1
|
Vazvaei-Smith F, Li W, Barnaby OS, Bhardwaj S, Dong J, Dumont C, Fernández-Metzler C, Geist B, Hassanein M, Hays A, Ilinskaya A, Kadar EP, King K, Kulagina N, Matta MK, Midde K, Pan R, Pathania D, Tarnowski T, Tewalt E, Thomas E, Wickremsinhe E, Xiao D. Best Practices and Recommendations for Non-Liquid Matrices Bioanalysis. AAPS J 2025; 27:57. [PMID: 40069400 DOI: 10.1208/s12248-025-01033-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 01/31/2025] [Indexed: 03/25/2025] Open
Abstract
The analysis of Non-Liquid Matrices (NLMs) can provide key information on many aspects in drug discovery and development. These include but are not limited to drug uptake and distribution, engagement and modulation, and target exposure. A thorough understanding of these aspects is fundamental to the progression of drug development. In many cases, such an understanding can only be achieved through quantitative analysis of NLMs. Such dependence can lead to bottlenecks in the drug development process-as the practices and regulations that govern bioanalysis of conventional liquid matrices typically cannot be directly applied to NLMs. This paper strives to fill this crucial gap. To this end, subject matter experts from across the industry, through the auspices of the AAPS Bioanalytical Community, have combined their collective best practices for NLM bioanalysis in this paper. Certainly, this endeavor came with challenges, the most prominent of which also serves as the impetus for this project, the lack of literature on NLM bioanalysis dealing with different types of NLM, analysis rigor, and best practices to draw from. This paper aims to serve as a comprehensive set of best practices drawn from the experiences of leading scientists across the industry-for NLM bioanalysis in drug development.
Collapse
Affiliation(s)
| | - Wenkui Li
- Novartis Biomedical Research, East Hanover, New Jersey, 07936, USA.
| | | | | | - Juyao Dong
- Intellia Therapeutics, Cambridge, Massachusetts, 02139, USA
| | | | | | - Brian Geist
- Johnson & Johnson Innovative Medicine, Spring House, PA, 19477, USA
| | | | - Amanda Hays
- BioAgilytix Labs, Durham, North Carolina, 27713, USA
| | | | | | - Kris King
- Valo Health, Lexington, Massachusetts, 02421, USA
| | | | - Murali K Matta
- Merck & Co., Inc., West Point, Pennsylvania, 19486, USA
- Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | | | - Rina Pan
- Incyte Research Institute, Wilmington, Delaware, 19803, USA
| | | | | | - Eric Tewalt
- PPD, part of Thermo Fisher Scientific, Richmond, Virginia, 23230, USA
| | | | | | - Deqing Xiao
- Gilead Sciences, Inc., Foster City, California, 94404, USA
| |
Collapse
|
2
|
Luettel DM, Terluk MR, Roh J, Weinreb NJ, Kartha RV. Emerging biomarkers in Gaucher disease. Adv Clin Chem 2025; 124:1-56. [PMID: 39818434 DOI: 10.1016/bs.acc.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Gaucher disease (GD) is a rare lysosomal disorder characterized by the accumulation of glycosphingolipids in macrophages resulting from glucocerebrosidase (GCase) deficiency. The accumulation of toxic substrates, which causes the hallmark symptoms of GD, is dependent on the extent of enzyme dysfunction. Accordingly, three distinct subtypes have been recognized, with type 1 GD (GD1) as the common and milder form, while types 2 (GD2) and 3 (GD3) are categorized as neuronopathic and severe. Manifestations variably include hepatosplenomegaly, anemia, thrombocytopenia, easy bruising, inflammation, bone pain and other skeletal pathologies, abnormal eye movements and neuropathy. Although the molecular basis of GD is relatively well understood, currently used biomarkers are nonspecific and inadequate for making finer distinctions between subtypes and in evaluating changes in disease status and guiding therapy. Thus, there is continued effort to investigate and identify potential biomarkers to improve GD diagnosis, monitoring and potential identification of novel therapeutic targets. Here, we provide a comprehensive review of emerging biomarkers in GD that can enhance current understanding and improve quality of life through better testing, disease management and treatment.
Collapse
Affiliation(s)
- Danielle M Luettel
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Marcia R Terluk
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Jaehyeok Roh
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Neal J Weinreb
- Department of Human Genetics, Leonard Miller School of Medicine of University of Miami, Miami, FL, United States
| | - Reena V Kartha
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
3
|
Zararsiz GE, Lintelmann J, Cecil A, Kirwan J, Poschet G, Gegner HM, Schuchardt S, Guan XL, Saigusa D, Wishart D, Zheng J, Mandal R, Adams K, Thompson JW, Snyder MP, Contrepois K, Chen S, Ashrafi N, Akyol S, Yilmaz A, Graham SF, O’Connell TM, Kalecký K, Bottiglieri T, Limonciel A, Pham HT, Koal T, Adamski J, Kastenmüller G. Interlaboratory comparison of standardised metabolomics and lipidomics analyses in human and rodent blood using the MxP ® Quant 500 kit. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.619447. [PMID: 39605511 PMCID: PMC11601468 DOI: 10.1101/2024.11.13.619447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Metabolomics and lipidomics are pivotal in understanding phenotypic variations beyond genomics. However, quantification and comparability of mass spectrometry (MS)-derived data are challenging. Standardised assays can enhance data comparability, enabling applications in multi-center epidemiological and clinical studies. Here we evaluated the performance and reproducibility of the MxP® Quant 500 kit across 14 laboratories. The kit allows quantification of 634 different metabolites from 26 compound classes using triple quadrupole MS. Each laboratory analysed twelve samples, including human plasma and serum, lipaemic plasma, NIST SRM 1950, and mouse and rat plasma, in triplicates. 505 out of the 634 metabolites were measurable above the limit of detection in all laboratories, while eight metabolites were undetectable in our study. Out of the 505 metabolites, 412 were observed in both human and rodent samples. Overall, the kit exhibited high reproducibility with a median coefficient of variation (CV) of 14.3 %. CVs in NIST SRM 1950 reference plasma were below 25 % and 10 % for 494 and 138 metabolites, respectively. To facilitate further inspection of reproducibility for any compound, we provide detailed results from the in-depth evaluation of reproducibility across concentration ranges using Deming regression. Interlaboratory reproducibility was similar across sample types, with some species-, matrix-, and phenotype-specific differences due to variations in concentration ranges. Comparisons with previous studies on the performance of MS-based kits (including the AbsoluteIDQ p180 and the Lipidyzer) revealed good concordance of reproducibility results and measured absolute concentrations in NIST SRM 1950 for most metabolites, making the MxP® Quant 500 kit a relevant tool to apply metabolomics and lipidomics in multi-center studies.
