1
|
Li W, Huang W, Yu X, Chen C, Yuan Y, Liu D, Wang F, Yu J, Diao X. A validated LC-MS/MS method for the quantitation of daratumumab in rat serum using rapid tryptic digestion without IgG purification and reduction. J Pharm Biomed Anal 2024; 243:116083. [PMID: 38447348 DOI: 10.1016/j.jpba.2024.116083] [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: 12/21/2023] [Revised: 02/15/2024] [Accepted: 03/01/2024] [Indexed: 03/08/2024]
Abstract
Daratumumab, a humanized monoclonal antibody utilized in treating immunoglobulin light-chain amyloidosis and relapsed/refractory multiple myeloma, was quantified in rat serum through a simple, economical and effective liquid chromatography tandem-mass spectrometry (LC-MS/MS) method. A surrogate peptide, LLIYDASNR, derived from trypsin hydrolysis, was quantitatively analyzed with LLIYDASN [13C6, 15N4] RAT as an internal standard. This corrected variations from sample pretreatment and mass spectrometry response, involving denaturation and trypsin hydrolysis in a two-step process lasting approximately 1 hour. Methodological validation demonstrated a linear range of 1 µg/mL to 1000 µg/mL in rat serum. Precision, accuracy, matrix effect, sensitivity, stability, selectivity, carryover, and interference met acceptance criteria. The validated LC-MS/MS approach was successfully applied to a pharmacokinetic study of daratumumab in rats at an intravenous dose of 15 mg/kg.
Collapse
Affiliation(s)
- Weiqiang Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wensi Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiong Yu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Chong Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yali Yuan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Donghui Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Feiyu Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinghua Yu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Xingxing Diao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
2
|
Werth EG, Roos D, Philip ET. Immunocapture LC-MS methods for pharmacokinetics of large molecule drugs. Bioanalysis 2024; 16:165-177. [PMID: 38348660 DOI: 10.4155/bio-2023-0261] [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] [Indexed: 03/26/2024] Open
Abstract
Implementation of immunocapture LC-MS methods to characterize the pharmacokinetic profile of large molecule drugs has become a widely used technique over the past decade. As the pharmaceutical industry strives for speediness into clinical development without jeopardizing quality, robust assays with generic application across the pipeline are becoming instrumental in bioanalysis, especially in early-stage development. This review highlights the capabilities and challenges involved in hybrid immunocapture LC-MS techniques and its continued applications in nonclinical and clinical pharmacokinetic assay design. This includes a comparison of LC-MS-based approaches to conventional ligand-binding assays and the driving demands in large molecule drug portfolios including growing sensitivity requirements and the unique challenges of new modalities requiring innovation in the bioanalytical laboratory.
Collapse
Affiliation(s)
- Emily G Werth
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT 06877, USA
| | - David Roos
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT 06877, USA
| | - Elsy T Philip
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT 06877, USA
| |
Collapse
|
3
|
Li X, Wang Y, Hu W, Song Q, Ding L. Development and validation of pharmacokinetics assays for a novel HER2-targeting antibody-drug conjugate (SHR-A1201): Application to its dose-escalation pharmacokinetic study. J Pharm Biomed Anal 2024; 240:115964. [PMID: 38219442 DOI: 10.1016/j.jpba.2024.115964] [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: 11/27/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/16/2024]
Abstract
Approximately 25% of breast cancer patients with HER2 overexpression tend to have a high risk of disease progression and death. Various HER2-targeting therapies have been approved for treatment. Recently, a novel antibody-drug conjugate, SHR-A1201, is being researched and developed. For the pharmacokinetic study of SHR-A1201, suitable bioanalytical methods are needed for quantifying unconjugated cytotoxin, cytotoxin-conjugated antibodies and total antibodies. In this research, bioanalytical methods involving a highly sensitive LC-MS/MS assay for unconjugated cytotoxic payload DM1 in human plasma, ELISA strategies for DM1-conjugated trastuzumab and total trastuzumab in human serum were developed, validated and successfully applied to a phase I dose-escalation pharmacokinetic study of SHR-A1201. The pharmacokinetic properties and exposure-to-dose proportionality was evaluated for SHR-A1201. According to the bioanalytical method validation guidance, the bioanalytical methods were fully validated and the validation results met the acceptance criteria. The nonspecific binding of DM1 and dimer was avoided for the LC-MS/MS assay. In the dose-escalation pharmacokinetic study of SHR-A1201, a potential dose-proportional pharmacokinetics was observed over the dose from 1.2 mg/kg to 4.8 mg/kg. The validated bioanalytical strategies are robust and reproducible and these bioanalytical methods will contribute to better understanding of the pharmacokinetic properties of SHR-A1201.
