Immunotherapy resistance

免疫治疗耐药
  • 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
    AXL受体表达被提议在非小细胞肺癌(NSCLC)患者中赋予免疫检查点抑制剂(ICI)抗性。我们试图结合突变和肿瘤微环境特征询问AXL表达,以揭示ICI治疗的NSCLC患者耐药的预测机制。
    通过免疫组织化学分析来自用ICI单一疗法治疗的111例NSCLC患者的肿瘤样品的肿瘤-和免疫-AXL表达。通过全外显子组测序(n=44)和成像质量细胞计数(n=14)分析患者的亚组。结果与ICI结果测量相关。
    肿瘤细胞AXL表达与侵袭性表型特征相关,包括化疗进展后ICIs治疗的患者OS降低(P=0.04),但与改善ICI治疗的疾病控制(P=0.045)相反,PD-L1高一线患者。AXL+免疫细胞浸润与总免疫细胞浸润相关,并改善总体预后(PFS:P=0.044,OS:P=0.054)。肿瘤细胞AXL上调显示与PD-L1上调和ICI反应相关的突变如MUC4和ZNF469的富集,以及与免疫抑制肿瘤表型和较差ICI预后相关的不利突变,包括CSMD1和LRP1B,特别是在化疗治疗的患者中。肿瘤突变负荷对ICI结果没有影响,并且与缺乏肿瘤浸润性免疫细胞有关。空间免疫表型提供了证据,表明肿瘤细胞AXL上调和不良突变可调节肿瘤微环境,有利于浸润,激活的中性粒细胞超过抗肿瘤免疫亚群,包括CD4和CD8T细胞。
    肿瘤细胞AXL上调与不同的基因型和定义化疗诱导ICI耐药机制的微环境免疫谱相关,这表明AXL抑制剂与目前的化学免疫治疗方案联合使用可以使NSCLC患者受益.
    UNASSIGNED: AXL receptor expression is proposed to confer immune-checkpoint inhibitor (ICI)-resistance in non-small cell lung cancer (NSCLC) patients. We sought to interrogate AXL expression in conjunction with mutational and tumor-microenvironmental features to uncover predictive mechanisms of resistance in ICI-treated NSCLC patients.
    UNASSIGNED: Tumor samples from 111 NSCLC patients treated with ICI-monotherapy were analyzed by immunohistochemistry for tumor- and immune-AXL expression. Subsets of patients were analyzed by whole-exome sequencing (n = 44) and imaging mass cytometry (n = 14). Results were related to ICI-outcome measurements.
    UNASSIGNED: Tumor-cell AXL expression correlated with aggressive phenotypic features including reduced OS in patients treated with ICIs (P = 0.04) after chemotherapy progression, but conversely associated with improved disease control (P = 0.045) in ICI-treated, PD-L1 high first-line patients. AXL+ immune-cell infiltration correlated with total immune-cell infiltration and improved overall outcomes (PFS: P = 0.044, OS: P = 0.054). Tumor-cell AXL-upregulation showed enrichment in mutations associated with PD-L1-upregulation and ICI-response such as MUC4 and ZNF469, as well as adverse mutations including CSMD1 and LRP1B which associated with an immune-suppressed tumor phenotype and poor ICI prognosis particularly within chemotherapy-treated patients. Tumor mutational burden had no effect on ICI-outcomes and was associated with a lack of tumor-infiltrating immune cells. Spatial-immunophenotyping provided evidence that tumor-cell AXL-upregulation and adverse mutations modulate the tumor microenvironment in favor of infiltrating, activated neutrophils over anti-tumor immune-subsets including CD4 and CD8 T-cells.
    UNASSIGNED: Tumor-cell AXL-upregulation correlated with distinct oncotypes and microenvironmental immune-profiles that define chemotherapy-induced mechanisms of ICI-resistance, which suggests the combination of AXL inhibitors with current chemoimmunotherapy regimens can benefit NSCLC patients.
