anti-angiogenesis

抗血管生成
  • 文章类型: Journal Article
    肝细胞癌(HCC)是一种高度血管化的癌,靶向其新血管形成代表了一种有效的治疗方法。我们先前的研究表明,杆状病毒介导的内皮抑素和血管抑素融合蛋白(BDS-hEA)有效抑制血管内皮细胞的血管生成和HCC肿瘤的生长。然而,其抗血管生成作用的潜在机制尚不清楚.越来越多的证据表明,自噬对血管内皮细胞的功能和对癌症治疗的反应具有重要影响。因此,本研究的目的是探讨BDS-hEA诱导的血管生成抑制与自噬,以及潜在的监管机制。我们的结果表明BDS-hEA在EA中诱导自噬。hy926细胞,自噬体和活性氧数量的增加证明了这一点,伴随着Beclin-1,LC3-II/LC3-I的上调,和p62蛋白表达。使用3-甲基腺嘌呤抑制自噬减弱了BDS-hEA诱导的EA的功能。hy926细胞,包括生存能力,扩散,入侵,迁移,和血管生成。此外,BDS-hEA通过下调CD31、VEGF、和VEGFR2,以及磷酸化蛋白激酶B(p-AKT)和磷酸化哺乳动物雷帕霉素靶蛋白(p-mTOR),同时上调磷酸化AMP激活蛋白激酶(p-AMPK)。体内结果进一步表明,氯喹对自噬的抑制作用显着阻碍了BDS-hEA抑制小鼠HCC肿瘤生长的能力。机械上,BDS-hEA显著促进肿瘤组织的自噬凋亡,降低肿瘤组织中ki67、CD31、VEGF、MMP-9,p-AKT,和p-mTOR,同时增强p-AMPK表达。总之,我们的研究结果表明,BDS-hEA通过调节AMPK/AKT/mTOR信号通路诱导自噬作为细胞毒性反应,从而对HCC发挥抗血管生成作用。
    Hepatocellular carcinoma (HCC) is a highly vascularized carcinoma, and targeting its neovascularization represents an effective therapeutic approach. Our previous study demonstrated that the baculovirus-mediated endostatin and angiostatin fusion protein (BDS-hEA) effectively inhibits the angiogenesis of vascular endothelial cells and the growth of HCC tumors. However, the mechanism underlying its anti-angiogenic effect remains unclear. Increasing evidence suggests that autophagy has a significant impact on the function of vascular endothelial cells and response to cancer therapy. Hence, the objective of this research was to investigate the correlation between BDS-hEA-induced angiogenesis inhibition and autophagy, along with potential regulatory mechanisms. Our results demonstrated that BDS-hEA induced autophagy in EA.hy926 cells, as evidenced by the increasing number of autophagosomes and reactive oxygen species, accompanied by an upregulation of Beclin-1, LC3-II/LC3-I, and p62 protein expression. Suppression of autophagy using 3-methyladenine attenuated the functions of BDS-hEA-induced EA.hy926 cells, including the viability, proliferation, invasion, migration, and angiogenesis. Moreover, BDS-hEA induced autophagy by downregulating the expression of CD31, VEGF, and VEGFR2, as well as phosphorylated protein kinase B (p-AKT) and phosphorylated mammalian target of rapamycin (p-mTOR), while concurrently upregulating phosphorylated AMP-activated protein kinase (p-AMPK). The in vivo results further indicated that inhibition of autophagy by chloroquine significantly impeded the ability of BDS-hEA to suppress HCC tumor growth in mice. Mechanistically, BDS-hEA prominently facilitated autophagic apoptosis in tumor tissues and decreased the levels of ki67, CD31, VEGF, MMP-9, p-AKT, and p-mTOR while simultaneously enhancing the p-AMPK expression. In conclusion, our findings suggest that BDS-hEA induces autophagy as a cytotoxic response by modulating the AMPK/AKT/mTOR signaling pathway, thereby exerting anti-angiogenic effects against HCC.
