1
|
Bouguerra Zina B, Rousseau F, Fauquier S, Sabatier R, Kfoury M. Practical clinical management of ocular adverse events related to Antibody-Drug Conjugates in gynaecological malignancies. Cancer Treat Rev 2025; 134:102867. [PMID: 39970828 DOI: 10.1016/j.ctrv.2024.102867] [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: 09/16/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 02/21/2025]
Abstract
BACKGROUND The advent of Antibody-Drug Conjugates (ADC) represents a significant advancement in targeted therapy for gynaecological malignancies. However, the ocular toxicities associated with ADCs, particularly Tisotumab Vedotin (TV) and Mirvetuximab Soravtansine (MIRV) necessitate effective mitigation in order to optimise patient care. METHODS This review synthesises findings from clinical trials to delineate the spectrum of ocular adverse events induced by ADCs. The analysis focuses on the incidence, onset, severity and reversibility of adverse events. It examines the underlying mechanisms of toxicity and provides management strategies based on study protocols. RESULTS Adverse events mainly impact the anterior ocular segment, resulting in conjunctivitis and keratopathy. They affect up to 56 % of patients treated with MIRV and 50 to 60 % of those receiving TV. Symptoms like blurred vision, dryness and pain hinder the patient's quality of life. Events are CTCAE grade 3 or higher in less than 10 % of cases. The median time to onset is 1.3 months. However, ocular toxicity may appear up to 10 months after treatment initiation, indicating a need for prolonged vigilance. Primary prophylaxis calls for local corticotherapy, lubricants and in some cases, vasoconstrictors. Despite the potential for severity, most cases are reversible with local treatment and transient dose reduction and/or delay. Close monitoring is crucial for early detection and subsequent management. CONCLUSIONS Clinicians ought to be cognizant of the potential ocular toxicity of ADCs. Proactive prophylaxis, patient education and a multidisciplinary approach involving ophthalmologists are paramount to minimising the impact of these AEs. Further research is essential to measure the real outcome of preventive strategies and balance their benefits with potential short and long-term risks.
Collapse
Affiliation(s)
- Bochra Bouguerra Zina
- Department of Medical Oncology, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France
| | - Frédérique Rousseau
- Department of Medical Oncology, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France
| | | | - Renaud Sabatier
- Department of Medical Oncology, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France; CRCM, Predictive Oncology Laboratory, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France
| | - Maria Kfoury
- Department of Medical Oncology, Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, Marseille, France.
| |
Collapse
|
2
|
Mishra A, Sharma AK, Gupta K, Banka DR, Johnson BA, Hoffman-Censits J, Huang P, McConkey DJ, Nimmagadda S. NECTIN-4 PET FOR OPTIMIZING ENFORTUMAB VEDOTIN DOSE-RESPONSE IN UROTHELIAL CARCINOMA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.25.630315. [PMID: 39763905 PMCID: PMC11703263 DOI: 10.1101/2024.12.25.630315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
The optimization of dosing strategies is critical for maximizing efficacy and minimizing toxicity in drug development, particularly for drugs with narrow therapeutic windows such as antibody-drug conjugates (ADCs). This study demonstrates the utility of Nectin-4-targeted positron emission tomography (PET) imaging using [68Ga]AJ647 as a non-invasive tool for real-time assessment of target engagement in enfortumab vedotin (EV) therapy for urothelial carcinoma (UC). By leveraging the specificity of [68Ga]AJ647 for Nectin-4, we quantified dynamic changes in target engagement across preclinical models and established its correlation with therapeutic outcomes. PET imaging revealed dose-dependent variations in Nectin-4 engagement, with suboptimal EV doses resulting in incomplete Nectin-4 engagement and reduced tumor growth. Importantly, target engagement measured by PET emerged as a more reliable predictor of therapeutic efficacy than dose or baseline Nectin-4 expression alone. Receiver operating characteristic (ROC) analysis identified a target engagement threshold that is determinant of response, providing a quantitative benchmark for dose optimization. Furthermore, PET imaging measures provide a promising framework to account for key challenges in ADC development, including tumor heterogeneity, declining drug-to-antibody ratios over time, and limitations of systemic pharmacokinetic measurements to account for tumor-drug interactions. These findings underscore the transformative potential of integrating PET pharmacodynamic measures as early biomarkers to refine dosing strategies, improve patient outcomes, and accelerate the clinical translation of next-generation targeted therapeutics.
