Cancer vaccine

癌症疫苗
  • 文章类型: Journal Article
    长期以来,卵巢癌(OC)的死亡率一直是妇科恶性肿瘤中最高的。虽然OC被认为是一种免疫原性肿瘤,免疫治疗效果不理想。免疫抑制微环境是其中一个原因,另一个原因是缺乏公认的疫苗有效抗原。化疗,作为OC最常用的治疗方法之一,在治疗过程中可以产生化疗相关抗原(CAAs),并显示出原位疫苗的效果。在这里,我们设计了一种抗原捕获纳米疫苗NP-TP1@M-M,其肿瘤靶向肽TMTP1和树突状细胞(DC)受体甘露糖组装在表面,佐剂单磷酰脂质A(MPLA)包裹在聚(D,L-丙交酯-共-乙交酯)(PLGA)纳米颗粒。PLGA本身具有抗原捕获能力。TMTP1是由我们的研究小组筛选的肿瘤归巢肽,具有广泛而优异的肿瘤靶向能力。经过这些修改,NP-TP1@M-M可以在化疗后捕获和富集更多的肿瘤特异性抗原,刺激DC成熟,激活适应性免疫,并结合免疫检查点封锁,以最大限度地释放身体的免疫潜能,为OC的治疗提供了一种良好的治疗策略。
    The mortality of ovarian cancer (OC) has long been the highest among gynecological malignancies. Although OC is considered to be an immunogenic tumor, the effect of immunotherapy is not satisfactory. The immunosuppressive microenvironment is one reason for this, and the absence of recognized effective antigens for vaccines is another. Chemotherapy, as one of the most commonly used treatment for OC, can produce chemotherapy-associated antigens (CAAs) during treatment and show the effect of in situ vaccine. Herein, we designed an antigen capture nano-vaccine NP-TP1@M-M with tumor targeting peptide TMTP1 and dendritic cell (DC) receptor mannose assembled on the surface and adjuvant monophosphoryl lipid A (MPLA) encapsulated in the core of poly (D, L-lactide-co-glycolide) (PLGA) nanoparticles. PLGA itself possessed the ability of antigen capture. TMTP1 was a tumor-homing peptide screened by our research team, which held extensive and excellent tumor targeting ability. After these modifications, NP-TP1@M-M could capture and enrich more tumor-specific antigens after chemotherapy, stimulate DC maturation, activate the adaptive immunity and combined with immune checkpoint blockade to maximize the release of the body\'s immune potential, providing an eutherapeutic strategy for the treatment of OC.
    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
    胰腺导管腺癌(PDAC)是美国第三大致死性癌症,导致中位生存期为6个月,5年总生存期(OS)不到5%。作为唯一可能治愈的治疗方法,由于诊断较晚,手术切除不适合多达90%的PDAC患者。具有免疫抑制性肿瘤微环境的高度纤维化PDAC限制了细胞毒性T淋巴细胞(CTL)的浸润和功能,从而导致全身疗法如基于树突状细胞(DC)的免疫疗法的成功有限。在这项研究中,我们研究了不可逆电穿孔(IRE)消融治疗联合DC疫苗治疗抗PDAC的潜在益处.
    我们进行了文献检索,以确定专注于DC疫苗治疗和IRE消融的研究,以增强针对PubMed索引的PDAC的治疗反应,WebofScience,和Scopus直到2月20日,2023年。
    IRE消融破坏肿瘤结构,同时保留细胞外基质和血管促进局部炎症。研究表明,IRE消融除了增强啮齿动物模型中的免疫反应外,还可以减少肿瘤纤维化并促进CTL肿瘤向PDAC肿瘤的浸润。与单独使用DC疫苗接种或IRE相比,在IRE消融后施用DC疫苗协同地增强治疗反应并延长OS率。此外,数据驱动方法的实施进一步允许在IRE+DC疫苗免疫消融后动态和纵向监测治疗反应和OS.
