1
|
Carlisle JW, Wolner Z, Pannu S, Mitchell C, Hsu M, Aijaz A, Johnson M, Naqash AR. Bispecific Antibodies in Non-Small Cell Lung Cancer: From Targeted Innovation to Real-World Integration. Am Soc Clin Oncol Educ Book 2025; 45:e472792. [PMID: 40397846 DOI: 10.1200/edbk-25-472792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Bispecific antibodies have ushered in a transformative era in treating non-small cell lung cancer (NSCLC), enabling dual-pathway targeting with promising clinical outcomes in previously refractory disease subsets. Recent US Food and Drug Administration approvals-including amivantamab, an epidermal growth factor receptor (EGFR)/mesenchymal-epithelial transition factor-targeting monoclonal antibody for EGFR exon 20 insertions and frontline EGFR-mutant (Exon 19 and 21) NSCLC, and zenocutuzumab for tumors harboring neuregulin 1 fusions-highlight their expanding therapeutic footprint. However, a new spectrum of on-target toxicities and implementation challenges are essential considerations as part of this innovation. This review dissects the evolving clinical data for bispecific antibodies in NSCLC, focusing on amivantamab, and provides a practical framework for managing dermatologic, infusion-related, and class-specific adverse events. We explore quality-of-life outcomes, financial toxicity, and the role of subcutaneous formulations in improving patient adherence and treatment experience. Furthermore, we highlight an emerging PD-1/vascular endothelial growth factor-A bispecific antibody (ivonescimab) and its potential to reshape frontline therapy paradigms in NSCLC. By integrating clinical trial evidence with real-world considerations, this review aims to equip oncologists with the tools to optimize the use of bispecific antibodies in NSCLC and guide future therapeutic integration.
Collapse
Affiliation(s)
- Jennifer W Carlisle
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA
- Winship Cancer Institute of Emory University, Atlanta, GA
| | - Zachary Wolner
- Winship Cancer Institute of Emory University, Atlanta, GA
| | - Sagal Pannu
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK
| | | | - Melinda Hsu
- Case Western Reserve University, Cleveland, OH
| | - Ayesha Aijaz
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK
| | | | | |
Collapse
|
2
|
Rahal Z, El Darzi R, Moghaddam SJ, Cascone T, Kadara H. Tumour and microenvironment crosstalk in NSCLC progression and response to therapy. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01021-1. [PMID: 40379986 DOI: 10.1038/s41571-025-01021-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2025] [Indexed: 05/19/2025]
Abstract
The treatment landscape of non-small-cell lung cancer (NSCLC) is evolving rapidly, driven by advances in the development of targeted agents and immunotherapies. Despite this progress, some patients have suboptimal responses to treatment, highlighting the need for new therapeutic strategies. In the past decade, the important role of the tumour microenvironment (TME) in NSCLC progression, metastatic dissemination and response to treatment has become increasingly evident. Understanding the complexity of the TME and its interactions with NSCLC can propel efforts to improve current treatment modalities, overcome resistance and develop new treatments, which will ultimately improve the outcomes of patients. In this Review, we provide a comprehensive view of the NSCLC TME, examining its components and highlighting distinct archetypes characterized by spatial niches within and surrounding tumour nests, which form complex neighbourhoods. Next, we explore the interactions within these components, focusing on how inflammation and immunosuppression shape the dynamics of the NSCLC TME. We also address the emerging influences of patient-related factors, such as ageing, sex and health disparities, on the NSCLC-TME crosstalk. Finally, we discuss how various therapeutic strategies interact with and are influenced by the TME in NSCLC. Overall, we emphasize the interconnectedness of these elements and how they influence therapeutic outcomes and tumour progression.
Collapse
Affiliation(s)
- Zahraa Rahal
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Roy El Darzi
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate School of Biomedical Sciences (GSBS), UTHealth Houston, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tina Cascone
- Graduate School of Biomedical Sciences (GSBS), UTHealth Houston, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Thoracic-Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Graduate School of Biomedical Sciences (GSBS), UTHealth Houston, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
3
|
Zhang M, Liu C, Tu J, Tang M, Ashrafizadeh M, Nabavi N, Sethi G, Zhao P, Liu S. Advances in cancer immunotherapy: historical perspectives, current developments, and future directions. Mol Cancer 2025; 24:136. [PMID: 40336045 PMCID: PMC12057291 DOI: 10.1186/s12943-025-02305-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 03/15/2025] [Indexed: 05/09/2025] Open
Abstract
Cancer immunotherapy, encompassing both experimental and standard-of-care therapies, has emerged as a promising approach to harnessing the immune system for tumor suppression. Experimental strategies, including novel immunotherapies and preclinical models, are actively being explored, while established treatments, such as immune checkpoint inhibitors (ICIs), are widely implemented in clinical settings. This comprehensive review examines the historical evolution, underlying mechanisms, and diverse strategies of cancer immunotherapy, highlighting both its clinical applications and ongoing preclinical advancements. The review delves into the essential components of anticancer immunity, including dendritic cell activation, T cell priming, and immune surveillance, while addressing the challenges posed by immune evasion mechanisms. Key immunotherapeutic strategies, such as cancer vaccines, oncolytic viruses, adoptive cell transfer, and ICIs, are discussed in detail. Additionally, the role of nanotechnology, cytokines, chemokines, and adjuvants in enhancing the precision and efficacy of immunotherapies were explored. Combination therapies, particularly those integrating immunotherapy with radiotherapy or chemotherapy, exhibit synergistic potential but necessitate careful management to reduce side effects. Emerging factors influencing immunotherapy outcomes, including tumor heterogeneity, gut microbiota composition, and genomic and epigenetic modifications, are also examined. Furthermore, the molecular mechanisms underlying immune evasion and therapeutic resistance are analyzed, with a focus on the contributions of noncoding RNAs and epigenetic alterations, along with innovative intervention strategies. This review emphasizes recent preclinical and clinical advancements, with particular attention to biomarker-driven approaches aimed at optimizing patient prognosis. Challenges such as immunotherapy-related toxicity, limited efficacy in solid tumors, and production constraints are highlighted as critical areas for future research. Advancements in personalized therapies and novel delivery systems are proposed as avenues to enhance treatment effectiveness and accessibility. By incorporating insights from multiple disciplines, this review aims to deepen the understanding and application of cancer immunotherapy, ultimately fostering more effective and widely accessible therapeutic solutions.
Collapse
Affiliation(s)
- Meiyin Zhang
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Chaojun Liu
- Department of Breast Surgery, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University; People's Hospital of Henan University, Zhengzhou, Henan, 450003, China
| | - Jing Tu
- Department of Pulmonary and Critical Care Medicine, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Min Tang
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, V8 V 1P7, Canada
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR) Yong Loo Lin, School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Peiqing Zhao
- Translational Medicine Center, Zibo Central Hospital Affiliated to Binzhou Medical University, No. 54 Communist Youth League Road, Zibo, China.
| | - Shijian Liu
- Department of General Medicine, The 2nd Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Harbin, 150081, China.
| |
Collapse
|
4
|
Zhang Y, Liu X, Ren S. Ivonescimab in non-small cell lung cancer: harmonizing immunotherapy and anti-angiogenesis. Expert Opin Biol Ther 2025; 25:1-7. [PMID: 40162997 DOI: 10.1080/14712598.2025.2487512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Immunotherapy combined with anti-angiogenesis has become a useful strategy in cancer treatment. Ivonescimab, the first-approved bispecific antibody targeting both immune checkpoint inhibition and anti-angiogenesis, represents a breakthrough over the conventional dual-drug combination approach. The emerging clinical evidence demonstrates promising efficacy and manageable safety profile of ivonescimab in the treatment of non-small cell lung cancer (NSCLC), suggesting its potential role as a cornerstone in the next generation of cancer immunotherapy. AREAS COVERED This review presents the pharmacological characteristics of ivonescimab, revisits relevant clinical trials and key data, and provides an in-depth analysis. Additionally, the potential of ivonescimab in NSCLC treatment is discussed, along with its clinical prospects. EXPERT OPINION The available clinical data demonstrate that simultaneously targeting both immune checkpoint inhibition and angiogenesis pathways through a single bispecific antibody represents a significant therapeutic advancement in NSCLC treatment. Ivonescimab's innovative dual-targeting mechanism, supported by promising efficacy data from the HARMONi trials and its manageable safety profile, appears to be fundamental to its potential to challenge current standards of care. As the first approved bispecific antibody with this unique mechanism of action, ivonescimab may not only transform current treatment paradigms but also pioneer a new direction in cancer immunotherapy.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Xinyu Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Cancer Institute, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Lizama-Muñoz A, Plaza-Diaz J. Bispecific Antibodies, Nanobodies and Extracellular Vesicles: Present and Future to Cancer Target Therapy. Biomolecules 2025; 15:639. [PMID: 40427532 PMCID: PMC12109199 DOI: 10.3390/biom15050639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Cancer remains one of the leading causes of mortality worldwide, with a growing need for precise and effective treatments. Traditional therapies such as chemotherapy and radiotherapy have limitations, including off-target effects and drug resistance. In recent years, targeted therapies have emerged as promising alternatives, aiming to improve treatment specificity and reduce systemic toxicity. Among the most innovative approaches, bispecific antibodies, nanobodies, and extracellular vesicles offer distinct and complementary mechanisms for cancer therapy. Bispecific antibodies enhance immune responses and enable dual-targeting of cancer cells, nanobodies provide superior tumor penetration due to their small size, and extracellular vesicles present a novel platform for drug and RNA delivery. This work aims to review and analyze these three approaches, assessing their current applications, advantages, challenges, and future perspectives.
