1
|
Xie H, Sun L, Yao S, Tian X, Jin L, Dai Y, Li Y, Li Y, Fang J, Guo P, Zhang Y. Therapeutically targeting endometrial cancer in preclinical models by ICAM1 antibody-drug conjugates. Gynecol Oncol 2025; 196:16-27. [PMID: 40147093 DOI: 10.1016/j.ygyno.2025.03.027] [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: 01/10/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE The incidence of mortality and morbidity from endometrial cancer (EC) is increasing annually, and there is a paucity of effective targeted therapies for the condition. Antibody-drug conjugates (ADCs) represent a promising approach to tumor-targeted therapy. In this study, we aim to identify a novel molecular target for the preclinical development of EC-targeted ADCs. METHODS Through quantitative and unbiased bioinformatics analyses intercellular adhesion molecule-1 (ICAM1) was identified as a potential cell membrane target. Two ADCs, ICAM1-MMAE and ICAM1-DXd, were subsequently developed by conjugating ICAM1 monoclonal antibodies with microtubule inhibitors and DNA topoisomerase inhibitors, respectively. The preclinical efficacy and biosafety of these ICAM1 ADCs were validated in both in vitro and in vivo models. Furthermore, transcriptomic analysis was conducted to elucidate the therapeutic effects of the ICAM1 ADCs. RESULTS Quantitative flow screening and bioinformatics analyses revealed significant overexpression of ICAM1 in EC. ICAM1-MMAE and ICAM1-DXd were developed using clinically effective linkers and payloads. In preclinical models, ICAM1 ADCs showed superior antitumor efficacy compared to standard chemotherapy, achieving sustained tumor regression with an excellent safety profile in both subcutaneous and orthotopic xenograft models. Transcriptomic analysis further revealed that ICAM1-DXd potently activated tumor immunity. CONCLUSIONS ICAM1 was identified as a promising cell membrane protein target for ADC development in EC. As-synthesized ICAM1 ADCs demonstrated potent antitumor activity, favorable biosafety profiles in vitro and in vivo, and the ability to activate tumor immunity. These findings support the potential of ICAM1 ADCs as a therapeutic strategy and warrant further investigation in clinical studies.
Collapse
Affiliation(s)
- Hanfei Xie
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou 310022, China
| | - Lu Sun
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China
| | - Shili Yao
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Xuefei Tian
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, Fudan University, Shanghai 201203, China
| | - Liming Jin
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Yujie Dai
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; MOE Frontier Science Centre for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Yuanzheng Li
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| | - Yuxuan Li
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China
| | - Jianmin Fang
- School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Peng Guo
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China; Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China; School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China; MOE Frontier Science Centre for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China; Eye Research Center, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Eye Hospital, Wenzhou Medical University, Hangzhou, Zhejiang 310018, China.
| | - Yingli Zhang
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou 310022, China.
| |
Collapse
|
2
|
Zhang P, Tao C, Xie H, Yang L, Lu Y, Xi Y, Yao S, Yuan L, Guo P, Cheng X. Identification of CD66c as a potential target in gastroesophageal junction cancer for antibody-drug conjugate development. Gastric Cancer 2025; 28:422-441. [PMID: 39918687 PMCID: PMC11993476 DOI: 10.1007/s10120-025-01584-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 12/27/2024] [Indexed: 04/13/2025]
Abstract
BACKGROUND Gastroesophageal junction (GEJ) cancer exhibits unique biological characteristics and currently lacks specific targeted therapies. Given the clinical efficacy of antibody-drug conjugates (ADCs) in solid tumor treatment, we aimed to identify a novel ADC target and suitable payload for GEJ-targeted therapy. METHODS In this study, we conducted bioinformatic analyses of multi-omics data, including transcriptomics, proteomics, and phosphoproteomics, to identify CD66c as a promising ADC target for GEJ cancer. We then engineered a CD66c-directed antibody-drug conjugate (CD66c-DXd) incorporating a GGFG linker. The preclinical efficacy of CD66c-DXd was determined in multi GEJ xenograft models. RESULTS Proteomic analyses of 103 cases of GEJ cancer revealed that CD66c expression was significantly higher in tumoral tissues compared to normal tissues. Proteomic and phosphoproteomic analyses identified deruxtecan (DXd) as a potentially potent payload for ADCs targeting GEJ cancer. Furthermore, high CD66c expression in GEJ was associated with a significantly lower proportion of plasma cells. The drug-to-antibody ratio (DAR) of CD66c-DXd was determined to be 3.6. CD66c-DXd effectively and selectively ablated multiple human GEJ cell lines (OE-19, OE33 and SK-GT-4) without affecting non-malignant cells (GES-1) in vitro. Eventually, CD66c-DXd mediated potent and durable tumor regression in vivo with excellent safety profiles. CONCLUSIONS This preclinical study provides a strong rationale for the further development of CD66c-DXd as promising therapeutic candidates to treat advanced GEJ cancer. Additionally, the study demonstrates the robustness of the multi-omics data in identifying novel potential ADC targets and payloads.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Department of Medical Oncology, Zhejiang Provincial People's Hospital, Hangzhou, 310022, Zhejiang, China
| | - Changjuan Tao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Hanfei Xie
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Postgraduate Training Base Alliance of Wenzhou Medical University, Hangzhou, 310022, Zhejiang, China
| | - Liu Yang
- Department of Medical Oncology, Zhejiang Provincial People's Hospital, Hangzhou, 310022, Zhejiang, China
| | - Ye Lu
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Yun Xi
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, China
| | - Shili Yao
- School of Materials Science and Engineering, Tianjin University, Tianjin, 300072, China
| | - Li Yuan
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Peng Guo
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China.
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China.
