1
|
Szarka S, Thorsteinsdottir M, Wheller R. Design of experiments for the optimization of sample preparation for bottom-up targeted protein LC-MS/MS workflows. Bioanalysis 2025:1-10. [PMID: 40492479 DOI: 10.1080/17576180.2025.2515004] [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: 02/11/2025] [Accepted: 05/23/2025] [Indexed: 06/12/2025] Open
Abstract
AIMS Design of experiments (DOE) is a versatile and efficient approach to tackle complex scientific problems. We aimed to assess its feasibility in the optimization of the multistep, involved bottom-up sample preparation for the UPLC-MS/MS analysis of proteins. MATERIALS AND METHODS The model analyte was a human IgG1 monoclonal antibody which was spiked into rat plasma and processed further by reduction, alkylation, and digestion for the subsequent UPLC-MS/MS analysis. The Modde Go software was used for the generation of experimental designs and for processing, analyzing and the interpretation of the data. RESULTS DOE screening revealed that urea made the biggest improvement on the surrogate peptide responses, while guanidine significantly suppressed them. DOE optimization resulted in a 2-, 10-, 10- and 50-fold response increase for the respective DTLM, FNWY, TPEV and VVSV surrogate peptide even after a short, <3-h sample preparation, when compared to a legacy method that required 2-day preparation. CONCLUSIONS The DOE approach was applied successfully for the comprehensive optimization of eight denaturation, reduction and digestion parameters. DOE was found to be an efficient tool for protein sample preparation optimization, and the predictive power of the DOE models was successfully demonstrated.
Collapse
|
2
|
Gatmaitan AN, Weaver K, Badal SP, Bird HE, Huhn A, Hussey C, Clarke WA, Eberlin LS. Development and Application of the Modular MasSpec Pen System for Clinical Opioid Screening. Anal Chem 2025. [PMID: 40395096 DOI: 10.1021/acs.analchem.5c00558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Mass spectrometry (MS) is an important tool for opioid screening due to the high analytical sensitivity and chemical specificity provided by MS for small molecule analysis. MS techniques that can provide direct detection of opioids are of high interest to simplify and expedite patient screening workflows. Here, we describe the development and application of the MasSpec Pen (MSPen) technology as a modular system for opioid screening in which sampling and MS analysis are decoupled. With the modular MSPen system, the rapid, direct, and non-destructive nature of sample collection provided by the MSPen device are retained, followed by analysis using a MS method suitable for the intended use. We used the modular MSPen system and electrospray ionization MS to detect oxycodone, hydrocodone, fentanyl, hydromorphone, and morphine both individually and as mixtures. Detection and quantification limits in the single nanogram range were achieved when analyzing standards deposited on surfaces as well as ex vivo human skin doped with opioids. We then tested the feasibility of the modular MSPen system at the Johns Hopkins Bayview Medical Center to analyze opioids from direct sampling of patient skin, compared to results obtained from the patients' urine and saliva. Altogether, the modular MSPen system allowed direct sampling and semi-quantitation of opioids from surfaces and patients' skin, with the opportunity to be further developed for other screening applications.
