cellular vaccine

  • 文章类型: Journal Article
    B细胞,尽管它们有几个独特的功能,在用作过继细胞疗法方面仍未开发,并且仅限于体外用于抗体生产。B细胞可以很容易地来源,它们具有出色的淋巴归巢能力,它们可以作为抗原呈递细胞(APC),提供树突状细胞(DC)的替代品,在临床上显示出有限的疗效。可溶性因子如IL-4和抗CD40抗体可增强其活性,生存,和B细胞的抗原呈递能力;然而,很难获得足够高浓度的这些生物制品以刺激体内B细胞。作为细胞参与剂的微贴片(MACE)是聚合物微粒,表面用抗CD40和抗IgM功能化,它可以附着于B细胞并同时接合多个B细胞受体(BCR)和CD40受体。通过MACE刺激这些受体,与游离抗体不同,增强了共刺激分子在B细胞表面的展示,B细胞活力增加,并在体外改善B细胞向T细胞的抗原呈递。通过MACE的B细胞活化进一步与可溶性IL-4和抗CD40协同。MACE还引起B细胞分泌T细胞趋化因子。静脉注射过继转移后,结合MACE的B细胞归巢于脾脏和淋巴结,抗原呈递给T细胞的关键位点。在小鼠皮下EG7-OVA肿瘤模型中,用卵清蛋白的CD4和CD8表位脉冲的MACE-B细胞的过继转移显着延迟了肿瘤进展,证明MACE赋予B细胞的功能益处。
    B cells, despite their several unique functionalities, remain largely untapped for use as an adoptive cell therapy and are limited to in vitro use for antibody production. B cells can be easily sourced, they possess excellent lymphoid-homing capabilities, and they can act as antigen-presenting cells (APCs), offering an alternative to dendritic cells (DCs), which have shown limited efficacy in the clinical setting. Soluble factors such as IL-4 and anti-CD40 antibody can enhance the activation, survival, and antigen-presenting capabilities of B cells; however, it is difficult to attain sufficiently high concentrations of these biologics to stimulate B cells in vivo. Micropatches as Cell Engagers (MACE) are polymeric microparticles, surface functionalized with anti-CD40 and anti-IgM, which can attach to B cells and simultaneously engage multiple B-cell receptors (BCR) and CD40 receptors. Stimulation of these receptors through MACE, unlike free antibodies, enhanced the display of costimulatory molecules on the B-cell surface, increased B-cell viability, and improved antigen presentation by B cells to T cells in vitro. B-cell activation by MACE further synergized with soluble IL-4 and anti-CD40. MACE also elicited T-cell chemokine secretion by B cells. Upon intravenous adoptive transfer, MACE-bound B cells homed to the spleen and lymph nodes, key sites for antigen presentation to T cells. Adoptive transfer of MACE-B cells pulsed with the CD4+ and CD8+ epitopes of ovalbumin significantly delayed tumor progression in a murine subcutaneous EG7-OVA tumor model, demonstrating the functional benefit conferred to B cells by MACE.
    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
    间充质基质细胞(MSC)通常以其免疫抑制能力而闻名。然而,我们小组提供的证据表明,利用基因工程或药理学手段将MSCs转化为有效的抗原呈递细胞(APC)是可能的.鉴于UM171a在MSC中触发APC样功能的能力,以及最近的发现,这种药物可能通过抑制赖氨酸特异性去甲基酶1(LSD1)来调节表观基因组,我们探讨了LSD1的直接药理学抑制是否可以在MSCs中灌输类似于UM171a的APC样功能.用LSD1抑制剂tranylcypromine(TC)处理MSC引起双链(ds)RNA应激反应及其相关的响应元件,包括模式识别受体(PRR),I型干扰素(IFN),和IFN刺激基因(ISGs)。最终结果是H2-Kb的表达增强,和增加的细胞表面肽:MHCI复合物的稳定性。因此,TC处理的MSCs有效刺激CD8T细胞活化,并在预防性疫苗接种的情况下引发针对EG.7T细胞淋巴瘤的有效抗肿瘤反应。总之,我们的研究结果揭示了一种新的药理学方案,通过该方案,在MSCs中靶向LSD1可引发APC样功能,该功能可在未来基于MSC的抗癌疫苗的设计中轻松利用.
