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
    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: 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
    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
    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)

  • 文章类型: Journal Article
    靶向治疗如单克隆抗体的出现,过继性T细胞疗法,在过去的二十年中,抗体-药物偶联物(ADC)极大地改变了弥漫性大B细胞淋巴瘤(DLBCL)的治疗前景。利妥昔单抗是第一个批准的。嵌合抗原受体T细胞目前被批准为一线化学免疫疗法难治性DLBCL患者的二线治疗。Polatuzumab,以CD79b为目标的ADC,被批准为高风险患者的一线治疗与化学免疫疗法的组合。双特异性抗体(BsAb)是一类新的药物,也正在改变DLBCL患者的治疗模式。它们被设计为同时结合两个不同的靶标。迄今为止,两种BsAb(glofitamab和epcoritamab)被批准作为DLBCL三线治疗的单药治疗.联合化疗策略,免疫疗法,和ADC目前正在调查中,在一线或后续治疗线中取得了令人鼓舞的结果。在下面的审查中,我们专注于BsAbs的结构,作用机制,临床疗效,以及对BsAb的抗性机制。
    The advent of targeted therapies such as monoclonal antibodies, adoptive T-cell therapies, and antibody-drug conjugates (ADCs) dramatically changed the treatment landscape of diffuse large B-cell lymphoma (DLBCL) over the last two decades. Rituximab was the first one approved. Chimeric antigen receptor T-cells are currently approved as second-line treatment in patients with DLBCL refractory to first-line chemo-immunotherapy. Polatuzumab, a CD79b-targeting ADC, is approved as first-line treatment in high-risk patients in combination with chemo-immunotherapy. Bispecific antibodies (BsAbs) are a novel category of drugs that are also changing the treatment paradigm of patients with DLBCL. They are engineered to bind to two different targets at the same time. To date, two BsAbs (glofitamab and epcoritamab) are approved as monotherapy in third-line treatment in DLBCL. Combination strategies with chemotherapy, immunotherapy, and ADCs are currently under investigation with encouraging results in first-line or subsequent lines of treatment. In the following review, we focus on the structure of BsAbs, the mechanism of action, clinical efficacy, and the mechanisms of resistance to BsAbs.
    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
    众所周知的X连锁遗传疾病之一是血友病,可能是由于F8(因子VIII)基因突变导致的血友病A或由于F9(因子IX)基因突变导致的血友病B,导致凝血级联必需的蛋白质水平不足。在患有严重血友病的患者中,血浆中因子VIII或因子IX的活性相当低,估计不到1%。这是自发性或创伤后出血事件的原因,或者两者兼而有之,导致疾病并发症和死亡。目前血友病的治疗依赖于预防出血,其中包括昂贵的终身替代输注治疗血浆凝血因子,他们的重组版本,或者用重组单克隆抗体治疗。最近出现的基因治疗方法可能是潜在的游戏规则改变者,可以使用一次性载体在体内递送重塑血友病A或B的治疗结果,并旨在实现因子VIII或IX的长期内源性表达。这篇综述探讨了血友病治疗的传统方法和现代方法,主要专注于基因治疗,更新这方面的知识。对最近的技术进步和正在进行的基因治疗进行了严格的回顾和总结。我们认为基因治疗是最有前途的方法,因为它可以克服与更传统治疗相关的问题。例如需要持续和昂贵的输注,以及对用于治疗血友病的抗体药物的免疫反应。
    One of the well-known X-linked genetic disorders is hemophilia, which could be hemophilia A as a result of a mutation in the F8 (factor VIII) gene or hemophilia B as a result of a mutation in the F9 (factor IX) gene, leading to insufficient levels of the proteins essential for blood coagulation cascade. In patients with severe hemophilia, factor VIII or factor IX activities in the blood plasma are considerably low, estimated to be less than 1%. This is responsible for spontaneous or post-traumatic bleeding episodes, or both, leading to disease complications and death. Current treatment of hemophilia relies on the prevention of bleeding, which consists of expensive lifelong replacement infusion therapy of blood plasma clotting factors, their recombinant versions, or therapy with recombinant monoclonal antibodies. Recently emerged gene therapy approaches may be a potential game changer that could reshape the therapeutic outcomes of hemophilia A or B using a one-off vector in vivo delivery and aim to achieve long-term endogenous expression of factor VIII or IX. This review examines both traditional approaches to the treatment of hemophilia and modern methods, primarily focusing on gene therapy, to update knowledge in this area. Recent technological advances and gene therapeutics in the pipeline are critically reviewed and summarized. We consider gene therapy to be the most promising method as it may overcome the problems associated with more traditional treatments, such as the need for constant and expensive infusions and the presence of an immune response to the antibody drugs used to treat hemophilia.
