1
|
Hidaka N, Oyama Y, Koga M, Kondo N, Yasunaga Y, Shimakura T, Yamamoto N, Takahashi HE, Iwafuchi Y, Watanabe S, Kimura S, Hoshino Y, Kato H, Kinoshita Y, Kobayashi H, Tanaka T, Ushiku T, Nangaku M, Tanaka S, Makita N, Saito T, Ito N. Excess fibroblast growth factor 23 in alcoholic osteomalacia is derived from the bone. JBMR Plus 2025; 9:ziaf010. [PMID: 39963340 PMCID: PMC11831984 DOI: 10.1093/jbmrpl/ziaf010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 02/20/2025] Open
Abstract
Excess fibroblast growth factor 23 (FGF23), a mature osteocyte-derived phosphaturic hormone, causes chronic hypophosphatemic osteomalacia in adults. This rare condition was recently reported in 2 alcoholic patients, with marked improvement upon cessation of alcohol consumption, suggesting a link between alcohol and FGF23-related hypophosphatemia within the highly limited cases. This study aimed to investigate whether the source of excess FGF23 in alcohol-induced FGF23-related hypophosphatemic osteomalacia is the bone or the other organs. To achieve this goal, an immunohistochemical approach for the bone obtained from a patient was employed. Initial attempts at quantifying FGF23 in the bone using conventional immunohistochemistry (IHC) faced issues in quantifiability and sensitivity for low FGF23 expression levels. Therefore, next-generation IHC with phosphor-integrated dots (PIDs) was applied, which enabled the quantification of FGF23 expression in the bone across a broad range. Preliminary analyses using IHC with PIDs on normal bone samples (n = 12) provided a reference level (154.5 PID particles per cell). IHC with PIDs quantified suppressed physiological FGF23 expression in the bone samples from 3 patients with tumor-induced osteomalacia, where FGF23 is oversecreted from a tumor (13.6 PID particles per cell). Subsequently, bone samples obtained from a 70-yr-old male with alcohol-induced FGF23-related hypophosphatemic osteomalacia were analyzed, showing a higher number of PID particles per cell (199.4 PID particles per cell) than the reference level. This study suggests that orthotopic, bone-derived FGF23 is implicated in alcohol-induced FGF23-related hypophosphatemic osteomalacia. Furthermore, the study also demonstrated that highly sensitive IHC with PIDs could aid in the differential diagnosis of FGF23-related hypophosphatemia of unknown origin. Specifically, a bone sample with a low number of PID particles per cell indicates an excess ectopic secretion of FGF23; a bone sample with a normal to high number of PID particles per cell indicates an excess orthotopic secretion of FGF23.
Collapse
Affiliation(s)
- Naoko Hidaka
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo 113-8655, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Yuko Oyama
- Department of Nephrology, Saiseikai Niigata Kenoh Kikan Hospital, Niigata 955-0091, Japan
| | - Minae Koga
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo 113-8655, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Naoki Kondo
- Division of Orthopedic Surgery, Department of Regenerative and Transplant Medicine, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
| | - Yoichi Yasunaga
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | | | | | | | - Yoichi Iwafuchi
- Department of Nephrology, Saiseikai Niigata Kenoh Kikan Hospital, Niigata 955-0091, Japan
| | - So Watanabe
- Osteoporosis Center, The University of Tokyo Hospital, Tokyo 113-8655, Japan
- Department of Geriatric Medicine, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Soichiro Kimura
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo 113-8655, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Yoshitomo Hoshino
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo 113-8655, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Hajime Kato
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo 113-8655, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Yuka Kinoshita
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo 113-8655, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Hiroshi Kobayashi
- Osteoporosis Center, The University of Tokyo Hospital, Tokyo 113-8655, Japan
- Department of Orthopedic Surgery, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Takeyuki Tanaka
- Orthopedic Surgery, Sensory and Motor System Medicine, Surgical Sciences, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Sakae Tanaka
- Department of Orthopedic Surgery, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Noriko Makita
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, Tokyo 113-8655, Japan
- Osteoporosis Center, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Taku Saito
- Osteoporosis Center, The University of Tokyo Hospital, Tokyo 113-8655, Japan
- Department of Orthopedic Surgery, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Nobuaki Ito
- Osteoporosis Center, The University of Tokyo Hospital, Tokyo 113-8655, Japan
- Division of Therapeutic Development for Intractable Bone Diseases, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
2
|
Ge H, Liu C, Shen C, Hu D, Zhao X, Wang Y, Ge H, Qin R, Ma X, Wang Y. The effectiveness and safety of RC48 alone or in combination with PD-1 inhibitors for locally advanced or metastatic urothelial carcinoma: a multicenter, real-world study. J Transl Med 2025; 23:243. [PMID: 40022107 PMCID: PMC11871675 DOI: 10.1186/s12967-025-06237-4] [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: 10/04/2024] [Accepted: 02/11/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND RC48 is an antibody-drug conjugate (ADC) specifically targeting HER2. Phase II and III clinical trials have proven its significant anti-tumor effect against locally advanced or metastatic urothelial carcinoma (la/mUC). This study aims to further assess the effectiveness and safety of RC48 for patients with la/mUC and provide insights for further clinical practice. METHODS Retrospective analysis for 42 patients with la/mUC who underwent RC48 alone or in combination with PD-1 inhibitors therapy between 18 October 2022 and 1 May 2024 were conducted to assess effectiveness and safety of RC48. Descriptive statistics were used to summarize baseline characteristics, treatment-related adverse events, etc. Cox proportional risk model and the Kaplan-Meier method were applied to analyze patients' survival. RESULTS We observed a median progression-free survival (mPFS) of 6.2 months, although median overall survival (mOS) has not been reached so far. An objective response rate (ORR) of 54.8% and a disease control rate (DCR) of 83.3% was also observed. Patients with first-line therapy, second- or later-line therapy and neoadjuvant therapy were observed disease remission with ORRs of 47.7%, 40.0% and 100.0%, respectively. The most common treatment-related adverse events (TRAEs) include hypoesthesia and elevated transaminases which affect over 90.0% of patients and mostly grade 1-2 in severity, and no treatment-related fatalities were found. CONCLUSIONS This multicenter, real-world study confirms that RC48 alone or in combination with PD-1 inhibitors exerted a promising effectiveness and manageable safety for first-line, second- and post-line, and neoadjuvant therapy with la/mUC.
