Solid tumor

实体瘤
  • 文章类型: Journal Article
    在以前的文献中,我们发现,关于其他系统同步脑转移(S-BM)短期预后的类似研究很少.我们的目的是从监测中评估S-BM患者的早期死亡率,流行病学,和最终结果(SEER)数据库,并探讨早期死亡率(≤1年)的危险因素。我们使用Kaplan-Meier(KM)曲线评估SEER数据库中S-BM患者的早期死亡率。Logistic回归分析用于确定随访时间>12个月的患者的显著独立预后因素。并将有意义的因素用于构建总体早期死亡的列线图。用受试者工作特征(ROC)曲线检验模型的预测能力,用决策曲线分析(DCA)曲线验证模型的临床应用能力。共使用47,284例患者进行单因素和多因素logistic回归分析以筛选变量以构建列线图。在全因早期死亡率特定模型中,训练集的ROC(AUC)曲线下面积为0.764(95%置信区间(CI):0.758-0.769),验证集的AUC为0.761(95%CI:0.752-0.770)。训练集和验证集的DCA校准曲线表明,该模型预测的1年早期死亡率与实际情况相符。我们发现1年早期死亡率为76.4%。我们使用这些协变量构建了一个有效的列线图,以有效预测S-BM患者的1年早期死亡率。此列线图可帮助临床工作者筛选高危患者,制定更合理的治疗方案。
    In previous literatures, we found that similar studies on the short-term prognosis of synchronous brain metastases (S-BM) from other systems are rare. Our aim was to evaluate the early mortality rate of patients with S-BM from the Surveillance, Epidemiology, and End Result (SEER) database and explore the risk factors for early mortality (≤ 1 year). We used Kaplan-Meier (KM) curves to evaluate early mortality in patients with S-BM from the SEER database. Logistic regression analyses were used to identify significant independent prognostic factors in patients with a follow-up time > 12 months. And the meaningful factors were used to construct a nomogram of overall early death. The receiver operating characteristic (ROC) curve was used to test the predictive ability of the model, while the decision curve analysis (DCA) curve was used to validate the clinical application ability of the model. A total of 47,284 patients were used for univariate and multivariate logistic regression analysis to screen variables to constructing a nomogram. In the all-cause early mortality specific model, the area under the ROC (AUC) curve of the training set was 0.764 (95% confidence interval (CI): 0.758-0.769), and the AUC of the validation set was 0.761 (95% CI: 0.752-0.770). The DCA calibration curves of the training set and validation set indicate that the 1-year early mortality rate predicted by this model is consistent with the actual situation. We found that the 1-year early mortality rate was 76.4%. We constructed a validated nomogram using these covariates to effectively predict 1-year early mortality in patients with S-BM. This nomogram can help clinical workers screen high-risk patients to develop more reasonable treatment plans.
    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
    背景:先前的研究表明,在各种实体瘤中,将免疫检查点抑制剂与抗血管生成剂组合使用时,具有潜在的协同抗肿瘤活性。我们旨在评估卡姆瑞珠单抗(人源化程序性细胞死亡-1抗体)联合阿帕替尼(血管内皮生长因子受体酪氨酸激酶抑制剂)治疗晚期粘膜黑色素瘤(MM)患者的疗效和安全性。和探索相关的生物标志物。
    方法:我们进行了单中心,开放标签,单臂,第二阶段研究。不可切除或复发/转移性MM患者接受卡利单抗和阿帕替尼治疗。主要终点是确认的客观反应率(ORR)。
    结果:在2019年4月至2022年6月之间,共招募了32名患者,50.0%以前接受过全身治疗。在28例反应可评估的患者中,确认的ORR为42.9%,疾病控制率为82.1%,中位无进展生存期(PFS)为8.05个月。在未治疗和先前治疗的患者中,确认的ORR为42.9%(6/14)。值得注意的是,初治患者的中位PFS为11.89个月,接受过治疗的患者的中位PFS为6.47个月.3级治疗相关不良事件为转氨酶升高,皮疹,高胆红素血症,蛋白尿,高血压,血小板减少症,手足综合征和腹泻。没有观察到治疗相关的死亡。较高的肿瘤突变负荷(TMB),T细胞受体(TCR)多样性增加,受体酪氨酸激酶(RTK)/RAS途径的改变与更好的肿瘤反应相关。
    结论:卡利珠单抗联合阿帕替尼在晚期MM患者中具有良好的抗肿瘤活性和可接受的毒性。TMB,TCR多样性和RTK/RAS途径基因被鉴定为潜在的预测性生物标志物,并需要进一步验证。
    背景:中国临床试验注册中心,ChiCTR1900023277。
    BACKGROUND: Previous studies have suggested the potential synergistic antitumor activity when combining immune checkpoint inhibitors with anti-angiogenic agents in various solid tumors. We aimed to assess the efficacy and safety of camrelizumab (a humanized programmed cell death-1 antibody) plus apatinib (a vascular endothelial growth factor receptor tyrosine kinase inhibitor) for patients with advanced mucosal melanoma (MM), and explore-related biomarkers.
