首页 > 最新文献

BioDrugs最新文献

英文 中文
Correction: Dystrophin- and Utrophin-Based Therapeutic Approaches for Treatment of Duchenne Muscular Dystrophy: A Comparative Review. 修正:以营养不良蛋白和营养不良蛋白为基础的治疗杜氏肌营养不良的方法:比较回顾。
IF 5.4 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-01 DOI: 10.1007/s40259-024-00693-y
Sylwia Szwec, Zuzanna Kapłucha, Jeffrey S Chamberlain, Patryk Konieczny
{"title":"Correction: Dystrophin- and Utrophin-Based Therapeutic Approaches for Treatment of Duchenne Muscular Dystrophy: A Comparative Review.","authors":"Sylwia Szwec, Zuzanna Kapłucha, Jeffrey S Chamberlain, Patryk Konieczny","doi":"10.1007/s40259-024-00693-y","DOIUrl":"10.1007/s40259-024-00693-y","url":null,"abstract":"","PeriodicalId":9022,"journal":{"name":"BioDrugs","volume":" ","pages":"167"},"PeriodicalIF":5.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11750941/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142944073","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Accelerating CAR-T Cell Therapies with Small-Molecule Inhibitors. 利用小分子抑制剂加速 CAR-T 细胞疗法。
IF 5.4 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-01 Epub Date: 2024-11-26 DOI: 10.1007/s40259-024-00688-9
Katrin Mestermann, Andoni Garitano-Trojaola, Michael Hudecek

Chimeric antigen receptor T-cell therapies have markedly improved the survival rates of patients with B-cell malignancies. However, their efficacy in other hematological cancers, such as acute myeloid leukemia, and in solid tumors has been limited. Key obstacles include the downregulation or loss of antigen expression on cancer cells, restricted accessibility to target cells, and the poor persistence of these "living drugs" because of the highly immunosuppressive tumor microenvironment. Additionally, manufacturing these immunotherapies presents significant challenges, and patients frequently experience side effects such as cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. This review emphasizes the potential of small-molecule inhibitors, many of which are already approved for clinical use, to facilitate chimeric antigen receptor T-cell manufacturing, enhance their anti-tumor efficacy, and mitigate their side effects. Although substantial work remains, the robust pre-clinical data and the growing clinical interest suggest significant promise for using cancer signaling pathway inhibitors to enhance and refine chimeric antigen receptor T-cell therapy for both hematological and solid tumors. Exploring these combination strategies could lead to more effective therapies, offering new hope for patients with resistant forms of cancer.

嵌合抗原受体 T 细胞疗法显著提高了 B 细胞恶性肿瘤患者的生存率。然而,嵌合抗原受体 T 细胞疗法对其他血液肿瘤(如急性髓性白血病)和实体瘤的疗效有限。主要障碍包括癌细胞抗原表达下调或丧失、靶细胞的可及性受到限制,以及这些 "活药 "因高度免疫抑制的肿瘤微环境而难以持久。此外,制造这些免疫疗法也是一大挑战,患者经常会出现细胞因子释放综合征和免疫效应细胞相关神经毒性综合征等副作用。本综述强调了小分子抑制剂在促进嵌合抗原受体 T 细胞制造、提高其抗肿瘤疗效和减轻其副作用方面的潜力,其中许多抑制剂已被批准用于临床。尽管仍有大量工作要做,但可靠的临床前数据和日益增长的临床兴趣表明,使用癌症信号通路抑制剂来增强和完善血液肿瘤和实体瘤的嵌合抗原受体 T 细胞疗法大有可为。探索这些组合策略可能会带来更有效的疗法,为耐药性癌症患者带来新的希望。
{"title":"Accelerating CAR-T Cell Therapies with Small-Molecule Inhibitors.","authors":"Katrin Mestermann, Andoni Garitano-Trojaola, Michael Hudecek","doi":"10.1007/s40259-024-00688-9","DOIUrl":"10.1007/s40259-024-00688-9","url":null,"abstract":"<p><p>Chimeric antigen receptor T-cell therapies have markedly improved the survival rates of patients with B-cell malignancies. However, their efficacy in other hematological cancers, such as acute myeloid leukemia, and in solid tumors has been limited. Key obstacles include the downregulation or loss of antigen expression on cancer cells, restricted accessibility to target cells, and the poor persistence of these \"living drugs\" because of the highly immunosuppressive tumor microenvironment. Additionally, manufacturing these immunotherapies presents significant challenges, and patients frequently experience side effects such as cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. This review emphasizes the potential of small-molecule inhibitors, many of which are already approved for clinical use, to facilitate chimeric antigen receptor T-cell manufacturing, enhance their anti-tumor efficacy, and mitigate their side effects. Although substantial work remains, the robust pre-clinical data and the growing clinical interest suggest significant promise for using cancer signaling pathway inhibitors to enhance and refine chimeric antigen receptor T-cell therapy for both hematological and solid tumors. Exploring these combination strategies could lead to more effective therapies, offering new hope for patients with resistant forms of cancer.</p>","PeriodicalId":9022,"journal":{"name":"BioDrugs","volume":" ","pages":"33-51"},"PeriodicalIF":5.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11750903/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142715052","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Interleukin-17 Inhibitors in the Treatment of Hidradenitis Suppurativa. 白细胞介素-17抑制剂在治疗化脓性扁桃体炎中的应用
IF 5.4 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-01 Epub Date: 2024-11-28 DOI: 10.1007/s40259-024-00687-w
Gonçalo Pinto Salgueiro, Orhan Yilmaz, Miguel Nogueira, Tiago Torres

