Bacterial therapy

细菌疗法
  • 文章类型: Journal Article
    口服免疫疗法(OIT),到目前为止,是食物过敏评估最多的治疗方法。然而,OIT不知道会导致治愈,它有过敏反应的风险。目前正在研究OIT的辅助疗法,以评估其对安全性和结果的影响。在这些疗法中,奥马珠单抗是评价最高的生物制剂。有越来越多的证据表明,奥马珠单抗在单食物和多过敏原OIT中都能有效诱导OIT的快速脱敏。同时减少不良反应的发生率。其他辅助生物制剂的评估,如dupilumab和细菌疗法,正在进行中。
    Oral immunotherapy (OIT), thus far, is the most evaluated therapeutic approach for food allergy. However, OIT is not known to lead to a cure, and it carries a risk for allergic reactions. Adjunct therapies to OIT are currently being investigated to evaluate their effect on safety and outcome. Of these therapies, omalizumab is the most evaluated biologic. There is mounting evidence that omalizumab is effective in inducing rapid desensitization of OIT in both single-food and multiallergen OIT, while diminishing the rate of adverse reactions. Evaluation of other adjunct biologics, such as dupilumab and bacterial therapy, is underway.
    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
    细菌疗法被认为是几种疾病的具有成本效益的治疗方法。然而,它的发展受到有限功能的阻碍,弱的固有治疗效果,以及对恶劣微环境条件的脆弱性,导致次优的治疗活动。增强细菌活性和治疗结果是一个关键的挑战。纳米酶由于其酶模拟活性和高稳定性而引起了极大的关注。它们使细菌能够模拟表达相同功能酶的基因编辑细菌的功能,从而提高细菌活性和治疗功效。这篇综述描述了细菌和纳米酶的治疗机制,其次是制备细菌/纳米酶复合材料的策略总结。此外,强调了这种复合材料在生物医学应用如胃肠道疾病和肿瘤中的协同作用。最后,讨论了细菌/纳米酶复合材料的挑战,并提出了潜在的解决方案。本研究旨在提供有价值的见解,为纳米材料辅助细菌治疗的发展提供理论指导。
    Bacterial therapy is recognized as a cost-effective treatment for several diseases. However, its development is hindered by limited functionality, weak inherent therapeutic effects, and vulnerability to harsh microenvironmental conditions, leading to suboptimal treatment activity. Enhancing bacterial activity and therapeutic outcomes emerges as a pivotal challenge. Nanozymes have garnered significant attention due to their enzyme-mimic activities and high stability. They enable bacteria to mimic the functions of gene-edited bacteria expressing the same functional enzymes, thereby improving bacterial activity and therapeutic efficacy. This review delineates the therapeutic mechanisms of bacteria and nanozymes, followed by a summary of strategies for preparing bacteria/nanozyme composites. Additionally, the synergistic effects of such composites in biomedical applications such as gastrointestinal diseases and tumors are highlighted. Finally, the challenges of bacteria/nanozyme composites are discussed and propose potential solutions. This study aims to provide valuable insights to offer theoretical guidance for the advancement of nanomaterial-assisted bacterial 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
    某些细菌显示出靶向和定植肿瘤微环境的能力,将它们定位为在癌症治疗中提供各种治疗剂的创新载体的特征。然而,我们对细菌如何使其生理状况适应肿瘤微环境的理解仍然难以捉摸。在这项工作中,我们采用液相色谱-串联质谱法检测小鼠肿瘤中定植的大肠杆菌的蛋白质组。与在丰富培养基中培养的大肠杆菌相比,我们发现在肿瘤中定植的大肠杆菌显著上调与三价铁离子相关的过程,包括肠杆菌素生物合成和铁稳态。这一发现表明肿瘤是大肠杆菌缺铁的环境。我们还发现,大肠杆菌在肿瘤中的定植导致脂质运载蛋白2(LCN2)的表达增加,一种可以隔离肠杆菌素的宿主蛋白。因此,我们改造大肠杆菌以逃避由LCN2提供的营养免疫。通过引入IroA集群,大肠杆菌合成糖基化的肠杆菌素,这会产生空间位阻以避免LCN2封存。IroA-E.大肠杆菌对LCN2的抗性增强,并显着提高了小鼠的抗肿瘤活性。此外,IroA-E治愈的小鼠大肠杆菌治疗对肿瘤再次挑战产生了抗性,表明免疫记忆的建立。总的来说,我们的研究强调了细菌在肿瘤微环境中获取三价铁离子的能力对于有效的癌症治疗的关键作用。
