chimeric antigen receptor (CAR)

嵌合抗原受体 (CAR)
  • 文章类型: Journal Article
    CART细胞由于其在选定的血液恶性肿瘤中的显着临床反应率而产生了极大的兴奋。然而,这些工程免疫细胞是活的药物,给药后难以控制。
    我们讨论了小分子调节的开关系统,该系统可能用于控制患者体内的CAR-T细胞功能,以及CAR-T细胞领域最重要的障碍,这些开关系统可能会克服。
    迫切需要开发先进的交换机系统。一旦可用,我们预计它们将为未来的CAR-T细胞世代开辟新的途径。
    UNASSIGNED: CAR T cells have generated great excitement due to their remarkable clinical response rates in selected hematologic malignancies. However, these engineered immune cells are living drugs which are hard to control after administration.
    UNASSIGNED: We discuss small molecule-regulated switch systems which can potentially be used to control CAR T cell function within the patient, as well as the most important obstacles in the CAR T cell field, which might be overcome with those switch systems.
    UNASSIGNED: There is an urgent need to develop advanced switch systems. Once available, we expect that they will open up new avenues for future CAR T cell generations.
    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
    CAR-T细胞疗法在治疗B细胞恶性肿瘤方面显示出非凡的前景,这引发了人们对其治疗其他类型癌症的潜力的乐观。然而,CAR-T细胞治疗实体肿瘤和非B细胞血液系统恶性肿瘤的期望尚未实现。此外,关于病毒载体使用和当前个性化生产过程的安全问题是限制其广泛使用的其他瓶颈。近年来,基因编辑技术在CAR-T细胞治疗中的应用为释放CAR-T细胞治疗的潜在潜力和减轻其相关挑战开辟了一条新途径。此外,基因编辑工具为以完全非病毒的方法制造CAR-T细胞铺平了道路,并提供了一种通用的,现成的产品。尽管基因编辑策略有很多优势,经典基因编辑工具的脱靶活性(ZFN,TLENs,和CRISPR/Cas9)仍然是一个主要问题。因此,近年来已经做出了一些努力来减少它们的脱靶活性和遗传毒性,导致引入具有改进安全性的先进基因编辑工具。在这次审查中,我们首先研究先进的基因编辑工具,概述了这些技术目前如何应用于CAR-T细胞疗法的临床试验。在此之后,我们探索了旨在增强CAR-T细胞治疗安全性和有效性的各种基因编辑策略.
    CAR-T cell therapy has shown remarkable promise in treating B-cell malignancies, which has sparked optimism about its potential to treat other types of cancer as well. Nevertheless, the Expectations of CAR-T cell therapy in solid tumors and non-B cell hematologic malignancies have not been met. Furthermore, safety concerns regarding the use of viral vectors and the current personalized production process are other bottlenecks that limit its widespread use. In recent years the use of gene editing technology in CAR-T cell therapy has opened a new way to unleash the latent potentials of CAR-T cell therapy and lessen its associated challenges. Moreover, gene editing tools have paved the way to manufacturing CAR-T cells in a fully non-viral approach as well as providing a universal, off-the-shelf product. Despite all the advantages of gene editing strategies, the off-target activity of classical gene editing tools (ZFNs, TALENs, and CRISPR/Cas9) remains a major concern. Accordingly, several efforts have been made in recent years to reduce their off-target activity and genotoxicity, leading to the introduction of advanced gene editing tools with an improved safety profile. In this review, we begin by examining advanced gene editing tools, providing an overview of how these technologies are currently being applied in clinical trials of CAR-T cell therapies. Following this, we explore various gene editing strategies aimed at enhancing the safety and efficacy of 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: Journal Article