Collapse
Affiliation(s)
- Gözde Ertürk Zararsiz
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Biostatistics, Erciyes University School of Medicine, Kayseri, Turkey
- Drug Application and Research Center (ERFARMA), Erciyes University, Kayseri, Turkey
| | - Jutta Lintelmann
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Alexander Cecil
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Jennifer Kirwan
- Metabolomics Platform, Berlin Institute of Health at Charité, Berlin, Germany
| | - Gernot Poschet
- Metabolomics Core Technology Platform, Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany
| | - Hagen M. Gegner
- Metabolomics Core Technology Platform, Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany
| | - Sven Schuchardt
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Xue Li Guan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Daisuke Saigusa
- Laboratory of Biomedical and Analytical Sciences, Faculty of Pharmaceutical Science, Teikyo University, Tokyo, Japan
| | - David Wishart
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
| | - Jiamin Zheng
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
| | - Rupasri Mandal
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
| | - Kendra Adams
- Duke Proteomics and Metabolomics Shared Resource, Center for Genomic and Computational Biology, Duke University, Durham (NC), USA
| | - J. Will Thompson
- Duke Proteomics and Metabolomics Shared Resource, Center for Genomic and Computational Biology, Duke University, Durham (NC), USA
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford (CA), USA
| | - Kevin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford (CA), USA
| | - Songjie Chen
- Department of Genetics, Stanford University School of Medicine, Stanford (CA), USA
| | - Nadia Ashrafi
- Corewell Health Research Institute, Metabolomics Department, Royal Oak (MI), USA
- Corewell Health William Beaumont University Hospital, Royal Oak (MI), USA
| | - Sumeyya Akyol
- Corewell Health Research Institute, Metabolomics Department, Royal Oak (MI), USA
| | - Ali Yilmaz
- Corewell Health Research Institute, Metabolomics Department, Royal Oak (MI), USA
- Corewell Health William Beaumont University Hospital, Royal Oak (MI), USA
- Oakland University-William Beaumont School of Medicine, Rochester (MI), USA
| | - Stewart F. Graham
- Corewell Health Research Institute, Metabolomics Department, Royal Oak (MI), USA
- Corewell Health William Beaumont University Hospital, Royal Oak (MI), USA
- Oakland University-William Beaumont School of Medicine, Rochester (MI), USA
| | | | - Karel Kalecký
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas (TX), USA
| | - Teodoro Bottiglieri
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas (TX), USA
| | | | | | | | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
4
|
Saito K, Goda R, Arai K, Asahina K, Kawabata M, Uchiyama H, Andou T, Shimizu H, Takahara K, Kakehi M, Yamauchi S, Nitta SI, Suga T, Fujita H, Ishikawa R, Saito Y. Interlaboratory evaluation of LC-MS-based biomarker assays. Bioanalysis 2024; 16:389-402. [PMID: 38334082 DOI: 10.4155/bio-2023-0173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024] Open
Abstract
Validation of biomarker assays is crucial for effective drug development and clinical applications. Interlaboratory reproducibility is vital for reliable comparison and combination of data from different centers. This review summarizes interlaboratory studies of quantitative LC-MS-based biomarker assays using reference standards for calibration curves. The following points are discussed: trends in reports, reference and internal standards, evaluation of analytical validation parameters, study sample analysis and normalization of biomarker assay data. Full evaluation of these parameters in interlaboratory studies is limited, necessitating further research. Some reports suggest methods to address variations in biomarker assay data among laboratories, facilitating organized studies and data combination. Method validation across laboratories is crucial for reducing interlaboratory differences and reflecting target biomarker responses.
Collapse
Affiliation(s)
- Kosuke Saito
- National Institute of Health Sciences, Kanagawa, Japan
| | - Ryoya Goda
- Daiichi Sankyo Company Ltd, Tokyo, Japan
| | - Koji Arai
- LSI Medience Corporation, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | - Rika Ishikawa
- National Institute of Health Sciences, Kanagawa, Japan
| | - Yoshiro Saito
- National Institute of Health Sciences, Kanagawa, Japan
| |
Collapse
|
5
|
Hickford ES, Dejager L, Yuill D, Kotian A, Shankar S, Staelens L, Ulrichts H, Lewis S, Louber J, Williams A, Le Provost GS, Cutler P. A biomarker assay validation approach tailored to the context of use and bioanalytical platform. Bioanalysis 2023; 15:757-771. [PMID: 37526064 DOI: 10.4155/bio-2023-0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023] Open
Abstract
It is widely acknowledged by the bioanalytical and biomarker community that biomarker assay validations should be fit-for-purpose depending on the context of use. The challenge is how to consistently apply these principles in teams responsible for measuring a disparate array of biomarkers, often on multiple analytical platforms, at various stages of the drug discovery and development pipeline and across diverse biology focus areas. To drive consistency, while maintaining the necessary flexibility to allow validations to be driven by scientific rationale and taking into consideration the context of use and associated biological and (pre)analytical factors, a framework applicable across biomarker assays was developed. Herein the authors share their perspective to engage in the ongoing conversation around fit-for-purpose biomarker assay validation.