- Zhejiang Key Lab of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Zhejiang Cancer Hospital, Hangzhou, China.
| |
Collapse
|
3
|
Mu D, Chen B, Liu X, Zheng S, Zhang Y, Ni H, Zhou D. Exploring the potential mechanisms of Da ChaiHu decoction against pancreatic cancer based on network pharmacology prediction and molecular docking approach. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04107-w. [PMID: 40266298 DOI: 10.1007/s00210-025-04107-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 03/25/2025] [Indexed: 04/24/2025]
Abstract
Da ChaiHu decoction (DCHD) is used in Chinese medicine to treat pancreatic cancer (PC), but its exact mechanism is not known. The aim of this study was to investigate the main active ingredients and specific mechanisms of DCHD against PC. Firstly, the active ingredients and targets of DCHD and PC-related targets were searched from the TCMSP, DrugBank, NCBI and GeneCards databases, respectively. The intersected targets of both were then taken to construct a PPI network using STRING, and this network was visualized by Cytoscape 3.8.2. GO and KEGG enrichment analyses of the intersected targets were performed using R 4.2.1 "clusterProfiler", "enrichplot", and "ggplot2" packages. Molecular docking was performed utilizing MOE software to detect the binding capacity between compounds and targets. Cell proliferation, apoptosis, invasion and migration were examined through a CCK8 kit, Muse® Cell Analyzer, transwell and wound healing experiment, respectively. The expression levels of five core targets were assessed by RT-qPCR in PANC-1 cells treated with stigmasterol. Molecular dynamic simulations analysis was conducted to analyze the binding affinities and modes of interaction between molecules and stigmasterol using the GROMACS 5.1.4 program package. In this study, 141 common targets of DCHD and PC were obtained. GO-MF items indicated that DCHD exerts its effects on PC primarily by influencing the binding activity of DNA-binding transcription factors. The KEGG analysis revealed that these genes were implicated in various signaling pathways, including the IL-17 signaling pathway and the PI3K/Akt signaling pathway. Stigmasterol was chosen as the final ingredient for subsequent investigation due to its derivation from herb (Da ChaiHu), its encompassment of more common targets, and the scarcity of existing research on its role in PC. The results of molecular docking and Molecular dynamic simulations analysis showed that stigmasterol had good binding activity with BCL2, and ICAM1. In vitro experiments suggested that stigmasterol could effectively inhibit the proliferation, invasion and migration of PANC-1 cells, and promote cell apoptosis. Moreover, stigmasterol treatment led to the reduced expression of AKT1, HIF1A, BCL2, IL1B, and ICAM1. This study is the first to reveal the main active components and potential mechanisms of DCHD against PC, which provides a theoretical basis for studying the role of DCHD in the treatment of PC. Especially, the anti-PC mechanism of active compound stigmasterol might be associated with inhibiting proliferation, invasion and migration and accelerating apoptosis. Furthermore, five targets (AKT1, HIF1A, BCL2, IL1B, and ICAM1) were identified as key targets of stigmasterol, and the mRNA expressions of these genes were down-regulated by stigmasterol through in vitro experiments.
Collapse
Affiliation(s)
- Dong Mu
- Department of Gastroenterology and Hepatology, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Beijin Chen
- Department of Gastroenterology and Hepatology, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Xiaoli Liu
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, China
| | - Shumei Zheng
- Department of Gastroenterology and Hepatology, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Yong Zhang
- Department of Gastroenterology and Hepatology, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Hua Ni
- Department of Gastroenterology and Hepatology, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Dejiang Zhou
- Department of Gastroenterology and Hepatology, The General Hospital of Western Theater Command, Chengdu, 610083, China.
- , Chengdu, China.
| |
Collapse
|
4
|
Zhou K, Liu Y, Tang C, Zhu H. Pancreatic Cancer: Pathogenesis and Clinical Studies. MedComm (Beijing) 2025; 6:e70162. [PMID: 40182139 PMCID: PMC11965705 DOI: 10.1002/mco2.70162] [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: 06/24/2024] [Revised: 03/08/2025] [Accepted: 03/12/2025] [Indexed: 04/05/2025] Open
Abstract
Pancreatic cancer (PC) is a highly lethal malignancy, with pancreatic ductal adenocarcinoma (PDAC) being the most common and aggressive subtype, characterized by late diagnosis, aggressive progression, and resistance to conventional therapies. Despite advances in understanding its pathogenesis, including the identification of common genetic mutations (e.g., KRAS, TP53, CDKN2A, SMAD4) and dysregulated signaling pathways (e.g., KRAS-MAPK, PI3K-AKT, and TGF-β pathways), effective therapeutic strategies remain limited. Current treatment modalities including chemotherapy, targeted therapy, immunotherapy, radiotherapy, and emerging therapies such as antibody-drug conjugates (ADCs), chimeric antigen receptor T (CAR-T) cells, oncolytic viruses (OVs), cancer vaccines, and bispecific antibodies (BsAbs), face significant challenges. This review comprehensively summarizes these treatment approaches, emphasizing their mechanisms, limitations, and potential solutions, to overcome these bottlenecks. By integrating recent advancements and outlining critical challenges, this review aims to provide insights into future directions and guide the development of more effective treatment strategies for PC, with a specific focus on PDAC. Our work underscores the urgency of addressing the unmet needs in PDAC therapy and highlights promising areas for innovation in this field.