Hidradenitis suppurativa (HS) is a chronic, debilitating, inflammatory dermatosis that significantly impacts patients' quality of life, primarily manifesting as inflammatory nodules, abscesses, and tunnels. The pathogenesis of HS is not fully understood and appears to be multifactorial, involving genetic, immunological, and endocrinological factors, as well as dysbiosis of skin microbiota. Increasing evidence highlights the role of the interleukin (IL)-17 pathway in the inflammatory process and pathogenesis of HS. Consequently, IL-17 inhibitors have emerged as a promising alternative to current therapies. Recently, secukinumab received approval from both the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA), while bimekizumab received approval from the EMA, for the treatment of moderate-to-severe HS in adults, with ongoing clinical trials aiming to further clarify the efficacy and safety of other drugs within this class. IL-17 inhibitors have shown effectiveness in treating moderate-to-severe HS, with safety profiles of drugs such as secukinumab and bimekizumab being comparable to their use in other dermatological conditions. On the other hand, innovative drugs such as sonelokimab and izokibep show promising results and are currently in phase III clinical trials. This review provides a comprehensive overview of current knowledge and scientific advances in HS, focusing on the IL-17 pathway's role and its inhibition as a treatment strategy, alongside examining the most recent and significant clinical studies on various IL-17 inhibitors in the treatment of HS.

化脓性扁平湿疹(HS)是一种慢性、使人衰弱的炎症性皮肤病,严重影响患者的生活质量,主要表现为炎性结节、脓肿和隧道。HS 的发病机制尚未完全明了,似乎是多因素的,涉及遗传、免疫和内分泌因素,以及皮肤微生物菌群失调。越来越多的证据表明,白细胞介素(IL)-17通路在HS的炎症过程和发病机制中起着重要作用。因此,IL-17抑制剂已成为目前疗法的一种有前途的替代疗法。最近,secukinumab获得了美国食品药品管理局(FDA)和欧洲药品管理局(EMA)的批准,而bimekizumab也获得了EMA的批准,用于治疗成人中度至重度HS,目前正在进行的临床试验旨在进一步明确该类药物的疗效和安全性。IL-17抑制剂已显示出治疗中重度HS的疗效,secukinumab和bimekizumab等药物的安全性与它们用于其他皮肤病的疗效相当。另一方面,sonelokimab 和 izokibep 等创新药物也显示出良好的疗效,目前正在进行 III 期临床试验。这篇综述全面概述了目前有关HS的知识和科学进展,重点关注IL-17通路的作用及其作为一种治疗策略的抑制作用,同时还探讨了有关各种IL-17抑制剂治疗HS的最新重要临床研究。
{"title":"Interleukin-17 Inhibitors in the Treatment of Hidradenitis Suppurativa.","authors":"Gonçalo Pinto Salgueiro, Orhan Yilmaz, Miguel Nogueira, Tiago Torres","doi":"10.1007/s40259-024-00687-w","DOIUrl":"10.1007/s40259-024-00687-w","url":null,"abstract":"<p><p>Hidradenitis suppurativa (HS) is a chronic, debilitating, inflammatory dermatosis that significantly impacts patients' quality of life, primarily manifesting as inflammatory nodules, abscesses, and tunnels. The pathogenesis of HS is not fully understood and appears to be multifactorial, involving genetic, immunological, and endocrinological factors, as well as dysbiosis of skin microbiota. Increasing evidence highlights the role of the interleukin (IL)-17 pathway in the inflammatory process and pathogenesis of HS. Consequently, IL-17 inhibitors have emerged as a promising alternative to current therapies. Recently, secukinumab received approval from both the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA), while bimekizumab received approval from the EMA, for the treatment of moderate-to-severe HS in adults, with ongoing clinical trials aiming to further clarify the efficacy and safety of other drugs within this class. IL-17 inhibitors have shown effectiveness in treating moderate-to-severe HS, with safety profiles of drugs such as secukinumab and bimekizumab being comparable to their use in other dermatological conditions. On the other hand, innovative drugs such as sonelokimab and izokibep show promising results and are currently in phase III clinical trials. This review provides a comprehensive overview of current knowledge and scientific advances in HS, focusing on the IL-17 pathway's role and its inhibition as a treatment strategy, alongside examining the most recent and significant clinical studies on various IL-17 inhibitors in the treatment of HS.</p>","PeriodicalId":9022,"journal":{"name":"BioDrugs","volume":" ","pages":"53-74"},"PeriodicalIF":5.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11750882/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142738331","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Evolving Applications of Bispecific Antibodies: Reaping the Harvest of Early Sowing and Planting New Seeds. 双特异性抗体应用的发展:收获早播的收获,播下新的种子。
IF 5.4 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-01 Epub Date: 2024-12-13 DOI: 10.1007/s40259-024-00691-0
Diego A Ellerman

