) test was confirmed. The patient did not experience recurrence of COVID-19 symptoms or severe cytokine release syndrome. Based on the analysis and comparison of the previous reports with this case, we believe that CAR-T therapy should be postponed until a negative PCR test is confirmed. In addition, Tixagevimab-Cilgavimab and long term direct-acting antiviral agent treatment can be effective prophylaxis for severe COVID-19 and shortening the duration of infection.
    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
    尽管嵌合抗原受体T(CAR-T)细胞疗法在血液恶性肿瘤中取得了巨大的进展,由于肿瘤微环境(TME)中的T细胞耗尽和细胞因子过度释放引起的全身毒性,它们在实体瘤中的应用受到很大限制.作为免疫抑制TME的关键调节剂,TGF-β通过NF-κB途径促进细胞因子合成。这里,我们共表达SMAD7,TGF-β信号的抑制因子,在工程化T细胞中使用HER2靶向CAR。这些新型CAR-T细胞显示出高的细胞溶解功效,并且对TGF-β触发的耗竭具有抗性。这使得持续抗原暴露后具有持续的杀肿瘤能力。此外,SMAD7基本上减少了抗原引发的CAR-T细胞产生的炎性细胞因子。机械上,SMAD7下调TGF-β受体I并消除了CAR-T细胞中TGF-β和NF-κB途径之间的相互作用。因此,这些CAR-T细胞持续抑制肿瘤生长并促进肿瘤攻击小鼠的存活,而不管高浓度TGF-β引起的不利肿瘤微环境如何.SMAD7共表达还增强了患者来源的肿瘤类器官中的CAR-T细胞浸润和持续激活。因此,我们的研究证明了SMAD7共表达作为提高CAR-T细胞治疗实体肿瘤疗效和安全性的新方法的可行性.
    Despite the tremendous progress of chimeric antigen receptor T (CAR-T) cell therapy in hematological malignancies, their application in solid tumors has been limited largely due to T-cell exhaustion in the tumor microenvironment (TME) and systemic toxicity caused by excessive cytokine release. As a key regulator of the immunosuppressive TME, TGF-β promotes cytokine synthesis via the NF-κB pathway. Here, we coexpressed SMAD7, a suppressor of TGF-β signaling, with a HER2-targeted CAR in engineered T cells. These novel CAR-T cells displayed high cytolytic efficacy and were resistant to TGF-β-triggered exhaustion, which enabled sustained tumoricidal capacity after continuous antigen exposure. Moreover, SMAD7 substantially reduced the production of inflammatory cytokines by antigen-primed CAR-T cells. Mechanistically, SMAD7 downregulated TGF-β receptor I and abrogated the interplay between the TGF-β and NF-κB pathways in CAR-T cells. As a result, these CAR-T cells persistently inhibited tumor growth and promoted the survival of tumor-challenged mice regardless of the hostile tumor microenvironment caused by a high concentration of TGF-β. SMAD7 coexpression also enhanced CAR-T-cell infiltration and persistent activation in patient-derived tumor organoids. Therefore, our study demonstrated the feasibility of SMAD7 coexpression as a novel approach to improve the efficacy and safety of CAR-T-cell therapy for solid tumors.
    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CAR-T细胞疗法在B-ALL中首次取得巨大成功以来,工程免疫细胞治疗血液恶性肿瘤一直是研究的主要焦点。现在可以在高度治疗难治性或复发性病症中治疗几种疾病。目前,许多CD19或BCMA特异性CAR-T细胞疗法被批准用于急性淋巴细胞白血病(ALL),弥漫性大B细胞淋巴瘤(DLBCL),套细胞淋巴瘤(MCL),多发性骨髓瘤(MM),和滤泡性淋巴瘤(FL)。即使在先前预后非常差的情况下,这些疗法的实施也显着改善了患者的预后和生存率。在这次全面审查中,我们介绍了研究的现状,最近的创新,以及CAR-T细胞疗法在一组选定的血液系统恶性肿瘤中的应用。我们专注于B细胞和T细胞恶性肿瘤,包括皮肤和外周T细胞淋巴瘤(T-ALL,PTCL,CTCL),急性髓系白血病(AML),慢性粒细胞白血病(CML),慢性淋巴细胞白血病(CLL),经典霍奇金淋巴瘤(HL),伯基特淋巴瘤(BL),毛细胞白血病(HCL),和Waldenström的巨球蛋白血症(WM)。虽然这些疾病是高度异质性的,我们强调了几种类似使用的方法(与既定的疗法相结合,健康细胞上的目标消耗),用于多种疾病的靶标(CD30,CD38,TRBC1/2),和独特的功能,需要个性化的方法。此外,我们关注当前CAR-T细胞疗法在个体疾病和实体中的局限性,例如免疫受损的肿瘤微环境(TME),上靶-外肿瘤效应的风险,以及不良事件发生的差异。最后,我们对CAR-T细胞工程方面的创新进行了展望,如人工智能的使用,以及CAR-T细胞在日常临床实践治疗方案中的未来作用.