Collapse
Affiliation(s)
- Kexun Zhou
- Department of Medical OncologyCancer CenterWest China HospitalSichuan UniversityChengduChina
| | - Yingping Liu
- Department of RadiotherapyCancer HospitalChinese Academy of Medical SciencesBeijingChina
| | - Chuanyun Tang
- The First Clinical Medical College of Nanchang UniversityNanchang UniversityNanchangChina
| | - Hong Zhu
- Department of Medical OncologyCancer CenterWest China HospitalSichuan UniversityChengduChina
- Division of Abdominal Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
5
|
Mathiyazhagan J, Rajesh C, Sagar S, Caffrey TC, Huang Y, Mohs AM, Swanson BJ, Hollingsworth MA, Brooks CL, Radhakrishnan P. Humanized Anti-MUC16 Antibody-Conjugated Contrast Agents for Magnetic Resonance Imaging of Pancreatic Cancer. Cancers (Basel) 2025; 17:957. [PMID: 40149293 PMCID: PMC11940418 DOI: 10.3390/cancers17060957] [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: 02/06/2025] [Revised: 03/05/2025] [Accepted: 03/08/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Pancreatic ductal adenocarcinoma (PDAC) is diagnosed at a late stage with distant metastasis in an overwhelming 50% of cases, and the prognosis is poor. Treating this extremely aggressive disease with standard-of-care therapies has led to modest benefits in overall survival, mainly due to a lack of targeted early treatment modalities, as early detection has not yet been possible. Mucin-16 (MUC16) is a glycoprotein overexpressed in more than 60% of patients with PDAC and is a tumor-specific biomarker. Methods: In this study, a magnetic resonance imaging (MRI) probe to facilitate the detection of early and late lesions of PDAC is developed by conjugating a MUC16-targeted humanized antibody (huAR9.6) with gadolinium. Results: In preclinical mouse models, this MUC16-targeted MRI probe demonstrates effective contrast enhancement in early lesions of PDAC in the subcutaneous setting and allows for the detection of late-stage pancreatic cancer tumors in an orthotopic model. The probe did not induce any toxicity in vital organs at the administered doses. Conclusions: This study establishes that synthesizing a MUC16-targeted MRI probe is feasible and allows for the better high-resolution contrast enhancement of MUC16+ PDAC lesions to facilitate detection and possibly better treatment strategies.
Collapse
Affiliation(s)
- Jayasindu Mathiyazhagan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Christabelle Rajesh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Satish Sagar
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Thomas C. Caffrey
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ying Huang
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Aaron M. Mohs
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Benjamin J. Swanson
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Cory L. Brooks
- Department of Chemistry and Biochemistry, California State University Fresno, Fresno, CA 93740, USA
| | - Prakash Radhakrishnan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
6
|
Yao S, Sun L, Lu Y, Zhu X, Xu R, Yang T, Tang H, Guo P, Zhu T. Eliminating VEGFA+ tumor-associated neutrophils by antibody-drug conjugates boosts antitumor immunity and potentiates PD-1 immunotherapy in preclinical models of cervical cancer. Cell Death Dis 2025; 16:115. [PMID: 39971940 PMCID: PMC11840153 DOI: 10.1038/s41419-025-07402-9] [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: 08/28/2024] [Revised: 01/08/2025] [Accepted: 01/27/2025] [Indexed: 02/21/2025]
Abstract
Tumor-associated neutrophils (TANs) actively interact with antibody-drug conjugates (ADCs) within the tumor microenvironment (TME), though the detailed mechanisms governing their response to ADC treatment remain to be fully elucidated. Herein, we explored how ICAM1-targeted ADCs affect TAN dynamics in preclinical models of cervical cancer. We discovered that I-DXd, our in-house ADC targeting cervical cancer, effectively eliminates tumor cells through specific antigen recognition while concurrently depleting pro-tumor VEGFA + TANs via Fcγ receptor-mediated phagocytosis. This dual action promotes an immunologically favorable TME. Through comprehensive preclinical studies, we established a foundational understanding of the synergistic benefits of combining I-DXd treatment with PD-1 immune checkpoint inhibition, thereby opening new avenues for therapeutic intervention in advanced cervical cancer.
Collapse
Affiliation(s)
- Shili Yao
- School of Materials Science and Engineering, Faculty of Medicine, Tianjin University, Tianjin, China
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
| | - Lu Sun
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ye Lu
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
| | - Xiu Zhu
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Rui Xu
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
- Institute of Molecular Medicine, Hangzhou Institute for Advanced Study (UCAS), Hangzhou, China
| | - Tong Yang
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China
| | - Huarong Tang
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China.
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, China.
- Department of Gynecological Radiotherapy, Zhejiang Cancer Hospital, Hangzhou, China.
| | - Peng Guo
- School of Materials Science and Engineering, Faculty of Medicine, Tianjin University, Tianjin, China.
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China.
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, China.
| | - Tao Zhu
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, China.
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, China.
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, China.
| |
Collapse
|
7
|
Liu Y, Wang X, Zhang N, He S, Zhang J, Xu X, Song S. Utility of 131I-HLX58-Der for the Precision Treatment: Evaluation of a Preclinical Radio-Antibody-Drug-Conjugate Approach in Mouse Models. Int J Nanomedicine 2025; 20:723-739. [PMID: 39839455 PMCID: PMC11748935 DOI: 10.2147/ijn.s501689] [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: 10/20/2024] [Accepted: 01/09/2025] [Indexed: 01/23/2025] Open
Abstract
Purpose None of the antibody-drug conjugates (ADCs) targeting Claudin 18.2 (CLDN18.2) have received approval from regulatory authorities due to their limited clinical benefits. Leveraging the radiosensitizing ability of Deruxtecan (DXd) and the internal radiation therapy of 131I for tumors, we aimed to develop the first radio-antibody-drug conjugates (RADCs) for the treatment of gastric cancer. Methods The CLDN18.2-specific antibody HLX58 was conjugated with the payload DXd through a cleavable maleimide glycynglycyn-phenylalanyn-glycyn (GGFG) peptide linker. HLX58-Der was labeled with 131I to produce RADC-131I-HLX58-Der. HLX58 was labeled with 125I for imaging CLDN18.2-positive tumors, providing a reference for RADC treatment in solid tumors. The antigen-binding properties and biodistribution of the RADC were studied both in vitro and in vivo. The cytotoxic effects of the RADC were evaluated in CLDN18.2-positive tumor cell lines and xenografts. Results HLX58 was successfully conjugated with DXd using the cleavable maleimide GGFG peptide linker and labeled with 131I to produce RADC-131I-HLX58-Der. HLX58 was labeled with 125I for imaging CLDN18.2-positive tumors. Both 125I-HLX58 and 131I-HLX58-Der exhibited significant binding affinity for the CLDN18.2-positive cancer cell line. The cytotoxic effect of 131I-HLX58-Der was observed in the CLDN18.2-positive cell line, with an IC50 of 11.28 ng/mL. In terms of cytotoxicity, 131I-HLX58-Der exhibited greater activity compared to HLX58-Der. 125I-HLX58 and 131I-HLX58-Der demonstrated similar biodistribution profiles in CLDN18.2-positive tumor models, achieving 5.72 ± 0.41%ID/g (48 h) and 5.83 ± 0.41%ID/g (72 h) in the tumor tissues postinjection, respectively. The average tumor size in groups treated with 131I-HLX58-Der and HLX58-Der was reduced by factors of 12.15 and 4.80, respectively, compared to the control group. 131I-HLX58-Der demonstrated no toxic effects on hepatorenal function, routine blood tests, or major organs in mice when compared to the control group. Conclusion These findings validate the potential of RADCs targeting CLDN18.2 in treating CLDN18.2-expressing solid tumors.
