checkpoint blockade

检查站封锁
  • 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

    求助全文

  • 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)

  • 文章类型: Journal Article
    具有PD-1/PD-L1抑制的免疫检查点阻断(ICB)彻底改变了非小细胞肺癌(NSCLC)的治疗。持久的响应,然而,仅在患者亚群中观察到。有缺陷的抗原呈递和免疫抑制性肿瘤微环境(TME)可导致T细胞募集不足和ICB抗性。我们评估了CXCL9和CXCL10工程化树突状细胞(CXCL9/10-DC)的肿瘤内(IT)疫苗接种作为克服耐药性的策略。ITCXCL9/10-DC导致TME中增强的T细胞浸润和活化以及小鼠NSCLC模型中的肿瘤抑制。ITCXCL9/10-DC的抗肿瘤功效依赖于CD4+和CD8+T细胞,以及CXCR3依赖性T细胞从淋巴结运输。ITCXCL9/10-DC,结合ICB,克服抗性并在鼠模型中建立系统性肿瘤特异性免疫。这些研究提供了对CXCL9/10-DC介导的宿主免疫激活的机制理解,并支持ITCXCL9/10-DC的临床翻译以增强NSCLC中的ICB功效。
    Immune checkpoint blockade (ICB) with PD-1/PD-L1 inhibition has revolutionized the treatment of non-small cell lung cancer (NSCLC). Durable responses, however, are observed only in a subpopulation of patients. Defective antigen presentation and an immunosuppressive tumor microenvironment (TME) can lead to deficient T cell recruitment and ICB resistance. We evaluate intratumoral (IT) vaccination with CXCL9- and CXCL10-engineered dendritic cells (CXCL9/10-DC) as a strategy to overcome resistance. IT CXCL9/10-DC leads to enhanced T cell infiltration and activation in the TME and tumor inhibition in murine NSCLC models. The antitumor efficacy of IT CXCL9/10-DC is dependent on CD4+ and CD8+ T cells, as well as CXCR3-dependent T cell trafficking from the lymph node. IT CXCL9/10-DC, in combination with ICB, overcomes resistance and establishes systemic tumor-specific immunity in murine models. These studies provide a mechanistic understanding of CXCL9/10-DC-mediated host immune activation and support clinical translation of IT CXCL9/10-DC to augment ICB efficacy in NSCLC.
    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
    放射疗法(RT)和抗PD-L1协同作用以增强局部和远处(横隔)肿瘤控制。然而,人类的临床结果是可变的。为了改善临床结果,我们研究了肿瘤引流淋巴结(TdLN)中CD8+PD-1+Tcf-1+干细胞样T细胞亚群的潜在协同机制.使用小鼠黑色素瘤模型,我们发现RT+抗PD-L1在TdLN茎样细胞中诱导了一种新的分化程序,这导致它们在肿瘤内扩增并分化成效应细胞.我们的数据表明,RT+抗PD-L1之间的最佳协同作用取决于TdLN干细胞样T细胞群,因为TdLN出口的阻断或特异性干细胞样T细胞耗竭降低了肿瘤控制。一起,这些数据表明,在TdLN中开始并在肿瘤中完成的联合治疗后,干细胞样T细胞的多步骤刺激.
    Radiotherapy (RT) and anti-PD-L1 synergize to enhance local and distant (abscopal) tumor control. However, clinical results in humans have been variable. With the goal of improving clinical outcomes, we investigated the underlying synergistic mechanism focusing on a CD8+ PD-1+ Tcf-1+ stem-like T cell subset in the tumor-draining lymph node (TdLN). Using murine melanoma models, we found that RT + anti-PD-L1 induces a novel differentiation program in the TdLN stem-like population which leads to their expansion and differentiation into effector cells within the tumor. Our data indicate that optimal synergy between RT + anti-PD-L1 is dependent on the TdLN stem-like T cell population as either blockade of TdLN egress or specific stem-like T cell depletion reduced tumor control. Together, these data demonstrate a multistep stimulation of stem-like T cells following combination therapy which is initiated in the TdLN and completed in the tumor.
    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
    癌症患者通常接受靶向程序性死亡配体1(PD-L1)和细胞毒性T淋巴细胞抗原4(CTLA4)的抗体组合。我们在头颈部鳞状细胞癌(HNSCC)中进行了机会窗研究,以检查抗CTLA4对抗PD-L1治疗的贡献。与治疗前活检的单细胞谱分析将T细胞扩增鉴定为早期反应标志物。在肿瘤中,抗PD-L1触发了大部分CD8+T细胞的扩增,而联合治疗扩增CD4+和CD8+T细胞。这样的CD4+T细胞表现出活化的T辅助1(Th1)表型。CD4+和CD8+T细胞与表达T细胞归巢因子的树突状细胞或产生抗体的浆细胞共定位并被其包围。T细胞受体追踪表明,抗CTLA4,而不是抗PD-L1,触发CD4+幼稚/中枢记忆T细胞从肿瘤引流淋巴结(tdLN)的运输,通过血,T细胞获得Th1表型的肿瘤。因此,CD4+T细胞活化和来自tdLN的募集是HNSCC中对抗PD-L1加抗CTLA4的早期反应的标志。
    Cancer patients often receive a combination of antibodies targeting programmed death-ligand 1 (PD-L1) and cytotoxic T lymphocyte antigen-4 (CTLA4). We conducted a window-of-opportunity study in head and neck squamous cell carcinoma (HNSCC) to examine the contribution of anti-CTLA4 to anti-PD-L1 therapy. Single-cell profiling of on- versus pre-treatment biopsies identified T cell expansion as an early response marker. In tumors, anti-PD-L1 triggered the expansion of mostly CD8+ T cells, whereas combination therapy expanded both CD4+ and CD8+ T cells. Such CD4+ T cells exhibited an activated T helper 1 (Th1) phenotype. CD4+ and CD8+ T cells co-localized with and were surrounded by dendritic cells expressing T cell homing factors or antibody-producing plasma cells. T cell receptor tracing suggests that anti-CTLA4, but not anti-PD-L1, triggers the trafficking of CD4+ naive/central-memory T cells from tumor-draining lymph nodes (tdLNs), via blood, to the tumor wherein T cells acquire a Th1 phenotype. Thus, CD4+ T cell activation and recruitment from tdLNs are hallmarks of early response to anti-PD-L1 plus anti-CTLA4 in HNSCC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号