    Engineering immune cells to treat hematological malignancies has been a major focus of research since the first resounding successes of CAR-T-cell therapies in B-ALL. Several diseases can now be treated in highly therapy-refractory or relapsed conditions. Currently, a number of CD19- or BCMA-specific CAR-T-cell therapies are approved for acute lymphoblastic leukemia (ALL), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), multiple myeloma (MM), and follicular lymphoma (FL). The implementation of these therapies has significantly improved patient outcome and survival even in cases with previously very poor prognosis. In this comprehensive review, we present the current state of research, recent innovations, and the applications of CAR-T-cell therapy in a selected group of hematologic malignancies. We focus on B- and T-cell malignancies, including the entities of cutaneous and peripheral T-cell lymphoma (T-ALL, PTCL, CTCL), acute myeloid leukemia (AML), chronic myeloid leukemia (CML), chronic lymphocytic leukemia (CLL), classical Hodgkin-Lymphoma (HL), Burkitt-Lymphoma (BL), hairy cell leukemia (HCL), and Waldenström\'s macroglobulinemia (WM). While these diseases are highly heterogenous, we highlight several similarly used approaches (combination with established therapeutics, target depletion on healthy cells), targets used in multiple diseases (CD30, CD38, TRBC1/2), and unique features that require individualized approaches. Furthermore, we focus on current limitations of CAR-T-cell therapy in individual diseases and entities such as immunocompromising tumor microenvironment (TME), risk of on-target-off-tumor effects, and differences in the occurrence of adverse events. Finally, we present an outlook into novel innovations in CAR-T-cell engineering like the use of artificial intelligence and the future role of CAR-T cells in therapy regimens in everyday clinical practice.
    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
    急性肾损伤和慢性肾病在多发性骨髓瘤中很常见。氟达拉滨是CAR-T细胞疗法之前淋巴清除的一部分,不建议用于严重肾功能损害的患者,其定义为肾小球滤过率低于30ml/min/1.73ml。我们在Ideecabtagenvicleucel输注前,将氟达拉滨作为淋巴耗竭的一部分,对一名58岁的骨髓瘤相关严重肾功能不全的女性患者进行了治疗。以减少的剂量(15mg/m2)施用氟达拉滨,并以300mg/m2的剂量施用环磷酰胺,随后使用更大的过滤器(FX-100)在6小时内进行血液透析。该疗法具有良好的耐受性,具有出色的CAR-T细胞扩增和完全缓解,目前正在超过12个月。
    Acute kidney injury and chronic kidney disease is common in multiple myeloma. Fludarabine which is part of lymphodepletion before CAR-T cell therapy is renally eliminated and its use is not recommended for patients with severe renal impairment defined as a glomerular filtration rate below 30ml/min/1.73m2. We administered fludarabine to a 58-year-old female patient with myeloma-associated severe renal impairment as part of lymphodepletion before Idecabtagen vicleucel infusion. Fludarabine was administered in reduced dose (15mg/m2) and cyclophosphamide with a dose of 300mg/m2 followed by hemodialysis over six hours using a larger filter (FX-100). The therapy was well tolerated with excellent CAR-T cell expansion and complete remission which is ongoing now beyond 12 months.
