neoantigens

新抗原
  • 文章类型: Journal Article
    宿主免疫遗传学(人类白细胞抗原,HLA)在人类对黑色素瘤的免疫反应中发挥关键作用,影响黑色素瘤患病率和免疫治疗结果。有益的结果取决于黑素瘤抗原的表位与HLAI类分子的成功结合,以有效地参与细胞毒性CD8淋巴细胞并随后消除癌细胞。这项研究评估了HLAI类对黑色素瘤肿瘤抗原的结合亲和力和免疫原性,以鉴定最适合于消除黑色素瘤抗原的等位基因。
    在这项研究中,我们使用免费提供的软件工具来确定2462个报告的HLAI类等位基因与黑色素瘤肿瘤中表达的11种已知抗原的所有线性非聚体表位的结合亲和力和免疫原性(TRP2,S100,酪氨酸酶,TRP1,PMEL(17),MAGE1,MAGE4,CTA,BAGE,GAGE/SSX2,Melan)。
    我们鉴定了以下9种针对所有11种黑色素瘤抗原具有非常高的免疫原性和结合亲和力的HLAI类等位基因:A*02:14,B*07:10,B*35:10,B*40:10,B*40:12,B*44:10,C*07:11和C*07:13和C*07:14。
    这9个HLA等位基因具有通过自身和通过增强免疫检查点抑制剂的有益作用来帮助消除黑色素瘤的潜力。
    UNASSIGNED: Host immunogenetics (Human Leukocyte Antigen, HLA) play a critical role in the human immune response to melanoma, influencing both melanoma prevalence and immunotherapy outcomes. Beneficial outcomes hinge on the successful binding of epitopes of melanoma antigens to HLA Class I molecules for an effective engagement of cytotoxic CD8+ lymphocytes and subsequent elimination of the cancerous cell. This study evaluated the binding affinity and immunogenicity of HLA Class I to melanoma tumor antigens to identify alleles best suited to facilitate elimination of melanoma antigens.
    UNASSIGNED: In this study, we used freely available software tools to determine in silico the binding affinity and immunogenicity of 2462 reported HLA Class I alleles to all linear nonamer epitopes of 11 known antigens expressed in melanoma tumors (TRP2, S100, Tyrosinase, TRP1, PMEL(17), MAGE1, MAGE4, CTA, BAGE, GAGE/SSX2, Melan).
    UNASSIGNED: We identified the following 9 HLA Class I alleles with very high immunogenicity and binding affinity against all 11 melanoma antigens: A*02:14, B*07:10, B*35:10, B*40:10, B*40:12, B*44:10, C*07:11, and C*07:13, and C*07:14.
    UNASSIGNED: These 9 HLA alleles possess the potential to aid in the elimination of melanoma both by themselves and by enhancing the beneficial effect of immune checkpoint inhibitors.
    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
    结直肠癌构成了巨大的全球健康负担。关于世卫组织,到2040年,结直肠癌的全球负担将是约320万新病例。同时,它表明这种癌症每年将导致600万人死亡。尽管化疗和单克隆抗体治疗取得了进展,由于癌症干细胞的耐药性,该疾病仍然是一个重大挑战。本研究致力于设计一种靶向TLR4/MD2复合物的多表位肽(9聚体表位)新抗原疫苗作为潜在的疫苗候选物。这些肿瘤特异性新抗原(TSA)被认为是可用于癌症疫苗开发的新型抗原。为了开发新抗原候选疫苗,我们使用了SPENCER数据库,并检索了140个lncRNA来源的表位。从140个表位,我们为疫苗构建体选择了7种具有高抗原特性的新抗原.一种含有表位的新型疫苗,接头(EAAAK和CPCPG),利用免疫信息学工具配制佐剂(核糖体[50S]蛋白L7L12)。对疫苗的生物物理特性进行了评估,显示其抗原性(0.6469),稳定性(不稳定性指数:37.05),和免疫系统相互作用的潜力。深入的结构分析,分子对接研究,支持ML的免疫模拟分析强调了疫苗的结构完整性,与TLR4的结合亲和力,以及引发针对结直肠癌抗原的强大免疫应答的能力。这些发现表明,多表位疫苗有望成为对抗结直肠癌的下一代方法。我们的计算机模拟研究显示了候选疫苗的潜力;然而,进一步的体内和体外研究对于验证免疫原性至关重要,安全,临床实施前的疗效。我们的研究首次开发了lncRNA衍生的基于新抗原的癌症疫苗。
    Colorectal cancer poses a substantial global health burden. Regarding WHO, the global burden of colorectal cancer will be about 3.2 million new cases by the year 2040. Simultaneously, it indicated that this cancer will cause 6 million deaths per year. Despite advancements in chemotherapy and monoclonal antibody therapy, the disease remains a significant challenge due to the resistance of cancer stem cells. This study endeavors to design a multi-epitopic peptide (9-mer epitopes) neoantigen-based vaccine targeting the TLR4/MD2 complex as a potential vaccine candidate. These tumor-specific neoantigens (TSA) are considered novel antigens that can be used for vaccine development against cancer. To develop the neoantigen vaccine candidate, we used the SPENCER database, and 140 lncRNA-derived epitopes were retrieved. From 140 epitopes, we selected seven neoantigens with high antigenic properties for the vaccine construct. A novel vaccine containing epitopes, linkers (EAAAK