    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:血管生成因子通常在实体瘤中被激活,并且是可行的治疗靶标。然而,血管生成抑制剂(AGI)的抗癌治疗仅限于少数癌症,主要作为单一疗法,而不是根据分子适应症选择。我们旨在确定基于多分析物肿瘤询问(ETA:百科全书肿瘤分析)选择的AGI和其他抗癌剂的患者特异性组合方案是否可以扩大AGI在晚期难治性实体器官癌中的范围,并改善治疗反应。方法:我们评估了60例晚期患者的治疗结果,接受ETA指导的AGI与其他靶向联合治疗方案的难治性实体器官癌,内分泌或细胞毒性剂。治疗反应的放射学评估,然后测定客观反应率(ORR),疾病控制率(DCR),无进展生存期(PFS)和总生存期(OS)。结果:60例患者中,部分缓解(PR)28例(46.7%),29例(48.3%)观察到稳定的疾病(SD)和疾病进展(PD,在60天内)观察到3例(5.0%)。ORR为46.7%,DCR为95.0%。在最近的随访中,中位PFS(mPFS)为5.0个月,中位OS(mOS)为8.9个月。没有4级治疗相关的不良事件或治疗相关的死亡。结论:ETA指导的患者特异性联合方案与AGI和其他抗肿瘤药,与AGI单药治疗相比,可以改善预后。试验注册:所有试验的详细信息可在WHO-ICTRP:https://apps上获得。谁。int/trialsearch/。电阻IDCTRI/2018/02/011,808。液体冲击IDCTRI/2019/02/017,548。
    Background: Angiogenic factors are commonly activated in solid tumors and present a viable therapeutic target. However, anticancer treatment with angiogenesis inhibitors (AGI) is limited to a few cancers, mostly as monotherapy and not selected based on molecular indications. We aimed to determine whether patient-specific combination regimens with AGI and other anticancer agents when selected based on multi-analyte tumor interrogation (ETA: Encyclopedic Tumor Analysis) can expand the scope of AGIs in advanced refractory solid organ cancers with improved treatment responses. Methods: We evaluated treatment outcomes in 60 patients with advanced, refractory solid organ cancers who received ETA-guided combination regimens of AGI with other targeted, endocrine or cytotoxic agents. Radiological evaluation of treatment response was followed by determination of Objective Response Rate (ORR), Disease Control Rate (DCR), Progression Free Survival (PFS) and Overall Survival (OS). Results: Among the 60 patients, Partial Response (PR) was observed in 28 cases (46.7%), Stable Disease (SD) was observed in 29 cases (48.3%) and Disease Progression (PD, within 60 days) was observed in 3 cases (5.0%). The ORR was 46.7% and DCR was 95.0%. At the most recent follow-up the median PFS (mPFS) was 5.0 months and median OS (mOS) was 8.9 months. There were no Grade 4 therapy related adverse events or treatment related deaths. Conclusion: ETA-guided patient-specific combination regimens with AGI and other anti-neoplastic agents, can yield improved outcomes over AGI monotherapy. Trial Registration: Details of all trials are available at WHO-ICTRP: https://apps.who.int/trialsearch/. RESILIENT ID CTRI/2018/02/011,808. LIQUID IMPACT ID CTRI/2019/02/017,548.
