checkpoint blockade

检查站封锁
  • 文章类型: Journal Article
    针对程序性细胞死亡蛋白1(anti-PD-1)的抗体已成为三阴性乳腺癌(TNBC)的有希望的免疫疗法,通过T细胞PD-1受体结合阻断肿瘤细胞的PD-L1信号传导。然而,只有10-20%符合标准的PD-L1+转移性TNBC患者受益于ICB,预测患者反应的生物标志物一直难以捉摸。我们以前开发了一种免疫学利基,由皮下空间的微孔植入物组成,支持组织形成,其免疫成分与重要器官内的免疫成分一致。在这里,我们研究了该免疫生态位内的动态基因表达,以提供抗PD-1反应的生物标志物。在转移性TNBC的4T1模型中,我们根据原发肿瘤的生长和生存率观察了抗PD-1的敏感性和耐药性.利基以前是活检的,during,抗PD-1治疗后,并分析指示治疗折射率的细胞类型和基因表达。在ICB敏感性和耐药性之间,髓系细胞与淋巴细胞的比率发生了变化。基因表达的纵向分析涉及动态骨髓细胞功能,将敏感性与耐药性分层。生态位衍生的基因标签预测治疗前的敏感性或抗性。对监测免疫疗法反应的利基分析为个性化护理和研究潜在的治疗抵抗机制提供了新的机会。
    Antibodies to programmed cell death protein1 (anti-PD-1) have become a promising immunotherapy for triple negative breast cancer (TNBC), blocking PD-L1 signaling from pro-tumor cells through T cell PD-1 receptor binding. Nevertheless, only 10-20% of PD-L1+ metastatic TNBC patients who meet criteria benefit from ICB, and biomarkers to predict patient response have been elusive. We have previously developed an immunological niche, consisting of a microporous implant in the subcutaneous space, that supports tissue formation whose immune composition is consistent with that within vital organs. Herein, we investigated dynamic gene expression within this immunological niche to provide biomarkers of response to anti-PD-1. In a 4T1 model of metastatic TNBC, we observed sensitivity and resistance to anti-PD-1 based on primary tumor growth and survival. The niche was biopsied before, during, and after anti-PD-1 therapy, and analyzed for cell types and gene expression indicative of treatment refractivity. Myeloid cell-to-lymphocyte ratios were altered between ICB-sensitivity and resistance. Longitudinal analysis of gene expression implicated dynamic myeloid cell function that stratified sensitivity from resistance. A niche-derived gene signature predicted sensitivity or resistance prior to therapy. Analysis of the niche to monitor immunotherapy response presents a new opportunity to personalize care and investigate mechanisms underlying treatment resistance.
    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
    Gasdermin介导的炎性细胞死亡(焦亡)可以在免疫冷肿瘤中激活保护性免疫。这里,我们对可以激活gasderminD(GSDMD)的化合物进行了高通量筛选,在肿瘤中广泛表达。我们确定了6,7-二氯-2-甲基磺酰基-3-N-叔丁基氨基喹喔啉(DMB)作为直接和选择性的GSDMD激动剂,可激活GSDMD孔形成和焦亡而不裂解GSDMD。在小鼠肿瘤模型中,DMB诱导的脉冲和低水平的焦亡抑制肿瘤生长而不损害表达GSDMD的免疫细胞。保护是免疫介导的,在缺乏淋巴细胞的小鼠中被废除。用DMB处理的癌细胞接种疫苗可保护小鼠免受继发性肿瘤攻击,表明免疫原性细胞死亡被诱导。DMB治疗与抗PD-1协同作用。DMB治疗不会改变循环的促炎细胞因子或白细胞数量或导致体重减轻。因此,我们的研究揭示了一种策略,该策略依赖于低水平的肿瘤细胞焦亡来诱导抗肿瘤免疫,并提高了利用焦亡而不引起明显毒性的可能性。
    Gasdermin-mediated inflammatory cell death (pyroptosis) can activate protective immunity in immunologically cold tumors. Here, we performed a high-throughput screen for compounds that could activate gasdermin D (GSDMD), which is expressed widely in tumors. We identified 6,7-dichloro-2-methylsulfonyl-3-N-tert-butylaminoquinoxaline (DMB) as a direct and selective GSDMD agonist that activates GSDMD pore formation and pyroptosis without cleaving GSDMD. In mouse tumor models, pulsed and low-level pyroptosis induced by DMB suppresses tumor growth without harming GSDMD-expressing immune cells. Protection is immune-mediated and abrogated in mice lacking lymphocytes. Vaccination with DMB-treated cancer cells protects mice from secondary tumor challenge, indicating that immunogenic cell death is induced. DMB treatment synergizes with anti-PD-1. DMB treatment does not alter circulating proinflammatory cytokine or leukocyte numbers or cause weight loss. Thus, our studies reveal a strategy that relies on a low level of tumor cell pyroptosis to induce antitumor immunity and raise the possibility of exploiting pyroptosis without causing overt toxicity.