    IRE消融和DC疫苗免疫疗法的组合是增强PDAC患者治疗结果的有效策略。
    UNASSIGNED: Pancreatic ductal adenocarcinoma (PDAC) is 3rd most lethal cancer in the USA leading to a median survival of six months and less than 5% 5-year overall survival (OS). As the only potentially curative treatment, surgical resection is not suitable for up to 90% of the patients with PDAC due to late diagnosis. Highly fibrotic PDAC with an immunosuppressive tumor microenvironment restricts cytotoxic T lymphocyte (CTL) infiltration and functions causing limited success with systemic therapies like dendritic cell (DC)-based immunotherapy. In this study, we investigated the potential benefits of irreversible electroporation (IRE) ablation therapy in combination with DC vaccine therapy against PDAC.
    UNASSIGNED: We performed a literature search to identify studies focused on DC vaccine therapy and IRE ablation to boost therapeutic response against PDAC indexed in PubMed, Web of Science, and Scopus until February 20th, 2023.
    UNASSIGNED: IRE ablation destructs tumor structure while preserving extracellular matrix and blood vessels facilitating local inflammation. The studies demonstrated IRE ablation reduces tumor fibrosis and promotes CTL tumor infiltration to PDAC tumors in addition to boosting immune response in rodent models. The administration of the DC vaccine following IRE ablation synergistically enhances therapeutic response and extends OS rates compared to the use of DC vaccination or IRE alone. Moreover, the implementation of data-driven approaches further allows dynamic and longitudinal monitoring of therapeutic response and OS following IRE plus DC vaccine immunoablation.
    UNASSIGNED: The combination of IRE ablation and DC vaccine immunotherapy is a potent strategy to enhance the therapeutic outcomes in patients with PDAC.
    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(CRC)仍然是一个重大的全球健康负担,强调创新治疗策略的必要性。95%的CRC种群是微卫星稳定的(MSS),对抗PD-1等经典免疫疗法不敏感;另一方面,反应者可以变得耐药和复发。最近,癌症疫苗的使用增强了对肿瘤细胞的免疫反应。在这种情况下,我们开发了一种基于刺激肿瘤细胞(STC)平台技术的治疗性疫苗。该疫苗由选定的肿瘤细胞系组成,这些细胞系在体外应激和半抗原化,以产生免疫原性癌症相关抗原的工厂,通过与患者活检的蛋白质组交叉分析进行验证。该技术允许对免疫系统进行多特异性教育,以靶向具有抗性克隆的肿瘤细胞。这里,我们报道了小鼠版STC疫苗在CT26BALB/c同系CRC小鼠模型上的安全性和抗肿瘤效力.我们表明一种基于细胞系(1CL)的STC疫苗抑制肿瘤生长并延长存活。此外,基于三种细胞系(3CL)的STC疫苗通过呈递额外的肿瘤相关抗原诱导多特异性抗肿瘤免疫应答,显著改善了这些参数.此外,蛋白质组学分析证实,与基于1CL的STC疫苗相比,基于3CL的STC疫苗代表的肿瘤相关蛋白的质量范围更广,涵盖了与肿瘤可塑性和治疗抗性相关的关键肿瘤抗原类别.我们还评估了STC疫苗在MC38抗PD-1抗性同系小鼠模型中的功效。使用基于3CL的STC疫苗的疫苗接种显着提高了存活率,并显示出通过CD8淋巴细胞T细胞和M1巨噬细胞浸润的增加而具有抗肿瘤活性的证实的完全应答。这些结果证明了该技术生产用于治疗CRC患者的人类疫苗的潜力。
    Colorectal cancer (CRC) remains a significant global health burden, emphasizing the need for innovative treatment strategies. 95% of the CRC population are microsatellite stable (MSS), insensitive to classical immunotherapies such as anti-PD-1; on the other hand, responders can become resistant and relapse. Recently, the use of cancer vaccines enhanced the immune response against tumor cells. In this context, we developed a therapeutic vaccine based on Stimulated Tumor Cells (STC) platform technology. This vaccine is composed of selected tumor cell lines stressed and haptenated in vitro to generate a factory of immunogenic cancer-related antigens validated by a proteomic cross analysis with patient\'s biopsies. This technology allows a multi-specific education of the immune system to target tumor cells harboring resistant clones. Here, we report safety and antitumor efficacy of the murine version of the STC vaccine on CT26 BALB/c CRC syngeneic murine models. We showed that one cell line (1CL)-based STC vaccine suppressed tumor growth and extended survival. In addition, three cell lines (3CL)-based STC vaccine significantly improves these parameters by presenting additional tumor-related antigens inducing a multi-specific anti-tumor immune response. Furthermore, proteomic analyses validated that the 3CL-based STC vaccine represents a wider quality range of tumor-related proteins than the 1CL-based STC vaccine covering key categories of tumor antigens related to tumor plasticity and treatment resistance. We also evaluated the efficacy of STC vaccine in an MC38 anti-PD-1 resistant syngeneic murine model. Vaccination with the 3CL-based STC vaccine significantly improved survival and showed a confirmed complete response with an antitumor activity carried by the increase of CD8+ lymphocyte T cells and M1 macrophage infiltration. These results demonstrate the potential of this technology to produce human vaccines for the treatment of patients with CRC.