Collapse
Affiliation(s)
- Asier Lizama-Muñoz
- Department of Biochemistry, Molecular Biology and Immunology III, Faculty of Medicine, University of Granada, 18016 Granada, Spain;
- Clinical Analysis and Immunology Department, University Hospital Virgen de las Nieves, 18014 Granada, Spain
| | - Julio Plaza-Diaz
- ANUT-DSM (Alimentaciò, Nutrició Desenvolupament i Salut Mental), Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, 43201 Reus, Spain
- School of Health Sciences, Universidad Internacional de La Rioja, Avenida de la Paz 137, 26006 Logroño, Spain
| |
Collapse
|
6
|
Xiong X, Liu W, Yao C. Development of an alkaliptosis-related lncRNA risk model and immunotherapy target analysis in lung adenocarcinoma. Front Genet 2025; 16:1573480. [PMID: 40264452 PMCID: PMC12011837 DOI: 10.3389/fgene.2025.1573480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/28/2025] [Indexed: 04/24/2025] Open
Abstract
Background Lung cancer has the highest mortality rate among all cancers worldwide. Alkaliptosis is characterized by a pH-dependent form of regulated cell death. In this study, we constructed a model related to alkaliptosis-associated long non-coding RNAs (lncRNAs) and developed a prognosis-related framework, followed by the identification of potential therapeutic drugs. Methods The TCGA database was utilized to obtain RNA-seq-based transcriptome profiling data, clinical information, and mutation data. We conducted multivariate Cox regression analysis to identify alkaliptosis-related lncRNAs. Subsequently, we employed the training group to construct the prognostic model and utilized the testing group to validate the model's accuracy. Calibration curves were generated to illustrate the discrepancies between predicted and observed outcomes. Principal Component Analysis (PCA) was performed to investigate the distribution of LUAD patients across high- and low-risk groups. Additionally, Gene Ontology (GO) and Gene Set Enrichment Analysis (GSEA) were conducted. Immune cell infiltration and Tumor Mutational Burden (TMB) analyses were carried out using the CIBERSORT and maftools algorithms. Finally, the "oncoPredict" package was employed to predict immunotherapy sensitivity and to further forecast potential anti-tumor immune drugs. qPCR was used for experimental verification. Results We identified 155 alkaliptosis-related lncRNAs and determined that 5 of these lncRNAs serve as independent prognostic factors. The progression-free survival (PFS) and overall survival (OS) rates of the low-risk group were significantly higher than those of the high-risk group. The risk signature functions as a prognostic factor that is independent of other variables. Different stages (I-II and III-IV) effectively predict the survival rates of lung adenocarcinoma (LUAD) patients, and these lncRNAs can reliably forecast these signatures. GSEA revealed that processes related to chromosome segregation and immune response activation were significantly enriched in both the high- and low-risk groups. The high-risk group exhibited a lower fraction of plasma cells and a higher proportion of activated CD4 memory T cells. Additionally, the OS of the low TMB group was significantly lower compared to the high TMB group. Furthermore, drug sensitivity was significantly greater in the high-risk group than in the low-risk group. These lncRNAs may serve as biomarkers for treating LUAD patients. Conclusion In summary, the construction of an alkaliptosis-related lncRNA prognostic model and drug sensitivity analysis in LUAD patients provides new insights into the clinical diagnosis and treatment of advanced LUAD patients.
Collapse
Affiliation(s)
| | | | - Chuan Yao
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| |
Collapse
|
7
|
Yang S, Fang Y, Ma Y, Wang F, Wang Y, Jia J, Yang Y, Sun W, Zhou Q, Li Z. Angiogenesis and targeted therapy in the tumour microenvironment: From basic to clinical practice. Clin Transl Med 2025; 15:e70313. [PMID: 40268524 PMCID: PMC12017902 DOI: 10.1002/ctm2.70313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
Angiogenesis, as a core marker of cancer survival and growth, is integral to the processes of tumour growth, invasion and metastasis. In recent years, targeted angiogenesis treatment strategies have gradually become an important direction in cancer treatment. Single-cell sequencing technology can provide new insights into targeted angiogenesis by providing a deeper understanding of the heterogeneity of tumour endothelial cells and exploring the interactions between endothelial cells and surrounding cells in the tumour microenvironment. Here, we systematically review the research progress in endothelial cell pathophysiology and its endothelial‒mesenchymal transition and illustrate the heterogeneity of endothelial cells from a single-cell perspective. Finally, we examine the contributions of different cell types within the tumour microenvironment in relation to tumour angiogenesis, as well as the latest progress and strategies in targeted angiogenesis therapy, hoping to provide useful insights into the clinical application of antiangiogenic treatment. Furthermore, a summary of the present progress in the development of potential angiogenesis inhibitors and the ongoing clinical trials for combination therapies is provided. KEY POINTS: Angiogenesis plays a key role in tumour progression, invasion and metastasis, so strategies targeting angiogenesis are gradually becoming an important direction in cancer therapy. Interactions between endothelial cells and stromal cells and immune cells in the tumour microenvironment are significant in angiogenesis. The application of antiangiogenic immunotherapy and nanotechnology in antiangiogenic therapy provides a vital strategy for prolonging the survival of cancer patients.
Collapse
Affiliation(s)
- Shuaixi Yang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yingshuai Fang
- The First Clinical School of MedicineZhengzhou UniversityZhengzhouChina
| | - Yangcheng Ma
- Department of OrthopedicsThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Fuqi Wang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yuhang Wang
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jiachi Jia
- The First Clinical School of MedicineZhengzhou UniversityZhengzhouChina
| | - Yabing Yang
- The First Clinical School of MedicineZhengzhou UniversityZhengzhouChina
| | - Weipeng Sun
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Quanbo Zhou
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Zhen Li
- Department of Colorectal SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
8
|
Chu T, Zhong H, Yu Z, Wang J, Zhao Y, Mu X, Yu X, Shi X, Shi Q, Guan M, Ding C, Geng N, Qian J, Han B. Efficacy and safety of first-line sintilimab plus anlotinib versus chemotherapy for metastatic non-small cell lung cancer: a phase II, open-label, randomized controlled trial. Cancer Commun (Lond) 2025; 45:442-455. [PMID: 39791315 PMCID: PMC11999892 DOI: 10.1002/cac2.12654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 12/03/2024] [Accepted: 12/24/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND The prognosis for non-small cell lung cancer (NSCLC) patients treated with standard platinum-based chemotherapy was suboptimal, with safety concerns. Following encouraging results from a preliminary phase I study, this phase II trial investigated the efficacy and safety of first-line sintilimab and anlotinib in metastatic NSCLC. METHODS In this open-label, randomized controlled trial (NCT04124731), metastatic NSCLC without epithelial growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), or proto-oncogene tyrosine-protein kinase ROS (ROS1) mutations, and previous treatments for metastatic disease were enrolled. Participants were randomly assigned in a 1:1 ratio to either sintilimab (200 mg every 3 weeks) plus anlotinib (12 mg D1-14 every 3 weeks) or a standard platinum-based chemotherapy regimen. Patients in the chemotherapy group were permitted to switch to sintilimab after disease progression. The primary endpoint was the objective response rate (ORR). RESULTS From November 2019 to March 2023, 99 patients were randomized into the sintilimab plus anlotinib group (n = 49) and the chemotherapy group (n = 50). The ORR was significantly higher in the sintilimab plus anlotinib group (44.9%; 95% confidence interval [CI] = 30.7%-59.8%) compared to the chemotherapy group (18.0%; 95% CI = 8.6%-31.4%, P = 0.003). Progression-free survival (PFS) was also notably longer (median: 14.4 vs. 5.6 months; hazard ratio [HR] = 0.39; 95% CI = 0.23-0.67; P < 0.001). The 24-month overall survival rate was 58.4% (95% CI = 40.4%-72.6%) and 43.2% (95% CI = 26.0%-59.2%), respectively. The rate of grade 3 or higher treatment-related adverse events was lower in the sintilimab plus anlotinib group (28.0%) than in the chemotherapy group (49.0%), especially for the hematological toxicities. CONCLUSION First-line sintilimab plus anlotinib showed improved ORR and PFS, alongside a superior safety profile, compared to the standard platinum-based chemotherapy for metastatic NSCLC patients.
