1
|
Hou Y, Miao J, Sun Y, Shi L, Ouyang L, Chen X, Li Z, Liu T, Qin G, Qin Q, Gong L. Ligand-binding assays validated for quantitative bioanalysis of a novel antibody-drug conjugate in monkey serum and related application in a nonclinical study. J Pharmacol Toxicol Methods 2024; 131:107580. [PMID: 39743180 DOI: 10.1016/j.vascn.2024.107580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/16/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Antibody-drug conjugates (ADCs) are an emerging class of targeted therapeutics and are receiving growing attention in the pharmaceutical field. Here we aimed to validate two ligand binding assays for the quantitation of GQ1001, an ADC made of Trastuzumab site-specifically conjugated with DM1, in cynomolgus monkey serum, and then apply the validated assays to a nonclinical study. METHODS The quantitative methods for conjugated GQ1001 and total GQ1001 were validated against regulatory guidance documents on bioanalytical method validation under a Good Laboratory Practice (GLP)-compliant environment. The validated assays were applied to a single-dose pharmacokinetic (PK) study of GQ1001 conducted in cynomolgus monkeys. RESULTS Both intra- and inter-assay precision and accuracy met the priori-defined acceptance criteria. Neither matrix effect nor hemolysis effect were observed, and the impact of specific interferents on the assays was evaluated. Dilution linearity was good with the expected dilution factors and no hook effect till up to 20.2 mg/mL of GQ1001 was noted. Besides, the stability of the ADC in monkey serum was found to be sufficient to cover the time required for sample storage and analysis. Furthermore, the assays demonstrated good parallelism determined with a study sample and good reproducibility acquired by incurred sample reanalysis (ISR). Using the validated assays, we obtained serum concentrations for the conjugated GQ1001 and the total GQ1001 in the single-dose PK study, and thereafter, evaluated their exposures over the dosing period. CONCLUSIONS All tested performance parameters of the assays met the validation acceptance criteria, which supported the application of the two assays in the nonclinical PK study and allowed the evaluation of the related PK parameters for GQ1001.
Collapse
Affiliation(s)
- Yingying Hou
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Miao
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yajun Sun
- GeneQuantum Healthcare (Suzhou) Co., Ltd., Suzhou 215000, China
| | - Lili Shi
- GeneQuantum Healthcare (Suzhou) Co., Ltd., Suzhou 215000, China
| | - Lu Ouyang
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaoqiang Chen
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ziyi Li
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Tingting Liu
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Gang Qin
- GeneQuantum Healthcare (Suzhou) Co., Ltd., Suzhou 215000, China.
| | - Qiuping Qin
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Likun Gong
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 101408, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China.
| |
Collapse
|
2
|
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
|
3
|
Greenway H, Wang J. Evaluation of High-Affinity Monoclonal Antibodies and Antibody-Drug Conjugates by Homogenous Time-Resolved FRET. ACS Med Chem Lett 2024; 15:1598-1605. [PMID: 39291004 PMCID: PMC11403740 DOI: 10.1021/acsmedchemlett.4c00317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
The rapid growth of therapeutic monoclonal antibodies demands greater accessibility to scalable methods of evaluating antigen binding. Homogenous TR-FRET is ideal for preliminary screening but has not been reported to assay these interactions due to their high-affinity and complex solution-phase kinetics. Here we report the development of a competition assay to rank-order the relative affinities of these drugs for a common antigen. The assay is compatible with automation, requires no modification of the analytes, and measures affinities as low as single-digit picomolar. We further demonstrate applications to inform the development of antibody-drug conjugates. The assay may aid discovery and manufacturing of therapeutic antibodies as a low-cost, high-throughput alternative to existing technologies.
Collapse
Affiliation(s)
- Harmon Greenway
- The Verna
and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas 77030, United States
- Center
for
NextGen Therapeutics, Baylor College of
Medicine, Houston, Texas 77030, United States
| | - Jin Wang
- The Verna
and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas 77030, United States
- Center
for
NextGen Therapeutics, Baylor College of
Medicine, Houston, Texas 77030, United States
| |
Collapse
|
4
|
Greenway H, Wang J. Evaluation of High-Affinity Monoclonal Antibodies and Antibody-Drug Conjugates by Homogenous Time-Resolved FRET. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606727. [PMID: 39149296 PMCID: PMC11326180 DOI: 10.1101/2024.08.05.606727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The rapid growth of therapeutic monoclonal antibodies demands greater accessibility to scalable methods of evaluating antigen binding. Homogenous TR-FRET is ideal for preliminary screening but has not been reported to assay these interactions due to their high-affinity and complex solution-phase kinetics. Here we report the development of a competition assay to rank-order the relative affinities of these drugs for a common antigen. The assay is compatible with automation, requires no modification of the analytes, and measures affinities as low as single-digit picomolar. We further demonstrate applications to inform the development of antibody-drug conjugates. The assay may aid discovery and manufacturing of therapeutic antibodies as a low-cost, high-throughput alternative to existing technologies.
Collapse
Affiliation(s)
- Harmon Greenway
- The Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas 77030, United States
- Center for NextGen Therapeutics, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Jin Wang
- The Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, Texas 77030, United States
- Center for NextGen Therapeutics, Baylor College of Medicine, Houston, Texas 77030, United States
| |
Collapse
|
5
|
Liao MZ, Leipold DD, Chen SC, Li Z, Kamath AV, Li C. Translational PK/PD framework for antibody-drug conjugates to inform drug discovery and development. Xenobiotica 2024; 54:543-551. [PMID: 38738473 DOI: 10.1080/00498254.2024.2351044] [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: 02/07/2024] [Accepted: 04/30/2024] [Indexed: 05/14/2024]
Abstract
ADCs represent a transformative class of medicine that combines the specificity of monoclonal antibodies with the potency of highly cytotoxic agents through linkers, aiming to enhance the therapeutic index of cytotoxic drugs. Given the complex molecular structures of ADCs, combining the molecular characteristics of small-molecule drugs and those of large-molecule biotherapeutics, there are several unique considerations when designing nonclinical-to-clinical PK/PD translation strategies.This complexity also demands a thorough understanding of the ADC's components - antibody, linker, and payload - to the overall toxicological, PK/PD, and efficacy profile. ADC development is a multidisciplinary endeavour requiring a strategic integration of nonclinical safety, pharmacology, and PK/PD modelling to translate from bench to bedside successfully.The ADC development underscores the necessity for a robust scientific foundation, leveraging advanced analytical and modelling tools to predict human responses and optimise therapeutic outcomes.This review aims to provide an ADC translational PK/PD framework by discussing unique aspects of ADC nonclinical to clinical PK translation, starting dose determination, and leveraging PK/PD modelling for human efficacious dose prediction and potential safety mitigation.
Collapse
Affiliation(s)
| | | | | | - Zao Li
- Genentech Inc, South San Francisco, CA, USA
| | | | - Chunze Li
- Genentech Inc, South San Francisco, CA, USA
| |
Collapse
|
6
|
Zhang Y, Yun X, Ouyang L, Zhang X, Gong L, Qin Q. Development of an ELISA with acidification treatment for an antibody conjugate incorporating Exatecans. Anal Biochem 2024; 690:115530. [PMID: 38570023 DOI: 10.1016/j.ab.2024.115530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/17/2024] [Accepted: 03/31/2024] [Indexed: 04/05/2024]
Abstract
The successful development of Sacituzumab Govitecan and Trastuzumab Deruxtecan has made camptothecin derivatives one of the most popular payloads for antibody-drug conjugates (ADCs). Camptothecin and its derivatives all exist in a pH-dependent equilibrium between the carboxylate and lactone forms. Such transformation may lead to differences in the ratio of the two molecular forms in calibration standards and biological matrix (bio-matrix) samples, thereby leading to inaccurate conjugated antibody results. In this study, we reported an enzyme-linked immunosorbent assay (ELISA) free of the aforementioned influence for the detection of the Exatecans-conjugated antibody (conjugated SM001) in cynomolgus monkey serum. The assay was developed by first acidifying all samples with glacial acetic acid (HAc), then performing neutralization and thereafter capturing conjugated SM001 with anti-Exatecan monoclonal antibody (mAb) and detecting it with biotinylated Nectin4 (hNectin4-Bio) and horseradish peroxidase-labeled streptavidin (SA-HRP). Results showed that all tested performance parameters met the acceptance criteria. The conjugated SM001 concentrations obtained were in parallel to but slightly lower than total antibody (TAb) throughout the pharmacokinetic (PK) study, revealing that the assay strategy implemented for conjugated SM001 measurement worked well for the elimination of interference triggered by the heterogeneous existence of the lactone and carboxylate forms of Exatecan (lactone-Exatecan and carboxylate-Exatecan).
Collapse
Affiliation(s)
- Yingying Zhang
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Pudong, Shanghai, 201203, China; Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210023, China
| | - Xi Yun
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Pudong, Shanghai, 201203, China
| | - Lu Ouyang
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Pudong, Shanghai, 201203, China
| | - Xianjing Zhang
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Pudong, Shanghai, 201203, China
| | - Likun Gong
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Pudong, Shanghai, 201203, China; Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, 210023, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 101408, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China.
| | - Qiuping Qin
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Pudong, Shanghai, 201203, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
7
|
Zarnoosheh Farahani T, Nejadmoghaddam MR, Sari S, Ghahremanzadeh R, Minai-Tehrani A. Generation of anti-SN38 antibody for loading efficacy and therapeutic monitoring of SN38-containing therapeutics. Heliyon 2024; 10:e33232. [PMID: 39021912 PMCID: PMC11253049 DOI: 10.1016/j.heliyon.2024.e33232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
SN38, one of the most potent anti-tumor analogues of the camptothecins (CPTs), has limitations in its direct formulation as an anticancer agent due to its super toxicity and poor solubility in water and pharmaceutically approved solvents. However, it has garnered significant scientific interest as a payload in conjugated nanomedicine platforms (e.g., SN-38lip, NK012, SNB-101, and ADCs) to enhance their effectiveness and safety. The development of these platforms necessitates a convenient quantitative determination of SN38 in preclinical and clinical studies, a need that our study directly addresses, offering a practical solution to a pressing problem in cancer research and drug development. This study details the meticulous process of generating poly and monoclonal antibodies (pAb and mAb) against SN38 and their application to measure the SN38 in naked and conjugated forms of SN38-conjugated ADCs. For this purpose, two haptens of SN38 were synthesized by introducing the glycine or 4-amino-4-oxobutanyol(glycine) moiety as a conjugation functional group of the SN38. IR, NMR and mass spectrometric techniques confirmed the chemical modifications of the haptens. The haptens were then conjugated to each bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH) protein. The SN38-KLH conjugates were meticulously examined for immunization and generation of pAb and mAb. The immunization efficiency, reactivity, binding affinity, specificity, and cross-reactivity of purified pAb and mAb against Irinotecan, a model for the emergence of an SN38 derivative in clinical settings, were evaluated using ELISA and western blotting (WB) techniques. Conjugation efficiency of the SN38 to the KLH was increased using 4-amino-4-oxobutanyol(glycine) moiety, as its immunization efficacy was more to generate pAb. Furthermore, only this hapten could immunized mice to generate mAb recognizing SN38 with nanomolar equilibrium affinity. Our recent findings strongly support the notion that the generated pAb employed in developing an ELISA effectively ascertains the presence of SN38 in SN38-conjugated ADC, with a test midpoint EC50 of 2.5 μg/mL. Our study's unique contribution to the field lies in the development of specific antibodies against SN38 for measuring it on ADC, a feat that has not been achieved before. These immunoassays can be readily applied to detect other SN38-conjugate therapeutic platforms, thereby enhancing their clinical knowledge translation. The affinity of both pAb and mAb also meets the acceptance criteria for quantifying SN38 in fluidic material, as well as in Therapeutic drug monitoring (TDM) studies, a crucial aspect of personalized medicine. The potential applications of the anti-SN38 antibodies extend to reducing SN38-induced systemic toxicity through an inverse targeting strategy, a novel approach that piques further interest in our findings.
