Bispecific antibodies

双特异性抗体
  • 文章类型: Journal Article
    小细胞肺癌(SCLC)占所有肺癌诊断的约10%至15%,并且由于其高死亡率而代表了紧迫的全球公共卫生挑战。常规治疗SCLC的疗效欠佳,以有限的抗肿瘤作用和频繁复发为特征。在这种情况下,新兴的研究转向免疫疗法与化疗相结合,这是一个快速发展的领域,在改善SCLC患者的临床结局方面显示出希望。通过最初开发用于非小细胞肺癌(NSCLC),这些疗法拓展了SCLC的新治疗途径.目前,一系列新兴热点治疗方法已显示出显著的治疗效果.基于化疗和免疫治疗的融合,以及新的免疫治疗剂的开发,SCLC的治疗已经看到了希望的未来。通过伴随使用化疗,在增强肿瘤免疫微环境方面取得了进展。免疫疗法,和酪氨酸激酶抑制剂(TKI),正如新兴的临床试验数据所证明的那样。此外,涉及免疫疗法的三方方法,靶向治疗,化疗对未来的临床应用来说似乎是吉祥的。克服对免疫治疗后方案的耐药性仍然是一个紧迫的探索领域。最后,双特异性抗体,过继细胞转移(ACT),溶瘤病毒,单一疗法,包括Delta样配体3(DLL3)和具有Ig和ITIM结构域的T细胞免疫受体(TIGIT),以及精准医学,可能是在SCLC中实现治愈结果的前瞻性途径。这篇综述旨在综合现有文献,并强调SCLC治疗的未来方向,承认该领域的持续挑战。此外,新的治疗药物和技术的不断发展使得SCLC治疗的未来越来越乐观.
    Small cell lung cancer (SCLC) constitutes approximately 10% to 15% of all lung cancer diagnoses and represents a pressing global public health challenge due to its high mortality rates. The efficacy of conventional treatments for SCLC is suboptimal, characterized by limited anti-tumoral effects and frequent relapses. In this context, emerging research has pivoted towards immunotherapy combined with chemotherapy, a rapidly advancing field that has shown promise in ameliorating the clinical outcomes of SCLC patients. Through originally developed for non-small cell lung cancer (NSCLC), these therapies have extended new treatment avenues for SCLC. Currently, a nexus of emerging hot-spot treatments has demonstrated significant therapeutic efficacy. Based on the amalgamation of chemotherapy and immunotherapy, and the development of new immunotherapy agents, the treatment of SCLC has seen the hoping future. Progress has been achieved in enhancing the tumor immune microenvironment through the concomitant use of chemotherapy, immunotherapy, and tyrosine kinase inhibitors (TKI), as evinced by emerging clinical trial data. Moreover, a tripartite approach involving immunotherapy, targeted therapy, and chemotherapy appears auspicious for future clinical applications. Overcoming resistance to post-immunotherapy regimens remains an urgent area of exploration. Finally, bispecific antibodies, adoptive cell transfer (ACT), oncolytic virus, monotherapy, including Delta-like ligand 3 (DLL3) and T cell immunoreceptor with Ig and ITIM domains (TIGIT), as well as precision medicine, may present a prospective route towards achieving curative outcomes in SCLC. This review aims to synthesize extant literature and highlight future directions in SCLC treatment, acknowledging the persistent challenges in the field. Furthermore, the continual development of novel therapeutic agents and technologies renders the future of SCLC treatment increasingly optimistic.
    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
    近年来,在发现用于治疗中枢神经系统(CNS)疾病的治疗性蛋白质和肽方面取得了快速进展。然而,它们的临床应用受到诸如低生物膜渗透性等挑战的极大阻碍,稳定性差,循环时间短,和强大的血脑屏障(BBB)。最近,在了解BBB的动力学和开发将蛋白质和肽递送到CNS的有效方法方面已经取得了实质性的进步。特别是通过使用各种纳米粒子。在这里,我们概述了在生理和病理条件下对BBB的最新认识,强调它们对大脑药物输送的影响。我们总结了先进的策略,并阐明了将蛋白质和肽输送到大脑的潜在机制。我们重点介绍了纳米载体在通过BBB杂交治疗CNS疾病中的发展和应用。我们还对当前策略的局限性和障碍提出了批评意见,并对未来的研究提出了展望。
    Recent years have witnessed rapid progress in the discovery of therapeutic proteins and peptides for the treatment of central nervous system (CNS) diseases. However, their clinical applications have been considerably hindered by challenges such as low biomembrane permeability, poor stability, short circulation time, and the formidable blood-brain barrier (BBB). Recently, substantial improvements have been made in understanding the dynamics of the BBB and developing efficient approaches for delivering proteins and peptides to the CNS, especially by using various nanoparticles. Herein, we present an overview of the up-to-date understanding of the BBB under physiological and pathological conditions, emphasizing their effects on brain drug delivery. We summarize advanced strategies and elucidate the underlying mechanisms for delivering proteins and peptides to the brain. We highlight the developments and applications of nanocarriers in treating CNS diseases via BBB crossing. We also provide critical opinions on the limitations and obstacles of the current strategies and put forward prospects for future research.
