Cowpea mosaic virus

  • 文章类型: Journal Article
    许多病毒基因组编码促进感染的蛋白酶。植物识别病毒蛋白酶的分子机制在很大程度上尚未被探索。使用Vignaunguiculata和cow豆花叶病毒(CPMV)的系统,我们鉴定了一种与CPMV编码的24KPro相互作用的of豆脂质转移蛋白(LTP1),一种半胱氨酸蛋白酶,但不具有酶活性的突变体24KPro(C166A)。生化分析表明,LTP1抑制了外壳蛋白前体大外壳蛋白-小外壳蛋白的24KPro蛋白水解裂解。一过性LTP1在cow豆中的过表达减少了CPMV感染,而RNA干扰介导的LTP1沉默增加了cow豆中CPMV的积累。LTP1主要位于未感染植物细胞的质外体中,在CPMV感染后,大部分LTP1被重新定位到细胞内区室,包括叶绿体.此外,在稳定的LTP1转基因烟草植物中,LTP1抑制大豆花叶病毒(SMV)核包涵有蛋白酶活性,SMV积累量显著降低。我们建议cow豆LTP1通过直接抑制病毒半胱氨酸蛋白酶活性来抑制CPMV和SMV的积累。
    Many virus genomes encode proteases that facilitate infection. The molecular mechanism of plant recognition of viral proteases is largely unexplored. Using the system of Vigna unguiculata and cowpea mosaic virus (CPMV), we identified a cowpea lipid transfer protein (LTP1) which interacts with CPMV-encoded 24KPro, a cysteine protease, but not with the enzymatically inactive mutant 24KPro(C166A). Biochemical assays showed that LTP1 inhibited 24KPro proteolytic cleavage of the coat protein precursor large coat protein-small coat protein. Transient overexpression of LTP1 in cowpea reduced CPMV infection, whereas RNA interference-mediated LTP1 silencing increased CPMV accumulation in cowpea. LTP1 is mainly localized in the apoplast of uninfected plant cells, and after CPMV infection, most of the LTP1 is relocated to intracellular compartments, including chloroplast. Moreover, in stable LTP1-transgenic Nicotiana benthamiana plants, LTP1 repressed soybean mosaic virus (SMV) nuclear inclusion a protease activity, and accumulation of SMV was significantly reduced. We propose that cowpea LTP1 suppresses CPMV and SMV accumulation by directly inhibiting viral cysteine protease activity.
    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
    卵巢癌(OvCa)是妇科恶性肿瘤死亡的主要原因,通常表现为产生转移的腹膜内球体,腹水,和免疫抑制肿瘤微环境。在这项研究中,我们使用OvCa腹膜球体的体外模型,探索了cow豆花叶病毒(CPMV)作为佐剂免疫治疗剂的免疫调节特性。先前的发现强调了肿瘤内CPMV对小鼠肿瘤模型中OvCa的有效功效。利用基于数字光处理(DLP)的生物打印对材料沉积和细胞图案化的精确控制,我们使用明胶甲基丙烯酸酯(GelMA)构建了OvCa-巨噬细胞球体来模拟腹膜球体,胶原衍生的可光聚合生物材料以模拟细胞外基质。CPMV治疗后,生物打印的球体显示抑制巨噬细胞激活介导的OvCa进展。我们的分析表明CPMV调节和激活巨噬细胞以诱导OvCa细胞杀伤和恢复正常的细胞-细胞连接。这项研究加深了我们对OvCa背景下CPMV肿瘤内免疫治疗机制的理解。这项研究还强调了通过DLP生物打印使用高通量组织模型研究免疫疗法的潜力。
    Ovarian cancer (OvCa) is a leading cause of mortality among gynecological malignancies and usually manifests as intraperitoneal spheroids that generate metastases, ascites, and an immunosuppressive tumor microenvironment. In this study, we explore the immunomodulatory properties of cowpea mosaic virus (CPMV) as an adjuvant immunotherapeutic agent using an in vitro model of OvCa peritoneal spheroids. Previous findings highlighted the potent efficacy of intratumoral CPMV against OvCa in mouse tumor models. Leveraging the precision control over material deposition and cell patterning afforded by digital-light-processing (DLP) based bioprinting, we constructed OvCa-macrophage spheroids to mimic peritoneal spheroids using gelatin methacrylate (GelMA), a collagen-derived photopolymerizable biomaterial to mimic the extracellular matrix. Following CPMV treatment, bioprinted spheroids exhibited inhibited OvCa progression mediated by macrophage activation. Our analysis indicates that CPMV regulates and activates macrophage to both induce OvCa cell killing and restore normal cell-cell junctions. This study deepened our understanding of the mechanism of CPMV intratumoral immunotherapy in the setting of OvCa. This study also highlights the potential of studying immunotherapies using high throughput tissue models via DLP bioprinting.