Collapse
Affiliation(s)
- Tahereh Zarnoosheh Farahani
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | | | - Soyar Sari
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ramin Ghahremanzadeh
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Arash Minai-Tehrani
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| |
Collapse
|
8
|
Lami I, Wiemer AJ. Antibody-Drug Conjugates in the Pipeline for Treatment of Melanoma: Target and Pharmacokinetic Considerations. Drugs R D 2024; 24:129-144. [PMID: 38951479 PMCID: PMC11315830 DOI: 10.1007/s40268-024-00473-7] [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: 06/11/2024] [Indexed: 07/03/2024] Open
Abstract
Melanoma is an aggressive, rapidly developing form of skin cancer that affects about 22 per 100,000 individuals. Treatment options for melanoma patients are limited and typically involve surgical excision of moles and chemotherapy. Survival has been improved in recent years through targeted small molecule inhibitors and antibody-based immunotherapies. However, the long-term side effects that arise from taking chemotherapies can negatively impact the lives of patients because they lack specificity and impact healthy cells along with the cancer cells. Antibody-drug conjugates are a promising new class of drugs for the treatment of melanoma. This review focuses on the development of antibody-drug conjugates for melanoma and discusses the existing clinical trials of antibody-drug conjugates and their use as a melanoma treatment. So far, the antibody-drug conjugates have struggled from efficacy problems, with modest effects at best, leading many to be discontinued for melanoma. At the same time, conjugates such as AMT-253, targeting melanoma cell adhesion molecule, and mecbotamab vedotin targeting AXL receptor tyrosine kinase, are among the most exciting for melanoma treatment in the future.
Collapse
Affiliation(s)
- Ina Lami
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 N Eagleville Road, Storrs, CT, 06269, USA
| | - Andrew J Wiemer
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 N Eagleville Road, Storrs, CT, 06269, USA.
| |
Collapse
|
9
|
Kumari S, Raj S, Babu MA, Bhatti GK, Bhatti JS. Antibody-drug conjugates in cancer therapy: innovations, challenges, and future directions. Arch Pharm Res 2024; 47:40-65. [PMID: 38153656 DOI: 10.1007/s12272-023-01479-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/20/2023] [Indexed: 12/29/2023]
Abstract
The emergence of antibody-drug conjugates (ADCs) as a potential therapeutic avenue in cancer treatment has garnered significant attention. By combining the selective specificity of monoclonal antibodies with the cytotoxicity of drug molecules, ADCs aim to increase the therapeutic index, selectively targeting cancer cells while minimizing systemic toxicity. Various ADCs have been licensed for clinical usage, with ongoing research paving the way for additional options. However, the manufacture of ADCs faces several challenges. These include identifying suitable target antigens, enhancing antibodies, linkers, and payloads, and managing resistance mechanisms and side effects. This review focuses on the strategies to overcome these hurdles, such as site-specific conjugation techniques, novel antibody formats, and combination therapy. Our focus lies on current advancements in antibody engineering, linker technology, and cytotoxic payloads while addressing the challenges associated with ADC development. Furthermore, we explore the future potential of personalized medicine, leveraging individual patients' molecular profiles, to propel ADC treatments forward. As our understanding of the molecular mechanisms driving cancer progression continues to expand, we anticipate the development of new ADCs that offer more effective and personalized therapeutic options for cancer patients.
Collapse
Affiliation(s)
- Shivangi Kumari
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Sonam Raj
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, U.P., India
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India.
| |
Collapse
|
10
|
Püsküllüoğlu M, Rudzińska A, Pacholczak-Madej R. Antibody-drug conjugates in HER-2 negative breast cancers with poor prognosis. Biochim Biophys Acta Rev Cancer 2023; 1878:188991. [PMID: 37758021 DOI: 10.1016/j.bbcan.2023.188991] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023]
Abstract
Antibody drug conjugates (ADCs) comprise a rapidly growing class of targeted drugs that selectively deliver a cytotoxic agent to cancer cells, reducing the side effects associated with conventional chemotherapy. Breast cancer (BC) is a heterogeneous entity. The need for effective therapies for HER-2 negative BCs with poor prognosis, such as triple-negative or endocrine-resistant BC, remains unmet due to the lack of potential targets for treatments. These BC subtypes are not candidates for hormonal or anti-HER-2 agents. However, ongoing clinical trials exploring the use of ADCs with a wide range of targets have shown potential for this treatment modality. In this review, we present the current state of knowledge regarding the role of ADC and speculate on novel approaches including ADC combination therapies, new molecular targets, and the role of other subclasses of ADCs (bicycle drug conjugates, bispecific ADCs, immune modulating ADCs) in this clinical scenario.
Collapse
Affiliation(s)
- Mirosława Püsküllüoğlu
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow Branch, Garncarska Street 11, 31-115 Krakow, Poland.
| | - Agnieszka Rudzińska
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow Branch, Garncarska Street 11, 31-115 Krakow, Poland
| | - Renata Pacholczak-Madej
- Department of Anatomy, Jagiellonian University, Medical College, Kopernika Street 12, 31-034 Krakow, Poland; Department of Chemotherapy, The District Hospital, 22 Szpitalna Street, 34-200 Sucha Beskidzka, Poland
| |
Collapse
|
11
|
Implementation of Systematic Bioanalysis of Antibody–Drug Conjugates for Preclinical Pharmacokinetic Study of Ado-Trastuzumab Emtansine (T-DM1) in Rats. Pharmaceutics 2023; 15:pharmaceutics15030756. [PMID: 36986616 PMCID: PMC10056844 DOI: 10.3390/pharmaceutics15030756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/15/2023] [Accepted: 02/22/2023] [Indexed: 02/26/2023] Open
Abstract
Antibody–drug conjugates (ADCs) are composed of monoclonal antibodies covalently bound to cytotoxic drugs by a linker. They are designed to selectively bind target antigens and present a promising cancer treatment without the debilitating side effects of conventional chemotherapies. Ado-trastuzumab emtansine (T-DM1) is an ADC that received US FDA approval for the treatment of HER2-positive breast cancer. The purpose of this study was to optimize methods for the quantification of T-DM1 in rats. We optimized four analytical methods: (1) an enzyme-linked immunosorbent assay (ELISA) to quantify the total trastuzumab levels in all drug-to-antibody ratios (DARs), including DAR 0; (2) an ELISA to quantify the conjugated trastuzumab levels in all DARs except DAR 0; (3) an LC–MS/MS analysis to quantify the levels of released DM1; and (4) a bridging ELISA to quantify the level of anti-drug antibodies (ADAs) of T-DM1. We analyzed serum and plasma samples from rats injected intravenously with T-DM1 (20 mg/kg, single dose) using these optimized methods. Based on these applied analytical methods, we evaluated the quantification, pharmacokinetics, and immunogenicity of T-DM1. This study establishes the systematic bioanalysis of ADCs with validated assays, including drug stability in matrix and ADA assay, for future investigation on the efficacy and safety of ADC development.
Collapse
|
12
|
Cahuzac H, Sallustrau A, Malgorn C, Beau F, Barbe P, Babin V, Dubois S, Palazzolo A, Thai R, Correia I, Lee KB, Garcia-Argote S, Lequin O, Keck M, Nozach H, Feuillastre S, Ge X, Pieters G, Audisio D, Devel L. Monitoring In Vivo Performances of Protein-Drug Conjugates Using Site-Selective Dual Radiolabeling and Ex Vivo Digital Imaging. J Med Chem 2022; 65:6953-6968. [PMID: 35500280 PMCID: PMC9833330 DOI: 10.1021/acs.jmedchem.2c00401] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In preclinical models, the development and optimization of protein-drug conjugates require accurate determination of the plasma and tissue profiles of both the protein and its conjugated drug. To this aim, we developed a bioanalytical strategy based on dual radiolabeling and ex vivo digital imaging. By combining enzymatic and chemical reactions, we obtained homogeneous dual-labeled anti-MMP-14 Fabs (antigen-binding fragments) conjugated to monomethyl auristatin E where the protein scaffold was labeled with carbon-14 (14C) and the conjugated drug with tritium (3H). These antibody-drug conjugates with either a noncleavable or a cleavable linker were then evaluated in vivo. By combining liquid scintillation counting and ex vivo dual-isotope radio-imaging, it was possible not only to monitor both components simultaneously during their circulation phase but also to quantify accurately their amount accumulated within the different organs.
Collapse
Affiliation(s)
- Héloïse Cahuzac
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, (France)
| | - Antoine Sallustrau
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-sur-Yvette, (France)
| | - Carole Malgorn
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, (France)
| | - Fabrice Beau
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, (France)
| | - Peggy Barbe
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, (France)
| | - Victor Babin
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-sur-Yvette, (France)
| | - Steven Dubois
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, (France)
| | - Alberto Palazzolo
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-sur-Yvette, (France)
| | - Robert Thai
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, (France)
| | - Isabelle Correia
- Sorbonne Université, Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, 75005 Paris, France
| | - Ki Baek Lee
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston 1825 Pressler St, Houston TX 77030
| | - Sébastien Garcia-Argote
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-sur-Yvette, (France)
| | - Olivier Lequin
- Sorbonne Université, Ecole Normale Supérieure, PSL University, CNRS, Laboratoire des Biomolécules, LBM, 75005 Paris, France
| | - Mathilde Keck
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, (France)
| | - Hervé Nozach
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, (France)
| | - Sophie Feuillastre
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-sur-Yvette, (France)
| | - Xin Ge
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston 1825 Pressler St, Houston TX 77030
| | - Gregory Pieters
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-sur-Yvette, (France)
| | - Davide Audisio
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-sur-Yvette, (France)
| | - Laurent Devel
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SIMoS, 91191 Gif-sur-Yvette, (France),
| |
Collapse
|
13
|
Li C, Menon R, Walles M, Singh R, Upreti VV, Brackman D, Lee AJ, Endres CJ, Kumar S, Zhang D, Barletta F, Suri A, Haninzl D, Liao KH, Lalovic B, Beaumont M, Zuo P, Mayer AP, Wei D. Risk-Based Pharmacokinetic and Drug-Drug Interaction Characterization of Antibody-Drug Conjugates in Oncology Clinical Development: An International Consortium for Innovation and Quality in Pharmaceutical Development Perspective. Clin Pharmacol Ther 2021; 112:754-769. [PMID: 34657311 DOI: 10.1002/cpt.2448] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/27/2021] [Indexed: 02/01/2023]
Abstract
Antibody-drug conjugates (ADCs) represent a rapidly evolving area of drug development and hold significant promise. To date, nine ADCs have been approved by the US Food and Drug Administration (FDA). These conjugates combine the target specificity of monoclonal antibodies with the anticancer activity of small-molecule therapeutics (also referred to as payload). Due to the complex structure, three analytes, namely ADC conjugate, total antibody, and unconjugated payload, are typically quantified during drug development; however, the benefits of measuring all three analytes at later stages of clinical development are not clear. The cytotoxic payloads, upon release from the ADC, are considered to behave like small molecules. Given the relatively high potency and low systemic exposure of cytotoxic payloads, drug-drug interaction (DDI) considerations for ADCs might be different from traditional small molecule therapeutics. The International Consortium for Innovation and Quality in Pharmaceutical Development (IQ Consortium) convened an ADC working group to create an IQ ADC database that includes 26 ADCs with six unique payloads. The analysis of the ADC data in the IQ database, as well as nine approved ADCs, supports the strategy of pharmacokinetic characterization of all three analytes in early-phase development and progressively minimizing the number of analytes to be measured in the late-phase studies. The systemic concentrations of unconjugated payload are usually too low to serve as a DDI perpetrator; however, the potential for unconjugated payloads as a victim still exists. A data-driven and risk-based decision tree was developed to guide the assessment of a circulating payload as a victim of DDI.