    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
    在多发性骨髓瘤(MM)的治疗干预领域取得了重大进展,导致其临床管理的变革性转变。虽然传统的方式,如手术,放射治疗,化疗改善了临床结果,为患有复发性和难治性MM(RRMM)的患者进行综合治疗的首要挑战仍然存在。值得注意的是,过继细胞疗法,特别是嵌合抗原受体T细胞(CAR-T)治疗,已在难治性或耐药性B细胞恶性肿瘤患者中显示出疗效,现在也正在MM患者中进行测试。在此背景下,B细胞成熟抗原(BCMA)已成为MM中CAR-T细胞抗原靶向的有希望的候选者。替代靶点包括SLAMF7、CD38、CD19、淋巴细胞活化信号分子CS1、NKG2D、CD138许多临床研究已经证明了这些CAR-T细胞疗法的临床疗效,虽然纵向随访显示一定程度的抗原逃逸。CAR-T细胞疗法的广泛实施受到几个障碍的阻碍,包括抗原逃避,实体癌瘤内浸润不均,细胞因子释放综合征,神经毒性,后勤实施,和财政负担。本文概述了CAR-T细胞治疗MM以及BCMA作为靶抗原的应用。以及其他潜在目标部分的概述。
    Significant progress has been achieved in the realm of therapeutic interventions for multiple myeloma (MM), leading to transformative shifts in its clinical management. While conventional modalities such as surgery, radiotherapy, and chemotherapy have improved the clinical outcomes, the overarching challenge of effecting a comprehensive cure for patients afflicted with relapsed and refractory MM (RRMM) endures. Notably, adoptive cellular therapy, especially chimeric antigen receptor T-cell (CAR-T) therapy, has exhibited efficacy in patients with refractory or resistant B-cell malignancies and is now also being tested in patients with MM. Within this context, the B-cell maturation antigen (BCMA) has emerged as a promising candidate for CAR-T-cell antigen targeting in MM. Alternative targets include SLAMF7, CD38, CD19, the signaling lymphocyte activation molecule CS1, NKG2D, and CD138. Numerous clinical studies have demonstrated the clinical efficacy of these CAR-T-cell therapies, although longitudinal follow-up reveals some degree of antigenic escape. The widespread implementation of CAR-T-cell therapy is encumbered by several barriers, including antigenic evasion, uneven intratumoral infiltration in solid cancers, cytokine release syndrome, neurotoxicity, logistical implementation, and financial burden. This article provides an overview of CAR-T-cell therapy in MM and the utilization of BCMA as the target antigen, as well as an overview of other potential target moieties.
    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细胞疗法彻底改变了急性淋巴细胞白血病的治疗,非霍奇金淋巴瘤,和多发性骨髓瘤,但以独特的毒性为代价,包括细胞因子释放综合征,免疫效应细胞相关神经毒性综合征,和长期的“靶向肿瘤外”效应。
    方法:所有这些因素都会增加已经高度免疫受损的患者人群的感染风险。的确,感染性并发症是CAR-T细胞术后非复发死亡率的关键决定因素.这些风险因素的时间分布形成了CAR-T细胞输注后早期与晚期的不同感染模式。此外,由于其靶标在不同分化阶段的B谱系细胞上的表达,CD19和B细胞成熟抗原(BCMA)CAR-T细胞诱导不同的免疫缺陷,可能需要不同的预防策略。感染发生率在输注后的第一个月期间最高,随后下降。然而,即使在输液一年后,感染仍然相对常见。
    结果:CD19后早期细菌感染占主导地位,而在BCMACAR-T细胞治疗后,细菌和病毒病因之间的分布更加平等。真菌感染普遍罕见。巨细胞病毒(CMV)和其他疱疹病毒的报道越来越多,但是是否有必要对所有人进行例行监测,或患者的一个亚组,还有待确定。不同中心的临床实践差异很大,许多不确定性仍然存在,包括CMV监测,抗菌和抗真菌预防和持续时间,使用免疫球蛋白替代疗法,接种疫苗的时间。
    结论:风险分层工具可用,可能有助于区分输液后发热的感染性和非感染性原因,并预测严重感染。这些工具需要前瞻性验证,它们在临床实践中的整合需要系统的研究。
    BACKGROUND: Chimeric antigen receptor (CAR)-T-cell therapies have revolutionized the management of acute lymphoblastic leukemia, non-Hodgkin lymphoma, and multiple myeloma but come at the price of unique toxicities, including cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and long-term \"on-target off-tumor\" effects.