    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
    使用抗PD-1和抗CTLA-4抗体的组合检查点阻断在黑素瘤中显示出有希望的功效。然而,人类的潜在机制尚不清楚。这里,我们对36例接受抗PD-1,抗CTLA-4或联合治疗的IV期黑色素瘤患者进行了跨时间的单细胞RNA和T细胞受体(TCR)配对测序.我们开发了Cyclone算法来跟踪时间克隆动力学和基础细胞状态。检查点阻断诱导在不同时间点达到峰值的克隆T细胞应答波。与单药治疗相比,联合治疗在6周和9周时导致更大的克隆反应。包括黑色素瘤特异性CD8+T细胞和耗尽的CD8+T细胞(TEX)克隆。TEX的集中分析确定抗CTLA-4诱导祖细胞TEX的强大扩增和增殖,在联合治疗期间与抗PD-1协同作用以恢复TEX的活力。这些下一代免疫分析方法可以指导药物的选择,时间表,和新型组合策略的剂量。
    Combination checkpoint blockade with anti-PD-1 and anti-CTLA-4 antibodies has shown promising efficacy in melanoma. However, the underlying mechanism in humans remains unclear. Here, we perform paired single-cell RNA and T cell receptor (TCR) sequencing across time in 36 patients with stage IV melanoma treated with anti-PD-1, anti-CTLA-4, or combination therapy. We develop the algorithm Cyclone to track temporal clonal dynamics and underlying cell states. Checkpoint blockade induces waves of clonal T cell responses that peak at distinct time points. Combination therapy results in greater magnitude of clonal responses at 6 and 9 weeks compared to single-agent therapies, including melanoma-specific CD8+ T cells and exhausted CD8+ T cell (TEX) clones. Focused analyses of TEX identify that anti-CTLA-4 induces robust expansion and proliferation of progenitor TEX, which synergizes with anti-PD-1 to reinvigorate TEX during combination therapy. These next generation immune profiling approaches can guide the selection of drugs, schedule, and dosing for novel combination strategies.
    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: Clinical Trial, Phase II
    Relatlimab(rela;抗LAG-3)加nivolumab(nivo;抗PD-1)可安全有效地治疗晚期黑色素瘤。我们设计了一项试验(NCT03743766),其中晚期黑色素瘤患者接受rela,尼沃,或rela+nivo询问rela+nivo的免疫机制。这项正在进行的试验的生物标本分析表明,rela+nivo导致CD8+T细胞受体信号传导能力增强,CD8+T细胞分化改变,尽管保留了耗尽曲线,但仍导致细胞毒性升高。细胞毒性和耗竭特征的共表达是由PRDM1、BATF、ETV7和TOX。在rela+nivo后出现的克隆扩增的CD8+T细胞中,效应子功能上调。rela+nivo肿瘤内CD8+T细胞特征与良好的预后相关。这种肿瘤内rela+nivo特征在外周血中被验证为CD38+TIM3+CD8+T细胞的频率升高。总的来说,我们证明,尽管保留了耗尽特征,但细胞毒性可以增强,这将为未来的治疗策略提供信息。
    Relatlimab (rela; anti-LAG-3) plus nivolumab (nivo; anti-PD-1) is safe and effective for treatment of advanced melanoma. We designed a trial (NCT03743766) where advanced melanoma patients received rela, nivo, or rela+nivo to interrogate the immunologic mechanisms of rela+nivo. Analysis of biospecimens from this ongoing trial demonstrated that rela+nivo led to enhanced capacity for CD8+ T cell receptor signaling and altered CD8+ T cell differentiation, leading to heightened cytotoxicity despite the retention of an exhaustion profile. Co-expression of cytotoxic and exhaustion signatures was driven by PRDM1, BATF, ETV7, and TOX. Effector function was upregulated in clonally expanded CD8+ T cells that emerged after rela+nivo. A rela+nivo intratumoral CD8+ T cell signature was associated with a favorable prognosis. This intratumoral rela+nivo signature was validated in peripheral blood as an elevated frequency of CD38+TIM3+CD8+ T cells. Overall, we demonstrated that cytotoxicity can be enhanced despite the retention of exhaustion signatures, which will inform future 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: Congress