Collapse
Affiliation(s)
- Chunze Li
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Rajeev Menon
- Department of Clinical Pharmacology and Pharmacometrics, AbbVie, Inc., North Chicago, Illinois, USA
| | - Markus Walles
- Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Renu Singh
- Global Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, Collegeville, Pennsylvania, USA.,Department of Metabolism and Pharmacokinetics, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Vijay V Upreti
- Clinical Pharmacology, Modeling and Simulation, AMGEN, South San Francisco, California, USA
| | - Deanna Brackman
- Department of Clinical Pharmacology and Pharmacometrics, AbbVie, Inc., North Chicago, Illinois, USA
| | - Anthony J Lee
- Quantitative Pharmacology and Disposition, Seagen Inc., Bothell, Washington, USA
| | | | - Seema Kumar
- Emmanuel Merck, Darmstadt Serono Research and Development Institute (A business of Merck, Darmstadt, Germany), Billerica, Massachusetts, USA
| | - Donglu Zhang
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| | - Frank Barletta
- Pre-Clinical Pharmacokinetics & Pharmacokinetics/Pharmacodynamics, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA.,Biomedicine Design, Pfizer Inc, Pearl River, New York, USA
| | - Ajit Suri
- Quantitative Clinical Pharmacology, Takeda Pharmaceuticals International Co., Cambridge, Massachusetts, USA
| | - Dominik Haninzl
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, Cambridge, Massachuetts, USA
| | - Kai H Liao
- Clinical Pharmacology, Arcus Biosciences, Hayward, California, USA.,Clinical Pharmacology, Early Clinical Development, Pfizer Inc., San Diego, California, USA
| | - Bojan Lalovic
- Modeling & Simulation Clinical Pharmacology Sciences, Eisai Inc., Woodcliff Lake, New Jersey, USA
| | - Maribel Beaumont
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Peiying Zuo
- Quantitative Pharmacology, Alexion Pharmaceuticals, Boston, Massachuetts, USA.,Pharmacometrics US, Clinical Pharmacology and Exploratory Development, Astellas Pharma Global Development, Inc., Northbrook, Illinois, USA
| | - Andrew P Mayer
- Bioanalysis, Immunogenicity & Biomarkers, In Vitro In Vivo Translation, GlaxoSmithKline Pharmaceuticals, Collegeville, Pennsylvania, USA
| | - Dong Wei
- Drug Metabolism and Pharmacokinetics, MPM NewCo., Cambridge, Massachusetts, USA.,Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Cambridge, Massachusetts, USA
| |
Collapse
|
14
|
Mayer AP, Licea-Perez H, Boram S, Pannullo KE, Kehler J, Evans CA. Overcoming challenges associated with the bioanalysis of cysteine-conjugated metabolites in the presence of antibody-drug conjugates. Bioanalysis 2021; 13:1427-1439. [PMID: 34551622 DOI: 10.4155/bio-2021-0171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Aim: Investigations have shown that for the antibody-drug conjugate (ADC) belantamab mafodotin, concentrations of the cysteine-conjugated metabolite, Cys-mcMMAF, were overestimated in the presence of the ADC during sample processing when utilizing a historical SPE method. Results: A new assay was developed utilizing an acidic protein precipitation to remove the ADC early in the extraction process, thus eliminating the risk of overestimating Cys-mcMMAF in the presence of belantamab mafodotin. In vitro experiments demonstrated a linear relationship between the concentration of belantamab mafodotin and the release of Cys-mcMMAF. Extensive stability assessments were performed to cover storage of study samples. Conclusion: This work emphasized the critical importance of understanding the performance of a bioanalytical method for free toxic payload in the presence of the ADC.
Collapse
Affiliation(s)
- Andrew P Mayer
- Bioanalysis, Immunogenicity & Biomarkers, IVIVT, GlaxoSmithKline Pharmaceuticals, 1250 S. Collegeville Rd, Collegeville, PA 19426, USA
| | - Hermes Licea-Perez
- Bioanalysis, Immunogenicity & Biomarkers, IVIVT, GlaxoSmithKline Pharmaceuticals, 1250 S. Collegeville Rd, Collegeville, PA 19426, USA
| | - Sharon Boram
- Bioanalysis, Immunogenicity & Biomarkers, IVIVT, GlaxoSmithKline Pharmaceuticals, 1250 S. Collegeville Rd, Collegeville, PA 19426, USA
| | - Kristen E Pannullo
- Non-Clinical Regulatory, GlaxoSmithKline Pharmaceuticals, 1250 S. Collegeville Rd, Collegeville, PA 19426, USA
| | - Jonathan Kehler
- Bioanalysis, Immunogenicity & Biomarkers, IVIVT, GlaxoSmithKline Pharmaceuticals, 1250 S. Collegeville Rd, Collegeville, PA 19426, USA
| | | |
Collapse
|
15
|
Ghosal S, Walker JE, Alabi CA. Predictive Platforms of Bond Cleavage and Drug Release Kinetics for Macromolecule–Drug Conjugates. Annu Rev Chem Biomol Eng 2021; 12:241-261. [DOI: 10.1146/annurev-chembioeng-091720-030636] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Macromolecule–drug conjugates (MDCs) occupy a critical niche in modern pharmaceuticals that deals with the assembly and combination of a macromolecular carrier, a drug cargo, and a linker toward the creation of effective therapeutics. Macromolecular carriers such as synthetic biocompatible polymers and proteins are often exploited for their inherent ability to improve drug circulation, prevent off-target drug cytotoxicity, and widen the therapeutic index of drugs. One of the most significant challenges in MDC design involves tuning their drug release kinetics to achieve high spatiotemporal precision. This level of control requires a thorough qualitative and quantitative understanding of the bond cleavage event. In this review, we highlight specific research findings that emphasize the importance of establishing a precise structure–function relationship for MDCs that can be used to predict their bond cleavage and drug release kinetic parameters.
Collapse
Affiliation(s)
- Souvik Ghosal
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14850, USA
| | - Javon E. Walker
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14850, USA
| | - Christopher A. Alabi
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14850, USA
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14850, USA
| |
Collapse
|
16
|
Clinical Pharmacology of Antibody-Drug Conjugates. Antibodies (Basel) 2021; 10:antib10020020. [PMID: 34063812 PMCID: PMC8161445 DOI: 10.3390/antib10020020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 12/30/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are biopharmaceutical products where a monoclonal antibody is linked to a biologically active drug (a small molecule) forming a conjugate. Since the approval of first ADC (Gemtuzumab ozogamicin (trade name: Mylotarg)) for the treatment of CD33-positive acute myelogenous leukemia, several ADCs have been developed for the treatment of cancer. The goal of an ADC as a cancer agent is to release the cytotoxic drug to kill the tumor cells without harming the normal or healthy cells. With time, it is being realized that ADCS can also be used to manage or cure other diseases such as inflammatory diseases, atherosclerosis, and bacteremia and some research in this direction is ongoing. The focus of this review is on the clinical pharmacology aspects of ADC development. From the selection of an appropriate antibody to the finished product, the entire process of the development of an ADC is a difficult and challenging task. Clinical pharmacology is one of the most important tools of drug development since this tool helps in finding the optimum dose of a product, thus preserving the safety and efficacy of the product in a patient population. Unlike other small or large molecules where only one moiety and/or metabolite(s) is generally measured for the pharmacokinetic profiling, there are several moieties that need to be measured for characterizing the PK profiles of an ADC. Therefore, knowledge and understanding of clinical pharmacology of ADCs is vital for the selection of a safe and efficacious dose in a patient population.
Collapse
|
17
|
Abedin MR, Powers K, Aiardo R, Barua D, Barua S. Antibody-drug nanoparticle induces synergistic treatment efficacies in HER2 positive breast cancer cells. Sci Rep 2021; 11:7347. [PMID: 33795712 PMCID: PMC8016985 DOI: 10.1038/s41598-021-86762-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 03/17/2021] [Indexed: 02/08/2023] Open
Abstract
Chemotherapeutic drugs suffer from non-specific binding, undesired toxicity, and poor blood circulation which contribute to poor therapeutic efficacy. In this study, antibody–drug nanoparticles (ADNs) are engineered by synthesizing pure anti-cancer drug nanorods (NRs) in the core of nanoparticles with a therapeutic monoclonal antibody, Trastuzumab on the surface of NRs for specific targeting and synergistic treatments of human epidermal growth factor receptor 2 (HER2) positive breast cancer cells. ADNs were designed by first synthesizing ~ 95 nm diameter × ~ 500 nm long paclitaxel (PTX) NRs using the nanoprecipitation method. The surface of PTXNRs was functionalized at 2′ OH nucleophilic site using carbonyldiimidazole and conjugated to TTZ through the lysine residue interaction forming PTXNR-TTZ conjugates (ADNs). The size, shape, and surface charge of ADNs were characterized using scanning electron microscopy (SEM), SEM, and zeta potential, respectively. Using fluorophore labeling and response surface analysis, the percentage conjugation efficiency was found > 95% with a PTX to TTZ mass ratio of 4 (molar ratio ≈ 682). In vitro therapeutic efficiency of PTXNR-TTZ was evaluated in two HER2 positive breast cancer cell lines: BT-474 and SK-BR-3, and a HER2 negative MDA-MB-231 breast cancer cell using MTT assay. PTXNR-TTZ inhibited > 80% of BT-474 and SK-BR-3 cells at a higher efficiency than individual PTX and TTZ treatments alone after 72 h. A combination index analysis indicated a synergistic combination of PTXNR-TTZ compared with the doses of single-drug treatment. Relatively lower cytotoxicity was observed in MCF-10A human breast epithelial cell control. The molecular mechanisms of PTXNR-TTZ were investigated using cell cycle and Western blot analyses. The cell cycle analysis showed PTXNR-TTZ arrested > 80% of BT-474 breast cancer cells in the G2/M phase, while > 70% of untreated cells were found in the G0/G1 phase indicating that G2/M arrest induced apoptosis. A similar percentage of G2/M arrested cells was found to induce caspase-dependent apoptosis in PTXNR-TTZ treated BT-474 cells as revealed using Western blot analysis. PTXNR-TTZ treated BT-474 cells showed ~ 1.3, 1.4, and 1.6-fold higher expressions of cleaved caspase-9, cytochrome C, and cleaved caspase-3, respectively than untreated cells, indicating up-regulation of caspase-dependent activation of apoptotic pathways. The PTXNR-TTZ ADN represents a novel nanoparticle design that holds promise for targeted and efficient anti-cancer therapy by selective targeting and cancer cell death via apoptosis and mitotic cell cycle arrest.