Collapse
Affiliation(s)
- Abigail N Gatmaitan
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712, United States
| | - Kathryn Weaver
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21251, United States
| | - Sunil P Badal
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712, United States
| | - H Elizabeth Bird
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21251, United States
| | - Andrew Huhn
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21251, United States
| | - Casey Hussey
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21251, United States
| | - William A Clarke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21251, United States
| | - Livia S Eberlin
- Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, United States
| |
Collapse
|
3
|
Nierves LA, Lin TT, Moradian A, Shen Q, Sechi S, MacCoss MJ, Qu J, van Eyk JE, Hoofnagle AN, Qian WJ. Biomarkers, Proteoforms, and Mass Spectrometry-Based Assays for Diabetes Clinical Research. J Clin Endocrinol Metab 2025; 110:1514-1523. [PMID: 40056450 DOI: 10.1210/clinem/dgaf159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/19/2025] [Accepted: 03/06/2025] [Indexed: 03/10/2025]
Abstract
The prevalence of diabetes, particularly type 2 diabetes, has reached epidemic proportions globally. The number of patients with type 1 diabetes (T1D) is also increasing rapidly. Despite advancements in understanding the pathogenesis of diabetes, the lack of circulating pancreatic biomarkers and reliable clinical-grade assays remains a major gap in diabetes research, often hindering the ability to adequately assess disease progression and therapeutic responses. This mini-review discusses emerging pancreatic biomarkers, with an emphasis on T1D, the limitations of current immunoassays, and the expanding role of mass spectrometry-based assays. Highlights include the recent work within the NIDDK-funded "Targeted Mass Spectrometry Assays for Diabetes and Obesity Research (TaMADOR)" consortium, which aims to develop robust, quantitative, and transferable assays for translational research. The review also emphasizes the importance of proteoform-specific assays for monitoring pancreatic function, including prohormone processing during disease progression or in responses to therapy.
Collapse
Affiliation(s)
- Lorenz A Nierves
- Translational Omics, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Tai-Tu Lin
- Translational Omics, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Annie Moradian
- Precision Biomarker Laboratories, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Qingqing Shen
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214, USA
| | - Salvatore Sechi
- Division of Diabetes, Endocrinology, and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael J MacCoss
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Jun Qu
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214, USA
| | - Jennifer E van Eyk
- Precision Biomarker Laboratories, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Andrew N Hoofnagle
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Wei-Jun Qian
- Translational Omics, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| |
Collapse
|
4
|
Kim SM, Oh H, Hong SN, Kim MJ, Choe YH, Lee SY. A Bottom-Up Liquid Chromatography-Tandem Mass Spectrometry Method for Therapeutic Drug Monitoring of Infliximab: Method Development, Comparison With 2 Enzyme-Linked Immunosorbent Assay Methods, and Evaluation of Anti-Drug Antibody Interference. Arch Pathol Lab Med 2025; 149:448-456. [PMID: 39041105 DOI: 10.5858/arpa.2023-0573-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2024] [Indexed: 07/24/2024]
Abstract
CONTEXT.— Therapeutic drug monitoring is recommended to optimize infliximab use and improve outcome in chronic inflammatory disorders. OBJECTIVE.— To describe a simple and affordable liquid chromatography-tandem mass spectrometry (LC-MS/MS) method to measure infliximab in serum. DESIGN.— Infliximab was measured using winged stable isotope-labeled peptides as internal standards. Linearity, lower limit of measuring interval, limit of detection, precision, accuracy, carryover, and ion suppression were evaluated. Method comparison against 2 enzyme-linked immunosorbent assay (ELISA) methods (Remsima Monitor and IDKmonitor Infliximab) and anti-drug antibody (ADA) interference were evaluated using clinical specimens from inflammatory bowel disease patients (N = 237). RESULTS.— Analytical run time and sample preparation time were 5 minutes per sample and 3 hours per batch, respectively. Analytical measurement interval and limit of detection were 0.50 to 50.0 μg/mL (R2 = 0.998) and 0.25 μg/mL, respectively. The intraday and interday imprecision percentage coefficients of variation were less than 6.1%. Accuracy was 94.2% to 98.7%. No significant ion suppression or carryover was observed. Infliximab concentrations measured by LC-MS/MS showed good agreement with those measured by Remsima Monitor (mean percentage difference, 5.7%; 95% CI, -1.2% to 12.6%) but were markedly lower than those measured by IDKmonitor (-32.6%; -35.8% to -29.4%), demonstrating significant bias between ELISAs. Although a good agreement between LC-MS/MS and ELISA was observed for ADA-negative samples (-3.5%; -12.8% to 5.9%), a significant bias was observed for ADA-positive samples (13.6%; 1.7% to 25.6%). CONCLUSIONS.— This simple, fast, and affordable LC-MS/MS method for infliximab quantitation could improve standardization of infliximab quantitation and optimization of infliximab use in patients with high-titer ADA.
