checkpoint blockade

检查站封锁
  • 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
    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
    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

  • 文章类型: Journal Article
    在实体瘤中,阻止“不要吃我”信号产生的强大抗肿瘤影响,源于CD47-SIRPα相互作用,受约束,特别是在造血系统恶性肿瘤中的疗效比较。激活巨噬细胞抗肿瘤活性不仅需要抑制“不要吃我”信号,而且还激活了“吃我”(吞噬细胞前)信号。有趣的是,细胞毒性T淋巴细胞相关抗原4(CTLA4)抗体(Ab)已被鉴定为刺激肿瘤微环境中Fc受体介导的活性吞噬细胞,从而产生“吃我”的信号。这项研究假设,同时靶向CD47和CTLA4可以通过同时阻断“不要吃我”信号同时触发“吃我”信号来增强抗肿瘤作用。来自该研究的实验数据证实,CD47和CTLA4的组合靶向增强了LLC细胞移植的荷瘤小鼠中针对实体瘤的免疫力。这种效果是通过减少骨髓来源的抑制细胞浸润,同时增加效应记忆CD8+T细胞的存在来实现的。NK1.1+CD8+T细胞,激活自然杀伤T细胞.同时,联合治疗也减轻了贫血。机械上,显示抗CD47Ab通过调节Foxp1上调NSCLC细胞中的CTLA4水平。此外,靶向CD47被证明通过增加CD4+T细胞的浸润促进肿瘤血管正常化。这些发现表明,CD47和CTLA4的双重靶向通过协调“吃我”和“不要吃我”信号发挥抗肿瘤作用。重塑免疫微环境,促进肿瘤血管正常化。这种联合治疗方法成为有效治疗实体瘤的有效策略。
    In solid tumors, the formidable anti-tumor impact resulting from blocking the \"don\'t eat me\" signal, arising from CD47-SIRPα interaction, is constrained, especially compared to its efficacy in hematopoietic malignancies. Activating macrophage anti-tumor activity not only necessitates the inhibition of the \"don\'t eat me\" signal, but also the activation of the \"eat me\" (pre-phagocyte) signal. Intriguingly, the cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) antibody (Ab) has been identified to stimulate Fc receptor-mediated active phagocytes in the tumor microenvironment, thereby generating \"eat me\" signals. This study postulates that concurrently targeting CD47 and CTLA4 could intensify the anti-tumor effects by simultaneously blocking the \"don\'t eat me\" signal while triggering the \"eat me\" signal. The experimental data from this investigation confirm that the combined targeting of CD47 and CTLA4 enhances immunity against solid tumors in LLC cell-transplanted tumor-bearing mice. This effect is achieved by reducing myeloid-derived suppressor cell infiltration while increasing the presence of effector memory CD8+ T cells, NK1.1+ CD8+ T cells, and activated natural killer T cells. Meanwhile, combination therapy also alleviated anemia. Mechanistically, the anti-CD47 Ab is shown to upregulate CTLA4 levels in NSCLC cells by regulating Foxp1. Furthermore, targeting CD47 is demonstrated to promote tumor vascular normalization through the heightened infiltration of CD4+ T cells. These findings suggest that the dual targeting of CD47 and CTLA4 exerts anti-tumor effects by orchestrating the \"eat me\" and \"don\'t eat me\" signals, reshaping the immune microenvironment, and fostering tumor vascular normalization. This combined therapeutic approach emerges as a potent strategy for effectively treating 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
    长链脂肪酸(LCFAs)及其激活酶的免疫调节作用,酰基辅酶A(CoA)合成酶长链家族(ACSL),肿瘤微环境在很大程度上仍然未知。这里,我们发现ACSL5具有免疫依赖性肿瘤抑制因子的功能。ACSL5表达通过调节主要组织相容性复合物I类(MHC-I)介导的抗原呈递使肿瘤体内对PD-1阻断治疗敏感,并在体外使CD8T细胞介导的细胞毒性敏感。通过筛选ACSL5的潜在底物,我们进一步确定反油酸(EA),一种长期以来被认为对人体健康有害的反式LCFA,表型以增强MHC-I表达。EA补充可以抑制肿瘤生长并使PD-1阻断疗法敏感。临床上,ACSL5表达与肺癌患者生存率改善呈正相关,血浆EA水平也是免疫治疗效率的预测指标。我们的发现为通过饮食EA补充靶向ACSL5或抗原呈递的代谢重编程来增强免疫治疗提供了基础。