Collapse
Affiliation(s)
- Akhilesh Mishra
- The Russell H. Morgan Department of Radiology and Radiological Science
| | - Ajay Kumar Sharma
- The Russell H. Morgan Department of Radiology and Radiological Science
| | - Kuldeep Gupta
- The Russell H. Morgan Department of Radiology and Radiological Science
| | - Dhanush R. Banka
- The Russell H. Morgan Department of Radiology and Radiological Science
| | - Burles A. Johnson
- The Johns Hopkins Greenberg Bladder Cancer Institute
- The James Buchanan Brady Urological Institute, Sidney Kimmel Comprehensive Cancer center
| | - Jeannie Hoffman-Censits
- The Johns Hopkins Greenberg Bladder Cancer Institute
- The James Buchanan Brady Urological Institute, Sidney Kimmel Comprehensive Cancer center
| | - Peng Huang
- Department of Biostatistics
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center and the Bloomberg–Kimmel Institute for Cancer Immunotherapy
| | - David J. McConkey
- The Johns Hopkins Greenberg Bladder Cancer Institute
- The James Buchanan Brady Urological Institute, Sidney Kimmel Comprehensive Cancer center
| | - Sridhar Nimmagadda
- The Russell H. Morgan Department of Radiology and Radiological Science
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center and the Bloomberg–Kimmel Institute for Cancer Immunotherapy
- Department of Pharmacology and Molecular Sciences
- Division of Clinical Pharmacology, Department of Medicine Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| |
Collapse
|
3
|
Hu Q, Wang L, Yang Y, Lee JB. Review of dose justifications for antibody-drug conjugate approvals from clinical pharmacology perspective: A focus on exposure-response analyses. J Pharm Sci 2024; 113:3434-3446. [PMID: 39374692 DOI: 10.1016/j.xphs.2024.10.002] [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: 05/29/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/09/2024]
Abstract
Antibody-drug conjugates (ADCs) are revolutionizing cancer treatment by specific targeting of the cancer cells thereby improving the therapeutic window of the drugs. Nevertheless, they are not free from unwanted toxicities mainly resulting from non-specific targeting and release of the payload. Therefore, the dosing regimen must be optimized through integrated analysis of the risk-benefit profile, to maximize the therapeutic potential. Exposure-response (E-R) analysis is one of the most widely used tools for risk-benefit assessment and it plays a pivotal role in dose optimization of ADCs. However, compared to conventional E-R analysis, ADCs pose unique challenges since they feature properties of both small molecules and antibodies. In this article, we review the E-R analyses that have formed the key basis of dose justification for each of the 12 ADCs approved in the USA. We discuss the multiple analytes and exposure metrics that can be utilized for such analysis and their relevance for safety and efficacy of the treatment. For the endpoints used for the E-R analysis, we were able to uncover commonalities across different ADCs for both safety and efficacy. Additionally, we discuss dose optimization strategies for ADCs which are now a critical component in clinical development of oncology drugs.
