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

       PDF(Pubmed)

  • 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
    目前缺乏具有特别是通过利用抗原呈递细胞(APC)改善适应性免疫的特征的疫苗佐剂。在先前的工作中,我们从Granolafrondosa的子实体中获得了一种新的可溶性300kDa均质β-葡聚糖,名为GFPBW1。GFPBW1可以通过靶向树突状细胞相关的C型凝集素1(Dectin-1)/Syk/NF-κB信号来激活巨噬细胞,从而实现抗肿瘤作用。在这项研究中,用OVA抗原和B16-OVA肿瘤模型探索了GFPBW1的佐剂作用。我们表明,GFPBW1(5、50、500μg/mL)通过增加CD80,CD86和MHCII表达而剂量依赖性地促进APC的体外激活和成熟。我们用OVA与GFPBW1(50或300μg)组合免疫雌性小鼠两次,间隔两周。GFPBW1显着和剂量依赖性地增加不同亚型的OVA特异性抗体滴度,包括IgG1,IgG2a,IgG2b和IgG3,表明它可以作为Th1和Th2型免疫应答的佐剂。此外,GFPBW1与铝组合显着增加OVA特异性IgG2a和IgG2b的滴度,而不是IgG1,这表明GFPBW1可以用作铝的辅助佐剂来补偿Th1缺乏。对于用OVA加GFPBW1免疫的小鼠,在主要器官或注射部位均未观察到明显的病理损伤。并且没有注意到任何血液学参数的异常。当GFPBW1在B16-OVA癌症疫苗模型中作为佐剂时,它可以用预防性疫苗完成整个肿瘤抑制,用治疗性疫苗增强抗肿瘤功效。发现差异表达基因在抗原加工过程中富集,特异性增加的DC肿瘤浸润,OVA加GFPBW1组的B1细胞和浆细胞,根据其对APC的激活和成熟功能。总的来说,这项研究系统地描述了GFPBW1作为一种新型有效和安全的佐剂的特性,并强调了其在疫苗开发中的巨大潜力。
    Adjuvants for vaccines with characteristics of improving adaptive immunity particularly via leverage of antigen presenting cells (APCs) are currently lacking. In a previous work we obtained a new soluble 300 kDa homogeneous β-glucan named GFPBW1 from the fruit bodies of Granola frondosa. GFPBW1 could activate macrophages by targeting dendritic cell associated C-type lectin 1 (Dectin-1)/Syk/NF-κB signaling to achieve antitumour effects. In this study the adjuvant effects of GFPBW1 were explored with OVA-antigen and B16-OVA tumor model. We showed that GFPBW1 (5, 50, 500 μg/mL) dose-dependently promoted activation and maturation of APCs in vitro by increasing CD80, CD86 and MHC II expression. We immunized female mice with OVA in combination with GFPBW1 (50 or 300 μg) twice with an interval of two weeks. GFPBW1 markedly and dose-dependently increased OVA-specific antibody titers of different subtypes including IgG1, IgG2a, IgG2b and IgG3, suggesting that it could serve as an adjuvant for both Th1 and Th2 type immune responses. Furthermore, GFPBW1 in combination with aluminum significantly increased the titers of OVA-specific IgG2a and IgG2b, but not those of IgG1, suggesting that GFPBW1 could be used as a co-adjuvant of aluminum to compensate for Th1 deficiency. For mice immunized with OVA plus GFPBW1, no obvious pathological injury was observed in either major organs or injection sites, and no abnormalities were noted for any of the hematological parameters. When GFPBW1 served as an adjuvant in the B16-OVA cancer vaccine models, it could accomplish entire tumor suppression with preventive vaccines, and enhance antitumour efficacy with therapeutic vaccines. Differentially expressed genes were found to be enriched in antigen processing process, specifically increased tumor infiltration of DCs, B1 cells and plasma cells in the OVA plus GFPBW1 group, in accordance with its activation and maturation function of APCs. Collectively, this study systematically describes the properties of GFPBW1 as a novel potent and safe adjuvant and highlights its great potential in vaccine development.