    Immunomodulatory effects of long-chain fatty acids (LCFAs) and their activating enzyme, acyl-coenzyme A (CoA) synthetase long-chain family (ACSL), in the tumor microenvironment remain largely unknown. Here, we find that ACSL5 functions as an immune-dependent tumor suppressor. ACSL5 expression sensitizes tumors to PD-1 blockade therapy in vivo and the cytotoxicity mediated by CD8+ T cells in vitro via regulation of major histocompatibility complex class I (MHC-I)-mediated antigen presentation. Through screening potential substrates for ACSL5, we further identify that elaidic acid (EA), a trans LCFA that has long been considered harmful to human health, phenocopies to enhance MHC-I expression. EA supplementation can suppress tumor growth and sensitize PD-1 blockade therapy. Clinically, ACSL5 expression is positively associated with improved survival in patients with lung cancer, and plasma EA level is also predictive for immunotherapy efficiency. Our findings provide a foundation for enhancing immunotherapy through either targeting ACSL5 or metabolic reprogramming of antigen presentation via dietary EA supplementation.
    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
    光动力疗法(PDT)面临的一个主要挑战是免疫诱导的浸润性CD8+T细胞的活性受到调节性T淋巴细胞(Tregs)的影响。使肿瘤在初次消融后有复发和转移的风险。为了增强抗肿瘤反应并重新编程免疫抑制肿瘤微环境(TME),超分子光动力纳米粒子(DACss)是通过去甲基斑三素偶联β-环糊精(DMC-CD)和金刚烷胺封端的二硫键偶联FFVLGGGC肽(Ad-ss-pep-Ce6)之间的主客体相互作用来构建的。用于乳腺癌治疗的光敏剂和免疫调节剂的智能递送。DMC-CD的酸不稳定的β-羧酰胺键响应于酸性TME而水解,导致DMC的局部释放和随后的Tregs抑制。客体分子Ad-ss-pep-Ce6可以被高水平的细胞内GSH裂解,减少光敏剂毒性和增加肿瘤中光敏剂的保留。随着CTL/Treg比率的显著增加,基于Ce6的PDT和DMC介导的免疫调节的组合充分实现了TME的时空调节和重塑,以及在PD-1抗体的帮助下改善原发肿瘤和原位肺转移抑制。
    A major challenge facing photodynamic therapy (PDT) is that the activity of the immune-induced infiltrating CD8+ T cells is subject to the regulatory T lymphocytes (Tregs), leaving the tumor at risk of recurrence and metastasis after the initial ablation. To augment the antitumor response and reprogram the immunosuppressive tumor microenvironment (TME), a supramolecular photodynamic nanoparticle (DACss) is constructed by the host-guest interaction between demethylcantharidin-conjugated β-cyclodextrin (DMC-CD) and amantadine-terminated disulfide-conjugated FFVLGGGC peptide with chlorin e6 decoration (Ad-ss-pep-Ce6) to achieve intelligent delivery of photosensitizer and immunomodulator for breast cancer treatment. The acid-labile β-carboxamide bond of DMC-CD is hydrolyzed in response to the acidic TME, resulting in the localized release of DMC and subsequent inhibition of Tregs. The guest molecule Ad-ss-pep-Ce6 can be cleaved by a high level of intracellular GSH, reducing photosensitizer toxicity and increasing photosensitizer retention in the tumor. With a significant increase in the CTL/Treg ratio, the combination of Ce6-based PDT and DMC-mediated immunomodulation adequately achieved spatiotemporal regulation and remodeling of the TME, as well as improved primary tumor and in situ lung metastasis suppression with the aid of PD-1 antibody.