Collapse
Affiliation(s)
- Yi Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Center for Biomedical Imaging, Fudan University, Shanghai, 200032, People’s Republic of China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032, People’s Republic of China
| | - Xiao Wang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Center for Biomedical Imaging, Fudan University, Shanghai, 200032, People’s Republic of China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen, 361102, People’s Republic of China
| | - Ni Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Center for Biomedical Imaging, Fudan University, Shanghai, 200032, People’s Republic of China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032, People’s Republic of China
| | - Simin He
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Center for Biomedical Imaging, Fudan University, Shanghai, 200032, People’s Republic of China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032, People’s Republic of China
| | - Jianping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Center for Biomedical Imaging, Fudan University, Shanghai, 200032, People’s Republic of China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032, People’s Republic of China
| | - Xiaoping Xu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Center for Biomedical Imaging, Fudan University, Shanghai, 200032, People’s Republic of China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032, People’s Republic of China
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, People’s Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People’s Republic of China
- Center for Biomedical Imaging, Fudan University, Shanghai, 200032, People’s Republic of China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, 200032, People’s Republic of China
| |
Collapse
|
8
|
Qian WJ, Yan JS, Gang XY, Xu L, Shi S, Li X, Na FJ, Cai LT, Li HM, Zhao MF. Intercellular adhesion molecule-1 (ICAM-1): From molecular functions to clinical applications in cancer investigation. Biochim Biophys Acta Rev Cancer 2024; 1879:189187. [PMID: 39317271 DOI: 10.1016/j.bbcan.2024.189187] [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: 03/18/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
Intercellular adhesion molecule-1 (ICAM-1) is a versatile molecule that plays a critical role in various physiological and pathological processes, particularly in tumor development where its impact is bidirectional. On the one hand, it augments the immune response by promoting immune cell migration, infiltration, and the formation of immunological synapses, thus facilitating potent antitumor effects. Simultaneously, it contributes to tumor immune evasion and influences metastasis by mediating transendothelial migration (TEM), epithelial-to-mesenchymal transition (EMT), and epigenetic modification of tumor cells. Despite its significant potential, the full clinical utility of ICAM-1 has yet to be fully realized. In this review, we thoroughly examine recent advancements in understanding the role of ICAM-1 in tumor development, its relevance in predicting therapeutic efficacy and prognosis, as well as the progress in clinical translational research on anti-ICAM-1-based therapies, encompassing including monoclonal antibodies, immunotherapy, antibody-drug conjugate (ADC), and conventional treatments. By shedding light on these innovative strategies, we aim to underscore ICAM-1's significance as a valuable and multifaceted target for cancer treatment, igniting enthusiasm for further research and facilitating translation into clinical applications.
Collapse
Affiliation(s)
- Wen-Jing Qian
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jin-Shan Yan
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Xiao-Yu Gang
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Lu Xu
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Sha Shi
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Xin Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Fang-Jian Na
- Network Information Center, China Medical University, Shenyang, China
| | - Lu-Tong Cai
- Psychological Medicine, Shenyang Medical College, Shenyang, China
| | - He-Ming Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China; Guangdong Association of Clinical Trials (GACT)/Chinese Thoracic Oncology Group (CTONG) and Guangdong Provincial Key Lab of Translational Medicine in Lung Cancer, Guangzhou, China.
| | - Ming-Fang Zhao
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
9
|
Alexander S, Aleem U, Jacobs T, Frizziero M, Foy V, Hubner RA, McNamara MG. Antibody-Drug Conjugates and Their Potential in the Treatment of Patients with Biliary Tract Cancer. Cancers (Basel) 2024; 16:3345. [PMID: 39409965 PMCID: PMC11476249 DOI: 10.3390/cancers16193345] [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: 08/31/2024] [Revised: 09/16/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Biliary tract cancers (BTCs) are aggressive in nature, often presenting asymptomatically until they are diagnosed at an advanced stage. Surgical resection or liver transplantation are potential curative options. However, a large proportion of patients present with incurable locally advanced or metastatic disease and most of these patients are only eligible for palliative chemotherapy or best supportive care. More recently, targeted therapies have proven beneficial in a molecularly selected subgroup of patients with cholangiocarcinoma who have progressed on previous lines of systemic treatment. However, only a minority of patients with BTCs whose tumours harbour specific molecular alterations can access these therapies. Methods: In relation to ADCs, studies regarding use of antibody-drug conjugates in cancer, particularly in BTCs, were searched in Embase (1974 to 2024) and Ovid MEDLINE(R) (1946 to 2024) to obtain relevant articles. Examples of current clinical trials utilising ADC treatment in BTCs were extracted from the ClinicalTrials.gov trial registry. Conclusions: Overall, this review has highlighted that ADCs have shown encouraging outcomes in cancer therapy, and this should lead to further research including in BTCs, where treatment options are often limited. The promising results observed with ADCs in various cancers underscore their potential as a transformative approach in oncology, warranting continued exploration and development and the need for education on the management of their specific toxicities. By addressing current challenges and optimising ADC design and application, future studies could potentially improve treatment outcomes for patients with BTCs and beyond, potentially in both early and advanced stage settings.