    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
    癌症的免疫检查点疗法(ICT)可以产生巨大的临床反应;然而,这些可能只在少数患者中观察到。这些反应可进一步受到随后的疾病复发和抗性的限制。正在开发组合免疫疗法策略以克服这些限制。我们以前曾报道过瘤内cow豆花叶病毒免疫疗法(CPMVIIT)和ICT方法的联合功效增强。淋巴细胞活化基因-3(LAG-3)是下一代抑制性免疫检查点,其在多个免疫细胞亚群中具有广泛表达。其表达在活化的T细胞上增加并有助于T细胞耗尽。我们在黑素瘤的小鼠模型中观察到CPMVIIT和抗LAG-3联合治疗的功效增强。Further,发现瘤内CPMV施用后TME内的LAG-3表达增加。CPMVIIT与LAG-3抑制的整合具有通过同时诱导全面的抗肿瘤免疫反应来改善治疗结果的显着潜力。增强局部免疫激活,减轻T细胞衰竭。
    Immune checkpoint therapy (ICT) for cancer can yield dramatic clinical responses; however, these may only be observed in a minority of patients. These responses can be further limited by subsequent disease recurrence and resistance. Combination immunotherapy strategies are being developed to overcome these limitations. We have previously reported enhanced efficacy of combined intratumoral cowpea mosaic virus immunotherapy (CPMV IIT) and ICT approaches. Lymphocyte-activation gene-3 (LAG-3) is a next-generation inhibitory immune checkpoint with broad expression across multiple immune cell subsets. Its expression increases on activated T cells and contributes to T cell exhaustion. We observed heightened efficacy of a combined CPMV IIT and anti-LAG-3 treatment in a mouse model of melanoma. Further, LAG-3 expression was found to be increased within the TME following intratumoral CPMV administration. The integration of CPMV IIT with LAG-3 inhibition holds significant potential to improve treatment outcomes by concurrently inducing a comprehensive anti-tumor immune response, enhancing local immune activation, and mitigating T cell exhaustion.