and CPCPG), and adjuvants (ribosomal [50S] protein L7L12) was formulated utilizing immunoinformatics tools. The vaccine\'s biophysical properties were evaluated, revealing its antigenicity (0.6469), stability (instability index: 37.05), and potential for immune system interaction. In-depth structural analyses, molecular docking studies, and ML-enabled immune simulation profiling underscored the vaccine\'s structural integrity, binding affinity with TLR4, and ability to elicit robust immune responses against colorectal cancer antigens. These findings suggest that the multi-epitopic vaccine holds promise as a next-generation approach to combat colorectal cancer. Our in silico studies exhibit potentiality of the vaccine candidate; however, further in vivo and in vitro investigations are crucial to validate immunogenicity, safety, and efficacy before clinical implementation. Our study developed a first-time lncRNA-derived neoantigen-based cancer vaccine.
    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
    个性化新抗原疗法在特定患者群体中显示出长期和稳定的疗效。然而,并非所有患者都有足够水平的新抗原用于治疗。尽管体细胞突变常见于肿瘤中,这些突变的很大一部分不会触发免疫反应。具有低突变负担的患者继续表现出对这种治疗的无反应性。我们通过利用高度免疫原性的非天然氨基酸对硝基苯丙氨酸(pNO2Phe)对未能产生新表位的体细胞突变进行序列改变,提出了新抗原疫苗的设计范例。这增强了突变的免疫原性并将其转化为免疫疗法的合适候选者。根据该范例设计的硝化改变的表位疫苗能够激活循环CD8T细胞并诱导针对不同MHC背景中的自体突变表位的免疫交叉反应性(H-2Kb,H-2Kd,和人类HLA-A02:01),导致携带突变的肿瘤细胞的消除。用改变的表位免疫后,肿瘤生长被显著抑制。值得注意的是,硝化表位诱导肿瘤浸润巨噬细胞分化为M1表型,令人惊讶地增强巨噬细胞的MHCII分子呈递途径。硝化表位处理的巨噬细胞具有交叉激活CD4+和CD8+T细胞的潜力,这可以解释为什么pNO2Phe可以增强表位的免疫原性。同时,肿瘤的免疫抑制微环境由于巨噬细胞的激活而改变。硝化新抗原疫苗策略使得能够设计针对非免疫原性肿瘤突变的疫苗。扩大用于个性化和共享新型抗原治疗的潜在肽库。这种方法为以前不适合新抗原疫苗治疗的患者提供了治疗机会。
    Personalized neoantigen therapy has shown long-term and stable efficacy in specific patient populations. However, not all patients have sufficient levels of neoantigens for treatment. Although somatic mutations are commonly found in tumours, a significant portion of these mutations do not trigger an immune response. Patients with low mutation burdens continue to exhibit unresponsiveness to this treatment. We propose a design paradigm for neoantigen vaccines by utilizing the highly immunogenic unnatural amino acid p-nitrophenylalanine (pNO2Phe) for sequence alteration of somatic mutations that failed to generate neoepitopes. This enhances the immunogenicity of the mutations and transforms it into a suitable candidate for immunotherapy. The nitrated altered epitope vaccines designed according to this paradigm is capable of activating circulating CD8+ T cells and inducing immune cross-reactivity against autologous mutated epitopes in different MHC backgrounds (H-2Kb, H-2Kd, and human HLA-A02:01), leading to the elimination of tumour cells carrying the mutation. After immunization with the altered epitopes, tumour growth was significantly inhibited. It is noteworthy that nitrated epitopes induce tumour-infiltrating macrophages to differentiate into the M1 phenotype, surprisingly enhancing the MHC II molecule presenting pathway of macrophages. Nitrated epitope-treated macrophages have the potential to cross-activate CD4+ and CD8+ T cells, which may explain why pNO2Phe can enhance the immunogenicity of epitopes. Meanwhile, the immunosuppressive microenvironment of the tumour is altered due to the activation of macrophages. The nitrated neoantigen vaccine strategy enables the design of vaccines targeting non-immunogenic tumour mutations, expanding the pool of potential peptides for personalized and shared novel antigen therapy. This approach provides treatment opportunities for patients previously ineligible for new antigen vaccine therapy.
