ICb

ICB
  • 文章类型: Journal Article
    以下叙述性审查开始全面探索肠道微生物组在饮食-微生物群-免疫(dmi)三方中的作用,旨在增强抗癌免疫治疗的疗效。在彻底改变癌症治疗的同时,对免疫疗法的耐药性和免疫相关不良事件(irAEs)仍然是挑战。肿瘤微环境(TME),由癌细胞形成,影响免疫疗法耐药性。肠道微生物组,受遗传学的影响,环境,饮食,和干预措施,成为TME重塑的关键参与者,从而调节免疫反应和治疗结果。地中海饮食等饮食模式,热量限制修改,和特定的营养成分在影响肿瘤微环境和肠道微生物组以获得更好的治疗结果方面显示出希望。抗生素,破坏肠道微生物多样性,可能会损害免疫疗法的疗效。这篇综述强调需要量身定制的营养策略来操纵微生物群落,增强免疫调节,并提高免疫治疗的可及性,同时最大限度地减少副作用。正在进行的研究调查饮食干预对癌症免疫治疗的影响,指出个性化癌症护理的有希望的发展。这篇叙述性综述综合了现有的知识,并为未来的调查绘制了路线,提出了饮食干预之间动态相互作用的整体观点,肠道微生物组,和癌症免疫疗法在MDI三方内。
    The following narrative review embarks on a comprehensive exploration of the role played by the gut microbiome within the Diet-Microbiota-Immunity (DMI) tripartite, aiming to enhance anti-cancer immunotherapy efficacy. While revolutionizing cancer treatment, resistance to immunotherapy and immune-related adverse events (irAEs) remain challenges. The tumor microenvironment (TME), shaped by cancer cells, influences immunotherapy resistance. The gut microbiome, influenced by genetics, environment, diet, and interventions, emerges as a critical player in TME reshaping, thereby modulating immune responses and treatment outcomes. Dietary patterns like the Mediterranean diet, caloric restriction modifications, and specific nutritional components show promise in influencing the tumor microenvironment and gut microbiome for better treatment outcomes. Antibiotics, disrupting gut microbiota diversity, may compromise immunotherapy efficacy. This review emphasizes the need for tailored nutritional strategies to manipulate microbial communities, enhance immune regulation, and improve immunotherapy accessibility while minimizing side effects. Ongoing studies investigate the impact of dietary interventions on cancer immunotherapy, pointing toward promising developments in personalized cancer care. This narrative review synthesizes existing knowledge and charts a course for future investigations, presenting a holistic perspective on the dynamic interplay between dietary interventions, the gut microbiome, and cancer immunotherapy within the DMI tripartite.
    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:随着免疫治疗研究的发展,强调了免疫检查点阻断(ICB)在宫颈癌治疗中的作用,但许多患者仍然无法从ICB中获得长期益处。聚ADP核糖聚合酶抑制剂(PARPi)已被证明在多种实体瘤中具有显着的抗肿瘤作用。宫颈癌患者是否从PARPi联合ICB的治疗方案中获得了更好的益处尚不清楚。
    方法:采用westernblot、免疫荧光和实时定量聚合酶链反应(qRT-PCR)检测尼拉帕尼诱导宫颈癌细胞PD-L1表达的改变及其机制。使用染色质免疫沉淀(ChIP)测定和RNA干扰证实了KDM5A对PTEN的调节。通过搜索在线数据库分析PD-L1与免疫效应分子之间的关系。尼拉帕尼的疗效,在同基因肿瘤模型中评估PD-L1阻断或组合。免疫组织化学(IHC)和qRT-PCR检测体内免疫细胞和细胞因子的变化。
    结果:我们发现尼拉帕尼在小鼠宫颈癌模型中上调PD-L1表达并增强PD-L1阻断的抗肿瘤作用。尼拉帕尼通过增加KDM5A的丰度抑制Pten的表达,通过激活PI3K-AKT-S6K1途径扩大PD-L1的丰度。PD-L1与包括TNF-α在内的免疫效应分子呈正相关,IFN-γ,基于生物信息分析的颗粒酶A和颗粒酶B。尼拉帕尼增加了CD8+T细胞的浸润和IFN-γ的水平,颗粒酶B在体内。
    结论:我们的研究结果表明尼拉帕尼对宫颈癌局部免疫微环境的调节作用,为支持PARPi联合PD-L1阻断作为宫颈癌潜在治疗提供了理论依据。
    OBJECTIVE: With the development of immunotherapy research, the role of immune checkpoint blockade (ICB) in the treatment of cervical cancer has been emphasized, but many patients still can\'t receive long-term benefits from ICB. Poly ADP ribose polymerase inhibitor (PARPi) has been proved to exert significant antitumor effects in multiple solid tumors. Whether cervical cancer patients obtain better benefits from the treatment regimen of PARPi combined with ICB remains unclear.
