Peptibody

  • 文章类型: Journal Article
    化疗诱导的血小板减少症(CIT)是癌症治疗的常见挑战,可导致化疗剂量减少,延迟,和/或停药,影响相对剂量强度,并可能对癌症护理产生不利影响。除了改变抗癌方案,CIT的标准管理仅限于血小板输注和支持治疗.使用血小板生成素受体激动剂romiplostim,已经批准用于免疫性血小板减少症,在CIT.中显示出有希望的疗效迹象。在一项针对实体瘤患者的2期前瞻性随机研究中,由于CIT,血小板计数<100×109/L≥4周,每周一次romiplostim将93%(14/15)的患者在3周内的血小板计数校正为>100×109/L,而未治疗的患者为12.5%(1/8)(p<0.001)。将接受romiplostim治疗的患者包括在一个额外的单臂队列中,所有接受romiplostim治疗的患者中有85%(44/52)在3周内对血小板计数进行了校正。一些CIT的回顾性研究也显示了对每周romiplostim的反应,最大的研究发现,肿瘤对骨髓的侵袭可以预测对romiplostim的反应不佳(比值比,0.029;95%CI:0.0046-0.18;p<0.001),既往骨盆照射(赔率比,0.078;95%CI:0.0062-0.98;p=0.048),和先前的替莫唑胺治疗(比值比0.24;95%CI:0.061-0.96;p=0.043)。在其他地方,较低的基线TPO水平可预测romiplostim反应(p=0.036).romiplostimcCIT研究没有出现新的安全信号。最近的治疗指南,包括来自国家综合癌症网络的,现在支持在CIT中使用romiplostim的考虑。数据预计来自两个正在进行的3期romiplostimCIT试验。
    Chemotherapy-induced thrombocytopenia (CIT) is a common challenge of cancer therapy and can lead to chemotherapy dose reduction, delay, and/or discontinuation, affecting relative dose intensity, and possibly adversely impacting cancer care. Besides changing anticancer regimens, standard management of CIT has been limited to platelet transfusions and supportive care. Use of the thrombopoietin receptor agonist romiplostim, already approved for use in immune thrombocytopenia, has shown promising signs of efficacy in CIT. In a phase 2 prospective randomized study of solid tumor patients with platelet counts <100 × 109/L for ≥4 weeks due to CIT, weekly romiplostim corrected the platelet count to >100 × 109/L in 93% (14/15) of patients within 3 weeks versus 12.5% (1/8) of untreated patients (p < 0.001). Including patients treated with romiplostim in an additional single-arm cohort, 85% (44/52) of all romiplostim-treated patients responded with platelet count correction within 3 weeks. Several retrospective studies of CIT have also shown responses to weekly romiplostim, with the largest study finding that poor response to romiplostim was predicted by tumor invasion of the bone marrow (odds ratio, 0.029; 95% CI: 0.0046-0.18; p < 0.001), prior pelvic irradiation (odds ratio, 0.078; 95% CI: 0.0062-0.98; p = 0.048), and prior temozolomide treatment (odds ratio 0.24; 95% CI: 0.061-0.96; p = 0.043). Elsewhere, lower baseline TPO levels were predictive of romiplostim response (p = 0.036). No new safety signals have emerged from romiplostim CIT studies. Recent treatment guidelines, including those from the National Comprehensive Cancer Network, now support consideration of romiplostim use in CIT. Data are expected from two ongoing phase 3 romiplostim CIT trials.