    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
    免疫抑制性肿瘤微环境(TME)极大地限制了免疫治疗的实际结果。因此,迫切需要制定适当的策略来重塑TME并最终诱导强烈的免疫反应。这里,我们开发了一种双功能脂质体,该脂质体负载有近红外区(NIR)光热剂IR808和Toll样受体-7(TLR7)激动剂洛沙滨前药(Lipo@IR808@Loxo),以实现NIR光触发光热治疗(PTT)和免疫佐剂的靶向递送.在近红外辐射下,Lipo@IR808@Loxo可以大大提高PTT直接杀伤肿瘤细胞和释放肿瘤相关抗原的效率,它可以与装载的洛索立滨一起缓解免疫抑制性TME,有效促进抗原呈递细胞的活化和随后的抗原呈递。这样,Lipo@IR808@Loxo可以作为原位治疗癌症疫苗,最终诱导有效的抗肿瘤T细胞反应。当进一步结合免疫检查点阻断,Lipo@IR808@Loxo介导的光热免疫治疗不仅可以消除原发肿瘤,而且可以抑制远处肿瘤的生长。从而增强了横观效应。
    The immunosuppressive tumor microenvironment (TME) greatly limits the actual outcome of immunotherapy. Therefore, it is urgent to develop appropriate strategies to reshape the TME and ultimately induce a strong immune response. Here, we developed a dual-functional liposome loaded with the photothermal agent IR808 near the infrared region (NIR) and Toll-like-receptor-7 (TLR7) agonist loxoribine prodrug (Lipo@IR808@Loxo) to achieve NIR light-triggered photothermal therapy (PTT) and the targeted delivery of immune adjuvants. Under NIR irradiation, Lipo@IR808@Loxo could greatly improve the efficiency of PTT to directly kill tumor cells and release tumor-associated antigens, which could work together with loaded loxoribine to relieve the immunosuppressive TME, effectively promoting the activation of antigen-presenting cells and subsequent antigen presentation. In this way, Lipo@IR808@Loxo could act as an in situ therapeutic cancer vaccine, eventually inducing a potent antitumor T-cell response. When further combined with immune checkpoint blockade, Lipo@IR808@Loxo-mediated photothermal immunotherapy could not only eliminate the primary tumors but also inhibit the growth of distant tumors, thus enhancing the abscopal effect.
    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
    检查点阻断抗体的使用仍然限制在几种恶性肿瘤中,由于疗效适中,尽管在抗肿瘤免疫疗法方面取得了相当大的成功。癌细胞对免疫破坏的不良反应是检查点治疗失败的重要原因。我们假设将检查点疗法与天然产物化学增敏剂相结合可以增强免疫反应。在这里,靶向二萜衍生物与检查点阻断(抗CTLA-4)整合,以改善使用热敏脂质体作为载体的免疫治疗.在体内,脂质体能够将两种药物有效载荷共同递送到肿瘤中。因此,调节性T细胞增殖受到抑制,细胞毒性T细胞浸润增强,并取得了深远的免疫治疗效果。此外,另一种临床使用的检查点抗体的免疫治疗效果,抗PD-1,也受益于二萜衍生物。值得注意的是,我们的机制研究表明,靶向二萜衍生物通过THBS1下调和由此导致的THBS1-CD47相互作用的破坏,增加了癌细胞对免疫攻击的敏感性.总的来说,共同提供THBS1抑制剂和检查点阻断有望促进癌症免疫疗法。我们首次发现THBS1抑制可以加强检查点治疗。
    The use of checkpoint-blockade antibodies is still restricted in several malignancies due to the modest efficacy, despite considerable success in anti-tumor immunotherapy. The poor response of cancer cells to immune destruction is an essential contributor to the failure of checkpoint therapy. We hypothesized that combining checkpoint therapy with natural-product chemosensitizer could enhance immune response. Herein, a targeted diterpenoid derivative was integrated with the checkpoint blockade (anti-CTLA-4) to improve immunotherapy using thermosensitive liposomes as carriers. In vivo, the liposomes enabled the co-delivery of the two drug payloads