Collapse
Affiliation(s)
- Muhammad Raisul Abedin
- Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, 110 Bertelsmeyer Hall, 1101 N. State Street, Rolla, MO, 65409-1230, USA
| | - Kaitlyne Powers
- Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, 110 Bertelsmeyer Hall, 1101 N. State Street, Rolla, MO, 65409-1230, USA
| | - Rachel Aiardo
- Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, 110 Bertelsmeyer Hall, 1101 N. State Street, Rolla, MO, 65409-1230, USA
| | - Dibbya Barua
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO, 65409, USA
| | - Sutapa Barua
- Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, 110 Bertelsmeyer Hall, 1101 N. State Street, Rolla, MO, 65409-1230, USA.
| |
Collapse
|
18
|
Clinical pharmacology strategies in supporting drug development and approval of antibody-drug conjugates in oncology. Cancer Chemother Pharmacol 2021; 87:743-765. [PMID: 33792763 PMCID: PMC8110483 DOI: 10.1007/s00280-021-04250-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/18/2021] [Indexed: 11/12/2022]
Abstract
Antibody–drug conjugates (ADCs) are important molecular entities in the treatment of cancer. These conjugates combine the target specificity of monoclonal antibodies with the potent anti-cancer activity of small-molecule therapeutics. The complex structure of ADCs poses unique challenges to characterize the drug’s pharmacokinetics (PKs) and pharmacodynamics (PDs) since it requires a quantitative understanding of the PK and PD properties of multiple different molecular species (e.g., ADC conjugate, total antibody and unconjugated cytotoxic drug). As a result, clinical pharmacology strategy of an ADC is rather unique and dependent on the linker/cytotoxic drug technology, heterogeneity of the ADC, PK and safety/efficacy profile of the specific ADC in clinical development. In this review, we summarize the clinical pharmacology strategies in supporting development and approval of ADCs using the approved ADCs as specific examples to illustrate the customized approach to clinical pharmacology assessments in their clinical development.
Collapse
|
19
|
Li C, Zhang C, Li Z, Samineni D, Lu D, Wang B, Chen SC, Zhang R, Agarwal P, Fine BM, Girish S. Clinical pharmacology of vc-MMAE antibody-drug conjugates in cancer patients: learning from eight first-in-human Phase 1 studies. MAbs 2021; 12:1699768. [PMID: 31852341 PMCID: PMC6927763 DOI: 10.1080/19420862.2019.1699768] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
vc-MMAE antibody–drug conjugates (ADCs) consist of a monoclonal antibody (mAb) covalently bound with a potent anti-mitotic toxin (MMAE) through a protease-labile valine-citrulline (vc) linker. The objective of this study was to characterize the pharmacokinetics (PK) and explore exposure–response relationships of eight vc-MMAE ADCs, against different targets and for diverse tumor indications, using data from eight first-in-human Phase 1 studies. PK parameters of the three analytes, namely antibody-conjugated MMAE (acMMAE), total antibody, and unconjugated MMAE, were estimated using non-compartmental approaches and compared across the eight vc-MMAE ADCs. Relationships between analytes were assessed by linear regression. Exposure–response relationships were explored with key efficacy (objective response rate) and safety (Grade 2+ peripheral neuropathy) endpoints. PK profiles of acMMAE, total antibody and unconjugated MMAE following the first dose of 2.4 mg/kg were comparable across the eight ADCs; the exposure differences between molecules were small relative to the inter-subject variability. acMMAE exposure was strongly correlated with total antibody exposure for all the eight ADCs, but such correlation was less evident between acMMAE and unconjugated MMAE exposure. For multiple ADCs evaluated, efficacy and safety endpoints appeared to correlate well with acMMAE exposure, but not with unconjugated MMAE over the doses tested. PK of vc-MMAE ADCs was well characterized and demonstrated remarkable similarity at 2.4 mg/kg across the eight ADCs. Results from analyte correlation and exposure–response relationship analyses suggest that measurement of acMMAE analyte alone might be adequate for vc-MMAE ADCs to support the clinical pharmacology strategy used during late-stage clinical development.
Collapse
Affiliation(s)
- Chunze Li
- Genentech Research & Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Cindy Zhang
- Genentech Research & Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Zao Li
- Genentech Research & Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Divya Samineni
- Genentech Research & Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Dan Lu
- Genentech Research & Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Bei Wang
- Genentech Research & Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Shang-Chiung Chen
- Genentech Research & Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Rong Zhang
- Genentech Research & Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Priya Agarwal
- Genentech Research & Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Bernard M Fine
- Genentech Research & Early Development, Genentech, Inc, South San Francisco, CA, USA
| | - Sandhya Girish
- Genentech Research & Early Development, Genentech, Inc, South San Francisco, CA, USA
| |
Collapse
|
20
|
Cahuzac H, Devel L. Analytical Methods for the Detection and Quantification of ADCs in Biological Matrices. Pharmaceuticals (Basel) 2020; 13:ph13120462. [PMID: 33327644 PMCID: PMC7765153 DOI: 10.3390/ph13120462] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/01/2020] [Accepted: 12/11/2020] [Indexed: 12/27/2022] Open
Abstract
Understanding pharmacokinetics and biodistribution of antibody–drug conjugates (ADCs) is a one of the critical steps enabling their successful development and optimization. Their complex structure combining large and small molecule characteristics brought out multiple bioanalytical methods to decipher the behavior and fate of both components in vivo. In this respect, these methods must provide insights into different key elements including half-life and blood stability of the construct, premature release of the drug, whole-body biodistribution, and amount of the drug accumulated within the targeted pathological tissues, all of them being directly related to efficacy and safety of the ADC. In this review, we will focus on the main strategies enabling to quantify and characterize ADCs in biological matrices and discuss their associated technical challenges and current limitations.
Collapse
|
21
|
Lee MV, Kaur S, Saad OM. Conjugation Site Influences Antibody-Conjugated Drug PK Assays: Case Studies for Disulfide-Linked, Self-Immolating Next-Generation Antibody Drug Conjugates. Anal Chem 2020; 92:12168-12175. [PMID: 32786429 DOI: 10.1021/acs.analchem.0c00773] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Immunoaffinity (IA) LC-MS/MS pharmacokinetic (PK) assays are widely used in the field for antibody drug conjugates (ADCs) containing peptide linkers that are enzymatically cleavable, such as MC-ValCit-PAB. Conjugate PK assay strategies for these ADCs involve cleavage with cathepsin B or papain to release and measure the antibody-conjugated drug (acDrug) concentration. However, robust acDrug PK methods for disulfide-linked self-immolating ADCs are lacking as they are a different conjugation modality. We developed acDrug PK assays for next-generation disulfide-linked ADCs involving immunoaffinity capture, chemical cleavage, and LC-MS/MS. Disulfide-linked ADCs captured from plasma were chemically reduced at basic pH to release the linker-drug, followed by self-immolation to liberate the active drug, and quantified by MRM LC-MS/MS. Herein, we detail the development and optimization of this chemical cleavage acDrug PK assay, resulting in robust accuracy and precision (±20%). The conjugation site of the linker-drug on the antibody was found to affect the kinetics of drug release. Multiple biophysical and chemical characteristics, such as tertiary structure, fractional solvent accessibility, pKa of the conjugation site, surrounding residue's pI, and electrostatic charge, may directly impact the drug release kinetics. Similar site-specific stability has been previously reported for ADCs in vivo. The assay development and qualification data for this original assay format are presented along with its application to multiple in vitro and in vivo studies across species.
Collapse
Affiliation(s)
- M Violet Lee
- Department of Bioanalytical Sciences, Assay Development and Technology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Surinder Kaur
- Department of Bioanalytical Sciences, Assay Development and Technology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Ola M Saad
- Department of Bioanalytical Sciences, Assay Development and Technology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
22
|
Barfield RM, Kim YC, Chuprakov S, Zhang F, Bauzon M, Ogunkoya AO, Yeo D, Hickle C, Pegram MD, Rabuka D, Drake PM. A Novel HER2-targeted Antibody-drug Conjugate Offers the Possibility of Clinical Dosing at Trastuzumab-equivalent Exposure Levels. Mol Cancer Ther 2020; 19:1866-1874. [PMID: 32651200 DOI: 10.1158/1535-7163.mct-20-0190] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/24/2020] [Accepted: 07/02/2020] [Indexed: 11/16/2022]
Abstract
Trastuzumab and the related ADC, ado-trastuzumab emtansine (T-DM1), both target HER2-overexpressing cells. Together, these drugs have treatment indications in both early-stage and metastatic settings for HER2+ breast cancer. T-DM1 retains the antibody functionalities of trastuzumab and adds the potency of a cytotoxic maytansine payload. Interestingly, in the clinic, T-DM1 cannot always replace the use of trastuzumab plus chemotherapy administered together as single agents. We hypothesize that this failure may be due, in part, to the limited systemic exposure achieved by T-DM1 relative to trastuzumab because of toxicity-related dosing constraints on the ADC. We have developed a trastuzumab-based ADC site specifically conjugated to maytansine through a noncleavable linker. This construct, termed CAT-01-106, has a drug-to-antibody ratio (DAR) of 1.8, approximately half the average DAR of T-DM1, which comprises a mixture of antibodies variously conjugated with DARs ranging from 0 to 8. The high DAR species present in T-DM1 contribute to its toxicity and limit its clinical dose. CAT-01-106 showed superior in vivo efficacy compared with T-DM1 at equal payload dosing and was equally or better tolerated compared with T-DM1 at equal payload dosing up to 120 mg/kg in Sprague-Dawley rats and 60 mg/kg in cynomolgus monkeys. CAT-01-106 also showed improved pharmacokinetics in rats relative to T-DM1, with 40% higher ADC exposure levels. Together, the data suggest that CAT-01-106 may be sufficiently tolerable to enable clinical dosing at trastuzumab-equivalent exposure levels, combining the functions of both the antibody and the payload in one drug and potentially improving patient outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Mark D Pegram
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | | | | |
Collapse
|
23
|
Zhu X, Huo S, Xue C, An B, Qu J. Current LC-MS-based strategies for characterization and quantification of antibody-drug conjugates. J Pharm Anal 2020; 10:209-220. [PMID: 32612867 PMCID: PMC7322744 DOI: 10.1016/j.jpha.2020.05.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 05/21/2020] [Accepted: 05/21/2020] [Indexed: 01/28/2023] Open
Abstract
The past few years have witnessed enormous progresses in the development of antibody-drug conjugates (ADCs). Consequently, comprehensive analysis of ADCs in biological systems is critical in supporting discovery, development and evaluation of these agents. Liquid chromatography-mass spectrometry (LC-MS) has emerged as a promising and versatile tool for ADC analysis across a wide range of scenarios, owing to its multiplexing ability, rapid method development, as well as the capability of analyzing a variety of targets ranging from small-molecule payloads to the intact protein with a high, molecular resolution. However, despite this tremendous potential, challenges persist due to the high complexity in both the ADC molecules and the related biological systems. This review summarizes the up-to-date LC-MS-based strategies in ADC analysis and discusses the challenges and opportunities in this rapidly-evolving field.