    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
    Mendez-Gomez等人.最近证明了RNA-脂质颗粒聚集体(RNA-LPAs)在免疫治疗中的转化潜力。通过重新编程肿瘤微环境(TME)和增强抗肿瘤免疫力,RNA-LPAs靶向原发性肿瘤并引发强大的全身免疫。这一创新平台有望将临床前成功转化为切实的临床益处。
    Mendez-Gomez et al. recently demonstrated the transformative potential of RNA-lipid particle aggregates (RNA-LPAs) in immunotherapy. By reprogramming the tumor microenvironment (TME) and potentiating antitumor immunity, RNA-LPAs target primary tumors and elicit robust systemic immunity. This innovative platform holds promise for translating preclinical success into tangible clinical benefits.
    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: 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
    免疫原性死亡的肿瘤细胞作为癌症疫苗激活针对原发性和转移性肿瘤的全身免疫具有有希望的前景。尤其是,X射线诱导的垂死肿瘤细胞富含高度免疫原性的肿瘤相关抗原和作为有效佐剂的自生成dsDNA。然而,我们发现X射线诱导过程会导致癌症疫苗中磷脂酰丝氨酸的过度暴露,可以与巨噬细胞上的MerTK受体特异性结合,充当“检查点”,以促进肿瘤微环境中的免疫沉默。因此,我们开发了一种新的策略,将X射线诱导的癌症疫苗与巨噬细胞MerTK检查点抑制剂UNC2250相结合,“用于治疗结肠癌中的腹膜癌。通过将UNC2250纳入治疗方案,巨噬细胞的免疫抑制性细胞增生,依赖于MerTK对疫苗上磷脂酰丝氨酸的定向识别,被有效地封锁了。因此,免疫分析表明,这种组合策略促进了树突状细胞的成熟和巨噬细胞的M1样复极化,从而同时引发强大的适应性和先天免疫。这种利用X射线诱导疫苗与检查点抑制剂组合的创新方法可能为开发针对结肠癌的有效癌症疫苗和免疫疗法提供有价值的见解。
    Immunogenic dying tumor cells hold promising prospects as cancer vaccines to activate systemic immunity against both primary and metastatic tumors. Especially, X-ray- induced dying tumor cells are rich in highly immunogenic tumor-associated antigens and self-generated dsDNA as potent adjuvants. However, we found that the X-ray induction process can result in the excessive exposure of phosphatidylserine in cancer vaccines, which can specifically bind with the MerTK receptor on macrophages, acting as a \"checkpoint\" to facilitate immune silence in the tumor microenvironment. Therefore, we developed a novel strategy combining X-ray-induced cancer vaccines with UNC2250, a macrophage MerTK \"checkpoint inhibitor,\" for treating peritoneal carcinomatosis in colon cancer. By incorporating UNC2250 into the treatment regimen, immunosuppressive efferocytosis of macrophages, which relies on MerTK-directed recognition of phosphatidylserine on vaccines, was effectively blocked. Consequently, the immune analysis revealed that this combination strategy promoted the maturation of dendritic cells and M1-like repolarization of macrophages, thereby simultaneously eliciting robust adaptive and innate immunity. This innovative approach utilizing X-ray-induced vaccines combined with a checkpoint inhibitor may provide valuable insights for developing effective cancer vaccines and immunotherapies targeting colon cancer.
