1
|
Hashii N, Haruyama Y, Hirayama R, Kajita R, Kishino Y, Mochizuki T, Inoue K, Goda R, Hoshino M, Kuroiwa I, Aikawa H, Ueda N, Nagumo K, Oda Y, Saito Y, Ishii-Watabe A. Versatile intact LC-MS method for evaluating the drug-antibody ratio and drug load distribution of antibody-drug conjugates in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1258:124608. [PMID: 40267607 DOI: 10.1016/j.jchromb.2025.124608] [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: 02/08/2025] [Revised: 04/04/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
The average drug-antibody ratio (DAR) and drug load distribution (DLD) of an antibody-drug conjugate (ADC) can be altered by biotransformation after administration. In addition, drug loading affects the clearance and exposure of the ADC. Evaluating alterations in the average DAR and DLD of an ADC in vivo would provide valuable information to better understand of the pharmacokinetic (PK) profile of the ADC. Although the quantitation of antibodies/ADCs using LC-MS is often coupled with affinity capture methods, here, we aimed to develop a versatile intact LC-MS method for evaluating the average DAR and DLD of ADCs in human plasma. The development of the affinity purification process and method validation were performed using healthy human pooled plasma spiked with the model ADCs, commercially available trastuzumab emtansine (T-DM1) and brentuximab vedotin (B-MMAE), and the recombinant proteins HER2 and CD30 were used to capture T-DM1 and B-MMAE, respectively. As unique points of this study, initially, a two-step gradient was established for the sensitive detection of a small amount of ADC. The ADC elution conditions after affinity capture were also optimized considering its application for LC-MS analysis. Furthermore, a validation study of the intact LC-MS approach for analyzing the average DAR and DLD of ADCs in human plasma sample was proposed for the first time. Using the validation study, our analytical method was validated by verifying its performance characteristics, including sensitivity, intermediate precision, accuracy, carryover and autosampler stability. In addition, the feasibility of applying our method was demonstrated by a collaborative study with six laboratories. Finally, our method was shown to be versatile for evaluating the average DAR and DLD of ADCs in human plasma.
Collapse
Affiliation(s)
- Noritaka Hashii
- National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki 210-9501, Japan.
| | - Yusuke Haruyama
- CMIC Pharma Science Co., Ltd., 17-18, Nakahata, Nishiwaki 677-0032, Japan
| | - Ryu Hirayama
- CMIC Pharma Science Co., Ltd., 17-18, Nakahata, Nishiwaki 677-0032, Japan
| | - Ryo Kajita
- Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yuki Kishino
- Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | | | - Kazuko Inoue
- Eisai Co., Ltd, 5-1-3 Tokodai, Tsukuba 300-2635, Japan
| | - Ryoya Goda
- Future Peak Co., Ltd., 3-48, Imainakamachi, Nakahara-ku, Kawasaki 211-0065, Japan
| | - Masaki Hoshino
- Mediford Corp., 1-15-10 Shimura, Itabashi-ku, Tokyo 174-0056, Japan
| | - Itsuki Kuroiwa
- Mediford Corp., 1-15-10 Shimura, Itabashi-ku, Tokyo 174-0056, Japan
| | - Hiroaki Aikawa
- Mediford Corp., 1-15-10 Shimura, Itabashi-ku, Tokyo 174-0056, Japan
| | - Natsuki Ueda
- Ono Pharmaceutical Co., Ltd., 17-2 Wadai, Tsukuba 300-4247, Japan
| | - Kaori Nagumo
- Ono Pharmaceutical Co., Ltd., 17-2 Wadai, Tsukuba 300-4247, Japan
| | - Yuki Oda
- Ono Pharmaceutical Co., Ltd., 17-2 Wadai, Tsukuba 300-4247, Japan
| | - Yoshiro Saito
- National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki 210-9501, Japan
| | - Akiko Ishii-Watabe
- National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki 210-9501, Japan
| |
Collapse
|
2
|
Song W, Yin L, Ren J, Jiang L, Huang ZA, Fan Y, Tao R, Duan T, Su Z, Cao Y, Song J, Xu A. Simultaneous Analysis of the Drug-to-Antibody Ratio, Free-Drug-Related Impurities, and Purity of Antibody-Drug Conjugates Based on Size Exclusion Chromatography. Anal Chem 2025; 97:9748-9754. [PMID: 40299313 DOI: 10.1021/acs.analchem.4c06661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Drug-to-antibody ratio (DAR), free-drug-related impurities (FDRI) content, and purity are critical quality attributes of antibody-drug conjugates (ADCs), which substantially impact product safety and efficacy. However, growing efforts in developing ADCs with higher complexity in a faster timeline impose a great challenge to their analytical support. Herein, for the ADC carries an antibody and linker-payload with distinct UV/vis absorption maxima, we propose a high-throughput and multiattribute ADC analysis strategy based on size exclusion chromatography coupled with UV detection (SEC-UV). Briefly, in addition to the quantitation of aggregates and fragments, SEC excludes FDRI from ADC-related peaks and improves the accuracy of DAR determination via dual-wavelength UV detection. Relative FDRI content can be determined subsequently by comparing the free-drug-related and ADC-related peaks at a wavelength where UV absorbance is exclusively attributed to the linker-payload. Afterward, a quantitative consistency in DAR and FDRI analysis was established between SEC-UV and orthogonal methodologies widely adopted by the pharmaceutical industry. The applicability of the developed SEC-UV method was further extended through feasiblity studies on ADC process-intermediates, bispecific ADC, and photodegraded ADC. Despite lacking characterization of drug load distribution and profiling of individual FDRI species, its efficiency, simplicity, and high throughput make SEC-UV a phase-appropriate strategy for supporting in-process monitoring and early-stage process development of diverse ADC projects.
Collapse
Affiliation(s)
- Woran Song
- Department of Global Technical Operations (GTO), BeiGene (Shanghai) Co. Ltd., Shanghai 200131, China
| | - Lifeng Yin
- Department of Global Technical Operations (GTO), BeiGene (Shanghai) Co. Ltd., Shanghai 200131, China
| | - Jun Ren
- Department of Global Technical Operations (GTO), BeiGene (Shanghai) Co. Ltd., Shanghai 200131, China
| | - Lai Jiang
- Department of Global Technical Operations (GTO), BeiGene (Shanghai) Co. Ltd., Shanghai 200131, China
| | - Zi-Ao Huang
- Department of Global Technical Operations (GTO), BeiGene (Shanghai) Co. Ltd., Shanghai 200131, China
| | - Yamin Fan
- Department of Global Technical Operations (GTO), BeiGene (Shanghai) Co. Ltd., Shanghai 200131, China
| | - Rong Tao
- Department of Global Technical Operations (GTO), BeiGene (Shanghai) Co. Ltd., Shanghai 200131, China
| | - Tingting Duan
- Department of Global Technical Operations (GTO), BeiGene (Shanghai) Co. Ltd., Shanghai 200131, China
| | - Zijun Su
- Department of Global Technical Operations (GTO), BeiGene (Beijing) Co. Ltd., Beijing 102206, China
| | - Yanjing Cao
- Department of Global Technical Operations (GTO), BeiGene (Shanghai) Co. Ltd., Shanghai 200131, China
| | - Jing Song
- Department of Global Technical Operations (GTO), BeiGene (Beijing) Co. Ltd., Beijing 102206, China
| | - April Xu
- Department of Global Technical Operations (GTO), BeiGene (Shanghai) Co. Ltd., Shanghai 200131, China
| |
Collapse
|
3
|
Li Z, Zhang J, Wu Y, Wang F, Cai T. Integrated strategy for biotransformation of antibody-drug conjugates and multidimensional interpretation via high-resolution mass spectrometry. Drug Metab Dispos 2025; 53:100081. [PMID: 40354712 DOI: 10.1016/j.dmd.2025.100081] [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: 03/20/2025] [Revised: 04/13/2025] [Accepted: 04/14/2025] [Indexed: 05/14/2025] Open
Abstract
The characterization of the release mechanism and stability in circulation for novel antibody-drug conjugates (ADCs) has become essential to address the complex variables (linker or payload selection, antibody, conjugating site). Understanding the integrated biotransformation of released catabolites and intact ADCs is necessary to elucidate the mechanism of action and mitigate the premature payload release and formation of inactive ADCs during circulation, which can lead to pharmacokinetic/pharmacodynamic disconnection. Herein, we present a comprehensive strategy for the biotransformation of ADCs from both small- and large-molecule perspectives. Two ADCs with common cleavable linkers were investigated: ADC-1 (maleimidocaproyl glycine-glycine-phenylalanine-glycine deruxtecan) and ADC-2 (maleimidocaproyl valine-citrulline-p-aminobenzyl carbamate monomethyl auristatin E). First, the payload-related catabolites released from lysosome, S9, and tumor cells were identified by sensitive data mining based on high-resolution mass spectrometry to reveal the pharmacologically active components. Second, we demonstrated the biotransformation occurring in intact ADCs using middle-down and bottom-up approaches, which particularly contributed to their instability in incubation. The combined immune capture with subunit or peptide analysis enables a comprehensive evaluation of the structural integrity of ADCs, whereas solely quantitative payload release is insufficient to determine the ADCs' stability. Although subunit analysis can visualize the deconjugation by mass shift directly, the precise and low percentage of biotransformation tended to be imperceptible in intact mass spectra. Therefore, a bottom-up method was developed to analyze three representative peptides conjugated with linker-payload. These multiple dimensional biotransformation approaches provide overall insights into the ADC development. SIGNIFICANCE STATEMENT: A thorough understanding of the release mechanism and stability is pivotal for advancing antibody drug conjugates (ADCs) therapeutics. However, the inherent complexity of ADCs poses significant challenges in biotransformation analysis. We developed an integrated strategy to systematically evaluate ADC biotransformation, encompassing payload release mechanisms and plasma stability assessments with middle-down and bottom-up approaches. This advancement not only clarifies the mechanism of action but also supports the rational design of ADC candidates, such as linker chemistry, payload selection, and antibody engineering.