Collapse
Affiliation(s)
- Xiaoyu Zhu
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
- New York State Center of Excellence in Bioinformatics & Life Sciences, Buffalo, NY, 14203, USA
| | - Shihan Huo
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
- New York State Center of Excellence in Bioinformatics & Life Sciences, Buffalo, NY, 14203, USA
| | - Chao Xue
- New York State Center of Excellence in Bioinformatics & Life Sciences, Buffalo, NY, 14203, USA
- Department of Chemical and Biological Engineering, School of Engineering and Applied Science, University at Buffalo, State University of New York, Buffalo, NY, 14260, USA
| | - Bo An
- Exploratory Biomarker, In-vitro/In-vivo Translation, R&D Research, GlaxoSmithKline Pharmaceuticals, 1250 South Collegeville Rd, Collegeville, PA, 19426, USA
| | - Jun Qu
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
- New York State Center of Excellence in Bioinformatics & Life Sciences, Buffalo, NY, 14203, USA
| |
Collapse
|
24
|
Pharmacokinetic/pharmacodynamic relationship of therapeutic monoclonal antibodies used in oncology: Part 1, monoclonal antibodies, antibody-drug conjugates and bispecific T-cell engagers. Eur J Cancer 2020; 128:107-118. [DOI: 10.1016/j.ejca.2020.01.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/02/2020] [Accepted: 01/07/2020] [Indexed: 01/31/2023]
|
25
|
Hayat SMG, Sahebkar A. Antibody-drug conjugates: smart weapons against cancer. Arch Med Sci 2020; 16:1257-1262. [PMID: 32864020 PMCID: PMC7444717 DOI: 10.5114/aoms.2019.83020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 03/08/2017] [Indexed: 01/28/2023] Open
Affiliation(s)
- Seyed Mohammad Gheibi Hayat
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Science, Yazd, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
26
|
Antibody-Drug Conjugates: Pharmacokinetic/Pharmacodynamic Modeling, Preclinical Characterization, Clinical Studies, and Lessons Learned. Clin Pharmacokinet 2019; 57:687-703. [PMID: 29188435 DOI: 10.1007/s40262-017-0619-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Antibody-drug conjugates are an emerging class of biopharmaceuticals changing the landscape of targeted chemotherapy. These conjugates combine the target specificity of monoclonal antibodies with the anti-cancer activity of small-molecule therapeutics. Several antibody-drug conjugates have received approval for the treatment of various types of cancer including gemtuzumab ozogamicin (Mylotarg®), brentuximab vedotin (Adcetris®), trastuzumab emtansine (Kadcyla®), and inotuzumab ozogamicin, which recently received approval (Besponsa®). In addition to these approved therapies, there are many antibody-drug conjugates in the drug development pipeline and in clinical trials, although these fall outside the scope of this article. Understanding the pharmacokinetics and pharmacodynamics of antibody-drug conjugates and the development of pharmacokinetic/pharmacodynamic models is indispensable, albeit challenging as there are many parameters to incorporate including the disposition of the intact antibody-drug conjugate complex, the antibody, and the drug agents following their dissociation in the body. In this review, we discuss how antibody-drug conjugates progressed over time, the challenges in their development, and how our understanding of their pharmacokinetics/pharmacodynamics led to greater strides towards successful targeted therapy programs.
Collapse
|
27
|
Li C, Zhang C, Deng R, Leipold D, Li D, Latifi B, Gao Y, Zhang C, Li Z, Miles D, Chen SC, Samineni D, Wang B, Agarwal P, Lu D, Prabhu S, Girish S, Kamath AV. Prediction of Human Pharmacokinetics of Antibody-Drug Conjugates From Nonclinical Data. Clin Transl Sci 2019; 12:534-544. [PMID: 31115997 PMCID: PMC6742937 DOI: 10.1111/cts.12649] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/26/2019] [Indexed: 12/16/2022] Open
Abstract
Prediction of human pharmacokinetics (PK) based on preclinical information for antibody–drug conjugates (ADCs) provide important insight into first‐in‐human (FIH) study design. This retrospective analysis was conducted to identify an appropriate scaling method to predict human PK for ADCs from animal PK data in the linear range. Different methods for projecting human clearance (CL) from animal PK data for 11 ADCs exhibiting linear PK over the tested dose ranges were examined: multiple species allometric scaling (CL vs. body weight), allometric scaling with correction factors, allometric scaling based on rule of exponent, and scaling from only cynomolgus monkey PK data. Two analytes of interest for ADCs, namely total antibody and conjugate (measured as conjugated drug or conjugated antibody), were assessed. Percentage prediction errors (PEs) and residual sum of squares (RSS) were compared across methods. Human CL was best estimated using cynomolgus monkey PK data alone and an allometric scaling exponent of 1.0 for CL. This was consistently observed for both conjugate and total antibody analytes. Other scaling methods either underestimated or overestimated human CL, or produced larger average absolute PEs and RSS. Human concentration‐time profiles were also reasonably predicted from the cynomolgus monkey data using species‐invariant time method with a fixed exponent of 1.0 for CL and 1.0 for volume of distribution. In conclusion, results from this retrospective analysis of 11 ADCs indicate that allometric scaling of CL with an exponent of 1.0 using cynomolgus monkey PK data alone can successfully project human PK profiles of an ADC within linear range.
Collapse
Affiliation(s)
- Chunze Li
- Genentech Inc., South San Francisco, California, USA
| | - Cindy Zhang
- Genentech Inc., South San Francisco, California, USA
| | - Rong Deng
- Genentech Inc., South San Francisco, California, USA
| | | | - Dongwei Li
- Genentech Inc., South San Francisco, California, USA
| | | | - Yuying Gao
- Certara USA, Inc., Menlo Park, California, USA
| | - Crystal Zhang
- Genentech Inc., South San Francisco, California, USA
| | - Zao Li
- Genentech Inc., South San Francisco, California, USA
| | - Dale Miles
- Genentech Inc., South San Francisco, California, USA
| | | | | | - Bei Wang
- Genentech Inc., South San Francisco, California, USA
| | - Priya Agarwal
- Genentech Inc., South San Francisco, California, USA
| | - Dan Lu
- Genentech Inc., South San Francisco, California, USA
| | | | | | | |
Collapse
|
28
|
Chitosan oligosaccharide (COS): An overview. Int J Biol Macromol 2019; 129:827-843. [DOI: 10.1016/j.ijbiomac.2019.01.192] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/14/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023]
|
29
|
ADME Considerations and Bioanalytical Strategies for Pharmacokinetic Assessments of Antibody-Drug Conjugates. Antibodies (Basel) 2018; 7:antib7040041. [PMID: 31544891 PMCID: PMC6698957 DOI: 10.3390/antib7040041] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 12/19/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a unique class of biotherapeutics of inherent heterogeneity and correspondingly complex absorption, distribution, metabolism, and excretion (ADME) properties. Herein, we consider the contribution of various components of ADCs such as various classes of warheads, linkers, and conjugation strategies on ADME of ADCs. Understanding the metabolism and disposition of ADCs and interpreting exposure-efficacy and exposure-safety relationships of ADCs in the context of their various catabolites is critical for design and subsequent development of a clinically successful ADCs. Sophisticated bioanalytical assays are required for the assessments of intact ADC, total antibody, released warhead and relevant metabolites. Both ligand-binding assays (LBA) and hybrid LBA-liquid chromatography coupled with tandem mass spectrometry (LBA-LC-MS/MS) methods have been employed to assess pharmacokinetics (PK) of ADCs. Future advances in bioanalytical techniques will need to address the rising complexity of this biotherapeutic modality as more innovative conjugation strategies, antibody scaffolds and novel classes of warheads are employed for the next generation of ADCs. This review reflects our considerations on ADME of ADCs and provides a perspective on the current bioanalytical strategies for pharmacokinetic assessments of ADCs.
Collapse
|
30
|
Hu XY, Wang R, Jin J, Liu XJ, Cui AL, Sun LQ, Li YP, Li Y, Wang YC, Zhen YS, Miao QF, Li ZR. An EGFR-targeting antibody-drug conjugate LR004-VC-MMAE: potential in esophageal squamous cell carcinoma and other malignancies. Mol Oncol 2018; 13:246-263. [PMID: 30372581 PMCID: PMC6360372 DOI: 10.1002/1878-0261.12400] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/27/2018] [Accepted: 10/16/2018] [Indexed: 12/18/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) is a rational target for cancer therapy, because its overexpression plays an important oncogenic role in a variety of solid tumors; however, EGFR‐targeted antibody–drug conjugate (ADC) therapy for esophageal squamous cell carcinoma (ESCC) is exceedingly rare. LR004 is a novel anti‐EGFR antibody with the advantages of improved safety and fewer hypersensitivity reactions. It may be of great value as a carrier in ADCs with high binding affinity and internalization ability. Here, we prepared an EGFR‐targeting ADC, LR004‐VC‐MMAE, and evaluated its antitumor activities against ESCC and EGFR‐positive cells. LR004 was covalently conjugated with monomethyl auristatin E (MMAE) via a VC linker by antibody interchain disulfide bond reduction. VC‐MMAE was conjugated with LR004 with approximately 4.0 MMAE molecules per ADC. LR004‐VC‐MMAE showed a potent antitumor effect against ESCC and other EGFR‐positive cells with IC50 values of nM concentrations in vitro. The in vivo antitumor effects of LR004‐VC‐MMAE were investigated in ESCC KYSE520 and A431 xenograft nude mice models. Significant activity was seen at 5 mg·kg−1, and complete tumor regression was observed at 15 mg·kg−1 in the KYSE520 xenograft nude mice after four injections, while the naked antibody LR004 had little effect on inhibiting tumor growth. Similar promising results were obtained in the A431 models. In addition, the tumors also remained responsive to LR004‐VC‐MMAE for large tumor experiments (tumor volume 400–500 mm3). The study results demonstrated that LR004‐VC‐MMAE could be a potential therapeutic agent for ESCC and other EGFR‐expressing malignancies. We also evaluated PK profile of LR004‐VC‐MMAE ADC in the mice model, which would provide qualitative guiding significance for the further research.