    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
    背景:黑色素瘤是一种异质性癌症,受黑色素瘤细胞可塑性及其对微环境线索的动态适应的影响。黑色素瘤细胞在明确定义的转录细胞状态之间过渡,影响治疗反应和抗性。
    方法:在本研究中,我们应用单细胞RNA测序技术,对BRAF/MEK抑制剂离体治疗后未经免疫治疗和免疫治疗耐药的黑色素瘤肿瘤的分子特征进行了研究.
    结果:我们确认存在四种不同的黑素瘤细胞状态-黑素细胞,暂时性的,神经脊状和未分化,并确定免疫疗法抗性肿瘤中神经crest样和未分化黑色素瘤细胞的富集。此外,我们引入了一种整合的计算方法来识别转录细胞状态中应答和非应答黑色素瘤细胞的亚群.
    结论:在所有转录细胞状态下都鉴定出无反应的黑素瘤细胞,并且由于促炎性IL6和TNFα信号传导而倾向于对BRAF/MEK抑制剂耐药。我们的研究提供了一个框架来研究不同黑素瘤细胞状态下的治疗反应,并表明肿瘤固有的促炎信号有助于BRAF/MEK抑制剂抵抗。
    背景:这项工作得到了麦格理大学的支持,澳大利亚黑色素瘤研究所,和澳大利亚国家卫生和医学研究委员会(NHMRC;赠款2012860,2028055)。
    BACKGROUND: Melanoma is a heterogeneous cancer influenced by the plasticity of melanoma cells and their dynamic adaptations to microenvironmental cues. Melanoma cells transition between well-defined transcriptional cell states that impact treatment response and resistance.
    METHODS: In this study, we applied single-cell RNA sequencing to interrogate the molecular features of immunotherapy-naive and immunotherapy-resistant melanoma tumours in response to ex vivo BRAF/MEK inhibitor treatment.
    RESULTS: We confirm the presence of four distinct melanoma cell states - melanocytic, transitory, neural-crest like and undifferentiated, and identify enrichment of neural crest-like and undifferentiated melanoma cells in immunotherapy-resistant tumours. Furthermore, we introduce an integrated computational approach to identify subsets of responding and nonresponding melanoma cells within the transcriptional cell states.
    CONCLUSIONS: Nonresponding melanoma cells are identified in all transcriptional cell states and are predisposed to BRAF/MEK inhibitor resistance due to pro-inflammatory IL6 and TNFɑ signalling. Our study provides a framework to study treatment response within distinct melanoma cell states and indicate that tumour-intrinsic pro-inflammatory signalling contributes to BRAF/MEK inhibitor resistance.
    BACKGROUND: This work was supported by Macquarie University, Melanoma Institute Australia, and the National Health and Medical Research Council of Australia (NHMRC; grant 2012860, 2028055).
    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
    肺癌仍然是全球癌症相关死亡的最高百分比。非小细胞肺癌治疗如免疫检查点抑制剂的进展显著改善了生存率和长期疾病反应。即使在治疗和围手术期设置。不幸的是,耐药性发展为对治疗的初始反应,或更常见的发展为初始反应后的进展。已经利用了几种方式来解决这一问题。这篇综述将集中在各种联合治疗与免疫检查点抑制剂,包括增加化疗,各种免疫疗法,辐射,抗体-药物缀合物,双特异性抗体,新抗原疫苗,和肿瘤浸润淋巴细胞.我们讨论了这些药物与免疫检查点抑制剂联合使用时的状态,重点是肺癌。早期毒性信号,耐受性,并对实施的可行性进行了审查。最后,我们讨论了治疗的后续步骤。
    Lung cancer continues to contribute to the highest percentage of cancer-related deaths worldwide. Advancements in the treatment of non-small cell lung cancer like immune checkpoint inhibitors have dramatically improved survival and long-term disease response, even in curative and perioperative settings. Unfortunately, resistance develops either as an initial response to treatment or more commonly as a progression after the initial response. Several modalities have been utilized to combat this. This review will focus on the various combination treatments with immune checkpoint inhibitors including the addition of chemotherapy, various immunotherapies, radiation, antibody-drug conjugates, bispecific antibodies, neoantigen vaccines, and tumor-infiltrating lymphocytes. We discuss the status of these agents when used in combination with immune checkpoint inhibitors with an emphasis on lung cancer. The early toxicity signals, tolerability, and feasibility of implementation are also reviewed. We conclude with a discussion of the next steps in 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
