cellular immunotherapy

细胞免疫疗法
  • 文章类型: Case Reports
    背景:本研究详细介绍了一例晚期肺腺癌患者的EGFR基因缺失外显子19。
    方法:一名46岁女性患者被诊断为IVb期左肺腺癌,多发骨及淋巴结转移.在鉴定肿瘤特异性抗原肽后,患者接受了免疫治疗(TSA-DC-CTL)和口服奥希替尼的联合治疗.治疗前后监测外周血循环免疫细胞和循环肿瘤细胞(CTC)。PET-CT和CT扫描用于评估肿瘤对治疗的反应。
    结果:观察到患者的总淋巴细胞百分比显着增加,CTC数量减少。影像学研究显示肿瘤转移明显减少。
    结论:本报告证明了TSA-DC-CTL细胞免疫治疗联合奥希替尼治疗EGFR外显子19缺失的晚期肺腺癌患者的安全性和有效性。这项研究为EGFR突变的晚期肺癌患者描述了一种有希望的新治疗选择。
    BACKGROUND: This study details a case of a patient with advanced lung adenocarcinoma harboring an exon 19 deletion in the EGFR gene.
    METHODS: A 46-year-old female patient was diagnosed with stage IVb left lung adenocarcinoma, with multiple bone and lymph node metastases. Following the identification of tumor-specific antigen peptides, the patient received a combination treatment of immunotherapy (TSA-DC-CTL) and oral osimertinib. Peripheral blood circulating immune cells and circulating tumor cells (CTCs) were monitored before and after treatment. PET-CT and CT scans were used to assess the tumor response to treatment.
    RESULTS: A significant increase in total lymphocyte percentage and decrease in the number of CTCs in the patient was observed. Imaging studies showed a notable reduction in tumor metastases.
    CONCLUSIONS: This report demonstrates the safety and efficacy of TSA-DC-CTL cell immunotherapy combined with osimertinib in the treatment of a patient with advanced lung adenocarcinoma with an EGFR exon 19 deletions. This study describes a promising new treatment option for patients with advanced lung cancer with EGFR mutations.
    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: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
    无名指蛋白43(RNF43),跨膜E3泛素连接酶,已被证明是胃癌治疗的潜在生物标志物,因为这种蛋白质增加肿瘤细胞凋亡并抑制细胞增殖。RNF43在细胞免疫治疗中的作用尚不清楚。在这里,我们旨在探讨RNF43在胃癌细胞系中的表达水平及其在细胞免疫治疗中的作用。分析RNF43和PD-L1在胃癌细胞系中的表达水平及其相关性。胃癌细胞系中PD-L1的表达与RNF43的表达呈负相关。RNF43与PD-L1相互作用以增强胃癌细胞系中PD-L1的K48和K63连接的泛素化。此外,RNF43在胃癌细胞系中的表达可以增强T细胞的抗肿瘤活性。总之,这项研究表明,RNF43可以抑制PD-L1的表达,从而增强细胞免疫治疗的抗肿瘤活性。
    Ring finger protein 43 (RNF43), a transmembrane E3 ubiquitin ligase, has been indicated to be a potential biomarker for gastric cancer treatment, as this protein increases tumour cell apoptosis and suppresses cellular proliferation. The role of RNF43 in cellular immunotherapy remains unclear. Herein, we aimed to explore the expression level of RNF43 in gastric cancer cell lines and its role in cellular immunotherapy. The expression level of RNF43 and PD-L1 and their correlation in gastric cancer cell lines were analysed. The expression of PD-L1 was negatively correlated with that of RNF43 in gastric cancer cell lines. RNF43 interacted with PD-L1 to augment both K48- and K63-linked ubiquitination of PD-L1 in gastric cancer cell lines. In addition, RNF43 expression in gastric cancer cell lines could enhance the antitumour activity of T cells. In conclusion, this study reveals that RNF43 can inhibit PD-L1 expression to enhance the antitumour activity of cellular 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