    表观遗传机制涉及几种细胞功能,它们在免疫系统中的作用至关重要。组蛋白脱乙酰酶(HDAC)是调节和催化脱乙酰过程的一组重要酶。HDAC已被证明是改善免疫疗法功效的有益靶标。HDAC11是一种参与T细胞功能负调节的酶。这里,我们研究了在CAR-T细胞中使用RNA干扰下调HDAC11以改善针对前列腺癌的免疫治疗结果的潜力.我们设计并测试了靶向HDAC11的四种不同的短发夹RNA(shRNA)序列,以鉴定对后续分析最有效的一种。HDAC11缺陷型CAR-T细胞(shD-NKG2D-CAR-T)对前列腺癌细胞系表现出比野生型CAR-T细胞更好的细胞毒性。这种效应归因于增强的激活,脱粒,shD-NKG2D-CAR-T与前列腺癌细胞系共培养时的细胞因子释放能力。我们的研究结果表明,HDAC11干扰显着增强CAR-T细胞增殖,减少耗竭标志物PD-1和TIM3,促进T中枢记忆中医群体的形成。对潜在分子机制的进一步探索揭示了转录因子Eomes的表达增加,提供对CAR-T细胞分化调控的见解。最后,shD-NKG2D-CAR-T细胞提供了有效的肿瘤控制,导致与野生型对应物相比,荷瘤小鼠的体内存活率提高.目前的研究强调了HDAC11下调在改善CAR-T细胞治疗中的潜力。该研究将为进一步研究铺平道路,重点是理解和利用免疫治疗结果的表观遗传机制。
    Epigenetic mechanisms are involved in several cellular functions, and their role in the immune system is of prime importance. Histone deacetylases (HDACs) are an important set of enzymes that regulate and catalyze the deacetylation process. HDACs have been proven beneficial targets for improving the efficacy of immunotherapies. HDAC11 is an enzyme involved in the negative regulation of T cell functions. Here, we investigated the potential of HDAC11 downregulation using RNA interference in CAR-T cells to improve immunotherapeutic outcomes against prostate cancer. We designed and tested four distinct short hairpin RNA (shRNA) sequences targeting HDAC11 to identify the most effective one for subsequent analyses. HDAC11-deficient CAR-T cells (shD-NKG2D-CAR-T) displayed better cytotoxicity than wild-type CAR-T cells against prostate cancer cell lines. This effect was attributed to enhanced activation, degranulation, and cytokine release ability of shD-NKG2D-CAR-T when co-cultured with prostate cancer cell lines. Our findings reveal that HDAC11 interference significantly enhances CAR-T cell proliferation, diminishes exhaustion markers PD-1 and TIM3, and promotes the formation of T central memory TCM populations. Further exploration into the underlying molecular mechanisms reveals increased expression of transcription factor Eomes, providing insight into the regulation of CAR-T cell differentiation. Finally, the shD-NKG2D-CAR-T cells provided efficient tumor control leading to improved survival of tumor-bearing mice in vivo as compared to their wild-type counterparts. The current study highlights the potential of HDAC11 downregulation in improving CAR-T cell therapy. The study will pave the way for further investigations focused on understanding and exploiting epigenetic mechanisms for immunotherapeutic 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
    实体肿瘤微环境中T细胞有限的扩增能力和功能失活是体内使用肿瘤浸润淋巴细胞(TIL)的应用中面临的主要问题。我们试图确定携带PD-1-CD28增强受体和CD19CAR的TIL是否可以克服这一限制并介导肿瘤消退。首先,PD-1-CD28增强受体或CD19CAR修饰的NY-ESO-1-TCR-T细胞模拟TIL功能的抗肿瘤作用(以下简称“PD-1-CD28-TCR-T”或“CD19CAR-TCR-T”细胞,分别)在体外和荷瘤模型中使用NY-ESO-1过表达的肿瘤细胞系进行了测试。此外,在体内评估了S-TIL(通过用编码PD-1-CD28-T2A-CD19CAR的质粒转导修饰的TIL)的安全性和抗肿瘤能力.PD-1-CD28-TCR-T细胞显示出强大的抗肿瘤能力,该能力在体内不受PD-1/PD-L1信号传导的影响。用CD19+B细胞刺激的CD19CAR-TCR-T细胞在体外和体内都表现出强大的扩增和抗肿瘤能力。3例难治性实体瘤患者接受了S-TIL输注。没有观察到治疗相关的死亡率,而且没有患者出现严重的副作用。一名黑色素瘤患者获得了部分反应,2例结肠癌或肾癌患者在接受S-TIL治疗后病情长期稳定.据我们所知,这是第一项描述自体S-TIL过继转移以控制晚期癌症患者疾病的安全性和有效性的研究,这表明S-TIL可能是一种有前途的癌症替代疗法。