Collapse
Affiliation(s)
- Xianjing Li
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmaceutical Analysis, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Yiya Wang
- Nanjing Clinical Tech Laboratories Inc., 18 Zhilan Road, Jiangning District, Nanjing 211100, China
| | - Wenhui Hu
- Nanjing Jiening Pharmaceutical Technology, 18 Zhilan Road, Jiangning District, Nanjing 211100, China
| | - Qinxin Song
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmaceutical Analysis, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.
| | - Li Ding
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Nanjing Clinical Tech Laboratories Inc., 18 Zhilan Road, Jiangning District, Nanjing 211100, China; Nanjing Jiening Pharmaceutical Technology, 18 Zhilan Road, Jiangning District, Nanjing 211100, China; Department of Pharmaceutical Analysis, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.
| |
Collapse
|
4
|
Beaumont K, Pike A, Davies M, Savoca A, Vasalou C, Harlfinger S, Ramsden D, Ferguson D, Hariparsad N, Jones O, McGinnity D. ADME and DMPK considerations for the discovery and development of antibody drug conjugates (ADCs). Xenobiotica 2022; 52:770-785. [PMID: 36314242 DOI: 10.1080/00498254.2022.2141667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The therapeutic concept of antibody drug conjugates (ADCs) is to selectively target tumour cells with small molecule cytotoxic drugs to maximise cell kill benefit and minimise healthy tissue toxicity.An ADC generally consists of an antibody that targets a protein on the surface of tumour cells chemically linked to a warhead small molecule cytotoxic drug.To deliver the warhead to the tumour cell, the antibody must bind to the target protein and in general be internalised into the cell. Following internalisation, the cytotoxic agent can be released in the endosomal or lysosomal compartment (via different mechanisms). Diffusion or transport out of the endosome or lysosome allows the cytotoxic drug to express its cell-killing pharmacology. Alternatively, some ADCs (e.g. EDB-ADCs) rely on extracellular cleavage releasing membrane permeable warheads.One potentially important aspect of the ADC mechanism is the 'bystander effect' whereby the cytotoxic drug released in the targeted cell can diffuse out of that cell and into other (non-target expressing) tumour cells to exert its cytotoxic effect. This is important as solid tumours tend to be heterogeneous and not all cells in a tumour will express the targeted protein.The combination of large and small molecule aspects in an ADC poses significant challenges to the disposition scientist in describing the ADME properties of the entire molecule.This article will review the ADC landscape and the ADME properties of successful ADCs, with the aim of outlining best practice and providing a perspective of how the field can further facilitate the discovery and development of these important therapeutic modalities.
Collapse
Affiliation(s)
- Kevin Beaumont
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, Cambridge, UK
| | - Andy Pike
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, Cambridge, UK
| | - Michael Davies
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, Cambridge, UK
| | - Adriana Savoca
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, Cambridge, UK
| | - Christina Vasalou
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, AstraZeneca, Boston, MA, USA
| | - Steffi Harlfinger
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, Cambridge, UK
| | - Diane Ramsden
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, AstraZeneca, Boston, MA, USA
| | - Douglas Ferguson
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, AstraZeneca, Boston, MA, USA
| | - Niresh Hariparsad
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, AstraZeneca, Boston, MA, USA
| | - Owen Jones
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, Cambridge, UK
| | - Dermot McGinnity
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, Cambridge, UK
| |
Collapse
|
5
|
Conlon PR, Gurijala VR, Kaufman M, Li D, Li J, Li Y, Yin M, Reddy BS, Wagler T, Wang Z, Xu Z, Yurkovetskiy AV, Zhu L. Process Development and GMP Production of a Conjugate Warhead: Auristatin F-HPA-Ala/TFA (XMT-1864/TFA). Org Process Res Dev 2022. [DOI: 10.1021/acs.oprd.1c00449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Patrick R. Conlon
- Former Mersana Therapeutics, Inc., 840 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Venu Reddy Gurijala
- Drug Substance Development, Mersana Therapeutics, Inc., 840 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Michael Kaufman
- Former Mersana Therapeutics, Inc., 840 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Dachang Li
- Chemical Macromolecule Division, Asymchem Life Science (Tianjin) Co., Ltd. No. 71, Seventh Avenue, TEDA Tianjin 300457, P.R. China
| | - Jiuyuan Li
- Chemical Macromolecule Division, Asymchem Life Science (Tianjin) Co., Ltd. No. 71, Seventh Avenue, TEDA Tianjin 300457, P.R. China
| | - Yuanyuan Li
- Drug Substance Development, Mersana Therapeutics, Inc., 840 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Mao Yin
- Former Mersana Therapeutics, Inc., 840 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Bollu Satyanarayan Reddy
- Drug Substance Development, Mersana Therapeutics, Inc., 840 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Thomas Wagler
- Drug Substance Development, Mersana Therapeutics, Inc., 840 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Zedong Wang
- Chemical Macromolecule Division, Asymchem Life Science (Tianjin) Co., Ltd. No. 71, Seventh Avenue, TEDA Tianjin 300457, P.R. China
| | - Zhongmin Xu
- Chemdiscover, 10 Carlton Circle, Princeton, New Jersey 08540, United States