Collapse
Affiliation(s)
- Xin-Yue Hu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Rong Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jie Jin
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiu-Jun Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - A-Long Cui
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lian-Qi Sun
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yan-Ping Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yi Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yu-Cheng Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yong-Su Zhen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Qing-Fang Miao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhuo-Rong Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
31
|
Vezina HE, Cotreau M, Han TH, Gupta M. Antibody-Drug Conjugates as Cancer Therapeutics: Past, Present, and Future. J Clin Pharmacol 2018; 57 Suppl 10:S11-S25. [PMID: 28921650 DOI: 10.1002/jcph.981] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/19/2017] [Indexed: 12/22/2022]
Abstract
Antibody-drug conjugates (ADCs) represent an innovative therapeutic approach that provides novel treatment options and hope for patients with cancer. By coupling monoclonal antibodies (mAbs) to cytotoxic small-molecule payloads with a plasma-stable linker, ADCs offer the potential for increased drug specificity and fewer off-target effects than systemic chemotherapy. As evidence for the potential of these therapies, many new ADCs are in various stages of clinical development. Because their structure poses unique challenges to pharmacokinetic and pharmacodynamic characterization, it is critical to recognize the differences between ADCs and conventional chemotherapy in the design of ADC clinical development strategies. Although some properties may be determined mainly by either the mAb or the small-molecule portion, the behavior of these agents is not always predictable. Furthermore, because the absorption, distribution, metabolism, and excretion (ADME) of ADCs are influenced by all 3 of its components (mAb, linker, and payload), it is important to characterize the intact molecule, any target-mediated catabolic clearance of the mAb, and the ADME properties of the small-molecule payload. Here we describe key issues in the clinical development of ADCs, including considerations for designing first-in-human studies for ADCs. We discuss some difficulties of ADC pharmacokinetic characterization and current approaches to overcoming these challenges. Finally, we consider all aspects of clinical pharmacology assessment required during drug development, using examples from the literature to illustrate the discussion.
Collapse
Affiliation(s)
| | | | - Tae H Han
- AbbVie Stemcentrx LLC, South San Francisco, CA, USA
| | | |
Collapse
|
32
|
Dan N, Setua S, Kashyap VK, Khan S, Jaggi M, Yallapu MM, Chauhan SC. Antibody-Drug Conjugates for Cancer Therapy: Chemistry to Clinical Implications. Pharmaceuticals (Basel) 2018; 11:ph11020032. [PMID: 29642542 PMCID: PMC6027311 DOI: 10.3390/ph11020032] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/02/2018] [Accepted: 04/03/2018] [Indexed: 01/01/2023] Open
Abstract
Chemotherapy is one of the major therapeutic options for cancer treatment. Chemotherapy is often associated with a low therapeutic window due to its poor specificity towards tumor cells/tissues. Antibody-drug conjugate (ADC) technology may provide a potentially new therapeutic solution for cancer treatment. ADC technology uses an antibody-mediated delivery of cytotoxic drugs to the tumors in a targeted manner, while sparing normal cells. Such a targeted approach can improve the tumor-to-normal tissue selectivity and specificity in chemotherapy. Considering its importance in cancer treatment, we aim to review recent efforts for the design and development of ADCs. ADCs are mainly composed of an antibody, a cytotoxic payload, and a linker, which can offer selectivity against tumors, anti-cancer activity, and stability in systemic circulation. Therefore, we have reviewed recent updates and principal considerations behind ADC designs, which are not only based on the identification of target antigen, cytotoxic drug, and linker, but also on the drug-linker chemistry and conjugation site at the antibody. Our review focuses on site-specific conjugation methods for producing homogenous ADCs with constant drug-antibody ratio (DAR) in order to tackle several drawbacks that exists in conventional conjugation methods.
Collapse
Affiliation(s)
- Nirnoy Dan
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Saini Setua
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Vivek K Kashyap
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Sheema Khan
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Meena Jaggi
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences and Cancer Research Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
33
|
A Polar Sulfamide Spacer Significantly Enhances the Manufacturability, Stability, and Therapeutic Index of Antibody-Drug Conjugates. Antibodies (Basel) 2018; 7:antib7010012. [PMID: 31544864 PMCID: PMC6698870 DOI: 10.3390/antib7010012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/08/2018] [Accepted: 02/17/2018] [Indexed: 11/17/2022] Open
Abstract
Despite tremendous efforts in the field of targeted cancer therapy with antibody–drug conjugates (ADCs), attrition rates have been high. Historically, the priority in ADC development has been the selection of target, antibody, and toxin, with little focus on the nature of the linker. We show here that a short and polar sulfamide spacer (HydraSpace™, Oss, The Netherlands) positively impacts ADC properties in various ways: (a) efficiency of conjugation; (b) stability; and (c) therapeutic index. Different ADC formats are explored in terms of drug-to-antibody ratios (DAR2, DAR4) and we describe the generation of a DAR4 ADC by site-specific attachment of a bivalent linker–payload construct to a single conjugation site in the antibody. A head-to-head comparison of HydraSpace™-containing DAR4 ADCs to marketed drugs, derived from the same antibody and toxic payload components, indicated a significant improvement in both the efficacy and safety of several vivo models, corroborated by in-depth pharmacokinetic analysis. Taken together, HydraSpace™ technology based on a polar sulfamide spacer provides significant improvement in manufacturability, stability, and ADC design, and is a powerful platform to enable next-generation ADCs with enhanced therapeutic index.
Collapse
|
34
|
Braun AC, Gutmann M, Lühmann T, Meinel L. Bioorthogonal strategies for site-directed decoration of biomaterials with therapeutic proteins. J Control Release 2018; 273:68-85. [PMID: 29360478 DOI: 10.1016/j.jconrel.2018.01.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/16/2018] [Accepted: 01/17/2018] [Indexed: 01/04/2023]
Abstract
Emerging strategies targeting site-specific protein modifications allow for unprecedented selectivity, fast kinetics and mild reaction conditions with high yield. These advances open exciting novel possibilities for the effective bioorthogonal decoration of biomaterials with therapeutic proteins. Site-specificity is particularly important to the therapeutics' end and translated by targeting specific functional groups or introducing new functional groups into the therapeutic at predefined positions. Biomimetic strategies are designed for modification of therapeutics emulating enzymatic strategies found in Nature. These strategies are suitable for a diverse range of applications - not only for protein-polymer conjugation, particle decoration and surface immobilization, but also for the decoration of complex biomaterials and the synthesis of bioresponsive drug delivery systems. This article reviews latest chemical and enzymatic strategies for the biorthogonal decoration of biomaterials with therapeutic proteins and inter-positioned linker structures. Finally, the numerous reports at the interface of biomaterials, linkers, and therapeutic protein decoration are integrated into practical advice for design considerations intended to support the selection of productive ligation strategies.
Collapse
Affiliation(s)
- Alexandra C Braun
- Institute for Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, DE-97074 Würzburg, Germany
| | - Marcus Gutmann
- Institute for Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, DE-97074 Würzburg, Germany
| | - Tessa Lühmann
- Institute for Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, DE-97074 Würzburg, Germany
| | - Lorenz Meinel
- Institute for Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, DE-97074 Würzburg, Germany.
| |
Collapse
|
35
|
Singh AP, Shah DK. Measurement and Mathematical Characterization of Cell-Level Pharmacokinetics of Antibody-Drug Conjugates: A Case Study with Trastuzumab-vc-MMAE. Drug Metab Dispos 2017; 45:1120-1132. [PMID: 28821484 DOI: 10.1124/dmd.117.076414] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/11/2017] [Indexed: 12/12/2022] Open
Abstract
The main objective of this work was to understand and mathematically characterize the cellular disposition of a tool antibody-drug conjugate (ADC), trastuzumab-valine-citrulline-monomethyl auristatin E (T-vc-MMAE). Toward this goal, three different analytical methods were developed to measure the concentrations of different ADC-related analytes in the media and cell lysate. A liquid chromatography-tandem mass spectrometry method was developed to quantify unconjugated drug (i.e., MMAE) concentrations, a forced deconjugation method was developed to quantify total drug concentrations, and an enzyme-linked immunosorbent assay method was developed to quantify total antibody (i.e., trastuzumab) concentrations. Cellular disposition studies were conducted in low-HER2-(GFP-MCF7) and high-HER2-expressing (N87) cell lines, following continuous or 2-hour exposure to MMAE and T-vc-MMAE. Similar intracellular accumulation of MMAE was observed between two cell lines following incubation with plain MMAE. However, when incubated with T-vc-MMAE, much higher intracellular exposures of unconjugated drug, total drug, and total antibody were observed in N87 cells compared with GFP-MCF7 cells. A novel single-cell disposition model was developed to simultaneously characterize in vitro pharmacokinetics of all three analytes of the ADC in the media and cellular space. The model was able to characterize all the data well and provided robust estimates of MMAE influx rate, MMAE efflux rate, and intracellular degradation rate for T-vc-MMAE. ADC internalization and degradation rates, HER2 expression, and MMAE efflux rate were found to be the key parameters responsible for intracellular exposure to MMAE, on the basis of a global sensitivity analysis. The single-cell pharmacokinetics model for ADCs presented here is expected to provide a better framework for characterizing bystander effect of ADCs.
Collapse
Affiliation(s)
- Aman P Singh
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, New York
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
36
|
McNiff ML, Chadwick JS. Metal-bound claMP Tag inhibits proteolytic cleavage. Protein Eng Des Sel 2017; 30:467-475. [PMID: 28541524 DOI: 10.1093/protein/gzx030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 05/05/2017] [Indexed: 11/13/2022] Open
Abstract
Biologics can be an improvement to small molecule drugs, providing high specificity for an identified target, lowering toxicity and limiting side effects. To achieve effective delivery, the biologic must have sufficient time to reach the target tissue. A prolonged half-life in the circulating environment is desired, but often serum stability is limited by proteases. Proteolysis in the serum causes degradation and inactivation as the biologic is fragmented and more rapidly cleared from the body. To improve the circulating half-life, large, hydrophilic polymers may be conjugated or stable fusion tags may be engineered to increase the effective size of the peptide and to hinder degradation by proteases. Improved resistance to proteases is essential for effective delivery. Here, a proof of concept study is presented using a metal-binding tripeptide tag known as the claMP Tag to create an inline conjugate and the ability of the tag to inhibit proteolysis was examined.
Collapse
Affiliation(s)
- Michaela L McNiff
- Department of Pharmaceutical Chemistry, University of Kansas, 2093 Constant Ave, Lawrence, KS 66047
| | - Jennifer S Chadwick
- Department of Pharmaceutical Chemistry, University of Kansas, 2093 Constant Ave, Lawrence, KS 66047.,BioAnalytix Inc., 790 Memorial Dr., Cambridge, MA 02139
| |
Collapse
|
37
|
Mariathasan S, Tan MW. Antibody-Antibiotic Conjugates: A Novel Therapeutic Platform against Bacterial Infections. Trends Mol Med 2017; 23:135-149. [PMID: 28126271 DOI: 10.1016/j.molmed.2016.12.008] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/15/2016] [Accepted: 12/16/2016] [Indexed: 11/26/2022]
Abstract
Antibodies are potent components of the immune repertoire and have been successfully exploited to treat bacterial infections. Recently an antibody-antibiotic conjugate (AAC) that combines key attributes of an antibody and antibiotic has been shown to be efficacious against Staphylococcus aureus infection. An AAC has three components: an antibiotic payload to kill bacteria, an antibody to target delivery of the payload to bacteria, and a linker attaching the payload to the antibody. With increasing understanding of the biology and pathophysiology of S. aureus, this article highlights how this knowledge has led to the design principles of an efficacious AAC, and discusses how the AAC platform could be translationally applied to treat other perilous infectious diseases.