    Certain bacteria demonstrate the ability to target and colonize the tumor microenvironment, a characteristic that positions them as innovative carriers for delivering various therapeutic agents in cancer therapy. Nevertheless, our understanding of how bacteria adapt their physiological condition to the tumor microenvironment remains elusive. In this work, we employed liquid chromatography-tandem mass spectrometry to examine the proteome of E. coli colonized in murine tumors. Compared to E. coli cultivated in the rich medium, we found that E. coli colonized in tumors notably upregulated the processes related to ferric ions, including the enterobactin biosynthesis and iron homeostasis. This finding indicated that the tumor is an iron-deficient environment to E. coli. We also found that the colonization of E. coli in the tumor led to an increased expression of lipocalin 2 (LCN2), a host protein that can sequester the enterobactin. We therefore engineered E. coli in order to evade the nutritional immunity provided by LCN2. By introducing the IroA cluster, the E. coli synthesizes the glycosylated enterobactin, which creates steric hindrance to avoid the LCN2 sequestration. The IroA-E. coli showed enhanced resistance to LCN2 and significantly improved the anti-tumor activity in mice. Moreover, the mice cured by the IroA-E. coli treatment became resistant to the tumor re-challenge, indicating the establishment of immunological memory. Overall, our study underscores the crucial role of bacteria\'s ability to acquire ferric ions within the tumor microenvironment for effective cancer therapy.
    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
    背景:细菌癌症治疗在19世纪末首次在患者中试验。最近,已经利用肿瘤靶向细菌将质粒表达的治疗性干扰RNA递送至一系列实体瘤.其临床翻译的主要限制是由于质粒不稳定性导致的RNA干扰在体内的短期性质。为了克服这一点,我们试图开发靶向肿瘤的减毒细菌,其通过在细菌染色体内整合表达盒而稳定表达shRNA,并在体外和体内证明治疗功效.
    结果:对减毒的肿瘤靶向鼠伤寒沙门氏菌SL7207菌株进行修饰,使其在xylA位点携带染色体整合的shRNA表达盒。结肠直肠癌细胞系SW480、HCT116和乳腺癌细胞系MCF7用于证明这些修饰的菌株进行细胞内感染并递送靶基因c-Myc的有效RNA和蛋白质敲低的能力。使用Lgr5creERT2Apcflx/flx和BlgCreBrca2flx/flp53flx/flx原位免疫活性小鼠结直肠癌和乳腺癌模型证明了体内治疗功效,分别。乳腺癌和结肠直肠癌细胞系与经修饰的SL7207的体外共培养显示与SL7207/c-Myc靶向菌株的RNA和蛋白质表达显著50-95%(P<0.01)减少。在体内,在肿瘤组织建立之后,单次腹膜内施用1×106CFU的SL7207/c-Myc足以允许肿瘤定植并显着延长存活,而在对照动物中没有明显的毒性。
    结论:总之,我们已经证明了嗜肿瘤细菌可以被修饰以安全地提供治疗水平的基因敲低。这项技术有可能通过个性化的治疗有效载荷专门针对原发性和继发性实体瘤,提供新的多癌症检测和治疗选择,具有最小的脱靶效应。需要进一步了解向性机制以及对宿主免疫和微生物组的影响,才能发展到临床翻译。
    BACKGROUND: Bacterial cancer therapy was first trialled in patients at the end of the nineteenth century. More recently, tumour-targeting bacteria have been harnessed to deliver plasmid-expressed therapeutic interfering RNA to a range of solid tumours. A major limitation to clinical translation of this is the short-term nature of RNA interference in vivo due to plasmid instability. To overcome this, we sought to develop tumour-targeting attenuated bacteria that stably express shRNA by virtue of integration of an expression cassette within the bacterial chromosome and demonstrate therapeutic efficacy in vitro and in vivo.