    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
    体外蛋白质重折叠是制备融合蛋白(例如使用大肠杆菌表达的肽体)中的关键限速单元操作之一。稀释辅助重折叠是最常用的工业实践,以实现可溶性,来自包涵体的重组蛋白的天然功能形式。这项研究的重点是开发一种色谱辅助的体外重折叠平台,以产生生物活性,重组肽体的天然形式。选择重组Romiplostim作为研究的模型蛋白。将塞流管式反应器与捕获步骤亲和色谱法串联连接,以同时实现重组Romiplostim的体外重折叠和捕获步骤纯化。各种关键工艺参数的影响,如倍数稀释,温度,停留时间,使用基于中心复合物的实验策略设计来研究半胱氨酸:DTT比率,以实现所选择的肽体的最大重折叠产率。在最佳重折叠条件下,在25倍稀释时达到57.0±1.5%的最大重折叠产率和超过79.73±3.4%的纯度,温度15°C,6小时的停留时间与6mM和10mM的半胱氨酸和DTT,分别。使用各种正交分析工具检查天然肽体结构的形成,以研究蛋白质的初级,次要,和三级结构。使用碰撞诱导解离(CID)对二硫键连接的肽的氨基酸序列进行定位,以确认Cys7-Cys7和Cys10-Cys10之间的链内二硫键的形成,类似于Cys42-Cys102和Cys148-Cys206之间的链内二硫键。这里开发的方案是鉴定涉及域间二硫键的多域蛋白的高产率可扩展重折叠条件的有价值的工具。
    In-vitro protein refolding is one of the key rate-limiting unit operations in manufacturing of fusion proteins such as peptibodies expressed using E. coli. Dilution-assisted refolding is the most commonly used industrial practice to achieve the soluble, native functional form of the recombinant protein from the inclusion bodies. This study is focused on developing a chromatography-assisted in-vitro refolding platform to produce the biologically active, native form of recombinant peptibody. Recombinant Romiplostim was selected as a model protein for the study. A plug flow tubular reactor was connected in series with capture step affinity chromatography to achieve simultaneous in-vitro refolding and capture step purification of recombinant Romiplostim. Effect of various critical process parameters like fold dilution, temperature, residence time, and Cysteine: DTT ratio was studied using a central composite based design of experiment strategy to achieve a maximum refolding yield of selected peptibody. Under optimum refolding conditions, the maximum refolding yield of 57.0 ± 1.5 % and a purity of over 79.73 ± 3.4 % were achieved at 25-fold dilution, 15 °C temperature, 6 h residence time with 6 mM and 10 mM of cysteine and DTT, respectively. The formation of native peptibody structure was examined using various orthogonal analytical tools to study the protein\'s primary, secondary, and tertiary structure. The amino acid sequence for the disulfide-linked peptide was mapped using collision-induced dissociation (CID) to confirm the formation of interchain disulfide bonds between Cys7-Cys7 and Cys10-Cys10 similarly for intra-chain disulfide bonds between Cys42-Cys102, and Cys148-Cys206. The developed protocol here is a valuable tool to identify high-yield scalable refolding conditions for multi-domain proteins involving inter-domain disulfide bonds.
    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
    背景:人表皮生长因子受体2(HER2)靶向策略的临床功效由于免疫抑制细胞负调节的抗肿瘤应答受损而受到限制。因此,我们,在4个T1-HER2肿瘤模型中,研究了抗HER2单克隆抗体(1个T0mAb)与CD11b/Gr-1骨髓细胞耗竭的抑制作用。
    方法:BALB/c小鼠用表达人HER2的4T1鼠乳腺癌细胞系攻击。肿瘤挑战后一周,每只小鼠每隔一天接受50微克的骨髓细胞特异性肽体,或10毫克/千克1T0单克隆抗体,每周两次,和他们的组合两个星期。通过计算肿瘤大小来测量治疗对肿瘤生长的影响。此外,流式细胞术检测CD11b+/Gr-1+细胞和T淋巴细胞的频率。
    结果:肽体处理的小鼠显示肿瘤消退,40%的小鼠根除其原发性肿瘤。肽体能够显着消耗脾CD11b/Gr-1细胞以及肿瘤内CD11b/Gr-1细胞(P<0.0001),并导致肿瘤浸润的CD8T细胞数量增加(3.3倍)以及常驻肿瘤引流淋巴结(TDLN)(3倍)。肽体和1T0mAb的组合导致肿瘤浸润CD4+和CD8+T细胞的扩增增强,这与60%的小鼠中的肿瘤根除相关。
    结论:Peptibody能够消耗CD11b/Gr-1细胞,并增加1T0mAb在肿瘤根除中的抗肿瘤作用。因此,这种髓样群体在肿瘤的发展中起关键作用,它们的消耗与抗肿瘤反应的诱导相关.
    BACKGROUND: Clinical efficacy of Human Epidermal growth factor Receptor 2 (HER2) targeted strategies is limited due to impaired anti-tumor responses negatively regulated by immunosuppressive cells. We thus, investigated the inhibitory effects of an anti-HER2 monoclonal antibody (1 T0 mAb) in combination with CD11b+/Gr-1+ myeloid cells depletion in 4 T1-HER2 tumor model.
    METHODS: BALB/c mice were challenged with human HER2-expressing 4 T1 murine breast cancer cell line. A week post tumor challenge, each mouse received 50 µg of a myeloid cells specific peptibody every other day, or 10 mg/kg of 1 T0 mAb two times a week, and their combination for two weeks. The treatments effect on tumor growth was measured by calculating tumor size. Also, the frequencies of CD11b+/Gr-1+ cells and T lymphocytes were measured by flow cytometry.