Collapse
Affiliation(s)
- Huaixi Ge
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Changxue Liu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Chengquan Shen
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ding Hu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xinzhao Zhao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yanhua Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Huimin Ge
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ruize Qin
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaocheng Ma
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yonghua Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
- Urinary Diseases Clinical Medical Research Center of Qingdao, Qingdao, Shandong, China.
- Shandong Province Medical and Health Key Laboratory of Urology, Qingdao, Shandong, China.
| |
Collapse
|
3
|
Lee HS. Spatial and Temporal Tumor Heterogeneity in Gastric Cancer: Discordance of Predictive Biomarkers. J Gastric Cancer 2025; 25:192-209. [PMID: 39822175 PMCID: PMC11739643 DOI: 10.5230/jgc.2025.25.e3] [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: 11/13/2024] [Accepted: 12/09/2024] [Indexed: 01/19/2025] Open
Abstract
Gastric cancer (GC) is a highly heterogeneous disease that varies in both histological presentation and genetic characteristics. Recent advances in the treatment of metastatic and unresectable GC have made several biomarker tests essential for patient management. Predictive biomarkers such as human epidermal growth factor receptor 2 (HER2), programmed death-ligand 1 (PD-L1), mismatch-repair (MMR) proteins, claudin 18.2, and fibroblast growth factor receptor 2b (FGFR2b) are commonly evaluated using immunohistochemistry. However, the expression levels of these biomarkers may vary across different tumor areas, and the accuracy of biomarker diagnosis can be affected by sample quantity, sample location, and collection method. Therefore, tumor heterogeneity presents substantial challenges for accurate biomarker-based diagnosis and prediction of therapeutic responses. Tumor heterogeneity can be categorized into spatial heterogeneity, which refers to variations within the primary tumor (intra-tumoral) or between primary and metastatic sites, and temporal heterogeneity, which encompasses changes over time. This review addresses the tumor heterogeneity in predictive biomarker expression in GC, focusing on HER2, PD-L1, MMR, the Epstein-Barr virus, claudin 18.2, and FGFR2b.
Collapse
Affiliation(s)
- Hye Seung Lee
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
4
|
Grairi M, Le Borgne M. Antibody-drug conjugates: prospects for the next generation. Drug Discov Today 2024; 29:104241. [PMID: 39542204 DOI: 10.1016/j.drudis.2024.104241] [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: 07/02/2024] [Revised: 10/28/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
The concept of a 'magic bullet' was first introduced by Paul Ehrlich in the early 1900s, he foresaw the advent of targeted therapies and the specific killing of harmful cells and/or microorganisms. However, these therapies were only used in the clinic after the second half of the 20th century with the development of specific monoclonal antibodies. To date, 13 antibody-drug conjugates (ADCs) are commercially available. Many advances have been made by modifying one or several of the three main components of an ADC, namely the antibody, the cleavable or non-cleavable linker or the payload, and by integrating conjugation chemistry. Despite these efforts, some problems have emerged and thus limit their effectiveness. New strategies could overcome these problems and identify the next generation of ADC.
Collapse
Affiliation(s)
- Meriem Grairi
- Institut des Sciences Pharmaceutiques et Biologiques (ISPB), Faculté de Pharmacie, Université Claude Bernard Lyon 1, Univ Lyon, 69373 Lyon, France
| | - Marc Le Borgne
- Institut des Sciences Pharmaceutiques et Biologiques (ISPB), Faculté de Pharmacie, Université Claude Bernard Lyon 1, Univ Lyon, 69373 Lyon, France; Small Molecules for Biological Targets Team, Centre de recherche en cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, Univ Lyon, 69373 Lyon, France.
| |
Collapse
|
5
|
Zhang Y, Wang L, Cao X, Song R, Yin S, Cheng Z, Li W, Shen K, Zhao T, Xu J, Liu S, Xie Q, Wu Y, Gao B, Guo Q, Wu J, Qiu X, Wang B, Zhang W, Yang T, Lu W, Zhu S. Evaluation of Double Self-Immolative Linker-Based Antibody-Drug Conjugate FDA022-BB05 with Enhanced Therapeutic Potential. J Med Chem 2024; 67:19852-19873. [PMID: 39444220 DOI: 10.1021/acs.jmedchem.4c02243] [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: 10/25/2024]
Abstract
Typical antibody-drug conjugates (ADCs) with valine-alanine linkage, often conjugated with the amino group in payloads, face challenges when interacting with hydroxyl group-containing payloads. Herein, we introduced a transformative Val-Ala-based double self-immolative linker-payload platform, reshaping ADCs by optimizing hydroxyl group-containing payload integration. Utilizing this platform, FDA022-BB05 was successfully conjugated with the hydroxyl group-containing payload DXd using trastuzumab (FDA022) as the monoclonal antibody (mAb). FDA022-BB05 demonstrated enhanced stability, effective cathepsin B sensitivity, reduced cell proliferation, increased bystander killing, and targeted delivery. Notably, acute toxicity evaluations in diverse preclinical models indicated favorable safety profiles and tolerability, with a broad therapeutic index in HER2-positive and -negative xenografts. Overall, these compelling findings support the promising therapeutic potential of FDA022-BB05, emphasizing the significance of diverse linker-payload platform strategies. This ADC is a valuable addition to targeted cancer therapy development, currently advancing through phase I clinical trials.
Collapse
Affiliation(s)
- Yifan Zhang
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Lei Wang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
| | - Xuemei Cao
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Ruiwen Song
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Sicheng Yin
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Zhiyang Cheng
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
| | - Weinan Li
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Keyu Shen
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Teng Zhao
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Jun Xu
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Shuangxi Liu
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Qian Xie
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Yinghan Wu
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Bei Gao
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Qingsong Guo
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Jingsong Wu
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Xuefei Qiu
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Baoxia Wang
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Wenbo Zhang
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Tong Yang
- R&D Department of Genetic Engineering, Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai 201210, P. R. China
| | - Wei Lu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
| | - Shulei Zhu
- Innovation Center for AI and Drug Discovery, School of Pharmacy, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, P. R. China
| |
Collapse
|
6
|
Mathiot L, Baldini C, Letissier O, Hollebecque A, Bahleda R, Gazzah A, Smolenschi C, Sakkal M, Danlos FX, Henon C, Beshiri K, Goldschmidt V, Parisi C, Patrikidou A, Michot JM, Marabelle A, Postel-Vinay S, Bernard-Tessier A, Loriot Y, Ponce S, Champiat S, Ouali K. Exploring the Role of Target Expression in Treatment Efficacy of Antibody-Drug Conjugates (ADCs) in Solid Cancers: A Comprehensive Review. Curr Oncol Rep 2024; 26:1236-1248. [PMID: 39066847 DOI: 10.1007/s11912-024-01576-9] [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] [Accepted: 06/29/2024] [Indexed: 07/30/2024]
Abstract
PURPOSE OF REVIEW Antibody-drug conjugates (ADCs) offer a promising path for cancer therapy, leveraging the specificity of monoclonal antibodies and the cytotoxicity of linked drugs. The success of ADCs hinges on precise targeting of cancer cells based on protein expression levels. This review explores the relationship between target protein expression and ADC efficacy in solid tumours, focusing on results of clinical trials conducted between January 2019 and May 2023. RECENT FINDINGS We hereby highlight approved ADCs, revealing their effectiveness even in low-expressing target populations. Assessing target expression poses challenges, owing to variations in scoring systems and biopsy types. Emerging methods, like digital image analysis, aim to standardize assessment. The complexity of ADC pharmacokinetics, tumour dynamics, and off-target effects emphasises the need for a balanced approach. This review underscores the importance of understanding target protein dynamics and promoting standardized evaluation methods in shaping the future of ADC-based cancer therapies.