Collapse
Affiliation(s)
- Ziyi Li
- Department of DMPK-BA, BeiGene (Beijing) Co, Ltd, Beijing, China
| | - Jingxian Zhang
- Department of DMPK-BA, BeiGene (Beijing) Co, Ltd, Beijing, China
| | - Yue Wu
- Department of DMPK-BA, BeiGene (Beijing) Co, Ltd, Beijing, China
| | - Fan Wang
- Department of DMPK-BA, BeiGene (Beijing) Co, Ltd, Beijing, China
| | - Tingting Cai
- Department of DMPK-BA, BeiGene (Beijing) Co, Ltd, Beijing, China.
| |
Collapse
|
4
|
Yu X, Li W, Huang W, Xiao B, Long J, Wang Q, Wang G, Wang C, Yu M, Yu J, Diao X. Simultaneous Quantification of Total Antibody and Conjugated Payload for DS001 in Rat Serum Using a Hybrid Immuno-Capture LC-MS/MS. AAPS J 2025; 27:23. [PMID: 39775231 DOI: 10.1208/s12248-024-01007-4] [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: 08/30/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Antibody-drug conjugates (ADCs) are intricate compounds that pose significant challenges in bioanalytical characterization. Therefore, multiple bioanalytical methods are required to comprehensively elucidate their pharmacokinetic (PK) profiles. In this study, we investigated DS001, an ADC consisting of a humanized monoclonal antibody (hRS7), a cleavable chemical linker, and the microtubule inhibitor monomethyl auristatin E (MMAE), with a drug-to-antibody ratio (DAR) of 8. This study established a rapid and sensitive hybrid immunoaffinity liquid-chromatography-tandem-mass-spectrometry (LC-MS/MS) approach for the simultaneous quantification of the total antibody and the enzymatically cleavable conjugated payload of DS001. This method is capable of monitoring fluctuations in average DAR values during PK assessments. The sample preparation procedure involved immunocapture, denaturation, trypsin digestion, papain digestion, and termination, all completed within a total processing time of less than 4 h. The method demonstrated linearity for the total antibody in the range of 100 ng/mL (lower-limit-of-quantification, LLOQ) to 100,000 ng/mL, and for the conjugated payload from 3.495 ng/mL (LLOQ) to 3495 ng/mL in rat serum. Both analytes exhibited standard curve correlation coefficients (r) greater than 0.990 within their respective linear ranges. The precision and accuracy of the method were within ± 15% (± 20% for LLOQ). The verified LC-MS/MS approach was successfully employed in the PK analysis following intravenous administration of 0.2 mg/kg DS001 in rats via tail vein injection.
Collapse
MESH Headings
- Animals
- Tandem Mass Spectrometry/methods
- Immunoconjugates/pharmacokinetics
- Immunoconjugates/administration & dosage
- Immunoconjugates/blood
- Immunoconjugates/chemistry
- Rats
- Chromatography, Liquid/methods
- Rats, Sprague-Dawley
- Oligopeptides/pharmacokinetics
- Oligopeptides/blood
- Oligopeptides/chemistry
- Oligopeptides/administration & dosage
- Male
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/blood
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/blood
- Antibodies, Monoclonal/administration & dosage
- Liquid Chromatography-Mass Spectrometry
Collapse
Affiliation(s)
- Xiong Yu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201210, China
| | - 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
| | - Bo Xiao
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Jing Long
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Qi Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Guifeng Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Chunhe Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Mingming Yu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266003, 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
|
5
|
Bardia A, Jhaveri K, Kalinsky K, Pernas S, Tsurutani J, Xu B, Hamilton E, Im SA, Nowecki Z, Sohn J, Laurentiis MD, Jañez NM, Adamo B, Lee KS, Jung KH, Rubovszky G, Tseng LM, Lu YS, Yuan Y, Maxwell MJ, Haddad V, Khan SS, Rugo HS, Pistilli B. TROPION-Breast01: Datopotamab deruxtecan vs chemotherapy in pre-treated inoperable or metastatic HR+/HER2- breast cancer. Future Oncol 2024; 20:423-436. [PMID: 37387213 DOI: 10.2217/fon-2023-0188] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023] Open
Abstract
Improving the prognosis for patients with metastatic HR+/HER2- breast cancer remains an unmet need. Patients with tumors that have progressed on endocrine therapy and/or are not eligible for endocrine therapy had limited treatment options beyond chemotherapy. Antibody-drug conjugates are a novel and promising treatment class in this setting. Datopotamab deruxtecan (Dato-DXd) consists of a TROP2-directed humanized IgG1 monoclonal antibody attached via a serum-stable cleavable linker to a topoisomerase I inhibitor payload. TROPION-Breast01 is an ongoing phase III study that is evaluating the efficacy and safety of Dato-DXd compared with investigator's choice of standard-of-care chemotherapy in patients with inoperable or metastatic HR+/HER2- breast cancer who have received one or two prior lines of systemic chemotherapy in the inoperable or metastatic setting. Clinical Trial Registration: NCT05104866 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Aditya Bardia
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Komal Jhaveri
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Kevin Kalinsky
- Winship Cancer Institute at Emory University, Atlanta, GA, USA
| | - Sonia Pernas
- Institut Català d'Oncologia, IDIBELL, L'Hospitalet, Barcelona, Spain
| | | | - Binghe Xu
- National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Erika Hamilton
- Sarah Cannon Research Institute / Tennessee Oncology, Nashville, TN, USA
| | - Seock-Ah Im
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Zbigniew Nowecki
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Joohyuk Sohn
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | - Noelia Martínez Jañez
- Ramón y Cajal University Hospital, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Barbara Adamo
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Keun Seok Lee
- Center for Breast Cancer, National Cancer Center, Goyang, Republic of Korea
| | - Kyung Hae Jung
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | - Ling-Ming Tseng
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yen-Shen Lu
- National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yuan Yuan
- Formerly City of Hope Comprehensive Cancer Center, Duarte, CA, USA; Currently: Cedars-Sinai Cancer Center, Los Angeles, CA, USA
| | | | | | | | - Hope S Rugo
- University of California San Francisco Comprehensive Cancer Center, San Francisco, CA, USA
| | | |
Collapse
|
6
|
Chang HP, Le HK, Shah DK. Pharmacokinetics and Pharmacodynamics of Antibody-Drug Conjugates Administered via Subcutaneous and Intratumoral Routes. Pharmaceutics 2023; 15:pharmaceutics15041132. [PMID: 37111619 PMCID: PMC10142912 DOI: 10.3390/pharmaceutics15041132] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/14/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023] Open
Abstract
We hypothesize that different routes of administration may lead to altered pharmacokinetics/pharmacodynamics (PK/PD) behavior of antibody-drug conjugates (ADCs) and may help to improve their therapeutic index. To evaluate this hypothesis, here we performed PK/PD evaluation for an ADC administered via subcutaneous (SC) and intratumoral (IT) routes. Trastuzumab-vc-MMAE was used as the model ADC, and NCI-N87 tumor-bearing xenografts were used as the animal model. The PK of multiple ADC analytes in plasma and tumors, and the in vivo efficacy of ADC, after IV, SC, and IT administration were evaluated. A semi-mechanistic PK/PD model was developed to characterize all the PK/PD data simultaneously. In addition, local toxicity of SC-administered ADC was investigated in immunocompetent and immunodeficient mice. Intratumoral administration was found to significantly increase tumor exposure and anti-tumor activity of ADC. The PK/PD model suggested that the IT route may provide the same efficacy as the IV route at an increased dosing interval and reduced dose level. SC administration of ADC led to local toxicity and reduced efficacy, suggesting difficulty in switching from IV to SC route for some ADCs. As such, this manuscript provides unprecedented insight into the PK/PD behavior of ADCs after IT and SC administration and paves the way for clinical evaluation of these routes.