    第21届癌症免疫治疗协会(CIMT)年会于5月15日至5月17日在美因茨举行,德国,来自33个不同国家的855名学术和临床专业人员参加了会议。会议为这些专家召集和讨论癌症免疫学和免疫疗法研究的最新突破提供了平台。专门的会议涵盖了用于癌症免疫治疗研究的人工智能工具的进步,以及非洲大陆癌症治疗和癌症免疫治疗试验的前景,在与会者中引发了生动而翔实的讨论。本报告旨在概述MT2024年CI最值得注意的亮点和主要收获。
    The 21st Association for Cancer Immunotherapy (CIMT) Annual Meeting took place from May 15th to May 17th in Mainz, Germany, and was attended by a total of 855 academic and clinical professionals hailing from 33 different countries. The conference served as a platform for these experts to convene and discuss the latest breakthroughs in cancer immunology and immunotherapy research. Dedicated sessions covering advancements in artificial intelligence tools for cancer immunotherapy research, as well as the landscape of cancer care and cancer immunotherapy trials on the African continent, prompted lively and informative discussions among the attendees. This report aims to provide an overview of the most noteworthy highlights and key takeaways from CIMT2024.
    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
    纤维化间质和血管生成肿瘤血管在调节肿瘤免疫中起重要作用。我们先前报道了一种合理设计的蛋白质(ProAgio),该蛋白质在新位点靶向整合素αvβ3。ProAgio诱导表达高水平整合素的细胞的凋亡。肿瘤中活化的癌症相关成纤维细胞(CAF)和血管生成内皮细胞(aEC)均表达高水平的整合素αvβ3。ProAgio同时并特异性诱导肿瘤中CAF和aECs的凋亡。我们在此提供证据,证明ProAgio耗尽CAF和消除渗漏的肿瘤血管生成血管会改变肿瘤免疫力。ProAgio减少CD4+Treg和髓系来源的抑制细胞(MDSCs),增加CD8+T细胞,并增加肿瘤中M1/M2巨噬细胞的比例。ProAgio对致密纤维化基质(CAFs)的消耗降低了肿瘤周围基质区域的程序性死亡配体1(PDL-1)水平,并因此强烈增加抗PDL-1抗体向靶癌细胞的递送。ProAgio对肿瘤免疫的影响提供了检查点抑制剂对肺癌治疗的强协同作用。
    Fibrotic stroma and angiogenic tumor vessels play an important role in modulating tumor immunity. We previously reported a rationally designed protein (ProAgio) that targets integrin αvβ3 at a novel site. ProAgio induces the apoptosis of cells that express high levels of the integrin. Both activated cancer-associated fibroblasts (CAFs) and angiogenic endothelial cells (aECs) in tumors express high levels of integrin αvβ3. ProAgio simultaneously and specifically induces apoptosis in CAFs and aECs in tumors. We provide evidence here that the depletion of CAFs and the elimination of leaky tumor angiogenic vessels by ProAgio alter tumor immunity. ProAgio reduces CD4+ Treg and Myeloid-derived suppressor cells (MDSCs), increases CD8+ T-cells, and increases the M1/M2 macrophage ratio in the tumor. The depletion of dense fibrotic stroma (CAFs) by ProAgio decreases the Programmed Death Ligand 1 (PDL-1) levels in the stroma areas surrounding the tumors, and thus strongly increases the delivery of anti-PDL-1 antibody to the target cancer cells. The impact of ProAgio on tumor immunity provides strong synergistical effects of checkpoint inhibitors on lung 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
    过去十年见证了癌症治疗的革命,从常规药物(化学疗法)转向靶向分子疗法和基于免疫的疗法,特别是免疫检查点抑制剂(ICIs)。这些免疫疗法释放宿主对肿瘤的免疫系统,并对那些被认为无法治愈的癌症患者显示出前所未有的持久缓解,例如转移性黑色素瘤,转移性肾细胞癌(RCC),微卫星不稳定性(MSI)高结直肠癌和晚期非小细胞肺癌(NSCLC)。然而,约80%的患者对这些免疫疗法没有反应,因此只能接受其他效果较低且潜在毒性的治疗。识别和理解使癌细胞适应并最终克服治疗的机制可以帮助规避耐药性并改善治疗。在这次审查中,我们描述了控制肿瘤微环境的肿瘤免疫过程及其对PD-1/PD-L1检查点阻断耐药性的影响的最新发现.