    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
    背景:目前髓母细胞瘤的护理治疗标准是不够的,因为这些标准没有考虑到肿瘤的异质性。较新,更安全,对于标准疗法无法治愈的高危髓母细胞瘤患者,需要采用针对患者的治疗方法.免疫疗法是一种有前途的治疗方式,可能是提高生存率和避免发病率的关键。为了有效的免疫反应,必须靶向适当的肿瘤抗原。虽然先前已经报道了具有亚组特异性遗传替换的髓母细胞瘤患者,这些遗传改变的免疫原性仍然未知。这项研究的目的是确定潜在的肿瘤排斥抗原,以开发针对髓母细胞瘤的抗原定向细胞疗法。
    方法:我们开发了癌症免疫基因组学流程,并对髓母细胞瘤亚组特异性转录谱进行了全面分析(n=170,18WNT,46SHH,41组3和65组4患者肿瘤)可通过国际癌症基因组联盟(ICGC)和欧洲基因组-表型档案(EGA)获得。我们在包括新抗原在内的广泛抗原类别中进行了计算机抗原预测,肿瘤相关抗原(TAA),和融合蛋白。此外,我们使用最新的计算反卷积方法评估了肿瘤细胞中的抗原加工和呈递途径以及免疫浸润细胞景观.
    结果:发现髓母细胞瘤患者表达多种私有和共有的免疫原性抗原。预测的TAA的比例高于所有分子亚群的新抗原和基因融合,除了声波刺猬(SHH),具有较高的新抗原负担。重要的是,癌症-睾丸抗原,以及以前未被重视的神经发育抗原,发现所有髓母细胞瘤亚组的大多数患者都有表达。尽管免疫寒冷,发现髓母细胞瘤亚组具有不同的免疫细胞基因特征。
    结论:使用自定义抗原预测管道,我们确定了潜在的肿瘤排斥抗原,这对于髓母细胞瘤免疫治疗的发展具有重要意义.
    BACKGROUND: The current standard of care treatments for medulloblastoma are insufficient as these do not take tumor heterogeneity into account. Newer, safer, patient-specific treatment approaches are required to treat high-risk medulloblastoma patients who are not cured by the standard therapies. Immunotherapy is a promising treatment modality that could be key to improving survival and avoiding morbidity. For an effective immune response, appropriate tumor antigens must be targeted. While medulloblastoma patients with subgroup-specific genetic substitutions have been previously reported, the immunogenicity of these genetic alterations remains unknown. The aim of this study is to identify potential tumor rejection antigens for the development of antigen-directed cellular therapies for medulloblastoma.
    METHODS: We developed a cancer immunogenomics pipeline and performed a comprehensive analysis of medulloblastoma subgroup-specific transcription profiles (n = 170, 18 WNT, 46 SHH, 41 Group 3, and 65 Group 4 patient tumors) available through International Cancer Genome Consortium (ICGC) and European Genome-Phenome Archive (EGA). We performed in silico antigen prediction across a broad array of antigen classes including neoantigens, tumor-associated antigens (TAAs), and fusion proteins. Furthermore, we evaluated the antigen processing and presentation pathway in tumor cells and the immune infiltrating cell landscape using the latest computational deconvolution methods.