Collapse
Affiliation(s)
- Hsuan-Ping Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14241, USA
| | - Huyen Khanh Le
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14241, USA
| | - Dhaval K. Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14241, USA
| |
Collapse
|
7
|
Moran AB, Domínguez-Vega E, Wuhrer M, Lageveen-Kammeijer GSM. Software-Assisted Data Processing Workflow for Intact Glycoprotein Mass Spectrometry. J Proteome Res 2023; 22:1367-1376. [PMID: 36857466 PMCID: PMC10088042 DOI: 10.1021/acs.jproteome.2c00762] [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: 03/03/2023]
Abstract
Intact protein analysis by mass spectrometry is important for several applications such as assessing post-translational modifications and biotransformation. In particular, intact protein analysis allows the detection of proteoforms that are commonly missed by other approaches such as proteolytic digestion followed by bottom-up analysis. Two quantification methods are mainly used for intact protein data quantification, namely the extracted ion and deconvolution approaches. However, a consensus with regard to a single best practice for intact protein data processing is lacking. Furthermore, many data processing tools are not fit-for-purpose and, as a result, the analysis of intact proteins is laborious and lacks the throughput required to be implemented for the analysis of clinical cohorts. Therefore, in this study, we investigated the application of a software-assisted data analysis and processing workflow in order to streamline intact protein integration, annotation, and quantification via deconvolution. In addition, the assessment of orthogonal data sets generated via middle-up and bottom-up analysis enabled the cross-validation of cleavage proteoform assignments present in seminal prostate-specific antigen (PSA). Furthermore, deconvolution quantification of PSA from patients' urine revealed results that were comparable with manually performed quantification based on extracted ion electropherograms. Overall, the presented workflow allows fast and efficient processing of intact protein data. The raw data is available on MassIVE using the identifier MSV000086699.
Collapse
Affiliation(s)
- Alan B Moran
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Elena Domínguez-Vega
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Guinevere S M Lageveen-Kammeijer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.,Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, 9713 AV Groningen, The Netherlands
| |
Collapse
|
8
|
Marei HE, Cenciarelli C, Hasan A. Potential of antibody-drug conjugates (ADCs) for cancer therapy. Cancer Cell Int 2022; 22:255. [PMID: 35964048 PMCID: PMC9375290 DOI: 10.1186/s12935-022-02679-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/05/2022] [Indexed: 11/10/2022] Open
Abstract
The primary purpose of ADCs is to increase the efficacy of anticancer medications by minimizing systemic drug distribution and targeting specific cells. Antibody conjugates (ADCs) have changed the way cancer is treated. However, because only a tiny fraction of patients experienced long-term advantages, current cancer preclinical and clinical research has been focused on combination trials. The complex interaction of ADCs with the tumor and its microenvironment appear to be reliant on the efficacy of a certain ADC, all of which have significant therapeutic consequences. Several clinical trials in various tumor types are now underway to examine the potential ADC therapy, based on encouraging preclinical results. This review tackles the potential use of ADCs in cancer therapy, emphasizing the essential processes underlying their positive therapeutic impacts on solid and hematological malignancies. Additionally, opportunities are explored to understand the mechanisms of ADCs action, the mechanism of resistance against ADCs, and how to overcome potential resistance following ADCs administration. Recent clinical findings have aroused interest, leading to a large increase in the number of ADCs in clinical trials. The rationale behind ADCs, as well as their primary features and recent research breakthroughs, will be discussed. We then offer an approach for maximizing the potential value that ADCs can bring to cancer patients by highlighting key ideas and distinct strategies.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.
| | | | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
| |
Collapse
|
9
|
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: 5] [Impact Index Per Article: 1.7] [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
|
10
|
Pouzin C, Gibiansky L, Fagniez N, Chadjaa M, Tod M, Nguyen L. Integrated multiple analytes and semi-mechanistic population pharmacokinetic model of tusamitamab ravtansine, a DM4 anti-CEACAM5 antibody-drug conjugate. J Pharmacokinet Pharmacodyn 2022; 49:381-394. [PMID: 35166967 PMCID: PMC9098589 DOI: 10.1007/s10928-021-09799-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/20/2021] [Indexed: 01/01/2023]
Abstract
Tusamitamab ravtansine (SAR408701) is an antibody-drug conjugate (ADC), combining a humanized monoclonal antibody (IgG1) targeting carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) and a potent cytotoxic maytansinoid derivative, DM4, inhibiting microtubule assembly. SAR408701 is currently in clinical development for the treatment of advanced solid tumors expressing CEACAM5. It is administered intravenously as a conjugated antibody with an average Drug Antibody Ratio (DAR) of 3.8. During SAR408701 clinical development, four entities were measured in plasma: conjugated antibody (SAR408701), naked antibody (NAB), DM4 and its methylated metabolite (MeDM4), both being active. Average DAR and proportions of individual DAR species were also assessed in a subset of patients. An integrated and semi-mechanistic population pharmacokinetic model describing the time-course of all entities in plasma and DAR measurements has been developed. All DAR moieties were assumed to share the same drug disposition parameters, excepted for clearance which differed for DAR0 (i.e. NAB entity). The conversion of higher DAR to lower DAR resulted in a DAR-dependent ADC deconjugation and was represented as an irreversible first-order process. Each conjugated antibody was assumed to contribute to DM4 formation. All data were fitted simultaneously and the model developed was successful in describing the pharmacokinetic profile of each entity. Such a structural model could be translated to other ADCs and gives insight of mechanistic processes governing ADC disposition. This framework will further be expanded to evaluate covariates impact on SAR408701 pharmacokinetics and its derivatives, and thus can help identifying sources of pharmacokinetic variability and potential efficacy and safety pharmacokinetic drivers.
Collapse
Affiliation(s)
- Clemence Pouzin
- Sanofi R&D, Pharmacokinetics Dynamics and Metabolism Department, 1 Avenue Pierre Brossolette, Chilly-Mazarin, 91380, Paris, France.
- Oncology department EMR3738, PKPD modelling unit, University of Claude Bernard Lyon 1, Lyon, France.
| | | | - Nathalie Fagniez
- Sanofi R&D, Pharmacokinetics Dynamics and Metabolism Department, 1 Avenue Pierre Brossolette, Chilly-Mazarin, 91380, Paris, France
| | | | - Michel Tod
- Oncology department EMR3738, PKPD modelling unit, University of Claude Bernard Lyon 1, Lyon, France
| | - Laurent Nguyen
- Sanofi R&D, Pharmacokinetics Dynamics and Metabolism Department, 1 Avenue Pierre Brossolette, Chilly-Mazarin, 91380, Paris, France
| |
Collapse
|
11
|
Pettinato MC. Introduction to Antibody-Drug Conjugates. Antibodies (Basel) 2021; 10:antib10040042. [PMID: 34842621 PMCID: PMC8628511 DOI: 10.3390/antib10040042] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/30/2021] [Accepted: 10/06/2021] [Indexed: 02/05/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are innovative biopharmaceutical products in which a monoclonal antibody is linked to a small molecule drug with a stable linker. Most of the ADCs developed so far are for treating cancer, but there is enormous potential for using ADCs to treat other diseases. Currently, ten ADCs have been approved by the United States Food and Drug Administration (FDA), and more than 90 ADCs are under worldwide clinical development. Monoclonal antibodies have evolved from research tools to powerful therapeutics in the past 30 years. Tremendous strides have been made in antibody discovery, protein bioengineering, formulation, and delivery devices. This manuscript provides an overview of the biology, chemistry, and biophysical properties of each component of ADC design. This review summarizes the advances and challenges in the field to date, with an emphasis on antibody conjugation, linker-payload chemistry, novel payload classes, drug-antibody ratio (DAR), and product development. The review emphasizes the lessons learned in the development of oncology antibody conjugates and look towards future innovations enabling other therapeutic indications. The review discusses resistance mechanisms to ADCs, and give an opinion on future perspectives.
Collapse
Affiliation(s)
- Mark C Pettinato
- Department of Biomedical Engineering, School of Engineering, Catholic University of America, 620 Michigan Avenue NE, Washington, DC 20064-0001, USA
| |
Collapse
|
12
|
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: 11] [Impact Index Per Article: 2.8] [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
|
13
|
Nagornov KO, Gasilova N, Kozhinov AN, Virta P, Holm P, Menin L, Nesatyy VJ, Tsybin YO. Drug-to-Antibody Ratio Estimation via Proteoform Peak Integration in the Analysis of Antibody-Oligonucleotide Conjugates with Orbitrap Fourier Transform Mass Spectrometry. Anal Chem 2021; 93:12930-12937. [PMID: 34519496 DOI: 10.1021/acs.analchem.1c02247] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The therapeutic efficacy and pharmacokinetics of antibody-drug conjugates (ADCs) in general, and antibody-oligonucleotide conjugates (AOCs) in particular, depend on the drug-to-antibody ratio (DAR) distribution and average value. The DAR is considered a critical quality attribute, and information pertaining to it needs to be gathered during ADC/AOC development, production, and storage. However, because of the high structural complexity of ADC/AOC samples, particularly in the initial drug-development stages, the application of the current state-of-the-art mass spectrometric approaches can be limited for DAR analysis. Here, we demonstrate a novel approach for the analysis of complex ADC/AOC samples, following native size-exclusion chromatography Orbitrap Fourier transform mass spectrometry (FTMS). The approach is based on the integration of the proteoform-level mass spectral peaks in order to provide an estimate of the DAR distribution and its average value with less than 10% error. The peak integration is performed via a truncation of the Orbitrap's unreduced time-domain ion signals (transients) before mass spectra generation via FT processing. Transient recording and processing are undertaken using an external data acquisition system, FTMS Booster X2, coupled to a Q Exactive HF Orbitrap FTMS instrument. This approach has been applied to the analysis of whole and subunit-level trastuzumab conjugates with oligonucleotides. The obtained results indicate that ADC/AOC sample purification or simplification procedures, for example, deglycosylation, could be omitted or minimized prior to the DAR analysis, streamlining the drug-development process.