Collapse
Affiliation(s)
- Elizabeth S Hickford
- Translational Biomarkers & Bioanalysis, Development Sciences, UCB Biopharma UK, Bath Road, Slough, SL1 3WE, UK
| | - Lien Dejager
- Precision Medicine & Biomarkers, Translational Medicine, UCB Pharma, Chemin du Foriest, B-1420 Braine-l'Alleud, Belgium
| | - Daisy Yuill
- Translational Biomarkers & Bioanalysis, Development Sciences, UCB Biopharma UK, Bath Road, Slough, SL1 3WE, UK
| | - Apoorva Kotian
- Translational Biomarkers & Bioanalysis, Development Sciences, UCB Biopharma UK, Bath Road, Slough, SL1 3WE, UK
| | - Sucharita Shankar
- Translational Biomarkers & Bioanalysis, Development Sciences, UCB Biopharma UK, Bath Road, Slough, SL1 3WE, UK
| | - Ludovicus Staelens
- Translational Biomarkers & Bioanalysis, Development Sciences, UCB Pharma, Chemin du Foriest, B-1420 Braine l'Alleud, Belgium
| | - Hans Ulrichts
- Translational Biomarkers & Bioanalysis, Development Sciences, UCB Pharma, Chemin du Foriest, B-1420 Braine l'Alleud, Belgium
- Employed by UCB Pharma, Belgium or UCB Biopharma UK at the time the work was undertaken
| | - Sion Lewis
- Translational Biomarkers & Bioanalysis, Development Sciences, UCB Biopharma UK, Bath Road, Slough, SL1 3WE, UK
| | - Jade Louber
- Translational Biomarkers & Bioanalysis, Development Sciences, UCB Biopharma UK, Bath Road, Slough, SL1 3WE, UK
- Employed by UCB Pharma, Belgium or UCB Biopharma UK at the time the work was undertaken
| | - Amanda Williams
- Translational Biomarkers & Bioanalysis, Development Sciences, UCB Biopharma UK, Bath Road, Slough, SL1 3WE, UK
| | - Gabrielle S Le Provost
- Translational Biomarkers & Bioanalysis, Development Sciences, UCB Biopharma UK, Bath Road, Slough, SL1 3WE, UK
| | - Paul Cutler
- Translational Biomarkers & Bioanalysis, Development Sciences, UCB Biopharma UK, Bath Road, Slough, SL1 3WE, UK
| |
Collapse
|
6
|
Mulvanny A, Pattwell C, Beech A, Southworth T, Singh D. Validation of Sputum Biomarker Immunoassays and Cytokine Expression Profiles in COPD. Biomedicines 2022; 10:biomedicines10081949. [PMID: 36009496 PMCID: PMC9405928 DOI: 10.3390/biomedicines10081949] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Immunoassays are commonly used to assess airway inflammation in sputum samples from chronic obstructive pulmonary disease (COPD) patients. However, assay performance and validation in this complex matrix is inconsistently reported. The aim of this study was to assess the suitability of various immunoassays for use with sputum samples, followed by use of validated immunoassays to evaluate biomarker levels in COPD patients. Assays were assessed for recombinant reference standard suitability, optimal sample dilution, standard recovery in the biological matrix and reproducibility. Validated assays were used to assess sputum supernatants in Cohort A (n = 30 COPD, n = 10 smokers, n = 10 healthy) and Cohort B (n = 81 COPD, n = 15 smokers, n = 26 healthy). Paired baseline and exacerbation samples from 14 COPD patients were assessed in cohort A, and associations with sputum cell counts and bacterial colonisation investigated in cohort B. 25/32 assays passed validation; the primary reason for validation failure was recombinant reference standard suitability and sample dilution effects. Interleukin (IL-)6 and IL-8 were significantly increased in COPD patients compared to healthy subjects and smokers for both cohorts. Tumour necrosis factor (TNF)α and IL-1β were higher in COPD compared to smokers using one immunoassay but not another, partly explained by different absolute recovery rates. IL-1β, IL-2, IL-4, IL-8, IL-17A, Granulocyte colony stimulating factor (G-CSF), Interferon (IFN-)γ, Interferon gamma induced protein (IP-)10, Macrophage inflammatory protein (MIP)-1α, MIP-1β and TNF-α levels correlated with sputum neutrophil percentage in COPD patients. IL-1β, IL-4, IL-8, G-CSF and IFN-γ levels were associated with Haemophilus influenzae colonisation in COPD patients. Current smokers had lower levels of IL-1β, IL-4, IL-8, G-CSF, IFN-γ, IP-10, Monocyte chemoattractant protein (MCP)-1, MIP-1α, MIP-1β and TNF-α. Validated immunoassays applied to sputum supernatants demonstrated differences between COPD patients and controls, the effects of current smoking and associations between Haemophilus influenzae colonisation and higher levels of selected cytokines. Immunoassay validation enabled inflammatory mediators associated with different COPD characteristics to be determined.
Collapse
Affiliation(s)
- Alex Mulvanny
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and Manchester University NHS Foundation Trust, Manchester M13 9PL, UK
- Medicines Evaluation Unit, Manchester University NHS Foundation Trust, Manchester M23 9QZ, UK
- Correspondence: ; Tel.: +44-0161-946-4050
| | - Caroline Pattwell
- Medicines Evaluation Unit, Manchester University NHS Foundation Trust, Manchester M23 9QZ, UK
| | - Augusta Beech
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and Manchester University NHS Foundation Trust, Manchester M13 9PL, UK
| | - Thomas Southworth
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and Manchester University NHS Foundation Trust, Manchester M13 9PL, UK
- Medicines Evaluation Unit, Manchester University NHS Foundation Trust, Manchester M23 9QZ, UK
| | - Dave Singh
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and Manchester University NHS Foundation Trust, Manchester M13 9PL, UK
- Medicines Evaluation Unit, Manchester University NHS Foundation Trust, Manchester M23 9QZ, UK
| |
Collapse
|
7
|
Validation of a method to analyze size distribution of crovalimab-complement C5-eculizumab complexes in human serum. Bioanalysis 2022; 14:935-947. [PMID: 35904159 DOI: 10.4155/bio-2022-0116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: Crovalimab is a humanized monoclonal antibody targeting human complement C5. Patients switching from eculizumab to crovalimab are expected to form drug-target-drug complexes (DTDCs), since these antibodies each bind to a different epitope on complement C5. An analytical method to evaluate the size distribution of these DTDCs was developed and validated. Methods: Human serum samples were separated by size-exclusion chromatography (SEC) into eight fractions, and the concentration of crovalimab in each fraction was measured by ELISA. We evaluated SEC, ELISA and the combination of both methods (SEC-ELISA). Results: Predetermined validation acceptance criteria were met. Conclusion: The DTDC assay method was successfully validated. It enables us to evaluate the impact of DTDCs on clinical outcomes.
Collapse
|
8
|
Fernández-Metzler C, Ackermann B, Garofolo F, Arnold ME, DeSilva B, Gu H, Laterza O, Mao Y, Rose M, Vazvaei-Smith F, Steenwyk R. Biomarker Assay Validation by Mass Spectrometry. AAPS J 2022; 24:66. [PMID: 35534647 DOI: 10.1208/s12248-022-00707-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/31/2022] [Indexed: 11/30/2022] Open
Abstract
Decades of discussion and publication have gone into the guidance from the scientific community and the regulatory agencies on the use and validation of pharmacokinetic and toxicokinetic assays by chromatographic and ligand binding assays for the measurement of drugs and metabolites. These assay validations are well described in the FDA Guidance on Bioanalytical Methods Validation (BMV, 2018). While the BMV included biomarker assay validation, the focus was on understanding the challenges posed in validating biomarker assays and the importance of having reliable biomarker assays when used for regulatory submissions, rather than definition of the appropriate experiments to be performed. Different from PK bioanalysis, analysis of biomarkers can be challenging due to the presence of target analyte(s) in the control matrices used for calibrator and quality control sample preparation, and greater difficulty in procuring appropriate reference standards representative of the endogenous molecule. Several papers have been published offering recommendations for biomarker assay validation. The situational nature of biomarker applications necessitates fit-for-purpose (FFP) assay validation. A unifying theme for FFP analysis is that method validation requirements be consistent with the proposed context of use (COU) for any given biomarker. This communication provides specific recommendations for biomarker assay validation (BAV) by LC-MS, for both small and large molecule biomarkers. The consensus recommendations include creation of a validation plan that contains definition of the COU of the assay, use of the PK assay validation elements that support the COU, and definition of assay validation elements adapted to fit biomarker assays and the acceptance criteria for both.