Collapse
Affiliation(s)
- Sang-Mi Kim
- From the Departments of Laboratory Medicine and Genetics (S.-M. Kim, Oh, Lee), Internal Medicine (Hong), and Pediatrics (M. J. Kim, Choe), Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- the Department of Laboratory Medicine, Chosun University Hospital, Chosun University School of Medicine, Gwangju, Republic of Korea (S.-M. Kim)
| | - Hyeonju Oh
- From the Departments of Laboratory Medicine and Genetics (S.-M. Kim, Oh, Lee), Internal Medicine (Hong), and Pediatrics (M. J. Kim, Choe), Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung Noh Hong
- From the Departments of Laboratory Medicine and Genetics (S.-M. Kim, Oh, Lee), Internal Medicine (Hong), and Pediatrics (M. J. Kim, Choe), Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Mi Jin Kim
- From the Departments of Laboratory Medicine and Genetics (S.-M. Kim, Oh, Lee), Internal Medicine (Hong), and Pediatrics (M. J. Kim, Choe), Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yon Ho Choe
- From the Departments of Laboratory Medicine and Genetics (S.-M. Kim, Oh, Lee), Internal Medicine (Hong), and Pediatrics (M. J. Kim, Choe), Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Soo-Youn Lee
- From the Departments of Laboratory Medicine and Genetics (S.-M. Kim, Oh, Lee), Internal Medicine (Hong), and Pediatrics (M. J. Kim, Choe), Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
5
|
Liu M, Jiao F, He Y. Rapid classification of proteins suspended in an evaporated sessile drop. Talanta 2025; 294:128224. [PMID: 40311480 DOI: 10.1016/j.talanta.2025.128224] [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: 01/15/2025] [Revised: 04/20/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025]
Abstract
Reliable and accessible technologies for biological sample analysis are crucial for rapid, accurate illness diagnosis in resource-limited settings. Conventional methods like mass spectrometry and enzyme-linked immunosorbent assay (ELISA) often face challenges of technological complexity and equipment immobility, leading to delayed diagnoses and elevated costs. This study introduces evaporative deposition patterns from sessile droplets as a simple, cost-effective approach for classifying proteins in protein-salt mixtures, leveraging quantitative texture analysis to achieve high diagnostic accuracy. Using mixtures of potassium chloride (KCl) or sodium chloride (NaCl) with Bovine Serum Albumin (BSA) or Ovalbumin (OVA) as model systems, we varied salt (0.25-2 wt%) and protein (0.5-1.5 wt%) concentrations, observing distinct deposition morphologies such as coffee rings, cruciform/dendritic crystals at high salt levels, and volcano-shaped /spoke-like patterns at low or high protein levels. A novel quantitative framework integrating radial density distributions, gray-level co-occurrence matrix (GLCM) texture features, and receiver operating characteristic (ROC) curves achieved a 96.74 % classification accuracy for NaCl-protein mixtures, surpassing traditional texture analysis in simplicity and efficacy. These findings demonstrate the potential of evaporative deposition analysis as a simple tool for distinguishing molecular mixtures, laying the groundwork for future diagnostic applications. Unlike prior qualitative studies, this work provides a robust, quantitative method for protein classification, advancing the understanding of protein-salt interactions.
Collapse
Affiliation(s)
- Maoxi Liu
- School of Chemical Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Feng Jiao
- School of Chemical Engineering, Kunming University of Science and Technology, Kunming, 650500, China.
| | - Yongqing He
- Chongqing Key Laboratory of Micro-Nano System and Intelligent Transduction, National Research Base of Intelligent Manufacturing Service, Chongqing Technology and Business University, Chongqing, 400067, China.