    RESULTS: Peptibody treated mice indicated tumor regression and 40 % of the mice eradicated their primary tumors. The peptibody was capable to deplete notably splenic CD11b+/Gr-1+ cells as well as intratumoral CD11b+/Gr-1+ cells (P < 0.0001) and led to an increased number of tumor infiltrating CD8+ T cells (3.3 folds) and also that of resident tumor draining lymph nodes (TDLNs) (3 folds). Combination of peptibody and 1 T0 mAb resulted in enhanced expansion of tumor infiltrating CD4 + and CD8+ T cells which was associated with tumor eradication in 60 % of the mice.
    CONCLUSIONS: Peptibody is able to deplete CD11b+/Gr-1+ cells and increase anti-tumoral effects of the 1 T0 mAb in tumor eradication. Thus, this myeloid population have critical roles in development of tumors and their depletion is associated with induction of anti-tumoral responses.
    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
    我们设计并生产了离子通道阻断肽体,目标是乙酰胆碱激活的向内整流钾电流(IKACh)。肽体是通过将生物活性肽与人免疫球蛋白G(IgG)的片段可结晶(Fc)区融合而产生的嵌合蛋白。IKACh阻断肽体被设计为人IgG1Fc片段和IKACh抑制剂tertiapinQ(TP)之间的融合体,一种21个氨基酸的合成肽毒素,最初是从欧洲蜜蜂毒液中分离出来的。从用肽体构建体转染的人胚肾(HEK)细胞的培养上清液中纯化肽体。我们测试了生物工程肽体具有生物活性和IKACh的有效阻断剂的假设。在用Kir3.1和Kir3.4转染的HEK细胞中,IKACh的分子相关性,膜片钳显示,肽体的效力比TP高300倍。分子动力学模拟表明,增加的效力可能是由于与tertiapin-Kir3.1/3.4通道相比,肽体-Kir3.1/3.4通道形成的复合物的稳定性增加。在分离的小鼠肌细胞中,肽体阻断了心房细胞中卡巴胆碱(Cch)激活的IKACh,但不影响心室肌细胞中的钾向内整流背景电流。在麻醉小鼠中,肽体消除了腹膜内注射Cch的心动过缓作用。此外,在老年小鼠中,肽体降低了心房颤动的诱导性,可能通过阻断组成型活性IKACh。生物工程抗离子通道肽体可以是强大和高度有效的离子通道阻断剂,有可能指导离子通道调节剂或抗心律失常方式的发展。
    We engineered and produced an ion channel blocking peptibody, that targets the acetylcholine-activated inwardly rectifying potassium current (IKACh). Peptibodies are chimeric proteins generated by fusing a biologically active peptide with the fragment crystallizable (Fc) region of the human immunoglobulin G (IgG). The IKACh blocking peptibody was engineered as a fusion between the human IgG1 Fc fragment and the IKACh inhibitor tertiapinQ (TP), a 21-amino acid synthetic peptidotoxin, originally isolated from the European honey bee venom. The peptibody was purified from the culture supernatant of human embryonic kidney (HEK) cells transfected with the peptibody construct. We tested the hypothesis that the bioengineered peptibody is bioactive and a potent blocker of IKACh. In HEK cells transfected with Kir3.1 and Kir3.4, the molecular correlates of IKACh, patch clamp showed that the peptibody was ~300-fold more potent than TP. Molecular dynamics simulations suggested that the increased potency could be due to an increased stabilization of the complex formed by peptibody-Kir3.1/3.4 channels compared to tertiapin-Kir3.1/3.4 channels. In isolated mouse myocytes, the peptibody blocked carbachol (Cch)-activated IKACh in atrial cells but did not affect the potassium inwardly rectifying background current in ventricular myocytes. In anesthetized mice, the peptibody abrogated the bradycardic effects of intraperitoneal Cch injection. Moreover, in aged mice, the peptibody reduced the inducibility of atrial fibrillation, likely via blocking constitutively active IKACh. Bioengineered anti-ion channel peptibodies can be powerful and highly potent ion channel blockers, with the potential to guide the development of modulators of ion channels or antiarrhythmic modalities.