    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
    血液恶性肿瘤(HMs)包括具有显著发病率和死亡率的不同组的血液肿瘤。免疫疗法已成为HMs患者的一种有效和关键的治疗方式。尽管在过去十年中,在理解和实施HMs的免疫疗法方面取得了显著进步,几个挑战依然存在。这些挑战包括免疫相关的不良反应,治疗性抗原在体内的精确生物分布和消除,肿瘤的免疫耐受,和肿瘤细胞在肿瘤微环境(TME)内的免疫逃避。纳米技术,具有在纳米尺度上操纵材料特性的能力,有可能通过改善药物靶向和稳定性等各个方面来解决这些障碍并彻底改变治疗结果。纳米技术和免疫治疗的融合催生了纳米免疫治疗,抗肿瘤治疗的一个专门分支。纳米技术已经在嵌合抗原受体T细胞(CAR-T)治疗中找到了应用,癌症疫苗,免疫检查点抑制剂,以及其他针对HMs的免疫治疗策略。在这篇综述中,我们描述了最近的发展,并讨论了目前在纳米免疫治疗领域的挑战,为这些疾病的基于纳米技术的治疗方法的潜力提供了新的见解。
    Hematological malignancies (HMs) encompass a diverse group of blood neoplasms with significant morbidity and mortality. Immunotherapy has emerged as a validated and crucial treatment modality for patients with HMs. Despite notable advancements having been made in understanding and implementing immunotherapy for HMs over the past decade, several challenges persist. These challenges include immune-related adverse effects, the precise biodistribution and elimination of therapeutic antigens in vivo, immune tolerance of tumors, and immune evasion by tumor cells within the tumor microenvironment (TME). Nanotechnology, with its capacity to manipulate material properties at the nanometer scale, has the potential to tackle these obstacles and revolutionize treatment outcomes by improving various aspects such as drug targeting and stability. The convergence of nanotechnology and immunotherapy has given rise to nano-immunotherapy, a specialized branch of anti-tumor therapy. Nanotechnology has found applications in chimeric antigen receptor T cell (CAR-T) therapy, cancer vaccines, immune checkpoint inhibitors, and other immunotherapeutic strategies for HMs. In this review, we delineate recent developments and discuss current challenges in the field of nano-immunotherapy for HMs, offering novel insights into the potential of nanotechnology-based therapeutic approaches for these diseases.
    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
    宫颈癌,在全球影响女性的最致命癌症中,主要起因于高危型人乳头瘤病毒(HPV)的持续性感染.有效对抗持续性感染,防止癌前病变进展为恶性肿瘤,治疗性HPV疫苗正在开发中。这项研究利用免疫信息学方法,以HPV16菌株的E6和E7癌蛋白为靶抗原,预测细胞毒性T淋巴细胞(CTL)和辅助T淋巴细胞(HTL)的表位。随后,通过精心选择T细胞表位和其他必要元件,构建了多表位疫苗,表现出良好的免疫原性,物理化学,和结构特征。此外,计算机模拟表明,该疫苗不仅与toll样受体(TLR2/TLR3/TLR4)相互作用良好,但也诱导了以升高的Th1型细胞因子为特征的强烈的先天和适应性免疫应答,例如干扰素-γ(IFN-γ)和白细胞介素-2(IL2)。此外,我们的研究调查了不同免疫间隔对免疫反应的影响,旨在优化省时的免疫计划。在动物模型实验中,疫苗表现出强大的免疫原性,治疗性的,和预防效果。管理三次,它持续诱导特异性CD4和CD8T细胞的扩增,导致大量细胞因子释放和脾细胞中记忆T细胞亚群的增殖增加。总的来说,我们的研究结果支持了这种多表位疫苗在对抗HPV16感染方面的潜力,并表明其可用于未来HPV疫苗开发.
    通过对T细胞表位和其他必要元件的严格选择,使用免疫信息学方法构建了一种靶向HPV16E6和E7癌蛋白的新型多表位疫苗.设计的疫苗可以诱导细胞和体液免疫反应,包括所有必需的免疫原性,物理化学,和理想疫苗设计的结构特征。此外,它提供了体面的全球报道。在动物研究中,疫苗表现出强烈的免疫反应,包括CD4和CD8T细胞的扩增,细胞因子释放,增强记忆T细胞增殖,产生长期的抗肿瘤作用,抑制肿瘤生长,和延长荷瘤小鼠的生存期。设计的疫苗的免疫学评估表明其作为针对HPV16的新型候选疫苗的潜力。
    Cervical cancer, among the deadliest cancers affecting women globally, primarily arises from persistent infection with high-risk human papillomavirus (HPV). To effectively combat persistent infection and prevent the progression of precancerous lesions into malignancy, a therapeutic HPV vaccine is under development. This study utilized an immunoinformatics approach to predict epitopes of cytotoxic T lymphocytes (CTLs) and helper T lymphocytes (HTLs) using the E6 and E7 oncoproteins of the HPV16 strain as target antigens. Subsequently, through meticulous selection of T-cell epitopes and other necessary elements, a multi-epitope vaccine was constructed, exhibiting good immunogenic, physicochemical, and structural characteristics. Furthermore, in silico simulations showed that the vaccine not only interacted well with toll-like receptors (TLR2/TLR3/TLR4), but also induced a strong innate and adaptive immune response characterized by elevated Th1-type cytokines, such as interferon-gamma (IFN-γ) and interleukin-2 (IL2). Additionally, our study investigated the effects of different immunization intervals on immune responses, aiming to optimize a time-efficient immunization program. In animal model experiments, the vaccine exhibited robust immunogenic, therapeutic, and prophylactic effects. Administered thrice, it consistently induced the expansion of specific CD4 and CD8 T cells, resulting in substantial cytokines release and increased proliferation of memory T cell subsets in splenic cells. Overall, our findings support the potential of this multi-epitope vaccine in combating HPV16 infection and signify its candidacy for future HPV vaccine development.