| |
Collapse
|
6
|
Paris J, Tavernier A, Bethegnies S, Descloux S, Fedeli O. Quantification of total sBCMA in human plasma by peptide-level immunocapture LC-MS/MS. J Mass Spectrom Adv Clin Lab 2025; 36:46-51. [PMID: 40331169 PMCID: PMC12051699 DOI: 10.1016/j.jmsacl.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 03/24/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Background B-cell maturation antigen (BCMA) is a membrane protein that is overexpressed in multiple myeloma cells and can be targeted with biotherapeutic agents. BCMA is naturally shed by γ-secretase, leading to the formation of soluble BCMA (sBCMA), which circulates in the blood. sBCMA can affect the efficacy of BCMA-targeted therapies and act as a drug sink. Additionally, sBCMA can interfere with pharmacokinetic measurements when BCMA is directly targeted. Therefore, quantification of this biomarker during clinical trials is essential to assess the effective dose and understand pharmacokinetic results. When quantifying sBCMA using ligand binding assays or hybrid assays, the biotherapeutic can interfere with the capture of sBCMA, leading to an underestimation of its levels. Methods Samples were denatured, reduced, and alkylated prior to trypsin digestion. sBCMA peptide enrichment was performed using anti-peptide polyclonal antibodies. Reversed-phase chromatographic separation was conducted on a biocompatible C18 column with an analysis time of sixteen minutes per sample. The SCIEX QTRAP 5500 mass spectrometer operated in multiple reaction monitoring mode. The calibration curve was prepared by spiking recombinant sBCMA into monkey plasma. Results The parallelism between the authentic and surrogate matrices, as well as between the endogenous and recombinant proteins, was validated. Comparisons were made between protein and peptide level hybrid assays, with the peptide level approach effectively removing the interference of the biotherapeutic. Additionally, the peptide level immunocapture LC-MS/MS demonstrated ligand tolerance. Conclusion The peptide level immunocapture LC-MS/MS analysis eliminated the interference of anti-BCMA biotherapeutics, allowing for the quantification of total sBCMA in clinical samples while achieving a LLOQ of 10 ng/mL.
Collapse
Affiliation(s)
| | | | - Sylvie Bethegnies
- Sanofi, TMU-Labsciences, 371 rue du Professeur Blayac, 34184 Montpellier, France
| | - Sandrine Descloux
- Sanofi, TMU-Labsciences, 371 rue du Professeur Blayac, 34184 Montpellier, France
| | - Olivier Fedeli
- Sanofi, TMU-Labsciences, 371 rue du Professeur Blayac, 34184 Montpellier, France
| |
Collapse
|
7
|
Becker JO, Shijo SK, Huynh HH, Forrest KL, MacCoss MJ, Emrick MA, Goonatilleke E, Hoofnagle AN. Quantification of glucagon and oxyntomodulin by protein precipitation-immunoaffinity enrichment-LC-MS/MS. J Mass Spectrom Adv Clin Lab 2025; 36:37-45. [PMID: 40385697 PMCID: PMC12083559 DOI: 10.1016/j.jmsacl.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 05/20/2025] Open
Abstract
Introduction Glucagon and oxyntomodulin are peptide hormones differentially released from proglucagon that function in regulating blood glucose. Their overlapping amino acid sequences make the development of specific immunoassays difficult, but the specificity of liquid chromatography-tandem mass spectrometry can be used to distinguish the peptides. We aimed to develop a sensitive and specific mass spectrometric assay that uses non-proprietary reagents and normal-flow liquid chromatography in the simultaneous quantification of both analytes. Methods Bulk plasma proteins were precipitated in ethanol/ammonium hydroxide. Analytes were enriched using monoclonal antibodies generated in-house and analyzed using liquid chromatography-tandem mass spectrometry. A glucagon calibration material was sourced commercially and characterized for purity and concentration by high-performance liquid chromatography-ultraviolet detection and amino acid analysis. Single-point calibration was used to minimize between-day variability. Results The novel antibodies performed acceptably (peptide recovery 45-59 %). The assay was precise (<13 %CV) and linear over the range of 1.3-14.7 pM and 1.1-13.7 pM for glucagon and oxyntomodulin, respectively. The glucagon calibration material concentration was determined to be 1.596 mg/g. Tube-type studies supported the use of protease inhibitor tubes at the time of blood draw. Patients with type 1 diabetes had lower concentrations of glucagon when maintained on an insulin pump, but not with injectable insulin. Conclusion We have validated a method with a highly detailed standard operating procedure. We have characterized calibration materials to help maintain accuracy and achieve between-day and between-laboratory harmonization. The assay will be beneficial in better understanding α-cell health and glycemic control in diabetes and other diseases.