Collapse
Affiliation(s)
- Sanjeev Mariathasan
- Department of Late-Stage Oncology Biomarkers Development, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Man-Wah Tan
- Department of Infectious Diseases, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
38
|
Wang YJ, Li YY, Liu XY, Lu XL, Cao X, Jiao BH. Marine Antibody-Drug Conjugates: Design Strategies and Research Progress. Mar Drugs 2017; 15:E18. [PMID: 28098746 PMCID: PMC5295238 DOI: 10.3390/md15010018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/30/2016] [Accepted: 01/04/2017] [Indexed: 01/22/2023] Open
Abstract
Antibody-drug conjugates (ADCs), constructed with monoclonal antibodies (mAbs), linkers, and natural cytotoxins, are innovative drugs developed for oncotherapy. Owing to the distinctive advantages of both chemotherapy drugs and antibody drugs, ADCs have obtained enormous success during the past several years. The development of highly specific antibodies, novel marine toxins' applications, and innovative linker technologies all accelerate the rapid R&D of ADCs. Meanwhile, some challenges remain to be solved for future ADCs. For instance, varieties of site-specific conjugation have been proposed for solving the inhomogeneity of DARs (Drug Antibody Ratios). In this review, the usages of various natural toxins, especially marine cytotoxins, and the development strategies for ADCs in the past decade are summarized. Representative ADCs with marine cytotoxins in the pipeline are introduced and characterized with their new features, while perspective comments for future ADCs are proposed.
Collapse
Affiliation(s)
- Yu-Jie Wang
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai 200433, China.
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China.
| | - Yu-Yan Li
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiao-Yu Liu
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai 200433, China.
| | - Xiao-Ling Lu
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai 200433, China.
| | - Xin Cao
- Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China.
| | - Bing-Hua Jiao
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
39
|
LC-MS/MS method for the simultaneous determination of Lys-MCC-DM1, MCC-DM1 and DM1 as potential intracellular catabolites of the antibody-drug conjugate trastuzumab emtansine (T-DM1). J Pharm Biomed Anal 2017; 137:170-177. [PMID: 28131055 DOI: 10.1016/j.jpba.2017.01.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/04/2017] [Accepted: 01/06/2017] [Indexed: 01/14/2023]
Abstract
Lysine-MCC-DM1, MCC-DM1 and DM1 are potential catabolites of trastuzumab emtansine (T-DM1). A convenient liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed and validated to detect these catabolites simultaneously in in vitro investigations for the first time. Protein precipitation was utilized to prepare the samples. Chromatographic separation was achieved on a Phenomenex Kinetex C18 column (100×2.1mm, 2.6μm) with mobile-phase gradient elution. The calibration curves of each analyte ranging from 1 to 100nM showed good linearity (r2>0.995). The method was validated successfully and applied to the intracellular catabolism and regulation of T-DM1.
Collapse
|
40
|
Coming-of-Age of Antibodies in Cancer Therapeutics. Trends Pharmacol Sci 2016; 37:1009-1028. [DOI: 10.1016/j.tips.2016.09.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 09/07/2016] [Accepted: 09/09/2016] [Indexed: 02/07/2023]
|
41
|
Stagg NJ, Shen BQ, Brunstein F, Li C, Kamath AV, Zhong F, Schutten M, Fine BM. Peripheral neuropathy with microtubule inhibitor containing antibody drug conjugates: Challenges and perspectives in translatability from nonclinical toxicology studies to the clinic. Regul Toxicol Pharmacol 2016; 82:1-13. [PMID: 27773754 DOI: 10.1016/j.yrtph.2016.10.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/15/2016] [Accepted: 10/18/2016] [Indexed: 11/18/2022]
Abstract
Antibody drug conjugates (ADC) consist of potent cytotoxic drugs conjugated to antibodies via chemical linkers, which enables specific targeting of tumor cells while reducing systemic exposure to the cytotoxic drug and improving the therapeutic window. The valine citrulline monomethyl auristatin E (vcMMAE, conventional linker-drug) ADC platform has shown promising clinical activity in several cancers, but peripheral neuropathy (PN) is a frequent adverse event leading to treatment discontinuation and dose reduction. This was not predicted based on nonclinical toxicology studies in monkeys or rats treated with vcMMAE ADCs. We evaluated four hypotheses for the lack of translatability of PN with vcMMAE ADCs: 1) species differences in exposure; 2) insensitivity of animal models; 3) species differences in target biology and other vcMMAE ADC properties in peripheral nerves and 4) increased susceptibility of patient population. The result of this hypothesis-based approach identified opportunities to improve the predictivity of PN in our animal models by increasing duration of exposure and adding an expanded neurohistopathology assessment of peripheral nerves. The utility of a predictive animal model would be to provide possible mitigation strategies in the clinic with vcMMAE ADCs and help to screen the next generation microtubule inhibitor (MTI) ADCs for reduced PN.
Collapse
Affiliation(s)
- Nicola J Stagg
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Ben-Quan Shen
- Department of Preclinical & Translational Pharmacokinetics & Pharmacodynamics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Flavia Brunstein
- Drug Safety, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Chunze Li
- Clinical Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Amrita V Kamath
- Department of Preclinical & Translational Pharmacokinetics & Pharmacodynamics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Fiona Zhong
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Melissa Schutten
- Safety Assessment, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Bernard M Fine
- Clinical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
42
|
Terwisscha van Scheltinga AGT, Ogasawara A, Pacheco G, Vanderbilt AN, Tinianow JN, Gupta N, Li D, Firestein R, Marik J, Scales SJ, Williams SP. Preclinical Efficacy of an Antibody-Drug Conjugate Targeting Mesothelin Correlates with Quantitative 89Zr-ImmunoPET. Mol Cancer Ther 2016; 16:134-142. [PMID: 27760836 DOI: 10.1158/1535-7163.mct-16-0449] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/27/2016] [Accepted: 09/29/2016] [Indexed: 11/16/2022]
Abstract
Antibody-drug conjugates (ADC) use monoclonal antibodies (mAb) as vehicles to deliver potent cytotoxic drugs selectively to tumor cells expressing the target. Molecular imaging with zirconium-89 (89Zr)-labeled mAbs recapitulates similar targeting biology and might help predict the efficacy of these ADCs. An anti-mesothelin antibody (AMA, MMOT0530A) was used to make comparisons between its efficacy as an ADC and its tumor uptake as measured by 89Zr immunoPET imaging. Mesothelin-targeted tumor growth inhibition by monomethyl auristatin E (MMAE), ADC AMA-MMAE (DMOT4039A), was measured in mice bearing xenografts of ovarian cancer OVCAR-3×2.1, pancreatic cancers Capan-2, HPAC, AsPC-1, and HPAF-II, or mesothelioma MSTO-211H. Ex vivo analysis of mesothelin expression was performed using immunohistochemistry. AMA-MMAE showed the greatest growth inhibition in OVCAR-3×2.1, Capan-2, and HPAC tumors, which showed target-specific tumor uptake of 89Zr-AMA. The less responsive xenografts (AsPC-1, HPAF-II, and MSTO-211H) did not show 89Zr-AMA uptake despite confirmed mesothelin expression. ImmunoPET can demonstrate the necessary delivery, binding, and internalization of an ADC antibody in vivo and this correlates with the efficacy of mesothelin-targeted ADC in tumors vulnerable to the cytotoxic drug delivered. Mol Cancer Ther; 16(1); 134-42. ©2016 AACR.
Collapse
Affiliation(s)
- Anton G T Terwisscha van Scheltinga
- Genentech Research and Early Development, Genentech, Inc,. South San Francisco, California.,Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Annie Ogasawara
- Genentech Research and Early Development, Genentech, Inc,. South San Francisco, California
| | - Glenn Pacheco
- Genentech Research and Early Development, Genentech, Inc,. South San Francisco, California
| | - Alexander N Vanderbilt
- Genentech Research and Early Development, Genentech, Inc,. South San Francisco, California
| | - Jeff N Tinianow
- Genentech Research and Early Development, Genentech, Inc,. South San Francisco, California
| | - Nidhi Gupta
- Genentech Research and Early Development, Genentech, Inc,. South San Francisco, California
| | - Dongwei Li
- Genentech Research and Early Development, Genentech, Inc,. South San Francisco, California
| | - Ron Firestein
- Genentech Research and Early Development, Genentech, Inc,. South San Francisco, California
| | - Jan Marik
- Genentech Research and Early Development, Genentech, Inc,. South San Francisco, California
| | - Suzie J Scales
- Genentech Research and Early Development, Genentech, Inc,. South San Francisco, California
| | - Simon-Peter Williams
- Genentech Research and Early Development, Genentech, Inc,. South San Francisco, California.
| |
Collapse
|
43
|
Singh AP, Sharma S, Shah DK. Quantitative characterization of in vitro bystander effect of antibody-drug conjugates. J Pharmacokinet Pharmacodyn 2016; 43:567-582. [PMID: 27670282 DOI: 10.1007/s10928-016-9495-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 09/21/2016] [Indexed: 11/28/2022]
Abstract
Antibody-drug conjugates (ADCs) are designed to target antigen expressing (Ag+) cells in a tumor. Once processed by the Ag+ cells, ADCs can release cytotoxic drug molecules that can diffuse out of Ag+ cells into the neighboring antigen-negative (Ag-) cells to induce their cytotoxicity. This additional efficacy of ADCs on Ag- cells in the presence of Ag+ cells is known as the 'bystander effect'. Although the importance of this phenomena is widely acknowledged for effective killing of a heterogeneous tumor, the rate and extent of the bystander killing in a heterogeneous system is not quantitatively understood yet. Thus, the objectives of this manuscript were to: (1) synthesize and characterize a tool ADC Trastuzumab-vc-MMAE that is capable of exhibiting bystander effect, (2) quantify the time course of the bystander effect for the tool ADC using in vitro co-culture systems created using mixture of various HER2-expressing cell lines, and (3) develop a pharmacodynamic (PD) model that is capable of characterizing the bystander effect of ADCs. Co-culture studies conducted using GFP labelled MCF7 cells as Ag- cells and N87, BT474, and SKBR3 as Ag+ cells revealed that the bystander effect of ADC increases with increasing fraction of Ag+ cells in a co-culture system, and with increased expression level of target on Ag+ cells. A notable lag time after ADC incubation was also observed prior to significant bystander killing of Ag- cells. Based on our results we hypothesize that there may be other determinants apart from the antigen expression level that can also influence the ability of Ag+ cells to demonstrate the bystander effect in a co-culture system. The co-culture analysis also suggested that the bystander effect of the ADC can dissipate over the period of time as the population of Ag+ cells declines. A novel PD model was developed to mathematically characterize the bystander effect of ADCs by combining two different cell distribution models to represent the population of Ag+ and Ag- cells in a co-culture system. This PD model can be integrated with the systems PK model for ADCs in the future to generate a quantitative framework that is capable of supporting the discovery and development of novel ADCs with optimal bystander killing capabilities.