    RESULTS: Medulloblastoma patients were found to express multiple private and shared immunogenic antigens. The proportion of predicted TAAs was higher than neoantigens and gene fusions for all molecular subgroups, except for sonic hedgehog (SHH), which had a higher neoantigen burden. Importantly, cancer-testis antigens, as well as previously unappreciated neurodevelopmental antigens, were found to be expressed by most patients across all medulloblastoma subgroups. Despite being immunologically cold, medulloblastoma subgroups were found to have distinct immune cell gene signatures.
    CONCLUSIONS: Using a custom antigen prediction pipeline, we identified potential tumor rejection antigens with important implications for the development of immunotherapy for medulloblastoma.
    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
    免疫肽正在成为一个越来越重要的研究领域。鉴定在人类免疫系统中具有关键作用的免疫肽的能力对于将当前的治疗医学转向个性化医学至关重要。多年来,该领域已经成熟,深入了解当前的陷阱。如今,人们普遍认为,将shot弹枪蛋白质组学工作流程进行概括是一种弊端,因为免疫功能疗法面临许多挑战。虽然这些困难中的许多已经得到解决,通往理想工作流程的道路仍然很复杂。尽管已证明免疫肽组中存在翻译后修饰(PTM),尽管它们对免疫疗法具有重要意义,但它们的鉴定仍然极具挑战性。免疫肽组中大量不可预测的修饰在功能和这些挑战中起着关键作用。这篇综述全面概述了免疫肽学中的最新进展。我们深入研究了与在免疫肽组中识别PTM相关的挑战,旨在解决该领域的当前状态。
    Immunopeptidomics is becoming an increasingly important field of study. The capability to identify immunopeptides with pivotal roles in the human immune system is essential to shift the current curative medicine towards personalized medicine. Throughout the years, the field has matured, giving insight into the current pitfalls. Nowadays, it is commonly accepted that generalizing shotgun proteomics workflows is malpractice because immunopeptidomics faces numerous challenges. While many of these difficulties have been addressed, the road towards the ideal workflow remains complicated. Although the presence of Posttranslational modifications (PTMs) in the immunopeptidome has been demonstrated, their identification remains highly challenging despite their significance for immunotherapies. The large number of unpredictable modifications in the immunopeptidome plays a pivotal role in the functionality and these challenges. This review provides a comprehensive overview of the current advancements in immunopeptidomics. We delve into the challenges associated with identifying PTMs within the immunopeptidome, aiming to address the current state of the field.
    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: English Abstract
    BACKGROUND: Neoantigen reactive T cell (NRT) has the ability to inhibit the growth of tumors expressing specific neoantigens. However, due to the difficult immune infiltration and the inhibition of tumor microenvironment, the therapeutic effect of NRT in solid tumors is limited. In this study, we designed NRT cells (7×19 NRT) that can express both interleukin-7 (IL-7) and chemokine C-C motif ligand 19 (CCL19) in mouse lung cancer cells, and evaluated the difference in anti-tumor effect between 7×19 NRT cells and conventional NRT cells.
    METHODS: We performed next-generation sequencing and neoantigen prediction for mouse Lewis lung carcinoma (LLC), prepared RNA vaccine, cultured NRT cells, constructed retroviral vectors encoding IL-7 and CCL19, transduced NRT cells and IL-7 and CCL19 were successfully expressed, and 7×19 NRT was successfully obtained. The anti-tumor effect was evaluated in vivo and in vitro in mice.
    RESULTS: The 7×19 NRT cells significantly enhanced the proliferation and invasion ability of T cells by secreting IL-7 and CCL19, achieved significant tumor inhibition in the mouse lung cancer and extended the survival period of mice. The T cell infiltration into tumor tissue and the necrosis of tumor tissue increased significantly after 7×19 NRT treatment. In addition, both 7×19 NRT treatment and conventional NRT treatment were safe.