Collapse
Affiliation(s)
| | - Sara K. Shijo
- Department of Laboratory Medicine and Pathology, USA
| | | | | | | | | | | | - Andrew N. Hoofnagle
- Department of Laboratory Medicine and Pathology, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Box 357110, Seattle, WA 98195-7110, USA
| |
Collapse
|
8
|
Pu J, Yang X, Lin TT, Fillmore TL, Gritsenko MA, Kelly SS, Swensen AC, Shi T, Master SR, DeLany JP, Goodpaster BH, Qian WJ, Qu J. A multiplex assay of leptin, resistin, and adiponectin by immunoaffinity enrichment and targeted mass spectrometry. J Mass Spectrom Adv Clin Lab 2025; 36:11-18. [PMID: 40093568 PMCID: PMC11910354 DOI: 10.1016/j.jmsacl.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 01/18/2025] [Accepted: 01/31/2025] [Indexed: 03/19/2025] Open
Abstract
Background Leptin, resistin, and adiponectin are critical adipokines involved in the pathophysiology of obesity and its related disorders, including type 2 diabetes. Although these biomarkers have historically been quantified using immunoassays, the specificity of antibody-based methods has frequently been questioned. As a result, there is an increasing interest in developing reliable, multiplexed clinical assays that utilize mass spectrometry for improved accuracy. In this study, we present a multiplexed immunoaffinity liquid chromatography-tandem mass spectrometry (multi-IA-LC-MS/MS) assay designed for the sensitive and selective measurement of leptin, resistin, and adiponectin in human plasma. Methods Leptin, resistin, and adiponectin were selectively enriched from plasma samples using an antibody cocktail composed of monoclonal antibodies targeting each respective adipokine. The enriched adipokines underwent enzymatic digestion, and the resulting tryptic peptides were quantified using LC-MS/MS. The validated assay was subsequently applied to plasma samples collected from a cohort of subjects representing various weight categories, including normal weight, overweight, and obesity. Results The lower limits of quantification for the assay were determined to be 0.5 ng/mL for both leptin and resistin, and 50 ng/mL for adiponectin. Intra- day, inter- day, and total imprecision measurements were all < 15 %, while spike recovery consistently exceeded 83 %. Comparative analysis with individual immunoassays demonstrated strong correlation, with all correlation coefficients (r) being equal to or greater than 0.869. Notably, when comparing subjects with obesity to those with normal weight, there was an approximately nine-fold increase in circulating leptin levels and a ∼1.6-fold decrease in circulating adiponectin levels. Conclusions A multi-IA-LC-MS/MS assay was developed for the simultaneous and sensitive measurement of leptin, resistin, and adiponectin in clinical samples. This quantitative method shows significant potential for applications related to obesity and could facilitate improved clinical management and understanding of obesity-related conditions.