Collapse
Affiliation(s)
- Laurent Mathiot
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Capucine Baldini
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Octave Letissier
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Antoine Hollebecque
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Rastislav Bahleda
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Anas Gazzah
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Cristina Smolenschi
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Madona Sakkal
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - François-Xavier Danlos
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Villejuif, France
- Centre d'Investigations Cliniques Biothérapies Pour Une Immunisation in Situ (BIOTHERIS), INSERM, CIC1428, Villejuif, France
| | - Clémence Henon
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Kristi Beshiri
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Vincent Goldschmidt
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Claudia Parisi
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Anna Patrikidou
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Jean-Marie Michot
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Aurélien Marabelle
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Sophie Postel-Vinay
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | | | - Yohann Loriot
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U981, Villejuif, France
| | - Santiago Ponce
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Stéphane Champiat
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Villejuif, France
- Centre d'Investigations Cliniques Biothérapies Pour Une Immunisation in Situ (BIOTHERIS), INSERM, CIC1428, Villejuif, France
| | - Kaïssa Ouali
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France.
| |
Collapse
|
7
|
Guo Q, Gao B, Song R, Li W, Zhu S, Xie Q, Lou S, Wang L, Shen J, Zhao T, Zhang Y, Wu J, Lu W, Yang T. FZ-AD005, a Novel DLL3-Targeted Antibody-Drug Conjugate with Topoisomerase I Inhibitor, Shows Potent Antitumor Activity in Preclinical Models. Mol Cancer Ther 2024; 23:1367-1377. [PMID: 38940283 PMCID: PMC11443207 DOI: 10.1158/1535-7163.mct-23-0701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/07/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
Delta-like ligand 3 (DLL3) is overexpressed in small cell lung cancer (SCLC) and has been considered an attractive target for SCLC therapy. Rovalpituzumab tesirine was the first DLL3-targeted antibody-drug conjugate (ADC) to enter clinical studies. However, serious adverse events limited progress in the treatment of SCLC with rovalpituzumab tesirine. In this study, we developed a novel DLL3-targeted ADC, FZ-AD005, by using DXd with potent cytotoxicity and a relatively better safety profile to maximize the therapeutic index. FZ-AD005 was generated by a novel anti-DLL3 antibody, FZ-A038, and a valine-alanine (Val-Ala) dipeptide linker to conjugate DXd. Moreover, Fc-silencing technology was introduced in FZ-AD005 to avoid off-target toxicity mediated by FcγRs and showed negligible Fc-mediated effector functions in vitro. In preclinical evaluation, FZ-AD005 exhibited DLL3-specific binding and demonstrated efficient internalization, bystander killing, and excellent in vivo antitumor activities in cell line-derived xenograft and patient-derived xenograft models. FZ-AD005 was stable in circulation with acceptable pharmacokinetic profiles in cynomolgus monkeys. FZ-AD005 was well tolerated in rats and monkeys. The safety profile of FZ-AD005 was favorable, and the highest nonseverely toxic dose was 30 mg/kg in cynomolgus monkeys. In conclusion, FZ-AD005 has the potential to be a superior DLL3-targeted ADC with a wide therapeutic window and is expected to provide clinical benefits for the treatment of patients with SCLC.
Collapse
Affiliation(s)
- Qingsong Guo
- Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai, China.
| | - Bei Gao
- Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai, China.
| | - Ruiwen Song
- Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai, China.
| | - Weinan Li
- Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai, China.
| | - Shulei Zhu
- School of Pharmacy, East China Normal University, Shanghai, China
| | - Qian Xie
- Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai, China.
| | - Sensen Lou
- Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai, China.
| | - Lei Wang
- School of Pharmacy, East China Normal University, Shanghai, China
| | - Jiafei Shen
- Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai, China.
| | - Teng Zhao
- Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai, China.
| | - Yifan Zhang
- Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai, China.
| | - Jinsong Wu
- Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai, China.
| | - Wei Lu
- School of Pharmacy, East China Normal University, Shanghai, China
| | - Tong Yang
- Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai, China.
| |
Collapse
|
8
|
Greenman M, Chang YE, McNamara B, Mutlu L, Santin AD. Unmet needs in cervical cancer - can biological therapies plug the gap? Expert Opin Biol Ther 2024; 24:995-1003. [PMID: 39311611 DOI: 10.1080/14712598.2024.2408754] [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: 06/10/2024] [Accepted: 09/22/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION Cervical cancer remains one of the most common gynecologic malignancies worldwide. A disproportionate burden of cases occurs in developing countries due to inadequate screening and treatment. Even among patients adequately treated, in the presence of locally advanced or recurrent disease, outcomes tend to be poor. The introduction of biologic therapy into treatment has increased overall survival; however, a considerable opportunity still exists to improve current standards in treatment. Biologics have shown antitumor activity in multiple tumor types and are actively being pursued for the management of cervical cancer. AREAS COVERED In this article, we will discuss the historical evolution of biologic therapy in cervical cancer including use of angiogenesis inhibitors, immune checkpoint inhibitors, antibody-drug conjugates, and vaccines. We will review how these therapies have been integrated into current treatment recommendations and discuss ongoing investigations intended to improve clinical outcomes. We also postulate on persistent gaps in care. EXPERT OPINION Biologic therapies have had a tremendous impact on our current approach to managing cervical cancer. We anticipate that significant more research and development will be committed to the continued investigation of biologics in cervical cancer in an effort to improve a historically difficult to treat malignancy.