Collapse
Affiliation(s)
- Tianqing Chu
- Department of Respiratory and Critical Care MedicineChest Hospital Affiliated to Shanghai Jiao Tong UniversityShanghaiP. R. China
| | - Hua Zhong
- Department of Respiratory and Critical Care MedicineChest Hospital Affiliated to Shanghai Jiao Tong UniversityShanghaiP. R. China
| | - Zhuang Yu
- Department of Oncologythe Affiliated Hospital of Qingdao UniversityQingdaoShandongP. R. China
| | - Jing Wang
- Department of Oncologythe Affiliated Hospital of Qingdao UniversityQingdaoShandongP. R. China
| | - Yanqiu Zhao
- Department of Respiratory MedicineHenan Cancer Hospital/Affiliated Cancer Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Xiaoqian Mu
- Department of Respiratory MedicineHenan Cancer Hospital/Affiliated Cancer Hospital of Zhengzhou UniversityZhengzhouHenanP. R. China
| | - Xinmin Yu
- Department of Thoracic OncologyCancer Hospital Affiliated to the University of Chinese Academy of SciencesHangzhouZhejiangP. R. China
| | - Xun Shi
- Department of Thoracic OncologyCancer Hospital Affiliated to the University of Chinese Academy of SciencesHangzhouZhejiangP. R. China
| | - Qingming Shi
- Department of Medical OncologyAnhui Chest HospitalHefeiAnhuiP. R. China
| | - Maojing Guan
- Department of Medical OncologyAnhui Chest HospitalHefeiAnhuiP. R. China
| | - Cuimin Ding
- Department of Respiratory MedicineThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiP. R. China
| | - Nan Geng
- Department of Respiratory MedicineThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiP. R. China
| | - Jialin Qian
- Department of Respiratory and Critical Care MedicineChest Hospital Affiliated to Shanghai Jiao Tong UniversityShanghaiP. R. China
| | - Baohui Han
- Department of Respiratory and Critical Care MedicineChest Hospital Affiliated to Shanghai Jiao Tong UniversityShanghaiP. R. China
| |
Collapse
|
9
|
Zhong T, Zhang L, Huang Z, Pang X, Jin C, Liu W, Du J, Yin W, Chen N, Min J, Xia M, Li B. Design of a fragment crystallizable-engineered tetravalent bispecific antibody targeting programmed cell death-1 and vascular endothelial growth factor with cooperative biological effects. iScience 2025; 28:111722. [PMID: 40034861 PMCID: PMC11872405 DOI: 10.1016/j.isci.2024.111722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/08/2024] [Accepted: 12/30/2024] [Indexed: 03/05/2025] Open
Abstract
Clinical studies have shown that combination therapy of PD-1 and VEGF antibodies significantly improves clinical benefit over PD-1 antibody alone in certain settings. Ivonescimab, an on-market tetravalent anti-PD-1/VEGF bispecific antibody, was designed to improve efficacy and safety over combo therapy. In this study, the mechanism of action is investigated. In the presence of VEGF, ivonescimab forms soluble complexes with VEGF dimers, leading to the enhanced binding avidity of ivonescimab to PD-1 therefore promoting its increased potency on PD-1/PD-L1-signaling blockade. Likewise, PD-1 binding enhanced ivonescimab binding to VEGF, therefore enhancing VEGF-signaling blockade. Furthermore, ivonescimab treatment demonstrated statistically significant anti-tumor response in vivo. Moreover, ivonescimab contains Fc-silencing mutations abrogating FcγRI/IIIa binding and showed significantly reduced effector function in vitro which is consistent with the better safety profile of ivonescimab in monkeys and humans. Briefly, ivonescimab displays unique cooperative binding and promotes the increased in vitro functional bioactivities with a favorable safety profile.
Collapse
Affiliation(s)
- Tingting Zhong
- Research and Development Department, Akeso Biopharma Inc., No.6 Shennong Road, Torch Development Zone, Zhongshan, Guangdong 528400, China
| | - Lingzhi Zhang
- Research and Development Department, Akeso Biopharma Inc., No.6 Shennong Road, Torch Development Zone, Zhongshan, Guangdong 528400, China
| | - Zhaoliang Huang
- Research and Development Department, Akeso Biopharma Inc., No.6 Shennong Road, Torch Development Zone, Zhongshan, Guangdong 528400, China
| | - Xinghua Pang
- Research and Development Department, Akeso Biopharma Inc., No.6 Shennong Road, Torch Development Zone, Zhongshan, Guangdong 528400, China
| | - Chunshan Jin
- Research and Development Department, Akeso Biopharma Inc., No.6 Shennong Road, Torch Development Zone, Zhongshan, Guangdong 528400, China
| | - Wenrong Liu
- Research and Development Department, Akeso Biopharma Inc., No.6 Shennong Road, Torch Development Zone, Zhongshan, Guangdong 528400, China
| | - Juan Du
- Research and Development Department, Akeso Biopharma Inc., No.6 Shennong Road, Torch Development Zone, Zhongshan, Guangdong 528400, China
| | - Wen Yin
- Research and Development Department, Akeso Biopharma Inc., No.6 Shennong Road, Torch Development Zone, Zhongshan, Guangdong 528400, China
| | - Na Chen
- Research and Development Department, Akeso Biopharma Inc., No.6 Shennong Road, Torch Development Zone, Zhongshan, Guangdong 528400, China
| | - Jing Min
- Research and Development Department, Akeso Biopharma Inc., No.6 Shennong Road, Torch Development Zone, Zhongshan, Guangdong 528400, China
| | - Michelle Xia
- President’s Office, Akeso Biopharma Inc., No.6 Shennong Road, Torch Development Zone, Zhongshan, Guangdong 528400, China
| | - Baiyong Li
- Research and Development Department, Akeso Biopharma Inc., No.6 Shennong Road, Torch Development Zone, Zhongshan, Guangdong 528400, China
| |
Collapse
|
10
|
Xiong A, Wang L, Chen J, Wu L, Liu B, Yao J, Zhong H, Li J, Cheng Y, Sun Y, Ge H, Yao J, Shi Q, Zhou M, Chen B, Han Z, Wang J, Bu Q, Zhao Y, Chen J, Nie L, Li G, Li X, Yu X, Ji Y, Sun D, Ai X, Chu Q, Lin Y, Hao J, Huang D, Zhou C, Shan J, Yang H, Liu X, Wang J, Shang Y, Mei X, Yang J, Lu D, Hu M, Wang ZM, Li B, Xia M, Zhou C. Ivonescimab versus pembrolizumab for PD-L1-positive non-small cell lung cancer (HARMONi-2): a randomised, double-blind, phase 3 study in China. Lancet 2025; 405:839-849. [PMID: 40057343 DOI: 10.1016/s0140-6736(24)02722-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 05/13/2025]
Abstract
BACKGROUND Ivonescimab is a bispecific antibody against programmed cell death protein 1 and vascular endothelial growth factor, yielding promising clinical outcomes for patients with advanced non-small cell lung cancer in early-phase studies. We compared the efficacy and safety of ivonescimab with pembrolizumab in patients with programmed cell death ligand-1 (PD-L1)-positive advanced non-small cell lung cancer. METHODS HARMONi-2 is a randomised, double-blind, phase 3 trial across 55 hospitals in China. Eligible patients were aged 18 years or older and had locally advanced or metastatic PD-L1-positive non-small cell lung cancer without sensitising epidermal growth factor receptor mutations or anaplastic lymphoma kinase translocations and an Eastern Cooperative Oncology Group performance-status of 0 or 1. Patients were randomly assigned (1:1) to receive 20 mg/kg ivonescimab or 200 mg pembrolizumab intravenously every 3 weeks. Randomisation was stratified by histology, clinical stage, and PD-L1 expression. The primary endpoint was progression-free survival (PFS) assessed by a masked independent radiographic review committee per RECIST v1.1 in the intention-to-treat population. This study is registered with ClinicalTrials.gov, NCT05499390; recruitment is complete, with the trial ongoing and final analysis to be reported later. FINDINGS Between Nov 9, 2022, and Aug 26, 2023, 398 (45%) of 879 screened patients were randomly assigned to receive ivonescimab (n=198) or pembrolizumab (n=200). At the preplanned interim analysis, median PFS was significantly longer with ivonescimab than with pembrolizumab (11·1 vs 5·8 months; stratified hazard ratio [HR] 0·51 [95% CI 0·38-0·69]; one-sided p<0·0001). The PFS benefit of ivonescimab over pembrolizumab was broadly consistent within prespecified subgroups, including patients with PD-L1 tumour proportion score (TPS) 1-49% (HR 0·54 [95% CI 0·37-0·78]) and PD-L1 TPS of 50% of higher (HR 0·48 [0·29-0·79]). Grade 3 or higher treatment-related adverse events occurred in 58 (29%) patients with ivonescimab and 31 (16%) patients with pembrolizumab. Immune-related adverse events of grade 3 or higher were observed in 14 (7%) of 197 patients on ivonescimab and 16 (8%) of 199 patients on pembrolizumab. Ivonescimab demonstrated a manageable safety profile in patients with both squamous and non-squamous non-small cell lung cancer. In patients with squamous cell carcinoma, grade 3 or higher treatment-related adverse events were comparable between the two groups. INTERPRETATION Ivonescimab significantly improved PFS compared with pembrolizumab in previously untreated patients with advanced PD-L1 positive non-small cell lung cancer. Therefore, ivonescimab might represent another treatment option in the first-line setting for PD-L1-positive advanced non-small cell lung cancer. FUNDING Akeso Biopharma.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/administration & dosage