Collapse
Affiliation(s)
- Aman P Singh
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Kapoor Hall, Buffalo, NY, 14214-8033, USA
| | - Sharad Sharma
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Kapoor Hall, Buffalo, NY, 14214-8033, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Kapoor Hall, Buffalo, NY, 14214-8033, USA.
| |
Collapse
|
44
|
Giddabasappa A, Gupta VR, Norberg R, Gupta P, Spilker ME, Wentland J, Rago B, Eswaraka J, Leal M, Sapra P. Biodistribution and Targeting of Anti-5T4 Antibody-Drug Conjugate Using Fluorescence Molecular Tomography. Mol Cancer Ther 2016; 15:2530-2540. [PMID: 27466353 DOI: 10.1158/1535-7163.mct-15-1012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 07/06/2016] [Indexed: 11/16/2022]
Abstract
Understanding a drug's whole-body biodistribution and tumor targeting can provide important information regarding efficacy, safety, and dosing parameters. Current methods to evaluate biodistribution include in vivo imaging technologies like positron electron tomography and single-photon emission computed tomography or ex vivo quantitation of drug concentrations in tissues using autoradiography and standard biochemical assays. These methods use radioactive compounds or are cumbersome and do not give whole-body information. Here, for the first time, we show the utility of fluorescence molecular tomography (FMT) imaging to determine the biodistribution and targeting of an antibody-drug conjugate (ADC). An anti-5T4-antibody (5T4-Ab) and 5T4-ADC were conjugated with a near-infrared (NIR) fluorophore VivoTag 680XL (VT680). Both conjugated compounds were stable as determined by SEC-HPLC and plasma stability studies. Flow cytometry and fluorescence microscopy studies showed that VT680-conjugated 5T4-ADC specifically bound 5T4-expressing cells in vitro and also exhibited a similar cytotoxicity profile as the unconjugated 5T4-ADC. In vivo biodistribution and tumor targeting in an H1975 subcutaneous xenograft model demonstrated no significant differences between accumulation of VT680-conjugated 5T4-Ab or 5T4-ADC in either normal tissues or tumor. In addition, quantitation of heart signal from FMT imaging showed good correlation with the plasma pharmacokinetic profile suggesting that it (heart FMT imaging) may be a surrogate for plasma drug clearance. These results demonstrate that conjugation of VT680 to 5T4-Ab or 5T4-ADC does not change the behavior of native biologic, and FMT imaging can be a useful tool to understand biodistribution and tumor-targeting kinetics of antibodies, ADCs, and other biologics. Mol Cancer Ther; 15(10); 2530-40. ©2016 AACR.
Collapse
Affiliation(s)
- Anand Giddabasappa
- Global Science & Technology - Comparative Medicine, Pfizer Inc., San Diego, California.
| | - Vijay R Gupta
- Global Science & Technology - Comparative Medicine, Pfizer Inc., San Diego, California
| | - Rand Norberg
- Global Science & Technology - Comparative Medicine, Pfizer Inc., San Diego, California
| | - Parul Gupta
- Global Science & Technology - Comparative Medicine, Pfizer Inc., San Diego, California
| | - Mary E Spilker
- Pharmacokinetics Dynamics and Metabolism, Pfizer Inc., San Diego, California
| | - Joann Wentland
- Pharmacokinetics Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut
| | - Brian Rago
- Pharmacokinetics Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut
| | - Jeetendra Eswaraka
- Global Science & Technology - Comparative Medicine, Pfizer Inc., San Diego, California
| | - Mauricio Leal
- Pharmacokinetics Dynamics and Metabolism, Pfizer Inc., Pearl River, New York
| | - Puja Sapra
- Oncology Research Unit, Pfizer Inc., Pearl River, New York
| |
Collapse
|
45
|
Antibody–drug conjugate bioanalysis using LB-LC–MS/MS hybrid assays: strategies, methodology and correlation to ligand-binding assays. Bioanalysis 2016; 8:1383-401. [DOI: 10.4155/bio-2016-0017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: Antibody–drug conjugates (ADCs) are complex drug constructs with multiple species in the heterogeneous mixture that contribute to their efficacy and toxicity. The bioanalysis of ADCs involves multiple assays and analytical platforms. Methods: A series of ligand binding and LC–MS/MS (LB-LC–MS/MS) hybrid assays, through different combinations of anti-idiotype (anti-Id), anti-payload, or generic capture reagents, and cathepsin-B or trypsin enzyme digestion, were developed and evaluated for the analysis of conjugated-payload as well as for species traditionally measured by ligand-binding assays, total-antibody and conjugated-antibody. Results & conclusion: Hybrid assays are complementary or viable alternatives to ligand-binding assay for ADC bioanalysis and PK/PD modeling. The fit-for-purpose choice of analytes, assays and platforms and an integrated strategy from Discovery to Development for ADC PK and bioanalysis are recommended.
Collapse
|
46
|
Chromatography-based methods for determining molar extinction coefficients of cytotoxic payload drugs and drug antibody ratios of antibody drug conjugates. J Chromatogr A 2016; 1455:133-139. [DOI: 10.1016/j.chroma.2016.05.086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/24/2016] [Accepted: 05/25/2016] [Indexed: 01/02/2023]
|
47
|
Gould J, Callis CM, Dolan DG, Stanard B, Weideman PA. Special endpoint and product specific considerations in pharmaceutical acceptable daily exposure derivation. Regul Toxicol Pharmacol 2016; 79 Suppl 1:S79-93. [PMID: 27233924 DOI: 10.1016/j.yrtph.2016.05.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/19/2016] [Indexed: 12/12/2022]
Abstract
Recently, a guideline has been published by the European Medicines Agency (EMA) on setting safe limits, permitted daily exposures (PDE) [also called acceptable daily exposures (ADE)], for medicines manufactured in multi-product facilities. The ADE provides a safe exposure limit for inadvertent exposure of a drug due to cross-contamination in manufacturing. The ADE determination encompasses a standard risk assessment, requiring an understanding of the toxicological and pharmacological effects, the mechanism of action, drug compound class, and the dose-response as well as the pharmacokinetic properties of the compound. While the ADE concept has broad application in pharmaceutical safety there are also nuances and specific challenges associated with some toxicological endpoints or drug product categories. In this manuscript we discuss considerations for setting ADEs when the following specific adverse health endpoints may constitute the critical effect: genotoxicity, developmental and reproductive toxicity (DART), and immune system modulation (immunostimulation or immunosuppression), and for specific drug classes, including antibody drug conjugates (ADCs), emerging medicinal therapeutic compounds, and compounds with limited datasets. These are challenging toxicological scenarios that require a careful evaluation of all of the available information in order to establish a health-based safe level.
Collapse
|
48
|
Singh AP, Maass KF, Betts AM, Wittrup KD, Kulkarni C, King LE, Khot A, Shah DK. Evolution of Antibody-Drug Conjugate Tumor Disposition Model to Predict Preclinical Tumor Pharmacokinetics of Trastuzumab-Emtansine (T-DM1). AAPS JOURNAL 2016; 18:861-75. [PMID: 27029797 DOI: 10.1208/s12248-016-9904-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 03/08/2016] [Indexed: 01/17/2023]
Abstract
A mathematical model capable of accurately characterizing intracellular disposition of ADCs is essential for a priori predicting unconjugated drug concentrations inside the tumor. Towards this goal, the objectives of this manuscript were to: (1) evolve previously published cellular disposition model of ADC with more intracellular details to characterize the disposition of T-DM1 in different HER2 expressing cell lines, (2) integrate the improved cellular model with the ADC tumor disposition model to a priori predict DM1 concentrations in a preclinical tumor model, and (3) identify prominent pathways and sensitive parameters associated with intracellular activation of ADCs. The cellular disposition model was augmented by incorporating intracellular ADC degradation and passive diffusion of unconjugated drug across tumor cells. Different biomeasures and chemomeasures for T-DM1, quantified in the companion manuscript, were incorporated into the modified model of ADC to characterize in vitro pharmacokinetics of T-DM1 in three HER2+ cell lines. When the cellular model was integrated with the tumor disposition model, the model was able to a priori predict tumor DM1 concentrations in xenograft mice. Pathway analysis suggested different contribution of antigen-mediated and passive diffusion pathways for intracellular unconjugated drug exposure between in vitro and in vivo systems. Global and local sensitivity analyses revealed that non-specific deconjugation and passive diffusion of the drug across tumor cell membrane are key parameters for drug exposure inside a cell. Finally, a systems pharmacokinetic model for intracellular processing of ADCs has been proposed to highlight our current understanding about the determinants of ADC activation inside a cell.
Collapse
Affiliation(s)
- Aman P Singh
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Kapoor Hall, Buffalo, New York, 14214-8033, USA
| | - Katie F Maass
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,David H. Koch Institute of Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Alison M Betts
- Translational Research Group, Department of Pharmacokinetics Dynamics and Metabolism, Pfizer, Groton, Connecticut, USA
| | - K Dane Wittrup
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,David H. Koch Institute of Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Chethana Kulkarni
- Oncology Medicinal Chemistry, Worldwide Medicinal Chemistry, Pfizer, Groton, Connecticut, USA
| | - Lindsay E King
- Translational Research Group, Department of Pharmacokinetics Dynamics and Metabolism, Pfizer, Groton, Connecticut, USA
| | - Antari Khot
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Kapoor Hall, Buffalo, New York, 14214-8033, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Kapoor Hall, Buffalo, New York, 14214-8033, USA.
| |
Collapse
|
49
|
Ross PL, Wolfe JL. Physical and Chemical Stability of Antibody Drug Conjugates: Current Status. J Pharm Sci 2016; 105:391-397. [DOI: 10.1016/j.xphs.2015.11.037] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/13/2015] [Accepted: 11/17/2015] [Indexed: 01/24/2023]
|
50
|
Kraynov E, Kamath AV, Walles M, Tarcsa E, Deslandes A, Iyer RA, Datta-Mannan A, Sriraman P, Bairlein M, Yang JJ, Barfield M, Xiao G, Escandon E, Wang W, Rock DA, Chemuturi NV, Moore DJ. Current Approaches for Absorption, Distribution, Metabolism, and Excretion Characterization of Antibody-Drug Conjugates: An Industry White Paper. ACTA ACUST UNITED AC 2015; 44:617-23. [PMID: 26669328 DOI: 10.1124/dmd.115.068049] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 12/14/2015] [Indexed: 11/22/2022]
Abstract
An antibody-drug conjugate (ADC) is a unique therapeutic modality composed of a highly potent drug molecule conjugated to a monoclonal antibody. As the number of ADCs in various stages of nonclinical and clinical development has been increasing, pharmaceutical companies have been exploring diverse approaches to understanding the disposition of ADCs. To identify the key absorption, distribution, metabolism, and excretion (ADME) issues worth examining when developing an ADC and to find optimal scientifically based approaches to evaluate ADC ADME, the International Consortium for Innovation and Quality in Pharmaceutical Development launched an ADC ADME working group in early 2014. This white paper contains observations from the working group and provides an initial framework on issues and approaches to consider when evaluating the ADME of ADCs.
Collapse
Affiliation(s)
- Eugenia Kraynov
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Amrita V Kamath
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Markus Walles
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Edit Tarcsa
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Antoine Deslandes
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Ramaswamy A Iyer
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Amita Datta-Mannan
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Priya Sriraman
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Michaela Bairlein
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Johnny J Yang
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Matthew Barfield
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Guangqing Xiao
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Enrique Escandon
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Weirong Wang
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Dan A Rock
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Nagendra V Chemuturi
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - David J Moore
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| |
Collapse
|