antibody-drug conjugates

抗体 - 药物偶联物
  • 文章类型: Journal Article
    NA.
    NA.
    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
    这篇综述旨在讨论SacituzumabGovitecan在乳腺癌治疗中的作用和疗效。
    乳腺癌是全球女性中最常见的癌症类型。这篇全面的综述探讨了SacituzumabGovitecan在治疗转移性三阴性乳腺癌(TNBC)方面的进展。专注于它的行动模式,有效的作用,临床试验,和相对于传统化疗的优势,该综述强调了该疗法通过单克隆抗体靶向癌细胞的精确性。SacituzumabGovitecan\'s能力提供化疗有效负载,特别是癌细胞与Trop-2受体使其区别于传统的化疗。减少附带损害和减少严重的副作用。SacituzumabGovitecan对改善无进展生存期的影响,肿瘤反应率,and,显著,讨论了患者的生活质量。这篇文章还揭示了正在进行的试验,FDA认可,以及该疗法改变乳腺癌治疗的潜力。
    总而言之,SacituzumabGovitecan显示出作为乳腺癌的创新治疗选择的潜力,特别是在转移性乳腺癌和三阴性乳腺癌中,但它需要更多的研究。
    UNASSIGNED: This review aims to discuss the role and efficacy of Sacituzumab Govitecan in the management of breast cancer.
    UNASSIGNED: Breast cancer is the most prevalent type of cancer among women worldwide. This comprehensive review delves into the advancements brought about by Sacituzumab Govitecan in the treatment of metastatic triple-negative breast cancer (TNBC). With a focus on its mode of action, efficacious role, clinical trials, and comparative advantages over conventional chemotherapy, the review highlights the therapy\'s precision in targeting cancer cells through monoclonal antibodies. Sacituzumab Govitecan\'s ability to deliver a chemotherapeutic payload specifically to cancer cells with the Trop-2 receptor sets it apart from traditional chemotherapy, minimizing collateral damage and reducing severe side effects. The impact of Sacituzumab Govitecan on improving progression-free survival, tumor response rates, and, significantly, the quality of life for patients is discussed. This article also sheds light on ongoing trials, FDA recognition, and the therapy\'s potential to transform breast cancer treatment.
    UNASSIGNED: In conclusion, Sacituzumab Govitecan shows potential as an innovative therapeutic option for breast cancer, particularly in metastatic breast cancer and triple-negative breast cancer, but it warrants additional 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
    HER2扩增是诱导结直肠癌抗EGFR治疗耐药的机制之一。近年来,来自多项随机临床试验的数据显示,抗HER2治疗改善了HER2阳性结直肠癌患者的预后.这些结果表明HER2是晚期结肠直肠癌的有希望的治疗靶标。尽管抗HER2治疗包括单克隆抗体,酪氨酸激酶抑制剂,和抗体-药物缀合物改善结果,不到30%的患者达到客观反应并最终产生耐药性。有必要探索抗HER2疗法耐药的主要和次要机制,这将为克服耐药性铺平道路。一些研究已经报道了对抗HER2疗法的抗性的潜在机制。在这次审查中,我们全面概述了临床研究的最新进展,治疗耐药机制,以及逆转HER2阳性结直肠癌患者耐药的策略。
    HER2 amplification is one of the mechanisms that induce drug resistance to anti-EGFR therapy in colorectal cancer. In recent years, data from several randomized clinical trials show that anti-HER2 therapies improved the prognosis of patients with HER2-positive colorectal cancer. These results indicate that HER2 is a promising therapeutic target in advanced colorectal cancer. Despite the anti-HER2 therapies including monoclonal antibodies, tyrosine kinase inhibitors, and antibody-drug conjugates improving the outcomes, less than 30 % of the patients achieve objective response and eventually have drug resistance. It is necessary to explore the primary and secondary mechanisms for the resistance to anti-HER2 therapies, which will pave the way to overcome the drug resistance. Several studies have reported the potential mechanisms for the resistance to anti-HER2 therapies. In this review, we present a comprehensive overview of the recent advances in clinical research, mechanisms of treatment resistance, and strategies for reversing resistance in HER2-positive colorectal cancer patients.