    细胞免疫疗法已经成为一种令人兴奋的癌症治疗策略,因为它旨在通过改造免疫细胞和从头开始设计合成分子来增强人体对肿瘤细胞的免疫反应。由于细胞毒性,丰富的外周血,基因工程技术的成熟,T细胞已成为迄今为止最常见的工程化免疫细胞。以嵌合抗原受体(CAR)-T疗法为代表,基于T细胞的免疫疗法彻底改变了血液系统恶性肿瘤的临床治疗。然而,在实体瘤中严重的副作用和有限的疗效阻碍了细胞免疫疗法的临床应用。为了解决这些限制,已经开发了关于合成细胞和分子的各种创新策略。一方面,一些T细胞以外的细胞毒性免疫细胞已被改造,以探索靶向消除肿瘤细胞的潜力,而一些佐剂细胞也被改造以增强治疗效果。另一方面,不同的合成细胞成分和分子被添加到工程免疫细胞来调节它们的功能,促进细胞毒活性和限制副作用。此外,新的生物活性材料,如水凝胶,促进治疗性免疫细胞的递送也已被用于提高细胞免疫疗法的功效。这篇综述总结了目前可用于细胞免疫疗法的合成细胞和分子的创新策略。讨论的局限性,并提供对下一代细胞免疫疗法的见解。
    Cellular immunotherapy has emerged as an exciting strategy for cancer treatment, as it aims to enhance the body\'s immune response to tumor cells by engineering immune cells and designing synthetic molecules from scratch. Because of the cytotoxic nature, abundance in peripheral blood, and maturation of genetic engineering techniques, T cells have become the most commonly engineered immune cells to date. Represented by chimeric antigen receptor (CAR)-T therapy, T cell-based immunotherapy has revolutionized the clinical treatment of hematological malignancies. However, serious side effects and limited efficacy in solid tumors have hindered the clinical application of cellular immunotherapy. To address these limitations, various innovative strategies regarding synthetic cells and molecules have been developed. On one hand, some cytotoxic immune cells other than T cells have been engineered to explore the potential of targeted elimination of tumor cells, while some adjuvant cells have also been engineered to enhance the therapeutic effect. On the other hand, diverse synthetic cellular components and molecules are added to engineered immune cells to regulate their functions, promoting cytotoxic activity and restricting side effects. Moreover, novel bioactive materials such as hydrogels facilitating the delivery of therapeutic immune cells have also been applied to improve the efficacy of cellular immunotherapy. This review summarizes the innovative strategies of synthetic cells and molecules currently available in cellular immunotherapies, discusses the limitations, and provides insights into the next generation of cellular immunotherapies.
    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
    MET受体是“侵入性生长”的主要驱动因素之一,在胚胎发育和组织修复过程中必不可少的多方面生物学反应,被癌细胞篡夺以诱导和维持恶性表型。MET是癌症中激活的最重要的癌基因之一,自癌症靶向治疗的最初时代以来,人们一直在探索其抑制作用。已经开发了不同的方法来阻碍MET信号传导和/或减少作为转化标志的MET(过)表达。考虑到癌症免疫疗法获得的极大兴趣,这篇综述评估了基于免疫功能开发的治疗方法中靶向MET的机会,在MET损伤对诱导有效反应至关重要的情况下(即,当MET是恶性肿瘤的驱动因素时),或者当阻断MET代表一种增强治疗的方法(即,当MET是肿瘤适应性的佐剂时)。
    The MET receptor is one of the main drivers of \'invasive growth\', a multifaceted biological response essential during embryonic development and tissue repair that is usurped by cancer cells to induce and sustain the malignant phenotype. MET stands out as one of the most important oncogenes activated in cancer and its inhibition has been explored since the initial era of cancer-targeted therapy. Different approaches have been developed to hamper MET signaling and/or reduce MET (over)expression as a hallmark of transformation. Considering the great interest gained by cancer immunotherapy, this review evaluates the opportunity of targeting MET within therapeutic approaches based on the exploitation of immune functions, either in those cases where MET impairment is crucial to induce an effective response (i.e., when MET is the driver of the malignancy), or when blocking MET represents a way for potentiating the treatment (i.e., when MET is an adjuvant of tumor fitness).