    CONCLUSIONS: The anti-solid tumor ability of NRT cells is significantly enhanced by the arming of IL-7 and CCL19, which is a safe and effective genetic modification of NRT.
    【中文题目:共表达IL-7/CCL19的新抗原反应性T细胞
对小鼠肺癌的抗肿瘤研究】 【中文摘要:背景与目的 新抗原反应性T细胞(neoantigen reactive T cell, NRT)具有抑制表达特异性新抗原的肿瘤生长的能力。然而,由于免疫浸润困难和肿瘤微环境的抑制,NRT在实体瘤中的治疗效果有限。本研究针对小鼠肺癌细胞设计出可以同时表达白细胞介素7(interleukin 7, IL-7)和趋化因子19(chemokine C-C motif ligand 19, CCL19)的NRT细胞(7×19 NRT),并对7×19 NRT细胞和常规NRT细胞的抗肿瘤效果差异进行了评估。方法 针对小鼠Lewis肺癌细胞(Lewis lung carcinoma, LLC)进行了新一代测序和新抗原预测,制备了RNA疫苗,培养了NRT细胞,构建了编码IL-7和CCL19的逆转录病毒载体,转导NRT细胞并成功表达IL-7和CCL19,成功获得了7×19 NRT,并在小鼠体内外对其抗肿瘤效果进行了评估。结果 7×19 NRT细胞通过分泌IL-7和CCL19显著增强T细胞的增殖和侵袭能力,在小鼠肺癌中实现了显著的抑瘤作用,延长了小鼠的生存期。经7×19 NRT治疗后,T细胞浸润肿瘤组织及肿瘤组织坏死显著增加。此外,7×19 NRT治疗与常规NRT治疗均安全。结论 通过IL-7和CCL19的表达,NRT细胞的抗实体瘤能力显著增强,这是一种对NRT安全有效的基因修饰。
】 【中文关键词:新抗原;白细胞介素7;趋化因子19;肺肿瘤;新抗原反应性T细胞】.
    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
    个性化癌症疫苗已成为癌症治疗或预防策略的有希望的途径。这种方法针对个体患者肿瘤中的特定遗传改变,提供更个性化和有效的治疗选择。以前的研究表明,针对有限范围的基因突变的广义肽疫苗是无效的,强调需要个性化的方法。虽然研究已经探索了个性化的mRNA疫苗,在这种情况下,尚未研究个性化的肽疫苗。胰腺导管腺癌(PDAC)在肿瘤学中仍然具有挑战性,需要创新的治疗策略。在这项研究中,我们开发了一种个性化的肽疫苗设计方法,采用RNA测序(RNAseq)鉴定患者实体瘤组织中PDAC发展的流行基因突变。我们对修剪适配器进行了RNAseq分析,读取对齐,和体细胞变异呼叫。我们还开发了一个名为SCGeneID的Python程序,这验证了RNAseq分析的比对。Python程序可以免费下载。利用染色体数和基因座数据,SCGeneID沿着UCSChg38参考集鉴定靶基因。根据基因突变数据,我们开发了一种个性化的PDAC癌症疫苗,该疫苗针对两名患者的100个高度流行的基因突变.我们预测肽-MHC结合亲和力,免疫原性,抗原性,变应原性,和每个表位的毒性。然后,我们根据之前发表的疫苗设计方法选择了前50和100个表位.最后,我们产生了pMHC-TCR三维分子模型复杂结构,可免费下载。设计的个性化癌症疫苗含有PDAC实体瘤组织中常见的表位。我们的个性化疫苗由新抗原组成,允许针对癌细胞的更精确和有针对性的免疫反应。此外,我们发现了突变基因,在参考研究中也发现了这一点,我们获得了测序数据,从而验证我们的疫苗设计方法。这是第一个使用人类患者数据设计针对新抗原的个性化肽癌症疫苗的研究,以鉴定与感兴趣的特定肿瘤相关的基因突变。
    Personalized cancer vaccines have emerged as a promising avenue for cancer treatment or prevention strategies. This approach targets the specific genetic alterations in individual patient\'s tumors, offering a more personalized and effective treatment option. Previous studies have shown that generalized peptide vaccines targeting a limited scope of gene mutations were ineffective, emphasizing the need for personalized approaches. While studies have explored personalized mRNA vaccines, personalized peptide vaccines have not yet been studied in this context. Pancreatic ductal adenocarcinoma (PDAC) remains challenging in oncology, necessitating innovative therapeutic strategies. In this study, we developed a personalized peptide vaccine design methodology, employing RNA sequencing (RNAseq) to identify prevalent gene mutations underlying PDAC development in a patient solid tumor tissue. We performed RNAseq analysis for trimming adapters, read alignment, and somatic variant calling. We also developed a Python program called SCGeneID, which validates the alignment of the RNAseq analysis. The Python program is freely available to download. Using chromosome number and