    Mesenchymal stromal cells (MSCs) are commonly known for their immune-suppressive abilities. However, our group provided evidence that it is possible to convert MSCs into potent antigen presenting cells (APCs) using either genetic engineering or pharmacological means. Given the capacity of UM171a to trigger APC-like function in MSCs, and the recent finding that this drug may modulate the epigenome by inhibiting the lysine-specific demethylase 1 (LSD1), we explored whether the direct pharmacological inhibition of LSD1 could instill APC-like functions in MSCs akin to UM171a. The treatment of MSCs with the LSD1 inhibitor tranylcypromine (TC) elicits a double-stranded (ds)RNA stress response along with its associated responsive elements, including pattern recognition receptors (PRRs), Type-I interferon (IFN), and IFN-stimulated genes (ISGs). The net outcome culminates in the enhanced expression of H2-Kb, and an increased stability of the cell surface peptide: MHCI complexes. As a result, TC-treated MSCs stimulate CD8 T-cell activation efficiently, and elicit potent anti-tumoral responses against the EG.7 T-cell lymphoma in the context of prophylactic vaccination. Altogether, our findings reveal a new pharmacological protocol whereby targeting LSD1 in MSCs elicits APC-like capabilities that could be easily exploited in the design of future MSC-based anti-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
    间充质基质细胞(MSCs)由于其良好的组织修复能力,已被广泛用于临床。然而,它们在细胞疫苗接种领域也有希望,因为在特定的治疗方案下,它们可以表现为响应干扰素(IFN)-γ治疗的条件抗原呈递细胞.这表明MSC的免疫功能可以在药理学上调节。鉴于激动剂嘧啶吲哚衍生物UM171a在人造血祖细胞中触发各种抗原呈递相关基因表达的能力,我们探讨了UM171a作为一种药理学方法在MSCs中导入和/或促进抗原呈递的潜在用途.
    除了完成一系列基于流式细胞术的表型分析,使用SIINFEKL特异性T细胞克隆B3Z进行了几种功能性抗原呈递测定.抗氧化剂和电子传递链抑制剂也用于破译UM171a在MSCs中的作用模式。最后,使用免疫活性C57BL/6小鼠预先建立的同基因EG.7T细胞淋巴瘤,在治疗性疫苗接种的背景下评估了UM171a治疗的MSCs的效力.
    用UM171a处理MSC引发H2-Kb细胞表面水平的有效增加以及抗原交叉呈递能力的获得。机械上,这种效应发生在对UM171a介导的线粒体衍生活性氧产生的反应中,因为它们使用抗氧化剂或抗霉素-A中和作用减轻了MSCs交叉呈递抗原的能力.免疫原性卵清蛋白来源的SIINFEKL肽的加工和呈递是由响应于UM171a触发的氧化应激的Psmb8基因的从头表达引起的。当在治疗性疫苗接种的背景下评估它们的抗肿瘤特性时,UM171a处理的MSC施用至具有预先建立的T细胞淋巴瘤的免疫活性小鼠,控制肿瘤生长,导致40%存活率,而不需要额外的支持疗法和/或标准护理。
    总之,我们的发现揭示了UM171a的一种新的免疫相关功能,并清楚地暗示了UM171a介导的ROS诱导与MSCs的抗原交叉呈递之间的直接联系.UM171a处理在不使用IFN-γ的情况下调节MSC成为抗原呈递细胞的事实开辟了一条新的研究路线,以寻找能够将免疫抑制MSC转化为易于适应疫苗接种的细胞工具的其他试剂。
    Mesenchymal stromal cells (MSCs) have been extensively used in the clinic due to their exquisite tissue repair capacity. However, they also hold promise in the field of cellular vaccination as they can behave as conditional antigen presenting cells in response to interferon (IFN)-gamma treatment under a specific treatment regimen. This suggests that the immune function of MSCs can be pharmacologically modulated. Given the capacity of the agonist pyrimido-indole derivative UM171a to trigger the expression of various antigen presentation-related genes in human hematopoietic progenitor cells, we explored the potential use of UM171a as a means to pharmacologically instill and/or promote antigen presentation by MSCs.