Collapse
Affiliation(s)
- Jie Pu
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Xinxin Yang
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Tai-Tu Lin
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Thomas L Fillmore
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Marina A Gritsenko
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Shane S Kelly
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Adam C Swensen
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Tujin Shi
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Stephen R Master
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - James P DeLany
- AdventHealth, Translational Research Institute, Orlando, FL, United States
| | - Bret H Goodpaster
- AdventHealth, Translational Research Institute, Orlando, FL, United States
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Jun Qu
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, United States
- New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, United States
| |
Collapse
|
9
|
van der Burgt YEM, Romijn FPHTM, Treep MM, Ruhaak LR, Cobbaert CM. Strategies to verify equimolar peptide release in mass spectrometry-based protein quantification exemplified for apolipoprotein(a). Clin Chem Lab Med 2025; 63:780-789. [PMID: 39450666 DOI: 10.1515/cclm-2024-0539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
OBJECTIVES Quantitative protein mass spectrometry (MS) is ideally suited for precision diagnostics and for reference standardization of protein analytes. At the Leiden Apolipoprotein Reference Laboratory we apply MS strategies to obtain detailed insight into the protein-to-peptide conversion in order to verify that quantifier peptides are not partly concealed in miscleaved protein backbone. METHODS Apolipoprotein(a) (apo(a)) was digested in a non-optimal manner to enhance the number of miscleaved peptides that were identified by high resolution liquid chromatography tandem-MS measurements. The protein-to-peptide conversion was carefully mapped with specific attention for miscleaved peptides that contain an apo(a) quantifier peptide. Four different isotopologues of each apo(a)-quantifier peptide were applied to evaluate linearity of internal peptide standards during measurement of specific real-life samples. RESULTS Two apo(a) quantifier peptides that were concealed in two different miscleaved peptides were included into a multiple reaction monitoring list in our targeted MS-based apo(a) quantifications to alert for potential protein digestion discrepancies. The presence of miscleaved peptides could be ruled out when applying our candidate reference measurement procedure (RMP) for apo(a) quantification. CONCLUSIONS These data further corroborate the validity of our apo(a) candidate RMP as higher order method for certification of commercial Lp(a) tests that is endorsed by the International Federation of Clinical Chemistry and Laboratory Medicine. MS-based molecular detection and quantification of heterogeneous apo(a) proteoforms will allow manufacturers' transitioning from confounded lipoprotein(a) [Lp(a)] mass levels into accurate molar apo(a) levels.
Collapse
Affiliation(s)
- Yuri E M van der Burgt
- Department of Clinical Chemistry and Laboratory Medicine, 4501 Leiden University Medical Center , Leiden, The Netherlands
| | - Fred P H T M Romijn
- Department of Clinical Chemistry and Laboratory Medicine, 4501 Leiden University Medical Center , Leiden, The Netherlands
| | - Maxim M Treep
- Department of Clinical Chemistry and Laboratory Medicine, 4501 Leiden University Medical Center , Leiden, The Netherlands
| | - L Renee Ruhaak
- Department of Clinical Chemistry and Laboratory Medicine, 4501 Leiden University Medical Center , Leiden, The Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, 4501 Leiden University Medical Center , Leiden, The Netherlands
| |
Collapse
|
10
|
Kruijt M, Cobbaert CM, Ruhaak LR. Antithrombin: Deficiency, Diversity, and the Future of Diagnostics. MASS SPECTROMETRY REVIEWS 2025. [PMID: 40088190 DOI: 10.1002/mas.21929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 02/12/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
Our healthcare system provides reactive sick-care, treating patients after symptoms have appeared by prescription of generic and often suboptimal therapy. This strategy brings along high costs and high pressure which is not sustainable. Alternatively, P5 healthcare is proposed focusing on five key elements: prevention, personalization, prediction, participation, psychocognition, however, changes in current clinical care pathways are required, for which antithrombin deficiency is a prime example. Hereditary antithrombin deficiency (ATD) is a genetic disorder, for which screening is instigated after a thrombotic episode. Current diagnostic tests for ATD lack sensitivity and refinement to correctly classify patients, and generic treatments are prescribed. A molecular understanding of ATD through a molecular diagnostic test that analyzes all clinically relevant features of antithrombin is required. Here, clinically relevant molecular characteristics of antithrombin, the diversity of antithrombin (deficiency) in heath and disease, and the strengths and weaknesses of antithrombin tests are reviewed. A mass spectrometry test that molecularly characterizes a patients antithrombin proteoforms harbors the highest potential to improve the clinical pathway for ATD. Application of this MS-based test in a future enhanced clinical pathway will improve patient management and outcome through molecular characterization of antithrombin and enables the promise of P5 healthcare for ATD.