Collapse
Affiliation(s)
- Michelle Greenman
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Yifan Emily Chang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Blair McNamara
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Levent Mutlu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
9
|
Yoon J, Oh DY. HER2-targeted therapies beyond breast cancer - an update. Nat Rev Clin Oncol 2024; 21:675-700. [PMID: 39039196 DOI: 10.1038/s41571-024-00924-9] [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: 06/28/2024] [Indexed: 07/24/2024]
Abstract
The receptor tyrosine-kinase HER2 (also known as ErbB2) is a well-established therapeutic target in patients with breast or gastric cancer selected on the basis of HER2 overexpression on immunohistochemistry and/or ERBB2 amplification on in situ hybridization. With advances in cancer molecular profiling and increased implementation of precision medicine approaches into oncology practice, actionable HER2 alterations in solid tumours have expanded to include ERBB2 mutations in addition to traditional HER2 overexpression and ERBB2 amplification. These various HER2 alterations can be found in solid tumour types beyond breast and gastric cancer, although few HER2-targeted therapeutic options have been established for the other tumour types. Nevertheless, during the 5 years since our previous Review on this topic was published in this journal, obvious and fruitful progress in the development of HER2-targeted therapies has been made, including new disease indications, innovative drugs with diverse mechanisms of action and novel frameworks for approval by regulatory authorities. These advances have culminated in the recent histology-agnostic approval of the anti-HER2 antibody-drug conjugate trastuzumab deruxtecan for patients with HER2-overexpressing solid tumours. In this new Review, we provide an update on the current development landscape of HER2-targeted therapies beyond breast cancer, as well as anticipated future HER2-directed treatment strategies to overcome resistance and thereby improve efficacy and patient outcomes.
Collapse
Affiliation(s)
- Jeesun Yoon
- Division of Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Do-Youn Oh
- Division of Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Nagai Y, Oitate M, Shibayama T, Takakusa H, Watanabe N. Unveiling the intra-tumor fate of trastuzumab deruxtecan in a xenograft model to support its mechanism of action. Drug Metab Pharmacokinet 2024; 56:101001. [PMID: 38643548 DOI: 10.1016/j.dmpk.2024.101001] [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: 06/20/2023] [Revised: 01/08/2024] [Accepted: 01/17/2024] [Indexed: 04/23/2024]
Abstract
Trastuzumab deruxtecan (T-DXd) is an antibody-drug conjugate used for cancer treatment comprising an anti-human epidermal growth factor receptor type 2 (HER2) antibody and the topoisomerase I inhibitor DXd. The present study investigated the intratumor fate of T-DXd. Fluorescence-labeled T-DXd was found to accumulate in tumors of HER2-positive tumor xenograft mice and was observed to be distributed within lysosomes of in vitro tumor cells in accordance with their HER2 expression. DXd was released by cysteine proteases, including cathepsins, in lysosomal fractions in vitro in response to the pH. Tumor slices obtained from HER2-positive tumor xenograft mice treated with T-DXd were examined by semi-quantitative and three-dimensional immunohistochemical assays using phosphor-integrated dots, which visualized DXd-related signals in the nucleus, the site of topoisomerase I inhibition. In addition, based on the data showing the antibody component of T-DXd barely distributed in the nucleus, it was suggested that the DXd-related signals detected in the nucleus were predominantly derived from free DXd. These observations help support the mode of action of T-DXd from the perspective of drug disposition.
Collapse
Affiliation(s)
- Yoko Nagai
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan.
| | - Masataka Oitate
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Takahiro Shibayama
- Quantitative Clinical Pharmacology, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Hideo Takakusa
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Nobuaki Watanabe
- Precision Medicine Function, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| |
Collapse
|
11
|
Martín M, Pandiella A, Vargas-Castrillón E, Díaz-Rodríguez E, Iglesias-Hernangómez T, Martínez Cano C, Fernández-Cuesta I, Winkow E, Perelló MF. Trastuzumab deruxtecan in breast cancer. Crit Rev Oncol Hematol 2024; 198:104355. [PMID: 38621469 DOI: 10.1016/j.critrevonc.2024.104355] [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: 03/05/2023] [Revised: 02/06/2024] [Accepted: 04/10/2024] [Indexed: 04/17/2024] Open
Abstract
Trastuzumab deruxtecan (T-DXd) is an antibody-drug conjugate (ADC) consisting of a humanised, anti-human epidermal growth factor receptor 2 (HER2) monoclonal antibody covalently linked to a topoisomerase I inhibitor cytotoxic payload (DXd). The high drug-to-antibody ratio (8:1) ensures a high DXd concentration is delivered to target tumour cells, following internalisation of T-DXd and subsequent cleavage of its tetrapeptide-based linker. DXd's membrane-permeable nature enables it to cross cell membranes and potentially exert antitumour activity on surrounding tumour cells regardless of HER2 expression. T-DXd's unique mechanism of action is reflected in its efficacy in clinical trials in patients with HER2-positive advanced breast cancer (in heavily pretreated populations and in those previously treated with a taxane and trastuzumab), as well as HER2-low metastatic breast cancer. Thus, ADCs such as T-DXd have the potential to change the treatment paradigm of targeting HER2 in metastatic breast cancer, including eventually within the adjuvant/neoadjuvant setting.
Collapse
Affiliation(s)
- Miguel Martín
- Instituto de Investigación Sanitaria Hospital Gregorio Marañón, Universidad Complutense, CIBERONC, Calle Doctor Esquerdo, 46, Madrid 28007, Spain.