    METHODS: All of these factors increase infection risk in an already highly immunocompromised patient population. Indeed, infectious complications represent the key determinant of non-relapse mortality after CAR-T cells. The temporal distribution of these risk factors shapes different infection patterns early versus late post-CAR-T-cell infusion. Furthermore, due to the expression of their targets on B lineage cells at different stages of differentiation, CD19, and B-cell maturation antigen (BCMA) CAR-T cells induce distinct immune deficits that could require different prevention strategies. Infection incidence is the highest during the first month post-infusion and subsequently decreases thereafter. However, infections remain relatively common even a year after infusion.
    RESULTS: Bacterial infections predominate early after CD19, while a more equal distribution between bacterial and viral causes is seen after BCMA CAR-T-cell therapy, and fungal infections are universally rare. Cytomegalovirus (CMV) and other herpesviruses are increasingly breported, but whether routine monitoring is warranted for all, or a subgroup of patients, remains to be determined. Clinical practices vary substantially between centers, and many areas of uncertainty remain, including CMV monitoring, antibacterial and antifungal prophylaxis and duration, use of immunoglobulin replacement therapy, and timing of vaccination.
    CONCLUSIONS: Risk stratification tools are available and may help distinguish between infectious and non-infectious causes of fever post-infusion and predict severe infections. These tools need prospective validation, and their integration in clinical practice needs to be systematically studied.
    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
    自2019年冠状病毒病(COVID-19)被宣布为全球大流行以来,已经过去了3年多,然而,COVID-19仍然严重影响免疫功能低下的个体,包括接受造血细胞移植(HCT)和嵌合抗原受体-T细胞疗法治疗的个体,这些个体仍然存在严重COVID-19和死亡的高风险。尽管疫苗接种努力,这些患者由于免疫抑制反应不足,这强调了需要额外的预防措施。最佳时机,疫苗接种时间表,和免疫相关的保护性免疫仍然未知。疾病早期使用的抗病毒疗法可以降低COVID-19导致的死亡率和严重程度。抗病毒药物的组合或顺序使用可能有利于控制复制,并防止长期COVID-19中治疗相关突变的发展。尽管数据相互矛盾,COVID-19恢复期血浆仍然是免疫功能低下的轻中度疾病患者预防进展的一种选择。这些患者越来越认识到长期的COVID-19,并与SARS-CoV-2变体的宿主内出现有关。最后,新型SARS-CoV2特异性T细胞和自然杀伤细胞增强(或含有)产品可能对多种变体具有活性,并且是免疫功能低下患者的有希望的治疗方法.
    More than 3 years have passed since Coronavirus disease 2019 (COVID-19) was declared a global pandemic, yet COVID-19 still severely impacts immunocompromised individuals including those treated with hematopoietic cell transplantation (HCT) and chimeric antigen receptor-T-cell therapies who remain at high risk for severe COVID-19 and mortality. Despite vaccination efforts, these patients have inadequate responses due to immunosuppression, which underscores the need for additional preventive approaches. The optimal timing, schedule of vaccination, and immunological correlates for protective immunity remain unknown. Antiviral therapies used early during disease can reduce mortality and severity due to COVID-19. The combination or sequential use of antivirals could be beneficial to control replication and prevent the development of treatment-related mutations in protracted COVID-19. Despite conflicting data, COVID-19 convalescent plasma remains an option in immunocompromised patients with mild-to-moderate disease to prevent progression. Protracted COVID-19 has been increasingly recognized among these patients and has been implicated in intra-host emergence of SARS-CoV-2 variants. Finally, novel SARS-CoV2-specific T-cells and natural killer cell-boosting (or -containing) products may be active against multiple variants and are promising therapies in immunocompromised patients.
    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