    虽然现在乳腺癌的治疗有了显著的进步,然而转移性,尤其是三阴性乳腺癌(TNBC),低生存率仍然具有挑战性。癌症免疫疗法,HER2阳性和TNBC的一种有希望的方法,仍然面临阻力障碍。最近,许多研究都将目光投向了乳腺癌免疫治疗的耐药性。我们的研究提供了对当前研究环境的透彻理解,热点,通过细致的文献计量分析,在这一关键领域取得了新的突破。截至2024年3月26日,共收集了1341篇关于乳腺癌免疫学抗性的文章,来自WebofScienceCoreCollection。包括765篇文章和576条评论。Bibliometrix,CiteSpace和VOSviewer软件每年用于检查出版物和引文,多产的国家,有贡献的机构,高水平的期刊和学者,以及被高度引用的文章,引用和关键字。在过去的七年中,乳腺癌免疫治疗耐药性的研究出现了惊人的增长。美国和中国作出了重大贡献,哈佛医学院是最多产的机构,并积极参与合作。最有贡献的作者是库里利亚诺,G来自意大利欧洲肿瘤研究所,而Wucherpfennig,K.W.来自美国达纳-法伯癌症研究所,引用次数最高。高产期刊主要集中在临床上,免疫学和肿瘤学研究。常用关键词包括\"阻力\",\"表达式\",“肿瘤微环境”,\"癌症\",\"T细胞\",“治疗”,“化疗”和“细胞”。目前的研究试图通过整合生物信息学来揭示乳腺癌的免疫抵抗机制,基础实验,和临床试验。正在努力制定提高免疫疗法有效性的策略,包括联合疗法的探索和药物输送系统的进步。此外,人们非常关注鉴定能够预测患者对免疫学反应的新型生物标志物.这项研究将为研究人员提供有关乳腺癌免疫学耐药性的现有知识的最新概述,作为明智决策和进一步研究解决免疫疗法耐药性的创新方法的宝贵资源。
    While breast cancer treatments have advanced significantly nowadays, yet metastatic, especially triple-negative breast cancer (TNBC), remains challenging with low survival. Cancer immunotherapy, a promising approach for HER2-positive and TNBC, still faces resistance hurdles. Recently, numerous studies have set their sights on the resistance of immunotherapy for breast cancer. Our study provides a thorough comprehension of the current research landscape, hotspots, and emerging breakthroughs in this critical area through a meticulous bibliometric analysis. As of March 26, 2024, a total of 1341 articles on immunology resistance in breast cancer have been gathered from Web of Science Core Collection, including 765 articles and 576 reviews. Bibliometrix, CiteSpace and VOSviewer software were utilized to examine publications and citations per year, prolific countries, contributive institutions, high-level journals and scholars, as well as highly cited articles, references and keywords. The research of immunotherapy resistance in breast cancer has witnessed a remarkable surge over the past seven years. The United States and China have made significant contributions, with Harvard Medical School being the most prolific institution and actively engaging in collaborations. The most contributive author is Curigliano, G from the European Institute of Oncology in Italy, while Wucherpfennig, K. W. from the Dana-Farber Cancer Institute in the USA, had the highest citations. Journals highly productive primarily focus on clinical, immunology and oncology research. Common keywords include \"resistance\", \"expression\", \"tumor microenvironment\", \"cancer\", \"T cell\", \"therapy\", \"chemotherapy\" and \"cell\". Current research endeavors to unravel the mechanisms of immune resistance in breast cancer through the integration of bioinformatics, basic experiments, and clinical trials. Efforts are underway to develop strategies that improve the effectiveness of immunotherapy, including the exploration of combination therapies and advancements in drug delivery systems. Additionally, there is a strong focus on identifying novel biomarkers that can predict patient response to immunology. This study will provide researchers with an up-to-date overview of the present knowledge in drug resistance of immunology for breast cancer, serving as a valuable resource for informed decision-making and further research on innovative approaches to address immunotherapy 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