    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
    间质性肺病(ILD)是与使用抗体-药物缀合物(ADC)相关的严重不良事件(AE)。本研究旨在使用数据挖掘方法深入研究与与ADC相关的ILD相关的AE信号。
    根据FDA不良事件报告系统(FAERS)数据库,对2004年第一季度至2023年第四季度与ADC和ILD有关的AEreports进行了回顾性分析。使用报告比值比(ROR)方法和多项目伽玛泊松收缩器(MGPS)方法进行信号挖掘。数据管理,分析,和可视化是使用Python(版本3.8)进行的,R软件(4.2.1版),和MySQL(版本8.0.34)。
    共获得1389例与ILD相关的不良事件报告,其中ADC为主要可疑药物。这些报告中代表最多的年龄组是61-80岁年龄组和41-60岁年龄组。曲妥珠单抗deruxtecan的报告最多。数据挖掘结果表明,该研究中11个ADC检测到ILD相关的AE信号。曲妥珠单抗deruxtecan在ROR和MGPS方法中均显示最强的信号。与ILD相关的ADC的中位发病时间从8天到207天不等。
    与ADC相关的ILDAE信号异常强。在ADCs的临床使用中,应密切监测和评估ILD,同时充分考虑药物的疗效和风险以及患者的具体情况。
    UNASSIGNED: Interstitial lung diseases (ILD) is a seriousadverse event (AE) associated with the use of antibody-drug conjugates (ADCs).This study aims to delve deeply into the signals of AE associated with ILDlinked to ADCs using data mining methods.
    UNASSIGNED: A retrospective analysis was conducted on AEreports related to ADCs and ILD from the first quarter of 2004 to the fourthquarter of 2023 based on the FDA Adverse Event Reporting System (FAERS)database. Signal mining was performed using the reporting odds ratio (ROR)method and the multi-item gamma Poisson shrinker (MGPS) method. Data management, analysis, andvisualization were carried out using Python (Version 3.8), R software (Version4.2.1), and MySQL (Version 8.0.34).
    UNASSIGNED: A total of 1389 AE reports related to ILD with eleventypes of ADCs as the primary suspected drugs were obtained. The age groups mostrepresented in these reports were 61-80 age group and 41-60 age group. Trastuzumab deruxtecan had the most ofreports. The data mining results indicated that ILD-related AE signals weredetected for eleven ADCs in the study. Trastuzumab deruxtecan showed thestrongest signals in both for ROR and MGPS methods. The median onset time ofADCs associated with ILD vary from 8 days to 207 days.
    UNASSIGNED: The signals of ILD AE associated with ADCs arenotably strong. ILD should be closely monitored and assessed in the clinicaluse of ADCs taking full account of the efficacy and risks of the drugs as wellas the specific conditions of the patients.
    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
    在过去的十年中,随着下一代治疗性抗体的出现,癌症治疗领域取得了显着发展。化疗等常规治疗带来了巨大挑战,包括不良副作用。单克隆抗体为更有针对性和有效的干预措施铺平了道路。从嵌合抗体到人源化和完全人抗体的进化已经导致免疫原性的降低和增强的体内耐受性。下一代抗体的出现,包括双特异性抗体,纳米抗体,抗体-药物缀合物,糖工程抗体,和抗体片段,代表了癌症治疗的飞跃.这些创新提供了更高的效力,适应性,并降低耐药性。目标验证等挑战,免疫原性,和高生产成本存在。然而,抗体工程技术的技术进步为解决这些问题提供了乐观。未来预示着范式的转变,正在进行的研究将这些强大的抗体推向最前沿,彻底改变抗击癌症的斗争,创造新的预防和治疗方法。这篇综述概述了三种下一代基于抗体的分子,即双特异性抗体,抗体-药物缀合物,和纳米抗体在癌症治疗中显示出有希望的结果。它讨论了抗体从常规形式到下一代分子的进化,以及它们在癌症治疗中的应用,生产方法,和相关的挑战。该综述旨在为研究人员提供有关下一代基于抗体的癌症疗法的不断发展的前景及其彻底改变治疗策略的潜力的见解。
    The field of cancer treatment has evolved significantly over the last decade with the emergence of next-generation therapeutic antibodies. Conventional treatments like chemotherapy pose significant challenges, including adverse side effects. Monoclonal antibodies have paved the way for more targeted and effective interventions. The evolution from chimeric to humanized and fully human antibodies has led to a reduction in immunogenicity and enhanced tolerance in vivo. The advent of next-generation antibodies, including bispecific antibodies, nanobodies, antibody-drug conjugates, glyco-engineered antibodies, and antibody fragments, represents a leap forward in cancer therapy. These innovations offer increased potency, adaptability, and reduced drug resistance. Challenges such as target validation, immunogenicity, and high production costs exist. However, technological advancements in antibody engineering techniques provide optimism for addressing these issues. The future promises a paradigm shift, where ongoing research will propel these powerful antibodies to the forefront, revolutionizing the fight against cancer and creating new preventive and curative treatments. This review provides an overview of three next-generation antibody-based molecules, namely bispecific antibodies, antibody-drug conjugates, and nanobodies that have shown promising results in cancer treatment. It discusses the evolution of antibodies from conventional forms to next-generation molecules, along with their applications in cancer treatment, production methods, and associated challenges. The review aims to offer researchers insights into the evolving landscape of next-generation antibody-based cancer therapeutics and their potential to revolutionize treatment strategies.