Collapse
Affiliation(s)
| | - Brad Ackermann
- Eli Lilly & Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Fabio Garofolo
- BRI - a Frontage Company, 8898 Heather St, Vancouver, British Columbia, V6P 3S8, Canada
| | - Mark E Arnold
- Labcorp Drug Development, 221 Tulip Tree Drive, Westampton, NJ, 08060-5511, USA
| | - Binodh DeSilva
- Bristol-Myers Squibb Co., Route 206 & Province Line Road, Princeton, NJ, 08543, USA
| | - Huidong Gu
- Bristol-Myers Squibb Co., Route 206 & Province Line Road, Princeton, NJ, 08543, USA
| | - Omar Laterza
- Merck and Co Inc., 90 E Scott Ave, Rahway, NJ, 07065, USA
| | - Yan Mao
- Boehringer-Ingelheim Pharmaceuticals, 900 Ridgebury Road, Ridgefield, CT, 06877, USA
| | - Mark Rose
- Gossamer Bio Inc., 3013 Science Park Road, Suite 200, San Diego, CA, 92121, USA
| | | | - Rick Steenwyk
- Pfizer-Retired, 8739 N Homestead Circle, Irons, MI, 49644, USA
| |
Collapse
|
9
|
Ishikawa R, Saito K, Matsumura T, Arai K, Yamauchi S, Goda R, Tachiki H, Kawabata M, Nitta SI, Nagao A, Suga T, Uchiyama H, Nakai K, Asahina K, Yamaoka M, Saito Y. A multilaboratory validation study of LC/MS biomarker assays for three lysophosphatidylcholines. Bioanalysis 2021; 13:1533-1546. [PMID: 34696608 DOI: 10.4155/bio-2021-0150] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/13/2021] [Indexed: 11/17/2022] Open
Abstract
Aim: Although the fit-for-purpose approach has been proposed for validation procedures and acceptance criteria for biomarker assays, practical biomarker assays to facilitate clinical application and regulatory documents on biomarker assays remain limited. Materials & methods: We assigned six independent laboratories and selected three lysophosphatidylcholines (LPCs): LPC(16:0), LPC(18:0) and LPC(18:1) as model biomarkers. Using LC-MS, the following key validation parameters were evaluated: calibration curve, carryover, parallelism, precision and relative accuracy and these values were similar among all laboratories. Further, we determined LPC levels in six lots of rat plasma at unknown concentrations and compared them among the laboratories. Conclusion: Our multilaboratory validation and reproducibility data are useful for the development of future biomarker assay validation procedures, as well as regulatory documents.
Collapse
Affiliation(s)
- Rika Ishikawa
- Division of Medical Safety Science, National Institute of Health Sciences, Kanagawa, 210-9501, Japan
| | - Kosuke Saito
- Division of Medical Safety Science, National Institute of Health Sciences, Kanagawa, 210-9501, Japan
| | | | - Koji Arai
- LSI Medience Corporation, Tokyo, 101-8517, Japan
| | | | - Ryoya Goda
- Daiichi Sankyo Company, Ltd, Tokyo, 140-8710, Japan
| | | | | | | | | | | | | | - Keiko Nakai
- LSI Medience Corporation, Tokyo, 101-8517, Japan
| | | | | | - Yoshiro Saito
- Division of Medical Safety Science, National Institute of Health Sciences, Kanagawa, 210-9501, Japan
| |
Collapse
|
10
|
Huisinga M, Bertrand L, Chamanza R, Damiani I, Engelhardt J, Francke S, Freyberger A, Harada T, Harleman J, Kaufmann W, Keane K, Köhrle J, Lenz B, Marty MS, Melching-Kollmuss S, Palazzi X, Pohlmeyer-Esch G, Popp A, Rosol TJ, Strauss V, Van den Brink-Knol H, Wood CE, Yoshida M. Adversity Considerations for Thyroid Follicular Cell Hypertrophy and Hyperplasia in Nonclinical Toxicity Studies: Results From the 6th ESTP International Expert Workshop. Toxicol Pathol 2021; 48:920-938. [PMID: 33334259 DOI: 10.1177/0192623320972009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The European Society of Toxicologic Pathology organized an expert workshop in May 2018 to address adversity considerations related to thyroid follicular cell hypertrophy and/or hyperplasia (FCHH), which is a common finding in nonclinical toxicity studies that can have important implications for risk assessment of pharmaceuticals, food additives, and environmental chemicals. The broad goal of the workshop was to facilitate better alignment in toxicologic pathology and regulatory sciences on how to determine adversity of FCHH. Key objectives were to describe common mechanisms leading to thyroid FCHH and potential functional consequences; provide working criteria to assess adversity of FCHH in context of associated findings; and describe additional methods and experimental data that may influence adversity determinations. The workshop panel was comprised of representatives from the European Union, Japan, and the United States. Participants shared case examples illustrating issues related to adversity assessments of thyroid changes. Provided here are summary discussions, key case presentations, and panel recommendations. This information should increase consistency in the interpretation of adverse changes in the thyroid based on pathology findings in nonclinical toxicity studies, help integrate new types of biomarker data into the review process, and facilitate a more systematic approach to communicating adversity determinations in toxicology reports.
Collapse
Affiliation(s)
| | - Lise Bertrand
- 57146Charles River Laboratories, Saint-Germain-Nuelles, France
| | - Ronnie Chamanza
- 50148Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| | | | | | - Sabine Francke
- Center for Food Safety and Applied Nutrition (CFSAN), 4137US Food and Drug Administration, College Park, MD, USA
| | | | | | | | | | | | - Josef Köhrle
- 72217Charité University Medicine Berlin, Berlin, Germany
| | - Barbara Lenz
- Roche Pharma Research and Development, Basel, Switzerland
| | - M Sue Marty
- 540144The Dow Chemical Company, Midland, MI, USA
| | | | | | | | | | | | | | | | - Charles E Wood
- 6893Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| | | |
Collapse
|
11
|
A normalized signal calibration with a long-term reference improves the robustness of RPLC-MRM/MS lipidomics in plasma. Anal Bioanal Chem 2021; 413:4077-4090. [PMID: 33907864 DOI: 10.1007/s00216-021-03364-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/19/2021] [Indexed: 10/21/2022]
Abstract
Improving the reliability of quantification in lipidomic analyses is crucial for its successful application in the discovery of new biomarkers or in clinical practice. In this study, we propose a workflow to improve the accuracy and precision of lipidomic results issued by the laboratory. Lipid species from 11 classes were analyzed by a targeted RPLC-MRM/MS method. The peak areas of species were used to estimate concentrations by an internal standard calibration approach (IS-calibration) and by an alternative normalization signal calibration schema (NS-calibration). The latter uses a long-term reference plasma material as a matrix-matched external calibrator whose accuracy was compared to the NIST SRM-1950 mean consensus values reported by the Interlaboratory Lipidomics Comparison Exercise. The bias of lipid concentrations showed a good accuracy for 69 of 89 quantified lipids. The quantitation of species by the NS-calibration schema improved the within- and between-batch reproducibility in quality control samples, in comparison to the usual IS-calibration approach. Moreover, the NS-calibration workflow improved the robustness of the lipidomics measurements reducing the between-batch variability (relative standard deviation <10% for 95% of lipid species) in real conditions tested throughout the analysis of 120 plasma samples. In addition, we provide a free access web tool to obtain the concentration of lipid species by the two previously mentioned quantitative approaches, providing an easy follow-up of quality control tasks related to lipidomics.