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/pathology
- Male
- Female
- Lung Neoplasms/drug therapy
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Middle Aged
- Double-Blind Method
- Aged
- China
- B7-H1 Antigen/metabolism
- Antibodies, Bispecific/therapeutic use
- Antibodies, Bispecific/adverse effects
- Antibodies, Bispecific/administration & dosage
- Adult
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/administration & dosage
- Progression-Free Survival
- Immune Checkpoint Inhibitors/therapeutic use
- Immune Checkpoint Inhibitors/adverse effects
Collapse
Affiliation(s)
- Anwen Xiong
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lei Wang
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | | | - Lin Wu
- Hunan Cancer Hospital, Changsha, China
| | - Baogang Liu
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Jun Yao
- The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Hua Zhong
- Shanghai Chest Hospital, Shanghai, China
| | - Jie Li
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | | | - Yulan Sun
- Shandong Cancer Hospital and Institute, Jinan, China
| | - Hui Ge
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jifang Yao
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qin Shi
- Fuzhou Tuberculosis Prevention and Treatment Hospital, Fuzhou, China
| | - Ming Zhou
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | | | - Zhengxiang Han
- The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jinliang Wang
- The Fifth Medical Center of the Chinese People's Liberation Army General Hospital, Beijing, China
| | - Qing Bu
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | | | | | - Ligong Nie
- Peking University First Hospital, Beijing, China
| | | | - Xingya Li
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinmin Yu
- Zhejiang Cancer Hospital, Hangzhou, China
| | - Yinghua Ji
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | | | - Xiaohong Ai
- The First Affiliated Hospital of University of South China, Hengyang, China
| | - Qian Chu
- Huazhong University of Science and Technology Tongji Hospital, Tongji Medical College, Wuhan, China
| | - Yu Lin
- Fujian Cancer Hospital, Fuzhou, China
| | - Jiqing Hao
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dingzhi Huang
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Chengzhi Zhou
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinlu Shan
- Daping Hospital, Army Medical University, Chongqing, China
| | | | - Xuewen Liu
- Third Xiangya Hospital of Central South University, Changsha, China
| | - Jing Wang
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanhong Shang
- Affiliated Hospital of Hebei University, Baoding, China
| | - Xiaodong Mei
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, China
| | - Jie Yang
- Akeso Biopharma, Zhongshan, China
| | | | | | | | | | | | - Caicun Zhou
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China; Shanghai East Hospital, Shanghai, China.
| |
Collapse
|
11
|
Wang Y, Safi M, Hirsch FR, Lu S, Peters S, Govindan R, Rosell R, Park K, Zhang JJ. Immunotherapy for advanced-stage squamous cell lung cancer: the state of the art and outstanding questions. Nat Rev Clin Oncol 2025; 22:200-214. [PMID: 39762577 DOI: 10.1038/s41571-024-00979-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2024] [Indexed: 02/26/2025]
Abstract
Immune-checkpoint inhibitors (ICIs) have transformed the treatment paradigm for advanced-stage squamous non-small-cell lung cancer (LUSC), a histological subtype associated with inferior outcomes compared with lung adenocarcinoma. However, only a subset of patients derive durable clinical benefit. In the first-line setting, multiple ICI regimens are available, including anti-PD-(L)1 antibodies as monotherapy, in combination with chemotherapy, or with an anti-CTLA4 antibody with or without chemotherapy. Several important questions persist regarding the optimal regimen for individual patients, particularly how to identify patients who might benefit from adding chemotherapy and/or anti-CTLA4 antibodies to anti-PD-(L)1 antibodies. An urgent need exists for predictive biomarkers beyond PD-L1 to better guide precision oncology approaches. Deeper knowledge of the underlying molecular biology of LUSC and its implications for response to ICIs will be important in this regard. Integration of this knowledge into multi-omics methods coupled with artificial intelligence might enable the development of more robust biomarkers. Finally, several novel therapeutic strategies, including novel ICIs, bispecific antibodies and personalized cancer vaccines, are emerging. Addressing these unresolved questions through innovative clinical trials and translational research will be crucial to further improving the outcomes of patients with LUSC. In this Review, we provide a comprehensive overview of current immunotherapeutic approaches, unresolved challenges and emerging strategies for patients with LUSC.
Collapse
Affiliation(s)
- Yibei Wang
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mohammed Safi
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Fred R Hirsch
- Center for Thoracic Oncology, Tisch Cancer Institute and Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Shun Lu
- Department of Medical Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Solange Peters
- Oncology Department, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | | | - Rafael Rosell
- Dr. Rosell Oncology Institute, Dexeus University Hospital, Barcelona, Spain
| | - Keunchil Park
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
- Division of Hematology/Oncology, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Jianjun J Zhang
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, the University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
12
|
Wang F, Wei X, Zheng Y, Wang J, Ying J, Chen X, Luo S, Luo H, Yu X, Chen B, Ma L, Xu R. Safety, Pharmacokinetics, and Pharmacodynamics Evaluation of Ivonescimab, a Novel Bispecific Antibody Targeting PD-1 and VEGF, in Chinese Patients With Advanced Solid Tumors. Cancer Med 2025; 14:e70653. [PMID: 40114411 PMCID: PMC11925807 DOI: 10.1002/cam4.70653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Ivonescimab (AK112) is a first-in-class bispecific antibody that simultaneously targets programmed death-1 (PD-1) and vascular endothelial growth factor (VEGF) with cooperative binding. We report the safety, pharmacokinetics (PK), and pharmacodynamics (PD) profiles of ivonescimab in patients suffered from advanced solid tumors. METHODS A multicenter, open-label, dose-escalation, phase I study was conducted in five hospitals in China. Ivonescimab was used as a monotherapy. The dose of ivonescimab intravenously administered was 3, 5, 10, 20, and 30 mg/kg every 2 weeks (Q2W), and 10 and 20 mg/kg every 3 weeks (Q3W). Safety, PK, and PD of ivonescimab were evaluated. RESULTS A total of 59 patients treated in the study. Only one dose-limiting toxicity (DLT) occurred in 1 out of 9 patients in the 10 mg/kg Q2W cohort, indicating that no maximum tolerated dose was reached. Among the participants, 53 patients (89.8%) experienced treatment-related adverse events (TRAEs), with the most common being proteinuria (33.9%), aspartate aminotransferase elevation (27.1%), white blood cell count decrease (22.0%), alanine aminotransferase elevation (20.3%), and anemia (20.3%). Fourteen patients (23.7%) had ≥ Grade 3 TRAEs, and 7 patients (11.9%) experienced serious TRAEs. Notably, there were no reported deaths associated with the TRAEs, and no dose-dependent increase in adverse events was observed. The half-life of ivonescimab ranged from 5.0 to 7.3 days following single-dose administration across all dose levels. The serum concentrations of ivonescimab increased with escalating doses in an approximately dose-proportional manner. Following multiple doses, the accumulation ratio ranged from 1.1 to 1.7, suggesting mild accumulation of ivonescimab. The steady state was achieved after 5 doses. Ivonescimab occupancy on PD-1 sustained over 80% across the treatment period. Serum VEGF level was rapidly down-regulated after each administration. CONCLUSIONS In patients with advanced solid tumors, ivonescimab monotherapy was well-tolerated and demonstrated a linear PK characteristics. PD profiles showed the promising potential of ivonescimab for the management of advanced solid tumors. TRIAL REGISTRATION ClinicalTrials.gov (NCT04597541).