Collapse
Affiliation(s)
| | - Natalia Gasilova
- Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | | | - Pasi Virta
- Department of Chemistry, University of Turku, 20014 Turku, Finland
| | - Patrik Holm
- Protein and Antibody Engineering Unit, Orion Pharma, 20380 Turku, Finland
| | - Laure Menin
- Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Victor J Nesatyy
- Protein and Antibody Engineering Unit, Orion Pharma, 20380 Turku, Finland
| | | |
Collapse
|
14
|
Larson EJ, Roberts DS, Melby JA, Buck KM, Zhu Y, Zhou S, Han L, Zhang Q, Ge Y. High-Throughput Multi-attribute Analysis of Antibody-Drug Conjugates Enabled by Trapped Ion Mobility Spectrometry and Top-Down Mass Spectrometry. Anal Chem 2021; 93:10013-10021. [PMID: 34258999 PMCID: PMC8319120 DOI: 10.1021/acs.analchem.1c00150] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Antibody-drug conjugates (ADCs) are one of the fastest growing classes of anticancer therapies. Combining the high targeting specificity of monoclonal antibodies (mAbs) with cytotoxic small molecule drugs, ADCs are complex molecular entities that are intrinsically heterogeneous. Primary sequence variants, varied drug-to-antibody ratio (DAR) species, and conformational changes in the starting mAb structure upon drug conjugation must be monitored to ensure the safety and efficacy of ADCs. Herein, we have developed a high-throughput method for the analysis of cysteine-linked ADCs using trapped ion mobility spectrometry (TIMS) combined with top-down mass spectrometry (MS) on a Bruker timsTOF Pro. This method can analyze ADCs (∼150 kDa) by TIMS followed by a three-tiered top-down MS characterization strategy for multi-attribute analysis. First, the charge state distribution and DAR value of the ADC are monitored (MS1). Second, the intact mass of subunits dissociated from the ADC by low-energy collision-induced dissociation (CID) is determined (MS2). Third, the primary sequence for the dissociated subunits is characterized by CID fragmentation using elevated collisional energies (MS3). We further automate this workflow by directly injecting the ADC and using MS segmentation to obtain all three tiers of MS information in a single 3-min run. Overall, this work highlights a multi-attribute top-down MS characterization method that possesses unparalleled speed for high-throughput characterization of ADCs.
Collapse
Affiliation(s)
- Eli J Larson
- Department of Chemistry, University of Wisconsin - Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - David S Roberts
- Department of Chemistry, University of Wisconsin - Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Jake A Melby
- Department of Chemistry, University of Wisconsin - Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Kevin M Buck
- Department of Chemistry, University of Wisconsin - Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Yanlong Zhu
- Department of Cell and Regenerative Biology, University of Wisconsin - Madison, 1111 Highland Avenue., Madison, Wisconsin 53705, United States
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin, 1111 Highland Avenue., Madison, Wisconsin 53705, United States
| | - Shiyue Zhou
- Analytical R&D, AbbVie Inc., 1 Waukegan Rd, North Chicago, Illinois 60064, United States
| | - Linjie Han
- Analytical R&D, AbbVie Inc., 1 Waukegan Rd, North Chicago, Illinois 60064, United States
| | - Qunying Zhang
- Analytical R&D, AbbVie Inc., 1 Waukegan Rd, North Chicago, Illinois 60064, United States
| | - Ying Ge
- Department of Chemistry, University of Wisconsin - Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
- Department of Cell and Regenerative Biology, University of Wisconsin - Madison, 1111 Highland Avenue., Madison, Wisconsin 53705, United States
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin, 1111 Highland Avenue., Madison, Wisconsin 53705, United States
| |
Collapse
|
15
|
Su D, Zhang D. Linker Design Impacts Antibody-Drug Conjugate Pharmacokinetics and Efficacy via Modulating the Stability and Payload Release Efficiency. Front Pharmacol 2021; 12:687926. [PMID: 34248637 PMCID: PMC8262647 DOI: 10.3389/fphar.2021.687926] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/09/2021] [Indexed: 01/03/2023] Open
Abstract
The development of antibody-drug conjugates (ADCs) has significantly been advanced in the past decade given the improvement of payloads, linkers and conjugation methods. In particular, linker design plays a critical role in modulating ADC stability in the systemic circulation and payload release efficiency in the tumors, which thus affects ADC pharmacokinetic (PK), efficacy and toxicity profiles. Previously, we have investigated key linker parameters such as conjugation chemistry (e.g., maleimide vs. disulfide), linker length and linker steric hindrance and their impacts on PK and efficacy profiles. Herein, we discuss our perspectives on development of integrated strategies for linker design to achieve a balance between ADC stability and payload release efficiency for desired efficacy in antigen-expressing xenograft models. The strategies have been successfully applied to the design of site-specific THIOMABTM antibody-drug conjugates (TDCs) with different payloads. We also propose to conduct dose fractionation studies to gain guidance for optimal dosing regimens of ADCs in pre-clinical models.
Collapse
Affiliation(s)
- Dian Su
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, United States
| | - Donglu Zhang
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, United States
| |
Collapse
|
16
|
Huang Y, Mou S, Wang Y, Mu R, Liang M, Rosenbaum AI. Characterization of Antibody-Drug Conjugate Pharmacokinetics and in Vivo Biotransformation Using Quantitative Intact LC-HRMS and Surrogate Analyte LC-MRM. Anal Chem 2021; 93:6135-6144. [PMID: 33835773 DOI: 10.1021/acs.analchem.0c05376] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Antibody-drug conjugates (ADCs) pose challenges to bioanalysis because of their inherently intricate structures and potential for very complex catabolism. Common bioanalysis strategy is to measure the concentration of ADCs and Total Antibody (Ab) as well as deconjugated warhead in circulation. The ADCs and the Total Ab can be quantified with ligand binding assays (LBA) or with hybrid immunocapture-liquid chromatography coupled with multiple reaction monitoring mass spectrometry (LBA-LC-MRM). With the LBA-LC-MRM approach, a surrogate analyte, often the signature peptide, and released warhead can be used for the quantification of the Total Ab and ADCs, respectively. Recent advances in analytical instrumentation, especially the development of high resolution mass spectrometers (HRMS), have enabled characterization and quantification of intact macromolecules such as ADCs. The LBA-LC-HRMS approach employs immunocapture, followed by chromatographic separation at the macromolecule level and detection of the intact analyte. We developed an intact quantification method with 1-10 μg/mL linear dynamic range using 25 μL of plasma sample volume. This method was qualified for the measurement of naked monoclonal antibody (mAb), a site-specific cysteine-conjugated ADC with drug to antibody ratio ∼2 (DAR2) and a site-nonspecific cysteine-conjugated ADC (DAR8) in rat plasma. Samples from a rat pharmacokinetic (PK) study were analyzed with both methods. For the naked mAb, the results from both assays matched well. For ADCs, new species were observed from the LBA-HRMS method. The results demonstrated that potential biotransformation of the ADC was unveiled using the intact quantification approach while not being observed with traditional LBA-LC-MRM approach. Our work demonstrated an application of novel intact quantification by supporting animal PK studies. Moreover, our results suggest that the intact quantification method can provide novel perspectives on ADC in vivo characterization and quantification, which can benefit future drug candidate optimization as well as the immunogenicity impact evaluation and safety assessment.
Collapse
Affiliation(s)
- Yue Huang
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Si Mou
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Yadi Wang
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Ruipeng Mu
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Meina Liang
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Anton I Rosenbaum
- Integrated Bioanalysis, Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, 121 Oyster Point Boulevard, South San Francisco, California 94080, United States
| |
Collapse
|
17
|
Application of middle-down approach in quantitation and catabolite identification of protein by LC-high-resolution mass spectrometry. Bioanalysis 2021; 13:465-479. [PMID: 33719526 DOI: 10.4155/bio-2020-0315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: To further enhance the detection sensitivity and increase resolving power of top-down intact protein bioanalysis, middle-down approach was explored. Materials & methods: An monoclonal antibody (mAb) was used as a model protein to evaluate quantitative bioanalytical assay performance and a disulfide linked dimer protein was investigated for its pharmacokinetics properties and catabolism in vivo by middle-down approach. Results & Conclusion: For quantitation of the mAb, different subunits generated by middle-down approach provided different level of signal improvement in biological samples with Lc and half Fc giving five-times better sensitivity than intact mAb. For the dimer protein, middle-down analysis by reduction enabled effective differentiation of the unchanged protein and its oxidized form, and clearly elucidated their respective proteolytic catabolites.