    尽管抗程序性细胞死亡-1(抗PD-1)免疫疗法取得了成功,许多癌症患者仍然没有反应,缺乏可靠的预测生物标志物。这里,我们表明嘧啶能受体P2RY6的异常表达在人类癌症中很常见,并导致免疫逃避。在小鼠同基因和人类异种移植肿瘤模型中,P2RY6的异位表达形成免疫抑制性肿瘤微环境(TME),以增强肿瘤生长和对免疫治疗的抵抗力,而P2RY6从高P2RY6表达的肿瘤中缺失会使TME发炎以抑制肿瘤生长。作为G蛋白偶联受体,P2RY6激活Gq/磷脂酶C-β信号并刺激前列腺素E2的合成,前列腺素E2是TME中免疫抑制的关键介质。与P2RY6在肿瘤中的重要作用相反,从小鼠中全局缺失P2ry6不会损害生存能力。因此,我们的研究将P2RY6作为肿瘤固有P2RY6高表达患者的精确免疫疗法靶标。
    Despite the success of anti-programmed cell death-1 (anti-PD-1) immunotherapy, many cancer patients remain unresponsive, and reliable predictive biomarkers are lacking. Here, we show that aberrant expression of the pyrimidinergic receptor P2RY6 is frequent in human cancers and causes immune evasion. In mouse syngeneic and human xenograft tumor models, ectopic expression of P2RY6 shapes an immunosuppressive tumor microenvironment (TME) to enhance tumor growth and resistance to immunotherapy, whereas deletion of P2RY6 from tumors with high P2RY6 expression inflames the TME to inhibit tumor growth. As a G protein-coupled receptor, P2RY6 activates Gq/phospholipase C-β signaling and stimulates the synthesis of prostaglandin E2, which is a key mediator of immunosuppression in the TME. In contrast to the essential role of P2RY6 in tumors, global deletion of P2ry6 from mice does not compromise viability. Our study thus nominates P2RY6 as a precision immunotherapy target for patients with high tumor-intrinsic P2RY6 expression.
    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
    微卫星不稳定性高(MSI-H)结直肠癌(CRC)对免疫检查点抑制剂(ICI)的治疗反应确实令人惊讶;然而,获得性耐药性的出现对这些患者的生存构成了更大的威胁。在这里,MSI-HCRC样本的生物信息学分析显示,Wnt信号通路是获得性免疫再激活的一个有希望的目标,而随后的分析和生化测试证实了Wnt过度活跃的CRC细胞倾向于与人血清白蛋白(HSA)一起进行大胞吞作用。这些发现激励我们开发一种工程化的HSA,其不仅具有特异性靶向癌细胞的能力,而且还有效地抑制这些恶性细胞内的Wnt/β-连环蛋白级联。为了实现这个目标,对已报道的Wnt小分子抑制剂进行了全面筛选,以评估其与HSA的亲和力,并且发现鼠尾草酸(CA)表现出最高的亲和力,同时揭示多个结合位点。进一步的调查显示,CAHSA能够将HSA设计成球形和尺寸可调的纳米结构,称为eHSA(工程HSA颗粒)。这证明了优化的巨细胞增多依赖性细胞内化。如预期,eHSA有效抑制Wnt信号通路并在体内重新激活获得性免疫应答。此外,在MSI-HCRC的皮下和原位小鼠同源移植模型中,eHSA成功恢复了对抗PD1抗癌作用的敏感性,以及人源化hu-PBMC患者来源的MSI-HCRC原位异种移植(PDOX)小鼠模型,同时保持良好的安全性。这种临床上可行的免疫再激活策略的集体实施不仅能够提供用于CRC治疗的Wnt抑制剂。但也可以作为精确医学指导的纳米药物开发的示例性演示,该药物开发可以有效地利用病理状态下的特定细胞适应症。
    The therapeutic response of microsatellite instability-high (MSI-H) colorectal cancer (CRC) to immune checkpoint inhibitors (ICI) is indeed surprising; however, the emergence of acquired resistance poses an even greater threat to the survival of these patients. Herein, bioinformatics analysis of MSI-H CRC samples revealed that Wnt signaling pathway represents a promising target for acquired immune reactivation, while subsequent analysis and biochemical testing substantiated the inclination of Wnt-hyperactive CRC cells to engage in macropinocytosis with human serum albumin (HSA). These findings have inspired us to develop an engineered HSA that not only possesses the ability to specifically target cancer cells but also effectively suppresses the Wnt/β-catenin cascade within these malignant cells. In pursuit of this objective, a comprehensive screening of reported Wnt small-molecule inhibitors was conducted to evaluate their affinity with HSA, and