    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
    越来越多的证据表明,放疗可能会增强免疫治疗的疗效。本研究旨在评估靶向PD-L1和CTLA-4的双特异性抗体KN046联合化疗和姑息性放疗治疗晚期食管鳞状细胞癌(ESCC)的可能性。在这个开放标签中,Ib期试验,晚期ESCC患者接受化疗和姑息性放疗,和KN046,预定义的递增剂量为1、3或5mg/kg(在化疗周期中每3周一次,在KN046维持期间每2周一次)。化疗方案包括顺铂(75mg/m2,d1)和紫杉醇(135-175mg/m2ivgtt。,d1).放射治疗细节,包括网站,定时,剂量,和碎片模式,由调查员自行决定。主要结果是剂量限制性毒性(DLT)。从2019年5月到2021年4月,在剂量组中招募了25名患者:1mg/kg中的3名,3mg/kg中的12,和10在5mg/kg。在剂量递增期间未观察到DLT。客观缓解率为41.7%(95CI22.1-63.4),而疾病控制率为87.5%(95CI67.6~97.3)。中位随访时间为11.8个月,中位无进展生存期为7.8个月(95CI5.2~9.7),中位总生存期为15.9个月(95CI8.4-NE).48.0%的患者报告了严重不良事件,主要是白细胞减少症(16%),免疫介导性小肠结肠炎(12%),免疫介导的肺炎(8%),和中性粒细胞减少(8%)。KN046联合化疗和姑息性放疗可能是可行的,在晚期ESCC患者中显示出良好的安全性和显着的疗效。
    There is growing evidence to suggest that radiotherapy might enhance the efficacy of immunotherapy. This study aimed to assess the possibility of KN046, a bispecific antibody targeting PD-L1 and CTLA-4, combined with chemotherapy and palliative radiotherapy for advanced esophageal squamous cell carcinoma (ESCC). In this open-label, phase Ib trial, patients with advanced ESCC were administered chemotherapy with palliative radiotherapy, and KN046 in the predefined escalation dosages of 1, 3, or 5 mg/kg (every 3 weeks during chemotherapy cycles and every 2 weeks during KN046 maintenance). The chemotherapy regimen constituted cisplatin (75 mg/m2 i.v., d1) and paclitaxel (135-175 mg/m2 ivgtt., d1). Radiotherapy specifics, including site, timing, dose, and fragmentation pattern, were at the investigator\'s discretion. The primary outcome was dose-limiting toxicity (DLT). From May 2019 to April 2021, 25 patients were enrolled across the dosage groups: 3 in 1 mg/kg, 12 in 3 mg/kg, and 10 in 5 mg/kg. No DLT was observed during the dose escalation. The objective response rate was 41.7% (95%CI 22.1-63.4), while the disease control rate was 87.5% (95%CI 67.6-97.3). At a median follow-up of 11.8 months, the median progression-free survival was 7.8 months (95%CI 5.2-9.7) and median overall survival was 15.9 months (95%CI 8.4-NE). Serious adverse events were reported in 48.0% of patients, predominantly leukopenia (16%), immune-mediated enterocolitis (12%), immune-mediated pneumonitis (8%), and neutropenia (8%). Combining KN046 with chemotherapy and palliative radiotherapy might be feasible, showing a favorable safety profile and notable efficacy in advanced ESCC 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
    基于关键的KarMma-1和CARTITUDE-1研究,Idecabtagenevicleucel(Ide-cel)和Ciltacabtageneautoleucel(Cilta-cel)已被批准用于治疗多发性骨髓瘤患者,接触过至少一种蛋白酶体抑制剂的人,免疫调节药物和抗CD38抗体治疗后4或3线,分别。同时,在多个现实世界分析中证实了前所未有的深度和持久缓解率,最近,KarMma-3和CARTITUDE-4研究导致在早期治疗中获得批准.目前认为,最终所有患有复发性/难治性多发性骨髓瘤的患者在抗BCMACART细胞疗法后经历复发。有大量针对新型抗原的CAR-T细胞疗法,旨在克服当前的CAR-T细胞抗性。在这次审查中,我们将总结目前新抗原的证据及其临床潜力。结合当前的CAR-T细胞疗法和T细胞接合器,这些方法可能会引导我们进入多发性骨髓瘤的下一个前沿:完全免疫疗法和无化疗治愈之路.