    METHODS: The alteration of PD-L1 expression induced by niraparib in cervical cancer cells and its underlying mechanism were assessed by western blot and immunofluorescence and quantitative real-time polymerase chain reaction (qRT-PCR).The regulation of PTEN by KDM5A was confirmed using Chromatin immunoprecipitation (ChIP) assay and RNA interference. Analyzing the relationship between PD-L1 and immune effector molecules through searching online databases. Therapeutic efficacy of niraparib, PD-L1 blockade or combination was assessed in syngeneic tumor model. The changes of immune cells and cytokines in vivo was detected by immunohistochemistry (IHC) and qRT-PCR.
    RESULTS: We found that niraparib upregulated PD-L1 expression and potentiated the antitumor effects of PD-L1 blockade in a murine cervical cancer model. Niraparib inhibited the Pten expression by increasing the abundance of KDM5A, which expanded PD-L1 abundance through activating the PI3K-AKT-S6K1 pathway. PD-L1 was positively correlated with immune effector molecules including TNF-α, IFN-γ, granzyme A and granzyme B based on biological information analysis. Niraparib increased the infiltration of CD8+ T cells and the level of IFN-γ, granzyme B in vivo.
    CONCLUSIONS: Our findings demonstrates the regulation of niraparib on local immune microenvironment of cervical cancer, and provides theoretical basis for supporting the combination of PARPi and PD-L1 blockade as a potential treatment for cervical 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
    前胶原c-蛋白酶增强蛋白(PCLCE)在改善前胶原c-蛋白酶的再生和促进细胞外基质的重建方面具有重要作用。PCLCE高表达与胃癌的阴性预后相关,卵巢癌,和骨肉瘤.这项工作的目的是研究PCLCE在神经胶质瘤中的功能。多种生物信息学技术已被用于研究PCLCE在神经胶质瘤中的作用,由PCLCE和预后之间的相关性组成,免疫检查点,免疫细胞浸润,和肿瘤微环境(TME)。基因本体论(GO)注释和京都基因和基因组百科全书(KEGG)分析用于评估PCLCE在神经胶质瘤中的潜在功能。发现PCLCE在神经胶质瘤中增加。我们发现PCLCE是一个潜在的预后因素,与肿瘤分级有关。上调PCLCE与低级别胶质瘤(LGG)的不良预后相关,多形性胶质母细胞瘤(GBM),和复发性神经胶质瘤.PCLCE与免疫细胞浸润相关,特别是B细胞,CD4+T细胞,巨噬细胞,中性粒细胞,和LGG中的树突状细胞(DC),GBM中的DCs浸润。PCLCE与许多与免疫和免疫检查点相关的基因共表达。此外,PCLCE低表达的胶质瘤患者对免疫检查点阻断(ICB)的反应较高.此外,PCLCE可能通过几种免疫相关的生物学过程或途径发挥作用,如白细胞迁移,激活T细胞,适应性免疫反应,中性粒细胞介导的免疫,NF-κB,和TNF信号通路。总之,PCLCE可能是一个新的免疫相关基因,通过免疫学途径调控肿瘤的发展。
    Procollagen c-protease enhancer protein (PCOLCE) performs an essential action in improving the recreation of procollagen c-protease and promoting the reconstruction of extracellular matrix. High PCOLCE expression was associated with a negative prognosis of stomach cancer, ovarian cancer, and osteosarcoma. The goal of this work is to investigate the function of PCOLCE in glioma. Multiple bioinformatics techniques have been employed to investigate the roles of PCOLCE in glioma, consisting of the correlation between PCOLCE and prognosis, immune checkpoints, immune cell infiltrates, and tumor microenvironment (TME). The gene ontology (GO) annotations and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were used to assess the potential function of PCOLCE in glioma. PCOLCE was found to be increased in glioma. We revealed that PCOLCE was a potential prognostic factor and related to tumor grade. Up-regulated PCOLCE was related to poor prognosis in lower-grade glioma (LGG), glioblastoma multiforme (GBM), and recurrent glioma. PCOLCE was correlated with immune cell infiltration, particularly B cells, CD4+ T cells, macrophages, neutrophils, and dendritic cells (DCs) in LGG, and DCs infiltration in GBM. PCOLCE was co-expressed with many genes related to the immune and the immune checkpoint. In addition, glioma patients with low expression of PCOLCE had a higher response to the immunological checkpoint blockade (ICB). Additionally, PCOLCE may exert its roles via several immune-related biological processes or pathways, such as leukocyte migration, activation of T cells, adaptive immune response, neutrophil-mediated immunity, NF-κB, and TNF signaling pathways. In conclusion, PCOLCE may be a new immune-related gene and regulate tumor development through immunological pathways.