    Besides completing a series of flow-cytometry-based phenotypic analyses, several functional antigen presentation assays were conducted using the SIINFEKL-specific T-cell clone B3Z. Anti-oxidants and electron transport chain inhibitors were also used to decipher UM171a\'s mode of action in MSCs. Finally, the potency of UM171a-treated MSCs was evaluated in the context of therapeutic vaccination using immunocompetent C57BL/6 mice with pre-established syngeneic EG.7T-cell lymphoma.
    Treatment of MSCs with UM171a triggered potent increase in H2-Kb cell surface levels along with the acquisition of antigen cross-presentation abilities. Mechanistically, such effects occurred in response to UM171a-mediated production of mitochondrial-derived reactive oxygen species as their neutralization using anti-oxidants or Antimycin-A mitigated MSCs\' ability to cross-present antigens. Processing and presentation of the immunogenic ovalbumin-derived SIINFEKL peptide was caused by de novo expression of the Psmb8 gene in response to UM171a-triggered oxidative stress. When evaluated for their anti-tumoral properties in the context of therapeutic vaccination, UM171a-treated MSC administration to immunocompetent mice with pre-established T-cell lymphoma controlled tumor growth resulting in 40% survival without the need of additional supportive therapy and/or standard-of-care.
    Altogether, our findings reveal a new immune-related function for UM171a and clearly allude to a direct link between UM171a-mediated ROS induction and antigen cross-presentation by MSCs. The fact that UM171a treatment modulates MSCs to become antigen-presenting cells without the use of IFN-gamma opens-up a new line of investigation to search for additional agents capable of converting immune-suppressive MSCs to a cellular tool easily adaptable to vaccination.
    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
    我们最近开发了一种用于癌症治疗的基于单核细胞的细胞疫苗平台。与传统的利用单核细胞作为前体以产生用于疫苗接种目的的树突状细胞(DC)相反,我们发现没有分化过程的新鲜分离的单核细胞可以装载肿瘤抗原(Ag),并引发强大的抗肿瘤细胞毒性T淋巴细胞(CTL)反应。在这一章中,我们描述了准备的方法,administrate,并评估鼠Ly-6Chi单核细胞为基础的细胞疫苗的治疗效果。这包括隔离程序,纯度测定,Ag负载量,给予骨髓(BM)衍生的单核细胞,以及通过检测小鼠黑色素瘤模型中Ag特异性CD8+T细胞扩增和抗肿瘤反应来确定疫苗效力的方法。作为疫苗平台,未分化的单核细胞可以容易地适应具有多种靶抗原的不同肿瘤模型。本文描述的方法旨在促进基于单核细胞的疫苗接种作为癌症免疫治疗策略的临床前研究。
    We recently developed a monocyte-based cellular vaccine platform for cancer treatment. In contrast to the traditional utilization of monocytes as precursors to generate dendritic cells (DC) for vaccination purposes, we find that freshly isolated monocytes with no differentiation process can be loaded with tumor antigens (Ag) and trigger robust antitumor cytotoxic T lymphocyte (CTL) responses. In this chapter, we describe methods to prepare, administer, and evaluate murine Ly-6Chi monocyte-based cellular vaccines for their therapeutic efficacy. This includes procedures for isolation, purity determination, Ag loading, administration of bone marrow (BM)-derived monocytes, as well as methods to determine vaccine efficacy through the examination of Ag-specific CD8+ T cell expansion and antitumor responses in murine melanoma models. As a vaccine platform, undifferentiated monocytes can be easily adapted to different tumor models with a multitude of target antigens. The method described here seeks to facilitate preclinical research of monocyte-based vaccination as a strategy for cancer immunotherapy.
    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
    Cancer vaccines that utilize patient antigen-presenting cells to fight their own tumors have shown exciting promise in many preclinical studies, but have proven quite challenging to translate to clinical feasibility. Dendritic cells have typically been the cell of choice for such vaccine platforms, due to their ability to endocytose antigens nonspecifically, and their expression of multiple surface molecules that enhance antigen presentation. However, dendritic cells are present in low numbers in human peripheral blood and must be matured in culture before use in vaccines. Mature B lymphocytes, in contrast, are relatively abundant in peripheral blood, and can be quickly activated and expanded in overnight cultures. We devised an optimal stimulation cocktail that engages the B cell antigen receptor, CD40, TLR4 and TLR7, to activate B cells to present antigens from lysates of the recipient\'s tumor cells, precluding the need for known tumor antigens. This B cell vaccine (Bvac) improved overall survival from B16F1 melanoma challenge, as well as reduced tumor size and increased time to tumor appearance. Bvac upregulated B cell antigen presentation molecules, stimulated activation of both CD4+ and CD8+ T cells, and induced T cell migration. Bvac provides an alternative cellular vaccine strategy that has considerable practical advantages for translation to clinical settings.