    RESULTS: The attenuated tumour targeting Salmonella typhimurium SL7207 strain was modified to carry chromosomally integrated shRNA expression cassettes at the xylA locus. The colorectal cancer cell lines SW480, HCT116 and breast cancer cell line MCF7 were used to demonstrate the ability of these modified strains to perform intracellular infection and deliver effective RNA and protein knockdown of the target gene c-Myc. In vivo therapeutic efficacy was demonstrated using the Lgr5creERT2Apcflx/flx and BlgCreBrca2flx/flp53flx/flx orthotopic immunocompetent mouse models of colorectal and breast cancer, respectively. In vitro co-cultures of breast and colorectal cancer cell lines with modified SL7207 demonstrated a significant 50-95% (P < 0.01) reduction in RNA and protein expression with SL7207/c-Myc targeted strains. In vivo, following establishment of tumour tissue, a single intra-peritoneal administration of 1 × 106 CFU of SL7207/c-Myc was sufficient to permit tumour colonisation and significantly extend survival with no overt toxicity in control animals.
    CONCLUSIONS: In summary we have demonstrated that tumour tropic bacteria can be modified to safely deliver therapeutic levels of gene knockdown. This technology has the potential to specifically target primary and secondary solid tumours with personalised therapeutic payloads, providing new multi-cancer detection and treatment options with minimal off-target effects. Further understanding of the tropism mechanisms and impact on host immunity and microbiome is required to progress to clinical translation.
    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
    卵巢癌对女性健康构成重大威胁,常规治疗方法遇到许多限制,新兴的工程细菌抗肿瘤策略为卵巢癌治疗提供了新的希望。在这项研究中,我们构建了VNP20009-Abvec-Igκ-MIIP(VM)工程菌株,并对其体外生长性能和迁移/侵袭抑制蛋白(MIIP)的表达能力进行了初步评估。随后,进行ID8卵巢癌细胞和小鼠癌症模型以研究VM对卵巢癌的影响。我们的结果表明,VM菌株表现出优异的生长性能,成功侵入ID8卵巢癌细胞,并表示MIIP,从而抑制细胞增殖和迁移。此外,VM特异性靶向肿瘤部位并表达MIIP,进一步缩小卵巢癌小鼠肿瘤体积(p<0.01),通过表皮生长因子受体(EGFR)的下调,拉斯,p-MEK,还有p-ERK.PI3K/AKT信号通路的下调和Bcl-2/Bax水平的降低也表明VM对卵巢癌细胞的凋亡潜能。总之,我们的研究表明,VM在体外和体内都表现出有希望的抗肿瘤作用,强调其临床治疗卵巢癌的潜力。关键点:•本研究构建了能够表达抗癌蛋白的鼠伤寒沙门氏菌工程菌株•工程细菌可以在体内靶向和定植肿瘤部位•VM可以抑制增殖,迁移,和卵巢癌细胞的侵袭。
    Ovarian cancer poses a significant threat to women\'s health, with conventional treatment methods encountering numerous limitations, and the emerging engineered bacterial anti-tumor strategies offer newfound hope for ovarian cancer treatment. In this study, we constructed the VNP20009-Abvec-Igκ-MIIP (VM) engineered strain and conducted initial assessments of its in vitro growth performance and the expression capability of migration/invasion inhibitory protein (MIIP). Subsequently, ID8 ovarian cancer cells and mouse cancer models were conducted to investigate the impact of VM on ovarian cancer. Our results revealed that the VM strain demonstrated superior growth performance, successfully invaded ID8 ovarian cancer cells, and expressed MIIP, consequently suppressing cell proliferation and migration. Moreover, VM specifically targeted tumor sites and expressed MIIP which further reduced the tumor volume of ovarian cancer mice (p < 0.01), via the downregulation of epidermal growth factor receptor (EGFR), Ras, p-MEK, and p-ERK. The downregulation of the PI3K/AKT signaling pathway and the decrease in Bcl-2/Bax levels also indicated VM\'s apoptotic potency on ovarian cancer cells. In summary, our research demonstrated that VM exhibits promising anti-tumor effects both in vitro and in vivo, underscoring its potential for clinical treatment of ovarian cancer. KEY POINTS: • This study has constructed an engineered strain of Salmonella typhimurium capable of expressing anticancer proteins • The engineered bacteria can target and colonize tumor sites in vivo • VM can inhibit the proliferation, migration, and invasion of ovarian cancer cells.