    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
    角膜新生血管是视力丧失的重要原因,常导致角膜混浊和慢性炎症。鲨鱼软骨被广泛认为是抗血管生成化合物的重要天然来源。我们先前的研究表明,来自白斑猫鲨(ChiloscylliumplagiosumBonnet)的多肽具有抑制乳腺肿瘤血管生成的潜力。这项研究将该肽(SAIF)应用于角膜碱损伤模型,以评估其对角膜新生血管的影响。结果显示SAIF抑制内皮细胞增殖,迁移,和管的形成。SAIF抑制基质胶塞中VEGF诱导的血管生成。使用角膜碱损伤模型,SAIF显著抑制小鼠角膜血管新生。我们发现SAIF不仅显著抑制VEGF等促血管生成因子的上调,bFGF,碱损伤诱导的PDGF表达,还能促进抗血管生成因子PEDF的表达。此外,我们还分析了参与细胞外基质(ECM)重塑的MMPs和TIMPs,血管生成,和淋巴管生成。我们发现SAIF治疗抑制促血管生成因子如MMP1,MMP2,MMP3,MMP9,MMP13和MMP14的表达,并促进抗血管生成因子如MMP7,TIMP1,TIMP2和TIMP3的表达。总之,SAIF作为抗血管生成因子抑制增殖,迁移,和内皮细胞的管形成,抑制促血管生成因子,促进抗血管生成因子,并调节MMP的表达,最终抑制角膜新生血管形成。
    Corneal neovascularization is a significant cause of vision loss, often resulting in corneal clouding and chronic inflammation. Shark cartilage is widely recognized as a significant natural source of anti-angiogenic compounds. Our previous studies have shown that a polypeptide from white-spotted catshark (Chiloscyllium plagiosum Bonnet) has the potential to inhibit the angiogenesis of breast tumors. This study applied this peptide (SAIF) to a corneal alkali injury model to assess its effect on corneal neovascularization. Results revealed that SAIF inhibits endothelial cell proliferation, migration, and tube formation. SAIF inhibited VEGF-induced angiogenesis in the matrigel plug. Using the corneal alkali injury model, SAIF significantly inhibited corneal vascular neovascularization in mice. We found that SAIF not only significantly inhibited the upregulation of pro-angiogenic factors such as VEGF, bFGF, and PDGF expression induced by alkali injury, but also promoted the expression of anti-angiogenesis factor PEDF. Moreover, we also analyzed the MMPs and TIMPs involved in extracellular matrix (ECM) remodeling, angiogenesis, and lymphangiogenesis. We found that SAIF treatment inhibited the expression of pro-angiogenic factors like MMP1, MMP2, MMP3, MMP9, MMP13, and MMP14, and promoted the expression of anti-angiogenesis factors such as MMP7, TIMP1, TIMP2, and TIMP3. In conclusion, SAIF acts as an anti-angiogenic factor to inhibit the proliferation, migration, and tube formation of endothelial cells, inhibit pro-angiogenic factors, promote anti-angiogenic factors, and regulate the expression of MMPs, ultimately inhibiting corneal neovascularization.
    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
    由Dirofilariaimmitis引起的心虫病是一种媒介传播的人畜共患疾病,主要是家犬和猫的感染,或者这些是已知数据最多的。人类是一个偶然的宿主,无症状的肺结节可能起源于。Dirofilariaimmitis还含有Wolbachia属的内共生体细菌,在蜕皮中起作用,胚胎发生,炎症病理学,和免疫反应。当Wolbachiasp.被释放到血液中,内皮和肺损伤加剧,进一步促进血栓形成和肺动脉高压,促进充血性心力衰竭和动物死亡。先前的研究表明,寄生虫排泄/分泌产物能够激活促血管生成途径(新血管的形成)以促进寄生虫存活。这项研究的目的是分析Wolbachiasp的作用。及其与体外人内皮细胞模型中细胞过程和血管生成途径的关系。重组Wolbachia表面蛋白(rWSP)的使用表明,其刺激通过检测VEGFR-1/sFlt1和sEndoglin的产生增加而发挥了抗血管生成作用,并且不影响VEGFR-2和mEndoglin的产生(促血管生成分子)。此外,它不刺激细胞增殖或迁移,尽管它确实负面地刺激了假毛细血管的形成,减缓这个过程。这些细胞过程与血管生成途径直接相关,有了这些结果,我们可以得出Wolbachiasp.与抗血管生成途径的刺激有关,不促进血管内皮中D.immitis的存活。
    Heartworm disease caused by Dirofilaria immitis is a vector-borne zoonotic disease responsible for the infection of mainly domestic dogs and cats, or these are those for which the most data are known. Humans are an accidental host where a benign, asymptomatic pulmonary nodule may originate. Dirofilaria immitis also harbours the endosymbiont bacteria of the genus Wolbachia, which play a role in moulting, embryogenesis, inflammatory pathology, and immune response. When Wolbachia sp. is released into the bloodstream, endothelial and pulmonary damage is exacerbated, further encouraging thrombus formation and pulmonary hypertension, facilitating congestive heart failure and death of the animal. Previous studies have shown that parasite excretory/secretory products are able to activate the pro-angiogenic pathway (formation of new vessels) to facilitate parasite survival. The aim of this study was to analyse the role of Wolbachia sp. and its relationship with the cellular processes and the angiogenic pathway in a model of human endothelial cells in vitro. The use of recombinant Wolbachia Surface Protein (rWSP) showed that its stimulation exerted an anti-angiogenic effect by detecting an increase in the production of VEGFR-1/sFlt1 and sEndoglin and did not affect the production of VEGFR-2 and mEndoglin (pro-angiogenic molecules). Furthermore, it did not stimulate cell proliferation or migration, although it did negatively stimulate the formation of pseudocapillaries, slowing down this process. These cellular processes are directly related to the angiogenic pathway so, with these results, we can conclude that Wolbachia sp. is related to the stimulation of the anti-angiogenic pathway, not facilitating the survival of D. immitis in vascular endothelium.