    The limited expansion ability and functional inactivation of T cells within the solid tumor microenvironment are major problems faced during in the application of using tumor-infiltrating lymphocytes (TILs) in vivo. We sought to determine whether TILs carrying a PD-1-CD28-enhanced receptor and CD19 CAR could overcome this limitation and mediate tumor regression. First, anti-tumor effects of PD-1-CD28-enhanced receptor or CD19 CAR modified NY-ESO-1-TCR-T cells to mimic the TILs function (hereafter \"PD-1-CD28-TCR-T\" or \"CD19 CAR-TCR-T\" cells, respectively) were tested using the NY-ESO-1 over-expressed tumor cell line in vitro and in a tumor-bearing model. Furthermore, the safety and anti-tumor ability of S-TILs (TILs modified through transduction with a plasmid encoding the PD-1-CD28-T2A-CD19 CAR) were evaluated in vivo. PD-1-CD28-TCR-T cells showed a formidable anti-tumor ability that was not subject to PD-1/PD-L1 signaling in vivo. CD19 CAR-TCR-T cells stimulated with CD19+ B cells exhibited powerful expansion and anti-tumor abilities both in vitro and in vivo. Three patients with refractory solid tumors received S-TILs infusion. No treatment-related mortality was observed, and none of the patients experienced serious side effects. One patient with melanoma achieved a partial response, and two patients with colon or kidney cancer achieved long-term stable disease following S-TILs therapy. To the best of our knowledge, this is the first study describing the safety and efficacy of the adoptive transfer of autologous S-TILs to control disease in patients with advanced cancers, suggesting that S-TILs may be a promising alternative therapy for cancer.
    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(CAR)-NK细胞的最新进展在临床试验中以最小的毒性治疗CD19阳性淋巴肿瘤[包括移植物抗宿主病(GvHD)和细胞因子释放综合征(CRS)。然而,CAR-NK细胞在对抗病毒感染中的应用尚未得到充分探索.先前的研究表明,表达S309单链片段可变(scFv)的CAR-NK细胞,以下S309-CAR-NK细胞,可以与SARS-CoV-2野生型假病毒(PV)结合,并在体外有效杀死表达野生型刺突蛋白的细胞。在这项研究中,我们进一步证明了S309-CAR-NK细胞可以结合不同的SARS-CoV-2变体,包括B.1.617.2(Delta),B.1.621(Mu),和体外B.1.1.529(Omicron)变体。我们还表明,S309-CAR-NK细胞降低了表达人血管紧张素转换酶2(hACE2)受体的NOD/SCIDγ(NSG)小鼠的病毒载量,这些小鼠受到SARS-CoV-2野生型(USA/WA1/2020)的攻击。我们的研究表明,S309-CAR-NK细胞可能用于抑制SARS-CoV-2感染,并可能用于治疗对目前可用的其他疗法无反应的COVID-19患者。
    目的:嵌合抗原受体(CAR)-NK细胞可以成为治疗各种疾病的“现成”产品,包括癌症,感染,和自身免疫性疾病。在这项研究中,我们改造自然杀伤(NK)细胞表达S309单链片段可变(scFv),靶向SARS-CoV-2的刺突蛋白,以下称为S309-CAR-NK细胞。我们的研究表明,S309-CAR-NK细胞对不同的SARS-CoV-2变体有效,包括B.1.617.2(Delta),B.1.621(Mu),和B.1.1.529(Omicron)变体。S309-CAR-NK细胞可以(i)直接结合SARS-CoV-2假型病毒(PV),(ii)与表达人血管紧张素转换酶2(hACE2)受体的293T细胞(293T-hACE2细胞)竞争性结合SARS-CoV-2PV,(iii)特异性靶向并裂解表达刺突蛋白的A549细胞,和(iv)显著降低表达hACE2的NOD/SCIDγ(NSG)小鼠(hACE2-NSG小鼠)的肺中SARS-CoV-2野生型(菌株USA/WA1/2020)的病毒载量。总之,本研究表明,S309-CAR-NK免疫疗法可作为COVID-19患者的替代疗法.