    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
    犬炎性乳腺癌(IMC)是一种高度侵袭性和致命性的癌症,作为一种有价值的动物模型,炎性乳腺癌(IBC),都缺乏有效的治疗方法。肿瘤内免疫疗法(IT-IT)与空cow豆花叶病毒(eCPMV)纳米颗粒已显示出有希望的结果,证明肿瘤大小减小,更长的存活率,提高了生活质量。这项研究比较了接受eCPMVIT-IT和药物治疗(MT)的IMC与单独接受MT的雌性狗的肿瘤样品的转录组学谱。转录组学分析,基因表达谱,信号通路,使用NanoStringTechnologies使用犬免疫肿瘤学小组评估了来自四只eCPMV处理的IMC犬和四只接受MT处理的IMC犬的样品中免疫细胞群的细胞类型谱分析。比较分析揭示了处理和未处理样品之间的34个差异表达基因。参与中性粒细胞募集和活化的5个基因(CXCL8、S100A9、CCL20、IL6和PTGS2)在处理过的样品中上调,与IL17信号通路有关。细胞类型谱分析显示,eCPMV治疗后,肿瘤微环境中的中性粒细胞数量显着增加。这些发现强调了嗜中性粒细胞在eCPMVIT-IT介导的抗肿瘤反应中的作用,并建议eCPMV作为IBC/IMC的新型治疗方法。
    Canine inflammatory mammary cancer (IMC) is a highly aggressive and lethal cancer in dogs serving as a valuable animal model for its human counterpart, inflammatory breast cancer (IBC), both lacking effective therapies. Intratumoral immunotherapy (IT-IT) with empty cowpea mosaic virus (eCPMV) nanoparticles has shown promising results, demonstrating a reduction in tumor size, longer survival rates, and improved quality of life. This study compares the transcriptomic profiles of tumor samples from female dogs with IMC receiving eCPMV IT-IT and medical therapy (MT) versus MT alone. Transcriptomic analyses, gene expression profiles, signaling pathways, and cell type profiling of immune cell populations in samples from four eCPMV-treated dogs with IMC and four dogs with IMC treated with MT were evaluated using NanoString Technologies using a canine immune-oncology panel. Comparative analyses revealed 34 differentially expressed genes between treated and untreated samples. Five genes (CXCL8, S100A9, CCL20, IL6, and PTGS2) involved in neutrophil recruitment and activation were upregulated in the treated samples, linked to the IL17-signaling pathway. Cell type profiling showed a significant increase in neutrophil populations in the tumor microenvironment after eCPMV treatment. These findings highlight the role of neutrophils in the anti-tumor response mediated by eCPMV IT-IT and suggest eCPMV as a novel therapeutic approach for IBC/IMC.
    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
    缺乏评估新型药物的最佳模型正在延迟针对人类乳腺癌(BC)的有效免疫疗法的开发。在这项前瞻性开放标签研究中,我们应用新辅助瘤内免疫疗法与空cow豆花叶病毒样颗粒(eCPMV)对11只诊断为犬乳腺癌(CMC)的伴侣犬,类似于人类BC的自发性肿瘤。我们发现,两次新辅助肿瘤内注射eCPMV可导致所有患者的注射肿瘤以及位于注射犬同侧和对侧乳腺链中的非注射肿瘤的肿瘤减少。肿瘤缩小与临床分期无关,肿瘤大小,组织病理学分级,和肿瘤分子亚型。注射肿瘤的基于RNA-seq的分析表明DNA复制活性降低,肿瘤微环境中激活的树突状细胞浸润增加。免疫组织化学分析显示肿瘤内中性粒细胞显著增加,T和B淋巴细胞,和浆细胞。eCPMV肿瘤内免疫治疗显示抗肿瘤疗效,无任何不良反应。这种新型免疫疗法具有改善人类BC患者预后的潜力。
    The lack of optimal models to evaluate novel agents is delaying the development of effective immunotherapies against human breast cancer (BC). In this prospective open label study, we applied neoadjuvant intratumoral immunotherapy with empty cowpea mosaic virus-like particles (eCPMV) to 11 companion dogs diagnosed with canine mammary cancer (CMC), a spontaneous tumor resembling human BC. We found that two neoadjuvant intratumoral eCPMV injections resulted in tumor reduction in injected tumors in all patients and in noninjected tumors located in the ipsilateral and contralateral mammary chains of injected dogs. Tumor reduction was independent of clinical stage, tumor size, histopathologic grade, and tumor molecular subtype. RNA-seq-based analysis of injected tumors indicated a decrease in DNA replication activity and an increase in activated dendritic cell infiltration in the tumor microenvironment. Immunohistochemistry analysis demonstrated significant intratumoral increases in neutrophils, T and B lymphocytes, and plasma cells. eCPMV intratumoral immunotherapy demonstrated antitumor efficacy without any adverse effects. This novel immunotherapy has the potential for improving outcomes for human BC 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