    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
    Gliomas, especially glioblastomas, represent one of the most aggressive and difficult-to-treat human brain tumors. In the last few decades, clinical immunotherapy has been developed and has provided exceptional achievements in checkpoint inhibitors and vaccines for cancer treatment. Immunization with cellular vaccines has the advantage of containing specific antigens and acceptable safety to potentially improve cancer therapy. Based on T cells, dendritic cells (DC), tumor cells and natural killer cells, the safety and feasibility of cellular vaccines have been validated in clinical trials for glioma treatment. For TAA engineered T cells, therapy mainly uses chimeric antigen receptors (IL13Rα2, EGFRvIII and HER2) and DNA methylation-induced technology (CT antigen) to activate the immune response. Autologous dendritic cells/tumor antigen vaccine (ADCTA) pulsed with tumor lysate and peptides elicit antigen-specific and cytotoxic T cell responses in patients with malignant gliomas, while its pro-survival effect is biased. Vaccinations using autologous tumor cells modified with TAAs or fusion with fibroblast cells are characterized by both effective humoral and cell-mediated immunity. Even though few therapeutic effects have been observed, most of this therapy showed safety and feasibility, asking for larger cohort studies and better guidelines to optimize cellular vaccine efficiency in anti-glioma therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Mismatch-repair deficiency (MMR-D) is closely linked to hypermutation and accordingly, high immunogenicity. MMR-D-related tumors thus constitute ideal vaccination targets for both therapeutic and prophylactic approaches. Herein, the prophylactic and therapeutic impact of a cellular vaccine on tumor growth and tumor-immune microenvironment was studied in a murine MLH1-/- knockout mouse model. Prophylactic application of the lysate (+/- CpG ODN 1826) delayed tumor development, accompanied by increased levels of circulating T cell numbers. Therapeutic application of the vaccine prolonged overall survival (median time: 11.5 (lysate) and 12 weeks (lysate + CpG ODN) vs. 3 weeks (control group), respectively) along with reduced tumor burden, as confirmed by PET/CT imaging and immune stimulation (increased CD3+CD8+ T - and NK cell numbers, reduced levels of TIM-3+ cells in both treatment groups). Coding microsatellite analysis of MMR-D-related target genes revealed increased mutational load upon vaccination (total mutation frequency within 28 genes: 28.6% vaccine groups vs. 14.9% control group, respectively). Reactive immune cells recognized autologous tumor cells, but also NK cells target YAC-1 in IFNγ ELISpot and, even more importantly, in functional kill assays. Assessment of tumor microenvironment revealed infiltration of CD8+ T-cells and granulocytes, but also upregulation of immune checkpoint molecules (LAG-3, PD-L1). The present study is the first reporting in vivo results on a therapeutic cellular MMR-D vaccine. Vaccination-induced prolonged survival was achieved in a clinically-relevant mouse model for MMR-D-related diseases by long-term impairment of tumor growth and this could be attributed to re-activated immune responses.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    The success of active immunotherapies in the prevention of many infectious diseases over the course of over 200 years has lead scientists to wonder if the same principles could be applied to cancer. Antigen-specific active immunotherapies for the treatment of cancer have been researched for over two decades, however, the overwhelming majority of these studies have failed to stimulate robust clinical responses. It is clear that current active immunotherapy research should incorporate methods to increase the immunostimulatory capacity of these therapies. To directly address this need, we propose the addition of the immunostimulatory heat shock proteins (HSPs) to active immunotherapeutic strategies to augment their efficacy. Heat shock proteins are a family of highly conserved intracellular chaperone proteins, and are the most abundant family proteins inside cells. This ubiquity, and their robust immunostimulatory capacity, points to their importance in regulation of intracellular processes and, therefore, indicators of loss of cellular integrity if found extracellularly. Thus, we emphasize the importance of taking into consideration the location of vaccine-derived HSP/tumor-antigen complexes when designing active immunotheraputic strategies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号