    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
    胰腺癌是消化道肿瘤中存活率最低、预后最差的肿瘤之一。在诊断的时候,大多数患者错过了根治性手术切除的机会,选择化疗作为他们的主要治疗选择.在最广泛使用的化疗方案如改良FOLFIRINOX方案的应用过程中出现耐药性,吉西他滨单一疗法或5-氟尿嘧啶联合疗法,这进一步降低了治疗功效。因此,迫切需要探索更好的胰腺癌治疗策略。近年来,越来越多的研究发现天然产物具有显著的抗胰腺癌特性。在本文中,我们综述了天然产物抑制胰腺癌细胞增殖和侵袭的可能机制,包括靶向抑制胰腺癌细胞生长和增殖调节途径的可能机制,诱导胰腺癌细胞凋亡和自噬,抑制胰腺癌细胞的EMT过程,并抑制胰腺癌的血管生成。同时,天然产物由于其成分的复杂性和生物提取技术的局限性,也阻碍了其基础和临床研究的进展。进一步探索天然产物抑制胰腺癌细胞增殖和侵袭的具体分子机制,优化纯化和制备技术,并丰富基础和临床试验验证其疗效和安全性可能是未来天然产物在抗胰腺癌领域研究的方向。
    Pancreatic cancer is one of the gastrointestinal tumors with the lowest survival rate and the worst prognosis. At the time of diagnosis, the majority of patients have missed the opportunity for radical surgical resection and opt for chemotherapy as their primary treatment choice. And drug resistance emerges during the application of the most widely used chemotherapeutic regimens such as modified FOLFIRINOX regimen, gemcitabine monotherapy or 5-Fluorouracil combination therapy, which further reduces the therapeutic efficacy. Therefore, it is urgent to explore better treatment strategies for pancreatic cancer. In recent years, more and more studies have found that natural products have significant anti-pancreatic cancer properties. In this paper, we reviewed the possible mechanisms by which natural products inhibit the proliferation and invasion of pancreatic cancer cells, including the possible mechanisms of targeting the inhibition of the growth and proliferation regulatory pathways of pancreatic cancer cells, inducing apoptosis and autophagy of pancreatic cancer cells, inhibiting the EMT process of pancreatic cancer cells, and inhibiting the angiogenesis of pancreatic cancer. Meanwhile, natural products have also hindered the progress of their basic and clinical research due to the complexity of their composition and the limitation of biological extraction technology. Further exploration of the specific molecular mechanisms of natural products to inhibit the proliferation and invasion of pancreatic cancer cells, optimization of purification and preparation techniques, and enrichment of basic and clinical trials to verify their efficacy and safety may be the future direction of natural products in the field of anti-pancreatic cancer research.