    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
    Notch信号通路是治疗炎症性疾病和癌症的重要治疗靶点。我们之前创建了Notch信号的配体特异性抑制剂,该抑制剂由Fc融合到Notch1胞外域的特异性EGF样重复序列组成。叫做Notch诱饵,结合配体,阻塞的陷波信号,并显示出抗肿瘤活性,低毒性。然而,其功能的研究依赖于病毒介导的表达,这排除了剂量控制和限制临床适用性。我们改进了诱饵设计,以创建包含核心结合域的基于肽体的Notch抑制剂,Notch1或Notch4的EGF样重复10-14。这些Notch肽体显示出高分泌特性和产量,与以前的Notch诱饵相比提高了近100倍。使用表面等离子体共振光谱结合免疫共沉淀测定,我们观察到Notch1和Notch4肽体表现出对Notch配体DLL4和JAG1的强但不同的结合性质。Notch1和Notch4肽体均干扰内皮细胞中的Notch信号传导并在治疗后降低典型Notch靶标的表达。虽然以前的DLL4抑制剂会导致过度发芽,Notch1肽体在三维体外发芽测定中减少了血管生成。对新生小鼠施用Notch1肽体导致视网膜脉管系统的径向生长减少,确认抗血管生成特性。我们得出的结论是,包含EGF样重复10-14的纯化的Notch肽体结合DLL4和JAG1配体并表现出抗血管生成特性。根据它们的分泌情况,独特的Notch抑制活性,和抗血管生成特性,Notch肽体为治疗性Notch抑制提供了新的机会。
    The Notch signaling pathway is an important therapeutic target for the treatment of inflammatory diseases and cancer. We previously created ligand-specific inhibitors of Notch signaling comprised of Fc fusions to specific EGF-like repeats of the Notch1 extracellular domain, called Notch decoys, which bound ligands, blocked Notch signaling, and showed anti-tumor activity with low toxicity. However, the study of their function depended on virally mediated expression, which precluded dosage control and limited clinical applicability. We have refined the decoy design to create peptibody-based Notch inhibitors comprising the core binding domains, EGF-like repeats 10-14, of either Notch1 or Notch4. These Notch peptibodies showed high secretion properties and production yields that were improved by nearly 100-fold compared to previous Notch decoys. Using surface plasmon resonance spectroscopy coupled with co-immunoprecipitation assays, we observed that Notch1 and Notch4 peptibodies demonstrate strong but distinct binding properties to Notch ligands DLL4 and JAG1. Both Notch1 and Notch4 peptibodies interfere with Notch signaling in endothelial cells and reduce expression of canonical Notch targets after treatment. While prior DLL4 inhibitors cause hyper-sprouting, the Notch1 peptibody reduced angiogenesis in a 3-dimensional in vitro sprouting assay. Administration of Notch1 peptibodies to neonate mice resulted in reduced radial outgrowth of retinal vasculature, confirming anti-angiogenic properties. We conclude that purified Notch peptibodies comprising EGF-like repeats 10-14 bind to both DLL4 and JAG1 ligands and exhibit anti-angiogenic properties. Based on their secretion profile, unique Notch inhibitory activities, and anti-angiogenic properties, Notch peptibodies present new opportunities for therapeutic Notch inhibition.
    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:髓系来源的抑制细胞(MDSC)是一种未成熟的异种髓系群体,可减弱抗肿瘤免疫反应。MDSC的耗尽已被证明可提高癌症免疫治疗方法的功效。这里,我们表达并表征了一种先前通过噬菌体展示技术定义的能够识别和消耗鼠MDSCs的肽体。
    方法:使用重叠延伸(SOE)PCR剪接,合成MDSC结合肽和接头的编码序列,然后连接到含有小鼠IgG2aFc的自制表达质粒中。通过脂质体3000试剂将肽体构建体转染到CHO-K1细胞中,并用蛋白G柱纯化所得的融合蛋白,然后通过ELISA表征。SDS-PAGE和免疫印迹。然后通过流式细胞术测试肽体与脾MDSC的结合谱及其MDSC消耗能力。
    结果:纯化的肽体在Western印迹中出现70KDa条带。它可以与98.8%的脾CD11b+/Gr-1+MDSCs结合。此外,与PBS处理的阴性对照对应物相比,肽体处理后肿瘤内MDSCs显著耗尽(P<0.05).
    结论:在这项研究中,能够消耗肿瘤内MDSCs的肽体,成功表达和纯化。我们的结果暗示它可以被认为是癌症免疫治疗研究的潜在工具。
    BACKGROUND: Myeloid derived suppressor cells (MDSCs) are an immature heterogeneous population of myeloid lineage that attenuate the anti-tumor immune responses. Depletion of MDSCs has been shown to improve efficacy of cancer immunotherapeutic approaches. Here, we expressed and characterized a peptibody which had previously been defined by phage display technique capable of recognizing and depleting murine MDSCs.