locus data, SCGeneID identifies the target gene along the UCSC hg38 reference set. Based on the gene mutation data, we developed a personalized PDAC cancer vaccine that targeted 100 highly prevalent gene mutations in two patients. We predicted peptide-MHC binding affinity, immunogenicity, antigenicity, allergenicity, and toxicity for each epitope. Then, we selected the top 50 and 100 epitopes based on our previously published vaccine design methodology. Finally, we generated pMHC-TCR 3D molecular model complex structures, which are freely available to download. The designed personalized cancer vaccine contains epitopes commonly found in PDAC solid tumor tissue. Our personalized vaccine was composed of neoantigens, allowing for a more precise and targeted immune response against cancer cells. Additionally, we identified mutated genes, which were also found in the reference study, where we obtained the sequencing data, thus validating our vaccine design methodology. This is the first study designing a personalized peptide cancer vaccine targeting neoantigens using human patient data to identify gene mutations associated with the specific tumor of interest.
    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
    低表达抗原,尤其是新抗原,对低免疫原性胰腺癌的免疫治疗提出了重大挑战。增加肿瘤突变负担对于增强肿瘤抗原的表达和提高肿瘤免疫原性至关重要。然而,DNA稳定性的不完全干预阻碍了肿瘤突变负荷的有效升高,从而降低新抗原的可能性。为了解决这个问题,我们已经开发了一种新型的纳米调节剂,可以干预肿瘤细胞的DNA稳定性,从而增强肿瘤突变。这种纳米调节剂包括包裹DNA损伤剂阿霉素的金属有机框架(MOFs)和DNA损伤修复抑制剂siRNA-ATR,能够同时诱导DNA突变和抑制其修复。重要的是,这个调节器,命名为MOFDOX&siATR,可以调节肿瘤基因表达谱,诱导Atp8b1新抗原的产生,增强胰腺癌的免疫原性。MOFDOX和siATR的DNA稳定性干预的特征有望增强低免疫原性肿瘤的免疫应答,使其成为治疗胰腺癌的潜在纳米药物。
    Low-expression antigens, especially neoantigens, pose a significant challenge in immunotherapy for low immunogenicity pancreatic cancer. Increasing the tumor mutation burden is crucial to enhance the expression of tumor antigens and improve tumor immunogenicity. However, the incomplete intervention in DNA stability hampers effective elevation of the tumor mutation burden, thus reducing the probability of neoantigen. To address this issue, we have developed a novel nano-regulator that intervenes in the DNA stability of tumor cells, thereby enhancing tumor mutations. This nano-regulator comprises metal-organic frameworks (MOFs) encapsulating DNA damage agent doxorubicin and DNA damage repair inhibitor siRNA-ATR, enabling simultaneous induction of DNA mutations and inhibition of their repair. Importantly, this regulator, named as MOFDOX&siATR, can modulate the tumor gene expression profile, induce the production of neoantigens of Atp8b1, and enhance the immunogenicity of pancreatic cancer. The characteristics of DNA stability intervention by MOFDOX&siATR hold promise for augmenting the immune response in low immunogenic tumors, making it a potential nanomedicine for the treatment of pancreatic cancer.