Collapse
|
12
|
Wang B, Shen J, Zhou Q, Meng D, He Y, Chen F, Wang S, Ji W. Effects of naringenin on the pharmacokinetics of tofacitinib in rats. PHARMACEUTICAL BIOLOGY 2020; 58:225-230. [PMID: 32202190 PMCID: PMC7144329 DOI: 10.1080/13880209.2020.1738504] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 01/12/2020] [Accepted: 02/28/2020] [Indexed: 06/10/2023]
Abstract
Context: Naringenin and tofacitinib are often used together for treatment of rheumatoid arthritis in Chinese clinics.Objective: This experiment investigates the effect of naringenin on the pharmacokinetics of tofacitinib in rats.Materials and methods: Twelve Sprague-Dawley rats were randomly divided into two groups (experimental group and control group). The experimental group was pre-treated with naringenin (150 mg/kg/day) for two weeks before dosing tofacitinib, and equal amounts of CMC-Na solution in the control group. After a single oral administration of 5 mg/kg of tofacitinib, 50 μL blood samples were directly collected into 1.5 mL heparinized tubes via the caudal vein at 0.083, 0.5, 1, 2, 3, 4, 6, 8, 10, 12 and 24 h. The plasma concentration of tofacitinib was quantified by UPLC/MS-MS.Results: Results indicated that naringenin could significantly affect the pharmacokinetics of tofacitinib. The AUC0-24 of tofacitinib was increased from 1222.81 ± 222.07 to 2016.27 ± 481.62 ng/mL/h, and the difference was significant (p < 0.05). Compared with the control group, the Tmax was increased from 0.75 ± 0.29 to 3.00 ± 0.00 h (p < 0.05), and the MRT(0-24) was increased from 4.90 ± 0.51 to 6.57 ± 0.66 h (p < 0.05), but the clearance was obviously decreased from 4.10 ± 0.72 to 2.42 ± 0.70 L/h/kg (p < 0.05) in experimental group. Although the Cmax and t1/2 of tofacitinib were increased, there were no significant differences (p > 0.05).Conclusions: This research demonstrated a drug-drug interaction between naringenin and tofacitinib possibly when preadministered with naringenin; thus, we should pay attention to this possibility in the clinic.
Collapse
Affiliation(s)
- Bo Wang
- Department of Orthopaedics, The Sixth Affiliated Hospital of Wenzhou Medical University, The People’s Hospital of Lishui, Lishui, China
| | - Jiquan Shen
- Department of Orthopaedics, The Sixth Affiliated Hospital of Wenzhou Medical University, The People’s Hospital of Lishui, Lishui, China
| | - Quan Zhou
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People’s Hospital of Lishui, Lishui, China
| | - Deru Meng
- School of Medicine, Yichun University, Yichun, China
| | - Youwu He
- Department of Orthopaedics, The Sixth Affiliated Hospital of Wenzhou Medical University, The People’s Hospital of Lishui, Lishui, China
| | - Feifei Chen
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People’s Hospital of Lishui, Lishui, China
| | - Shuanghu Wang
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People’s Hospital of Lishui, Lishui, China
- School of Pharmaceutical Science, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, China
| | - Weiping Ji
- Department of Orthopaedics, The Sixth Affiliated Hospital of Wenzhou Medical University, The People’s Hospital of Lishui, Lishui, China
| |
Collapse
|
13
|
Conklin LS, Hoffman EP, van den Anker J. Developmental Pharmacodynamics and Modeling in Pediatric Drug Development. J Clin Pharmacol 2020; 59 Suppl 1:S87-S94. [PMID: 31502687 DOI: 10.1002/jcph.1482] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022]
Abstract
Challenges in pediatric drug development include small patient numbers, limited outcomes research, ethical barriers, and sparse biosamples. Increasingly, pediatric drug development is focusing on extrapolation: leveraging knowledge about adult disease and drug responses to inform projections of drug and clinical trial performance in pediatric subpopulations. Pharmacokinetic-pharmacodynamic (PK-PD) modeling and extrapolation aim to reduce the numbers of patients and data points needed to establish efficacy. Planning for PK-PD and biomarker studies should begin early in the adult drug development program. Extrapolation relies on the assumption that both the underlying disease and the mechanism of action of the drug used to treat that disease are similar in adults and pediatric subpopulations. Clearly, developmental changes in PK and PD need to be considered to enhance the quality of PK-PD modeling and, therefore, increase the success of extrapolation. This article focuses on the influence of differences in PD between adults and pediatric subpopulations that are highly relevant for the use of extrapolation.
Collapse
Affiliation(s)
- Laurie S Conklin
- Division of Gastroenterology, Hepatology, and Nutrition, Children's National Health System, Washington, DC, USA.,ReveraGen BioPharma, Rockville, MD, USA
| | - Eric P Hoffman
- ReveraGen BioPharma, Rockville, MD, USA.,Binghamton University-SUNY, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - John van den Anker
- ReveraGen BioPharma, Rockville, MD, USA.,Division of Clinical Pharmacology, Children's National Health System, Washington, DC, USA
| |
Collapse
|
14
|
Ruggiero C, Ramirez S, Ramazzotti E, Mancini R, Muratori R, Raggi MA, Conti M. Multiplexed therapeutic drug monitoring of antipsychotics in dried plasma spots by LC-MS/MS. J Sep Sci 2020; 43:1440-1449. [PMID: 32077627 DOI: 10.1002/jssc.201901200] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/31/2020] [Accepted: 02/01/2020] [Indexed: 01/19/2023]
Abstract
In this work, a convenient method for the therapeutic monitoring of seven common antipsychotic drugs in "dried plasma spot" samples has been developed. It is based on the liquid chromatography tandem mass spectrometry technique, operating in multiple reaction monitoring mode, and a straightforward procedure for the simultaneous extraction of all antipsychotics in a single step, with high extraction yield. The method was fully validated with proper accuracy, precision, selectivity and sensitivity, for all the drugs. Limits of quantification were 0.12, 1.09, 1.46, 1.47, 5.70, 1.32, 1.33 µg/L for haloperidol, aripiprazole, olanzapine, quetiapine, clozapine, risperidone, and paliperidone, respectively. Accuracy, intra- and interday precision values were <10% for all drugs at all concentration levels examined. The method was tested in the analysis of 30 plasma samples from real patients for each drug. The proposed analytical approach, by combining practical and logistical advantages of microsampling with liquid chromatography tandem mass spectrometry analytical performance, could offer an ideal strategy for accurate and timely therapeutic drug monitoring of antipsychotic drugs in most clinical settings, even in remote centers and/or in out-patient settings, bringing so many potential improvements in psychiatric patient care.