    Based on the pivotal KarMMa-1 and CARTITUDE-1 studies, Idecabtagene vicleucel (Ide-cel) and Ciltacabtagene autoleucel (Cilta-cel) have been approved to treat multiple myeloma patients, who have been exposed to at least 1 proteasome inhibitor, immunomodulatory drug and anti-CD38 antibody after 4 or 3 lines of therapy, respectively. The unprecedented rates of deep and long-lasting remissions have been meanwhile confirmed in multiple real-world analyses and more recently, the KarMMa-3 and CARTITUDE-4 studies lead to the approval in earlier lines of therapy. It is currently believed that ultimately all patients with relapsed/refractory multiple myeloma experience relapse after anti-BCMA CAR T-cell therapies. There is a plethora of CAR T-cell therapies targeting novel antigens, with the aim to overcome current CAR T-cell resistance. In this review, we will summarize current evidence of novel antigens and their clinical potential. Together with current CAR T-cell therapy and T-cell engagers, these approaches might lead us to the next frontier in multiple myeloma: total immunotherapy and the road to chemotherapy-free cure.
    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: Editorial
    暂无摘要。
    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
    靶向多种病毒蛋白对于持续抑制高度突变的病毒至关重要。近年来,已经开发了针对流感病毒血凝素和神经氨酸酶糖蛋白的广泛中和抗体,和抗体单一疗法已在临床前和临床研究中进行了测试,以治疗或预防流感病毒感染。然而,血凝素和神经氨酸酶双重中和对感染过程的影响,以及它的治疗潜力,还没有经过彻底的测试。为此,我们产生了一种能中和流感病毒血凝素和神经氨酸酶的双特异性抗体.我们证明了这种双特异性抗体具有双重抗病毒活性,因为它阻断感染并防止子代病毒从感染细胞中释放。我们表明,双特异性抗体对血凝素和神经氨酸酶的双重中和优于单克隆抗体组合,因为它提高了中和能力并增强了抗体效应子功能。值得注意的是,双特异性抗体在流感病毒感染的小鼠中显示出增强的抗病毒活性,降低小鼠死亡率并限制抗体施用后的病毒突变谱。因此,血凝素和神经氨酸酶的双重中和可有效控制流感病毒感染。
    Targeting multiple viral proteins is pivotal for sustained suppression of highly mutable viruses. In recent years, broadly neutralizing antibodies that target the influenza virus hemagglutinin and neuraminidase glycoproteins have been developed, and antibody monotherapy has been tested in preclinical and clinical studies to treat or prevent influenza virus infection. However, the impact of dual neutralization of the hemagglutinin and neuraminidase on the course of infection, as well as its therapeutic potential, has not been thoroughly tested. For this purpose, we generated a bispecific antibody that neutralizes both the hemagglutinin and the neuraminidase of influenza viruses. We demonstrated that this bispecific antibody has a dual-antiviral activity as it blocks infection and prevents the release of progeny viruses from the infected cells. We show that dual neutralization of the hemagglutinin and the neuraminidase by a bispecific antibody is advantageous over monoclonal antibody combination as it resulted an improved neutralization capacity and augmented the antibody effector functions. Notably, the bispecific antibody showed enhanced antiviral activity in influenza virus-infected mice, reduced mice mortality, and limited the virus mutation profile upon antibody administration. Thus, dual neutralization of the hemagglutinin and neuraminidase could be effective in controlling influenza virus infection.