    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: English Abstract
    Systemic treatment of urothelial carcinoma of the bladder requires complex approaches and is constantly evolving. Neoadjuvant cisplatin-based chemotherapy followed by radical cystectomy remains the current standard of care for muscle-invasive advanced bladder cancer. For patients ineligible for cisplatin, adjuvant treatment with nivolumab is recommended. Innovative perioperative combinations could transform the treatment landscape in the future. First-line treatment for metastatic urothelial carcinoma has long been dominated by platinum-based combinations, recently followed by the immune checkpoint inhibitor avelumab as maintenance therapy; however, recent results on the use of enfortumab vedotin and pembrolizumab in the first-line setting are expected to fundamentally change the treatment options. In subsequent lines of treatment, the not yet approved erdafitinib, as the first targeted therapy for advanced urothelial carcinoma, offers an important alternative and underscores the need for molecular testing.
    UNASSIGNED: Die Systemtherapie des Urothelkarzinoms der Harnblase erfordert komplexe Ansätze und befindet sich im stetigen Wandel. Die neoadjuvante cisplatinbasierte Chemotherapie, gefolgt von radikaler Zystektomie, bleibt der aktuelle Therapiestandard des muskelinvasiven, fortgeschrittenen Harnblasenkarzinoms. Für cisplatinungeeignete Patienten wird eine adjuvante Therapie mit Nivolumab empfohlen. Innovative perioperative Kombinationen könnten die Therapielandschaft in Zukunft verändern. Die Erstlinientherapie des metastasierten Urothelkarzinoms wurde lange durch platinhaltige Kombinationen geprägt, zuletzt gefolgt vom Immuncheckpoint-Inhibitor Avelumab als Erhaltungstherapie. Doch die jüngsten Ergebnisse zum Einsatz von Enfortumab-Vedotin und Pembrolizumab in der Erstlinie werden die Therapieoptionen grundlegend verändern. Auch in den Folgelinien bietet das noch nicht zugelassene Erdafitinib als erste zielgerichtete Therapie für das fortgeschrittene Urothelkarzinom eine wichtige Alternative und unterstreicht die Notwendigkeit molekularer Testungen.
    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
    抗体-药物缀合物(ADC)是一类新兴的肿瘤治疗,其将单克隆抗体的靶特异性与细胞毒性有效载荷相结合。这些药物与不同药物的独特皮肤毒性有关。目前,有11个ADC获得了实体和液体肿瘤的监管批准,80多个ADC目前正在临床开发中,皮肤科医生认识到并适当减轻与这些疗法相关的皮肤毒性是至关重要的。本临床综述将总结批准的ADC的新机制和适应症,讨论在临床试验和上市后研究中证明的皮肤病毒性,并在遇到这些反应时给予认可和管理指导,以帮助患者安全舒适地服用药物。
    Antibody-drug conjugates (ADCs) are a new and emerging category of oncologic treatments that combine the target specificity of a monoclonal antibody with a cytotoxic payload. These drugs are associated with unique cutaneous toxicities that vary across agents. Currently, there are eleven ADCs with regulatory approval for solid and liquid tumors and over 80 ADCs currently in clinical development, it is critical for dermatologists to recognize and appropriately mitigate the cutaneous toxicities associated with these therapies. This clinical review will summarize the novel mechanisms and indications of approved ADCs, discuss dermatologic toxicities demonstrated in clinical trials and post-marketing studies, and impart recognition and management guidance when encountering these reactions to help maintain patients safely and comfortably on their medications.
    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
    治疗性单克隆抗体的快速生长需要对评估抗原结合的可扩展方法的更大可接近性。同质TR-FRET是初步筛选的理想选择,但由于其高亲和力和复杂的溶液相动力学,尚未报道分析这些相互作用。在这里,我们报告了竞争测定法的开发,以对这些药物对常见抗原的相对亲和力进行排序。该测定与自动化兼容,不需要对分析物进行修饰,并测量亲和力低至个位数皮摩尔。我们进一步展示了为开发抗体-药物缀合物提供信息的应用。该测定可以帮助发现和制造治疗性抗体作为一种低成本,高通量替代现有技术。
    The rapid growth of therapeutic monoclonal antibodies demands greater accessibility to scalable methods of evaluating antigen binding. Homogenous TR-FRET is ideal for preliminary screening but has not been reported to assay these interactions due to their high-affinity and complex solution-phase kinetics. Here we report the development of a competition assay to rank-order the relative affinities of these drugs for a common antigen. The assay is compatible with automation, requires no modification of the analytes, and measures affinities as low as single-digit picomolar. We further demonstrate applications to inform the development of antibody-drug conjugates. The assay may aid discovery and manufacturing of therapeutic antibodies as a low-cost, high-throughput alternative to existing technologies.