Collapse
Affiliation(s)
- Carla Ruggiero
- LUM Metropolitan Laboratory, AUSL Bologna, Bologna, Italy
| | | | | | - Rita Mancini
- LUM Metropolitan Laboratory, AUSL Bologna, Bologna, Italy
| | | | | | - Matteo Conti
- LUM Metropolitan Laboratory, AUSL Bologna, Bologna, Italy
| |
Collapse
|
15
|
Glycosaminoglycans in biological samples – Towards identification of novel biomarkers. Trends Analyt Chem 2020. [DOI: 10.1016/j.trac.2019.115732] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
16
|
Development and evaluation of an ultrasensitive free VEGF-A immunoassay for analysis of human aqueous humor. Bioanalysis 2019; 11:875-886. [DOI: 10.4155/bio-2019-0044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aim: Novel bifunctional VEGF-A neutralizing therapies are being developed for the treatment of retinal vascular diseases such as age-related macular degeneration and diabetic retinopathy. In developing new therapeutic drugs, only small aqueous humor sample volumes are available for analyzing several parameters. Highly sensitive detection methods must be applied in analyzing VEGF-A levels in ocular fluids in order to demonstrate VEGF-A suppression following drug administration. Experimental: A highly sensitive immunoassay for VEGF-A was developed on the single molecule array (Simoa) platform, and validated before being used for the analysis of clinical aqueous humor samples from patients treated with anti-VEGF-A therapeutics. Results: This highly sensitive immunoassay allows the detection of baseline VEGF-A levels and suppression effects after drug administration, even in sample volumes as low as 12 μl. Conclusion: The Simoa VEGF-A assay is a valuable tool for the reliable monitoring of VEGF-A suppression after intravitreal administration of anti-VEGF-A drugs.
Collapse
|
17
|
Li AA, Makris SL, Marty MS, Strauss V, Gilbert ME, Blacker A, Zorrilla LM, Coder PS, Hannas B, Lordi S, Schneider S. Practical considerations for developmental thyroid toxicity assessments: What's working, what's not, and how can we do better? Regul Toxicol Pharmacol 2019; 106:111-136. [PMID: 31018155 DOI: 10.1016/j.yrtph.2019.04.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/11/2019] [Accepted: 04/14/2019] [Indexed: 12/26/2022]
Abstract
Thyroid hormones (THs; T3 and T4) play a role in development of cardiovascular, reproductive, immune and nervous systems. Thus, interpretation of TH changes from rodent studies (during pregnancy, in fetuses, neonates, and adults) is critical in hazard characterization and risk assessment. A roundtable session at the 2017 Society of Toxicology (SOT) meeting brought together academic, industry and government scientists to share knowledge and different perspectives on technical and data interpretation issues. Data from a limited group of laboratories were compiled for technical discussions on TH measurements, including good practices for reliable serum TH data. Inter-laboratory historical control data, derived from immunoassays or mass spectrometry methods, revealed: 1) assay sensitivities vary within and across methodologies; 2) TH variability is similar across animal ages; 3) laboratories generally achieve sufficiently sensitive TH quantitation levels, although issues remain for lower levels of serum TH and TSH in fetuses and postnatal day 4 pups; thus, assay sensitivity is critical at these life stages. Best practices require detailed validation of rat serum TH measurements across ages to establish assay sensitivity and precision, and identify potential matrix effects. Finally, issues related to data interpretation for biological understanding and risk assessment were discussed, but their resolution remains elusive.
Collapse
Affiliation(s)
- Abby A Li
- Exponent Inc., 1010 14th Street, San Francisco, CA, 94114, USA.
| | - Susan L Makris
- US Environmental Protection Agency Office of Research and Development, 1200 Pennsylvania Ave NW 8623R, Washington, DC, 20460, USA.
| | - M Sue Marty
- The Dow Chemical Company, Toxicology & Environmental Research and Consulting, 1803 Building, Midland, MI, 48674, USA.
| | - Volker Strauss
- BASF SE, Experimental Toxicology and Ecology, 67056, Ludwigshafen, Germany.
| | - Mary E Gilbert
- US Environmental Protection Agency, National Health Environmental Effects Research Lab, 109 T.W. Alexander Drive, MD B105 05, Research Triangle Park, NC, 27711, USA.
| | - Ann Blacker
- Bayer CropScience, P.O. Box 12014, RTP, NC, 27709, USA.
| | | | - Pragati S Coder
- Charles River Laboratories, Developmental and Reproductive Toxicology, 1407 George Road, Ashland, OH, 44805, USA.
| | - Bethany Hannas
- The Dow Chemical Company, Toxicology & Environmental Research and Consulting, 1803 Building, Midland, MI, 48674, USA.
| | - Sheri Lordi
- Charles River Laboratories International, 251 Ballardvale Street, Wilmington, MA, 01887, USA.
| | - Steffen Schneider
- BASF SE, Experimental Toxicology and Ecology, 67056, Ludwigshafen, Germany.
| |
Collapse
|
18
|
Poitout F, Colangelo JL, Lavallée S, Aulbach AD, Piché MS, Ennulat D, Ameri M, Boone LI. Current Practices and Challenges in Method Validation. Toxicol Pathol 2018; 46:847-856. [PMID: 30253718 DOI: 10.1177/0192623318801571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Method validation is a cornerstone on which biomarker development and utilization rest. However, given the abundance of biomarker candidates that are being identified and characterized, validation of these entities for the use in nonclinical studies can be complex. The objective of this continuing education course was to review current practices and challenges encountered during the validation of methods for the analysis of novel biomarkers. Additionally, the importance of biological validation and correlation with pathology end points for biomarker candidates was discussed. This article is a summary of the materials presented at the 36th Annual Symposium of the Society of Toxicologic Pathology for a continuing education course titled "Current Practices and Challenges in Method Validation." The speakers were subject-matter experts in the validation of quantitative mass spectrometry, multiplex binding assays, biological biomarkers, and immunophenotyping and anatomic and clinical pathology considerations in biomarker qualification.
Collapse
Affiliation(s)
- Florence Poitout
- 1 Preclinical Services, Charles River Laboratories, Senneville, Quebec, Canada
| | | | - Simon Lavallée
- 1 Preclinical Services, Charles River Laboratories, Senneville, Quebec, Canada
| | | | - Marie-Soleil Piché
- 1 Preclinical Services, Charles River Laboratories, Senneville, Quebec, Canada
| | | | - Mehrdad Ameri
- 4 GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | - Laura I Boone
- 5 Covance Laboratories, Inc., Greenfield, Indiana, USA
| |
Collapse
|
19
|
A novel LC-MS/MS assay to quantify dermatan sulfate in cerebrospinal fluid as a biomarker for mucopolysaccharidosis II. Bioanalysis 2018; 10:825-838. [PMID: 29863901 DOI: 10.4155/bio-2018-0025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
AIM The study aimed to develop an LC-MS/MS assay to measure dermatan sulfate (DS) in human cerebrospinal fluid (CSF). METHODS & RESULTS DS was quantified by ion pairing LC-MS/MS analysis of the major disaccharides derived from chondroitinase B digestion. Artificial CSF was utilized as a surrogate for calibration curve preparation. The assay was fully validated, with a linear range of 20.0-4000 ng/ml, accuracy within ±20%, and precision of ≤20%. CSF samples from mucopolysaccharidoses (MPS) II patients showed an average of 11-fold increase in DS levels compared with controls. CONCLUSION The described assay is capable of differentiating DS levels in the CSF of MPS II patients from controls and can be used to monitor disease progression and therapeutic responses.