    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
    口腔鳞状细胞癌(OSCC)影响全世界的大量个体。尽管手术取得了进步,辐射,和化疗,尚未取得令人满意的结果。近年来,靶向程序性细胞死亡1(PD-1)和程序性细胞死亡配体1(PD-L1)的药物的成功导致了癌症治疗的突破,但缺乏对其对抗OSCC有效性的系统总结。本文综述了PD-1/PD-L1通路的最新研究以及基于该通路的联合治疗OSCC的潜力。Further,它探讨了该途径与外泌体相互作用和蛋白质-蛋白质相互作用的机制,最后提出了未来潜在的OSCC治疗策略。
    Oral squamous cell carcinoma (OSCC) affects a large number of individuals worldwide. Despite advancements in surgery, radiation, and chemotherapy, satisfactory outcomes have not been achieved. In recent years, the success of drugs targeting programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PD-L1) has led to breakthroughs in cancer treatment, but systematic summaries on their effectiveness against OSCC are lacking. This article reviews the latest research on the PD-1/PD-L1 pathway and the potential of combination therapy based on this pathway in OSCC. Further, it explores the mechanisms involved in the interaction of this pathway with exosomes and protein-protein interactions, and concludes with potential future OSCC therapeutic strategies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    HIF1α-糖酵解在调节低氧T细胞中的IFN-γ诱导中的作用是未知的。鉴于缺氧是广泛的病理生理背景(如肿瘤)中的共同特征,并且IFN-γ有助于保护性免疫,对此有一个清晰的想法具有重要意义。结合药理学和遗传功能获得和功能丧失的方法,我们发现HIF1α-糖酵解控制缺氧激活的人和小鼠T细胞中IFN-γ的诱导。T细胞中HIF1α的特异性缺失(HIF1α-/-)和糖酵解抑制显著消除IFN-γ诱导。相反,通过缺氧和VHL缺失(VHL-/-)在T细胞中稳定HIF1α促进IFN-γ产生。机械上,低氧HIF1α-/-T细胞中IFN-γ产生的减少是由于减弱的活化诱导的细胞死亡而不是增殖缺陷。我们进一步表明,细胞内乙酰辅酶A的消耗是一个关键的代谢机制。缺氧HIF1α-/-T细胞对肿瘤细胞的杀伤能力较差,和HIF1α-/-荷瘤小鼠对免疫检查点阻断(ICB)治疗无反应,表明T细胞中HIF1α的丢失是ICB治疗抗性的主要机制。重要的是,补充乙酸盐可恢复低氧HIF1α-/-T细胞中IFN-γ的产生,并使HIF1α-/-荷瘤小鼠对ICB重新敏感,提供了克服ICB耐药性的有效策略。一起来看,我们的结果强调T细胞HIF1α-厌氧糖酵解是IFN-γ诱导和抗肿瘤免疫的主要介质。考虑到乙酸盐的补充(即,甘油三乙酸酯(GTA)被批准用于治疗患有Canavan病的婴儿,我们设想快速转化我们的发现,证明GTA作为ICB耐药性再利用药物的进一步测试,迫切的未满足的医疗需求。
    The role of HIF1α-glycolysis in regulating IFN-γ induction in hypoxic T cells is unknown. Given that hypoxia is a common feature in a wide array of pathophysiological contexts such as tumor and that IFN-γ is instrumental for protective immunity, it is of great significance to gain a clear idea on this. Combining pharmacological and genetic gain-of-function and loss-of-function approaches, we find that HIF1α-glycolysis controls IFN-γ induction in both human and mouse T cells activated under hypoxia. Specific deletion of HIF1α in T cells (HIF1α-/-) and glycolytic inhibition significantly abrogate IFN-γ induction. Conversely, HIF1α stabilization in T cells by hypoxia and VHL deletion (VHL-/-) promotes IFN-γ production. Mechanistically, reduced IFN-γ production in hypoxic HIF1α-/- T cells is due to attenuated activation-induced cell death but not proliferative defect. We further show that depletion of intracellular acetyl-CoA is a key metabolic underlying mechanism. Hypoxic HIF1α-/- T cells are less able to kill tumor cells, and HIF1α-/- tumor-bearing mice are not responsive to immune checkpoint blockade (ICB) therapy, indicating loss of HIF1α in T cells is a major mechanism of therapeutic resistance to ICBs. Importantly, acetate supplementation restores IFN-γ production in hypoxic HIF1α-/- T cells and re-sensitizes HIF1α-/- tumor-bearing mice to ICBs, providing an effective strategy to overcome ICB resistance. Taken together, our results highlight T cell HIF1α-anaerobic glycolysis as a principal mediator of IFN-γ induction and anti-tumor immunity. Considering that acetate supplementation (i.e., glycerol triacetate (GTA)) is approved to treat infants with Canavan disease, we envision a rapid translation of our findings, justifying further testing of GTA as a repurposed medicine for ICB resistance, a pressing unmet medical need.