    Recent progress on chimeric antigen receptor (CAR)-NK cells has shown promising results in treating CD19-positive lymphoid tumors with minimal toxicities [including graft versus host disease (GvHD) and cytokine release syndrome (CRS) in clinical trials. Nevertheless, the use of CAR-NK cells in combating viral infections has not yet been fully explored. Previous studies have shown that CAR-NK cells expressing S309 single-chain fragment variable (scFv), hereinafter S309-CAR-NK cells, can bind to SARS-CoV-2 wildtype pseudotyped virus (PV) and effectively kill cells expressing wild-type spike protein in vitro. In this study, we further demonstrate that the S309-CAR-NK cells can bind to different SARS-CoV-2 variants, including the B.1.617.2 (Delta), B.1.621 (Mu), and B.1.1.529 (Omicron) variants in vitro. We also show that S309-CAR-NK cells reduce virus loads in the NOD/SCID gamma (NSG) mice expressing the human angiotensin-converting enzyme 2 (hACE2) receptor challenged with SARS-CoV-2 wild-type (strain USA/WA1/2020). Our study demonstrates the potential use of S309-CAR-NK cells for inhibiting infection by SARS-CoV-2 and for the potential treatment of COVID-19 patients unresponsive to otherwise currently available therapeutics.
    OBJECTIVE: Chimeric antigen receptor (CAR)-NK cells can be \"off-the-shelf\" products that treat various diseases, including cancer, infections, and autoimmune diseases. In this study, we engineered natural killer (NK) cells to express S309 single-chain fragment variable (scFv), to target the Spike protein of SARS-CoV-2, hereinafter S309-CAR-NK cells. Our study shows that S309-CAR-NK cells are effective against different SARS-CoV-2 variants, including the B.1.617.2 (Delta), B.1.621 (Mu), and B.1.1.529 (Omicron) variants. The S309-CAR-NK cells can (i) directly bind to SARS-CoV-2 pseudotyped virus (PV), (ii) competitively bind to SARS-CoV-2 PV with 293T cells expressing the human angiotensin-converting enzyme 2 (hACE2) receptor (293T-hACE2 cells), (iii) specifically target and lyse A549 cells expressing the spike protein, and (iv) significantly reduce the viral loads of SARS-CoV-2 wild-type (strain USA/WA1/2020) in the lungs of NOD/SCID gamma (NSG) mice expressing hACE2 (hACE2-NSG mice). Altogether, the current study demonstrates the potential use of S309-CAR-NK immunotherapy as an alternative treatment for COVID-19 patients.
    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
    实体癌髓系细胞在实体癌中普遍存在,但它们经常表现出有助于免疫抑制肿瘤微环境(TME)的抗炎肿瘤表型,这阻碍了癌症免疫疗法的有效性。髓系细胞天然的肿瘤运输能力使得工程骨髓细胞治疗成为一种有趣的方法来应对实体癌症带来的挑战。包括肿瘤浸润,肿瘤细胞异质性和免疫抑制TME。一种这样的工程方法是靶向检查点分子PD-L1,其通常被实体癌上调以逃避免疫应答。
    在这里,我们设计了一种基于表达嵌合抗原受体(CAR)样免疫受体(CARIR)的骨髓细胞的过继细胞治疗策略。CARIR的胞外结构域源自天然抑制性受体PD-1,而胞内结构域源自CD40和/或CD3ζ。为了评估CARIR工程骨髓细胞的疗效,我们在体外使用共培养和基于流式细胞术的吞噬作用试验进行了原理验证实验.此外,我们采用了完全免疫活性的同系肿瘤小鼠模型来评估该策略在体内的有效性。
    将表达CARIR的人单核细胞THP-1细胞与表达PD-L1的靶细胞共培养导致共刺激分子CD86的上调以及促炎细胞因子TNF-1α和IL-1β的表达。此外,CARIR表达在体外显著增强多种表达PD-L1的癌细胞系的吞噬作用。用表达CARIR的人原代巨噬细胞观察到类似的结果。在携带4T1乳腺肿瘤的同基因BALB/c小鼠中进行的实验中,输注表达小鼠版本CARIR的小鼠骨髓细胞显着减慢了肿瘤的生长并延长了生存期。
    放在一起,这些结果表明,PD-1CARIR工程骨髓细胞的过继转移是治疗PD-L1阳性实体癌的一种有前景的策略.