Collapse
|
18
|
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: 56] [Impact Index Per Article: 11.2] [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
|
19
|
Chen Y, Yang W, Wu J, Sun W, Loh TP, Jiang Y. 2H-Azirines as Potential Bifunctional Chemical Linkers of Cysteine Residues in Bioconjugate Technology. Org Lett 2020; 22:2038-2043. [DOI: 10.1021/acs.orglett.0c00415] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Yang Chen
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, P. R. China
| | - Wenjie Yang
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, P. R. China
| | - Jiamin Wu
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, P. R. China
| | - Wangbin Sun
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, P. R. China
| | - Teck-Peng Loh
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, P. R. China
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637616, Singapore
| | - Yaojia Jiang
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing 211816, P. R. China
| |
Collapse
|
20
|
IQ consortium perspective: complementary LBA and LC–MS in protein therapeutics bioanalysis and biotransformation assessment. Bioanalysis 2020; 12:257-270. [DOI: 10.4155/bio-2019-0279] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Increasingly diverse large molecule modalities have driven the need for complex bioanalysis and biotransformation assessment involving both traditional ligand-binding assays (LBA) and more recent hybrid immunoaffinity LC–MS platforms. Given the scientific expertise in LBA and LC–MS typically resides in different functions within the industry, this has presented operational challenges for an integrated approach for bioanalysis and biotransformation assessment. Encouragingly, over time, the industry has recognized the complementary value of the two platforms. This has not been an easy transition as organizational structures vary widely within the industry. However, there are tremendous benefits in adopting fully integrated strategies for biopharma. This IQ consortium paper presents current perspectives across the biopharma industry. It highlights the technical and operational challenges in current large molecule bioanalysis, the value of collaborations across LBA and LC–MS, and scientific expertise for fully integrated strategies for bioanalysis and biotransformation.
Collapse
|
21
|
Immunoaffinity LC–MS/MS to quantify a PEGylated anti-Factor D Fab biotherapeutic in cynomolgus monkey serum. Bioanalysis 2019; 11:2161-2173. [DOI: 10.4155/bio-2019-0082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aim: To develop a sensitive hybrid immunoaffinity LC–MS/MS monkey serum assay to quantify multiple components of anti-Factor D; a complex PEGylated Fab biotherapeutic explored as a therapy for age-related macular degeneration. Materials & methods: Immunoaffinity enrichment of PEGylated anti-Factor D Fab, including fully conjugated, partially conjugated and unconjugated (i.e., free) Fab species, using a capture reagent coupled to magnetic beads was performed. The surrogate peptides derived from the therapeutic Fab via trypsin digestion were measured to obtain the total Fab concentrations. Results & conclusion: The method demonstrated the ability to accurately quantify both PEGylated and unconjugated Fab species. It was successfully validated with a LLOQ at 25.0 ng/ml.
Collapse
|
22
|
High-Resolution Characterization of ADCs by Orbitrap LCMS. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2019; 2078:213-219. [PMID: 31643059 DOI: 10.1007/978-1-4939-9929-3_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Antibody-drug conjugates (ADCs) have complex molecular structures as they are composed of both small and large molecules, and they often undergo biotransformation over time in circulation. Here we describe a high-resolution Orbitrap MS approach for the characterization of ADC biotransformation and stability. Compared with conventional approach by Q-TOF MS, the method described here significantly improved the mass resolution and enabled more comprehensive characterization of ADC catabolites. It is particularly beneficial for characterizing ADC biotransformations with small mass changes.
Collapse
|
23
|
Vallejo DD, Polasky DA, Kurulugama RT, Eschweiler JD, Fjeldsted JC, Ruotolo BT. A Modified Drift Tube Ion Mobility-Mass Spectrometer for Charge-Multiplexed Collision-Induced Unfolding. Anal Chem 2019; 91:8137-8146. [PMID: 31194508 DOI: 10.1021/acs.analchem.9b00427] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Collision-induced unfolding (CIU) of protein ions and their noncovalent complexes offers relatively rapid access to a rich portfolio of biophysical information, without the need to tag or purify proteins prior to analysis. Such assays have been characterized extensively for a range of therapeutic proteins, proving exquisitely sensitive to alterations in protein sequence, structure, and post-translational modification state. Despite advantages over traditional probes of protein stability, improving the throughput and information content of gas-phase protein unfolding assays remains a challenge for current instrument platforms. In this report, we describe modifications to an Agilent 6560 drift tube ion mobility-mass spectrometer in order to perform robust, simultaneous CIU across all precursor ions detected. This approach dramatically increases the speed associated with typical CIU assays, which typically involve mass selection of narrow m/ z regions prior to collisional activation, and thus their development requires a comprehensive assessment of charge-stripping reactions that can unintentionally pollute CIU data with chemical noise when more than one precursor ion is allowed to undergo simultaneous activation. By studying the unfolding and dissociation of intact antibody ions, a key analyte class associated with biotherapeutics, we reveal a predictive relationship between the precursor charge state, the amount of buffer components bound to the ions of interest, and the amount of charge stripping detected. We then utilize our knowledge of antibody charge stripping to rapidly capture CIU data for a range of antibody subclasses and subtypes across all charge states simultaneously, demonstrating a strong charge state dependence on the information content of CIU. Finally, we demonstrate that CIU data collection times can be further reduced by scanning fewer voltage steps, enabling us to optimize the throughput of our improved CIU methods and confidently differentiate antibody variant ions using ∼20% of the data typically collected during CIU. Taken together, our results characterize a new instrument platform for biotherapeutic stability measurements with dramatically improved throughput and information content.
Collapse
Affiliation(s)
- Daniel D Vallejo
- Department of Chemistry , University of Michigan , 930 North University Avenue , Ann Arbor , Michigan 48109 , United States
| | - Daniel A Polasky
- Department of Chemistry , University of Michigan , 930 North University Avenue , Ann Arbor , Michigan 48109 , United States
| | | | - Joseph D Eschweiler
- Department of Chemistry , University of Michigan , 930 North University Avenue , Ann Arbor , Michigan 48109 , United States.,AbbVie , North Chicago , Illinois 60064 , United States
| | - John C Fjeldsted
- Agilent Technologies , Santa Clara , California 95051 , United States
| | - Brandon T Ruotolo
- Department of Chemistry , University of Michigan , 930 North University Avenue , Ann Arbor , Michigan 48109 , United States
| |
Collapse
|
24
|
Li K, Lin ZJ, Shi H, Ma Y. Characterization of Positional Isomers of Interchain Cysteine Linked Antibody−Drug Conjugates by High-Resolution Mass Spectrometry. Anal Chem 2019; 91:8558-8563. [DOI: 10.1021/acs.analchem.9b01808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Ke Li
- Department of Chemistry and Center for Biomedical Research, Missouri University of Science and Technonlogy, Rolla, Missouri 65409, United States
| | - Zhongping John Lin
- Department of Bioanalysis, Frontage Laboratories, Inc., Exton, Pennsylvania 19341, United States
| | - Honglan Shi
- Department of Chemistry and Center for Biomedical Research, Missouri University of Science and Technonlogy, Rolla, Missouri 65409, United States
| | - Yinfa Ma
- Department of Chemistry and Center for Biomedical Research, Missouri University of Science and Technonlogy, Rolla, Missouri 65409, United States
- Department of Chemistry, California State University, Sacramento, California 95819, United States
| |
Collapse
|
25
|
Hyung SJ, Li D, Koppada N, Kaur S, Saad OM. Method development of a novel PK assay for antibody-conjugated drug measurement of ADCs using peptide-linker drug analyte. Anal Bioanal Chem 2019; 411:2587-2596. [PMID: 30828756 DOI: 10.1007/s00216-019-01701-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/11/2019] [Accepted: 02/18/2019] [Indexed: 01/24/2023]
Abstract
Pharmacokinetic analysis of antibody-drug conjugates (ADCs) requires characterization and quantification of both the antibody-conjugated cytotoxic drug molecule (acDrug) as well as the antibody vehicle, among other analytes, in order to assess the safety and efficacy of ADCs. Due to the complexity of biological matrices, immunoaffinity capture is widely used for enrichment of the biotherapeutic, followed by enzymatic or chemical release of the drug and LC-MS/MS analysis to provide the concentration of acDrug. This bioanalytical strategy has been used successfully with ADCs, but is limited to ADCs having cleavable linkers. Herein, we developed a sensitive and specific method that involved subjecting the ADC to tryptic digestion, and measured a peptide that included cysteine conjugated to the drug to provide quantification of acDrug. Using this method for a THIOMAB™ antibody-drug conjugate (TDC) conjugated to MMAE via a cleavable linker, valine-citrulline, we compared peptide-linker MMAE data from the new assay format with earlier MMAE data for acDrug. This showed that the new assay format provides robust acDrug as well as total antibody concentration to study in vitro stability of the TDC in multiple matrices and in vivo pharmacokinetic models of TDC in rat and mouse. The data from the two orthogonal modes of acDrug analysis showed good agreement with each other, allowing us to successfully quantify acDrug to study the stability in vitro and the pharmacokinetic parameters in vivo. This new assay strategy allows acDrug quantification for ADCs with non-cleavable linkers where the resulting acDrug analyte is a peptide-linker drug.