| | - Atanasio Pandiella
- Centro de Investigación del Cáncer, Universidad de Salamanca-CSIC-IBSAL and CIBERONC, Campus Miguel de Unamuno, Salamanca 37007, Spain
| | - Emilio Vargas-Castrillón
- Servicio de Farmacología Clínica, Hospital Clínico San Carlos, Calle del Prof Martín Lagos, S/N, Madrid 28040, Spain; Facultad de Medicina, Universidad Complutense de Madrid, Plaza de Ramón y Cajal, s/n, Madrid 28040, Spain
| | - Elena Díaz-Rodríguez
- Centro de Investigación del Cáncer, Universidad de Salamanca-CSIC-IBSAL and CIBERONC, Campus Miguel de Unamuno, Salamanca 37007, Spain
| | - Teresa Iglesias-Hernangómez
- Servicio de Farmacología Clínica, Hospital Clínico San Carlos, Calle del Prof Martín Lagos, S/N, Madrid 28040, Spain
| | - Concha Martínez Cano
- Daiichi Sankyo, Paseo Club Deportivo, 1, Edificio 14, Madrid, Pozuelo de Alarcón 28223, Spain
| | | | - Elena Winkow
- Daiichi Sankyo, Paseo Club Deportivo, 1, Edificio 14, Madrid, Pozuelo de Alarcón 28223, Spain
| | - Maria Francesca Perelló
- Daiichi Sankyo, Paseo Club Deportivo, 1, Edificio 14, Madrid, Pozuelo de Alarcón 28223, Spain
| |
Collapse
|
12
|
Saito N, Matsuo T, Tsuda H, Yokota H, Okada H. Novel approach to HER2 quantification using phosphor-integrated dots in human breast invasive cancer microarray. PLoS One 2024; 19:e0303614. [PMID: 38748758 PMCID: PMC11095758 DOI: 10.1371/journal.pone.0303614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/28/2024] [Indexed: 05/19/2024] Open
Abstract
HER2 expression in breast cancer is evaluated to select patients for anti-HER2 therapy. With the advent of newly approved HER2-targeted drugs for low HER2 expression breast cancer, more solid evidence on the whole spectrum of HER2 expression is needed. In this study, we quantitatively assessed HER2 expression from the whole core by combining high-intensity phosphor-integrated dot (PID) immunostaining and whole slide imaging (WSI) analysis. Two types of staining were performed using a 170-core tissue microarray of invasive breast cancer. First, HER2 was stained by immunohistochemistry (IHC), and IHC scores were determined by two practicing pathologists according to the ASCO/CAP HER2 guideline. Second, HER2 was stained with PID, and tentative PID scores were determined by quantitative analysis. The results show that PID can numerically classify HER2 expression status into scores 3+, 2+, 1+, and 0. The HER2 value quantified by PID strongly correlated with the 3, 3'-diaminobenzidine (DAB) IHC score determined by pathologists (R2 = 0.93). PID IHC score 1+ cases included both DAB IHC score 1+ and 0 cases, and low HER2 expression cases appeared to be often evaluated as DAB IHC score 0. Therefore, digital image analysis by PID and WSI can help stratify HER2 IHC. It may also help classify low HER2 expression.
Collapse
Affiliation(s)
- Naoya Saito
- Technology Development Headquarters, Advanced Core Technology Center, Konica Minolta, Inc., Hachioji, Japan
| | - Tsukasa Matsuo
- Technology Development Headquarters, Advanced Core Technology Center, Konica Minolta, Inc., Hachioji, Japan
| | - Hitoshi Tsuda
- Department of Basic Pathology, National Defense Medical College, Saitama, Japan
| | - Hiroyuki Yokota
- Technology Development Headquarters, Advanced Core Technology Center, Konica Minolta, Inc., Hachioji, Japan
| | - Hisatake Okada
- Technology Development Headquarters, Advanced Core Technology Center, Konica Minolta, Inc., Hachioji, Japan
| |
Collapse
|
13
|
Wei Q, Yang T, Zhu J, Zhang Z, Yang L, Zhang Y, Hu C, Chen J, Wang J, Tian X, Shimura T, Fang J, Ying J, Fan M, Guo P, Cheng X. Spatiotemporal Quantification of HER2-targeting Antibody-Drug Conjugate Bystander Activity and Enhancement of Solid Tumor Penetration. Clin Cancer Res 2024; 30:984-997. [PMID: 38113039 DOI: 10.1158/1078-0432.ccr-23-1725] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/03/2023] [Accepted: 12/15/2023] [Indexed: 12/21/2023]
Abstract
PURPOSE Antibody-drug conjugate (ADC) has had a transformative effect on the treatment of many solid tumors, yet it remains unclear how ADCs exert bystander activity in the tumor microenvironment. EXPERIMENTAL DESIGN Here, we directly visualized and spatiotemporally quantified the intratumor biodistribution and pharmacokinetics of different ADC components by developing dual-labeled fluorescent probes. RESULTS Mechanistically, we found that tumor penetration of ADCs is distinctly affected by their ability to breach the binding site barrier (BSB) in perivascular regions of tumor vasculature, and bystander activity of ADC can only partially breach BSB. Furthermore, bystander activity of ADCs can work in synergy with coadministration of their parental antibodies, leading to fully bypassing BSBs and enhancing tumor penetration via a two-step process. CONCLUSIONS These promising preclinical data allowed us to initiate a phase I/II clinical study of coadministration of RC48 and trastuzumab in patients with malignant stomach cancer to further evaluate this treatment strategy in humans.
Collapse
Affiliation(s)
- Qing Wei
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, P.R. China
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, P.R. China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, P.R. China
| | - Teng Yang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, P.R. China
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, P.R. China
| | - Jiayu Zhu
- Zhejiang Chinese Medical University, Hangzhou, P.R. China
| | - Ziwen Zhang
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, P.R. China
| | - Le Yang
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, P.R. China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, P.R. China
| | - Yuchao Zhang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, P.R. China
| | - Can Hu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, P.R. China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, P.R. China
| | - Jiahui Chen
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, P.R. China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, P.R. China
| | - Jinchao Wang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, P.R. China
| | - Xuefei Tian
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, P.R. China
- Shanghai Institute of Materia Medica, University of Chinese Academy of Sciences, Shanghai, P.R. China
- College of Molecular Medicine, Hangzhou Institute for Advanced Study (HIAS), University of Chinese Academy of Sciences, Hangzhou, P.R. China
| | - Takaya Shimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Jianmin Fang
- School of Life Science and Technology, Tongji University, Shanghai, P.R. China
| | - Jieer Ying
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, P.R. China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, P.R. China
| | - Mengyang Fan
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, P.R. China
| | - Peng Guo
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, P.R. China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, P.R. China
| | - Xiangdong Cheng
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, P.R. China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, P.R. China
| |
Collapse
|
14
|
McNamara B, Greenman M, Pebley N, Mutlu L, Santin AD. Antibody-Drug Conjugates (ADC) in HER2/neu-Positive Gynecologic Tumors. Molecules 2023; 28:7389. [PMID: 37959808 PMCID: PMC10650896 DOI: 10.3390/molecules28217389] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are a new class of targeted anti-cancer therapies that combine a monoclonal tumor-surface-receptor-targeting antibody with a highly cytotoxic molecule payload bonded through specifically designed cleavable or non-cleavable chemical linkers. One such tumor surface receptor is human epidermal growth factor 2 (HER2), which is of interest for the treatment of many gynecologic tumors. ADCs enable the targeted delivery of a variety of cytotoxic therapies to tumor cells while minimizing delivery to healthy tissues. This review summarizes the existing literature about HER2-targeting ADC therapies approved for use in gynecologic malignancies, relevant preclinical studies, strategies to address ADC resistance, and ongoing clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Alessandro D. Santin