Collapse
Affiliation(s)
- Mirjam Kruijt
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - L Renee Ruhaak
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
11
|
Yadav AS, Rubinow KB, Zelter A, Authement AK, Kestenbaum BR, Amory JK, Isoherranen N. Development and Validation of a Novel LC-MS/MS Based Proteomics Method for Quantitation of Retinol Binding Protein 4 (RBP4) and Transthyretin (TTR). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640583. [PMID: 40093066 PMCID: PMC11908197 DOI: 10.1101/2025.02.27.640583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Background Retinol binding protein 4 (RBP4) is the plasma carrier of retinol that complexes with transthyretin (TTR). RBP4 is a potential biomarker of cardiometabolic disease. However, RBP4 quantitation has relied on immunoassays and western blots without concurrent measurement of retinol and TTR. A liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for simultaneous absolute quantitation of serum and plasma RBP4 and TTR is needed to advance the understanding of RBP4 and TTR as biomarkers. Methods Surrogate peptides with reproducible, linear LC-MS/MS response were selected for RBP4 and TTR quantitation. Purified proteins were used as quantitation standards and heavy labelled peptides as internal standards. Matrix effects were evaluated for quantitation. The method was validated using pooled human serum and applied to measure inter- and intra-individual variability in RBP4 and TTR concentrations in healthy individuals and in patients with diabetic kidney disease. Results The quantitation was linear for the clinically relevant concentration ranges of RBP4 (0.5 - 6 μM) and TTR (5.8 - 69 μM). The assay inter-day variability was <12% and precision within 5%. The inter-individual variability for RBP4 and TTR concentrations was 18 - 26%, while intra-individual variability was similar to assay variability. RBP4 and TTR quantitation correlated with commercially available ELISA assays. Conclusions The developed LC-MS/MS method allows simultaneous absolute quantitation of RBP4 and TTR in human serum and plasma with reduced samples volumes. The method can be applied to clinical biomarker studies, for analysis of nutritional vitamin A status, and measurements of stoichiometry of RBP4, TTR and retinol in circulation.
Collapse
Affiliation(s)
- Aprajita S Yadav
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Katya B Rubinow
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Alex Zelter
- Department of Genome Sciences, School of Medicine, University of Washington, Seattle, Washington, USA
| | - Aurora K Authement
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Bryan R Kestenbaum
- Division of Nephrology, Kidney Research Institute, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - John K Amory
- Division of Internal Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington, USA
| |
Collapse
|
12
|
Nielsen MI, Wolters JC, Bringas OGR, Jiang H, Di Stefano LH, Oghbaie M, Hozeifi S, Nitert MJ, van Pijkeren A, Smit M, Ter Morsche L, Mourtzinos A, Deshpande V, Taylor MS, Chait BT, LaCava J. Targeted detection of endogenous LINE-1 proteins and ORF2p interactions. Mob DNA 2025; 16:3. [PMID: 39915890 PMCID: PMC11800616 DOI: 10.1186/s13100-024-00339-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 12/24/2024] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Both the expression and activities of LINE-1 (L1) retrotransposons are known to occur in numerous cell-types and are implicated in pathobiological contexts such as aging-related inflammation, autoimmunity, and in cancers. L1s encode two proteins that are translated from bicistronic transcripts. The translation product of ORF1 (ORF1p) has been robustly detected by immunoassays and shotgun mass spectrometry (MS). Yet, more sensitive detection methods would enhance the use of ORF1p as a clinical biomarker. In contrast, until now, no direct evidence of endogenous L1 ORF2 translation to protein (ORF2p) has been shown. Instead, assays for ORF2p have been limited to ectopic L1 ORF over-expression contexts and to indirect detection of endogenous ORF2p enzymatic activity, such as by the sequencing of de novo genomic insertions. Immunoassays for endogenous ORF2p have been problematic, producing apparent false positives due to cross-reactivities, and shotgun MS has not yielded reliable evidence of ORF2p peptides in biological samples. RESULTS Here we present targeted mass spectrometry assays, selected and parallel reaction monitoring (SRM and PRM, respectively) to detect and quantify L1 ORF1p and ORF2p at their endogenous abundances. We were able to quantify ORF1p and ORF2p present in our samples down to a range in the low attomoles. Confident in our ability to affinity enrich ORF2p, we describe an interactome associated with endogenous ORF2-containing macromolecular assemblies. CONCLUSIONS This is the first assay to demonstrate sensitive and robust quantitation of endogenous ORF2p. The ability to assay ORF2p directly and quantitatively will improve our understanding of the developmental and diseased cell states where L1 expression and its activity naturally occur. The ability to simultaneously assay endogenous L1 ORF1p and ORF2p is an important step forward for L1 analytical biochemistry. Endogenous ORF2p interactomes can now be presented with confidence that ORF2p is among the enriched proteins.