    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
    雌激素代表的逃学,黄体酮,和人表皮生长因子受体发生在TNBC。与其余乳腺癌(BC)类型相比,TNBC被认为是上复发,诊断较差。目前,因此,TNBC没有批准靶向治疗,治疗方案需要化疗和手术,死亡率很高。因此,本文重点介绍了TNBC的重要通路,并讨论了TNBC治疗的最新进展,包括免疫检查点抑制剂(ICIs),PARP抑制剂,和癌症疫苗。免疫治疗和ICIs,像PD1和PDL1抑制剂,在临床试验(CT)中显示出潜力。这些抑制剂阻碍了允许肿瘤细胞逃避系统的机制,从而增强了人体对TNBC的防御能力。免疫疗法,单独或联合化疗已证明患者的预后,例如生存率提高和治疗相关副作用减少。此外,靶向治疗方法包括BRCA/2突变多聚核糖聚合酶抑制剂,血管内皮生长因子受体(VEGFR)抑制剂,表皮生长因子受体抑制剂,成纤维细胞生长因子抑制剂,雄激素受体抑制剂,PIK3/AKT/mTOR通路抑制剂,细胞周期蛋白依赖性激酶(CDK)抑制剂,Notch信号通路抑制剂,信号转导和转录激活因子3(STAT3)信号通路抑制剂,嵌合抗原受体T(CAR-T)细胞疗法,转化生长因子(TGF)-β抑制剂,表观遗传修饰(EPM)极光激酶抑制剂和抗体-药物缀合物。我们还强调了正在进行的临床试验和TNBC治疗的潜在未来方向。尽管治疗TNBC面临挑战,了解TNBC分子和免疫特性的最新进展为靶向治疗开辟了新的机会,这有望改善这种侵袭性疾病的结果。
    The truancy of representation of the estrogen, progesterone, and human epidermal growth factor receptors occurs during TNBC. TNBC is recognized for the upper reappearance and has a poorer diagnosis compared with rest breast cancer (BC) types. Presently, as such, no targeted therapy is approved for TNBC and treatment options are subjected to chemotherapy and surgery, which have high mortality rates. Hence, the current article focuses on the scenario of TNBC vital pathways and discusses the latest advances in TNBC treatment, including immune checkpoint inhibitors (ICIs), PARP suppressors, and cancer vaccines. Immunotherapy and ICIs, like PD 1 and PD L1 suppressors, displayed potential in clinical trials (CTs). These suppressors obstruct the mechanisms which allow tumor cells to evade the system thereby boosting the body\'s defense against TNBC. Immunotherapy, either alone or combined with chemotherapy has demonstrated patient outcomes such as increased survival rates and reduced treatment-related side effects. Additionally, targeted therapy approaches include BRCA/2 mutation poly ribose polymerase inhibitors, Vascular Endothelial Growth Factor Receptor (VEGFR) inhibitors, Epidermal growth factor receptor inhibitors, Fibroblast growth factor inhibitors, Androgen Receptor inhibitors, PIK3/AKT/mTOR pathway inhibitors, Cyclin-dependent kinase (CDK) inhibitors, Notch signaling pathway inhibitors, Signal transducer and activator of transcription 3 (STAT3) signaling pathway inhibitors, Chimeric antigen receptor T (CAR-T) cell therapy, Transforming growth factor (TGF) -β inhibitors, Epigenetic modifications (EPM), Aurora Kinase inhibitors and antibody-drug conjugates. We also highlight ongoing clinical trials and potential future directions for TNBC therapy. Despite the challenges in treating TNBC, recent developments in understanding the molecular and immune characteristics of TNBC have opened up new opportunities for targeted therapies, which hold promise for improving outcomes in this aggressive disease.