    植物病毒纳米颗粒可作为药物载体,成像试剂,疫苗携带者,瘤内原位癌疫苗制剂中的免疫佐剂。一个例子是cow豆花叶病毒(CPMV),一种无包膜病毒,具有二分正链RNA基因组,每种RNA分别包装到相同的蛋白质衣壳中。根据它们的密度差异,例如,携带RNA-1(6kb)的组分表示为底部(B)组分或携带RNA-2(3.5kb)表示为中间(M)组分可以彼此分离并与顶部(T)组分分离。没有任何RNA。以前的临床前小鼠研究和犬癌试验使用CPMV的混合群体(含有B,M,和T分量),因此,目前尚不清楚粒子类型是否在其功效上有所不同。已知CPMVRNA基因组通过激活TLR7而有助于免疫刺激。为了确定具有不同大小和不相关序列的两个RNA基因组是否引起不同的免疫刺激,我们比较了B和M组分和普通CPMV在体外和小鼠癌症模型中的治疗效果。我们发现分离的B和M颗粒的行为与混合的CPMV相似,激活先天性免疫细胞以诱导促炎细胞因子如IFNα的分泌,IFNγ,IL-6和IL-12,同时抑制免疫抑制细胞因子如TGF-β和IL-10。在黑色素瘤和结肠癌的小鼠模型中,混合和分离的CPMV颗粒均显着降低了肿瘤的生长,延长了生存期,但没有显着差异。这表明,即使B颗粒具有比M颗粒多40%的RNA,特定RNA基因组也类似地刺激免疫系统;每种CPMV颗粒类型可以用作针对癌症的有效佐剂,具有与天然混合CPMV相同的功效。从平移的角度来看,B或M组分相对于混合CPMV制剂的使用提供了这样的优点,即单独分离的B或M对植物是非传染性的,因此提供了农艺安全性。
    Plant virus nanoparticles can be used as drug carriers, imaging reagents, vaccine carriers, and immune adjuvants in the formulation of intratumoral in situ cancer vaccines. One example is the cowpea mosaic virus (CPMV), a nonenveloped virus with a bipartite positive-strand RNA genome with each RNA packaged separately into identical protein capsids. Based on differences in their densities, the components carrying RNA-1 (6 kb) denoted as the bottom (B) component or carrying RNA-2 (3.5 kb) denoted as the middle (M) component can be separated from each other and from a top (T) component, which is devoid of any RNA. Previous preclinical mouse studies and canine cancer trials used mixed populations of CPMV (containing B, M, and T components), so it is unclear whether the particle types differ in their efficacies. It is known that the CPMV RNA genome contributes to immunostimulation by activation of TLR7. To determine whether the two RNA genomes that have different sizes and unrelated sequences cause different immune stimulation, we compared the therapeutic efficacies of B and M components and unfractionated CPMV in vitro and in mouse cancer models. We found that separated B and M particles behaved similarly to the mixed CPMV, activating innate immune cells to induce the secretion of pro-inflammatory cytokines such as IFNα, IFNγ, IL-6, and IL-12, while inhibiting immunosuppressive cytokines such as TGF-β and IL-10. In murine models of melanoma and colon cancer, the mixed and separated CPMV particles all significantly reduced tumor growth and prolonged survival with no significant difference. This shows that the specific RNA genomes similarly stimulate the immune system even though B particles have 40% more RNA than M particles; each CPMV particle type can be used as an effective adjuvant against cancer with the same efficacy as native mixed CPMV. From a translational point of view, the use of either B or M component vs the mixed CPMV formulation offers the advantage that separated B or M alone is noninfectious toward plants and thus provides agronomic safety.