After decades of gradual progress from conceptualization to early clinical trials (1960-2000), the therapeutic potential of bispecific antibodies (bisp Abs) is now being fully realized. Insights gained from both successful and unsuccessful trials are helping to identify which mechanisms of action, antibody formats, and targets prove most effective, and which may benefit from further refinement. While T-cell engagers remain the most commonly used class of bisp Abs, current efforts aim to increase their effectiveness by co-engaging costimulatory molecules, reducing escape mechanisms, and countering immunosuppression. Strategies to minimize cytokine release syndrome (CRS) are also actively under development. In addition, novel antibody formats that are selectively activated within tumors are an exciting area of research, as is the precise targeting of specific T-cell subsets. Beyond T cells, the recruitment of macrophages and dendritic cells is attracting increasing interest, with researchers exploring various macrophage receptors to promote phagocytosis or to carry out specialized functions, such as the immunologically silent clearance of amyloid-beta plaques in the brain. While bisp Abs targeting B cells are relatively limited, they are primarily aimed at inhibiting B-cell activity in autoimmune diseases. Another evolving application involves the forced interaction between proteins. Beyond the successful development of Hemlibra for hemophilia, bispecific antibodies that mimic cytokine activity are being explored. Additionally, the recruitment of cell surface ubiquitin ligases and other enzymes represents a novel and promising therapeutic strategy. In regard to antibody formats, some applications such as the combination of T-cell engagers with costimulatory molecules are driving the development of trispecific antibodies, at least in preclinical settings. However, the increasing structural complexity of multispecific antibodies often leads to more challenging development paths, and the number of multispecific antibodies in clinical trials remains low. The clinical success of certain applications and well-established production methods position this therapeutic class to expand its benefits into other therapeutic areas.