    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
    癌症疫苗已被开发为增强癌症免疫力的有希望的方法。然而,由于肿瘤抗原的不精确递送,它们的临床效力通常受到限制。为了克服这个问题,我们缀合了内源性Toll样受体(TLR)2/6配体,UNE-C1,人乳头瘤病毒16型(HPV-16)衍生的肽抗原,E7,并且发现与UNE-C1和E7的非共价组合相比,UNE-C1缀合的癌症疫苗(UCV)在体内显示出显著增强的抗肿瘤活性。UCV与PD-1阻断的组合进一步增强了其治疗功效。具体来说,UNE-C1与E7的结合增强了其在腹股沟引流淋巴结中的保留,向树突状细胞的特异性递送和E7抗原特异性T细胞应答,以及与两种肽的非共价组合相比的体内抗肿瘤功效。这些发现表明源自人半胱氨酰-tRNA合成酶1(CARS1)的UNE-C1作为通过TLR2/6将癌症抗原特异性递送至抗原呈递细胞以改善癌症疫苗的独特载体的潜力。
    Cancer vaccines have been developed as a promising way to boost cancer immunity. However, their clinical potency is often limited due to the imprecise delivery of tumor antigens. To overcome this problem, we conjugated an endogenous Toll-like receptor (TLR)2/6 ligand, UNE-C1, to human papilloma virus type 16 (HPV-16)-derived peptide antigen, E7, and found that the UNE-C1-conjugated cancer vaccine (UCV) showed significantly enhanced antitumor activity in vivo compared with the noncovalent combination of UNE-C1 and E7. The combination of UCV with PD-1 blockades further augmented its therapeutic efficacy. Specifically, the conjugation of UNE-C1 to E7 enhanced its retention in inguinal draining lymph nodes, the specific delivery to dendritic cells and E7 antigen-specific T cell responses, and antitumor efficacy in vivo compared with the noncovalent combination of the two peptides. These findings suggest the potential of UNE-C1 derived from human cysteinyl-tRNA synthetase 1 as a unique vehicle for the specific delivery of cancer antigens to antigen-presenting cells via TLR2/6 for the improvement of cancer vaccines.
    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
    黑色素瘤,以其侵袭性转移性而闻名,在癌症治疗中提出了巨大的挑战,传统疗法往往不足。这项研究介绍了使用基于肿瘤疫苗的纳米材料的开创性方法,突出了他们在革新黑色素瘤治疗方面的潜力。这项工作采用了有机氮氧化物,特别是4-羧基-TEMPO,与壳聚糖(CS)组合,创造一种新型的纳米复合材料-CS-TEMPO-OVA纳米疫苗。该组合物不仅改善了生物相容性并延长了血液循环时间,而且标志着在MRI技术中与传统的基于钆的造影剂的显著偏离。解决安全问题。CS-TEMPO-OVA纳米疫苗在细胞和类器官水平上都表现出优异的生物相容性。它们有效地刺激骨髓来源的树突状细胞(BMDC),进而促进T细胞的成熟和活化。这最终导致必需细胞因子的强烈产生。这些纳米疫苗具有双重目的:它们既具有治疗性又具有预防性。通过诱导免疫反应,激活细胞毒性T细胞,促进巨噬细胞M1极化,它们有效地抑制黑素瘤生长并提高小鼠模型的存活率。当与αPD-1结合时,CS-TEMPO-OVA纳米疫苗显着增强肿瘤内细胞毒性T淋巴细胞(CTL)的浸润,激发强大的系统性抗肿瘤反应,有效遏制肿瘤转移。这些纳米疫苗控制原发性(皮下)和转移性B16-OVA肿瘤的能力突出了它们的显著功效。此外,CS-TEMPO-OVA纳米疫苗可以通过静脉内和肌内途径在体内给药,两者均有效增强了肿瘤组织磁共振成像的T1对比度。这项研究为这些纳米疫苗在临床诊断和治疗中的综合应用提供了宝贵的见解。标志着癌症研究和患者护理的重大进步。
    Melanoma, known for its aggressive metastatic nature, presents a formidable challenge in cancer treatment, where conventional therapies often fall short. This study introduces a pioneering approach utilizing metal-free nanosystem as tumor vaccines, spotlighting their potential in revolutionizing melanoma treatment. This work employed organic nitroxides, specifically 4-carboxy-TEMPO, in combination with chitosan (CS), to create a novel nanocomposite material - the CS-TEMPO-OVA nanovaccines. This composition not only improves biocompatibility and extends blood circulation time of TEMPO but also marks a significant departure from traditional gadolinium-based contrast agents in MRI technology, addressing