    METHODS: Using splicing by overlap extension (SOE) PCR, the coding sequence of the MDSC binding peptide and linker were synthesized and then ligated into a home-made expression plasmid containing mouse IgG2a Fc. The peptibody construct was transfected into CHO-K1 cells by lipofectamine 3000 reagent and the resulting fusion protein was purified with protein G column and subsequently characterized by ELISA, SDS-PAGE and immunoblotting. The binding profile of the peptibody to splenic MDSCs and its MDSC depletion ability were then tested by flow cytometry.
    RESULTS: The purified peptibody appeared as a 70 KDa band in Western blot. It could bind to 98.8% of splenic CD11b+/Gr-1+ MDSCs. In addition, the intratumoral MDSCs were significantly depleted after peptibody treatment compared to their PBS-treated negative control counterparts (P < 0.05).
    CONCLUSIONS: In this study, a peptibody capable of depleting intratumoral MDSCs, was successfully expressed and purified. Our results imply that it could be considered as a potential tool for research on 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
    We describe, for the first time, a new splice variant of the human TGF-β type II receptor (TβRII). The new transcript lacks 149 nucleotides, resulting in a frameshift and the emergence of an early stop codon, rendering a truncated mature protein of 57 amino acids. The predicted protein, lacking the transmembrane domain and with a distinctive 13-amino-acid stretch at its C-terminus, was named TβRII-Soluble Endogenous (TβRII-SE). Binding predictions indicate that the novel 13-amino-acid stretch interacts with all three TGF-β cognate ligands and generates a more extensive protein-protein interface than TβRII. TβRII-SE and human IgG1 Fc domain were fused in frame in a lentiviral vector (Lv) for further characterization. With this vector, we transduced 293T cells and purified TβRII-SE/Fc by A/G protein chromatography from conditioned medium. Immunoblotting revealed homogeneous bands of approximately 37 kDa (reduced) and 75 kDa (non-reduced), indicating that TβRII-SE/Fc is secreted as a disulfide-linked homodimer. Moreover, high-affinity binding of TβRII-SE to the three TGF-β isoforms was confirmed by surface plasmon resonance (SPR) analysis. Also, intrahepatic delivery of Lv.TβRII-SE/Fc in a carbon tetrachloride-induced liver fibrosis model revealed amelioration of liver injury and fibrosis. Our results indicate that TβRII-SE is a novel member of the TGF-β signaling pathway with distinctive characteristics. This novel protein offers an alternative for the prevention and treatment of pathologies caused by the overproduction of TGF-β ligands.
    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
    成纤维细胞生长因子受体(FGFR)是癌症定向治疗的新兴靶标。这里提出了一种新的FGFR1靶向缀合物,使用肽体的肽体F2,肽与人IgG的Fc片段的融合物作为选择性靶向剂和药物载体。基于FGF2序列的短肽用于构建靶向FGFR1的肽体。我们已经表明,该肽确保将肽F2特异性递送到表达FGFR1的细胞中。为了使用peptibodyF2作为细胞毒性药物的递送载体,我们已经把它和MMAE结合了,一种广泛用于靶向治疗的抗体-药物缀合物的药物。所得的缀合物显示对FGFR1阳性细胞的高和特异性细胞毒性,即,鳞状细胞肺癌NCI-H520,同时对FGFR1阴性细胞保持无毒。这种肽体-药物缀合物可以作为开发具有过度表达或功能不良的FGFRs的肿瘤的疗法的基础。
    Fibroblast growth factor receptors (FGFRs) are emerging targets for directed cancer therapy. Presented here is a new FGFR1-targeting conjugate, the peptibodyF2, which employs peptibody, a fusion of peptide and the Fc fragment of human IgG as a selective targeting agent and drug carrier. Short peptide based on FGF2 sequence was used to construct a FGFR1-targeting peptibody. We have shown that this peptide ensures specific delivery of peptibodyF2 into FGFR1-expressing cells. In order to use peptibodyF2 as a delivery vehicle for cytotoxic drugs, we have conjugated it with MMAE, a drug widely used in antibody-drug conjugates for targeted therapy. Resulting conjugate shows high and specific cytotoxicity towards FGFR1-positive cells, i.e., squamous cell lung carcinoma NCI-H520, while remaining non-toxic for FGFR1-negative cells. Such peptibody-drug conjugate can serve as a basis for development of therapy for tumors with overexpressed or malfunctioning FGFRs.