Collapse
Affiliation(s)
- Qianqian Hu
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, 33620, USA
| | - Lujing Wang
- Clinical Pharmacology and Pharmacometrics, Bristol Myers Squibb, 3551 Lawrenceville Road, Princeton, NJ, 08543, USA
| | - Yuqing Yang
- Pharmacokinetic Sciences, Biomedical Research, Novartis, East Hanover, NJ, 07936, USA.
| | - Jong Bong Lee
- Pharmacokinetic Sciences, Biomedical Research, Novartis, East Hanover, NJ, 07936, USA.
| |
Collapse
|
4
|
Li X, Liu D, Liu S, Yu M, Wu X, Wang H. Application of Pharmacometrics in Advancing the Clinical Research of Antibody-Drug Conjugates: Principles and Modeling Strategies. Clin Pharmacokinet 2024; 63:1373-1387. [PMID: 39325307 DOI: 10.1007/s40262-024-01423-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 09/27/2024]
Abstract
Antibody-drug conjugates (ADCs) have become a pivotal area in the research and development of antitumor drugs. They provide innovative possibilities for tumor therapy by integrating the tumor-targeting capabilities of monoclonal antibodies with the cytotoxic effect of small molecule drugs. Pharmacometrics, an important discipline, facilitates comprehensive understanding of the pharmacokinetic characteristics of ADCs by integrating clinical trial data through modeling and simulation. However, due to the complex structure of ADCs, their modeling approaches are still unclear. In this review, we analyzed published population pharmacokinetic models for ADCs and classified them into single-analyte, two-analyte, and three-analyte models. We also described the benefits, limitations, and recommendations for each model. Furthermore, we suggested that the development of population pharmacokinetic models for ADCs should be rigorously considered and established based on four key aspects: (1) research objectives; (2) available in vitro and animal data; (3) accessible clinical information; and (4) the capability of bioanalytical methods. This review offered insights to guide the application of pharmacometrics in the clinical research of ADCs, thereby contributing to more effective therapeutic development.
Collapse
Affiliation(s)
- Xiuqi Li
- State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Dan Liu
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Shupeng Liu
- State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Mengyang Yu
- State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Xiaofei Wu
- State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Hongyun Wang
- State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
5
|
Hengel SM, Topletz-Erickson AR, Kadry H, Alley SC. A modelling approach to compare ADC deconjugation and systemic elimination rates of individual drug-load species using native ADC LC-MS data from human plasma. Xenobiotica 2024; 54:492-501. [PMID: 39329288 DOI: 10.1080/00498254.2024.2340741] [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: 02/19/2024] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 09/28/2024]
Abstract
Native liquid chromatography mass spectrometry (LC-MS) is a commonly used approach for intact analysis of inter-chain cysteine conjugated antibody-drug conjugates (ADCs). Coupling native LC-MS with affinity capture provides a platform for intact ADC analysis from in vivo samples and characterisation of individual drug load species, specifically the impact of drug linker deconjugation, hydrolysis, and differential clearance in a biological system.This manuscript describes data generated from native LC-MS analysis of ADCs from human plasma, both in vitro incubations and clinical samples. It also details the pharmacokinetic (PK) model built to specifically characterise the disposition of individual drug load species from MMAE and MMAF interchain cysteine conjugated ADCs.In vitro deconjugation and hydrolysis rates were similar across both ADCs. Differential clearance of higher loaded species in vivo was pronounced for the MMAE conjugated ADC, while systemic elimination after accounting for deconjugation was similar across drug loads for the MMAF conjugated ADC. This is the first report of affinity capture native LC-MS analysis, and subsequent modelling of deconjugation, hydrolysis and clearance rates of individual drug load species using clinical data from cysteine conjugated ADCs.