    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
    肝-胰腺-胆道(HPB)恶性肿瘤构成了一组具有不良预后的高度侵袭性癌症。不适合根治性手术切除的患者接受全身化疗,然而,几乎没有生存的好处。抗体-药物缀合物(ADC)是将单克隆抗体的复杂选择性和特异性与附着的超毒性有效载荷的细胞破坏效力合并的三部分化合物。鉴于对可提高HPB癌症患者生存率的药物的需求尚未得到满足,评估ADC治疗HPB恶性肿瘤已成为广泛的临床和临床前研究的重点,显示出令人鼓舞的初步结果。在当前的审查中,我们全面概述了用于HPB恶性肿瘤的ADC方法越来越多的证据.从简要讨论ADC的功能原理开始,我们在这里总结了来自评估HPB癌症ADC的临床前和临床研究的所有可用数据。
    Hepato-Pancreato-Biliary (HPB) malignancies constitute a highly aggressive group of cancers that have a dismal prognosis. Patients not amenable to curative intent surgical resection are managed with systemic chemotherapy which, however, confers little survival benefit. Antibody-Drug Conjugates (ADCs) are tripartite compounds that merge the intricate selectivity and specificity of monoclonal antibodies with the cytodestructive potency of attached supertoxic payloads. In view of the unmet need for drugs that will enhance the survival rates of HPB cancer patients, the assessment of ADCs for treating HPB malignancies has become the focus of extensive clinical and preclinical investigation, showing encouraging preliminary results. In the current review, we offer a comprehensive overview of the growing body of evidence on ADC approaches tested for HPB malignancies. Starting from a concise discussion of the functional principles of ADCs, we summarize here all available data from preclinical and clinical studies evaluating ADCs in HPB cancers.
    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
    抗体-药物缀合物(ADC)代表了治疗几种肿瘤类型的有效药物。包括乳腺癌(BC),具有批准的分子,如曲妥珠单抗-emtansine,曲妥珠单抗-deruxtecan,和sacituzumab-govitecan.免疫检查点抑制剂(ICIs)在选定的BC亚型中也显示出活性,还有两个探员,派姆单抗和阿特珠单抗,目前已被批准用于治疗三阴性BC患者。BC中ADC和免疫疗法之间的潜在协同作用仍然是活跃研究的领域。临床前研究表明,ADC促进免疫监视,调节肿瘤微环境,诱导免疫原性细胞死亡,增强抗肿瘤免疫力。转化证据表明,单独或与免疫治疗联合使用的ADC的潜在预测性生物标志物,包括靶抗原的表达,致癌途径,肿瘤浸润淋巴细胞,和中性粒细胞与淋巴细胞的比率。鉴于这一背景,几项临床试验评估了BC患者的ADC-ICI组合,证明了有希望的结果,具有整体可管理的毒性,目前正在进行许多研究以确认这种治疗方法的有效性和可行性。在本次审查中,我们总结了关于整合ADC和免疫治疗治疗BC的现有证据,强调需要进一步的转化和临床研究来优化这种治疗策略并阐明预测性生物标志物,最终改善患者预后。
    Antibody-drug conjugates (ADCs) represent an effective class of agents for the treatment of several tumor types, including breast cancer (BC), featuring approved molecules such as trastuzumab-emtansine, trastuzumab-deruxtecan, and sacituzumab-govitecan. Immune-checkpoint inhibitors (ICIs) also showed activity in selected BC subtypes, and two agents, pembrolizumab and atezolizumab, are currently approved for the treatment of triple-negative BC patients. The potential synergy between ADCs and immunotherapy in BC remains an area of active investigation. Preclinical studies suggest that ADCs promote immune surveillance, modulating tumor microenvironment, inducing immunogenic cell death, and enhancing antitumor immunity. Translational evidence has shown potential predictive biomarkers for ADCs alone or in combination with immunotherapy, including expression of target antigen, oncogenic pathways, tumor-infiltrating lymphocytes, and neutrophil-to-lymphocyte ratio. Given this background, several clinical trials evaluated ADC-ICI combinations in BC patients, demonstrating promising outcomes with an overall manageable toxicity profile, and many studies are currently ongoing to confirm the efficacy and feasibility of this therapeutic approach. In the present review, we summarized the available evidence about the integration of ADCs and immunotherapy for the management of BC, emphasizing the need for further translational and clinical investigations to optimize this treatment strategy and elucidate predictive biomarkers, eventually improving patient outcomes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号