Collapse
|
20
|
Definitive profiling of plasma bile acids as potential biomarkers for human liver diseases using UPLC–HRMS. Bioanalysis 2018; 10:917-932. [DOI: 10.4155/bio-2018-0018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aim: Investigation of bile acids (BAs) as biomarkers for liver and kidney diseases has gained momentum recently to fulfill the needs in drug development and clinical practice, but a thorough and rapid profiling of BAs in human plasma has been hindered by the large interindividual variability and lack of selective methods. Results: A selective and efficient UPLC-high resolution mass spectrometry method was developed and fully validated for the definitive profiling of 26 BAs in human plasma with a curve rage of 1–1000 ng/ml and a runtime of 7.2 min. Conclusion: Four BA combinations with good sensitivity and specificity show potential biomarker applications for liver injury and diseases.
Collapse
|
21
|
The FDA/critical path initiative/Duke-Margolis center for health policy public workshop on analytical validation of assays for biomarker qualification: an update on the White Paper. Bioanalysis 2018; 10:893-896. [DOI: 10.4155/bio-2018-0125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
22
|
Goettel M, Niessner R, Scherer M, Scherer G, Pluym N. Analysis of Urinary Eicosanoids by LC–MS/MS Reveals Alterations in the Metabolic Profile after Smoking Cessation. Chem Res Toxicol 2018; 31:176-182. [DOI: 10.1021/acs.chemrestox.7b00276] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Michael Goettel
- Chair for Analytical Chemistry, Technische Universität München, Marchioninistraße 17, 81377 Munich, Germany
- ABF, Analytisch-Biologisches Forschungslabor GmbH, Semmelweisstraße 5, 82152 Planegg, Germany
| | - Reinhard Niessner
- Chair for Analytical Chemistry, Technische Universität München, Marchioninistraße 17, 81377 Munich, Germany
| | - Max Scherer
- ABF, Analytisch-Biologisches Forschungslabor GmbH, Semmelweisstraße 5, 82152 Planegg, Germany
| | - Gerhard Scherer
- ABF, Analytisch-Biologisches Forschungslabor GmbH, Semmelweisstraße 5, 82152 Planegg, Germany
| | - Nikola Pluym
- ABF, Analytisch-Biologisches Forschungslabor GmbH, Semmelweisstraße 5, 82152 Planegg, Germany
| |
Collapse
|
23
|
Conti M, Matulli Cavedagna T, Ramazzotti E, Mancini R, Calza L, Rinaldi M, Badia L, Guardigni V, Viale P, Verucchi G. Multiplexed therapeutic drug monitoring (TDM) of antiviral drugs by LC-MS/MS. CLINICAL MASS SPECTROMETRY (DEL MAR, CALIF.) 2018; 7:6-17. [PMID: 39193552 PMCID: PMC11322761 DOI: 10.1016/j.clinms.2017.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 01/05/2023]
Abstract
Background Therapeutic drug monitoring (TDM) can be a useful tool in the clinical management of anti-hepatitis C virus (anti-HCV) drugs. Methods for the determination of various types of anti-HCV drugs in biological samples are, therefore, needed for clinical laboratories. Objective In this work, employing the LC-MS/MS approach, we aimed to develop a multiplexed method for identification of the following anti-HCV drugs: Ribavirin (RBV), Boceprevir (BOC), Telaprevir (TVR), Simeprevir (SIM), Daclatasvir (DAC), Sofosbuvir (SOF) and its metabolite GS 331007 (SOFM) in liquid plasma and in dried plasma spots (DPSs). Method A single-step extractive-deproteinization was employed for both liquid plasma and DPSs. Reverse-phase liquid chromatography coupled with MRM detection was developed for multiplexed drug detection and quantification. Results Sensitivities (expressed as LOQ) were 10 (±1.2), 10 (±4.9), 10 (±4.4), 10 (±4.4), 10 (±6.4), 10 (±3.4), 10 (±6.4) ng/ml for RBV, SOFM, SOF, DAC, BOC, TVR, and SIM, respectively; accuracy (expressed as BIAS%) was <10% for all drugs; reproducibility (intra- and inter-day CV%) was <10% for all drugs; dynamic range was 10-10,000 ng/ml for all drugs. Conclusions A novel, simple, rapid and robust LC-MS/MS multiplex assay for the TDM of various anti-HCV drugs that are currently in the clinic was successfully developed. Application to DPS samples enabled TDM to be used for outpatients as well.
Collapse
Affiliation(s)
- M. Conti
- LUM Metropolitan Laboratory – Azienda USL Bologna, Bologna, Italy
| | | | - E. Ramazzotti
- LUM Metropolitan Laboratory – Azienda USL Bologna, Bologna, Italy
| | - R. Mancini
- LUM Metropolitan Laboratory – Azienda USL Bologna, Bologna, Italy
| | - L. Calza
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - M. Rinaldi
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Research Center for the Study of Hepatitis, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - L. Badia
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Research Center for the Study of Hepatitis, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - V. Guardigni
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - P. Viale
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - G. Verucchi
- Infectious Diseases Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, Bologna, Italy
- Research Center for the Study of Hepatitis, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
24
|
Elander N, Aughton K, Greenhalf W. Development of Novel Therapeutic Response Biomarkers. PANCREATIC CANCER 2018:1273-1304. [DOI: 10.1007/978-1-4939-7193-0_59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
25
|
Broadhurst D, Goodacre R, Reinke SN, Kuligowski J, Wilson ID, Lewis MR, Dunn WB. Guidelines and considerations for the use of system suitability and quality control samples in mass spectrometry assays applied in untargeted clinical metabolomic studies. Metabolomics 2018; 14:72. [PMID: 29805336 PMCID: PMC5960010 DOI: 10.1007/s11306-018-1367-3] [Citation(s) in RCA: 530] [Impact Index Per Article: 75.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 05/03/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Quality assurance (QA) and quality control (QC) are two quality management processes that are integral to the success of metabolomics including their application for the acquisition of high quality data in any high-throughput analytical chemistry laboratory. QA defines all the planned and systematic activities implemented before samples are collected, to provide confidence that a subsequent analytical process will fulfil predetermined requirements for quality. QC can be defined as the operational techniques and activities used to measure and report these quality requirements after data acquisition. AIM OF REVIEW This tutorial review will guide the reader through the use of system suitability and QC samples, why these samples should be applied and how the quality of data can be reported. KEY SCIENTIFIC CONCEPTS OF REVIEW System suitability samples are applied to assess the operation and lack of contamination of the analytical platform prior to sample analysis. Isotopically-labelled internal standards are applied to assess system stability for each sample analysed. Pooled QC samples are applied to condition the analytical platform, perform intra-study reproducibility measurements (QC) and to correct mathematically for systematic errors. Standard reference materials and long-term reference QC samples are applied for inter-study and inter-laboratory assessment of data.