    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
    目前的免疫疗法对T细胞耗竭肿瘤的益处有限,呼吁治疗创新。使用癌症患者数据的多组学整合,我们预测I型干扰素(IFN)应答树突状细胞(DC)疫苗的高状态,具有有效的临床影响。然而,临床前DC疫苗通过将免疫原性癌细胞死亡与I型IFN反应的诱导相结合来重现这种状态,但无法逆转缺乏T细胞浸润的小鼠肿瘤。这里,在淋巴结(LN),而不是激活CD4+/CD8+T细胞,DC刺激免疫抑制性程序性死亡-配体1阳性(PD-L1+)LN相关巨噬细胞(LAMs)。此外,DC疫苗还刺激PD-L1+肿瘤相关巨噬细胞(TAMs)。这产生了抑制CD8+T细胞的PD-L1+巨噬细胞的两个解剖学上不同的生态位。因此,PD-L1阻断与DC疫苗的组合通过消耗PD-L1+巨噬细胞实现显著的肿瘤消退,抑制骨髓炎症,和去抑制效应物/干细胞样记忆T细胞。重要的是,临床DC疫苗还可增强胶质母细胞瘤患者的T细胞抑制性PD-L1+TAMs.我们建议必须采用多模式免疫疗法和疫苗接种方案来克服T细胞耗尽的肿瘤。
    Current immunotherapies provide limited benefits against T cell-depleted tumors, calling for therapeutic innovation. Using multi-omics integration of cancer patient data, we predict a type I interferon (IFN) responseHIGH state of dendritic cell (DC) vaccines, with efficacious clinical impact. However, preclinical DC vaccines recapitulating this state by combining immunogenic cancer cell death with induction of type I IFN responses fail to regress mouse tumors lacking T cell infiltrates. Here, in lymph nodes (LNs), instead of activating CD4+/CD8+ T cells, DCs stimulate immunosuppressive programmed death-ligand 1-positive (PD-L1+) LN-associated macrophages (LAMs). Moreover, DC vaccines also stimulate PD-L1+ tumor-associated macrophages (TAMs). This creates two anatomically distinct niches of PD-L1+ macrophages that suppress CD8+ T cells. Accordingly, a combination of PD-L1 blockade with DC vaccines achieves significant tumor regression by depleting PD-L1+ macrophages, suppressing myeloid inflammation, and de-inhibiting effector/stem-like memory T cells. Importantly, clinical DC vaccines also potentiate T cell-suppressive PD-L1+ TAMs in glioblastoma patients. We propose that a multimodal immunotherapy and vaccination regimen is mandatory to overcome T cell-depleted 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
    这里,我们展示了两个现实世界的例子,以人为本,多健康状况方法-也称为多发病率方法-已在大曼彻斯特的一般实践中应用于长期状况管理。第一个例子是在曼彻斯特市的一般实践中,通过人口健康管理方法实施有针对性的多种健康状况审查。第二个例子是以人为中心的风险分层工具的开发,专注于未满足的心血管需求,名为“CVNeed”。该工具提供了一种独特的方法来突出最高的未满足需求,因此,积极努力对系统的最大投资回报。这些例子展示了如何以人为本,长期病情管理的多种健康状况方法,通过数据智能来驱动临床需求的优先级,可以帮助解决长期的健康不平等和健康结果的不合理变化。这项工作还强调了综合护理系统(ICS)协同工作以解决系统中的健康不平等问题的潜力,地区和邻里水平,从而在实现大曼彻斯特综合护理伙伴关系(ICP)战略中提出的愿景方面取得了重大进展。
    Here, we present two real-world examples of how a prioritised, person-centred, multiple health condition approach - also termed a multimorbidity approach - has been applied to long-term condition management in general practice in Greater Manchester. The first example is the implementation of targeted multiple health condition reviews via a population health management approach across general practice in the City of Manchester. The second example is the development of a person-centred risk stratification tool, focused on unmet cardiovascular need, called \'CVNeed\'. This tool provides a unique approach to highlighting the highest unmet need and, thus, the largest return on investment to the system from proactive efforts. These examples demonstrate how a person-centred, multiple health condition approach to long-term condition management, enabled by data intelligence to drive prioritisation of clinical need, can help to address longstanding health inequalities and unwarranted variation in health outcomes. This work also highlights the potential for integrated care systems (ICS) to work collaboratively to tackle health inequalities at a system, locality and neighbourhood level, thus making significant strides toward achieving the vision set out in the Greater Manchester Integrated Care Partnership (ICP) Strategy.