Collapse
Affiliation(s)
- Shaun Alexander
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK; (U.A.); (M.F.); (V.F.); (R.A.H.)
| | - Umair Aleem
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK; (U.A.); (M.F.); (V.F.); (R.A.H.)
| | - Timothy Jacobs
- The Library, The Christie NHS Foundation Trust, Manchester M20 4BX, UK;
| | - Melissa Frizziero
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK; (U.A.); (M.F.); (V.F.); (R.A.H.)
| | - Victoria Foy
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK; (U.A.); (M.F.); (V.F.); (R.A.H.)
| | - Richard A. Hubner
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK; (U.A.); (M.F.); (V.F.); (R.A.H.)
| | - Mairéad G. McNamara
- Division of Cancer Sciences, School of Medical Sciences, University of Manchester, The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| |
Collapse
|
10
|
Zhang P, Tao C, Lu Y, Li P, Wang X, Dai Y, Xi Y, Shimura T, Li X, Fang J, Yang L, He D, Guo P. Epigenetic Reprogramming Potentiates ICAM1 Antibody Drug Conjugates in Preclinical Models of Melanoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400203. [PMID: 38874532 PMCID: PMC11321650 DOI: 10.1002/advs.202400203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 05/15/2024] [Indexed: 06/15/2024]
Abstract
Therapeutic benefits and underlying biomechanism(s) of antibody drug conjugates (ADC) in combination with other targeted therapeutics are largely unknown. Here, the synergy between ADC and epigenetic drug decitabine (DAC), a clinically approved DNA methylation inhibitor, in multiple preclinical models of melanoma specifically investigated. Mechanistically, the underlying biomechanisms of how DAC cooperatively worked with ICAM1 antibody conjugated DNA topoisomerase I inhibitor DXd (I1-DXd) is elucidated. DAC treatment significantly enhanced anti-tumor efficacy of I1-DXd by upregulating antigen expression, enhancing antibody internalization and potentiating tumor sensitivity by epigenetically reprogramming of melanoma. Meanwhile, I1-DXd/DAC combination also exerted regulatory effects on tumor microenvironment (TME) by enhancing tumor infiltration of innate and adaptive immune cells and improving penetration of ADCs with a boosted antitumor immunity. This study provides a rational ADC combination strategy for solid tumor treatment.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Medical OncologyZhejiang Provincial People's HospitalHangzhouZhejiang310022China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang ProvinceZhejiang Provincial People's HospitalPeople's Hospital of Hangzhou Medical CollegeHangzhouZhejiang310014China
- Hangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
| | - Changjuan Tao
- Department of Radiation OncologyThe Cancer Hospital of the University of Chinese Academy of SciencesZhejiang Cancer HospitalHangzhouZhejiang310022China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang ProvinceZhejiang Cancer HospitalHangzhouZhejiang310022China
| | - Ye Lu
- Hangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
| | - Peijing Li
- Department of Radiation OncologyThe Cancer Hospital of the University of Chinese Academy of SciencesZhejiang Cancer HospitalHangzhouZhejiang310022China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang ProvinceZhejiang Cancer HospitalHangzhouZhejiang310022China
| | - Xing Wang
- Department of Head and Neck SurgeryThe Cancer Hospital of the University of Chinese Academy of SciencesZhejiang Cancer HospitalHangzhouZhejiang310022China
| | - Yujie Dai
- MabPlex InternationalYantaiShandong264006China
| | - Yun Xi
- Department of PathologyThe Cancer Hospital of the University of Chinese Academy of SciencesZhejiang Cancer HospitalHangzhouZhejiang310022China
| | - Takaya Shimura
- Department of Gastroenterology and MetabolismNagoya City University Graduate School of Medical SciencesNagoya467–8601Japan
| | - Xinfang Li
- MabPlex InternationalYantaiShandong264006China
| | - Jianmin Fang
- School of Materials Science and EngineeringTianjin UniversityTianjin300072China
| | - Liu Yang
- Department of Medical OncologyZhejiang Provincial People's HospitalHangzhouZhejiang310022China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang ProvinceZhejiang Provincial People's HospitalPeople's Hospital of Hangzhou Medical CollegeHangzhouZhejiang310014China
| | - Dawei He
- Department of UrologyChildren's Hospital of Chongqing Medical UniversityChongqingChina
| | - Peng Guo
- Hangzhou Institute of Medicine (HIM)Chinese Academy of SciencesHangzhouZhejiang310022China
| |
Collapse
|
11
|
Wittwer NL, Brown MP, Liapis V, Staudacher AH. Antibody drug conjugates: hitting the mark in pancreatic cancer? J Exp Clin Cancer Res 2023; 42:280. [PMID: 37880707 PMCID: PMC10598980 DOI: 10.1186/s13046-023-02868-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/16/2023] [Indexed: 10/27/2023] Open
Abstract
Pancreatic cancer is one of the most common causes of cancer-related death, and the 5-year survival rate has only improved marginally over the last decade. Late detection of the disease means that in most cases the disease has advanced locally and/or metastasized, and curative surgery is not possible. Chemotherapy is still the first-line treatment however, this has only had a modest impact in improving survival, with associated toxicities. Therefore, there is an urgent need for targeted approaches to better treat pancreatic cancer, while minimizing treatment-induced side-effects. Antibody drug conjugates (ADCs) are one treatment option that could fill this gap. Here, a monoclonal antibody is used to deliver extremely potent drugs directly to the tumor site to improve on-target killing while reducing off-target toxicity. In this paper, we review the current literature for ADC targets that have been examined in vivo for treating pancreatic cancer, summarize current and on-going clinical trials using ADCs to treat pancreatic cancer and discuss potential strategies to improve their therapeutic window.
Collapse
Affiliation(s)
- Nicole L Wittwer
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology, University of South Australia, Adelaide, SA, 5000, Australia.