Collapse
Affiliation(s)
- Suk-Joon Hyung
- Department of BioAnalytical Sciences - Assay Development and Technologies, Genentech, Inc., South San Francisco, CA, 94080, USA.
| | - Dongwei Li
- Pre-clinical and Translational Pharmacokinetics, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Neelima Koppada
- Department of BioAnalytical Sciences - Assay Development and Technologies, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Surinder Kaur
- Department of BioAnalytical Sciences - Assay Development and Technologies, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Ola M Saad
- Department of BioAnalytical Sciences - Assay Development and Technologies, Genentech, Inc., South San Francisco, CA, 94080, USA.
| |
Collapse
|
26
|
Protein quantification by LC–MS: a decade of progress through the pages of Bioanalysis. Bioanalysis 2019; 11:629-644. [DOI: 10.4155/bio-2019-0032] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Over the past 10 years, there has been a remarkable increase in the use of LC–MS for the quantitative determination of proteins, and this technique can now be considered an established bioanalytical platform for the quantification of macromolecular drugs and biomarkers, next to the traditional ligand-binding assays. Many researchers have contributed to the field and helped improve both the technical possibilities of LC–MS-based workflows and our understanding of the meaning of the results that are obtained. As a tribute to Bioanalysis, which has published many important contributions, this report gives a high-level overview of the most important trends in the field of protein LC–MS, as published in this journal since its inauguration a decade ago. It describes the major technical developments with regard to sample handling, separation and MS detection of both digested and intact protein analysis. In addition, the relevance of the complex structure and in vivo behavior of proteins is discussed and the effect of protein–protein interactions, biotransformation and the occurrence of isoforms on the analytical result is addressed.
Collapse
|
27
|
Biotherapeutic Antibody Subunit LC-MS and Peptide Mapping LC-MS Measurements to Study Possible Biotransformation and Critical Quality Attributes In Vivo. J Pharm Sci 2019; 108:1415-1422. [DOI: 10.1016/j.xphs.2018.11.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 10/29/2018] [Accepted: 11/14/2018] [Indexed: 12/13/2022]
|
28
|
Hayat SMG, Sahebkar A. Antibody-drug conjugates: smart weapons against cancer. Arch Med Sci 2019; 16:1257-1262. [PMID: 32864020 PMCID: PMC7444717 DOI: 10.5114/aoms.2019.83020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [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
|
29
|
Multiplex LC-MS/MS Assays for Clinical Bioanalysis of MEDI4276, an Antibody-Drug Conjugate of Tubulysin Analogue Attached via Cleavable Linker to a Biparatopic Humanized Antibody against HER-2. Antibodies (Basel) 2019; 8:antib8010011. [PMID: 31544817 PMCID: PMC6640689 DOI: 10.3390/antib8010011] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/06/2018] [Accepted: 12/06/2018] [Indexed: 01/14/2023] Open
Abstract
Bioanalysis of complex biotherapeutics, such as antibody-drug conjugates (ADCs), is challenging and requires multiple assays to describe their pharmacokinetic (PK) profiles. To enable exposure-safety and exposure-efficacy analyses, as well as to understand the metabolism of ADC therapeutics, three bioanalytical methods are typically employed: Total Antibody, Antibody Conjugated Toxin or Total ADC and Unconjugated Toxin. MEDI4276 is an ADC comprised of biparatopic humanized antibody attached via a protease-cleavable peptide-based maleimidocaproyl linker to a tubulysin toxin (AZ13599185) with an approximate average drug-antibody ratio of 4. The conjugated payload of MEDI4276 can undergo ester hydrolysis to produce the conjugated payload AZ13687308, leading to the formation of MEDI1498 (de-acetylated MEDI4276). In this report, we describe the development, validation and application of three novel multiplex bioanalytical methods. The first ligand-binding liquid chromatography coupled with tandem mass spectrometry (LBA-LC-MS/MS) method was developed and validated for simultaneous measurement of total antibody and total ADC (antibody-conjugated AZ13599185) from MEDI4276. The second LBA-LC-MS/MS assay quantified total ADC (antibody-conjugated AZ13687308) from MEDI1498. The third multiplex LC-MS/MS assay was used for simultaneous quantification of unconjugated AZ13599185 and AZ13687308. Additional stability experiments confirmed that quantification of the released warhead in the presence of high concentrations of MEDI4276 was acceptable. Subsequently, the assays were employed in support of a first-in-human clinical trial (NCT02576548).
Collapse
|
30
|
Wang J, Zhang W, Salter R, Lim HK. Reductive Desulfuration as an Important Tool in Detection of Small Molecule Modifications to Payload of Antibody Drug Conjugates. Anal Chem 2019; 91:2368-2375. [DOI: 10.1021/acs.analchem.8b05134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Jianyao Wang
- Department of Drug Metabolism and Pharmacokinetics, Janssen Research & Development, Welsh & McKean Roads, Spring House, Pennsylvania 19477, United States
| | - Wei Zhang
- Department of Drug Metabolism and Pharmacokinetics, Janssen Research & Development, Welsh & McKean Roads, Spring House, Pennsylvania 19477, United States
| | - Rhys Salter
- Department of Drug Metabolism and Pharmacokinetics, Janssen Research & Development, Welsh & McKean Roads, Spring House, Pennsylvania 19477, United States
| | - Heng-Keang Lim
- Department of Drug Metabolism and Pharmacokinetics, Janssen Research & Development, Welsh & McKean Roads, Spring House, Pennsylvania 19477, United States
| |
Collapse
|
31
|
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: 26] [Impact Index Per Article: 3.7] [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
|
32
|
Optimizing hybrid LC-MS/MS binding conditions is critical: impact of biotransformation on quantification of trastuzumab. Bioanalysis 2018; 10:1819-1831. [PMID: 30325201 DOI: 10.4155/bio-2018-0196] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background: Hybrid ligand-binding (LB) LC-MS/MS protein quantitative assays involve a LB step for analyte enrichment that has less stringent requirements than the conventional LB assays. Results: Herceptin™(trastuzumab) binding to HER2 extracellular domain was evaluated using on-bead and off-bead capture formats. The two formats yielded significantly different trastuzumab concentrations in human and monkey serum pharmacokinetic samples. Biotransformations, including deamidation of asparagine and isomerization of aspartic acid near the complementarity-determining regions of trastuzumab, had a profound impact on the LB step for analyte enrichment and trastuzumab quantification. Conclusion: Quantitative measurements were profoundly impacted by LB conditions in a hybrid LB LC-MS/MS protein assay due to biotransformations. Therefore, similar to conventional LB assays, binding conditions should be carefully evaluated during assay development.
Collapse
|
33
|
Review of approaches and examples for monitoring biotransformation in protein and peptide therapeutics by MS. Bioanalysis 2018; 10:1877-1890. [PMID: 30325207 DOI: 10.4155/bio-2018-0113] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Biotherapeutic drugs have emerged in quantity in pharmaceutical pipelines, and increasingly diverse biomolecules are progressed through preclinical and clinical development. As purification, separation, mass spectrometer detection and data processing capabilities improve, there is opportunity to monitor drug concentration by traditional ligand-binding assay or MS measurement and to monitor metabolism, catabolism or other biomolecular mass variants present in circulation. This review highlights approaches and examples of monitoring biotransformation of biotherapeutics by MS as these techniques are poised to add value to drug development in years to come. The increased use of such approaches, and the successful quantitation of biotherapeutic structural modifications, will provide insightful data for the benefit of both researchers and patients.