Collapse
Affiliation(s)
- Shawna M Hengel
- Clinical Pharmacology and Translational Science, Pfizer Inc, Bothell, Washington, USA
| | | | - Hossam Kadry
- Clinical Pharmacology and Translational Science, Pfizer Inc, Bothell, Washington, USA
| | - Stephen C Alley
- Clinical Pharmacology and Translational Science, Pfizer Inc, Bothell, Washington, USA
| |
Collapse
|
6
|
Tang M, Garg A, Bonate PL, Rosenberg JE, Matsangou M, Kadokura T, Yamada A, Choules M, Pavese J, Nagata M, Tenmizu D, Koibuchi A, Heo N, Wang L, Wojtkowski T, Hanley WD, Poondru S. Clinical Pharmacology of the Antibody-Drug Conjugate Enfortumab Vedotin in Advanced Urothelial Carcinoma and Other Malignant Solid Tumors. Clin Pharmacokinet 2024; 63:423-438. [PMID: 38609704 PMCID: PMC11052883 DOI: 10.1007/s40262-024-01369-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 04/14/2024]
Abstract
Enfortumab vedotin is an antibody-drug conjugate comprised of a human monoclonal antibody directed to Nectin-4 and monomethyl auristatin E (MMAE), a microtubule-disrupting agent. The objectives of this review are to summarize the clinical pharmacology of enfortumab vedotin monotherapy and demonstrate that the appropriate dose has been selected for clinical use. Pharmacokinetics (PK) of enfortumab vedotin (antibody-drug conjugate and total antibody) and free MMAE were evaluated in five clinical trials of patients with locally advanced or metastatic urothelial carcinoma (n = 748). Intravenous enfortumab vedotin 0.5-1.25 mg/kg on days 1, 8, and 15 of a 28-day cycle showed linear, dose-proportional PK. No significant differences in exposure or safety of enfortumab vedotin and free MMAE were observed in mild, moderate, or severe renal impairment versus normal renal function. Patients with mildly impaired versus normal hepatic function had a 37% increase in area under the concentration-time curve (0-28 days), a 31% increase in maximum concentration of free MMAE, and a similar adverse event profile. No clinically significant PK differences were observed based on race/ethnicity with weight-based dosing, and no clinically meaningful QT prolongation was observed. Concomitant use with dual P-glycoprotein and strong cytochrome P450 3A4 inhibitors may increase MMAE exposure and the risk of adverse events. Approximately 3% of patients developed antitherapeutic antibodies against enfortumab vedotin 1.25 mg/kg. These findings support enfortumab vedotin 1.25 mg/kg monotherapy on days 1, 8, and 15 of a 28-day cycle. No dose adjustments are required for patients with renal impairment or mild hepatic impairment, or by race/ethnicity.
Collapse
Affiliation(s)
- Mei Tang
- Clinical Pharmacology and Exploratory Development, Astellas Pharma Global Development Inc., 2375 Waterview Drive, Northbrook, IL, 60062-6111, USA
| | - Amit Garg
- Clinical Pharmacology Oncology, Pfizer Inc., 181 Oyster Point Boulevard, South San Francisco, CA, 94080, USA.
| | - Peter L Bonate
- Clinical Pharmacology and Exploratory Development, Astellas Pharma Global Development Inc., 2375 Waterview Drive, Northbrook, IL, 60062-6111, USA
| | - Jonathan E Rosenberg
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria Matsangou
- Therapeutic Area-Oncology, Astellas Pharma Global Development, Astellas Pharma, Inc., Northbrook, IL, USA
| | - Takeshi Kadokura
- Clinical Pharmacology and Exploratory Development, Astellas Pharma Inc., Tokyo, Japan
| | - Akihiro Yamada
- Clinical Pharmacology and Exploratory Development, Astellas Pharma Inc., Tokyo, Japan
| | - Mary Choules
- Clinical Pharmacology and Exploratory Development, Astellas Pharma Global Development Inc., 2375 Waterview Drive, Northbrook, IL, 60062-6111, USA
| | - Janet Pavese
- Clinical Pharmacology and Exploratory Development, Astellas Pharma Global Development Inc., 2375 Waterview Drive, Northbrook, IL, 60062-6111, USA
| | - Masanori Nagata
- Analysis and Pharmacokinetics Research Laboratories, Drug Discovery Research, Astellas Pharma, Inc, Tsukuba, Japan
| | - Daisuke Tenmizu
- Analysis and Pharmacokinetics Research Laboratories, Drug Discovery Research, Astellas Pharma, Inc, Tsukuba, Japan
| | - Akira Koibuchi
- Clinical Pharmacology and Exploratory Development, Astellas Pharma Inc., Tokyo, Japan
| | - Nakyo Heo
- Clinical Pharmacology and Exploratory Development, Astellas Pharma Global Development Inc., 2375 Waterview Drive, Northbrook, IL, 60062-6111, USA
| | - Lu Wang
- Statistical and Real-World Data Science, Astellas Pharma Global Development Inc, Northbrook, IL, USA
| | - Tomasz Wojtkowski
- Data Science Development, Astellas Pharma Global Development Inc., Northbrook, IL, USA
| | - William D Hanley
- Clinical Pharmacology Oncology, Pfizer Inc., 181 Oyster Point Boulevard, South San Francisco, CA, 94080, USA
| | - Srinivasu Poondru
- Clinical Pharmacology and Exploratory Development, Astellas Pharma Global Development Inc., 2375 Waterview Drive, Northbrook, IL, 60062-6111, USA.