    METHODS: We conducted a single-center, open-label, single-arm, phase II study. Patients with unresectable or recurrent/metastatic MM received camrelizumab and apatinib. The primary endpoint was the confirmed objective response rate (ORR).
    RESULTS: Between April 2019 and June 2022, 32 patients were enrolled, with 50.0% previously received systemic therapy. Among 28 patients with evaluable response, the confirmed ORR was 42.9%, the disease control rate was 82.1%, and the median progression-free survival (PFS) was 8.05 months. The confirmed ORR was 42.9% (6/14) in both treatment-naïve and previously treated patients. Notably, treatment-naïve patients had a median PFS of 11.89 months, and those with prior treatment had a median PFS of 6.47 months. Grade 3 treatment-related adverse events were transaminase elevation, rash, hyperbilirubinemia, proteinuria, hypertension, thrombocytopenia, hand-foot syndrome and diarrhea. No treatment-related deaths were observed. Higher tumor mutation burden (TMB), increased T-cell receptor (TCR) diversity, and altered receptor tyrosine kinase (RTK)/RAS pathway correlated with better tumor response.
    CONCLUSIONS: Camrelizumab plus apatinib provided promising antitumor activity with acceptable toxicity in patients with advanced MM. TMB, TCR diversity and RTK/RAS pathway genes were identified as potential predictive biomarkers and warrant further validation.
    BACKGROUND: Chinese Clinical Trial Registry, ChiCTR1900023277.
    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
    癌症免疫疗法在过去的10-15年里蓬勃发展,改变肿瘤学的实践,并为一些患者提供长期的临床益处。在此期间,三类不同的免疫检查点抑制剂,对两个靶标具有特异性的嵌合抗原受体-T细胞疗法,和两类不同的双特异性T细胞衔接者,疫苗,和溶瘤病毒已经加入细胞因子作为癌症治疗的标准。同时,科学进步带来了大量新知识。例如,单细胞测序和空间转录组学等技术的进步为肿瘤微环境的免疫生物学提供了深刻的见解。随着这种快速的临床和科学进步,癌症免疫治疗领域目前正处于一个关键的拐点,具有未来十年指数增长的潜力。认识到这一点,癌症免疫治疗学会召集了代表学术界的癌症免疫治疗专家,制药和生物技术行业,病人的倡导,和监管社区确定当前的机遇和挑战,目标是优先考虑具有最高临床影响潜力的领域。共识小组确定了该领域当前机会的七个高度优先领域:抗肿瘤活性和毒性机制;耐药性机制;生物标志物和生物标本;新疗法的独特方面;宿主和环境相互作用;癌前免疫,免疫拦截,和免疫预防措施;和临床试验设计,端点,和行为。此外,讨论了进展的潜在障碍,并确定了几个主题作为优化的交叉工具,每个都有可能影响多个科学优先领域。这些交叉工具包括临床前模型,数据管理和共享,活检和生物标本,资金来源多样化,定义和标准,和病人的参与。最后,确定了三项关键指导原则,这些原则将优化和最大限度地提高该领域的进展。这些包括参与患者社区;培养多样性,股本,inclusion,和可访问性;并利用人工智能的力量加速进步。这里,我们将这些讨论的结果作为激发该领域未来十年癌症免疫疗法呈指数级进展的战略愿景.