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia.
| | - Michael P Brown
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology, University of South Australia, Adelaide, SA, 5000, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
| | - Vasilios Liapis
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology, University of South Australia, Adelaide, SA, 5000, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Alexander H Staudacher
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology, University of South Australia, Adelaide, SA, 5000, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
| |
Collapse
|
12
|
Zhu B, Wang X, Shimura T, Huang AC, Kong N, Dai Y, Fang J, Guo P, Ying JE. Development of potent antibody drug conjugates against ICAM1 + cancer cells in preclinical models of cholangiocarcinoma. NPJ Precis Oncol 2023; 7:93. [PMID: 37717087 PMCID: PMC10505223 DOI: 10.1038/s41698-023-00447-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 09/05/2023] [Indexed: 09/18/2023] Open
Abstract
As a highly lethal adenocarcinoma of the hepatobiliary system, outcomes for cholangiocarcinoma (CCA) patients remain prominently poor with a 5-year survival of <10% due to the lack of effective treatment modalities. Targeted therapeutics for CCA are limited and surgical resection of CCA frequently suffers from a high recurrence rate. Here we report two effective targeted therapeutics in this preclinical study for CCA. We first performed a quantitative and unbiased screening of cancer-related antigens using comparative flow cytometry in a panel of human CCA cell lines, and identified intercellular adhesion molecule-1 (ICAM1) as a therapeutic target for CCA. After determining that ICAM1 has the ability to efficiently mediate antibody internalization, we constructed two ICAM1 antibody-drug conjugates (ADCs) by conjugating ICAM1 antibodies to different cytotoxic payloads through cleavable chemical linkers. The efficacies of two ICAM1 ADCs have been evaluated in comparison with the first-line chemodrug Gemcitabine in vitro and in vivo, and ICAM1 antibodies coupled with warhead DX-8951 derivative (DXd) or monomethyl auristatin E (MMAE) elicit a potent and consistent tumor attenuation. In summary, this study paves the road for developing a promising targeted therapeutic candidate for clinical treatment of CCA.
Collapse
Affiliation(s)
- Bing Zhu
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310018, China
| | - Xinyan Wang
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310018, China
- Institute of Molecular Medicine, Hangzhou Institute for Advanced Study (UCAS), Hangzhou, Zhejiang, 310000, China
| | - Takaya Shimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | | | - Nana Kong
- MabPlex International, Yantai, Shandong, 264006, China
| | - Yujie Dai
- MabPlex International, Yantai, Shandong, 264006, China
| | - Jianmin Fang
- School of Life Science and Technology, Tongji University, Shanghai, 200092, China
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Peng Guo
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China.
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310018, China.
| | - Jie-Er Ying
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
13
|
Zhang P, Tao C, Shimura T, Huang AC, Kong N, Dai Y, Yao S, Xi Y, Wang X, Fang J, Moses MA, Guo P. ICAM1 antibody drug conjugates exert potent antitumor activity in papillary and anaplastic thyroid carcinoma. iScience 2023; 26:107272. [PMID: 37520726 PMCID: PMC10371847 DOI: 10.1016/j.isci.2023.107272] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/27/2023] [Accepted: 06/28/2023] [Indexed: 08/01/2023] Open
Abstract
Treatment options for anaplastic thyroid cancer (ATC) and refractory papillary thyroid carcinoma (PTC) are limited and outcomes remain poor. In this study, we determined via bioinformatic expression analyses and immunohistochemistry staining that intercellular adhesion molecule-1(ICAM1) is an attractive target for ATC and PTC. We designed and engineered two ICAM1-directed antibody-drug conjugate (I1-MMAE and I1-DXd), both of which potently and selectively ablate multiple human ATC and PTC cell lines without affecting non-plastic cells in vitro. Furthermore, I1-MMAE and I1-DXd mediated a potent tumor regression in ATC and PTC xenograft models. To develop a precision medicine, we also explored magnetic resonance imaging (MRI) as a non-invasive biomarker detection method to quantitatively map ICAM1 antigen expression in heterogeneous thyroid tumors. Taken together, this study provides a strong rationale for the further development of I1-MMAE and I1-DXd as promising therapeutic candidates to treat advanced PTC and ATC.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Medical Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Changjuan Tao
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Department of Radiation Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Takaya Shimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | | | - Nana Kong
- MabPlex International, Yantai, Shandong 264006, China
| | - Yujie Dai
- MabPlex International, Yantai, Shandong 264006, China
| | - Shili Yao
- School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Yun Xi
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Xing Wang
- Department of Head and Neck Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
| | - Jianmin Fang
- School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Marsha A. Moses
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Peng Guo
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
14
|
Wang Y, Huang N, Yang Z. Revealing the Role of Zinc Ions in Atherosclerosis Therapy via an Engineered Three-Dimensional Pathological Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300475. [PMID: 37092571 PMCID: PMC10288231 DOI: 10.1002/advs.202300475] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/09/2023] [Indexed: 05/03/2023]
Abstract
An incomplete understanding of the cellular functions and underlying mechanisms of zinc ions released from zinc-based stents in atherosclerosis (AS) therapy is one of the major obstacles to their clinical translation. The existing evaluation methodology using cell monolayers has limitations on accurate results due to the lack of vascular architectures and pathological features. Herein, the authors propose a 3D biomimetic AS model based on a multi-layer vascular structure comprising endothelial cells and smooth muscle cells with hyperlipidemic surroundings and inflammatory stimulations as AS-prone biochemical conditions to explore the biological functions of zinc ions in AS therapy. Concentration-dependent biphasic effects of zinc ions on cell growth are observed both in cell monolayers and 3D AS models. Nevertheless, the cells within 3D AS model exhibit more accurate biological assessments of the zinc ions, as evidenced by augmented pathological features and significantly higher half-maximal inhibitory concentration values against zinc ions. Based on such a developed 3D biomimetic AS model, the inhibitory effects on the deoxyribonucleic acid (DNA) synthesis, significantly influenced biological processes like cell motility, proliferation, and adhesion, and several potential bio-targets of zinc ions of cells are revealed.