从概念到早期临床试验(1960-2000),经过几十年的逐步发展,双特异性抗体(bisp Abs)的治疗潜力现在正在充分实现。从成功和不成功的试验中获得的见解有助于确定哪些作用机制、抗体形式和靶点被证明是最有效的,哪些可能从进一步改进中受益。虽然t细胞接合物仍然是最常用的一类双抗,但目前的努力旨在通过共接合共刺激分子、减少逃逸机制和对抗免疫抑制来提高其有效性。减少细胞因子释放综合征(CRS)的策略也在积极开发中。此外,在肿瘤内选择性激活的新型抗体格式是一个令人兴奋的研究领域,就像精确靶向特定t细胞亚群一样。除了T细胞外,巨噬细胞和树突状细胞的募集也引起了越来越多的兴趣,研究人员探索了各种巨噬细胞受体来促进吞噬或执行特殊功能,例如免疫沉默清除大脑中的淀粉样蛋白斑块。虽然针对B细胞的bisp抗体相对有限,但它们主要用于抑制自身免疫性疾病中的B细胞活性。另一个不断发展的应用涉及蛋白质之间的强制相互作用。除了Hemlibra治疗血友病的成功开发,模拟细胞因子活性的双特异性抗体也在探索中。此外,细胞表面泛素连接酶和其他酶的募集代表了一种新的和有前途的治疗策略。在抗体形式方面,一些应用,如t细胞接合体与共刺激分子的结合,正在推动三特异性抗体的发展,至少在临床前环境中是这样。然而,随着多特异性抗体结构复杂性的增加,其发展路径往往更具挑战性,而且临床试验中的多特异性抗体数量仍然很低。临床成功的某些应用和完善的生产方法定位这类治疗扩大其效益到其他治疗领域。
{"title":"The Evolving Applications of Bispecific Antibodies: Reaping the Harvest of Early Sowing and Planting New Seeds.","authors":"Diego A Ellerman","doi":"10.1007/s40259-024-00691-0","DOIUrl":"10.1007/s40259-024-00691-0","url":null,"abstract":"<p><p>After decades of gradual progress from conceptualization to early clinical trials (1960-2000), the therapeutic potential of bispecific antibodies (bisp Abs) is now being fully realized. Insights gained from both successful and unsuccessful trials are helping to identify which mechanisms of action, antibody formats, and targets prove most effective, and which may benefit from further refinement. While T-cell engagers remain the most commonly used class of bisp Abs, current efforts aim to increase their effectiveness by co-engaging costimulatory molecules, reducing escape mechanisms, and countering immunosuppression. Strategies to minimize cytokine release syndrome (CRS) are also actively under development. In addition, novel antibody formats that are selectively activated within tumors are an exciting area of research, as is the precise targeting of specific T-cell subsets. Beyond T cells, the recruitment of macrophages and dendritic cells is attracting increasing interest, with researchers exploring various macrophage receptors to promote phagocytosis or to carry out specialized functions, such as the immunologically silent clearance of amyloid-beta plaques in the brain. While bisp Abs targeting B cells are relatively limited, they are primarily aimed at inhibiting B-cell activity in autoimmune diseases. Another evolving application involves the forced interaction between proteins. Beyond the successful development of Hemlibra for hemophilia, bispecific antibodies that mimic cytokine activity are being explored. Additionally, the recruitment of cell surface ubiquitin ligases and other enzymes represents a novel and promising therapeutic strategy. In regard to antibody formats, some applications such as the combination of T-cell engagers with costimulatory molecules are driving the development of trispecific antibodies, at least in preclinical settings. However, the increasing structural complexity of multispecific antibodies often leads to more challenging development paths, and the number of multispecific antibodies in clinical trials remains low. The clinical success of certain applications and well-established production methods position this therapeutic class to expand its benefits into other therapeutic areas.</p>","PeriodicalId":9022,"journal":{"name":"BioDrugs","volume":" ","pages":"75-102"},"PeriodicalIF":5.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142821785","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CAR T-Cell Therapy for Rheumatic Diseases: What Does the Future Hold? CAR - t细胞治疗风湿病:未来如何?
IF 5.4 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-01 Epub Date: 2024-12-29 DOI: 10.1007/s40259-024-00692-z
Jérôme Avouac, Marc Scherlinger

Chimeric antigen receptor (CAR) T-cell therapy, initially successful in treating hematological malignancies, is emerging as a potential treatment for autoimmune diseases, including rheumatic conditions. CAR T cells, engineered to target and eliminate autoreactive B cells, offer a novel approach to managing diseases like systemic lupus erythematosus (SLE), systemic sclerosis (SSc), and inflammatory myopathies, where B cells play a pivotal role in disease pathology. Early case reports have demonstrated promising results, with patients achieving significant disease remission, normalization of serological markers, and the ability to discontinue traditional immunosuppressive therapies, which supported the initiation of several clinical trials. However, the application of CAR T-cell therapy in chronic inflammatory rheumatic disorders poses unique challenges, including patient heterogeneity, the risk of adverse effects such as cytokine release syndrome, and the high costs associated with the therapy. Despite these challenges, the potential for CAR T cells to provide long-term remission or even a cure in refractory autoimmune diseases is significant. Ongoing research aims to optimize CAR T-cell constructs and improve safety profiles, paving the way for broader application in rheumatic diseases. If these challenges can be addressed, CAR T-cell therapy could revolutionize the treatment landscape for chronic inflammatory rheumatic disorders, offering new hope for patients with severe, treatment-resistant conditions.