Collapse
Affiliation(s)
- Fenghua Wang
- Department of Medical Oncology, Cancer Prevention CenterSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Xiaoli Wei
- Department of Medical Oncology, Cancer Prevention CenterSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Yulong Zheng
- Department of Medical OncologyThe First Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
| | - Jing Wang
- Department of GynecologyHunan Cancer HospitalChangshaHunanChina
| | - Jieer Ying
- Department of Hepatobiliary Pancreatic Gastric MedicineZhejiang Cancer HospitalZhejiangHangzhouChina
| | - Xiaozhong Chen
- The Department of Head and Neck RadiationZhejiang Cancer HospitalZhejiangHangzhouChina
| | - Suxia Luo
- Department of Medical OncologyHenan Cancer HospitalZhengzhouHenanChina
| | - Huiyan Luo
- Department of Medical Oncology, Cancer Prevention CenterSun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Xufang Yu
- Akeso Biopharma, Inc.ZhongshanGuangdongChina
| | | | - Lei Ma
- Akeso Biopharma, Inc.ZhongshanGuangdongChina
| | - Ruihua Xu
- Department of Medical Oncology, Cancer Prevention CenterSun Yat‐Sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
13
|
Yang P, He S, Fan L, Ye L, Weng H. Risk factors for immunoresistance in advanced non-small cell lung cancer and the advantages of targeted therapy in improving prognosis. Am J Cancer Res 2025; 15:573-586. [PMID: 40084370 PMCID: PMC11897627 DOI: 10.62347/fgay1920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/17/2025] [Indexed: 03/16/2025] Open
Abstract
OBJECTIVES The advent of immunotherapy has transformed the therapeutic landscape for advanced non-small cell lung cancer (NSCLC); nonetheless, the emergence of resistance to immunotherapy poses a considerable obstacle. Our research sought to identify factors contributing to immunotherapy resistance and to assess the effectiveness of subsequent treatments in patients with advanced NSCLC who have been exposed to immune checkpoint inhibitors (ICIs). METHODS This retrospective study analyzed data from 232 individuals with advanced NSCLC who were treated with ICIs during January 2020 to December 2023. Based on their response to ICIs, these patients were classified into two groups: immunoresistance group (IM group) and non-immunoresistance group (NIM group). Data collected included demographics, clinical parameters, cytokine profiles, tumor mutational burden (TMB), PD-L1 expression, overall survival (OS), progression-free survival (PFS), and adverse events. The association between risk factors and immunoresistance were assessed, and second-line treatment outcomes were evaluated. RESULTS Key risk factors for immunoresistance included lower TMB, higher levels of interleukin-10 (IL-10), and PD-L1 expression ≥ 50%. TMB was inversely correlated with immunoresistance (rho = -0.838, P < 0.001). In multivariate analysis, IL-10 remained a significant risk factor (OR = 33.654, P = 0.021), whereas TMB was protective (OR = 0.786, P < 0.001). Second-line targeted therapy significantly improved OS (8.72 ± 2.02 months) and PFS (5.37 ± 2.15 months) compared to chemotherapy (OS: 7.93 ± 2.13 months; PFS: 4.86 ± 1.68 months) (P < 0.05). The targeted therapy group experienced distinct side effects, notably increased hypertension and hand-foot syndrome, while chemotherapy group had higher rates of fatigue (P < 0.05). CONCLUSION Immunoresistance in advanced NSCLC is influenced by IL-10, TMB, and PD-L1 expression. Targeted therapies offer superior outcomes than chemotherapy, though side effect management remains crucial.
Collapse
Affiliation(s)
- Ping Yang
- Department of Respiratory and Critical Care Medicine, The People’s Hospital Affiliated to Fujian University of Traditional Chinese MedicineFuzhou 350000, Fujian, China
| | - Shangxiang He
- Department of Medical Oncology, Shanghai GoBroad Cancer Hospital, China Pharmaceutical UniversityShanghai 200100, China
| | - Linyin Fan
- Department of Radiology, Zhejiang Cancer HospitalHangzhou 310022, Zhejiang, China
| | - Ling Ye
- Department of Respiratory and Critical Care Medicine, The People’s Hospital Affiliated to Fujian University of Traditional Chinese MedicineFuzhou 350000, Fujian, China
| | - Heng Weng
- Department of Respiratory and Critical Care Medicine, The People’s Hospital Affiliated to Fujian University of Traditional Chinese MedicineFuzhou 350000, Fujian, China
| |
Collapse
|
14
|
Chen Z, Wu L, Wang Q, Yu Y, Liu X, Ma R, Li T, Li Y, Song X, Li L, Zhao W, Wang Q, Xu X, Lu S. Brief Report: Ivonescimab Combined With Etoposide Plus Carboplatin as First-Line Treatment for Extensive-Stage SCLC: Results of a Phase 1b Clinical Trial. J Thorac Oncol 2025; 20:233-239. [PMID: 39490738 DOI: 10.1016/j.jtho.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/15/2024] [Accepted: 10/19/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION Ivonescimab is a humanized IgG1 bispecific anti-programmed cell death protein 1/vascular endothelial growth factor antibody. This study aimed to evaluate the safety and tolerance of ivonescimab combined with etoposide and carboplatin as first-line treatment in patients with extensive-stage SCLC and to explore the primary efficacy of this regimen. METHODS Eligible patients received intravenous ivonescimab 3 mg/kg, 10 mg/kg, or 20 mg/kg every 3 weeks combined with etoposide and carboplatin for up to four cycles, followed by ivonescimab as maintenance. The primary end points were safety and objective response rate (ORR). RESULTS Between April 23, 2021, and December 2, 2021, 35 patients were enrolled. At data cutoff (October 25, 2023), the median follow-up was 13.3 (range: 0.3-28.5) months. For all patients, the confirmed ORR and disease control rate were 80% and 91.4%, respectively. The ORR was 66.7%, 90.9%, and 76.2% at the dose of 3 mg/kg, 10 mg/kg, and 20 mg/kg, respectively. Grade more than or equal to 3 treatment-related adverse events (TRAEs) were observed in 21 patients (60%), and the most frequent toxicities were decreased neutrophil count (n = 8, 22.9%), decreased white blood cell count (n = 5, 14.3%), and anemia (n = 5, 14.3%). Grade more than or equal to 3 TRAEs occurred in 66.7%, 54.5%, and 61.9% of patients in 3, 10, and 20 mg/kg groups, respectively. TRAEs leading to death were reported in two patients (5.7%). Immune-related adverse events, most of them grade 1 or 2, occurred in 14 patients (40.0%). CONCLUSIONS Ivonescimab in combination with etoposide and carboplatin was well tolerated and found to have promising antitumor activity in extensive-stage SCLC.