    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
    免疫疗法显示出巨大的希望,但并非对所有类型的肿瘤都有效,对不到3%的胰腺导管腺癌(PDAC)患者有效.为了对更多的癌症患者和患有PDAC的患者进行有效的免疫治疗,我们对沙门氏菌进行基因工程改造,将外源抗原直接传递到肿瘤细胞的细胞质中。我们假设外源免疫抗原的细胞内递送将激活抗原特异性CD8T细胞并减少免疫小鼠中的肿瘤。
    为了检验这一假设,我们给予细胞内递送(ID)沙门氏菌,将卵清蛋白作为模型抗原递送到荷瘤中,卵清蛋白接种的小鼠。ID沙门氏菌在诱导细胞侵袭后通过在细胞中自主裂解来递送抗原。
    我们表明所递送的卵清蛋白分散在培养物和肿瘤中的细胞的整个细胞质中。这种进入细胞质的递送对于抗原交叉呈递是必需的。我们表明,卵清蛋白受体癌细胞与卵清蛋白特异性CD8T细胞的共培养触发了细胞毒性T细胞反应。OT-ICD8T细胞过继转移后,卵清蛋白的细胞内递送减少了肿瘤生长并消除了肿瘤。这种作用取决于卵清蛋白特异性T细胞的存在。在小鼠中接种外源抗原后,抗原的细胞内递送清除了43%的已建立的KPC胰腺肿瘤,增加生存率,并防止肿瘤重新植入。
    免疫抑制KPC模型中的这种反应证明了治疗对检查点抑制剂无反应的肿瘤的潜力,并且对再攻击的反应表明针对内在肿瘤抗原建立了新的免疫力。在诊所里,ID沙门氏菌可用于从儿童免疫中递送蛋白质抗原,以重新集中针对肿瘤的预先存在的T细胞免疫。作为一种现成的免疫疗法,这种细菌系统有可能在广泛的癌症患者中有效。
    Immunotherapies have shown great promise, but are not effective for all tumors types and are effective in less than 3% of patients with pancreatic ductal adenocarcinomas (PDAC). To make an immune treatment that is effective for more cancer patients and those with PDAC specifically, we genetically engineered Salmonella to deliver exogenous antigens directly into the cytoplasm of tumor cells. We hypothesized that intracellular delivery of an exogenous immunization antigen would activate antigen-specific CD8 T cells and reduce tumors in immunized mice.
    To test this hypothesis, we administered intracellular delivering (ID) Salmonella that deliver ovalbumin as a model antigen into tumor-bearing, ovalbumin-vaccinated mice. ID Salmonella delivers antigens by autonomously lysing in cells after the induction of cell invasion.
    We showed that the delivered ovalbumin disperses throughout the cytoplasm of cells in culture and in tumors. This delivery into the cytoplasm is essential for antigen cross-presentation. We showed that co-culture of ovalbumin-recipient cancer cells with ovalbumin-specific CD8 T cells triggered a cytotoxic T cell response. After the adoptive transfer of OT-I CD8 T cells, intracellular delivery of ovalbumin reduced tumor growth and eliminated tumors. This effect was dependent on the presence of the ovalbumin-specific T cells. Following vaccination with the exogenous antigen in mice, intracellular delivery of the antigen cleared 43% of established KPC pancreatic tumors, increased survival, and prevented tumor re-implantation.