    UNASSIGNED: Solid cancers Myeloid cells are prevalent in solid cancers, but they frequently exhibit an anti-inflammatory pro-tumor phenotype that contribute to the immunosuppressive tumor microenvironment (TME), which hinders the effectiveness of cancer immunotherapies. Myeloid cells\' natural ability of tumor trafficking makes engineered myeloid cell therapy an intriguing approach to tackle the challenges posed by solid cancers, including tumor infiltration, tumor cell heterogenicity and the immunosuppressive TME. One such engineering approach is to target the checkpoint molecule PD-L1, which is often upregulated by solid cancers to evade immune responses.
    UNASSIGNED: Here we devised an adoptive cell therapy strategy based on myeloid cells expressing a Chimeric Antigen Receptor (CAR)-like immune receptor (CARIR). The extracellular domain of CARIR is derived from the natural inhibitory receptor PD-1, while the intracellular domain(s) are derived from CD40 and/or CD3ζ. To assess the efficacy of CARIR-engineered myeloid cells, we conducted proof-of-principle experiments using co-culture and flow cytometry-based phagocytosis assays in vitro. Additionally, we employed a fully immune-competent syngeneic tumor mouse model to evaluate the strategy\'s effectiveness in vivo.
    UNASSIGNED: Co-culturing CARIR-expressing human monocytic THP-1 cells with PD-L1 expressing target cells lead to upregulation of the costimulatory molecule CD86 along with expression of proinflammatory cytokines TNF-1α and IL-1β. Moreover, CARIR expression significantly enhanced phagocytosis of multiple PD-L1 expressing cancer cell lines in vitro. Similar outcomes were observed with CARIR-expressing human primary macrophages. In experiments conducted in syngeneic BALB/c mice bearing 4T1 mammary tumors, infusing murine myeloid cells that express a murine version of CARIR significantly slowed tumor growth and prolonged survival.
    UNASSIGNED: Taken together, these results demonstrate that adoptive transfer of PD-1 CARIR-engineered myeloid cells represents a promising strategy for treating PD-L1 positive solid cancers.
    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-自然杀伤(CAR-NK)细胞疗法是一种新型的免疫疗法,通过在NK细胞上产生识别特定癌症抗原的嵌合抗原受体来靶向癌细胞。如今,由于CAR-NK细胞能够释放针对癌细胞的有效细胞毒性而无副作用,例如细胞因子释放综合征(CRS),CAR-NK细胞疗法正受到关注。神经毒性和移植物抗宿主病(GvHD)。CAR-NK细胞不需要抗原引发,从而使它们能够被用作“现成的”疗法。尽管如此,CAR-NK细胞疗法在消除存在于缺氧和免疫抑制肿瘤微环境中的癌细胞方面仍然具有若干挑战。因此,这篇综述旨在探讨当前CAR-NK细胞治疗的进展和局限性,并讨论克服CAR-NK细胞治疗所面临挑战的策略.这篇综述还旨在剖析CAR-NK细胞临床试验的现状,以及提高CAR-NK细胞治疗有效性和安全性的未来建议。
    Chimeric antigen receptor-natural killer (CAR-NK) cell therapy is a novel immunotherapy targeting cancer cells via the generation of chimeric antigen receptors on NK cells which recognize specific cancer antigens. CAR-NK cell therapy is gaining attention nowadays owing to the ability of CAR-NK cells to release potent cytotoxicity against cancer cells without side effects such as cytokine release syndrome (CRS), neurotoxicity and graft-versus-host disease (GvHD). CAR-NK cells do not require antigen priming, thus enabling them to be used as \"off-the-shelf\" therapy. Nonetheless, CAR-NK cell therapy still possesses