    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
    双特异性抗体(bsAb)是在一个分子中组合两个不同抗原结合位点的工程化免疫球蛋白。BsAb可分为两种分子形式:IgG样抗体和非IgG样抗体。每种形式的结构元素对参与免疫系统都有影响。T细胞接合抗体(TCE)是设计用于使T细胞与靶细胞接合的bsAb。TCE不仅可以应用于癌症,还可以应用于传染病治疗以激活T细胞反应。在这次审查中,我们专注于目前的文献设计和使用bsAb作为一种创新策略,以增强适应性抗病毒免疫应答。我们总结了新型T细胞相关免疫疗法,重点是TCEs,为治疗慢性乙型肝炎而开发的乙型肝炎病毒(HBV)慢性感染在2022年造成110万人死亡,主要是由于肝硬化和肝细胞癌在超过250万人中发展慢性感染。缺乏治疗慢性乙型肝炎的治疗方法。将抗病毒治疗与激活T细胞反应的免疫治疗相结合被认为是治愈HBV和预防慢性感染后遗症的最有希望的治疗方法。吸引功能完整的T细胞,不是HBV特异性的,因此,尚未暴露于调节机制,激活肝脏靶位点的调节机制是一种非常有趣的治疗方法,可以通过TCEs实现。因此,TCEs将T细胞重定向到HBV阳性细胞代表了治疗慢性乙型肝炎和HBV相关肝细胞癌的有希望的策略。
    Bispecific antibodies (bsAbs) are engineered immunoglobulins that combine two different antigen-binding sites in one molecule. BsAbs can be divided into two molecular formats: IgG-like and non-IgG-like antibodies. Structural elements of each format have implications for engaging the immune system. T cell engager antibodies (TCEs) are bsAbs designed to engage T cells with target cells. TCEs can be applied not only in cancer but also in infectious disease therapy to activate T-cell responses. In this review, we focus on current literature on the design and use of bsAbs as an innovative strategy to enhance adaptive antiviral immune responses. We summarized the novel T cell-related immunotherapies with a focus on TCEs, that are developed for the treatment of chronic hepatitis B. Chronic infection with the hepatitis B virus (HBV) had a death toll of 1.1 million humans in 2022, mainly due to liver cirrhosis and hepatocellular carcinoma developing in the more than 250 million humans chronically infected. A curative treatment approach for chronic hepatitis B is lacking. Combining antiviral therapy with immune therapies activating T-cell responses is regarded as the most promising therapeutic approach to curing HBV and preventing the sequelae of chronic infection. Attracting functionally intact T cells that are not HBV-specific and, therefore, have not yet been exposed to regulatory mechanisms and activating those at the target site in the liver is a very interesting therapeutic approach that could be achieved by TCEs. Thus, TCEs redirecting T cells toward HBV-positive cells represent a promising strategy for treating chronic hepatitis B and HBV-associated hepatocellular carcinoma.
    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
    尽管在急性髓细胞性白血病(AML)治疗方面取得了相当大的进展,异基因造血干细胞移植(HSCT)后复发仍然很常见,并且与不良预后相关。已显示复发与HSCT之前CD34+白血病干细胞的不完全根除相关。以前,我们已经证明了一种新颖的CD34定向,双特异性T细胞接合器(BTE)可以有效地将T细胞效应子功能重定向到癌细胞,从而在体外和体内消除白血病细胞。然而,其对γδT细胞的影响尚不清楚。在这项研究中,我们使用体外扩增的γδT细胞作为效应子测试了CD34特异性BTE的功效。我们表明,BTE与γδT细胞和CD34白血病细胞系结合,并以剂量依赖性方式诱导靶细胞杀伤。此外,发现γδT细胞介导的杀伤优于αβT细胞介导的细胞毒性。此外,我们观察到,只有在BTE的存在下,γδT细胞在体外诱导原发性AML母细胞杀伤。重要的是,我们的结果表明,γδT细胞不能靶向健康的CD34中间内皮血脑屏障细胞系(hCMEC/D3),也不能裂解健康骨髓样本中的CD34+HSC.
    Despite the considerable progress in acute myeloid leukemia (AML) treatment, relapse after allogeneic hematopoietic stem cell transplantation (HSCT) is still frequent and associated with a poor prognosis. Relapse has been shown to be correlated with an incomplete eradication of CD34+ leukemic stem cells prior to HSCT. Previously, we have shown that a novel CD34-directed, bispecific T-cell engager (BTE) can efficiently redirect the T-cell effector function toward cancer cells, thus eliminating leukemic cells in vitro and in vivo. However, its impact on γδ T-cells is still unclear. In this study, we tested the efficacy of the CD34-specific BTE using in vitro expanded γδ T-cells as effectors. We showed that the BTEs bind to γδ T-cells and CD34+ leukemic cell lines and induce target cell killing in a dose-dependent manner. Additionally, γδ T-cell mediated killing was found to be superior to αβ T-cell mediated cytotoxicity. Furthermore, we observed that only in the presence of BTE the γδ T-cells induced primary AML blast killing in vitro. Importantly, our results show that γδ T-cells did not target the healthy CD34intermediate endothelial blood-brain barrier cell line (hCMEC/D3) nor lysed CD34+ HSCs from healthy bone marrow samples.