into the tumor. Consequently, the regulatory T cell proliferation was restrained, the cytotoxic T cell infiltration was enhanced, and the profound immunotherapeutic effect was achieved. In addition, the immunotherapeutic effect of another clinically used checkpoint antibody, anti-PD-1, also benefited from the diterpenoid derivative. Of note, our mechanism study revealed that the targeted diterpenoid derivative increased the sensitivity of cancer cells to immune attack via THBS1 downregulation and the resultant destruction of THBS1-CD47 interaction. Collectively, co-delivering THBS1 inhibitor and checkpoint blockade is promising to boost cancer immunotherapy. We first time discovered that THBS1 suppression could strengthen checkpoint therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    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
    胶质母细胞瘤(GBM)的代谢重编程由于其对免疫抑制微环境的影响而对有效的免疫治疗构成了巨大障碍。这里,我们开发了专门为GBM免疫疗法设计的氢键有机框架(HOF),利用中枢神经系统(CNS)中相对孤立的胆固醇代谢微环境。基于HOF的生物调节剂调节细胞外胆固醇代谢,有效阻断程序性细胞死亡蛋白1/程序性死亡配体1(PD-1/PD-L1)通路并降低2B4表达。这种代谢破坏GBM的免疫抑制微环境并使CD8+T细胞恢复活力。此外,胆固醇代谢调节在治疗GBM侵袭方面提供了额外的益处。此外,响应于肿瘤微环境(TEM)启动的化学激发光动力疗法(PDT),在调节胆固醇代谢的过程中,该生物调节剂可有效触发免疫原性细胞死亡(ICD)并增加GBM中细胞毒性T淋巴细胞(CTL)的浸润。通过逆转免疫抑制微环境和增强化学激发的PDT,我们的方法激发了GBM的有效抗体非依赖性免疫治疗。本研究为通过调节胆固醇代谢增强免疫治疗提供了模型,并探讨了GBM治疗中“代谢检查点”策略的可行性。本文受版权保护。保留所有权利。
    The metabolic reprogramming of glioblastoma (GBM) poses a tremendous obstacle to effective immunotherapy due to its impact on the immunosuppressive microenvironment. In this work, a hydrogen-bonded organic framework (HOF) specifically designed for GBM immunotherapy is developed, taking advantage of the relatively isolated cholesterol metabolism microenvironment in the central nervous system (CNS). The HOF-based biotuner regulates extra/intracellular cholesterol metabolism, effectively blocking the programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) pathway and reducing 2B4 expression. This metabolically disrupts the immunosuppressive microenvironment of GBM and rejuvenates CD8+ T cells. Moreover, cholesterol metabolism regulation offers additional benefits in treating GBM invasion. Furthermore, tumor microenvironment (TME)-initiated chemiexcited photodynamic therapy (PDT) is enhanced during the regulation of cholesterol metabolism, and the biotuner can effectively trigger immunogenic cell death (ICD) and increase the infiltration of cytotoxic T lymphocytes (CTLs) in GBM. By reversing the immunosuppressive microenvironment and bolstering chemiexcited-PDT, this approach invigorates efficient antibody non-dependent immunotherapy for GBM. This study provides a model for enhancing immunotherapy through cholesterol metabolism regulation and explores the feasibility of a \"metabolic checkpoint\" strategy in GBM treatment.
    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