several challenges in eliminating cancer cells which reside in hypoxic and immunosuppressive tumor microenvironment. Therefore, this review is envisioned to explore the current advancements and limitations of CAR-NK cell therapy as well as discuss strategies to overcome the challenges faced by CAR-NK cell therapy. This review also aims to dissect the current status of clinical trials on CAR-NK cells and future recommendations for improving the effectiveness and safety of CAR-NK 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: Journal Article
    嵌合抗原受体(CAR)细胞疗法利用CAR将免疫细胞重定向到表达特异性抗原如人表皮生长因子受体2(HER2)的癌细胞。尽管有潜力,CART细胞疗法在一些患者中表现出可变的应答率和不良反应。非侵入性分子成像可以通过跟踪给药后输注的细胞来帮助预测患者结果。CAR-T细胞通常是自体的,增加制造复杂性和成本。另一种方法涉及开发CAR自然杀伤(CAR-NK)细胞作为现成的同种异体产品。在这项研究中,我们设计了HER2靶向的共表达正电子发射断层扫描(PET)报告基因人碘化钠转运体(NIS)的CAR-NK细胞,并在HER2卵巢癌小鼠模型中评估了其治疗效果和PET成像能力.NK-92细胞被遗传修饰以表达HER2靶向的CAR,生物发光成像记者Antares,和NIS。将表达HER2的卵巢癌细胞工程化以表达生物发光报道萤火虫荧光素酶(Fluc)。共培养实验表明,与幼稚NK细胞相比,CAR-NK细胞的细胞毒性显著增强。涉及具有表达Fluc的肿瘤的小鼠的体内研究显示,与对照相比,用CAR-NK细胞处理的那些表现出降低的肿瘤负荷和延长的存活。纵向生物发光成像显示了随着时间的推移来自CAR-NK细胞的稳定信号。使用NIS靶向示踪剂18F-四氟硼酸酯([18F]TFB)的PET成像在用表达NIS的CAR-NK细胞处理的小鼠中显示出显著更高的PET信号。总的来说,我们的研究展示了HER2靶向CAR-NK细胞在侵袭性卵巢癌模型中的治疗潜力,并强调了在患者中使用人源PET报告基因成像非侵入性监测这些细胞的可行性.
    Chimeric antigen receptor (CAR) cell therapies utilize CARs to redirect immune cells towards cancer cells expressing specific antigens like human epidermal growth factor receptor 2 (HER2). Despite their potential, CAR T cell therapies exhibit variable response rates and adverse effects in some patients. Non-invasive molecular imaging can aid in predicting patient outcomes by tracking infused cells post-administration. CAR-T cells are typically autologous, increasing manufacturing complexity and costs. An alternative approach involves developing CAR natural killer (CAR-NK) cells as an off-the-shelf allogeneic product. In this study, we engineered HER2-targeted CAR-NK cells co-expressing the positron emission tomography (PET) reporter gene human sodium-iodide symporter (NIS) and assessed their therapeutic efficacy and PET imaging capability in a HER2 ovarian cancer mouse model.NK-92 cells were genetically modified to express a HER2-targeted CAR, the bioluminescence imaging reporter Antares, and NIS. HER2-expressing ovarian cancer cells were engineered to express the bioluminescence reporter Firefly luciferase (Fluc). Co-culture experiments demonstrated significantly enhanced cytotoxicity of CAR-NK cells compared to naive NK cells. In vivo studies involving mice with Fluc-expressing tumors revealed that those treated with CAR-NK cells exhibited reduced tumor burden and prolonged survival compared to controls. Longitudinal bioluminescence imaging demonstrated stable signals from CAR-NK cells over time. PET imaging using the NIS-targeted tracer 18F-tetrafluoroborate ([18F]TFB) showed significantly higher PET signals in mice treated with NIS-expressing CAR-NK cells.Overall, our study showcases the therapeutic potential of HER2-targeted CAR-NK cells in an aggressive ovarian cancer model and underscores the feasibility of using human-derived PET reporter gene imaging to monitor these cells non-invasively in patients.