    Cancer immunotherapy has flourished over the last 10-15 years, transforming the practice of oncology and providing long-term clinical benefit to some patients. During this time, three distinct classes of immune checkpoint inhibitors, chimeric antigen receptor-T cell therapies specific for two targets, and two distinct classes of bispecific T cell engagers, a vaccine, and an oncolytic virus have joined cytokines as a standard of cancer care. At the same time, scientific progress has delivered vast amounts of new knowledge. For example, advances in technologies such as single-cell sequencing and spatial transcriptomics have provided deep insights into the immunobiology of the tumor microenvironment. With this rapid clinical and scientific progress, the field of cancer immunotherapy is currently at a critical inflection point, with potential for exponential growth over the next decade. Recognizing this, the Society for Immunotherapy of Cancer convened a diverse group of experts in cancer immunotherapy representing academia, the pharmaceutical and biotechnology industries, patient advocacy, and the regulatory community to identify current opportunities and challenges with the goal of prioritizing areas with the highest potential for clinical impact. The consensus group identified seven high-priority areas of current opportunity for the field: mechanisms of antitumor activity and toxicity; mechanisms of drug resistance; biomarkers and biospecimens; unique aspects of novel therapeutics; host and environmental interactions; premalignant immunity, immune interception, and immunoprevention; and clinical trial design, endpoints, and conduct. Additionally, potential roadblocks to progress were discussed, and several topics were identified as cross-cutting tools for optimization, each with potential to impact multiple scientific priority areas. These cross-cutting tools include preclinical models, data curation and sharing, biopsies and biospecimens, diversification of funding sources, definitions and standards, and patient engagement. Finally, three key guiding principles were identified that will both optimize and maximize progress in the field. These include engaging the patient community; cultivating diversity, equity, inclusion, and accessibility; and leveraging the power of artificial intelligence to accelerate progress. Here, we present the outcomes of these discussions as a strategic vision to galvanize the field for the next decade of exponential progress in cancer 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
    最近的研究强调了干扰素γ受体(IFNγR)途径在T细胞介导的针对固体而非液体肿瘤的细胞毒性中的重要作用。IFNγ不仅直接促进T细胞的肿瘤细胞死亡,而且还通过肿瘤微环境中的骨髓吞噬作用间接促进细胞毒性。同时,完整的人体外免疫检查点药物筛选仍然具有挑战性.我们假设工程化的γ干扰素激活位点反应元件荧光素酶报告基因(GAS-Luc2)可用于多种离体T细胞-实体瘤细胞共培养系统中的免疫检查点药物筛选。我们全面分析了ATCC广泛收集的人类肿瘤和免疫细胞系中的细胞表面蛋白,鉴定那些具有已建立和新型免疫检查点分子和结合配体的内源性高表达。然后我们改造了三种表达免疫检查点PD-L1、CD155或B7-H3/CD276的GAS-Luc2报告肿瘤细胞系。在相关的免疫检查点-配体接合时抑制荧光素酶表达。在存在免疫检查点抑制剂的情况下,T细胞释放IFNγ,激活GAS-Luc2细胞中的JAK-STAT通路,并产生用于抑制剂评估的可量化生物发光信号。这些报告细胞系还检测到旁分泌IFNγ信号用于免疫检查点靶向ADCC药物筛选。进一步发展为人工抗原呈递细胞系(aAPC)显著增强T细胞信号传导,以在这些离体免疫检查点药物筛选平台中获得优异的性能。
    Recent studies highlight the integral role of the interferon gamma receptor (IFNγR) pathway in T cell-mediated cytotoxicity against solid but not liquid tumors. IFNγ not only directly facilitates tumor cell death by T cells but also indirectly promotes cytotoxicity via myeloid phagocytosis in the tumor microenvironment. Meanwhile, full human ex vivo immune checkpoint drug screening remains challenging. We hypothesized that an engineered gamma interferon activation site response element luciferase reporter (GAS-Luc2) can be utilized for immune checkpoint drug screening in diverse ex vivo T cell-solid tumor cell co-culture systems. We comprehensively profiled cell surface proteins in ATCC\'s extensive collection of human tumor and immune cell lines, identifying those with endogenously high expression of established and novel immune checkpoint molecules and binding ligands. We then engineered three GAS-Luc2 reporter tumor cell lines expressing immune checkpoints PD-L1, CD155, or B7-H3/CD276. Luciferase expression was suppressed upon relevant immune checkpoint-ligand engagement. In the presence of an immune checkpoint inhibitor, T cells released IFNγ, activating the JAK-STAT pathway in GAS-Luc2 cells, and generating a quantifiable bioluminescent signal for inhibitor evaluation. These reporter lines also detected paracrine IFNγ signaling for immune checkpoint-targeted ADCC drug screening. Further development into an artificial antigen-presenting cell line (aAPC) significantly enhanced T cell signaling for superior performance in these ex vivo immune checkpoint drug screening platforms.