    The past decade has witnessed a revolution in cancer treatment, shifting from conventional drugs (chemotherapies) towards targeted molecular therapies and immune-based therapies, in particular immune-checkpoint inhibitors (ICIs). These immunotherapies release the host\'s immune system against the tumor and have shown unprecedented durable remission for patients with cancers that were thought incurable, such as metastatic melanoma, metastatic renal cell carcinoma (RCC), microsatellite instability (MSI) high colorectal cancer and late stages of non-small cell lung cancer (NSCLC). However, about 80% of the patients fail to respond to these immunotherapies and are therefore left with other less effective and potentially toxic treatments. Identifying and understanding the mechanisms that enable cancerous cells to adapt to and eventually overcome therapy can help circumvent resistance and improve treatment. In this review, we describe the recent discoveries on the onco-immunological processes which govern the tumor microenvironment and their impact on the resistance to PD-1/PD-L1 checkpoint blockade.
    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
    免疫检查点抑制剂(CPI)诱导的糖尿病(CPI-DM)是一种罕见的免疫相关不良事件(irAE)。患者和提供者担心持续的CPIs会使患者面临其他irAE的风险,因此可能会停止治疗。目前,几乎没有数据可以告知这一决定。因此,本研究旨在阐明CPI-DM诊断后停止CPIs是否会影响未来irAE的发展以及进展和死亡等癌症结局.对2015年7月1日至2023年7月5日在UCSF治疗期间发生CPI-DM的患者进行癌症结果和irAE发展分析。费希尔的精确检验,学生t检验,Kaplan-Meier方法,适当时使用Cox回归。在43例CPI-DM患者中,20(47%)在IRAE发生后90天内恢复了CPIs,4例(9%)患者在90天后重新开始,和19(44%)患者从未重新启动。在恢复CPIs的24人中有9人(38%)和停止CPIs的19人中有3人(16%)被诊断出随后的irAE(p=0.17)。两组之间的死亡(p=0.74)或癌症进展(p=0.55)没有显着差异。虽然我们的单机构研究在停止CPIs后没有显示更坏的癌症结果,许多变量会影响结果,我们的研究没有足够的能力来评估。需要一种细致入微的方法来决定在CPI-DM等严重的irAE之后是否继续CPI治疗。
    Immune checkpoint inhibitor (CPI)-induced diabetes mellitus (CPI-DM) is a rare immune-related adverse event (irAE). Patients and providers fear that continuing CPIs puts patients at risk for additional irAEs and thus may discontinue therapy. Currently, there are little data to inform this decision. Therefore, this study aims to elucidate whether discontinuing CPIs after diagnosis of CPI-DM impacts the development of future irAEs and cancer outcomes such as progression and death. Patients who developed CPI-DM during cancer treatment at UCSF from 1 July 2015 to 5 July 2023 were analyzed for cancer outcomes and irAE development. Fisher\'s exact tests, Student t-tests, Kaplan-Meier methods, and Cox regression were used as appropriate. Of the 43 patients with CPI-DM, 20 (47%) resumed CPIs within 90 days of the irAE, 4 (9%) patients restarted after 90 days, and 19 (44%) patients never restarted. Subsequent irAEs were diagnosed in 9 of 24 (38%) who resumed CPIs and 3 of 19 (16%) who discontinued CPIs (p = 0.17). There was no significant difference in death (p = 0.74) or cancer progression (p = 0.55) between these two groups. While our single-institution study did not show worse cancer outcomes after discontinuing CPIs, many variables can impact outcomes, which our study was not adequately powered to evaluate. A nuanced approach is needed to decide whether to continue CPI treatment after a severe irAE like CPI-DM.