    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,在理解chrysin(CH)的潜在抗癌作用方面取得了显著进展,一种天然存在的类黄酮化合物,在各种植物来源如蜂蜜中大量发现,蜂胶,和某些水果和蔬菜。这种活性化合物由于其具有良好的治疗性能和最小的毒性而引起了极大的关注。CH抵抗癌症的能力源于其多方面的作用机制,包括细胞凋亡的启动和增殖的抑制,血管生成,转移,和细胞周期进程。CH还显示有效的抗氧化和抗炎特性,有效抵消导致DNA损伤和癌症发展的有害分子。此外,CH表现出了使癌细胞对传统化疗和放疗敏感的潜力,放大这些治疗的有效性,同时减少它们对健康细胞的负面影响。因此,在当前的审查中,组成,化学,行动机制,CH的安全问题,以及其纳米配方的可行性。最后,最近对CH的抗癌作用的调查提出了一个令人信服的一瞥这种天然化合物作为一系列抗癌方法的补充治疗元素的潜力,提供一种更安全、更全面的方法来对抗这种毁灭性的疾病。
    In recent times, there have been notable advancements in comprehending the potential anti-cancer effects of chrysin (CH), a naturally occurring flavonoid compound found abundantly in various plant sources like honey, propolis, and certain fruits and vegetables. This active compound has garnered significant attention due to its promising therapeutic qualities and minimal toxicity. CH\'s ability to combat cancer arises from its multifaceted mechanisms of action, including the initiation of apoptosis and the inhibition of proliferation, angiogenesis, metastasis, and cell cycle progression. CH also displays potent antioxidant and anti-inflammatory properties, effectively counteracting the harmful molecules that contribute to DNA damage and the development of cancer. Furthermore, CH has exhibited the potential to sensitize cancer cells to traditional chemotherapy and radiotherapy, amplifying the effectiveness of these treatments while reducing their negative impact on healthy cells. Hence, in this current review, the composition, chemistry, mechanisms of action, safety concerns of CH, along with the feasibility of its nanoformulations. To conclude, the recent investigations into CH\'s anti-cancer effects present a compelling glimpse into the potential of this natural compound as a complementary therapeutic element in the array of anti-cancer approaches, providing a safer and more comprehensive method of combating this devastating ailment.
    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
    血管生成是形成新血管的生理过程,在癌症等看似无关的疾病中具有病理重要性,糖尿病,和各种炎症性疾病。针对血管生成的治疗对这些类型的疾病有希望,但目前的抗血管生成药物在递送和副作用方面具有严重的局限性。这需要探索替代方法,如基于生物分子的药物。蛋白质,脂质,寡核苷酸最近在生物医学中变得流行,特别是作为治疗药物的生物相容性成分。其优异的生物利用度和潜在的生物活性和免疫原性特性使其成为药物发现或药物递送系统的主要候选者。基于脂质的脂质体已成为靶向纳米颗粒(NP)递送的标准载体,而蛋白质和核苷酸NP显示出作为智能NP的环境敏感性递送的希望。它们的治疗应用最初受到循环时间短和制造困难的阻碍,但纳米加工和NP工程的最新发展已经找到了规避这些缺点的方法。大大提高了生物分子NP的实用性。在这次审查中,我们将简要讨论基于生物分子的NP如何改善基于抗血管生成的治疗.
    Angiogenesis is a physiological process of forming new blood vessels that has pathological importance in seemingly unrelated illnesses like cancer, diabetes, and various inflammatory diseases. Treatment targeting angiogenesis has shown promise for these types of diseases, but current anti-angiogenic agents have critical limitations in delivery and side-effects. This necessitates exploration of alternative approaches like biomolecule-based drugs. Proteins, lipids, and oligonucleotides have recently become popular in biomedicine, specifically as biocompatible components of therapeutic drugs. Their excellent bioavailability and potential bioactive and immunogenic properties make them prime candidates for drug discovery or drug delivery systems. Lipid-based liposomes have become standard vehicles for targeted nanoparticle (NP) delivery, while protein and nucleotide NPs show promise for environment-sensitive delivery as smart NPs. Their therapeutic applications have initially been hampered by short circulation times and difficulty of fabrication but recent developments in nanofabrication and NP engineering have found ways to circumvent these disadvantages, vastly improving the practicality of biomolecular NPs. In this review, we are going to briefly discuss how biomolecule-based NPs have improved anti-angiogenesis-based 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