    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细胞过继免疫疗法是一种有前途的癌症治疗,使用基因工程T细胞攻击肿瘤。然而,这种疗法会有一些副作用。CAR-T细胞衍生的外泌体是CAR-T细胞的潜在替代品,可以克服一些限制。外泌体是细胞释放的小囊泡,可以携带多种分子,包括蛋白质,RNA,和DNA。它们在细胞间通讯中起重要作用,可用于向癌细胞递送治疗剂。CAR-T细胞来源的外泌体的应用可以使CAR-T细胞疗法在临床上更加可控和有效。外泌体是无细胞的,这意味着它们比CAR-T细胞更不容易引起不良反应。CAR-T细胞和外泌体的组合可能是比单独的任何一种疗法更有效的治疗癌症的方法。外泌体可以将治疗剂递送到CAR-T细胞无法到达的癌细胞。细胞和外泌体平台的适当应用可以使CAR-T细胞疗法成为更可行的癌症治疗方法。这种联合疗法可以提供一种安全有效的方法来治疗多种癌症。
    Chimeric antigen receptor (CAR)-T cell adoptive immunotherapy is a promising cancer treatment that uses genetically engineered T cells to attack tumors. However, this therapy can have some adverse effects. CAR-T cell-derived exosomes are a potential alternative to CAR-T cells that may overcome some limitations. Exosomes are small vesicles released by cells and can carry a variety of molecules, including proteins, RNA, and DNA. They play an important role in intercellular communication and can be used to deliver therapeutic agents to cancer cells. The application of CAR-T cell-derived exosomes could make CAR-T cell therapy more clinically controllable and effective. Exosomes are cell-free, which means that they are less likely to cause adverse reactions than CAR-T cells. The combination of CAR-T cells and exosomes may be a more effective way to treat cancer than either therapy alone. Exosomes can deliver therapeutic agents to cancer cells where CAR-T cells cannot reach. The appropriate application of both cellular and exosomal platforms could make CAR-T cell therapy a more practicable treatment for cancer. This combination therapy could offer a safe and effective way to treat a variety of 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
    造血细胞激酶(Hck),属于Src激酶家族的非受体酪氨酸激酶,与许多人类疾病的发病机理有着错综复杂的联系,与癌症有特别明显的关联。Hck不仅直接影响扩散,迁移,和癌细胞的凋亡,但也与JAK/STAT相互作用,MEK/ERK,PI3K/AKT,CXCL12/CXCR4等途径。Hck还影响肿瘤微环境以促进癌症的发作和进展。本文探讨了Hck在多种实体瘤中的功能作用和调控机制。此外,它探讨了Hck在血液恶性肿瘤中的意义。这篇综述总结了Hck抑制剂的研究现状,其中大多数处于研究的临床前阶段。值得注意的是,这些抑制剂主要用于白血病的治疗管理,它们的组合潜力为未来的研究指明了有希望的途径。总之,这篇综述强调了Hck在实体瘤中的作用机制的意义。这种见解对于理解有关Hck的当前研究趋势至关重要:针对Hck的靶向治疗在恶性肿瘤的诊断和治疗中显示出巨大的前景。进一步研究Hck在癌症中的作用,再加上特异性抑制剂的开发,有可能彻底改变癌症治疗方法。
    Hematopoietic cell kinase (Hck), a non-receptor tyrosine kinase belonging to the Src kinase family, is intricately linked to the pathogenesis of numerous human diseases, with a particularly pronounced association with cancer. Hck not only directly impacts the proliferation, migration, and apoptosis of cancer cells but also interacts with JAK/STAT, MEK/ERK, PI3K/AKT, CXCL12/CXCR4, and other pathways. Hck also influences the tumor microenvironment to facilitate the onset and progression of cancer. This paper delves into the functional role and regulatory mechanisms of Hck in various solid tumors. Additionally, it explores the implications of Hck in hematological malignancies. The review culminates with a summary of the current research status of Hck inhibitors, the majority of which are in the pre-clinical phase of investigation. Notably, these inhibitors are predominantly utilized in the therapeutic management of leukemia, with their combinatorial potential indicating promising avenues for future research. In conclusion, this review underscores the significance of the mechanism of Hck in solid tumors. This insight is crucial for comprehending the current research trends regarding Hck: targeted therapy against Hck shows great promise in both diagnosis and treatment of malignant tumors. Further investigation into the role of Hck in cancer, coupled with the development of specific inhibitors, has the potential to revolutionize approaches to cancer treatment.