    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
    自2017年双特异性抗体blinatumomab批准用于治疗复发的急性淋巴细胞白血病以来,许多双特异性抗体构建体的开发在血液系统恶性肿瘤中已显著扩展.许多人最近在淋巴瘤的不同治疗阶段获得了食品药品监督管理局和欧洲药品管理局的批准,白血病,和多发性骨髓瘤.
    这篇综述的目的是提供双特异性抗体治疗的概述,包括导致效应T细胞靶向肿瘤相关抗原的机制,治疗适应症,功效,毒性,以及不同结构的挑战。通过访问PubMed和clinicaltrials.gov进行了文献检索。
    虽然NHL的治疗取得了实质性的成功,MM,和所有,仍然存在进展有限的血液系统恶性肿瘤,如AML.重要的是要继续研究新的设计,肿瘤抗原靶标,并进一步完善目前批准的双特异性抗体在治疗测序方面的适合之处。希望,随着近年来获得的知识和这些疗法的爆炸式增长,血癌患者在未来几年将继续受益于这些治疗。
    UNASSIGNED: Since the approval of the bispecific antibody blinatumomab in 2017 for the treatment of acute lymphoblastic leukemia in relapse, the development of numerous bispecific antibody constructs has dramatically expanded in hematologic malignancies. Many have recently received Food Drug Administration and European Medicines Agency approvals in various stages of treatment for lymphomas, leukemias, and multiple myeloma.
    UNASSIGNED: The purpose of this review is to provide an overview of bispecific antibody treatment including the mechanisms leading to effector T cells targeting tumor-associated antigens, the treatment indications, efficacies, toxicities, and challenges of the different constructs. A literature search was performed through access to PubMed and clinicaltrials.gov.
    UNASSIGNED: While there has been substantial success in the treatment of NHL, MM, and ALL, there are still hematologic malignancies such as AML where there has been limited progress. It is important to continue to investigate new designs, tumor antigen targets, and further refine where current approved bispecific antibodies fit in terms of sequencing of therapy. Hopefully, with the knowledge gained in recent years and the explosion of these therapies, patients with blood cancers will continue to benefit from these treatments for years to come.
    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
    目前针对人类表皮生长因子受体(HER)家族的疗法,包括单克隆抗体(mAb)和酪氨酸激酶抑制剂(TKIs),受到耐药性和全身毒性的限制。抗体-药物缀合物(ADC)是最快速扩展的抗癌治疗剂类别之一,目前已获得FDA批准。重要的是,ADC代表了一种有希望的治疗选择,具有通过向HER过表达的癌细胞特异性递送高效细胞毒素并发挥mAb和有效载荷介导的抗肿瘤功效来克服传统HER靶向治疗抗性的潜力。HER靶向ADC的临床效用通过HER2靶向ADC(包括曲妥珠单抗依坦素和曲妥珠单抗deruxtecan)的巨大成功来举例说明。尽管如此,改善现有HER2靶向ADC以及开发针对其他HER家族成员的ADC的策略,特别是EGFR和HER3,引起了极大的兴趣。迄今为止,没有HER4靶向ADC的报道。在这次审查中,我们广泛详述临床阶段EGFR-,HER2-,和HER3靶向单特异性ADC以及针对该受体家族的新型临床和临床前双特异性ADC(bsADC)。最后,我们讨论了针对HER的ADC发展的新趋势,包括新型ADC有效载荷和HER配体靶向ADC。
    Current therapies targeting the human epidermal growth factor receptor (HER) family, including monoclonal antibodies (mAbs) and tyrosine kinase inhibitors (TKIs), are limited by drug resistance and systemic toxicities. Antibody-drug conjugates (ADCs) are one of the most rapidly expanding classes of anti-cancer therapeutics with 13 presently approved by the FDA. Importantly, ADCs represent a promising therapeutic option with the potential to overcome traditional HER-targeted therapy resistance by delivering highly potent cytotoxins specifically to HER-overexpressing cancer cells and exerting both mAb- and payload-mediated antitumor efficacy. The clinical utility of HER-targeted ADCs is exemplified by the immense success of HER2-targeted ADCs including trastuzumab emtansine and trastuzumab deruxtecan. Still, strategies to improve upon existing HER2-targeted ADCs as well as the development of ADCs against other HER family members, particularly EGFR and HER3, are of great interest. To date, no HER4-targeting ADCs have been reported. In this review, we extensively detail clinical-stage EGFR-, HER2-, and HER3-targeting monospecific ADCs as well as novel clinical and pre-clinical bispecific ADCs (bsADCs) directed against this receptor family. We close by discussing nascent trends in the development of HER-targeting ADCs, including novel ADC payloads and HER ligand-targeted ADCs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号