| | - Aleksandr V. Yurkovetskiy
- Former Mersana Therapeutics, Inc., 840 Memorial Drive, Cambridge, Massachusetts 02139, United States
| | - Lei Zhu
- Drug Substance Development, Mersana Therapeutics, Inc., 840 Memorial Drive, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
6
|
Petrilli R, Pinheiro DP, de Cássia Evangelista de Oliveira F, Galvão GF, Marques LGA, Lopez RFV, Pessoa C, Eloy JO. Immunoconjugates for Cancer Targeting: A Review of Antibody-Drug Conjugates and Antibody-Functionalized Nanoparticles. Curr Med Chem 2021; 28:2485-2520. [PMID: 32484100 DOI: 10.2174/0929867327666200525161359] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/07/2020] [Accepted: 04/16/2020] [Indexed: 11/22/2022]
Abstract
Targeted therapy has been recently highlighted due to the reduction of side effects and improvement in overall efficacy and survival from different types of cancers. Considering the approval of many monoclonal antibodies in the last twenty years, cancer treatment can be accomplished by the combination of monoclonal antibodies and small molecule chemotherapeutics. Thus, strategies to combine both drugs in a single administration system are relevant in the clinic. In this context, two strategies are possible and will be further discussed in this review: antibody-drug conjugates (ADCs) and antibody-functionalized nanoparticles. First, it is important to better understand the possible molecular targets for cancer therapy, addressing different antigens that can selectively bind to antibodies. After selecting the best target, ADCs can be prepared by attaching a cytotoxic drug to an antibody able to target a cancer cell antigen. Briefly, an ADC will be formed by a monoclonal antibody (MAb), a cytotoxic molecule (cytotoxin) and a chemical linker. Usually, surface-exposed lysine or the thiol group of cysteine residues are used as anchor sites for linker-drug molecules. Another strategy that should be considered is antibody-functionalized nanoparticles. Basically, liposomes, polymeric and inorganic nanoparticles can be attached to specific antibodies for targeted therapy. Different conjugation strategies can be used, but nanoparticles coupling between maleimide and thiolated antibodies or activation with the addition of ethyl-3-(3-dimethyl aminopropyl) carbodiimide (EDC)/ N-hydroxysuccinimide (NHS) (1:5) and further addition of the antibody are some of the most used strategies. Herein, molecular targets and conjugation strategies will be presented and discussed to better understand the in vitro and in vivo applications presented. Also, the clinical development of ADCs and antibody-conjugated nanoparticles are addressed in the clinical development section. Finally, due to the innovation related to the targeted therapy, it is convenient to analyze the impact on patenting and technology. Information related to the temporal evolution of the number of patents, distribution of patent holders and also the number of patents related to cancer types are presented and discussed. Thus, our aim is to provide an overview of the recent developments in immunoconjugates for cancer targeting and highlight the most important aspects for clinical relevance and innovation.
Collapse
Affiliation(s)
- Raquel Petrilli
- University for International Integration of the Afro-Brazilian Lusophony, Institute of Health Sciences, Ceara, Brazil
| | - Daniel Pascoalino Pinheiro
- Federal University of Ceara, College of Medicine, Department of Physiology and Pharmacology, Fortaleza, Ceara, Brazil
| | | | - Gabriela Fávero Galvão
- School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Av. Cafe s/n, Ribeirao Preto, SP, Brazil
| | - Lana Grasiela Alves Marques
- Institute of Communication and Scientific and Technological Information in Health, Oswaldo Cruz Foundation - FIOCRUZ, Rio de Janeiro, Brazil
| | - Renata Fonseca Vianna Lopez
- School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Av. Cafe s/n, Ribeirao Preto, SP, Brazil
| | - Claudia Pessoa
- Federal University of Ceara, College of Medicine, Department of Physiology and Pharmacology, Fortaleza, Ceara, Brazil
| | - Josimar O Eloy
- Federal University of Ceará, College of Pharmacy, Dentistry and Nursing, Department of Pharmacy, Fortaleza, Ceara, Brazil
| |
Collapse
|
7
|
Clardy SM, Lee DH, Schuck P. Determining the Stoichiometry of a Protein-Polymer Conjugate Using Multisignal Sedimentation Velocity Analytical Ultracentrifugation. Bioconjug Chem 2021; 32:942-949. [PMID: 33848127 DOI: 10.1021/acs.bioconjchem.1c00095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Advances in polymer science have broadened the applications of protein-polymer conjugates as biopharmaceuticals and biotechnology reagents. The complex nature of these conjugates makes characterization challenging. Here, we describe the use of multisignal sedimentation velocity analytical ultracentrifugation to measure the polymer-to-protein ratio. To demonstrate the principle, we applied this approach to a series of antibody-drug conjugates with different polymer-to-protein ratios and various degrees of heterogeneity, and validated results with orthogonal analytical techniques. We found that multisignal sedimentation velocity can provide accurate information on key attributes including polymer-to-protein ratio, which is important for maximizing the therapeutic potential of future protein-polymer conjugates.
Collapse
Affiliation(s)
- Susan M Clardy
- Mersana Therapeutics Inc., Cambridge, Massachusetts 02139, United States
| | - David H Lee
- Mersana Therapeutics Inc., Cambridge, Massachusetts 02139, United States
| | - Peter Schuck
- Dynamics of Macromolecular Assembly Section, Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
8
|
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
|