    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
    Cw豆花叶病毒(CPMV)已被开发为用于癌症免疫疗法的有前途的纳米平台技术;当用作原位疫苗时,CPMV表现出强效,系统性,和持久的功效。虽然CPMV对哺乳动物没有传染性,它对豆类有传染性;因此,需要解决农艺安全问题,以扩大CPMV的翻译应用。含RNA的制剂优于无RNA的病毒样颗粒,因为RNA和蛋白质,每个,有助于CPMV的有效抗肿瘤功效。我们先前已经优化了灭活方法以开发含有RNA但对植物没有传染性的CPMV。我们确定灭活的CPMV与未经处理的CPMV相比具有降低的功效,原生CPMV。然而,没有对天然CPMV和不同灭活形式的CPMV进行系统比较。因此,在这项研究中,我们直接比较了CPMV的治疗效果和免疫激活机制,紫外线-(UV-),和福尔马林(形式)灭活的CPMV来解释不同的功效。在B16F10黑色素瘤小鼠肿瘤模型中,形式-CPMV抑制肿瘤生长,延长存活(比较CPMV和形式-CPMV没有统计学差异)。相比之下,UV-CPMV显著抑制肿瘤生长,但不如形式-CPMV或CPMV。UV-CPMV的治疗功效降低可以通过RNA的交联程度和聚集状态来解释。这使得它无法通过Toll样受体(TLR)7/8来激活免疫应答。机理研究表明,UV-CPMV的高度聚集状态比形式-CPMV制剂更能抑制TLR7信号传导。减少白细胞介素-6(IL-6)和干扰素-α(IFN-α)的分泌,与TLR7信号相关的细胞因子。这些发现支持形式-CPMV作为非感染性免疫治疗剂的翻译发展。
    Cowpea mosaic virus (CPMV) has been developed as a promising nanoplatform technology for cancer immunotherapy; when applied as in situ vaccine, CPMV exhibits potent, systemic, and durable efficacy. While CPMV is not infectious to mammals, it is infectious to legumes; therefore, agronomic safety needs to be addressed to broaden the translational application of CPMV. RNA-containing formulations are preferred over RNA-free virus-like particles because the RNA and protein, each, contribute to CPMV\'s potent antitumor efficacy. We have previously optimized inactivation methods to develop CPMV that contains RNA but is not infectious to plants. We established that inactivated CPMV has reduced efficacy compared to untreated, native CPMV. However, a systematic comparison between native CPMV and different inactivated forms of CPMV was not done. Therefore, in this study, we directly compared the therapeutic efficacies and mechanisms of immune activation of CPMV, ultraviolet- (UV-), and formalin (Form)-inactivated CPMV to explain the differential efficacies. In a B16F10 melanoma mouse tumor model, Form-CPMV suppressed the tumor growth with prolonged survival (there were no statistical differences comparing CPMV and Form-CPMV). In comparison, UV-CPMV inhibited tumor growth significantly but not as well as Form-CPMV or CPMV. The reduced therapeutic efficacy of UV-CPMV is explained by the degree of cross-linking and aggregated state of the RNA, which renders it inaccessible for sensing by Toll-like receptor (TLR) 7/8 to activate immune responses. The mechanistic studies showed that the highly aggregated state of UV-CPMV inhibited TLR7 signaling more so than for the Form-CPMV formulation, reducing the secretion of interleukin-6 (IL-6) and interferon-α (IFN-α), cytokines associated with TLR7 signaling. These findings support the translational development of Form-CPMV as a noninfectious immunotherapeutic agent.