嵌合抗原受体(CAR) t细胞疗法最初在治疗血液系统恶性肿瘤方面取得成功,目前正在成为包括风湿病在内的自身免疫性疾病的潜在治疗方法。CAR - T细胞被设计用于靶向和消除自身反应性B细胞,为治疗系统性红斑狼疮(SLE)、系统性硬化症(SSc)和炎性肌病等疾病提供了一种新方法,其中B细胞在疾病病理中起着关键作用。早期病例报告显示了令人鼓舞的结果,患者获得了显着的疾病缓解,血清学标志物正常化,并且能够停止传统的免疫抑制治疗,这支持了几项临床试验的启动。然而,CAR - t细胞疗法在慢性炎症性风湿病中的应用面临着独特的挑战,包括患者异质性,细胞因子释放综合征等不良反应的风险,以及与治疗相关的高成本。尽管存在这些挑战,CAR - T细胞在难治性自身免疫性疾病中提供长期缓解甚至治愈的潜力是显著的。正在进行的研究旨在优化CAR - t细胞结构和提高安全性,为风湿病的更广泛应用铺平道路。如果这些挑战能够得到解决,CAR - t细胞疗法可能会彻底改变慢性炎症性风湿病的治疗前景,为患有严重、治疗难治性疾病的患者带来新的希望。
{"title":"CAR T-Cell Therapy for Rheumatic Diseases: What Does the Future Hold?","authors":"Jérôme Avouac, Marc Scherlinger","doi":"10.1007/s40259-024-00692-z","DOIUrl":"10.1007/s40259-024-00692-z","url":null,"abstract":"<p><p>Chimeric antigen receptor (CAR) T-cell therapy, initially successful in treating hematological malignancies, is emerging as a potential treatment for autoimmune diseases, including rheumatic conditions. CAR T cells, engineered to target and eliminate autoreactive B cells, offer a novel approach to managing diseases like systemic lupus erythematosus (SLE), systemic sclerosis (SSc), and inflammatory myopathies, where B cells play a pivotal role in disease pathology. Early case reports have demonstrated promising results, with patients achieving significant disease remission, normalization of serological markers, and the ability to discontinue traditional immunosuppressive therapies, which supported the initiation of several clinical trials. However, the application of CAR T-cell therapy in chronic inflammatory rheumatic disorders poses unique challenges, including patient heterogeneity, the risk of adverse effects such as cytokine release syndrome, and the high costs associated with the therapy. Despite these challenges, the potential for CAR T cells to provide long-term remission or even a cure in refractory autoimmune diseases is significant. Ongoing research aims to optimize CAR T-cell constructs and improve safety profiles, paving the way for broader application in rheumatic diseases. If these challenges can be addressed, CAR T-cell therapy could revolutionize the treatment landscape for chronic inflammatory rheumatic disorders, offering new hope for patients with severe, treatment-resistant conditions.</p>","PeriodicalId":9022,"journal":{"name":"BioDrugs","volume":" ","pages":"5-19"},"PeriodicalIF":5.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142908701","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Sequelae of B-Cell Depleting Therapy: An Immunologist's Perspective. B 细胞消耗疗法的后遗症:免疫学家的视角。
IF 5.4 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-01 Epub Date: 2024-12-16 DOI: 10.1007/s40259-024-00696-9
Mark Kacar, Adam Al-Hakim, Sinisa Savic

B-cell depleting therapy (BCDT) has revolutionised the treatment of B-cell malignancies and autoimmune diseases by targeting specific B-cell surface antigens, receptors, ligands, and signalling pathways. This narrative review explores the mechanisms, applications, and complications of BCDT, focusing on the therapeutic advancements since the introduction of rituximab in 1997. Various monoclonal antibodies and kinase inhibitors are examined for their roles in depleting B cells through antibody-dependent and independent mechanisms. The off-target effects, such as hypogammaglobulinemia, infections, and cytokine release syndrome, are discussed, emphasising the need for immunologists to identify and help manage these complications. The increasing prevalence of BCDT has necessitated the involvement of clinical immunologists in addressing treatment-associated immunological abnormalities, including persistent hypogammaglobulinemia and neutropenia. We highlight the importance of considering underlying inborn errors of immunity (IEI) in patients presenting with these complications. Furthermore, we discuss the impact of BCDT on other immune cell populations and the challenges in predicting and managing long-term immunological sequelae. The potential for novel BCDT agents targeting the BAFF/APRIL-TACI/BCMA axis and B-cell receptor signalling pathways to treat autoimmune disorders is also explored, underscoring the rapidly evolving landscape of B-cell targeted therapies.