    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
    在肿瘤组织中,碱性成纤维细胞生长因子(bFGF)和血管内皮生长因子A(VEGFA)通过激活血管生成促进肿瘤发生,但靶向单因子可能产生耐药性和代偿性血管生成。设计具有bFGF/VEGFA的肽体以同时阻断这两种因子。我们的目标是大规模生产这种Fc融合蛋白。将肽体菌株的生物学特性鉴定为大肠杆菌,并在摇瓶和10-L生物反应器中优化了发酵模式。发酵扩大到100升,湿细胞重量(WCW)126g/L,产量1.41g/L,IPTG诱导和生产率0.35g/(L·h)。通过阳离子交换分离目标蛋白,疏水和蛋白A色谱,总回收率为60.28%,HPLC纯度为86.71%。宿主细胞的蛋白质,DNA,内毒素残留在阈值内。在小鼠模型中,Peptibody疫苗的免疫可以显著抑制肿瘤的生长和血管生成,抑制率分别为57.73和39.34%。肽体疫苗能引发高滴度的抗bFGF和抗VEGFA抗体,通过降低Akt/MAPK信号通路抑制Lewis肺癌细胞的增殖和迁移。因此,具有bFGF/VEGFA的肽体可用作治疗性肿瘤疫苗。我们开发的大规模工艺可以支持其未来的工业生产和临床前研究。
    In tumor tissue, basic fibroblast growth factor (bFGF) and vascular endothelial growth factor A (VEGFA) promote tumorigenesis by activating angiogenesis, but targeting single factor may produce drug resistance and compensatory angiogenesis. The Peptibody with bFGF/VEGFA was designed to simultaneously blockade these two factors. We were aiming to produce this Fc fusion protein in a large scale. The biological characterizations of Peptibody strains were identified as Escherichia coli and the fermentation mode was optimized in the shake flasks and 10-L bioreactor. The fermentation was scaled up to 100 L, with wet cell weight (WCW) 126 g/L, production 1.41 g/L, and productivity 0.35 g/(L·h) of IPTG induction. The target protein was isolated by cation-exchange, hydrophobic and Protein A chromatography, with total recovery of 60.28% and HPLC purity of 86.71%. The host cells protein, DNA, and endotoxin residues were within the threshold. In mouse model, immunization of Peptibody vaccine could significantly suppressed the tumor growth and angiogenesis, with inhibition rate of 57.73 and 39.34%. The Peptibody vaccine could elicit high-titer anti-bFGF and anti-VEGFA antibodies, which inhibited the proliferation and migration of Lewis lung cancer cell cells by decreasing the Akt/MAPK signal pathways. Therefore, the Peptibody with bFGF/VEGFA might be used as a therapeutic tumor vaccine. The large-scale process we developed could support its industrial production and pre-clinical study in the future.
    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
    Tumor angiogenesis is dependent on growth factors, and inhibition of their pathways is one of the promising strategies in cancer therapy. However, resistance to single pathway has been a great concern in clinical trials so that it necessitates multiple targetable factors for developing tumor angiogenesis inhibitors. Moreover, the strategy of Fc fusion protein is an attractive platform for novel peptide agents, which gains increasing importance with FDA approval because of better immunogenicity and stability. Here, we applied the Fc fusion protein concept to bFGF/VEGFA pathways and designed a multi-epitope Peptibody with immunogenic peptides derived from human bFGF and VEGFA sequences. Immunization with Peptibody could elicit high-titer anti-bFGF and anti-VEGFA antibodies, activate T cells, and induce Th1/Th2-type cytokines. In in vitro experiments, the isolated anti-Peptibody antibody inhibited the proliferation and migration of A549 cells and human umbilical vein endothelial cells (HUVECs) by decreasing the MAPK/Akt/mTOR signal pathways. In the murine tumor model, pre-immunization with Peptibody suppressed the tumor growth and neovascularization of lung cancer by decreasing the production of bFGF/VEGFA/PDGF, the MAPK/Akt/mTOR signal pathways, and the activation of suppressive cells in tumor sites. Further, the biological characterizations of the recombinant Peptibody were investigated systematically, including protein primary structure, secondary structure, stability, and toxicity. Collectively, the results highlighted the strategy of bFGF/VEGFA pathways and Fc fusion protein in suppressing tumor progression and angiogenesis, which emphasized the potential of multiple targetable factors for producing enduring clinical responses in tumor patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号