    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
    Cw豆花叶病毒(CPMV)是一种核蛋白纳米颗粒,在瘤内给药时可用作高效的免疫调节剂,并用作原位疫苗。CPMV原位疫苗接种重塑了肿瘤微环境,并引发了高度有效的,系统性,和持久的抗肿瘤免疫应答对治疗和未治疗,远处转移部位(远隔效应)。在多种肿瘤小鼠模型中证明了有效的疗效,最重要的是,在犬癌症患者自发性肿瘤。数据表明抗CPMV抗体的存在不是中和的,并且实际上调理作用导致增强的功效。植物病毒是食物链的一部分,但迄今为止,没有关于人类接触CPMV的信息。因此,检测患者血清中是否存在抗CPMV的免疫球蛋白,事实上,>50%的去鉴定患者样品检测为CPMV抗体阳性。为了更广泛地了解植物病毒在人类中的暴露和免疫原性,我们还测试了血清中抗烟草花叶病毒的抗体(>90%的患者检测呈阳性),马铃薯病毒X(<20%的患者检测呈阳性),和cow豆褪绿斑驳病毒(未检测到抗体)。Further,分析患者血清中是否存在针对大肠杆菌噬菌体Qβ的抗体,目前正在进行原位疫苗接种临床试验的平台技术;我们发现60%的患者存在抗Qβ抗体.因此,数据表明人类暴露于CPMV和其他植物病毒和噬菌体。接下来,我们认为要解决农艺安全问题;即,我们检查了肿瘤内治疗和口服灌胃(模拟食物消耗)后CPMV的命运。因为使用了实时CPMV,一个重要的问题是,是否有任何证据表明小鼠或患者的感染性颗粒脱落。CPMV对哺乳动物是非传染性的;然而,它对包括黑眼豌豆和其他豆类在内的植物具有传染性。荷瘤小鼠和健康小鼠的生物分布数据表明,几乎没有从肿瘤中浸出,并通过网状内皮系统清除,然后进行胆汁排泄。虽然有证据表明粪便中的RNA脱落,当植物受到粪便提取物的攻击时,没有感染颗粒的证据,从而表明农艺安全。这些数据一起帮助CPMV作为癌症免疫疗法的候选药物的转化发展。
    Cowpea mosaic virus (CPMV) is a nucleoprotein nanoparticle that functions as a highly potent immunomodulator when administered intratumorally and is used as an in situ vaccine. CPMV in situ vaccination remodels the tumor microenvironment and primes a highly potent, systemic, and durable antitumor immune response against the treated and untreated, distant metastatic sites (abscopal effect). Potent efficacy was demonstrated in multiple tumor mouse models and, most importantly, in canine cancer patients with spontaneous tumors. Data indicate that presence of anti-CPMV antibodies are not neutralizing and that in fact opsonization leads to enhanced efficacy. Plant viruses are part of the food chain, but to date, there is no information on human exposure to CPMV. Therefore, patient sera were tested for the presence of immunoglobulins against CPMV, and indeed, >50% of deidentified patient samples tested positive for CPMV antibodies. To get a broader sense of plant virus exposure and immunogenicity in humans, we also tested sera for antibodies against tobacco mosaic virus (>90% patients tested positive), potato virus X (<20% patients tested positive), and cowpea chlorotic mottle virus (no antibodies were detected). Further, patient sera were analyzed for the presence of antibodies against the coliphage Qβ, a platform technology currently undergoing clinical trials for in situ vaccination; we found that 60% of patients present with anti-Qβ antibodies. Thus, data indicate human exposure to CPMV and other plant viruses and phages. Next, we thought to address agronomical safety; i.e., we examined the fate of CPMV after intratumoral treatment and oral gavage (to mimic consumption by food). Because live CPMV is used, an important question is whether there is any evidence of shedding of infectious particles from mice or patients. CPMV is noninfectious toward mammals; however, it is infectious toward plants including black-eyed peas and other legumes. Biodistribution data in tumor-bearing and healthy mice indicate little leaching from tumors and clearance via the reticuloendothelial system followed by biliary excretion. While there was evidence of shedding of RNA in stool, there was no evidence of infectious particles when plants were challenged with stool extracts, thus indicating agronomical safety. Together these data aid the translational development of CPMV as a drug candidate for cancer immunotherapy.