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
15
|
Ohkuma R, Miura S, Muto S, Toyomasu Y, Fujimoto Y, Ieguchi K, Onishi N, Shimizu T, Watanabe M, Takayanagi D, Goshima T, Horiike A, Hamada K, Ariizumi H, Shimokawa M, Hirasawa Y, Ishiguro T, Suzuki R, Iriguchi N, Tsurui T, Mura E, Takenoshita S, Numajiri K, Okabe N, Yoshimura K, Tsuji M, Kiuchi Y, Yajima T, Ishida H, Suzuki H, Yamochi T, Kobayashi S, Tsunoda T, Wada S. Novel quantitative immunohistochemical analysis for evaluating PD-L1 expression with phosphor-integrated dots for predicting the efficacy of patients with cancer treated with immune checkpoint inhibitors. Front Immunol 2023; 14:1260492. [PMID: 37790929 PMCID: PMC10544572 DOI: 10.3389/fimmu.2023.1260492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/04/2023] [Indexed: 10/05/2023] Open
Abstract
Introduction Programmed cell death ligand 1 (PD-L1) expression in tumor tissues is measured as a predictor of the therapeutic efficacy of immune checkpoint inhibitors (ICIs) in many cancer types. PD-L1 expression is evaluated by immunohistochemical staining using 3,3´-diaminobenzidine (DAB) chronogenesis (IHC-DAB); however, quantitative and reproducibility issues remain. We focused on a highly sensitive quantitative immunohistochemical method using phosphor-integrated dots (PIDs), which are fluorescent nanoparticles, and evaluated PD-L1 expression between the PID method and conventional DAB method. Methods In total, 155 patients with metastatic or recurrent cancer treated with ICIs were enrolled from four university hospitals. Tumor tissue specimens collected before treatment were subjected to immunohistochemical staining with both the PID and conventional DAB methods to evaluate PD-L1 protein expression. Results PD-L1 expression assessed using the PID and DAB methods was positively correlated. We quantified PD-L1 expression using the PID method and calculated PD-L1 PID scores. The PID score was significantly higher in the responder group than in the non-responder group. Survival analysis demonstrated that PD-L1 expression evaluated using the IHC-DAB method was not associated with progression-free survival (PFS) or overall survival (OS). Yet, PFS and OS were strikingly prolonged in the high PD-L1 PID score group. Conclusion Quantification of PD-L1 expression as a PID score was more effective in predicting the treatment efficacy and prognosis of patients with cancer treated with ICIs. The quantitative evaluation of PD-L1 expression using the PID method is a novel strategy for protein detection. It is highly significant that the PID method was able to identify a group of patients with a favorable prognosis who could not be identified by the conventional DAB method.
Collapse
Affiliation(s)
- Ryotaro Ohkuma
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
| | - Sakiko Miura
- Department of Pathology, Showa University School of Medicine, Tokyo, Japan
| | - Satoshi Muto
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Yoshitaka Toyomasu
- Department of Digestive Tract and General Surgery, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Yuki Fujimoto
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
| | - Katsuaki Ieguchi
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
| | - Nobuyuki Onishi
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
| | - Takashi Shimizu
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
| | - Makoto Watanabe
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
- Department of Pharmacology, School of Medicine, Showa University, Tokyo, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
| | - Daisuke Takayanagi
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
| | - Tsubasa Goshima
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
| | - Atsushi Horiike
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
| | - Kazuyuki Hamada
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Hirotsugu Ariizumi
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
| | - Masahiro Shimokawa
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
| | - Yuya Hirasawa
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
| | - Tomoyuki Ishiguro
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
| | - Risako Suzuki
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
| | - Nana Iriguchi
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
| | - Toshiaki Tsurui
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
- Department of Pharmacology, School of Medicine, Showa University, Tokyo, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
| | - Emiko Mura
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
| | - Sachiko Takenoshita
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
| | - Kazuki Numajiri
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Naoyuki Okabe
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Kiyoshi Yoshimura
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
- Department of Clinical Immuno Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
| | - Mayumi Tsuji
- Pharmacological Research Center, Showa University, Tokyo, Japan
| | - Yuji Kiuchi
- Department of Pharmacology, School of Medicine, Showa University, Tokyo, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
| | - Toshiki Yajima
- Department of General Thoracic Surgery, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Hideyuki Ishida
- Department of Digestive Tract and General Surgery, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Hiroyuki Suzuki
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Toshiko Yamochi
- Department of Pathology, Showa University School of Medicine, Tokyo, Japan
| | - Shinichi Kobayashi
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
| | - Takuya Tsunoda
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
| | - Satoshi Wada
- Division of Medical Oncology, Department of Medicine, School of Medicine, Showa University, Tokyo, Japan
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
- Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
- Department of Pharmacology, School of Medicine, Showa University, Tokyo, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
| |
Collapse
|
16
|
Dumontet C, Reichert JM, Senter PD, Lambert JM, Beck A. Antibody-drug conjugates come of age in oncology. Nat Rev Drug Discov 2023; 22:641-661. [PMID: 37308581 DOI: 10.1038/s41573-023-00709-2] [Citation(s) in RCA: 313] [Impact Index Per Article: 156.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2023] [Indexed: 06/14/2023]
Abstract
Antibody-drug conjugates (ADCs) combine the specificity of monoclonal antibodies with the potency of highly cytotoxic agents, potentially reducing the severity of side effects by preferentially targeting their payload to the tumour site. ADCs are being increasingly used in combination with other agents, including as first-line cancer therapies. As the technology to produce these complex therapeutics has matured, many more ADCs have been approved or are in late-phase clinical trials. The diversification of antigenic targets as well as bioactive payloads is rapidly broadening the scope of tumour indications for ADCs. Moreover, novel vector protein formats as well as warheads targeting the tumour microenvironment are expected to improve the intratumour distribution or activation of ADCs, and consequently their anticancer activity for difficult-to-treat tumour types. However, toxicity remains a key issue in the development of these agents, and better understanding and management of ADC-related toxicities will be essential for further optimization. This Review provides a broad overview of the recent advances and challenges in ADC development for cancer treatment.