    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
    在癌症免疫治疗的治疗策略中,涉及免疫调节抗体,癌症疫苗或过继性T细胞转移,T细胞由于其对肿瘤细胞的细胞毒性和受体的肿瘤抗原特异性结合而成为有吸引力的靶标。利用T细胞的专有特性,嵌合抗原受体(CAR)-T和T细胞受体(TCR)-T细胞通过其受体的遗传版本产生,这导致了T细胞疗法的特异性和有效性的提高。CAR-T细胞的过继细胞转移已成功用于血液恶性肿瘤的治疗。为了将T细胞疗法扩展到实体瘤,T细胞被修饰以表达确定的TCR靶向肿瘤相关抗原(TAA),这就是所谓的TCR-T疗法。在这里,这篇综述讨论了抗肿瘤T细胞治疗,重点是工程化TCR-T细胞治疗。我们描述了TCR-T细胞疗法的特点,以及TCR-T细胞治疗非血液系统恶性肿瘤的临床应用。
    Among the therapeutic strategies in cancer immunotherapy-such as immune-modulating antibodies, cancer vaccines, or adoptive T cell transfer-T cells have been an attractive target due to their cytotoxicity toward tumor cells and the tumor antigen-specific binding of their receptors. Leveraging the unique properties of T cells, chimeric antigen receptor-T cells and T cell receptor (TCR)-T cells were developed through genetic modification of their receptors, enhancing the specificity and effectiveness of T cell therapy. Adoptive cell transfer of chimeric antigen receptor-T cells has been successful for the treatment of hematological malignancies. To expand T cell therapy to solid tumors, T cells are modified to express defined TCR targeting tumor associated antigen, which is called TCR-T therapy. This review discusses anti-tumor T cell therapies, with a focus on engineered TCR-T cell therapy. We outline the characteristics of TCR-T cell therapy and its clinical application to non-hematological malignancies.
    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
    背景:Tebentafusp,双特异性(gp100×CD3)ImmTAC,HLA-A*02:01+未经治疗的转移性葡萄膜黑色素瘤(mUM)成年患者的总生存期(OS)结局显著改善,并且在IMCgp100-102研究的主要分析中,1年OS为62%的既往治疗mUM中显示有希望的生存期.在这里,我们报告了这项1/2期研究在预处理mUM中的长期结果。
    方法:先前治疗过mUM的患者每周静脉注射tebentafusp,剂量为20µg,剂量为30µg,剂量为2,剂量为54、64、68或73µg(1期)或68µg(2期)剂量为3+。主要目标是总体反应率。次要目标包括操作系统和安全性。OS通过Kaplan-Meier方法估计。操作系统和基线协变量之间的关联,实体瘤(RECIST)反应的治疗反应评估标准,评估基线肿瘤活检和循环肿瘤DNA(ctDNA)变化。
    结果:146例患者接受tebentafusp治疗:19在1期,127在2期。中位随访时间为48.5个月,中位OS为17.4个月(95%CI,13.1至22.8),和1年,2年,3年和4年OS率为62%,40%,23%和14%,分别。在治疗第9周时,生存率的提高与较低的ctDNA基线水平和更大的ctDNA减少相关。100%1年,ctDNA清除患者的2年和3年OS率分别为73%和45%。基线gp100表达与生存率无关,尽管在表达较高的患者中有更多的RECIST反应。长期给药没有新的安全性信号报告。
    结论:这项研究代表了迄今为止对Tcell受体双特异性的最长随访,并证实了tebentafusp在先前治疗的mUM中实现的持久生存益处,具有良好的长期耐受性。对于接受tebentafusp治疗的患者,再次提出了ctDNA减少作为临床获益的早期指标的作用。
    BACKGROUND: Tebentafusp, a bispecific (gp100×CD3) ImmTAC, significantly improved overall survival (OS) outcomes for HLA-A*02:01+ adult patients with untreated metastatic uveal melanoma (mUM) and showed promising survival in previously treated mUM with 1-year OS of 62% in the primary analysis of study IMCgp100-102. Here we report long-term outcomes from this phase 1/2 study in pretreated mUM.