    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
    尽管腹腔镜手术联合新辅助和辅助治疗取得了进展,结肠癌管理在肿瘤学中仍然具有挑战性。局部或远处器官中癌组织的复发(转移)是结肠癌治疗中的主要问题。疫苗和免疫疗法有望通过刺激免疫系统来预防癌症复发。我们和其他人已经证明来自植物病毒的纳米粒子,如Cw豆花叶病毒(CPMV)纳米颗粒,是癌症疫苗的有效免疫佐剂,并在治疗或预防肿瘤中用作免疫刺激剂。虽然对哺乳动物没有传染性,CPMV通过模式识别受体(PRR)的识别激活先天免疫系统。虽然CPMV的颗粒结构对于显著的免疫激活是必不可少的,蛋白质结构使CPMV在体内降解;因此,CPMV免疫疗法需要重复注射以获得最佳结果。然而,从临床角度来看,频繁的腹膜内(IP)注射并不是最佳的,并且由于IP给药需要住院治疗,可能会恶化患者的生活质量。为了克服重复IP注射的需要,我们在可注射壳聚糖/甘油磷酸酯(GP)水凝胶制剂中加载CPMV纳米颗粒,以它们的缓释潜力为特征,并评估了水凝胶中CPMV单剂量与可溶性CPMV的抗肿瘤预防功效(单次和初次加强给药)。使用荧光标记的水凝胶CPMV制剂,体内释放数据表明,在3周的研究期内,水凝胶制剂的单次IP注射产生了提供完整CPMV的凝胶储库,而可溶性CPMV仅持续一周。在BALB/c小鼠中,在CT26IP攻击后,用可溶性CPMV促进CPMV-in-hygar制剂的IP施用以组合立即和持续的免疫激活显著抑制结肠癌生长。观察到的抗肿瘤功效表明,CPMV可以在壳聚糖/GP水凝胶中配制,以实现延长的免疫刺激作用,作为针对结肠癌复发的单剂量免疫疗法。本发现说明了可注射水凝胶技术适应植物病毒纳米颗粒以促进有效抗肿瘤免疫疗法的转化发展的潜力。
    Despite advances in laparoscopic surgery combined with neoadjuvant and adjuvant therapy, colon cancer management remains challenging in oncology. Recurrence of cancerous tissue locally or in distant organs (metastasis) is the major problem in colon cancer management. Vaccines and immunotherapies hold promise in preventing cancer recurrence through stimulation of the immune system. We and others have shown that nanoparticles from plant viruses, such as cowpea mosaic virus (CPMV) nanoparticles, are potent immune adjuvants for cancer vaccines and serve as immunostimulatory agents in the treatment or prevention of tumors. While being noninfectious toward mammals, CPMV activates the innate immune system through recognition by pattern recognition receptors (PRRs). While the particulate structure of CPMV is essential for prominent immune activation, the proteinaceous architecture makes CPMV subject to degradation in vivo; thus, CPMV immunotherapy requires repeated injections for optimal outcome. Frequent intraperitoneal (IP) injections however are not optimal from a clinical point of view and can worsen the patient\'s quality of life due to the hospitalization required for IP administration. To overcome the need for repeated IP injections, we loaded CPMV nanoparticles in injectable chitosan/glycerophosphate (GP) hydrogel formulations, characterized their slow-release potential, and assessed the antitumor preventative efficacy of CPMV-in-hydrogel single dose versus soluble CPMV (single and prime-boost administration). Using fluorescently labeled CPMV-in-hydrogel formulations, in vivo release data indicated that single IP injection of the hydrogel formulation yielded a gel depot that supplied intact CPMV over the study period of 3 weeks, while soluble CPMV lasted only for one week. IP administration of the CPMV-in-hydrogel formulation boosted with soluble CPMV for combined immediate and sustained immune activation significantly inhibited colon cancer growth after CT26 IP challenge in BALB/c mice. The observed antitumor efficacy suggests that CPMV can be formulated in a chitosan/GP hydrogel to achieve prolonged immunostimulatory effects as single-dose immunotherapy against colon cancer recurrence. The present findings illustrate the potential of injectable hydrogel technology to accommodate plant virus nanoparticles to boost the translational development of effective antitumor 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