    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
    CD4+T辅助抗原是癌症疫苗的重要组成部分,但这些MHCII类限制性抗原来源的相关性仍未得到充分研究。为了比较肿瘤特异性辅助抗原与肿瘤无关辅助抗原的有效性,我们为小鼠MC-38结肠癌设计了三种DNA疫苗,单独编码CD8+T细胞新抗原(noHELP)或与“通用”辅助抗原(uniHELP)或辅助新抗原(neoHELP)联合编码。两种类型的帮助疫苗都增加了疫苗诱导的CD8+T细胞的频率,特别是uniHELP增加了KLRG1+和PD-1低效应细胞的比例。然而,当小鼠随后注射MC-38细胞时,只有neoHELP疫苗接种的肿瘤控制效果明显优于noHELP。与uniHELP相比,neoHELP诱导的肿瘤控制依赖于CD4+T细胞的存在,而两种疫苗都依赖于CD8+T细胞。与此相符,含有CD4+或CD8+T细胞新抗原的野生型对应物的neoHELP变体显示出降低的肿瘤控制。这些数据表明最佳的个性化癌症疫苗应包括MHCII类限制性新抗原以引发肿瘤特异性CD4+T细胞帮助。
    CD4+ T helper antigens are essential components of cancer vaccines, but the relevance of the source of these MHC class II-restricted antigens remains underexplored. To compare the effectiveness of tumor-specific versus tumor-unrelated helper antigens, we designed three DNA vaccines for the murine MC-38 colon carcinoma, encoding CD8+ T cell neoantigens alone (noHELP) or in combination with either \"universal\" helper antigens (uniHELP) or helper neoantigens (neoHELP). Both types of helped vaccines increased the frequency of vaccine-induced CD8+ T cells, and particularly uniHELP increased the fraction of KLRG1+ and PD-1low effector cells. However, when mice were subsequently injected with MC-38 cells, only neoHELP vaccination resulted in significantly better tumor control than noHELP. In contrast to uniHELP, neoHELP-induced tumor control was dependent on the presence of CD4+ T cells, while both vaccines relied on CD8+ T cells. In line with this, neoHELP variants containing wild-type counterparts of the CD4+ or CD8+ T cell neoantigens displayed reduced tumor control. These data indicate that optimal personalized cancer vaccines should include MHC class II-restricted neoantigens to elicit tumor-specific CD4+ T cell help.
    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(ICIs)表现出持久的反应,长期生存益处,与化疗相比,癌症患者的预后有所改善。然而,大多数癌症患者对ICI没有反应,对ICI治疗有反应的患者中有很大比例会对ICI产生先天或后天的耐药性,限制其临床效用。研究最多的ICI反应的预测组织生物标志物是PD-L1免疫组织化学表达,DNA错配修复缺陷,和肿瘤突变负担,尽管这些是ICI反应的弱预测因子。更好的预测性生物标志物的鉴定仍然是改善将受益于ICI的患者的鉴定的重要目标。这里,我们回顾了ICI反应的已建立和新兴的生物标志物,关注癌症患者的表观基因组和基因组改变,它们有可能帮助指导单药ICI免疫治疗或ICI免疫治疗与其他ICI免疫治疗或药物的组合。我们简要回顾了ICI反应生物标志物的现状,包括研究性生物标志物,我们提出了一些新兴的和有前途的表观基因组生物标志物候选的见解,包括目前在黑色素瘤患者ICI免疫治疗反应方面的知识差距。
    Immune checkpoint inhibitors (ICIs) demonstrate durable responses, long-term survival benefits, and improved outcomes in cancer patients compared to chemotherapy. However, the majority of cancer patients do not respond to ICIs, and a high proportion of those patients who do respond to ICI therapy develop innate or acquired resistance to ICIs, limiting their clinical utility. The most studied predictive tissue biomarkers for ICI response are PD-L1 immunohistochemical expression, DNA mismatch repair deficiency, and tumour mutation burden, although these are weak predictors of ICI response. The identification of better predictive biomarkers remains an important goal to improve the identification of patients who would benefit from ICIs. Here, we review established and emerging biomarkers of ICI response, focusing on epigenomic and genomic alterations in cancer patients, which have the potential to help guide single-agent ICI immunotherapy or ICI immunotherapy in combination with other ICI immunotherapies or agents. We briefly review the current status of ICI response biomarkers, including investigational biomarkers, and we present insights into several emerging and promising epigenomic biomarker candidates, including current knowledge gaps in the context of ICI immunotherapy response in melanoma patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号