safety concerns. CS-TEMPO-OVA nanovaccines demonstrate excellent biocompatibility at both the cellular and organoid level. They effectively stimulate bone marrow-derived dendritic cells (BMDCs), which in turn promote the maturation and activation of T cells. This ultimately leads to a strong production of essential cytokines. These nanovaccines serve a dual purpose as both therapeutic and preventive. By inducing an immune response, activating cytotoxic T cells, and promoting macrophage M1 polarization, they effectively inhibit melanoma growth and enhance survival in mouse models. When combined with αPD-1, the CS-TEMPO-OVA nanovaccines significantly bolster the infiltration of cytotoxic T lymphocytes (CTLs) within tumors, sparking a powerful systemic antitumor response that effectively curbs tumor metastasis. The ability of these nanovaccines to control both primary (subcutaneous) and metastatic B16-OVA tumors highlights their remarkable efficacy. Furthermore, the CS-TEMPO-OVA nanovaccine can be administered in vivo via both intravenous and intramuscular routes, both of which effectively enhance the T1 contrast of magnetic resonance imaging in tumor tissue. This study offers invaluable insights into the integrated application of these nanovaccines in both clinical diagnostics and treatment, marking a significant stride in cancer research and patient care.
    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
    mRNA递送系统,如脂质纳米颗粒(LNP),在改善mRNA表达方面取得了显著进展,而免疫系统激活作用在一个阈值上。维持抗原表达和树突状细胞(DC)活化之间的微妙平衡对于有效的免疫识别至关重要。这里,开发了用磷酸钙纳米颗粒(CaP-PME)稳定的水包油包水(w/o/w)皮克林乳液,用于癌症疫苗接种中的mRNA递送。CaP-PME有效地将mRNA转运到细胞质中,诱导促炎反应并通过破坏细胞内钙/钾离子平衡激活DC。与LNP不同,CaP-PME显示出对DC的偏好,增强它们的激活和向淋巴结的迁移。它引发干扰素-γ介导的CD8+T细胞反应,促进NK细胞增殖和活化,导致明显的NK细胞浸润和改善肿瘤微环境。制备的w/o/wPickering乳剂在E.G7和B16-OVA肿瘤模型中表现出优异的抗肿瘤效果,作为癌症疫苗接种的增强mRNA递送载体,提供了有希望的前景。
    mRNA delivery systems, such as lipid nanoparticle (LNP), have made remarkable strides in improving mRNA expression, whereas immune system activation operates on a threshold. Maintaining a delicate balance between antigen expression and dendritic cell (DC) activation is vital for effective immune recognition. Here, a water-in-oil-in-water (w/o/w) Pickering emulsion stabilized with calcium phosphate nanoparticles (CaP-PME) is developed for mRNA delivery in cancer vaccination. CaP-PME efficiently transports mRNA into the cytoplasm, induces pro-inflammatory responses and activates DCs by disrupting intracellular calcium/potassium ions balance. Unlike LNP, CaP-PME demonstrates a preference for DCs, enhancing their activation and migration to lymph nodes. It elicits interferon-γ-mediated CD8+ T cell responses and promotes NK cell proliferation and activation, leading to evident NK cells infiltration and ameliorated tumor microenvironment. The prepared w/o/w Pickering emulsion demonstrates superior anti-tumor effects in E.G7 and B16-OVA tumor models, offering promising prospects as an enhanced mRNA delivery vehicle for cancer vaccinations.