    METHODS: Patients with previously treated mUM received tebentafusp weekly intravenous at 20 µg dose 1, 30 µg dose 2 and either 54, 64, 68, or 73 µg (phase 1) or 68 µg (phase 2) dose 3+. The primary objective was overall response rate. Secondary objectives included OS and safety. OS was estimated by Kaplan-Meier methods. Association between OS and baseline covariates, on-treatment Response Evaluation Criteria in Solid Tumors (RECIST) response, baseline tumor biopsy and circulating-tumor DNA (ctDNA) changes were assessed.
    RESULTS: 146 patients were treated with tebentafusp: 19 in phase 1 and 127 in phase 2. With a median follow-up duration of 48.5 months, the median OS was 17.4 months (95% CI, 13.1 to 22.8), and the 1-year, 2-year, 3-year and 4-year OS rates were 62%, 40%, 23% and 14%, respectively. Improved survival was associated with lower ctDNA baseline levels and greater ctDNA reductions by week 9 on-treatment, with 100% 1-year, 73% 2-year and 45% 3-year OS rates for patients with ctDNA clearance. Baseline gp100 expression was not associated with survival, despite more RECIST responses among patients with higher expression. No new safety signals were reported with long-term dosing.
    CONCLUSIONS: This study represents the longest follow-up of a Tcell receptor bispecific to date and confirms the durable survival benefits achieved with tebentafusp in previously treated mUM with good tolerability long-term. A role for ctDNA reduction as an early indicator of clinical benefit was again suggested for patients treated with tebentafusp.
    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
    肿瘤微环境与启动密切相关,促销,和实体瘤的进展。在其宪法中,免疫细胞作为关键参与者出现,促进免疫逃避和肿瘤进展。除了它们对抗肿瘤免疫的间接影响外,免疫细胞直接影响肿瘤细胞,支持或阻碍肿瘤进展。然而,目前旨在减轻效应免疫细胞群的调节细胞免疫抑制的治疗模式可能无法在各种实体瘤中始终产生令人满意的结果。比如乳腺癌,结直肠癌,等。因此,这篇综述概述和总结了直接的,免疫细胞如T细胞的双重效应,先天的淋巴样细胞,B细胞,嗜酸性粒细胞,以及肿瘤微环境中肿瘤细胞上的肿瘤相关巨噬细胞。该综述还深入研究了所涉及的潜在机制,并介绍了基于这些直接影响的临床试验结果,旨在提出解决实体肿瘤的创新和有效的治疗策略。
    The tumor microenvironment is closely linked to the initiation, promotion, and progression of solid tumors. Among its constitutions, immunologic cells emerge as critical players, facilitating immune evasion and tumor progression. Apart from their indirect impact on anti-tumor immunity, immunocytes directly influence neoplastic cells, either bolstering or impeding tumor advancement. However, current therapeutic modalities aimed at alleviating immunosuppression from regulatory cells on effector immune cell populations may not consistently yield satisfactory results in various solid tumors, such as breast carcinoma, colorectal cancer, etc. Therefore, this review outlines and summarizes the direct, dualistic effects of immunocytes such as T cells, innate lymphoid cells, B cells, eosinophils, and tumor-associated macrophages on tumor cells within the tumor microenvironment. The review also delves into the underlying mechanisms involved and presents the outcomes of clinical trials based on these direct effects, aiming to propose innovative and efficacious therapeutic strategies for addressing 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
    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)

公众号