Collapse
Affiliation(s)
- Charles Dumontet
- CRCL INSERM 1052/CNRS 5286, University of Lyon, Hospices Civils de Lyon, Lyon, France.
| | | | | | | | - Alain Beck
- Institut de Recherche Pierre Fabre, CIPF, Saint-Julien-en-Genevois, France
| |
Collapse
|
17
|
Tarantino P, Ricciuti B, Pradhan SM, Tolaney SM. Optimizing the safety of antibody-drug conjugates for patients with solid tumours. Nat Rev Clin Oncol 2023:10.1038/s41571-023-00783-w. [PMID: 37296177 DOI: 10.1038/s41571-023-00783-w] [Citation(s) in RCA: 95] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2023] [Indexed: 06/12/2023]
Abstract
Over the past 5 years, improvements in the design of antibody-drug conjugates (ADCs) have enabled major advances that have reshaped the treatment of several advanced-stage solid tumours. Considering the intended rationale behind the design of ADCs, which is to achieve targeted delivery of cytotoxic molecules by linking them to antibodies targeting tumour-specific antigens, ADCs would be expected to be less toxic than conventional chemotherapy. However, most ADCs are still burdened by off-target toxicities that resemble those of the cytotoxic payload as well as on-target toxicities and other poorly understood and potentially life-threatening adverse effects. Given the rapid expansion in the clinical indications of ADCs, including use in curative settings and various combinations, extensive efforts are ongoing to improve their safety. Approaches currently being pursued include clinical trials optimizing the dose and treatment schedule, modifications of each ADC component, identification of predictive biomarkers for toxicities, and the development of innovative diagnostic tools. In this Review, we describe the determinants of the toxicities of ADCs in patients with solid tumours, highlighting key strategies that are expected to improve tolerability and enable improvements in the treatment outcomes of patients with advanced-stage and those with early stage cancers in the years to come.
Collapse
Affiliation(s)
- Paolo Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Biagio Ricciuti
- Harvard Medical School, Boston, MA, USA
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shan M Pradhan
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Nishikawa T, Hasegawa K, Matsumoto K, Mori M, Hirashima Y, Takehara K, Ariyoshi K, Kato T, Yagishita S, Hamada A, Kawasaki M, Kawashima S, Tomatsuri S, Nagasaka Y, Yoshida H, Machida R, Hirakawa A, Nakamura K, Yonemori K. Trastuzumab Deruxtecan for Human Epidermal Growth Factor Receptor 2-Expressing Advanced or Recurrent Uterine Carcinosarcoma (NCCH1615): The STATICE Trial. J Clin Oncol 2023; 41:2789-2799. [PMID: 36977309 PMCID: PMC10414746 DOI: 10.1200/jco.22.02558] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 02/01/2023] [Indexed: 03/30/2023] Open
Abstract
PURPOSE To investigate the efficacy and safety of trastuzumab deruxtecan, an antibody-drug conjugate targeting human epidermal growth factor receptor 2 (HER2) with a topoisomerase I inhibitor payload, in patients with uterine carcinosarcoma (UCS) expressing HER2. PATIENTS AND METHODS Patients with recurrent UCS with HER2 immunohistochemistry scores ≥1+ previously treated with chemotherapy were included. Patients were assigned to the HER2-high (immunohistochemistry score ≥2+; n = 22) or low (immunohistochemistry score of 1+; n = 10) groups for primary and exploratory analyses, respectively. Trastuzumab deruxtecan 6.4 or 5.4 mg/kg was administered intravenously once every 3 weeks until unacceptable toxicity or disease progression. Dose modification was based on the updated recommended phase II dose for breast cancer to be 5.4 mg/kg. The primary end point was the objective response rate by central review in the HER2-high group. Secondary end points included the overall response rate (ORR) in the HER2-high group by investigator assessment, ORR in the HER2-low group, progression-free survival (PFS), overall survival (OS), and safety. RESULTS The ORR by central review in the HER2-high and HER2-low groups were 54.5% (95% CI, 32.2 to 75.6) and 70.0% (95% CI, 34.8 to 93.3) and those by investigator assessments were 68.2% and 60.0%, respectively. The median PFS and OS in the HER2-high and HER2-low groups were 6.2 and 13.3 months and 6.7 months and not reached, respectively. Grade ≥ 3 adverse events occurred in 20 patients (61%). Grades 1-2 and 3 pneumonitis/interstitial lung disease occurred in eight (24%) and one (3%) patient, respectively. CONCLUSION Trastuzumab deruxtecan has efficacy in patients with UCS, regardless of HER2 status. The safety profile was generally consistent with that previously reported. Toxicities were manageable with appropriate monitoring and treatment.
Collapse
Affiliation(s)
- Tadaaki Nishikawa
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Koji Matsumoto
- Medical Oncology Division, Ambulatory Chemotherapy Center, Hyogo Cancer Center, Hyogo, Japan
| | - Masahiko Mori
- Department of Gynecologic Oncology, Aichi Cancer Center Hospital, Aichi, Japan
| | | | - Kazuhiro Takehara
- Department of Gynecologic Oncology, National Hospital Organization Shikoku Cancer Center, Ehime, Japan
| | - Kazuya Ariyoshi
- Gynecology Service, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Tomoyasu Kato
- Department of Gynecologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shigehiro Yagishita
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Akinobu Hamada
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Mamiko Kawasaki
- Clinical Research Support Office, National Cancer Center Hospital, Tokyo, Japan
| | - Satoshi Kawashima
- Clinical Research Support Office, National Cancer Center Hospital, Tokyo, Japan
| | - Sawako Tomatsuri
- Clinical Research Support Office, National Cancer Center Hospital, Tokyo, Japan
| | - Yukari Nagasaka
- Clinical Research Support Office, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroshi Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Ryunosuke Machida
- Biostatistics Division, Center for Research Administration and Support, National Cancer Center Hospital, Tokyo, Japan
| | - Akihiro Hirakawa
- Department of Clinical Biostatistics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kenichi Nakamura
- Clinical Research Support Office, National Cancer Center Hospital, Tokyo, Japan
| | - Kan Yonemori
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
19
|
Tatsumi A, Morichika K, Krueger J, Yokota H. Immunohistochemistry-Based Fluorescent Nanoparticle Assay for Determination of Antibody Binding Capacity and Its Correlation with Flow Cytometry Analysis. ANAL LETT 2022. [DOI: 10.1080/00032719.2022.2153365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Atsuro Tatsumi
- Precision Medicine Business Unit, Healthcare Business Headquarters, KONICA MINOLTA, INC.,Tokyo, Japan
| | - Keisuke Morichika
- Precision Medicine Business Unit, Healthcare Business Headquarters, KONICA MINOLTA, INC.,Tokyo, Japan
| | | | - Hiroyuki Yokota
- Precision Medicine Business Unit, Healthcare Business Headquarters, KONICA MINOLTA, INC.,Tokyo, Japan
| |
Collapse
|
20
|
Rassy E, Rached L, Pistilli B. Antibody drug conjugates targeting HER2: Clinical development in metastatic breast cancer. Breast 2022; 66:217-226. [PMID: 36334569 PMCID: PMC9636477 DOI: 10.1016/j.breast.2022.10.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/25/2022] [Accepted: 10/25/2022] [Indexed: 11/12/2022] Open
Abstract
The identification of the HER2 alteration as an actionable oncogenic driver in breast cancer has propelled the development of HER-targeting monoclonal antibodies (mAb) such as trastuzumab and pertuzumab, which led to dramatic improvements in survival outcomes. Lately, the great strides made toward developing antibody-conjugation methods have led to the development of a new class of compelling compounds, the antibody-drug conjugates (ADCs) targeting HER2 which have profoundly transformed the treatment landscape of breast cancer. HER2-targeting ADCs, trastuzumab-emtansine and trastuzumab-deruxtecan, have improved the overall survival in the second and third-line settings with manageable adverse events. Other HER2-targeting ADCs using novel technological advances in the antibody, linker and/or payload conception have shown promising activity in preclinical and clinical studies and some of them are now being evaluated in larger clinical trials. Multiple challenges still impede the success of ADCs in breast cancer namely the lack of a comprehensive understanding of resistance mechanisms as well as the mechanisms of action of ADCs in special subgroups of patients such as those with low or ultra-low HER2 expression and patients with brain or leptomeningeal metastases (BM). In this framework, we review the approved indications and ongoing trials for HER2-targeting ADCs, across patient subgroups, including those with BM and discuss the associated potential mechanisms of action and resistance. Last, we provide an overview of the future perspectives involving HER2-targeting ADCs in breast cancer.