针对特定 B 细胞表面抗原、受体、配体和信号通路的 B 细胞耗竭疗法(BCDT)彻底改变了 B 细胞恶性肿瘤和自身免疫性疾病的治疗方法。这篇叙述性综述探讨了 BCDT 的机制、应用和并发症,重点关注自 1997 年利妥昔单抗问世以来的治疗进展。文章研究了各种单克隆抗体和激酶抑制剂在通过抗体依赖和独立机制消耗 B 细胞方面的作用。还讨论了低丙种球蛋白血症、感染和细胞因子释放综合征等脱靶效应,强调免疫学家需要识别并帮助控制这些并发症。随着 BCDT 患病率的增加,临床免疫学家有必要参与处理与治疗相关的免疫异常,包括持续性低丙种球蛋白血症和中性粒细胞减少症。我们强调了在出现这些并发症的患者中考虑潜在先天性免疫错误(IEI)的重要性。此外,我们还讨论了 BCDT 对其他免疫细胞群的影响,以及预测和管理长期免疫后遗症所面临的挑战。我们还探讨了以 BAFF/APRIL-TACI/BCMA 轴和 B 细胞受体信号通路为靶点的新型 BCDT 药物治疗自身免疫性疾病的潜力,强调了 B 细胞靶向疗法快速发展的前景。
{"title":"Sequelae of B-Cell Depleting Therapy: An Immunologist's Perspective.","authors":"Mark Kacar, Adam Al-Hakim, Sinisa Savic","doi":"10.1007/s40259-024-00696-9","DOIUrl":"10.1007/s40259-024-00696-9","url":null,"abstract":"<p><p>B-cell depleting therapy (BCDT) has revolutionised the treatment of B-cell malignancies and autoimmune diseases by targeting specific B-cell surface antigens, receptors, ligands, and signalling pathways. This narrative review explores the mechanisms, applications, and complications of BCDT, focusing on the therapeutic advancements since the introduction of rituximab in 1997. Various monoclonal antibodies and kinase inhibitors are examined for their roles in depleting B cells through antibody-dependent and independent mechanisms. The off-target effects, such as hypogammaglobulinemia, infections, and cytokine release syndrome, are discussed, emphasising the need for immunologists to identify and help manage these complications. The increasing prevalence of BCDT has necessitated the involvement of clinical immunologists in addressing treatment-associated immunological abnormalities, including persistent hypogammaglobulinemia and neutropenia. We highlight the importance of considering underlying inborn errors of immunity (IEI) in patients presenting with these complications. Furthermore, we discuss the impact of BCDT on other immune cell populations and the challenges in predicting and managing long-term immunological sequelae. The potential for novel BCDT agents targeting the BAFF/APRIL-TACI/BCMA axis and B-cell receptor signalling pathways to treat autoimmune disorders is also explored, underscoring the rapidly evolving landscape of B-cell targeted therapies.</p>","PeriodicalId":9022,"journal":{"name":"BioDrugs","volume":" ","pages":"103-130"},"PeriodicalIF":5.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142827355","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Relevance of Neutralizing Antibodies for the Pharmacokinetics of Pegunigalsidase Alfa in Patients with Fabry Disease. 中和抗体与法布里病患者Pegunigalsidase Alfa药代动力学的相关性
IF 5.4 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-01 Epub Date: 2024-11-30 DOI: 10.1007/s40259-024-00690-1
Malte Lenders, Elise Raphaela Menke, Michael Rudnicki, Markus Cybulla, Eva Brand

Background: Pegunigalsidase alfa is a newly approved drug for the treatment of Fabry disease, designed to increase the plasma half-life and reduce immunogenicity of infused α-galactosidase A (AGAL). We provide the first comprehensive pharmacokinetic and immunogenic data apart from industry-initiated studies.

Methods: Pharmacokinetics of pegunigalsidase alfa, amino acid, and polyethylene glycol (PEG)-specific antibodies and immune complexes were measured in treated patients (11 switched, two naïve). Measurements were performed in serum samples drawn directly before and after infusions over three to ten consecutive infusions. Only three patients started directly with 1.0 mg/kg body weight.

Results: No infusion-associated reactions were reported under pegunigalsidase alfa during the observation. Patients without pre-existing neutralizing anti-AGAL antibodies showed high enzymatic AGAL peak activities and sustained AGAL serum concentrations until the next infusion, which was not observed in those with neutralizing anti-AGAL antibodies. Nine (69.2%) patients presented with pre-existing anti-PEG antibodies (IgG or IgM), which seemed to have no impact on pharmacokinetics during the observation. No new anti-PEG or anti-AGAL antibody formation was observed after treatment initiation. Three (75.0%) patients with pre-existing neutralizing anti-AGAL antibodies showed a titer increase and one (25.0%) patient a decrease. In patients with anti-AGAL antibodies (n = 4) immune-complex formation was detected.

Conclusion: The pharmacokinetics of pegunigalsidase alfa show different profiles depending on the presence of pre-existing neutralizing antibodies, with reduced plasma half-life and peak enzyme activity after infusion in patients with antibodies. The clinical significance of a reduced pegunigalsidase alfa half-life and the formation of immune complexes in antibody-positive patients needs to be analyzed in future studies.