    血管生成在肿瘤进展中起关键作用,特别是在黑色素瘤中,最致命的皮肤癌.这篇综述综合了有关黑色素瘤进展中血管生成与肿瘤微环境(TME)之间复杂相互作用的最新知识。促血管生成因子,包括VEGF,PlGF,FGF-2,IL-8,Ang,TGF-β,PDGF,整合素,MMPs,PAF,调节血管生成并有助于黑素瘤转移。此外,TME内的细胞,如癌症相关的成纤维细胞,肥大细胞,和黑色素瘤相关的巨噬细胞,影响肿瘤血管生成和进展。抗血管生成疗法,在显示承诺的同时,面临诸如耐药性和肿瘤诱导的替代血管生成途径激活等挑战。正在探索抗血管生成剂和免疫疗法的合理组合以克服抗性。用于治疗反应的生物标志物识别对于个性化治疗仍然至关重要。这篇综述强调了黑色素瘤中血管生成的复杂性,并强调了对针对动态TME的创新治疗方法的需求。
    Angiogenesis plays a pivotal role in tumor progression, particularly in melanoma, the deadliest form of skin cancer. This review synthesizes current knowledge on the intricate interplay between angiogenesis and tumor microenvironment (TME) in melanoma progression. Pro-angiogenic factors, including VEGF, PlGF, FGF-2, IL-8, Ang, TGF-β, PDGF, integrins, MMPs, and PAF, modulate angiogenesis and contribute to melanoma metastasis. Additionally, cells within the TME, such as cancer-associated fibroblasts, mast cells, and melanoma-associated macrophages, influence tumor angiogenesis and progression. Anti-angiogenic therapies, while showing promise, face challenges such as drug resistance and tumor-induced activation of alternative angiogenic pathways. Rational combinations of anti-angiogenic agents and immunotherapies are being explored to overcome resistance. Biomarker identification for treatment response remains crucial for personalized therapies. This review highlights the complexity of angiogenesis in melanoma and underscores the need for innovative therapeutic approaches tailored to the dynamic TME.
    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
    迫切需要肝细胞癌(HCC)的新治疗方式,鉴于缺乏特异性,严重的副作用,和单一化疗耐药。工程化细菌可以靶向并在肿瘤组织中积累,诱导免疫反应,充当药物输送工具。然而,传统的细菌疗法有局限性,如药物装载能力和货物释放困难,导致治疗结果不足。合成生物技术可以提高基于细菌的递送系统的精度和功效。这使得治疗有效负载在体内的选择性释放成为可能。
    在这项研究中,我们构建了一种非致病性大肠杆菌(E.大肠杆菌)与同步裂解回路作为药物/基因递送载体和原位(乙型肝炎表面抗原)Ag(ASEC)生产者。将负载有质粒编码的人硫酸酯酶1(hsulf-1)酶(PNP)的聚乙二醇(CHO-PEG2000-CHO)-聚(乙烯亚胺)(PEI25k)-柠康酸酐(CA)-多柔比星(DOX)纳米颗粒锚定在ASEC(ASEC@PNP)的表面。合成并表征了复合材料。在HepG2细胞系和小鼠皮下肿瘤模型中测试了ASEC@PNP的体外和体内抗肿瘤作用。
    结果表明,静脉注射荷瘤小鼠后,ASEC可以主动靶向和定植肿瘤部位。裂解基因实现Ag的爆发和集中释放显着增加了细胞因子的分泌和肿瘤内CD4/CD8T细胞的浸润,引发了特定的免疫反应。同时,PNP系统将hsulf-1和DOX释放到肿瘤细胞中,从而导致快速的肿瘤消退和预防转移。
    新型药物递送系统在体内显着抑制HCC,副作用减少,表明临床肝癌治疗的潜在策略。
    UNASSIGNED: New treatment modalities for hepatocellular carcinoma (HCC) are desperately critically needed, given the lack of specificity, severe side effects, and drug resistance with single chemotherapy. Engineered bacteria can target and accumulate in tumor tissues, induce an immune response, and act as drug delivery vehicles. However, conventional bacterial therapy has limitations, such as drug loading capacity and difficult cargo release, resulting in inadequate therapeutic outcomes. Synthetic biotechnology can enhance the precision and efficacy of bacteria-based delivery systems. This enables the selective release of therapeutic payloads in vivo.
    UNASSIGNED: In this study, we constructed a non-pathogenic Escherichia coli (E. coli) with a synchronized lysis circuit as both a drug/gene delivery vehicle and an in-situ (hepatitis B surface antigen) Ag (ASEc) producer. Polyethylene glycol (CHO-PEG2000-CHO)-poly(ethyleneimine) (PEI25k)-citraconic anhydride (CA)-doxorubicin (DOX) nanoparticles loaded with plasmid encoded human sulfatase 1 (hsulf-1) enzyme (PNPs) were anchored on the surface of ASEc (ASEc@PNPs). The composites were synthesized and characterized. The in vitro and in vivo anti-tumor effect of ASEc@PNPs was tested in HepG2 cell lines and a mouse subcutaneous tumor model.