Collapse
Affiliation(s)
- Zhiwei Chen
- Department of Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Lin Wu
- Department of Internal Thoracic Medicine, Hunan Cancer Hospital, Changsha, People's Republic of China
| | - Qiming Wang
- Department of Respiratory Medicine, Henan Cancer Hospital, Zhengzhou, People's Republic of China
| | - Yan Yu
- Department of Respiratory Medicine, Harbin Medical University Cancer Hospital, Harbin, People's Republic of China
| | - Xianling Liu
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Rui Ma
- Department of Internal Thoracic Medicine, Liaoning Cancer Hospital & Institute, Shenyang, People's Republic of China
| | - Tao Li
- Department of Radiology, Sichuan Cancer Hospital, Chengdu, People's Republic of China
| | - Yan Li
- Department of Medical Oncology, Shandong Provincial Qianfoshan Hospital/The First Affiliated Hospital of Shandong First Medical University, Jinan, People's Republic of China
| | - Xia Song
- Department of Respiratory Medicine, Shanxi Provincial Cancer Hospital, Taiyuan, People's Republic of China
| | - Lin Li
- Department of Medical Oncology, Beijing Hospital, Beijing, People's Republic of China
| | - Wei Zhao
- Department of Medical Oncology, Shandong Provincial Qianfoshan Hospital/The First Affiliated Hospital of Shandong First Medical University, Jinan, People's Republic of China
| | - Qiaoyun Wang
- Akeso Biopharma, Inc., Zhongshan, People's Republic of China
| | - Xiao Xu
- Akeso Biopharma, Inc., Zhongshan, People's Republic of China
| | - Shun Lu
- Department of Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China.
| |
Collapse
|
15
|
Daum S, Decristoforo L, Mousa M, Salcher S, Plattner C, Hosseinkhani B, Trajanoski Z, Wolf D, Carmeliet P, Pircher A. Unveiling the immunomodulatory dance: endothelial cells' function and their role in non-small cell lung cancer. Mol Cancer 2025; 24:21. [PMID: 39819502 PMCID: PMC11737145 DOI: 10.1186/s12943-024-02221-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 12/27/2024] [Indexed: 01/19/2025] Open
Abstract
The dynamic interactions between tumor endothelial cells (TECs) and the immune microenvironment play a critical role in the progression of non-small cell lung cancer (NSCLC). In general, endothelial cells exhibit diverse immunomodulatory properties, influencing immune cell recruitment, antigen presentation, and regulation of immune checkpoint expression. Understanding the multifaceted roles of TECs as well as assigning specific functional hallmarks to various TEC phenotypes offer new avenues for targeted development of therapeutic interventions, particularly in the context of advanced immunotherapy and anti-angiogenic treatments. This review provides insights into the complex interplay between TECs and the immune system in NSCLC including discussion of potential optimized therapeutic opportunities.
Collapse
Affiliation(s)
- Sophia Daum
- Internal Medicine 5, Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Tyrolean Cancer Research Institute (TKFI), Medical University Innsbruck, Innsbruck, Austria
| | - Lilith Decristoforo
- Internal Medicine 5, Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Tyrolean Cancer Research Institute (TKFI), Medical University Innsbruck, Innsbruck, Austria
| | - Mira Mousa
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Stefan Salcher
- Internal Medicine 5, Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Tyrolean Cancer Research Institute (TKFI), Medical University Innsbruck, Innsbruck, Austria
| | - Christina Plattner
- Institute of Bioinformatics, Biocenter Medical University Innsbruck, Innsbruck, Austria
| | - Baharak Hosseinkhani
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), VIB Center for Cancer Biology, KU Leuven, VIB, Leuven, Belgium
| | - Zlatko Trajanoski
- Institute of Bioinformatics, Biocenter Medical University Innsbruck, Innsbruck, Austria
| | - Dominik Wolf
- Internal Medicine 5, Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Tyrolean Cancer Research Institute (TKFI), Medical University Innsbruck, Innsbruck, Austria
| | - Peter Carmeliet
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), VIB Center for Cancer Biology, KU Leuven, VIB, Leuven, Belgium
| | - Andreas Pircher
- Internal Medicine 5, Department of Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Tyrolean Cancer Research Institute (TKFI), Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
16
|
Abdallah M, Voland R, Decamp M, Flickinger J, Pacioles T, Jamil M, Silbermins D, Shenouda M, Valsecchi M, Bir A, Shweihat Y, Bastidas J, Chowdhury N, Kachynski Y, Eldib H, Wright T, Mahdi A, Al-Nusair J, Nwanwene K, Varlotto J. Evaluation of Anti-Angiogenic Therapy Combined with Immunotherapy and Chemotherapy as a Strategy to Treat Locally Advanced and Metastatic Non-Small-Cell Lung Cancer. Cancers (Basel) 2024; 16:4207. [PMID: 39766108 PMCID: PMC11674749 DOI: 10.3390/cancers16244207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/23/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Immunotherapy has made recent improvements in disease-free survival (DFS) and/or overall survival (OS) in all stages of non-small-cell lung cancer (NSCLC). Here, we review the tumor microenvironment and its immunosuppressive effects and discuss how anti-angiogenic therapies may potentiate the anti-carcinogenic effects of immunotherapy. We also review all the past literature and discuss strategies of combining anti-angiogenic therapy and immunotherapy +/- chemotherapy and hypothesize how we can use this strategy for non-small-cell lung cancer in metastatic previously untreated/previously treated settings in previously treated EGFR-mutated NSCLC for the upfront treatment of brain metastases prior to radiation therapy and for the incorporation of this strategy into stage III unresectable disease. We assert the use of anti-angiogenic therapy and immunotherapy when combined appropriately with chemotherapy and radiotherapy has the potential to increase the long-term survivals in both the stage III and metastatic setting so that we can now consider more patients to experience curative treatment.
Collapse
Affiliation(s)
- Mahmoud Abdallah
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Rick Voland
- Department of Ophthalmology, University of Wisconsin, Madison, WI 53705, USA;
| | - Malcolm Decamp
- Division of Cardiothoracic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53726, USA;
| | - John Flickinger
- Department of Radiation Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA 15219, USA;
| | - Toni Pacioles
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Muhammad Jamil
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Damian Silbermins
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Mina Shenouda
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Matias Valsecchi
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Arvinder Bir
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Yousef Shweihat
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Juan Bastidas
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Nepal Chowdhury
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Yury Kachynski
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Howide Eldib
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - Thomas Wright
- Department of Internal Medicine, Marshall Health, Huntington, WV 25701, USA; (T.W.); (A.M.); (J.A.-N.)
| | - Ahmad Mahdi
- Department of Internal Medicine, Marshall Health, Huntington, WV 25701, USA; (T.W.); (A.M.); (J.A.-N.)
| | - Jowan Al-Nusair
- Department of Internal Medicine, Marshall Health, Huntington, WV 25701, USA; (T.W.); (A.M.); (J.A.-N.)
| | - Kemnasom Nwanwene
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| | - John Varlotto
- Department of Oncology, Edwards Comprehensive Cancer Institute, Marshall University, Huntington, WV 25701, USA; (M.A.); (T.P.); (M.J.); (D.S.); (M.S.); (M.V.); (A.B.); (Y.S.); (J.B.); (N.C.); (Y.K.); (H.E.); (K.N.)
| |
Collapse
|
17
|
Yao Y, Ren Y, Hou X, Wang P, Zhu J, Liu S, Ma X, Liu T, Yang Z, Zhu H, Li N. Construction and preclinical evaluation of a 124I-labelled bispecific antibody targeting PD-L1 and PD-L2. Eur J Nucl Med Mol Imaging 2024; 52:36-47. [PMID: 39155310 DOI: 10.1007/s00259-024-06886-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024]
Abstract
PURPOSE NB12 is a bispecific antibody that consists of two anti-programmed cell death-ligand 1 (PD-L1) nanobodies and two anti-programmed cell death-ligand 2 (PD-L2) nanobodies. The aim of this study was to design a novel tracer, [124I]I-NB12, targeting PD-L1/2 and perform preclinical evaluations to dynamically monitor PD-L1/2 expression for determining cancer patient responsiveness to ICI therapy. METHODS NB12 was labelled with the radionuclide 124I at room temperature (RT). An in vitro binding assay was performed to assess the affinity of [124I]I-NB12 for PD-L1 and PD-L2. Cellular uptake, pharmacokinetic, and biodistribution experiments were performed to evaluate the biological properties. Micro-PET/CT imaging with [124I]I-NB12 was conducted at different time points. Immunohistochemical and haematoxylin and eosin (HE) staining experiments were carried out using tumour tissues. Routine blood, biochemical indices and major organ pathology were used to evaluate the biosafety of the tracers. RESULTS The radiochemical yield of [124I]I-NB12 was 84.62 ± 3.90%, and the radiochemical purity (RCP) was greater than 99%. [124I]I-NB12 had a high affinity for the PD-L1 (Kd = 19.82 nM) and PD-L2 (Kd = 2.93 nM). Cellular uptake experiments confirmed that the uptake of [124I]I-NB12 by A549-PDL1/2 cells was greater than that by A549 cells. The half-lives of the distribution phase and elimination phase were 0.26 h and 4.08 h, respectively. Micro-PET/CT showed significant [124I]I-NB12 uptake in the tumour region of A549-PDL1/2 tumour-bearing mice compared with A549 tumour-bearing mice 24 h postinjection. Immunohistochemical and HE staining experiments confirmed that tumour-bearing mice was successfully constructed. CONCLUSION We constructed a bispecific antibody that targets PD-L1 and PD-L2, namely, [124I]I-NB12. Biological evaluation revealed its specificity and affinity for PD-L1/2, and micro-PET/CT confirmed the feasibility of visualizing tumour PD-L1/2 in vivo. Using [124I]I-NB12 may be a promising strategy for identifying cancer patients that can potentially benefit from ICI therapy.