    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
    膀胱癌(BC)约占所有恶性肿瘤的4%。非肌肉侵入性BC,75%的病例,经尿道电切术和辅助膀胱灌注治疗,而肌肉浸润性BC需要以顺铂为基础的围手术期化疗。虽然免疫检查点抑制剂,抗体药物缀合物和靶向药物提供了巨大的进步,转移性BC仍然是一种通常无法治愈的疾病,临床试验继续积极评估新分子.癌症疫苗旨在激活患者的免疫系统对抗肿瘤细胞。已经开发了几种递送新抗原的方法,包括肽,抗原呈递细胞,病毒,或核酸。各种改进不断被探索,如佐剂的使用和组合策略。近年来,以核酸为基础的疫苗越来越受到重视,对其他恶性肿瘤有希望的结果。然而,尽管最近的优势,许多障碍依然存在。这篇综述旨在描述不同类型的癌症疫苗,他们在UC患者中的评估以及该领域的最新创新。
    Bladder cancer (BC) accounts for about 4% of all malignancies. Non-muscle-invasive BC, 75% of cases, is treated with transurethral resection and adjuvant intravesical instillation, while muscle-invasive BC warrants cisplatin-based perioperative chemotherapy. Although immune-checkpoint inhibitors, antibody drug conjugates and targeted agents have provided dramatic advances, metastatic BC remains a generally incurable disease and clinical trials continue to vigorously evaluate novel molecules. Cancer vaccines aim at activating the patient\'s immune system against tumor cells. Several means of delivering neoantigens have been developed, including peptides, antigen-presenting cells, virus, or nucleic acids. Various improvements are constantly being explored, such as adjuvants use and combination strategies. Nucleic acids-based vaccines are increasingly gaining attention in recent years, with promising results in other malignancies. However, despite the recent advantages, numerous obstacles persist. This review is aimed at describing the different types of cancer vaccines, their evaluations in UC patients and the more recent innovations in this field.
    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
    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
    胰胆管癌,包括胰腺和胆道的恶性肿瘤,提出了一个可怕的临床挑战,其特征是一致的预后暗淡。其早期阶段的无症状性质通常导致延迟检测,导致不利的5年总生存率。传统的治疗方式显示出有限的疗效,强调迫切需要替代治疗方法。近年来,免疫疗法已成为对抗胰胆管癌的一个有希望的途径。诸如治疗性疫苗和肿瘤浸润性淋巴细胞的使用等策略因其引起更稳健和持久反应的潜力而引起关注。这篇综述旨在阐明新兴的免疫治疗干预措施的前景,提供从临床和研究角度的见解。通过加深对胰胆管癌的认识,探索创新的治疗方式,我们的目标是促进改善患者预后和生活质量.
    Pancreaticobiliary cancer, encompassing malignancies of both the pancreatic and biliary tract, presents a formidable clinical challenge marked by a uniformly bleak prognosis. The asymptomatic nature of its early stages often leads to delayed detection, contributing to an unfavorable 5-year overall survival rate. Conventional treatment modalities have shown limited efficacy, underscoring the urgent need for alternative therapeutic approaches. In recent years, immunotherapy has emerged as a promising avenue in the fight against pancreaticobiliary cancer. Strategies such as therapeutic vaccines and the use of tumor-infiltrating lymphocytes have garnered attention for their potential to elicit more robust and durable responses. This review seeks to illuminate the landscape of emerging immunotherapeutic interventions, offering insights from both clinical and research perspectives. By deepening our understanding of pancreaticobiliary cancer and exploring innovative treatment modalities, we aim to catalyze improvements in patient outcomes and quality of life.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号