背景:Pegunigalsidase alfa是一种新批准的治疗Fabry病的药物,旨在增加输注α-半乳糖苷酶a (AGAL)的血浆半衰期,降低免疫原性。我们提供第一个全面的药代动力学和免疫原性数据,除了行业发起的研究。方法:在治疗的患者中测定PEG特异性抗体和免疫复合物的药代动力学(11例切换,2例naïve)。在连续三到十次输注前后直接抽取血清样本进行测量。只有3例患者直接以1.0 mg/kg体重开始治疗。结果:观察过程中,未见pegunigalsidase - α输注相关反应。没有预先存在中和性抗AGAL抗体的患者表现出较高的酶促AGAL峰活性和持续的AGAL血清浓度,直到下一次输注,这在中和性抗AGAL抗体的患者中没有观察到。9例(69.2%)患者存在预先存在的抗peg抗体(IgG或IgM),在观察期间似乎对药代动力学没有影响。治疗开始后未观察到新的抗peg或抗agal抗体的形成。已有中和抗agal抗体的3例(75.0%)患者滴度升高,1例(25.0%)患者滴度降低。抗agal抗体患者(n = 4)检测到免疫复合物形成。结论:pegunigalsidase alfa的药代动力学表现出不同的特征,取决于预先存在的中和抗体的存在,抗体患者输注后血浆半衰期缩短,酶活性达到峰值。pegunigalsidase α半衰期缩短和抗体阳性患者免疫复合物形成的临床意义需要在未来的研究中分析。
{"title":"Relevance of Neutralizing Antibodies for the Pharmacokinetics of Pegunigalsidase Alfa in Patients with Fabry Disease.","authors":"Malte Lenders, Elise Raphaela Menke, Michael Rudnicki, Markus Cybulla, Eva Brand","doi":"10.1007/s40259-024-00690-1","DOIUrl":"10.1007/s40259-024-00690-1","url":null,"abstract":"<p><strong>Background: </strong>Pegunigalsidase alfa is a newly approved drug for the treatment of Fabry disease, designed to increase the plasma half-life and reduce immunogenicity of infused α-galactosidase A (AGAL). We provide the first comprehensive pharmacokinetic and immunogenic data apart from industry-initiated studies.</p><p><strong>Methods: </strong>Pharmacokinetics of pegunigalsidase alfa, amino acid, and polyethylene glycol (PEG)-specific antibodies and immune complexes were measured in treated patients (11 switched, two naïve). Measurements were performed in serum samples drawn directly before and after infusions over three to ten consecutive infusions. Only three patients started directly with 1.0 mg/kg body weight.</p><p><strong>Results: </strong>No infusion-associated reactions were reported under pegunigalsidase alfa during the observation. Patients without pre-existing neutralizing anti-AGAL antibodies showed high enzymatic AGAL peak activities and sustained AGAL serum concentrations until the next infusion, which was not observed in those with neutralizing anti-AGAL antibodies. Nine (69.2%) patients presented with pre-existing anti-PEG antibodies (IgG or IgM), which seemed to have no impact on pharmacokinetics during the observation. No new anti-PEG or anti-AGAL antibody formation was observed after treatment initiation. Three (75.0%) patients with pre-existing neutralizing anti-AGAL antibodies showed a titer increase and one (25.0%) patient a decrease. In patients with anti-AGAL antibodies (n = 4) immune-complex formation was detected.</p><p><strong>Conclusion: </strong>The pharmacokinetics of pegunigalsidase alfa show different profiles depending on the presence of pre-existing neutralizing antibodies, with reduced plasma half-life and peak enzyme activity after infusion in patients with antibodies. The clinical significance of a reduced pegunigalsidase alfa half-life and the formation of immune complexes in antibody-positive patients needs to be analyzed in future studies.</p>","PeriodicalId":9022,"journal":{"name":"BioDrugs","volume":" ","pages":"153-165"},"PeriodicalIF":5.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11750932/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142765602","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction: Monoclonal Antibody Generation Using Single B Cell Screening for Treating Infectious Diseases. 更正:利用单个 B 细胞筛选生成单克隆抗体治疗传染病。
IF 5.4 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-01 DOI: 10.1007/s40259-024-00695-w
John S Schardt, Neelan S Sivaneri, Peter M Tessier
{"title":"Correction: Monoclonal Antibody Generation Using Single B Cell Screening for Treating Infectious Diseases.","authors":"John S Schardt, Neelan S Sivaneri, Peter M Tessier","doi":"10.1007/s40259-024-00695-w","DOIUrl":"10.1007/s40259-024-00695-w","url":null,"abstract":"","PeriodicalId":9022,"journal":{"name":"BioDrugs","volume":" ","pages":"169"},"PeriodicalIF":5.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11750878/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142827346","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Investigational RNA Interference Agents for Hepatitis B. 乙型肝炎RNA干扰剂的研究。
IF 5.4 2区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-01 Epub Date: 2024-12-07 DOI: 10.1007/s40259-024-00694-x
Rex Wan-Hin Hui, Lung-Yi Mak, Wai-Kay Seto, Man-Fung Yuen