Collapse
|
34
|
Zhao WB, Qiu CX, Shen Y, Liu WH, Zhou J, Xu YC, Zhou Z, Chen SQ. In situ quantitative bioanalysis of monomethyl auristatin E-conjugated antibody-drug conjugates by flow cytometry. Eur J Pharm Sci 2018; 120:89-95. [PMID: 29727724 DOI: 10.1016/j.ejps.2018.04.046] [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: 01/30/2018] [Revised: 04/10/2018] [Accepted: 04/29/2018] [Indexed: 11/26/2022]
Abstract
Antibody-drug conjugates (ADCs) consist of cytotoxic agents covalently conjugated to monoclonal antibodies that substantially improve antitumour activity and reduce systemic toxicity. With the growing number of ADCs in clinical applications, more accurate bioanalysis data are urgently needed to facilitate the development and rational use of ADCs. Herein, we used antigen-positive cells as antigen carriers and ofatumumab (OFA-HL) and ofatumumab-based ADC (OFA-HL-MMAE) as examples to establish a new ligand-binding assay (LBA) method based on flow cytometry. We proved that the new method met the required analytical performance criteria and the lower limit of quantitation (LOQ) was 0.2 μg/mL. In addition, the LOQ of the quantitative OFA-HL flow cytometry method was reduced to 0.025 μg/mL by choosing an optimized fluorescent antibody, which indicated that the LOQ of the new method can be improved. What's more, the new method showed good stability and specificity when we used it to determine the concentrations of OFA-HL and OFA-HL-MMAE in mouse serum. During the bioanalysis of ADCs, various factors should be considered. Therefore, choosing optimal methods for ADC bioanalysis is necessary. This new method using in situ antigens not only extends the scope of application of the conventional LBA methods by avoiding the need for soluble antigens, but also improves the authenticity of ADC bioanalysis as a supplementary approach, which is valuable for developing accurate ADC assays.
Collapse
Affiliation(s)
- Wen-Bin Zhao
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chi-Xiao Qiu
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Shen
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wen-Hui Liu
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jie Zhou
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying-Chun Xu
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhan Zhou
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Shu-Qing Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
35
|
He J, Yu SF, Yee S, Kaur S, Xu K. Characterization of in vivo biotransformations for trastuzumab emtansine by high-resolution accurate-mass mass spectrometry. MAbs 2018; 10:960-967. [PMID: 29958059 DOI: 10.1080/19420862.2018.1494487] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate (ADC) designed for the treatment of HER2-positive cancers. T-DM1 is composed of the humanized monoclonal antibody trastuzumab connected to a maytansine derivative cytotoxic drug, via a nonreducible thioether linker at random lysine residues, and therefore has a very complex molecular structure. It was anticipated that T-DM1 undergoes biotransformations in circulation. However, there was limited knowledge on these structural changes due to bioanalytical challenges. Here, we have investigated the in vivo biotransformations of T-DM1 using a high-resolution accurate-mass (HR/AM) mass spectrometry approach. Three types of biotransformations were characterized for T-DM1 in circulation in tumor-bearing mice, including cysteine or glutathione adduct formation via maleimide exchange, loss of maytansinol via ester hydrolysis, as well as addition of H2O via linker-drug hydrolysis. These results provide new insights into in vivo catabolism of T-DM1.
Collapse
Affiliation(s)
- Jintang He
- a Genentech Research and Early Development, Genentech Inc ., South San Francisco, CA , USA
| | - Shang-Fan Yu
- a Genentech Research and Early Development, Genentech Inc ., South San Francisco, CA , USA
| | - Sharon Yee
- a Genentech Research and Early Development, Genentech Inc ., South San Francisco, CA , USA
| | - Surinder Kaur
- a Genentech Research and Early Development, Genentech Inc ., South San Francisco, CA , USA
| | - Keyang Xu
- a Genentech Research and Early Development, Genentech Inc ., South San Francisco, CA , USA
| |
Collapse
|
36
|
Quantitation of intact monoclonal antibody in biological samples: comparison of different data processing strategies. Bioanalysis 2018; 10:1055-1067. [DOI: 10.4155/bio-2018-0016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: Sample extraction using immuno-affinity capture coupled with LC–high-resolution mass spectrometer has recently emerged as a novel approach for the determination of concentrations of large molecules at intact level in biological matrix. Methodology: In the current work, different data processing strategies for intact protein bioanalysis, deconvoluted mass spectra or extracted ion chromatogram, were applied to quantitate monoclonal antibody in biological samples for comparison of assay performance. Conclusion: Both deconvolution and extracted ion chromatogram strategies showed similar selectivity, sensitivity, accuracy and precision. The monkey pharmacokinetics data obtained from both approaches agreed well with each other, and agreed with data obtained from surrogate peptide approach. The pros and cons, and optimal parameters of each approach were discussed.
Collapse
|
37
|
LC–HRMS quantitation of intact antibody drug conjugate trastuzumab emtansine from rat plasma. Bioanalysis 2018; 10:851-862. [DOI: 10.4155/bio-2018-0003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Aim: Compared with small molecules, LC–MS quantitation of larger biotherapeutic proteins such as antibodies and antibody–drug conjugates at the intact level presents many challenges in both LC and MS due to their higher molecular weight, bigger size, structural complexity and heterogeneity. Results & conclusion: In this study, quantitation of an intact lysine-linked antibody–drug conjugate, trastuzumab emtansine is presented. Trastuzumab emtansine was extracted from rat plasma using bead-based immunoaffinity capture; after elution from the beads, it was directly analyzed on a LC–HRMS system. Quantitation using both extracted ion chromatogram and deconvoluted mass peaks was evaluated. A limit of quantitation was approximately 20 ng on column with a linear dynamic range from 5 to 100 μg/ml. In addition, the reproducibility and distribution of the drug-to-antibody ratio at different trastuzumab emtansine concentrations were discussed.
Collapse
|
38
|
Shi C, Goldberg S, Lin T, Dudkin V, Widdison W, Harris L, Wilhelm S, Jmeian Y, Davis D, O’Neil K, Weng N, Jian W. LC/MS/MS Bioanalysis of Protein–Drug Conjugates—The Importance of Incorporating Succinimide Hydrolysis Products. Anal Chem 2018; 90:5314-5321. [DOI: 10.1021/acs.analchem.8b00411] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Chuan Shi
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Shalom Goldberg
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Tricia Lin
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Vadim Dudkin
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Wayne Widdison
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Luke Harris
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Sharon Wilhelm
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Yazen Jmeian
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Darryl Davis
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Karyn O’Neil
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Naidong Weng
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Wenying Jian
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| |
Collapse
|
39
|
LC–MS Challenges in Characterizing and Quantifying Monoclonal Antibodies (mAb) and Antibody-Drug Conjugates (ADC) in Biological Samples. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/s40495-017-0118-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
40
|
Toward best practices in data processing and analysis for intact biotherapeutics by MS in quantitative bioanalysis. Bioanalysis 2017; 9:1883-1893. [DOI: 10.4155/bio-2017-0179] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Aim: Typically, quantitation of biotherapeutics from biological matrices by LC–MS is based on a surrogate peptide approach to determine molecule concentration. Recent efforts have focused on quantitation of the intact protein molecules or larger mass subunits of monoclonal antibodies. To date, there has been limited guidance for large or intact protein mass quantitation for quantitative bioanalysis. Methodology: Intact- and subunit-level analyses of biotherapeutics from biological matrices are performed at 12–25 kDa mass range with quantitation data presented. Results: Linearity, bias and other metrics are presented along with recommendations made on the viability of existing quantitation approaches. Conclusion: This communication is intended to start a discussion around intact protein data analysis and processing, recognizing that other published contributions will be required.
Collapse
|
41
|
Xu L, Packer LE, Li C, Abdul-Hadi K, Veiby P. A generic approach for simultaneous measurements of total antibody and cleavable antibody-conjugated drug by LC/MS/MS. Anal Biochem 2017; 537:33-36. [DOI: 10.1016/j.ab.2017.08.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/10/2017] [Accepted: 08/30/2017] [Indexed: 11/16/2022]
|
42
|
Qu M, An B, Shen S, Zhang M, Shen X, Duan X, Balthasar JP, Qu J. Qualitative and quantitative characterization of protein biotherapeutics with liquid chromatography mass spectrometry. MASS SPECTROMETRY REVIEWS 2017; 36:734-754. [PMID: 27097288 DOI: 10.1002/mas.21500] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/02/2016] [Indexed: 06/05/2023]
Abstract
In the last decade, the advancement of liquid chromatography mass spectrometry (LC/MS) techniques has enabled their broad application in protein characterization, both quantitatively and qualitatively. Owing to certain important merits of LC/MS techniques (e.g., high selectivity, flexibility, and rapid method development), LC/MS assays are often deemed as preferable alternatives to conventional methods (e.g., ligand-binding assays) for the analysis of protein biotherapeutics. At the discovery and development stages, LC/MS is generally employed for two purposes absolute quantification of protein biotherapeutics in biological samples and qualitative characterization of proteins. For absolute quantification of a target protein in bio-matrices, recent work has led to improvements in the efficiency of LC/MS method development, sample treatment, enrichment and digestion, and high-performance low-flow-LC separation. These advances have enhanced analytical sensitivity, specificity, and robustness. As to qualitative analysis, a range of techniques have been developed to characterize intramolecular disulfide bonds, glycosylation, charge variants, primary sequence heterogeneity, and the drug-to-antibody ratio of antibody drug conjugate (ADC), which has enabled a refined ability to assess product quality. In this review, we will focus on the discussion of technical challenges and strategies of LC/MS-based quantification and characterization of biotherapeutics, with the emphasis on the analysis of antibody-based biotherapeutics such as monoclonal antibodies (mAbs) and ADCs. © 2016 Wiley Periodicals, Inc. Mass Spec Rev 36:734-754, 2017.