    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
    自然杀伤细胞治疗近年来在肿瘤学领域显示出较强的疗效,已应用于有转移的患者,旨在改善晚期胃癌的预后。一名59岁的男性患有胃腺癌伴胰腺转移(T4N0M1),他接受了胃癌伴肿瘤转移的根治性手术,接受了奥沙利铂和替加氟联合细胞回输的治疗。疗程结束后,连续监测CT扫描和血清肿瘤标志物。经过五个疗程的综合治疗,患者病情得到控制,无明显副作用.在最后一次随访中,甲胎蛋白已恢复到正常值,胰腺体内的低密度阴影显示不佳。胰腺癌起源隐匿,死亡率高。该报告为胰腺转移癌的细胞治疗提供了临床证据,并改善了生活质量。
    Nature killer cell therapy has shown strong efficacy in the field of oncology in recent years and has been applied to patients with metastases with the aim of improving the prognosis of advanced gastric cancer. A 59-year-old male with gastric adenocarcinoma with pancreatic metastasis (T4N0M1) who underwent radical surgery for gastric cancer with tumor metastasis was treated with oxaliplatin and tegafur combined with cellular reinfusion in stages. Computed tomograpy scan and serum tumor markers were monitored continuously after the treatment course. After five courses of combined treatment, the patient was in disease control with no significant side effects. At the last follow-up, the alpha fetoprotein had returned to its normal value with a poor display of low-density shadows in the body of the pancreas. Pancreatic cancer is insidious in origin and has a high mortality rate. The report provides clinical evidence for cell therapy of pancreatic metastatic cancer with improved 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
    目前自身免疫性疾病的治疗是免疫抑制剂,有许多令人衰弱的副作用。然而,树突状细胞(DC)可以诱导抗原特异性耐受。由离体产生的DC介导的耐受性恢复可以是治疗方法。因此,在这次审查中,我们总结了针对自身免疫性疾病开发离体产生的DC策略的概念框架.首先,我们将讨论DC在发展免疫耐受中的作用,作为开发基于树突状细胞的自身免疫性疾病免疫治疗的基础。然后,我们还讨论了体外产生的DCs治疗自身免疫性疾病的临床前和临床研究的相关发现.最后,我们讨论了自身免疫性疾病中树突状细胞治疗的问题和挑战。在整篇文章中,我们讨论自身免疫性疾病,强调SLE。
    Current therapies for autoimmune diseases are immunosuppressant agents, which have many debilitating side effects. However, dendritic cells (DCs) can induce antigen-specific tolerance. Tolerance restoration mediated by ex vivo-generated DCs can be a therapeutic approach. Therefore, in this review, we summarize the conceptual framework for developing ex vivo-generated DC strategies for autoimmune diseases. First, we will discuss the role of DCs in developing immune tolerance as a foundation for developing dendritic cell-based immunotherapy for autoimmune diseases. Then, we also discuss relevant findings from pre-clinical and clinical studies of ex vivo-generated DCs for therapy of autoimmune diseases. Finally, we discuss problems and challenges in dendritic cell therapy in autoimmune diseases. Throughout the article, we discuss autoimmune diseases, emphasizing SLE.
    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
    细胞免疫疗法(CIT)在癌症中表现出出色的功效水平,并提出了独特的商业化挑战。对用于开发第一个嵌合抗原受体T细胞(CAR-Ts)的上市(G2M)策略的历史分析可以提供有关公司如何利用伙伴关系或独立性来确保商业成功的见解。基于合作的战略,例如伙伴关系,收购,和许可交易,在行业中占主导地位,以最大限度地提高收入和患者的访问。Manufacturing,后勤,监管挑战阻碍了独立的商业化。尽管如此,该行业正在适应这些挑战:新技术显示出优越的可负担性和可实施性,和商业解决方案组织(CSO)越来越多地帮助CIT公司独立解决商业化问题。因此,这个行业的G2M战略可能会演变,独立成为商业成功的可行策略。
    Cellular immunotherapy (CIT) has both demonstrated outstanding levels of efficacy in cancer and presented unique commercialisation challenges. A historical analysis of go-to-market (G2M) strategies used to develop the first chimeric antigen receptor T cells (CAR-Ts) can offer insight into how companies leverage partnership or independence to ensure commercial success. Collaboration-based strategies, such as partnerships, acquisitions, and licensing deals, have predominated in the industry to maximise revenue and patient access. Manufacturing, logistical, and regulatory challenges have hindered independent commercialisation. Nonetheless, the industry is adapting to these challenges: novel technologies show superior affordability and implementability, and commercial solutions organisations (CSOs) increasingly help CIT companies navigate through commercialisation issues independently. G2M strategies in this industry are therefore likely to evolve, with independence becoming a feasible strategy for commercial success.