    This response in the immunosuppressive KPC model demonstrates the potential to treat tumors that do not respond to checkpoint inhibitors, and the response to re-challenge indicates that new immunity was established against intrinsic tumor antigens. In the clinic, ID Salmonella could be used to deliver a protein antigen from a childhood immunization to refocus pre-existing T cell immunity against tumors. As an off-the-shelf immunotherapy, this bacterial system has the potential to be effective in a broad range of cancer 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
    细菌介导的抗肿瘤治疗因其天然的肿瘤靶向能力和特异性的免疫激活特性而受到广泛关注。在打破单药治疗的局限性,有效根除肿瘤方面取得了重大进展,尤其是与传统疗法结合时,比如放疗。根据其不同的生物学特性,细菌及其衍生物不仅可以提高肿瘤放疗的敏感性,而且可以保护正常组织。此外,基因工程细菌和基于细菌的生物材料进一步扩大了其在放射治疗中的应用范围。在这次审查中,综述了近年来细菌及其衍生物在放疗中的应用研究,阐述细菌,细菌衍生物和基于细菌的生物材料不仅可以直接增强放疗,还可以通过改善肿瘤微环境(TME)和免疫效果来提高抗肿瘤效果。此外,一些益生菌还可以通过抗炎保护肠道等正常组织和器官免受辐射,抗氧化和凋亡抑制。总之,细菌在放射治疗中的前景将非常广泛,但其生物安全性和作用机制有待进一步评价和研究。
    Bacteria-mediated anti-tumor therapy has received widespread attention due to its natural tumor-targeting ability and specific immune-activation characteristics. It has made significant progress in breaking the limitations of monotherapy and effectively eradicating tumors, especially when combined with traditional therapy, such as radiotherapy. According to their different biological characteristics, bacteria and their derivatives can not only improve the sensitivity of tumor radiotherapy but also protect normal tissues. Moreover, genetically engineered bacteria and bacteria-based biomaterials have further expanded the scope of their applications in radiotherapy. In this review, we have summarized relevant researches on the application of bacteria and its derivatives in radiotherapy in recent years, expounding that the bacteria, bacterial derivatives and bacteria-based biomaterials can not only directly enhance radiotherapy but also improve the anti-tumor effect by improving the tumor microenvironment (TME) and immune effects. Furthermore, some probiotics can also protect normal tissues and organs such as intestines from radiation via anti-inflammatory, anti-oxidation and apoptosis inhibition. In conclusion, the prospect of bacteria in radiotherapy will be very extensive, but its biological safety and mechanism need to be further evaluated and studied.
    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
    胰腺癌是一种高度侵袭性的癌症,五年生存率仅为10%。胰腺导管腺癌(PDAC)占这些病例的90%。PDAC与致密基质相关,其赋予对当前治疗方式的抗性。增加对癌症治疗的抗性提出了挑战和对替代疗法的需要。细菌介导的癌症疗法是由WilliamColey博士在1800年代后期提出的,当时他向骨肉瘤患者注射了活链球菌或热灭活的化脓性链球菌和粘质沙雷菌,称为Coley毒素。从那以后,几种细菌已经获得了增强治疗反应的可能作用的认可,增强抗肿瘤免疫力,并减轻标准治疗方案的不良影响。这篇综述强调了促进抗肿瘤免疫的关键细菌机制和结构。与细菌介导的癌症治疗相关的挑战和风险,以及在PDAC管理中使用的应用程序和机会。
    Pancreatic cancer is a highly aggressive form of cancer with a five-year survival rate of only ten percent. Pancreatic ductal adenocarcinoma (PDAC) accounts for ninety percent of those cases. PDAC is associated with a dense stroma that confers resistance to current treatment modalities. Increasing resistance to cancer treatments poses a challenge and a need for alternative therapies. Bacterial mediated cancer therapies were proposed in the late 1800s by Dr. William Coley when he injected osteosarcoma patients with live streptococci or a fabrication of heat-killed Streptococcus pyogenes and Serratia marcescens known as Coley\'s toxin. Since then, several bacteria have gained recognition for possible roles in potentiating treatment response, enhancing anti-tumor immunity, and alleviating adverse effects to standard treatment options. This review highlights key bacterial mechanisms and structures that promote anti-tumor immunity, challenges and risks associated with bacterial mediated cancer therapies, and applications and opportunities for use in PDAC management.
    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
    最近发现细菌菌株,它可以专门对抗恶性细胞,可以有效地用于癌症治疗。许多合适的细菌菌株是致病性的或侵入性的,并且在体内监测以这种方式使用的细菌的分布的方法存在显著不足。这里,建议使用逐层(LbL)方法,该方法可以用荧光标记的聚电解质(PE)和磁铁矿纳米颗粒(NP)包裹单个细菌细胞。NP能够在体内远程引导到所讨论的部位,并且远红发射光谱中标记的外壳允许对体内细菌的分布进行非侵入性监测。经修饰的细菌的磁截留导致细菌的局部浓度增加至少5倍。PE制造了一个强大的屏障,体外实验表明,以这种方式包被的细菌细胞的分裂时间可以调节,从而控制它们侵入组织。研究中使用的动物存活下来,没有遭受感染性休克,这可以归因于防止细菌细胞释放内毒素的PE胶囊。
    It was shown recently that bacterial strains, which can act specifically against malignant cells, can be used efficiently in cancer therapy. Many appropriate bacterial strains are either pathogenic or invasive and there is a substantial shortage of methods with which to monitor in vivo the distribution of bacteria used in this way. Here, it is proposed to use a Layer-by-Layer (LbL) approach that can encapsulate individual bacterial cells with fluorescently labeled polyelectrolytes (PE)s and magnetite nanoparticles (NP)s. The NP enable remote direction in vivo to the site in question and the labeled shells in the far-red emission spectra allow non-invasive monitoring of the distribution of bacteria in the body. The magnetic entrapment of the modified bacteria causes the local concentration of the bacteria to increase by a factor of at least 5. The PEs create a strong barrier, and it has been shown in vitro experiments that the division time of bacterial cells coated in this way can be regulated, resulting in control of their invasion into tissues. That animals used in the study survived and did not suffer septic shock, which can be attributed to PE capsules that prevent release of endotoxins from bacterial cells.