    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
    尽管CAR-T细胞疗法已成为癌症免疫疗法中的游戏规则改变者,但一些瓶颈限制了其作为一线疗法的广泛使用。目前用于生产CAR-T细胞的方案主要依赖于使用慢病毒/逆转录病毒载体。然而,根据病毒载体使用的安全性问题,它们的临床使用有几个监管障碍。根据“当前良好生产规范”(cGMP)大规模生产病毒载体涉及严格的质量控制评估和监管要求,这给供应商带来了高昂的成本,因此,导致治疗费用显著增加。寻求有效的非病毒方法对免疫细胞进行遗传修饰是基于细胞的基因治疗的热门话题。这项研究旨在调查当前的最新状态在非病毒方法的CAR-T细胞制造。在本研究的第一部分,在回顾了病毒载体临床使用的优缺点之后,讨论了不同的非病毒载体及其临床翻译的途径。这些载体包括转座子(睡美人,piggyBac,Tol2和TcBuster),可编程核酸酶(ZFN,TLENs,和CRISPR/Cas9),mRNA质粒,微圈,和纳米质粒。之后,综述了将非病毒载体有效递送到细胞中的各种方法。
    Although CAR-T cell therapy has emerged as a game-changer in cancer immunotherapy several bottlenecks limit its widespread use as a front-line therapy. Current protocols for the production of CAR-T cells rely mainly on the use of lentiviral/retroviral vectors. Nevertheless, according to the safety concerns around the use of viral vectors, there are several regulatory hurdles to their clinical use. Large-scale production of viral vectors under \"Current Good Manufacturing Practice\" (cGMP) involves rigorous quality control assessments and regulatory requirements that impose exorbitant costs on suppliers and as a result, lead to a significant increase in the cost of treatment. Pursuing an efficient non-viral method for genetic modification of immune cells is a hot topic in cell-based gene therapy. This study aims to investigate the current state-of-the-art in non-viral methods of CAR-T cell manufacturing. In the first part of this study, after reviewing the advantages and disadvantages of the clinical use of viral vectors, different non-viral vectors and the path of their clinical translation are discussed. These vectors include transposons (sleeping beauty, piggyBac, Tol2, and Tc Buster), programmable nucleases (ZFNs, TALENs, and CRISPR/Cas9), mRNA, plasmids, minicircles, and nanoplasmids. Afterward, various methods for efficient delivery of non-viral vectors into the cells are reviewed.
    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
    多发性骨髓瘤(MM),大多数患者的浆细胞异常增殖和单克隆免疫球蛋白重链或轻链的产生,传统上与生存不良有关,尽管自引入新型药物以来,所有年龄组的中位生存期均有所改善。随着新药和新治疗方案的发展,生存率显著提高,如嵌合抗原受体T细胞疗法(CAR-T),在MM治疗中显示出了希望并给出了新的希望。汽车现在被列为第一-,第二-,和第三代CAR,基于整合到其设计中的单价至三价共刺激分子的数量。这篇综述的范围相对狭窄,因为它主要是关于CAR-T疗法在MM中的临床应用的文献比较。因此,我们的目标是概述CAR-T细胞在治疗MM方面的新进展,因此,在这篇综述中,我们综述了CAR-T细胞在MM中的临床使用进展,试图为其在管理MM时的临床使用提供参考。
    Multiple myeloma (MM), marked by abnormal proliferation of plasma cells and production of monoclonal immunoglobulin heavy or light chains in the majority of patients, has traditionally been associated with poor survival, despite improvements achieved in median survival in all age groups since the introduction of novel agents. Survival has significantly improved with the development of new drugs and new treatment options, such as chimeric antigen receptor T-cell therapy (CAR-T), which have shown promise and given new hope in MM therapy. CARs are now classified as first-, second-, and third-generation CARs based on the number of monovalent to trivalent co-stimulatory molecules incorporated into their design. The scope of this review was relatively narrow because it was mainly about a comparison of the literature on the clinical application of CAR-T therapy in MM. Thus, our goal is to provide an overview of the new advances of CAR-T cells in the cure of MM, so in this review we looked at the progress of the clinical use of CAR-T cells in MM to try to provide a reference for their clinical use when managing MM.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号