Collapse
Affiliation(s)
- Miao Qu
- Beijing University of Chinese Medicine, Beijing, 100029, China
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214
- New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, 14203
| | - Bo An
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214
- New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, 14203
| | - Shichen Shen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214
- New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, 14203
| | - Ming Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214
- New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, 14203
| | - Xiaomeng Shen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214
- New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, 14203
| | - Xiaotao Duan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Joseph P Balthasar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214
| | - Jun Qu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214
- New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, 14203
| |
Collapse
|
43
|
Immunoprecipitation middle-up LC–MS for in vivo drug-to-antibody ratio determination for antibody–drug conjugates. Bioanalysis 2017; 9:1535-1549. [DOI: 10.4155/bio-2017-0148] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Aim: Drug-to-antibody ratio (DAR) determination is critical for development of antibody–drug conjugates (ADCs). This work presents a middle-up LC–MS approach for DAR analysis using prelabeled capture beads and in-house fabricated slit-plates. Methodology & Results: Cysteine, engineered cysteine and disulfide-linked ADCs, each with two different linker payloads, were immunocaptured and digested to scFc and F(ab′)2 fragments. At this point, disulfide-linked ADCs were analyzed while cysteine and engineered cysteine ADCs were reduced to LC and Fd′ fragments for analysis. Results were precise, accurate and sensitive, allowing DAR to be determined out to 21 days. Conclusion: This work describes a method that is easily implemented, amenable to high-throughput analysis and does not require specialized reagents or equipment.
Collapse
|
44
|
Mollazadeh H, Cicero AFG, Blesso CN, Pirro M, Majeed M, Sahebkar A. Immune modulation by curcumin: The role of interleukin-10. Crit Rev Food Sci Nutr 2017; 59:89-101. [PMID: 28799796 DOI: 10.1080/10408398.2017.1358139] [Citation(s) in RCA: 269] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cytokines are small secreted proteins released by different types of cells with specific effects on cellular signaling and communication via binding to their receptors on the cell surface. IL-10 is known to be a pleiotropic and potent anti-inflammatory and immunosuppressive cytokine that is produced by both innate and adaptive immunity cells including dendritic cells, macrophages, mast cells, natural killer cells, eosinophils, neutrophils, B cells, CD8+ T cells, and TH1, TH2, and TH17 and regulatory T cells. Both direct and indirect activation of the stress axis promotes IL-10 secretion. IL-10 deregulation plays a role in the development of a large number of inflammatory diseases such as neuropathic pain, Parkinson's disease, Alzheimer's disease, osteoarthritis, rheumatoid arthritis, psoriasis, systemic lupus erythematosus, type 1 diabetes, inflammatory bowel disease, and allergy. Curcumin is a natural anti-inflammatory compound able to induce the expression and production of IL-10 and enhancing its action on a large number of tissues. In vitro and in pre-clinical models curcumin is able to modulate the disease pathophysiology of conditions such as pain and neurodegenerative diseases, bowel inflammation, and allergy, but also of infections and cancer through its effect on IL-10 secretion. In humans, at least one part of the positive effects of curcumin on health could be related to its ability to enhance IL-10 -mediated effects.
Collapse
Affiliation(s)
- Hamid Mollazadeh
- a Department of Physiology and Pharmacology, School of Medicine , North Khorasan University of Medical Sciences , Bojnurd , Iran
| | - Arrigo F G Cicero
- b Department of Medical and Surgical Sciences , University of Bologna , Via Albertoni 15, Bologna , Italy
| | | | - Matteo Pirro
- d Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine , University of Perugia , Perugia , Italy
| | | | - Amirhossein Sahebkar
- f Department of Medical Biotechnology, Biotechnology Research Center , Mashhad University of Medical Sciences , Mashhad , Iran
| |
Collapse
|
45
|
|
46
|
Protocols for the analytical characterization of therapeutic monoclonal antibodies. I – Non-denaturing chromatographic techniques. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1058:73-84. [DOI: 10.1016/j.jchromb.2017.05.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 11/17/2022]
|
47
|
Kang L, Camacho RC, Li W, D’Aquino K, You S, Chuo V, Weng N, Jian W. Simultaneous Catabolite Identification and Quantitation of Large Therapeutic Protein at the Intact Level by Immunoaffinity Capture Liquid Chromatography–High-Resolution Mass Spectrometry. Anal Chem 2017; 89:6065-6075. [DOI: 10.1021/acs.analchem.7b00674] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Lijuan Kang
- Department of Pharmacokinetics, Dynamics, and Metabolism (PDM) and ‡Department of Cardiovascular Metabolism, Janssen Research & Development, Johnson & Johnson, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Raul C. Camacho
- Department of Pharmacokinetics, Dynamics, and Metabolism (PDM) and ‡Department of Cardiovascular Metabolism, Janssen Research & Development, Johnson & Johnson, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Wenyu Li
- Department of Pharmacokinetics, Dynamics, and Metabolism (PDM) and ‡Department of Cardiovascular Metabolism, Janssen Research & Development, Johnson & Johnson, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Katharine D’Aquino
- Department of Pharmacokinetics, Dynamics, and Metabolism (PDM) and ‡Department of Cardiovascular Metabolism, Janssen Research & Development, Johnson & Johnson, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Seohee You
- Department of Pharmacokinetics, Dynamics, and Metabolism (PDM) and ‡Department of Cardiovascular Metabolism, Janssen Research & Development, Johnson & Johnson, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Vanessa Chuo
- Department of Pharmacokinetics, Dynamics, and Metabolism (PDM) and ‡Department of Cardiovascular Metabolism, Janssen Research & Development, Johnson & Johnson, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Naidong Weng
- Department of Pharmacokinetics, Dynamics, and Metabolism (PDM) and ‡Department of Cardiovascular Metabolism, Janssen Research & Development, Johnson & Johnson, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Wenying Jian
- Department of Pharmacokinetics, Dynamics, and Metabolism (PDM) and ‡Department of Cardiovascular Metabolism, Janssen Research & Development, Johnson & Johnson, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| |
Collapse
|
48
|
He J, Su D, Ng C, Liu L, Yu SF, Pillow TH, Del Rosario G, Darwish M, Lee BC, Ohri R, Zhou H, Wang X, Lu J, Kaur S, Xu K. High-Resolution Accurate-Mass Mass Spectrometry Enabling In-Depth Characterization of in Vivo Biotransformations for Intact Antibody-Drug Conjugates. Anal Chem 2017; 89:5476-5483. [DOI: 10.1021/acs.analchem.7b00408] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Jintang He
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Dian Su
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Carl Ng
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Luna Liu
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Shang-Fan Yu
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Thomas H. Pillow
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | | | - Martine Darwish
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Byoung-Chul Lee
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Rachana Ohri
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Hongxiang Zhou
- Wuxi Apptec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Xueji Wang
- Wuxi Apptec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Jiawei Lu
- Wuxi Apptec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai, 200131, China
| | - Surinder Kaur
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Keyang Xu
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
49
|
Su D, Ng C, Khosraviani M, Yu SF, Cosino E, Kaur S, Xu K. Custom-Designed Affinity Capture LC-MS F(ab′)2 Assay for Biotransformation Assessment of Site-Specific Antibody Drug Conjugates. Anal Chem 2016; 88:11340-11346. [DOI: 10.1021/acs.analchem.6b03410] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Dian Su
- Genentech, Inc. 1 DNA Way, South San Francisco, California 94080, United States
| | - Carl Ng
- Genentech, Inc. 1 DNA Way, South San Francisco, California 94080, United States
| | | | - Shang-Fan Yu
- Genentech, Inc. 1 DNA Way, South San Francisco, California 94080, United States
| | - Ely Cosino
- Genentech, Inc. 1 DNA Way, South San Francisco, California 94080, United States
| | - Surinder Kaur
- Genentech, Inc. 1 DNA Way, South San Francisco, California 94080, United States
| | - Keyang Xu
- Genentech, Inc. 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
50
|
Calculated conjugated payload from immunoassay and LC–MS intact protein analysis measurements of antibody-drug conjugate. Bioanalysis 2016; 8:2205-2217. [DOI: 10.4155/bio-2016-0160] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Aim: Complex nature of bioconjugates require multiple bioanalytical approaches to support PK and absorption, distribution, metabolism and excretion characterization. For antibody-drug conjugate (ADC) bioanalysis both LC–MS and ligand-binding assays (LBAs) are employed. Results: A method consisting of immunocapture extraction of ADC from biomatrices followed by LC–MS analysis of light and heavy chain is described. Drug antibody ratio (DAR) profiles of ADC Tras-mcVC-PF06380101 dosed at 0.3, 1 and 3 mg/kg in Sprague Dawley rats were obtained. Combined with total antibody (monoclonal antibody) measurement by LBA, conjugated payload concentration was calculated. Conclusion: PK profiles from LBA, ADC and calculated conjugated payload (DAR × monoclonal antibody) were in good agreement. We present a new tool for PK assessment of ADCs while also exploring ADC metabolism and DAR sensitivity of LBA ADC assay.
Collapse
|