Collapse
Affiliation(s)
- David Broadhurst
- School of Science, Centre for Integrative Metabolomics and Computational Biology, Edith Cowan University, Joondalup, Perth, Australia
| | - Royston Goodacre
- School of Chemistry, Manchester Institute of Biotechnology, University of Manchester, Manchester, M1 7DN, UK
| | - Stacey N Reinke
- School of Science, Centre for Integrative Metabolomics and Computational Biology, Edith Cowan University, Joondalup, Perth, Australia
- Separation Sciences and Metabolomics Laboratory, Murdoch University, Perth, WA, Australia
| | - Julia Kuligowski
- Neonatal Research Unit, Health Research Institute La Fe, Avda. Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Ian D Wilson
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, SW7 2AZ, UK
| | - Matthew R Lewis
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, SW7 2AZ, UK
| | - Warwick B Dunn
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
26
|
2017 White Paper on recent issues in bioanalysis: a global perspective on immunogenicity guidelines & biomarker assay performance (Part 3 – LBA: immunogenicity, biomarkers and PK assays). Bioanalysis 2017; 9:1967-1996. [DOI: 10.4155/bio-2017-4974] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The 2017 11th Workshop on Recent Issues in Bioanalysis took place in Los Angeles/Universal City, California, on 3–7 April 2017 with participation of close to 750 professionals from pharmaceutical/biopharmaceutical companies, biotechnology companies, contract research organizations and regulatory agencies worldwide. WRIB was once again a 5-day, week-long event – a full immersion week of bioanalysis, biomarkers and immunogenicity. As usual, it was specifically designed to facilitate sharing, reviewing, discussing and agreeing on approaches to address the most current issues of interest including both small- and large-molecule analysis involving LC–MS, hybrid ligand-binding assay (LBA)/LC–MS and LBA approaches. This 2017 White Paper encompasses recommendations emerging from the extensive discussions held during the workshop, and is aimed to provide the bioanalytical community with key information and practical solutions on topics and issues addressed, in an effort to enable advances in scientific excellence, improved quality and better regulatory compliance. Due to its length, the 2017 edition of this comprehensive White Paper has been divided into three parts for editorial reasons. This publication (Part 3) covers the recommendations for large-molecule bioanalysis, biomarkers and immunogenicity using LBA. Part 1 (LC–MS for small molecules, peptides and small molecule biomarkers) and Part 2 (hybrid LBA/LC–MS for biotherapeutics and regulatory agencies’ inputs) are published in volume 9 of Bioanalysis, issues 22 and 23 (2017), respectively.
Collapse
|
27
|
Current practices and future outlook on the integration of biomarkers in the drug development process. Bioanalysis 2017; 9:1827-1837. [PMID: 29120222 DOI: 10.4155/bio-2017-0155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Over the last decade, there has been broad incorporation of translational biomarkers into the early drug development process to predict safety concerns, measure target engagement and monitor disease progression. One goal of translational biomarkers is to create a cycle whereby preclinical readouts influence candidate selection and subsequent clinical data are fed back into research to facilitate better decision making. Successes have been limited and not as broad in scope as desired. Collaborations between industry and regulators have increased the number of qualified biomarkers; but the process is lengthy and expensive. A high level overview of translational biomarkers as well as a discussion of some of the successes and failures encountered in development is discussed here.
Collapse
|
28
|
Parallelism experiments to evaluate matrix effects, selectivity and sensitivity in ligand-binding assay method development: pros and cons. Bioanalysis 2017; 9:1107-1122. [DOI: 10.4155/bio-2017-0084] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Parallelism is an essential experiment characterizing relative accuracy for a ligand-binding assay (LBA). By assessing the effects of dilution on the quantitation of endogenous analyte(s) in matrix, selectivity, matrix effects, minimum required dilution, endogenous levels of healthy and diseased populations and the LLOQ are assessed in a single experiment. This review compares and discusses all available approaches that can be used to assess key assay parameters for pharmacokinetic and biomarker LBAs, as well as the advantages and disadvantages of each approach. This review also summarizes a systematic approach that can apply to guide endogenous LBA method development and optimization with a suggested way to interpret parallelism data.
Collapse
|
29
|
Ensuring quality and compliance in outsourcing of bioanalysis of clinical biomarkers. Bioanalysis 2017; 9:501-504. [PMID: 28300423 DOI: 10.4155/bio-2017-0008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
30
|
Welcome to volume 9 of Bioanalysis. Bioanalysis 2016; 9:151-154. [PMID: 27960542 DOI: 10.4155/bio-2016-4983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Welcome to volume 9 of Bioanalysis and Happy New Year to all of our readers. We would like to take the opportunity to look back 2016, which was another great year for us. We thank all our authors, readers and reviewers, as well as our Editorial Board members for their continued support. We very much look forward to working with everyone in 2017.
Collapse
|
31
|
Accuracy: a potential quandary in regulated bioanalysis of ‘endogenous’ analytes. Bioanalysis 2016; 8:2393-2397. [DOI: 10.4155/bio-2016-0247] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
32
|
Abstract
Tower Hall Funabori, Tokyo, Japan, 9-10 March 2016 At the 7th Japan Bioanalysis Forum symposium, bioanalytical methods and processes were discussed with authorities after the issuing of bioanalytical method validation guidelines (EMA and Japanese Ministry of Health, Labour and Welfare) and draft guidance (US FDA). Method establishment and bioanalysis of biomarkers were discussed with a focus on scientific validation. Bioanalytical methods for antibody-drug conjugates, the potential of MS imaging and microsampling activity in drug development were introduced. Discussion groups presented and openly discussed their results with about 300 attendees. This manuscript provides an overview of the highlights of the symposium.
Collapse
|
33
|
Arnold ME, Booth B, King L, Ray C. Workshop Report: Crystal City VI—Bioanalytical Method Validation for Biomarkers. AAPS JOURNAL 2016; 18:1366-1372. [DOI: 10.1208/s12248-016-9946-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/05/2016] [Indexed: 11/30/2022]
|
34
|
Adaptation of hybrid immunoaffinity LC–MS methods for protein bioanalysis in a Contract Research Organization. Bioanalysis 2016; 8:1545-1549. [DOI: 10.4155/bio-2016-0104] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
35
|
Implementing fit-for-purpose biomarker assay approaches: a bioanalytical perspective. Bioanalysis 2016; 8:1221-3. [DOI: 10.4155/bio-2016-0070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|