it was discovered that Carnosic acid (CA) exhibited the highest affinity while simultaneously revealing multiple binding sites. Further investigation revealed that CA HSA the capability to engineer HSA into spherical and size-tunable nanostructures known as eHSA (Engineering HSA particle), which demonstrated optimized macropinocytosis-dependent cellular internalization. As anticipated, eHSA effectively suppressed the Wnt signaling pathway and reactivated the acquired immune response in vivo. Furthermore, eHSA successfully restored sensitivity to Anti-PD1\'s anticancer effects in both subcutaneous and orthotopic mouse homograft models of MSI-H CRC, as well as a humanized hu-PBMC patient-derived orthotopic xenograft (PDOX) mouse model of MSI-H CRC, all while maintaining a favorable safety profile. The collective implementation of this clinically viable immune reactivation strategy not only enables the delivery of Wnt inhibitors for CRC therapy, but also serves as an exemplary demonstration of precision-medicine-guided nanopharmaceutical development that effectively harnesses specific cellular indications in pathological states.
    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
    抗程序性死亡1/程序性死亡配体1(抗PD-1/PD-L1)抗体通过克服肿瘤细胞免疫逃避和逆转T细胞耗竭而发挥显著的抗肿瘤作用。然而,耐药性的出现导致大多数患者对这些免疫检查点抑制剂(ICIs)的反应较差.研究表明,T细胞浸润不足,缺乏PD-1表达,缺乏干扰素信号,肿瘤抗原呈递的丧失,脂质代谢异常均被认为与免疫治疗耐药密切相关。为了解决肿瘤免疫治疗中的耐药性,许多研究集中在开发联合治疗策略上。目前,抗PD-1/PD-L1抗体等ICI联合化疗和靶向治疗已被批准用于临床治疗。在这次审查中,我们从肿瘤微环境的角度分析抗PD-1/PD-L1治疗的耐药机制,肠道菌群,表观遗传调控,和共抑制性免疫检查点受体。我们还讨论了各种有前途的联合治疗策略,以解决抗PD-1/PD-L1药物的耐药性,包括将这些疗法与传统中药相结合,非编码RNA,靶向治疗,其他ICIs,和个性化的癌症疫苗。此外,我们关注预测抗PD-1/PD-L1治疗耐药以及联合治疗疗效的生物标志物.最后,我们提出了通过使用生物标志物系统的个性化组合策略进一步扩大免疫治疗应用的方法.
    Anti-programmed death 1/programmed death ligand 1 (anti-PD-1/PD-L1) antibodies exert significant antitumor effects by overcoming tumor cell immune evasion and reversing T-cell exhaustion. However, the emergence of drug resistance causes most patients to respond poorly to these immune checkpoint inhibitors (ICIs). Studies have shown that insufficient T-cell infiltration, lack of PD-1 expression, deficient interferon signaling, loss of tumor antigen presentation, and abnormal lipid metabolism are all considered to be closely associated with immunotherapy resistance. To address drug resistance in tumor immunotherapy, a lot of research has concentrated on developing combination therapy strategies. Currently, ICIs such as anti-PD-1 /PD-L1 antibody combined with chemotherapy and targeted therapy have been approved for clinical treatment. In this review, we analyze the mechanisms of resistance to anti-PD-1/PD-L1 therapy in terms of the tumor microenvironment, gut microbiota, epigenetic regulation, and co-inhibitory immune checkpoint receptors. We also discuss various promising combination therapeutic strategies to address resistance to anti-PD-1/PD-L1 drugs, including combining these therapies with traditional Chinese medicine, non-coding RNAs, targeted therapy, other ICIs, and personalized cancer vaccines. Moreover, we focus on biomarkers that predict resistance to anti-PD-1/PD-L1 therapy as well as combination therapy efficacy. Finally, we suggest ways to further expand the application of immunotherapy through personalized combination strategies using biomarker systems.