Collapse
Affiliation(s)
- Ying Wang
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative MedicineThe Tenth Affiliated Hospital of Southern Medical UniversityDongguan523059P. R. China
- Guangdong Provincial Key Laboratory of Cardiac Function and MicrocirculationGuangzhou510080P. R. China
| | - Nan Huang
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative MedicineThe Tenth Affiliated Hospital of Southern Medical UniversityDongguan523059P. R. China
| | - Zhilu Yang
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative MedicineThe Tenth Affiliated Hospital of Southern Medical UniversityDongguan523059P. R. China
- Guangdong Provincial Key Laboratory of Cardiac Function and MicrocirculationGuangzhou510080P. R. China
- Department of CardiologyThird People's Hospital of Chengdu Affiliated to Southwest Jiaotong UniversityChengdu610031P. R. China
| |
Collapse
|
15
|
Guo P, Huang J, Zhu B, Huang AC, Jiang L, Fang J, Moses MA. A rationally designed ICAM1 antibody drug conjugate eradicates late-stage and refractory triple-negative breast tumors in vivo. SCIENCE ADVANCES 2023; 9:eabq7866. [PMID: 37146146 PMCID: PMC10162665 DOI: 10.1126/sciadv.abq7866] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Triple-negative breast cancer (TNBC) remains the most lethal form of breast cancer, and effective targeted therapeutics are in urgent need to improve the poor prognosis of TNBC patients. Here, we report the development of a rationally designed antibody drug conjugate (ADC) for the treatment of late-stage and refractory TNBC. We determined that intercellular adhesion molecule-1 (ICAM1), a cell surface receptor overexpressed in TNBC, efficiently facilitates receptor-mediated antibody internalization. We next constructed a panel of four ICAM1 ADCs using different chemical linkers and warheads and compared their in vitro and in vivo efficacies against multiple human TNBC cell lines and a series of standard, late-stage, and refractory TNBC in vivo models. An ICAM1 antibody conjugated with monomethyl auristatin E (MMAE) via a protease-cleavable valine-citrulline linker was identified as the optimal ADC formulation owing to its outstanding efficacy and safety, representing an effective ADC candidate for TNBC therapy.
Collapse
Affiliation(s)
- Peng Guo
- Vascular Biology Program, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Jing Huang
- Vascular Biology Program, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Bing Zhu
- Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | | | - Lingxiao Jiang
- School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Jianmin Fang
- School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Marsha A Moses
- Vascular Biology Program, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
16
|
Olajubutu O, Ogundipe OD, Adebayo A, Adesina SK. Drug Delivery Strategies for the Treatment of Pancreatic Cancer. Pharmaceutics 2023; 15:pharmaceutics15051318. [PMID: 37242560 DOI: 10.3390/pharmaceutics15051318] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/15/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Pancreatic cancer is fast becoming a global menace and it is projected to be the second leading cause of cancer-related death by 2030. Pancreatic adenocarcinomas, which develop in the pancreas' exocrine region, are the predominant type of pancreatic cancer, representing about 95% of total pancreatic tumors. The malignancy progresses asymptomatically, making early diagnosis difficult. It is characterized by excessive production of fibrotic stroma known as desmoplasia, which aids tumor growth and metastatic spread by remodeling the extracellular matrix and releasing tumor growth factors. For decades, immense efforts have been harnessed toward developing more effective drug delivery systems for pancreatic cancer treatment leveraging nanotechnology, immunotherapy, drug conjugates, and combinations of these approaches. However, despite the reported preclinical success of these approaches, no substantial progress has been made clinically and the prognosis for pancreatic cancer is worsening. This review provides insights into challenges associated with the delivery of therapeutics for pancreatic cancer treatment and discusses drug delivery strategies to minimize adverse effects associated with current chemotherapy options and to improve the efficiency of drug treatment.
Collapse
Affiliation(s)
| | - Omotola D Ogundipe
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, USA
| | - Amusa Adebayo
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, USA
| | - Simeon K Adesina
- Department of Pharmaceutical Sciences, Howard University, Washington, DC 20059, USA
| |
Collapse
|
17
|
Zettl I, Ivanova T, Zghaebi M, Rutovskaya MV, Ellinger I, Goryainova O, Kollárová J, Villazala-Merino S, Lupinek C, Weichwald C, Drescher A, Eckl-Dorna J, Tillib SV, Flicker S. Generation of high affinity ICAM-1-specific nanobodies and evaluation of their suitability for allergy treatment. Front Immunol 2022; 13:1022418. [DOI: 10.3389/fimmu.2022.1022418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
The nasal cavity is an important site of allergen entry. Hence, it represents an organ where trans-epithelial allergen penetration and subsequent IgE-mediated allergic inflammation can potentially be inhibited. Intercellular adhesion molecule 1 (ICAM-1) is highly expressed on the surface of respiratory epithelial cells in allergic patients. It was identified as a promising target to immobilize antibody conjugates bispecific for ICAM-1 and allergens and thereby block allergen entry. We have previously characterized a nanobody specific for the major birch pollen allergen Bet v 1 and here we report the generation and characterization of ICAM-1-specific nanobodies. Nanobodies were obtained from a camel immunized with ICAM-1 and a high affinity binder was selected after phage display (Nb44). Nb44 was expressed as recombinant protein containing HA- and His-tags in Escherichia coli (E.coli) and purified via affinity chromatography. SDS-PAGE and Western blot revealed a single band at approximately 20 kDa. Nb44 bound to recombinant ICAM-1 in ELISA, and to ICAM-1 expressed on the human bronchial epithelial cell line 16HBE14o- as determined by flow cytometry. Experiments conducted at 4°C and at 37°C, to mimic physiological conditions, yielded similar percentages (97.2 ± 1.2% and 96.7 ± 1.5% out of total live cells). To confirm and visualize binding, we performed immunofluorescence microscopy. While Texas Red Dextran was rapidly internalized Nb44 remained localized on the cell surface. Additionally, we determined the strength of Nb44 and ICAM-1 interaction using surface plasmon resonance (SPR). Nb44 bound ICAM-1 with high affinity (10-10 M) and had slow off-rates (10-4 s-1). In conclusion, our results showed that the selected ICAM-1-specific nanobody bound ICAM-1 with high affinity and was not internalized. Thus, it could be further used to engineer heterodimers with allergen-specific nanobodies in order to develop topical treatments of pollen allergy.