    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
    胶质瘤是世界上最常见的原发性恶性脑肿瘤,胶质母细胞瘤(GBM)是最常见和侵袭性的类型。尽管二十年来不懈地追求探索GBM的新治疗方法,在改善患者生存结局方面进展有限。许多障碍阻碍了GBM的有效治疗,包括免疫抑制肿瘤微环境(TME),血脑屏障,和广泛的异质性。尽管面临这些挑战,免疫疗法正在成为一个有希望的途径,可能为治疗神经胶质瘤提供新的希望。神经胶质瘤的免疫疗法有四种主要类型,免疫检查点封锁,嵌合抗原受体T细胞疗法,疫苗,和溶瘤病毒。此外,基因治疗,双特异性抗体治疗,本文还简要介绍了联合疗法。在许多研究中都强调了TME在免疫治疗过程中的重要作用。虽然免疫疗法是治疗神经胶质瘤的一种有希望的治疗方法,需要付出巨大的努力来克服现有的成功障碍。由于神经胶质瘤免疫治疗的迅速发展和日益受到重视,本文就神经胶质瘤免疫疗法的最新进展作一综述。
    Gliomas are the most prevalent primary malignant brain tumors worldwide, with glioblastoma (GBM) being the most common and aggressive type. Despite two decades of relentless pursuit in exploring novel therapeutic approaches for GBM, there is limited progress in improving patients\' survival outcomes. Numerous obstacles impede the effective treatment of GBM, including the immunosuppressive tumor microenvironment (TME), the blood-brain barrier, and extensive heterogeneity. Despite these challenges, immunotherapies are emerging as a promising avenue that may offer new hope for the treatment of gliomas. There are four main types of immunotherapies for gliomas, immune checkpoint blockades, chimeric antigen receptor T-cell therapies, vaccines, and oncolytic viruses. In addition, gene therapy, bispecific antibody therapy, and combine therapy are also briefly introduced in this review. The significant role of TME in the process of immunotherapies has been emphasized in many studies. Although immunotherapy is a promising treatment for gliomas, enormous effort is required to overcome the existing barriers to its success. Owing to the rapid development and increasing attention paid to immunotherapies for gliomas, this article aims to review the recent advances in immunotherapies for gliomas.