    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
    程序性细胞死亡蛋白1/程序性细胞死亡配体1(PD-1/PD-L1)免疫检查点抑制剂是目前最有前途的恶性肿瘤治疗方法,但低反应率限制了其进一步的临床应用。为了解决这个问题,我们小组构建了VNP20009-Abvec-Igκ-mPD-1[V-A-mPD-1(mPD-1,鼠PD-1)]工程菌株,将溶瘤细菌治疗与免疫治疗相结合.Further,我们在建立黑色素瘤小鼠模型的同时,评估其体外生长性能和mPD-1表达能力,以探索其在肿瘤治疗中的潜在抗癌作用。我们的结果表明,V-A-mPD-1菌株具有优越的生长性能,可以入侵B16F10黑色素瘤细胞并表达PD-1。此外,在黑色素瘤小鼠模型中,我们观察到肿瘤体积明显缩小,形成了更大的坏死区域。V-A-mPD-1给药导致mPD-1在肿瘤部位高表达,通过下调大鼠肉瘤(Ras)的表达抑制肿瘤细胞增殖,磷酸化丝裂原活化蛋白激酶(p-MEK)/MEK,磷酸化的细胞外信号调节激酶(p-ERK)/ERK表达显著抑制肿瘤细胞增殖。下调磷酸肌醇3激酶(PI3K)和蛋白激酶B(AKT)信号通路促进肿瘤细胞凋亡,如Bcl-2相关X蛋白/B细胞淋巴瘤-2(Bax/Bcl-2)表达比率增加所证明。同时,全身炎症细胞因子的表达水平,如白细胞介素-6(IL-6),白细胞介素-1β(IL-1β),和肿瘤坏死因子-α(TNF-α),大幅减少。总之,我们的研究表明,V-A-mPD-1具有优异的抗肿瘤作用,提示联合应用微生物疗法和免疫治疗是一种可行的癌症治疗策略。
    Programmed cell death protein 1/Programmed cell death ligand 1 (PD-1/PD-L1) immune checkpoint inhibitors are the most promising treatments for malignant tumors currently, but the low response rate limits their further clinical utilization. To address this problem, our group constructed an engineered strain of VNP20009-Abvec-Igκ-mPD-1 [V-A-mPD-1 (mPD-1, murine PD-1)] to combine oncolytic bacterial therapy with immunotherapy. Further, we evaluated its growth performance and mPD-1 expression ability in vitro while establishing the melanoma mice model to explore its potential anti-cancer effects in tumor therapy. Our results indicated that the V-A-mPD-1 strain has superior growth performance and can invade B16F10 melanoma cells and express PD-1. In addition, in the melanoma mice model, we observed a marked reduction in tumor volume and the formation of a larger necrotic area. V-A-mPD-1 administration resulted in a high expression of mPD-1 at the tumor site, inhibiting tumor cell proliferation via the down-regulation of the expression of rat sarcoma (Ras), phosphorylated mitogen-activated protein kinase (p-MEK)/MEK, and phosphorylated extracellular signal-regulated kinase (p-ERK)/ERK expression significantly inhibited tumor cell proliferation. Tumor cell apoptosis was promoted by down-regulating phosphoinositide 3 kinase (PI3K) and protein kinase B (AKT) signaling pathways, as evidenced by an increased Bcl-2-associated X protein/B cell lymphoma-2 (Bax/Bcl-2) expression ratio. Meanwhile, the expression levels of systemic inflammatory cytokines, such as interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α), were substantially reduced. In conclusion, our research demonstrated that V-A-mPD-1 has an excellent anti-tumor effect, prompting that the combined application of microbial therapy and immunotherapy is a feasible cancer treatment strategy.
    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