| |
Collapse
|
7
|
Joseph J, Sandel G, Kulkarni R, Alatrash R, Herrera BB, Jain P. Antibody and Cell-Based Therapies against Virus-Induced Cancers in the Context of HIV/AIDS. Pathogens 2023; 13:14. [PMID: 38251321 PMCID: PMC10821063 DOI: 10.3390/pathogens13010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/12/2023] [Accepted: 12/18/2023] [Indexed: 01/23/2024] Open
Abstract
Infectious agents, notably viruses, can cause or increase the risk of cancer occurrences. These agents often disrupt normal cellular functions, promote uncontrolled proliferation and growth, and trigger chronic inflammation, leading to cancer. Approximately 20% of all cancer cases in humans are associated with an infectious pathogen. The International Agency for Research on Cancer (IARC) recognizes seven viruses as direct oncogenic agents, including Epstein-Barr Virus (EBV), Kaposi's Sarcoma-associated herpesvirus (KSHV), human T-cell leukemia virus type-1 (HTLV-1), human papilloma virus (HPV), hepatitis C virus (HCV), hepatitis B virus (HBV), and human immunodeficiency virus type 1 (HIV-1). Most viruses linked to increased cancer risk are typically transmitted through contact with contaminated body fluids and high-risk behaviors. The risk of infection can be reduced through vaccinations and routine testing, as well as recognizing and addressing risky behaviors and staying informed about public health concerns. Numerous strategies are currently in pre-clinical phases or undergoing clinical trials for targeting cancers driven by viral infections. Herein, we provide an overview of risk factors associated with increased cancer incidence in people living with HIV (PLWH) as well as other chronic viral infections, and contributing factors such as aging, toxicity from ART, coinfections, and comorbidities. Furthermore, we highlight both antibody- and cell-based strategies directed against virus-induced cancers while also emphasizing approaches aimed at discovering cures or achieving complete remission for affected individuals.
Collapse
Affiliation(s)
- Julie Joseph
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (J.J.); (G.S.)
| | - Grace Sandel
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (J.J.); (G.S.)
| | - Ratuja Kulkarni
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (J.J.); (G.S.)
| | - Reem Alatrash
- Global Health Institute, Rutgers University, New Brunswick, NJ 08901, USA; (R.A.); (B.B.H.)
- Department of Medicine, Division of Allergy, Immunology and Infectious Diseases, Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Bobby Brooke Herrera
- Global Health Institute, Rutgers University, New Brunswick, NJ 08901, USA; (R.A.); (B.B.H.)
- Department of Medicine, Division of Allergy, Immunology and Infectious Diseases, Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Pooja Jain
- Department of Microbiology & Immunology, Drexel University College of Medicine, Philadelphia, PA 19129, USA; (J.J.); (G.S.)
| |
Collapse
|