Functional cure of chronic hepatitis B (CHB)-defined as sustained seroclearance of hepatitis B surface antigen (HBsAg) with unquantifiable hepatitis B virus (HBV) DNA at 24 weeks off treatment, is an optimal treatment endpoint. Nonetheless, it cannot be consistently attained by current treatment modalities. RNA interference (RNAi) is a novel treatment strategy using small-interfering RNA (siRNA) or antisense oligonucleotide (ASO) to target HBV post-transcriptional RNA, in turn suppressing viral protein production and replication. Hence, RNAi has indirect effects in promoting host immune reconstitution against HBV. Multiple RNAi therapeutics have entered phase II/III clinical trials, demonstrating potent, dose-dependent, and sustainable effects in suppressing HBsAg. Incidences of HBsAg seroclearance, particularly with the use of ASO, have also been documented. The combination of RNAi with other antivirals/immunomodulators (e.g. pegylated interferon), have shown promising results in potentiating RNAi effects and enhancing treatment durability. Further research will be required to establish predictors of response, optimal treatment protocols, and long-term outcomes in patients on RNAi. RNAi therapeutics have shown promising results and will likely be the keystone of future HBV treatment.

慢性乙型肝炎(CHB)的功能性治愈——定义为在治疗结束后24周,乙型肝炎表面抗原(HBsAg)和无法量化的乙型肝炎病毒(HBV) DNA的持续血清清除——是一个最佳的治疗终点。然而,目前的治疗方式并不能始终如一地达到这一目标。RNA干扰(RNAi)是一种新的治疗策略,利用小干扰RNA (siRNA)或反义寡核苷酸(ASO)靶向HBV转录后RNA,从而抑制病毒蛋白的产生和复制。因此,RNAi在促进宿主对HBV的免疫重建中具有间接作用。多种RNAi疗法已进入II/III期临床试验,显示出有效的、剂量依赖性的和可持续的抑制HBsAg的效果。HBsAg血清清除率的发生率,特别是使用ASO,也有文献记载。RNAi与其他抗病毒药物/免疫调节剂(如聚乙二醇化干扰素)联合使用,在增强RNAi效应和提高治疗持久性方面显示出有希望的结果。需要进一步的研究来确定RNAi患者的反应预测因素、最佳治疗方案和长期结果。RNAi疗法已经显示出有希望的结果,并可能成为未来HBV治疗的基石。
{"title":"Investigational RNA Interference Agents for Hepatitis B.","authors":"Rex Wan-Hin Hui, Lung-Yi Mak, Wai-Kay Seto, Man-Fung Yuen","doi":"10.1007/s40259-024-00694-x","DOIUrl":"10.1007/s40259-024-00694-x","url":null,"abstract":"<p><p>Functional cure of chronic hepatitis B (CHB)-defined as sustained seroclearance of hepatitis B surface antigen (HBsAg) with unquantifiable hepatitis B virus (HBV) DNA at 24 weeks off treatment, is an optimal treatment endpoint. Nonetheless, it cannot be consistently attained by current treatment modalities. RNA interference (RNAi) is a novel treatment strategy using small-interfering RNA (siRNA) or antisense oligonucleotide (ASO) to target HBV post-transcriptional RNA, in turn suppressing viral protein production and replication. Hence, RNAi has indirect effects in promoting host immune reconstitution against HBV. Multiple RNAi therapeutics have entered phase II/III clinical trials, demonstrating potent, dose-dependent, and sustainable effects in suppressing HBsAg. Incidences of HBsAg seroclearance, particularly with the use of ASO, have also been documented. The combination of RNAi with other antivirals/immunomodulators (e.g. pegylated interferon), have shown promising results in potentiating RNAi effects and enhancing treatment durability. Further research will be required to establish predictors of response, optimal treatment protocols, and long-term outcomes in patients on RNAi. RNAi therapeutics have shown promising results and will likely be the keystone of future HBV treatment.</p>","PeriodicalId":9022,"journal":{"name":"BioDrugs","volume":" ","pages":"21-32"},"PeriodicalIF":5.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11750937/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142790963","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Acknowledgement to Referees. 给推荐人的确认函。
IF 5.4 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-12-24 DOI: 10.1007/s40259-024-00697-8
{"title":"Acknowledgement to Referees.","authors":"","doi":"10.1007/s40259-024-00697-8","DOIUrl":"https://doi.org/10.1007/s40259-024-00697-8","url":null,"abstract":"","PeriodicalId":9022,"journal":{"name":"BioDrugs","volume":" ","pages":""},"PeriodicalIF":5.4,"publicationDate":"2024-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142881069","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
BioDrugs
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1