    用于癌症免疫疗法的原位疫苗接种使用小分子的肿瘤内给药,蛋白质,纳米粒子,或激活病原体识别受体(PRR)以重新编程肿瘤微环境并引发全身抗肿瘤免疫的病毒。Cw豆花叶病毒(CPMV)是一种植物病毒,尽管对哺乳动物没有传染性,但却激活了哺乳动物的PRR。CPMV作为原位疫苗(ISV)的应用在肿瘤小鼠模型和犬患者中产生有效和持久的功效;数据表明CPMV优于小分子PRR激动剂和其他非相关植物病毒和病毒样颗粒(VLP)。在这项工作中,我们着手比较CPMV与来自Secoviridae的其他植物病毒的效力。我们开发了从植物中生产和分离cow豆严重花叶病毒(CPSMV)和烟草环斑病毒(TRSV)的方案。CPSMV,像CPMV,是一种具有基因组和蛋白质同源性的Comovirus,而TRSV缺乏同源性,来自子病毒属。当在皮肤黑素瘤的小鼠模型中作为ISV应用时(使用B16F10细胞和C57Bl6J小鼠),CPMV的表现优于CPSMV和TRSV,再次凸显了CPMV的独特功效。机械上,效力的增加与通过toll样受体(TLRs)的信号增强有关,特别是,CPMV信号通过TLR2、4和7。使用敲除(KO)小鼠模型,我们在这里证明了所有三种植物病毒都通过衔接分子MyD88发出信号,CPSMV和TRSV主要激活TLR2和4。与TRSV和CPSMV相比,CPMV诱导的干扰素β(IFNβ)明显更多;因此,通过TLR7信号传导时释放的IFNβ可能是观察到的CPMV-ISV效力的区分因子。此外,CPMV诱导瘤内细胞因子产生的不同时间模式,其特征是在2次每周治疗的第二次治疗后4天,炎性细胞因子显着增加。就像CPMV诱发了“记忆反应”一样。这个更高,更持久的细胞因子诱导可能是解释CPMV-ISV独特效力的另一个关键区别因素。
    In situ vaccination for cancer immunotherapy uses intratumoral administration of small molecules, proteins, nanoparticles, or viruses that activate pathogen recognition receptors (PRRs) to reprogram the tumor microenvironment and prime systemic antitumor immunity. Cowpea mosaic virus (CPMV) is a plant virus that─while noninfectious toward mammals─activates mammalian PRRs. Application of CPMV as in situ vaccine (ISV) results in a potent and durable efficacy in tumor mouse models and canine patients; data indicate that CPMV outperforms small molecule PRR agonists and other nonrelated plant viruses and virus-like particles (VLPs). In this work, we set out to compare the potency of CPMV versus other plant viruses from the Secoviridae. We developed protocols to produce and isolate cowpea severe mosaic virus (CPSMV) and tobacco ring spot virus (TRSV) from plants. CPSMV, like CPMV, is a comovirus with genome and protein homology, while TRSV lacks homology and is from the genus nepovirus. When applied as ISV in a mouse model of dermal melanoma (using B16F10 cells and C57Bl6J mice), CPMV outperformed CPSMV and TRSV─again highlighting the unique potency of CPMV. Mechanistically, the increased potency is related to increased signaling through toll-like receptors (TLRs)─in particular, CPMV signals through TLR2, 4, and 7. Using knockout (KO) mouse models, we demonstrate here that all three plant viruses signal through the adaptor molecule MyD88─with CPSMV and TRSV predominantly activating TLR2 and 4. CPMV induced significantly more interferon β (IFNβ) compared to TRSV and CPSMV; therefore, IFNβ released upon signaling through TLR7 may be a differentiator for the observed potency of CPMV-ISV. Additionally, CPMV induced a different temporal pattern of intratumoral cytokine generation characterized by significantly increased inflammatory cytokines 4 days after the second of 2 weekly treatments, as if CPMV induced a \"memory response\". This higher, longer-lasting induction of cytokines may be another key differentiator that explains the unique potency of CPMV-ISV.
    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