    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
    近年来,由于某些患者的肿瘤清除率显着,癌症免疫疗法获得了广泛的关注。尽管免疫检查点阻断(ICB)在多种恶性肿瘤中取得了显著成功,免疫系统参与靶向前列腺癌(PCa)治疗仍处于起步阶段。多种因素导致反应有限,包括PCa的异质性,寒冷的肿瘤微环境,和低数量的新抗原。在改善基于免疫的PCa疗法方面投入了大量努力。本文就免疫治疗在PCa治疗中的应用现状作一综述。讨论了多种免疫模式,包括疫苗,过继转移的T细胞,和双特异性T细胞衔接者,其中一些正在进行临床试验。此外,这篇综述还侧重于新兴的基于机制的小分子酪氨酸激酶抑制剂,具有免疫调节特性,作为单一药物或与其他免疫疗法联合使用,有可能改善临床结果。
    Cancer immunotherapy has gained traction in recent years owing to remarkable tumor clearance in some patients. Despite the notable success of immune checkpoint blockade (ICB) in multiple malignancies, engagement of the immune system for targeted prostate cancer (PCa) therapy is still in its infancy. Multiple factors contribute to limited response, including the heterogeneity of PCa, the cold tumor microenvironment, and a low number of neoantigens. Significant effort is being invested in improving immune-based PCa therapies. This review is a summary of the status of immunotherapy in treating PCa, with a discussion of multiple immune modalities, including vaccines, adoptively transferred T cells, and bispecific T cell engagers, some of which are undergoing clinical trials. In addition, this review also focuses on emerging mechanism-based small-molecule tyrosine kinase inhibitors with immune modulatory properties that, either as single agents or in combination with other immunotherapies, have the potential to improve clinical 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
    高级别浆液性卵巢癌(HGSOC)是大多数妇科癌症相关死亡的原因。处于缓解期的患者通常在治疗后2年内以更具侵袭性的疾病形式复发。替代免疫肿瘤学(IO)策略,例如靶向PD-(L)1信号轴的免疫检查点阻断(ICB),到目前为止,事实证明效率低下。我们的目标是利用表观遗传调节剂在离体3D患者衍生平台和体内同源模型中最大化个性化IO组合的益处。使用患者来源的肿瘤腹水,我们优化了离体3D筛选平台(PDOTS),使用自体免疫细胞和循环腹水来源的肿瘤细胞,快速测试个性化IO组合。最重要的是,患者对铂类化疗和聚ADP核糖聚合酶抑制剂在3D平台中的反应概括了临床反应.此外,与临床试验结果相似,PDOTS中对ICB的反应往往较低,并且与CD3免疫细胞和EPCAM/PD-L1肿瘤细胞的频率呈正相关。因此,在患者来源的HGSOC腹水中观察到单独使用抗PD-1/抗PD-L1免疫疗法观察到的最大反应,在免疫和肿瘤细胞中呈现高水平的系统性CD3+和PD-L1+表达,分别。此外,在离体3D测试平台和体内肿瘤模型中,用表观遗传佐剂引发极大地增强ICB。我们进一步发现,表观遗传引发诱导肿瘤分泌增加的几种关键细胞因子,已知增强T和NK细胞活化和细胞毒性,包括IL-6、IP-10(CXCL10)、KC(CXCL1),和RANTES(CCL5)。此外,在患者来源的PDOTS中单独的表观遗传引发以及与ICB免疫疗法组合诱导CD69的快速上调,CD69是CD4+和CD8+T细胞中免疫标志物的可靠早期激活。因此,这种功能精准医学方法可以快速识别能够增强ICB的个性化治疗组合,这是一个很大的优势,特别是考虑到目前在患者中测试大量潜在组合的临床困难。
    High-grade serous ovarian cancer (HGSOC) is responsible for the majority of gynecology cancer-related deaths. Patients in remission often relapse with more aggressive forms of disease within 2 years post-treatment. Alternative immuno-oncology (IO) strategies, such as immune checkpoint blockade (ICB) targeting the PD-(L)1 signaling axis, have proven inefficient so far. Our aim is to utilize epigenetic modulators to maximize the benefit of personalized IO combinations in ex vivo 3D patient-derived platforms and in vivo syngeneic models. Using patient-derived tumor ascites, we optimized an ex vivo 3D screening platform (PDOTS), which employs autologous immune cells and circulating ascites-derived tumor cells, to rapidly test personalized IO combinations. Most importantly, patient responses to platinum chemotherapy and poly-ADP ribose polymerase inhibitors in 3D platforms recapitulate clinical responses. Furthermore, similar to clinical trial results, responses to ICB in PDOTS tend to be low and positively correlated with the frequency of CD3+ immune cells and EPCAM+/PD-L1+ tumor cells. Thus, the greatest response observed with anti-PD-1/anti-PD-L1 immunotherapy alone is seen in patient-derived HGSOC ascites, which present with high levels of systemic CD3+ and PD-L1+ expression in immune and tumor cells, respectively. In addition, priming with epigenetic adjuvants greatly potentiates ICB in ex vivo 3D testing platforms and in vivo tumor models. We further find that epigenetic priming induces increased tumor secretion of several key cytokines known to augment T and NK cell activation and cytotoxicity, including IL-6, IP-10 (CXCL10), KC (CXCL1), and RANTES (CCL5). Moreover, epigenetic priming alone and in combination with ICB immunotherapy in patient-derived PDOTS induces rapid upregulation of CD69, a reliable early activation of immune markers in both CD4+ and CD8+ T cells. Consequently, this functional precision medicine approach could rapidly identify personalized therapeutic combinations able to potentiate ICB, which is a great advantage, especially given the current clinical difficulty of testing a high number of potential combinations in patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号