    Through the stringent selection of T-cell epitopes and other necessary elements, a novel multi-epitope vaccine targeting HPV 16 E6 and E7 oncoproteins was constructed using an immunoinformatics approach.The vaccine designed can induce both cellular and humoral immune responses, encompassing all the required immunogenic, physicochemical, and structural characteristics for an ideal vaccine design. Moreover, it offers decent worldwide coverage.In animal studies, the vaccine demonstrated strong immune responses, including expansion of CD4 and CD8 T cells, cytokine release, and enhanced memory T cell proliferation, resulting in long-term anti-tumor effects, inhibition of tumor growth, and prolonged survival in tumor-bearing mice.The immunological evaluation of the designed vaccine suggests its potential as a novel vaccine candidate against HPV 16.
    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
    肿瘤免疫疗法已成为一种有效的治疗方法,可以动员患者的免疫系统来实现持久的肿瘤抑制。这里,我们设计了一种光动力疗法驱动的纳米疫苗(Dex-HDL/ALA-Fe3O4)共递送5-氨基乙酰丙酸和Fe3O4纳米酶,证明了一种长期持久的免疫治疗策略.接种疫苗后,纳米疫苗表现出明显的肿瘤部位积累,淋巴结归巢,和特异性和记忆抗肿瘤免疫唤起。激光照射后,Dex-HDL/ALA-Fe3O4在肿瘤部位有效地产生活性氧,不仅诱导免疫原性细胞死亡级联反应,而且触发全类型肿瘤抗原的按需释放。有趣的是,Fe3O4纳米酶催化过氧化氢产生的氧气用于缓解肿瘤缺氧和改变抑制性肿瘤微环境,从而表现出作为敏化剂的显著潜力。在不同的临床前癌症模型中静脉内施用纳米疫苗已经证明了显著的肿瘤消退和术后肿瘤复发和转移的抑制,从而实现针对高度异质性肿瘤的个性化治疗策略。
    Tumor immunotherapy has emerged as an efficacious therapeutic approach that mobilizes the patient\'s immune system to achieve durable tumor suppression. Here, we design a photodynamic therapy-motivated nanovaccine (Dex-HDL/ALA-Fe3O4) co-delivering 5-aminolevulinic acid and Fe3O4 nanozyme that demonstrate a long-term durable immunotherapy strategy. After vaccination, the nanovaccine exhibits obvious tumor site accumulation, lymph node homing, and specific and memory antitumor immunity evocation. Upon laser irradiation, Dex-HDL/ALA-Fe3O4 effectively generates reactive oxygen species at the tumor site not only to induce the immunogenic cell death-cascade but also to trigger the on-demand release of full types of tumor antigens. Intriguingly, Fe3O4 nanozyme-catalyzed hydrogen peroxide generated oxygen for alleviating tumor hypoxia and modifying the inhibitory tumor microenvironment, thereby exhibiting remarkable potential as a sensitizer. The intravenous administration of nanovaccines in diverse preclinical cancer models has demonstrated remarkable tumor regression and inhibition of postoperative tumor recurrence and metastasis, thereby enabling personalized treatment strategies against highly heterogeneous tumors.
    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