Collapse
|
18
|
Zhu M, Zhou L, Hu S, Miao Q, Gong J, Zhang N, Zhang G, Wang M, Wang J, He H, Wang Y. Rational Design and Systemic Appraisal of an EGFR-Targeting Antibody-Drug Conjugate LR-DM1 for Pancreatic Cancer. J Med Chem 2022; 65:7141-7153. [PMID: 35522590 DOI: 10.1021/acs.jmedchem.1c01920] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
By harnessing the payload DM1 and a monoclonal antibody LR004 through a noncleavable linker succinimidyl-4-(N-maleimidomethyl)-cyclohexane-1-carboxylate, we designed and evaluated an antibody-drug conjugate LR-DM1 with an appropriate drug-antibody ratio of 3.6. LR-DM1, which was targeted toward the epidermal growth factor receptor for pancreatic cancer, exhibited potent antiproliferation activity in vitro with a half-maximal inhibitory concentration value of 7.03 nM for Capan-2 cells. Particularly, it displayed prominent tumor growth inhibition in vivo under 20 mg/kg LR-DM1 dosage in a single administration or multiple administrations without apparent abnormality of pathological observation. Moreover, LR-DM1 possessed a relatively broad therapeutic index with a half-lethal dose above 300 mg/kg, which was over 15-fold higher than the highest administration dosage of 20 mg/kg. This initial study on LR-DM1 holds promise for further development of a new antibody drug conjugate that is transformative for treatment of patients concerned.
Collapse
Affiliation(s)
- Mei Zhu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Lei Zhou
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Shangjiu Hu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Qingfang Miao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Jianhua Gong
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Na Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Guoning Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Minghua Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Juxian Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Hongwei He
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Yucheng Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
19
|
Yang Y, McCloskey JE, Yang H, Puc J, Alcaina Y, Vedvyas Y, Gomez Gallegos AA, Ortiz-Sánchez E, de Stanchina E, Min IM, von Hofe E, Jin MM. Bispecific CAR T Cells against EpCAM and Inducible ICAM-1 Overcome Antigen Heterogeneity and Generate Superior Antitumor Responses. Cancer Immunol Res 2021; 9:1158-1174. [PMID: 34341066 PMCID: PMC8492509 DOI: 10.1158/2326-6066.cir-21-0062] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/26/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023]
Abstract
Adoptive transfer of chimeric antigen receptor (CAR) T cells has demonstrated unparalleled responses in hematologic cancers, yet antigen escape and tumor relapse occur frequently. CAR T-cell therapy for patients with solid tumors faces even greater challenges due to the immunosuppressive tumor environment and antigen heterogeneity. Here, we developed a bispecific CAR to simultaneously target epithelial cell adhesion molecule (EpCAM) and intercellular adhesion molecule 1 (ICAM-1) to overcome antigen escape and to improve the durability of tumor responses. ICAM-1 is an adhesion molecule inducible by inflammatory cytokines and elevated in many types of tumors. Our study demonstrates superior efficacy of bispecific CAR T cells compared with CAR T cells targeting a single primary antigen. Bispecific CAR T achieved more durable antitumor responses in tumor models with either homogenous or heterogenous expression of EpCAM. We also showed that the activation of CAR T cells against EpCAM in tumors led to upregulation of ICAM-1, which rendered tumors more susceptible to ICAM-1 targeting by bispecific CAR T cells. Our strategy of additional targeting of ICAM-1 may have broad applications in augmenting the activity of CAR T cells against primary tumor antigens that are prone to antigen loss or downregulation.
Collapse
MESH Headings
- Animals
- Antigenic Drift and Shift
- CRISPR-Cas Systems
- Cell Line, Tumor
- Cytotoxicity, Immunologic
- Epithelial Cell Adhesion Molecule/genetics
- Epithelial Cell Adhesion Molecule/metabolism
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Intercellular Adhesion Molecule-1/genetics
- Intercellular Adhesion Molecule-1/metabolism
- Male
- Mice
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Yanping Yang
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Jaclyn E McCloskey
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Huan Yang
- AffyImmune Therapeutics, Inc., Natick, Massachusetts
| | - Janusz Puc
- AffyImmune Therapeutics, Inc., Natick, Massachusetts
| | - Yago Alcaina
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Yogindra Vedvyas
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Angel A Gomez Gallegos
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México, México
| | - Elizabeth Ortiz-Sánchez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México, México
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Irene M Min
- Department of Surgery, Weill Cornell Medicine, New York, New York
| | - Eric von Hofe
- AffyImmune Therapeutics, Inc., Natick, Massachusetts
| | - Moonsoo M Jin
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, New York, New York.
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
20
|
Theocharopoulos C, Lialios PP, Samarkos M, Gogas H, Ziogas DC. Antibody-Drug Conjugates: Functional Principles and Applications in Oncology and Beyond. Vaccines (Basel) 2021; 9:1111. [PMID: 34696218 PMCID: PMC8538104 DOI: 10.3390/vaccines9101111] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/28/2022] Open
Abstract
In the era of precision medicine, antibody-based therapeutics are rapidly enriched with emerging advances and new proof-of-concept formats. In this context, antibody-drug conjugates (ADCs) have evolved to merge the high selectivity and specificity of monoclonal antibodies (mAbs) with the cytotoxic potency of attached payloads. So far, ten ADCs have been approved by FDA for oncological indications and many others are currently being tested in clinical and preclinical level. This paper summarizes the essential components of ADCs, from their functional principles and structure up to their limitations and resistance mechanisms, focusing on all latest bioengineering breakthroughs such as bispecific mAbs, dual-drug platforms as well as novel linkers and conjugation chemistries. In continuation of our recent review on anticancer implication of ADC's technology, further insights regarding their potential usage outside of the oncological spectrum are also presented. Better understanding of immunoconjugates could maximize their efficacy and optimize their safety, extending their use in everyday clinical practice.
Collapse
Affiliation(s)
| | | | | | | | - Dimitrios C. Ziogas
- First Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 115 27 Athens, Greece; (C.T.); (P.-P.L.); (M.S.); (H.G.)
| |
Collapse
|