Collapse
Affiliation(s)
- Yuan Yao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, No. 52 Fu-Cheng Rd, Beijing, 100142, People's Republic of China
| | - Yanan Ren
- Guizhou University School of Medicine, Guiyang, Guizhou, 550025, People's Republic of China
| | - Xingguo Hou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, No. 52 Fu-Cheng Rd, Beijing, 100142, People's Republic of China
| | - Pei Wang
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jinyu Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, No. 52 Fu-Cheng Rd, Beijing, 100142, People's Republic of China
| | - Song Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, No. 52 Fu-Cheng Rd, Beijing, 100142, People's Republic of China
| | - Xiaokun Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, No. 52 Fu-Cheng Rd, Beijing, 100142, People's Republic of China
| | - Teli Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, No. 52 Fu-Cheng Rd, Beijing, 100142, People's Republic of China
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, No. 52 Fu-Cheng Rd, Beijing, 100142, People's Republic of China.
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, No. 52 Fu-Cheng Rd, Beijing, 100142, People's Republic of China.
| | - Nan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, No. 52 Fu-Cheng Rd, Beijing, 100142, People's Republic of China.
| |
Collapse
|
18
|
Xu J, Tang Z. Progress on angiogenic and antiangiogenic agents in the tumor microenvironment. Front Oncol 2024; 14:1491099. [PMID: 39629004 PMCID: PMC11611712 DOI: 10.3389/fonc.2024.1491099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/31/2024] [Indexed: 12/06/2024] Open
Abstract
The development of tumors and their metastasis relies heavily on the process of angiogenesis. When the volume of a tumor expands, the resulting internal hypoxic conditions trigger the body to enhance the production of various angiogenic factors. These include vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), and transforming growth factor-α (TGF-α), all of which work together to stimulate the activation of endothelial cells and catalyze angiogenesis. Antiangiogenic therapy (AAT) aims to normalize tumor blood vessels by inhibiting these angiogenic signals. In this review, we will explore the molecular mechanisms of angiogenesis within the tumor microenvironment, discuss traditional antiangiogenic drugs along with their limitations, examine new antiangiogenic drugs and the advantages of combination therapy, and consider future research directions in the field of antiangiogenic drugs. This comprehensive overview aims to provide insights that may aid in the development of more effective anti-tumor treatments.
Collapse
Affiliation(s)
| | - Zhihua Tang
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing, China
| |
Collapse
|
19
|
Hockemeyer KG, Rusthoven CG, Pike LRG. Advances in the Management of Lung Cancer Brain Metastases. Cancers (Basel) 2024; 16:3780. [PMID: 39594735 PMCID: PMC11593022 DOI: 10.3390/cancers16223780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Lung cancer, both non-small cell and small cell, harbors a high propensity for spreading to the central nervous system. Radiation therapy remains the backbone of the management of brain metastases. Recent advances in stereotactic radiosurgery have expanded its indications and ongoing studies seek to elucidate optimal fractionation and coordination with systemic therapies, especially targeted inhibitors with intracranial efficacy. Efforts in whole-brain radiotherapy aim to preserve neurocognition and to investigate the need for prophylactic cranial irradiation. As novel combinatorial strategies are tested and prognostic/predictive biomarkers are identified and tested, the management of brain metastases in lung cancer will become increasingly personalized to optimally balance intracranial efficacy with preserving neurocognitive function and patient values.
Collapse
Affiliation(s)
- Kathryn G. Hockemeyer
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chad G. Rusthoven
- Department of Radiation Oncology, University of Colorado, Aurora, CO 80045, USA
| | - Luke R. G. Pike
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
20
|
Massariol Pimenta T, Carlos de Souza J, da Silva Martins B, Silva Butzene SM, Simões Padilha JM, Ganho Marçal M, Dos Santos Elias G, Rangel LBA. Emerging strategies to overcome ovarian cancer: advances in immunotherapy. Front Pharmacol 2024; 15:1490896. [PMID: 39564107 PMCID: PMC11573523 DOI: 10.3389/fphar.2024.1490896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024] Open
Abstract
Ovarian cancer is the second most common malignant neoplasm of gynecological origin and the leading cause of death from cancer in the female reproductive system worldwide. This scenario is largely due to late diagnoses, often in advanced stages, and the development of chemoresistance by cancer cells. These challenges highlight the need for alternative treatments, with immunotherapy being a promising option. Cancer immunotherapy involves triggering an anti-tumor immune response and developing immunological memory to eliminate malignant cells, prevent recurrence, and inhibit metastasis. Some ongoing research investigate potentially immunological advancements in the field of cancer vaccines, immune checkpoint blockade, CAR-T cell, and other strategies.
Collapse
Affiliation(s)
- Tatiana Massariol Pimenta
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Josiany Carlos de Souza
- Biotechnology Program/RENORBIO, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Bárbara da Silva Martins
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Solenny Maria Silva Butzene
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - José Matheus Simões Padilha
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Milleny Ganho Marçal
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Guilherme Dos Santos Elias
- Biochemistry Program, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Leticia Batista Azevedo Rangel
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
- Biotechnology Program/RENORBIO, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
- Biochemistry Program, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| |
Collapse
|
21
|
Croote D, Wong JJW, Creeks P, Aruva V, Landers JJ, Kwok M, Jama Z, Hamilton RG, Santos AF, O'Konek JJ, Ferrini R, Thomas GR, Lowman HB. Preclinical efficacy of peanut-specific IgG4 antibody therapeutic IGNX001. J Allergy Clin Immunol 2024; 154:1241-1248.e7. [PMID: 39069172 DOI: 10.1016/j.jaci.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/20/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Existing therapeutic strategies are challenged by long times to achieve effect and often require frequent administration. Peanut-allergic individuals would benefit from a therapeutic that provides rapid protection against accidental exposure within days of administration while carrying little risk of adverse reactions. OBJECTIVE Guided by the repertoire of human IgE mAbs from allergic individuals, we sought to develop a treatment approach leveraging the known protective effects of allergen-specific IgG4 antibodies. METHODS We applied our single-cell RNA-sequencing SEQ SIFTER platform (IgGenix, Inc, South San Francisco, Calif) to whole blood samples from peanut-allergic individuals to discover IgE mAbs. These were then class-switched by replacing the IgE constant region with IgG4 while retaining the allergen-specific variable regions. In vitro mast cell activation tests, basophil activation tests, ELISAs, and an in vivo peanut allergy mouse model were used to evaluate the specificity, affinity, and activity of these recombinant IgG4 mAbs. RESULTS We determined that human peanut-specific IgE mAbs predominantly target immunodominant epitopes on Ara h 2 and Ara h 6 and that recombinant IgG4 mAbs effectively block these epitopes. IGNX001, a mixture of 2 such high-affinity IgG4 mAbs, provided robust protection against peanut-mediated mast cell activation in vitro as well as against anaphylaxis upon intragastric peanut challenge in a peanut allergy mouse model. CONCLUSIONS We developed a peanut-specific IgG4 antibody therapeutic with convincing preclinical efficacy starting from a large repertoire of human IgE mAbs from demographically and geographically diverse individuals. These results warrant further clinical investigation of IGNX001 and underscore the opportunity for the application of this therapeutic development strategy in other food and environmental allergies.
Collapse
Affiliation(s)
| | | | | | - Venu Aruva
- IgGenix, Inc, South San Francisco, Calif
| | - Jeffrey J Landers
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich
| | - Matthew Kwok
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | - Zainab Jama
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | - Robert G Hamilton
- Division of Allergy and Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Alexandra F Santos
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom; Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, King's College London, London, United Kingdom; Children's Allergy Service, Evelina London, Guy's and St Thomas' Hospital, London, United Kingdom
| | - Jessica J O'Konek
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich
| | | | | | | |
Collapse
|
22
|
Wang L, Zhang L, Zhang Z, Wu P, Zhang Y, Chen X. Advances in targeting tumor microenvironment for immunotherapy. Front Immunol 2024; 15:1472772. [PMID: 39421736 PMCID: PMC11484021 DOI: 10.3389/fimmu.2024.1472772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
The tumor microenvironment (TME) provides essential conditions for the occurrence, invasion, and spread of cancer cells. Initial research has uncovered immunosuppressive properties of the TME, which include low oxygen levels (hypoxia), acidic conditions (low pH), increased interstitial pressure, heightened permeability of tumor vasculature, and an inflammatory microenvironment. The presence of various immunosuppressive components leads to immune evasion and affects immunotherapy efficacy. This indicates the potential value of targeting the TME in cancer immunotherapy. Therefore, TME remodeling has become an effective method for enhancing host immune responses against tumors. In this study, we elaborate on the characteristics and composition of the TME and how it weakens immune surveillance and summarize targeted therapeutic strategies for regulating the TME.