    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
    人乳头瘤病毒(HPV)16和18感染与许多人类癌症有关。尽管有几种针对高风险(hr)HPV的预防性疫苗,仍然迫切需要开发用于靶向预先存在的hrHPV感染和病变的治疗性HPV疫苗.在这项研究中,我们开发了一种脂质纳米颗粒(LNP)配制的基于mRNA的HPV治疗性疫苗(mHTV)-03E2,同时靶向HPV16和HPV18的E2/E6/E7.mHTV-03E2显著诱导抗原特异性细胞免疫反应,在来自表达HPVE6/E7抗原的C57BL/6小鼠的原代肺上皮细胞的TC-1肿瘤中导致显著的CD8+T细胞浸润和细胞毒性,介导的显著肿瘤消退,延长动物的存活时间,以剂量依赖的方式。在免疫和远处肿瘤挑战实验中,我们进一步证明了针对HPV16/18E6/E7抗原的显著T细胞免疫接种后长达4个月,提示针对复发的健壮记忆T细胞免疫。最后,mHTV-03E2与免疫检查点阻断协同抑制肿瘤生长并延长动物存活,表明联合治疗的潜力。我们得出的结论是,mHTV-03E2是治疗由HPV16或HPV18感染引起的恶性肿瘤的出色候选治疗性mRNA疫苗。
    Human papillomavirus (HPV) 16 and 18 infections are related to many human cancers. Despite several preventive vaccines for high-risk (hr) HPVs, there is still an urgent need to develop therapeutic HPV vaccines for targeting pre-existing hrHPV infections and lesions. In this study, we developed a lipid nanoparticle (LNP)-formulated mRNA-based HPV therapeutic vaccine (mHTV)-03E2, simultaneously targeting the E2/E6/E7 of both HPV16 and HPV18. mHTV-03E2 dramatically induced antigen-specific cellular immune responses, leading to significant CD8+ T cell infiltration and cytotoxicity in TC-1 tumors derived from primary lung epithelial cells of C57BL/6 mice expressing HPV E6/E7 antigens, mediated significant tumor regression, and prolonged animal survival, in a dose-dependent manner. We further demonstrated significant T cell immunity against HPV16/18 E6/E7 antigens for up to 4 months post-vaccination in immunological and distant tumor rechallenging experiments, suggesting robust memory T cell immunity against relapse. Finally, mHTV-03E2 synergized with immune checkpoint blockade to inhibit tumor growth and extend animal survival, indicating the potential in combination therapy. We conclude that mHTV-03E2 is an excellent candidate therapeutic mRNA vaccine for treating malignancies caused by HPV16 or HPV18 infections.
    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
    皮肤T细胞淋巴瘤(CTCL)是具有低存活率和有限治疗的皮肤癌。虽然免疫疗法已经显示出一定的疗效,对CTCL患者施用免疫激活剂的免疫学后果尚未得到系统表征.我们应用一套高维技术来调查当地,细胞,CTCL患者接受抗PD-1单药或联合干扰素-γ(IFN-γ)治疗的全身反应。肿瘤T细胞在免疫疗法后没有显示活化的证据。IFN-γ诱导沉默的内源性免疫反应,虽然抗PD-1引起更广泛的变化,包括CLA+CD39+T细胞的丰度增加。我们开发了一种无偏见的多维分析方法,能够发现对患者进行分层的免疫模块。我们确定了非应答者中激活的调节性CLACD39T细胞和白血病患者中激活的细胞毒性CLACD39T细胞的富集。我们的研究结果为CTCL患者的免疫治疗效果提供了见解,并为临床试验的多维分析提供了可推广的框架。
    Cutaneous T cell lymphomas (CTCLs) are skin cancers with poor survival rates and limited treatments. While immunotherapies have shown some efficacy, the immunological consequences of administering immune-activating agents to CTCL patients have not been systematically characterized. We apply a suite of high-dimensional technologies to investigate the local, cellular, and systemic responses in CTCL patients receiving either mono- or combination anti-PD-1 plus interferon-gamma (IFN-γ) therapy. Neoplastic T cells display no evidence of activation after immunotherapy. IFN-γ induces muted endogenous immunological responses, while anti-PD-1 elicits broader changes, including increased abundance of CLA+CD39+ T cells. We develop an unbiased multi-omic profiling approach enabling discovery of immune modules stratifying patients. We identify an enrichment of activated regulatory CLA+CD39+ T cells in non-responders and activated cytotoxic CLA+CD39+ T cells in leukemic patients. Our results provide insights into the effects of immunotherapy in CTCL patients and a generalizable framework for multi-omic analysis of clinical trials.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号