Collapse
Affiliation(s)
- Elie Rassy
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Layal Rached
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Barbara Pistilli
- Department of Medical Oncology, Gustave Roussy, Villejuif, France.
| |
Collapse
|
21
|
Dias e Silva D, Andriatte GM, Pestana RC. Antibody-Drug Conjugates and Tissue-Agnostic Drug Development. Cancer J 2022; 28:462-468. [DOI: 10.1097/ppo.0000000000000633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
22
|
Tatsumi A, Morichika K, Krueger J, Yokota H. High-Sensitivity Immunohistochemistry Method Using a Combination of Fluorescent Nanoparticles and Tyramide Linker. ANAL LETT 2022. [DOI: 10.1080/00032719.2022.2123498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Atsuro Tatsumi
- Precision Medicine Business Unit, Healthcare Business Headquarters, KONICA MINOLTA, INC., Hino-shi, Japan
| | - Keisuke Morichika
- Precision Medicine Business Unit, Healthcare Business Headquarters, KONICA MINOLTA, INC., Hino-shi, Japan
| | | | - Hiroyuki Yokota
- Precision Medicine Business Unit, Healthcare Business Headquarters, KONICA MINOLTA, INC., Hino-shi, Japan
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Summarizing the current preclinical and clinical evidence about bystander effect of antibody-drug conjugates (ADCs) in solid tumors. RECENT FINDINGS One of the main challenges of treating solid tumors with ADCs is the heterogeneous expression of the target antigen (Ag), which however may be overcome by the so-called bystander killing effect. This unique, but still debated, feature of certain ADCs is represented by the unintentional payload diffusion from Ag-positive tumor cells to adjacent Ag-negative tumor cells. Some pharmacological characteristics, such as a hydrophobic payload or a cleavable linker, seem to play a major role in this effect. Abundant preclinical evidence of the bystander effect has emerged, and the clinical activity of ADCs in tumors with a heterogeneous Ag expression suggests the relevance of this feature. Additional studies are required to investigate if the bystander effect is necessary for achieving a solid activity with ADCs.
Collapse
|
24
|
Tarantino P, Carmagnani Pestana R, Corti C, Modi S, Bardia A, Tolaney SM, Cortes J, Soria JC, Curigliano G. Antibody-drug conjugates: Smart chemotherapy delivery across tumor histologies. CA Cancer J Clin 2022; 72:165-182. [PMID: 34767258 DOI: 10.3322/caac.21705] [Citation(s) in RCA: 239] [Impact Index Per Article: 79.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/26/2021] [Accepted: 10/04/2021] [Indexed: 12/15/2022] Open
Abstract
As distinct cancer biomarkers have been discovered in recent years, a need to reclassify tumors by more than their histology has been proposed, and therapies are now tailored to treat cancers based on specific molecular aberrations and immunologic markers. In fact, multiple histology-agnostic therapies are currently adopted in clinical practice for treating patients regardless of their tumor site of origin. In parallel with this new model for drug development, in the past few years, several novel antibody-drug conjugates (ADCs) have been approved to treat solid tumors, benefiting from engineering improvements in the conjugation process and the introduction of novel linkers and payloads. With the recognition that numerous surface targets are expressed across various cancer histologies, alongside the remarkable activity of modern ADCs, this drug class has been increasingly evaluated as suitable for a histology-agnostic expansion of indication. For illustration, the anti-HER2 ADC trastuzumab deruxtecan has demonstrated compelling activity in HER2-overexpressing breast, gastric, colorectal, and lung cancer. Examples of additional novel and potentially histology-agnostic ADC targets include trophoblast cell-surface antigen 2 (Trop-2) and nectin-4, among others. In the current review article, the authors summarize the current approvals of ADCs by the US Food and Drug Administration focusing on solid tumors and discuss the challenges and opportunities posed by the multihistological expansion of ADCs.
Collapse
Affiliation(s)
- Paolo Tarantino
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Roberto Carmagnani Pestana
- Dayan-Daycoval Family Center for Oncology and Hematology, Albert Einstein Israelite Hospital, Sao Paulo, Brazil
| | - Chiara Corti
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Shanu Modi
- Breast Medicine Service, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, New York
| | - Aditya Bardia
- Harvard Medical School, Boston, Massachusetts
- Breast Cancer Treatment Program, Massachusetts General Hospital, Boston, Massachusetts
| | - Sara M Tolaney
- Harvard Medical School, Boston, Massachusetts
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Javier Cortes
- International Breast Cancer Center, Quironsalud Group, Barcelona, Spain
- Medica Scientia Innovation Research, Barcelona, Spain
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, European University of Madrid, Madrid, Spain
| | - Jean-Charles Soria
- Paris Saclay University, St Aubin, France
- Drug Development Department, Gustave Roussy, Villejuif, France
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|