Collapse
Affiliation(s)
- Lugang Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liubo Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhen Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Wu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xinfeng Chen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
23
|
Dhillon S. Ivonescimab: First Approval. Drugs 2024; 84:1135-1142. [PMID: 39073550 DOI: 10.1007/s40265-024-02073-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2024] [Indexed: 07/30/2024]
Abstract
Ivonescimab (®) is a first-in-class, humanized, tetravalent bispecific monoclonal antibody targeting programmed cell death protein 1 (PD-1) and vascular endothelial growth factor (VEGF)-A being developed by Akeso Biopharma for the treatment of non-small cell lung cancer (NSCLC) and other solid tumours, including breast cancer, liver cancer and gastric cancer. Ivonescimab simultaneously blocks the binding of PD-1 to its ligand (PD-L1), thereby relieving PD-1/PD-L1-mediated immunosuppression, and blocks the binding of VEGF-A to its receptor (VEGFR2), thus blocking tumour angiogenesis in the tumour microenvironment. In May 2024, ivonescimab, in combination with pemetrexed and carboplatin, received its first approval in China for the treatment of patients with EGFR-mutated locally advanced or metastatic non-squamous NSCLC who have progressed after tyrosine kinase inhibitor (TKI) therapy. Clinical studies of ivonescimab are underway in multiple countries worldwide. This article summarizes the milestones in the development of ivonescimab leading to this first approval for EGFR-mutated locally advanced or metastatic non-squamous NSCLC who have progressed after TKI therapy.
Collapse
Affiliation(s)
- Sohita Dhillon
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
24
|
Frentzas S, Austria Mislang AR, Lemech C, Nagrial A, Underhill C, Wang W, Wang ZM, Li B, Xia Y, Coward JIG. Phase 1a dose escalation study of ivonescimab (AK112/SMT112), an anti-PD-1/VEGF-A bispecific antibody, in patients with advanced solid tumors. J Immunother Cancer 2024; 12:e008037. [PMID: 38642937 PMCID: PMC11033648 DOI: 10.1136/jitc-2023-008037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Studies showed that vascular endothelial growth factor (VEGF) inhibitors could improve therapeutic efficacy of PD-1/PD-L1 antibodies by transforming the immunosuppressive tumor microenvironment (TME) into an immunoresponsive TME. Ivonescimab is a first-in-class, humanized tetravalent bispecific antibody targeting PD-1 and VEGF-A simultaneously. Here, we report the first-in-human, phase 1a study of ivonescimab in patients with advanced solid tumors. METHODS Patients with advanced solid tumors were treated with ivonescimab 0.3, 1, 3, 10, 20 or 30 mg/kg intravenously every 2 weeks using a 3+3+3 dose escalation design. Dose expansion occurred at 10 and 20 mg/kg in selected tumor types. The primary objective was to assess the safety and tolerability, and to determine the maximum tolerated dose (MTD). The secondary objectives included pharmacokinetics, pharmacodynamics and preliminary antitumor activity based on Response Evaluation Criteria in Solid Tumors V.1.1. RESULTS Between October 2, 2019 and January 14, 2021, a total of 51 patients were enrolled and received ivonescimab. Two dose-limiting toxicities were reported at 30 mg/kg. The MTD of ivonescimab was 20 mg/kg every 2 weeks. Grade≥3 treatment-related adverse events (TRAEs) occurred in 14 patients (27.5%). The most common TRAEs of any grade were rash (29.4%), arthralgia (19.6%), hypertension (19.6%), fatigue (17.6%), diarrhea (15.7%) and pruritus (11.8%). The most common grade≥3 TRAEs were hypertension (7/51, 13.7%), alanine aminotransferase increased (3/51, 5.2%), aspartate aminotransferase increased (2/51, 3.9%) and colitis (2/51, 3.9%). Of 47 patients who had at least one postbaseline assessment, the confirmed objective response rate was 25.5% (12/47) and disease control rate was 63.8% (30/47). Among 19 patients with platinum-resistant ovarian cancer, 5 patients (26.3%) achieved partial response (PR). Efficacy signals were also observed in patients with mismatch repair proficient (pMMR) colorectal cancer, non-small cell lung cancer, and both MMR deficient and pMMR endometrial cancer. CONCLUSIONS Ivonescimab demonstrated manageable safety profiles and promising efficacy signals in multiple solid tumors. Exploration of alternative dosing regimens of ivonescimab monotherapy and combination therapies is warranted. TRIAL REGISTRATION NUMBER NCT04047290.
Collapse
Affiliation(s)
- Sophia Frentzas
- Department of Medical Oncology, Monash Health, Melbourne, Victoria, Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
| | - Anna Rachelle Austria Mislang
- Icon (Adelaide) Cancer Centre, Kurralta Park, South Australia, Australia
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - Charlotte Lemech
- Scientia Clinical Research Ltd, Sydney, New South Wales, Australia
| | - Adnan Nagrial
- Blacktown Cancer and Haematology Centre, Blacktown Hospital, University of Sydney, Sydney, New South Wales, Australia
| | - Craig Underhill
- Border Medical Oncology and Haematology Research Unit, Albury-Wodonga Regional Cancer Centre, Albury Wodonga, New South Wales, Australia
- University of New South Wales, Rural Medical School, Albury Campus, Sydney, New South Wales, Australia
| | | | | | | | - Yu Xia
- Akeso Biopharma, Inc, Zhongshan, China
| | - Jermaine I G Coward
- Icon Cancer Centre, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
25
|
Dammeijer F, Dumoulin DW, Aerts JGJV. Anti-Vascular Endothelial Growth Factor/Programmed Cell Death Protein 1 Bispecific Antibodies: Using Nunchucks to Fight an Old Adversary. J Thorac Oncol 2024; 19:366-369. [PMID: 38453322 DOI: 10.1016/j.jtho.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 12/01/2023] [Indexed: 03/09/2024]
Affiliation(s)
- Floris Dammeijer
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Erasmus University MC, Rotterdam, The Netherlands
| | - Daphne W Dumoulin
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Erasmus University MC, Rotterdam, The Netherlands
| | - Joachim G J V Aerts
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Erasmus University MC, Rotterdam, The Netherlands.
| |
Collapse
|
26
|
Surowka M, Klein C. A pivotal decade for bispecific antibodies? MAbs 2024; 16:2321635. [PMID: 38465614 PMCID: PMC10936642 DOI: 10.1080/19420862.2024.2321635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/12/2024] Open
Abstract
Bispecific antibodies (bsAbs) are a class of antibodies that can mediate novel mechanisms of action compared to monospecific monoclonal antibodies (mAbs). Since the discovery of mAbs and their adoption as therapeutic agents in the 1980s and 1990s, the development of bsAbs has held substantial appeal. Nevertheless, only three bsAbs (catumaxomab, blinatumomab, emicizumab) were approved through the end of 2020. However, since then, 11 bsAbs received regulatory agency approvals, of which nine (amivantamab, tebentafusp, mosunetuzumab, cadonilimab, teclistamab, glofitamab, epcoritamab, talquetamab, elranatamab) were approved for the treatment of cancer and two (faricimab, ozoralizumab) in non-oncology indications. Notably, of the 13 currently approved bsAbs, two, emicizumab and faricimab, have achieved blockbuster status, showing the promise of this novel class of therapeutics. In the 2020s, the approval of additional bsAbs can be expected in hematological malignancies, solid tumors and non-oncology indications, establishing bsAbs as essential part of the therapeutic armamentarium.
Collapse
Affiliation(s)
- Marlena Surowka
- Roche Innovation Center Zurich, Roche Pharma Research & Early Development, Roche Glycart AG, Schlieren, Switzerland
| | - Christian Klein
- Roche Innovation Center Zurich, Roche Pharma Research & Early Development, Roche Glycart AG, Schlieren, Switzerland
| |
Collapse
|