    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)T细胞疗法已经改变了癌症免疫疗法。然而,重大挑战限制了其在B细胞驱动的恶性肿瘤之外的应用,包括有限的临床疗效,高毒性,和复杂的自体细胞产品制造。尽管努力改善CART细胞治疗结果,人们对利用替代免疫细胞来开发CAR细胞越来越感兴趣。这些免疫细胞提供了几个优点,如主要组织相容性复合体(MHC)独立功能,肿瘤微环境(TME)调制,和增加组织浸润能力。目前,来自各种T细胞亚型的CAR产品,先天免疫细胞,造血祖细胞,甚至外泌体也在探索中。这些CAR产品通常显示出增强的抗肿瘤功效,毒性降低,和优越的肿瘤渗透。考虑到这些好处,许多临床试验正在进行中,以获得这些创新的CAR细胞的潜力。这篇综述旨在彻底研究这些优势,挑战,以及对这些新的CAR产品在癌症治疗中的现有见解。
    Chimeric antigen receptor (CAR) T cell therapy has transformed cancer immunotherapy. However, significant challenges limit its application beyond B cell-driven malignancies, including limited clinical efficacy, high toxicity, and complex autologous cell product manufacturing. Despite efforts to improve CAR T cell therapy outcomes, there is a growing interest in utilizing alternative immune cells to develop CAR cells. These immune cells offer several advantages, such as major histocompatibility complex (MHC)-independent function, tumor microenvironment (TME) modulation, and increased tissue infiltration capabilities. Currently, CAR products from various T cell subtypes, innate immune cells, hematopoietic progenitor cells, and even exosomes are being explored. These CAR products often show enhanced antitumor efficacy, diminished toxicity, and superior tumor penetration. With these benefits in mind, numerous clinical trials are underway to access the potential of these innovative CAR cells. This review aims to thoroughly examine the advantages, challenges, and existing insights on these new CAR products in cancer 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
    间变性甲状腺癌(ATC)是最具侵袭性的甲状腺癌类型。虽然ATC很少见,它的死亡率很高。标准治疗,比如手术,放射治疗,和化疗,在管理ATC方面表现出有限的功效。然而,免疫治疗的出现显著改善了ATC患者的预后.免疫疗法通过利用人体免疫细胞的力量有效地靶向和消除肿瘤细胞。新抗原是由体细胞突变产生的非典型蛋白质,只在肿瘤细胞中观察到,并且缺乏中心耐受性。新抗原对肿瘤细胞表现出增强的特异性,并显示出强大的免疫原性。目前,新抗原治疗主要应用于免疫检查点抑制剂和细胞免疫治疗,包括过继免疫疗法和肿瘤疫苗。本研究讨论了这种机制,肿瘤微环境,临床试验,不良事件,与ATC免疫治疗相关的局限性和未来方向。
    Anaplastic thyroid cancer (ATC) is the most aggressive type of thyroid cancer. While ATC is rare, its mortality is high. Standard treatments, such as surgery, radiotherapy, and chemotherapy, have demonstrated limited efficacy in managing ATC. However, the advent of immunotherapy has significantly improved the prognosis for patients with ATC. Immunotherapy effectively targets and eliminates tumor cells by using the power of the body\'s immune cells. The neoantigen is an atypical protein generated by somatic mutation, is exclusively observed in neoplastic cells, and is devoid of central tolerance. Neoantigens exhibit enhanced specificity towards tumor cells and display robust immunogenic properties. Currently, neoantigen therapy is primarily applied in immune checkpoint inhibitors and cellular immunotherapy, encompassing adoptive immunotherapy and tumor vaccines. This study discusses the mechanism, tumor microenvironment, clinical trials, adverse events, limitations and future directions associated with ATC immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号