    UNASSIGNED: The results demonstrated that upon intravenous injection into tumor-bearing mice, ASEc can actively target and colonise tumor sites. The lytic genes to achieve blast and concentrated release of Ag significantly increased cytokine secretion and the intratumoral infiltration of CD4/CD8+T cells, initiated a specific immune response. Simultaneously, the PNPs system releases hsulf-1 and DOX into the tumor cell resulting in rapid tumor regression and metastasis prevention.
    UNASSIGNED: The novel drug delivery system significantly suppressed HCC in vivo with reduced side effects, indicating a potential strategy for clinical HCC 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
    胰腺导管腺癌(PDAC)中普遍存在的纤维增生性基质和免疫抑制微环境对治疗干预构成了重大挑战。尽管蛋白酪氨酸激酶(PTK)抑制剂在减轻促增生基质反应和增强免疫环境方面具有潜力,他们的疗效是由次优的药代动力学(PK)和不足的肿瘤渗透减少。为了克服这些障碍,我们开创了一个新的战略,采用脂质双层包覆的介孔二氧化硅纳米颗粒(称为“硅质体”)作为递送尼达尼布的载体。Nintedanib,一种靶向血管内皮生长因子的三联PTK抑制剂,血小板源性生长因子和成纤维细胞生长因子受体,通过弱碱的远程加载机制封装在硅体的孔中。这种创新的方法不仅增强了药代动力学和肿瘤内药物浓度,而且还在强大的原位KRAS介导的胰腺癌(KPC)模型中协调了组织增生和免疫景观的转变。我们的结果表明,通过封装的Nintedanib治疗,血管密度和胶原蛋白含量的衰减,伴随着CD8+/FoxP3+T细胞比率的显着增加。在KPC模型中,这种重塑与肿瘤消退显著相关。引人注目的是,封装的尼达尼布与抗PD-1免疫疗法之间的协同作用进一步增强了抗肿瘤作用。游离和包封的Nintedanib均通过细胞外信号调节激酶(ERK)途径诱导PD-L1的转录上调。总之,我们的开创性方法涉及硅质体载体,不仅改善了抗肿瘤血管生成,而且深刻地重塑了PDAC内的促生长基质和免疫景观。这些见解为PDAC治疗中创新组合策略的开发提供了极好的前景。
    The prevailing desmoplastic stroma and immunosuppressive microenvironment within pancreatic ductal adenocarcinoma (PDAC) pose substantial challenges to therapeutic intervention. Despite the potential of protein tyrosine kinase (PTK) inhibitors in mitigating the desmoplastic stromal response and enhancing the immune milieu, their efficacy is curtailed by suboptimal pharmacokinetics (PK) and insufficient tumor penetration. To surmount these hurdles, we have pioneered a novel strategy, employing lipid bilayer-coated mesoporous silica nanoparticles (termed \"silicasomes\") as a carrier for the delivery of Nintedanib. Nintedanib, a triple PTK inhibitor that targets vascular endothelial growth factor, platelet-derived growth factor and fibroblast growth factor receptors, was encapsulated in the pores of silicasomes via a remote loading mechanism for weak bases. This innovative approach not only enhanced pharmacokinetics and intratumor drug concentrations but also orchestrated a transformative shift in the desmoplastic and immune landscape in a robust orthotopic KRAS-mediated pancreatic carcinoma (KPC) model. Our results demonstrate attenuation of vascular density and collagen content through encapsulated Nintedanib treatment, concomitant with significant augmentation of the CD8+/FoxP3+ T-cell ratio. This remodeling was notably correlated with tumor regression in the KPC model. Strikingly, the synergy between encapsulated Nintedanib and anti-PD-1 immunotherapy further potentiated the antitumor effect. Both free and encapsulated Nintedanib induced a transcriptional upregulation of PD-L1 via the extracellular signal-regulated kinase (ERK) pathway. In summary, our pioneering approach involving the silicasome carrier not only improved antitumor angiogenesis but also profoundly reshaped the desmoplastic stromal and immune landscape within PDAC. These insights hold excellent promise for the development of innovative combinatorial strategies in PDAC therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号