    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
    尽管在过去的二十年中,对多发性骨髓瘤(MM)生物学的理解和新型治疗策略的发展取得了重大进展,MM仍然是一种不治之症。具有替代作用机制的新药,如核出口选择性抑制剂(SINE),泛素途径的调节剂[cereblonE3连接酶调节药物(CELMoDs)],和T细胞重定向(TCR)治疗,导致患者预后显着改善。然而,抵抗仍然出现,这对骨髓瘤患者的治疗构成了重大问题。这篇综述总结了目前用SINE治疗的数据,TCR治疗,和CELMoDs,并探讨其抗性机制。了解这些耐药机制对于制定克服治疗失败和改善治疗结果的策略至关重要。
    Despite significant advances in the understanding of multiple myeloma (MM) biology and the development of novel treatment strategies in the last two decades, MM is still an incurable disease. Novel drugs with alternative mechanisms of action, such as selective inhibitors of nuclear export (SINE), modulators of the ubiquitin pathway [cereblon E3 ligase modulatory drugs (CELMoDs)], and T cell redirecting (TCR) therapy, have led to significant improvement in patient outcomes. However, resistance still emerges, posing a major problem for the treatment of myeloma patients. This review summarizes current data on treatment with SINE, TCR therapy, and CELMoDs and explores their mechanism of resistance. Understanding these resistance mechanisms is critical for developing strategies to overcome treatment failure and improve therapeutic outcomes.
    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
    自然杀伤(NK)细胞是人类先天免疫的关键参与者。更有效地募集和激活NK细胞的细胞接合抗体形式已经成为一种有希望的免疫治疗方法,通过更有效的抗体依赖性细胞介导的细胞毒性(ADCC)靶向癌细胞。具有ADCC活性的单克隆抗体药物对于患有某些类型癌症的患者已经显示出临床益处和改善的结果。CD16a,FcγIII受体,是负责NK细胞ADCC活性的主要成分。筛选AvantGen的酵母展示的人抗体文库导致2个抗体克隆的分离,#1A2和#2-2A2,以高亲和力选择性识别原代NK细胞上CD16a的两种亚型(F和V),但与中性粒细胞表达的CD16b(NA1和NA2)的两种等位基因(#1A2)发生最小或不发生交叉反应(#2-2A2)。表位作图研究表明,它们与依赖于CD16a残基Y158的表位结合,因为Y158突变为相应的CD16b残基H158完全消除了与CD16a的结合。当格式化为靶向CD16a和肿瘤相关抗原(TAA,例如CD19),它们表现出与NK细胞的特异性结合,并在遇到肿瘤细胞时诱导有效的NK细胞活化,导致有效的肿瘤细胞杀伤。值得注意的是,这些双特异性抗体衔接剂在与靶细胞共培养过程中刺激NK细胞细胞因子释放,导致靶细胞的细胞毒性。这些抗CD16a抗体克隆是与任何感兴趣的TAA组合的有希望的候选者。提供了基于NK细胞衔接剂的新型癌症疗法的潜力,这些疗法受循环中高浓度人IgG的影响最小。
    Natural killer (NK) cells are key players in human innate immunity. Cell engager antibody formats that recruit and activate NK cells more effectively have emerged as a promising immunotherapy approach to target cancer cells through more effective antibody-dependent cell-mediated cytotoxicity (ADCC). Monoclonal antibody drugs with ADCC activity have shown clinical benefit and improved outcomes for patients with certain types of cancer. CD16a, a Fc gamma III receptor, is the major component that is responsible for the ADCC activity of NK cells. Screening AvantGen\'s yeast displayed human antibody libraries led to the isolation of 2 antibody clones, #1A2 and #2-2A2, that selectively recognize both isoforms (F and V) of CD16a on primary NK cells with high affinity, yet minimally (#1A2) or do not (#2-2A2) cross-react with both allelotypes of CD16b (NA1 and NA2) expressed by neutrophils. Epitope mapping studies revealed that they bind to an epitope dependent on residue Y158 of CD16a, since mutation of Y158 to the corresponding CD16b residue H158 completely abolishes binding to CD16a. When formatted as bispecific antibodies targeting CD16a and a tumor-associated antigen (TAA, e.g. CD19), they exhibit specific binding to NK cells and induce potent NK cell activation upon encountering tumor cells, resulting in effective tumor cell killing. Notably, these bispecific antibody engagers stimulate NK cell cytokine release during co-culture with target cells, resulting in target cell cytotoxicity. These anti-CD16a antibody clones are promising candidates for combination with any TAA of interest, offering the potential for novel NK cell engager-based cancer therapeutics that are minimally affected by the high concentrations of human IgG in the circulation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 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)

公众号