    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
    免疫疗法彻底改变了某些癌症的治疗方法,但是在克服免疫疗法耐药性方面仍然存在挑战。研究表明,肿瘤微环境的代谢调节可以增强抗肿瘤免疫力。这里,我们讨论了代谢修饰剂与免疫疗法联合应用的近期临床前和临床证据.虽然这种组合很有希望,必须解决几个关键领域,其中包括确定代谢调节剂对免疫细胞代谢的影响,治疗功效的推定生物标志物,修饰剂对具有代谢异质性的肿瘤的功效,以及由于肿瘤对替代代谢途径的依赖而导致的耐药性的潜在发展。我们提出了这些问题的解决方案,并认为评估这些参数对于考虑代谢调节剂在使肿瘤对免疫疗法敏感方面的潜力至关重要。
    Immunotherapies have revolutionized the treatment of certain cancers, but challenges remain in overcoming immunotherapy resistance. Research shows that metabolic modulation of the tumor microenvironment can enhance antitumor immunity. Here, we discuss recent preclinical and clinical evidence for the efficacy of combining metabolic modifiers with immunotherapies. While this combination holds great promise, a few key areas must be addressed, which include identifying the effects of metabolic modifiers on immune cell metabolism, the putative biomarkers of therapeutic efficacy, the efficacy of modifiers on tumors harboring metabolic heterogeneity, and the potential development of resistance due to tumor reliance on alternative metabolic pathways. We propose solutions to these problems and posit that assessing these parameters is crucial for considering the potential of metabolic modifiers in sensitizing tumors to immunotherapies.
    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
    通过以疫苗为基础的癌症免疫疗法,已经开始在实际临床应用中取得突破,它使用人体的免疫系统,体液和细胞,攻击恶性细胞和对抗疾病。然而,传统的疫苗方法仍然面临着引发有效的抗原特异性免疫反应的多重挑战,导致免疫疗法耐药。近年来,通过结合各种生物实体的天然结构,仿生纳米疫苗已成为传统疫苗方法的有希望的替代品,如细胞,病毒,和细菌。仿生纳米疫苗提供靶向抗原呈递细胞(APC)递送的好处,改进的抗原/佐剂负载,和生物相容性,从而提高免疫治疗的敏感性。本文综述了几种仿生纳米疫苗在抗肿瘤免疫反应中的作用,包括细胞膜包覆的纳米疫苗,基于蛋白质的自组装纳米疫苗,基于细胞外囊泡的纳米疫苗,天然配体修饰的纳米疫苗,基于人工抗原呈递细胞的纳米疫苗和基于脂质体的纳米疫苗。我们还讨论了与新兴的仿生纳米疫苗平台的临床翻译相关的观点和挑战,这些平台用于使癌细胞对免疫疗法敏感。
    Breakthroughs in actual clinical applications have begun through vaccine-based cancer immunotherapy, which uses the body\'s immune system, both humoral and cellular, to attack malignant cells and fight diseases. However, conventional vaccine approaches still face multiple challenges eliciting effective antigen-specific immune responses, resulting in immunotherapy resistance. In recent years, biomimetic nanovaccines have emerged as a promising alternative to conventional vaccine approaches by incorporating the natural structure of various biological entities, such as cells, viruses, and bacteria. Biomimetic nanovaccines offer the benefit of targeted antigen-presenting cell (APC) delivery, improved antigen/adjuvant loading, and biocompatibility, thereby improving the sensitivity of immunotherapy. This review presents a comprehensive overview of several kinds of biomimetic nanovaccines in anticancer immune response, including cell membrane-coated nanovaccines, self-assembling protein-based nanovaccines, extracellular vesicle-based nanovaccines, natural ligand-modified nanovaccines, artificial antigen-presenting cells-based nanovaccines and liposome-based nanovaccines. We also discuss the perspectives and challenges associated with the clinical translation of emerging biomimetic nanovaccine platforms for sensitizing cancer cells to immunotherapy.
    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