Collapse
Affiliation(s)
- Mathias I Nielsen
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY, USA
| | - Justina C Wolters
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| | - Omar G Rosas Bringas
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Hua Jiang
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY, USA
| | - Luciano H Di Stefano
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Mehrnoosh Oghbaie
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY, USA
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Samira Hozeifi
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Mats J Nitert
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Alienke van Pijkeren
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marieke Smit
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Lars Ter Morsche
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY, USA
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Apostolos Mourtzinos
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY, USA
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Vikram Deshpande
- Department of Pathology, Mass General Brigham and Harvard Medical School, Boston, MA, USA
| | - Martin S Taylor
- Department of Pathology, Mass General Brigham and Harvard Medical School, Boston, MA, USA
| | - Brian T Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY, USA
| | - John LaCava
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY, USA.
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| |
Collapse
|
13
|
Duarte CA, Cabrales A, Echevarría R, Paneque T, Ramírez AC, Casillas D, Sobrino-Iglesias X, Garay H, Besada V, Fernández-Ortega C. Stability in human serum and plasma of the HIV peptide drug candidate CIGB-210 and improved variants. Biotechnol Appl Biochem 2025; 72:187-195. [PMID: 39219226 DOI: 10.1002/bab.2655] [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: 08/04/2023] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
The peptide CIGB-210 inhibits HIV replication, inducing a rearrangement of vimentin intermediate filaments. The assessment of the in vitro serum and plasma stability of this peptide is important to develop an optimal pharmacological formulation. A half-life of 17.68 ± 0.59 min was calculated for CIGB-210 in human serum by reverse-phase high-performance liquid chromatography (HPLC) and mass spectrometry (MS). Eight metabolites of CIGB-210 were identified with this methodology, all of them lacking the N-terminal moiety. A previously developed CIGB-210 in-house competitive ELISA was used to compare the stability of CIGB-210 derivatives containing either D-amino acids, acetylation at the N-terminus, or both modifications. The half-life of CIGB-210 in serum was five times higher when measured by ELISA than by HPLC/MS, and twice higher in plasma as compared to serum. The substitution of D-asparagine on position 6 doubled the half-life, while D-amino acids on positions 8 and 9 did not improve the stability. The acetylation of the N-terminus resulted in a 24-fold more stable peptide in plasma. The positive effect of N-terminal acetylation on CIGB-210 serum stability was confirmed by the HPLC/MS method, as the half-life of the peptide was not reached after 2 h of incubation, which represents more than a 6.8-fold increase in the half-life with respect to the original peptide.
Collapse
Affiliation(s)
- Carlos A Duarte
- Departamento de Farmacéuticos, Centro de Ingeniería Genética y Biotecnología, La Habana, Cuba
| | - Ania Cabrales
- Departamento de Química-Física, Centro de Ingeniería Genética y Biotecnología, La Habana, Cuba
| | - Reina Echevarría
- Departamento de Farmacéuticos, Centro de Ingeniería Genética y Biotecnología, La Habana, Cuba
| | - Taimí Paneque
- Departamento de Farmacéuticos, Centro de Ingeniería Genética y Biotecnología, La Habana, Cuba
| | - Anna C Ramírez
- Departamento de Farmacéuticos, Centro de Ingeniería Genética y Biotecnología, La Habana, Cuba
| | - Dionne Casillas
- Departamento de Farmacéuticos, Centro de Ingeniería Genética y Biotecnología, La Habana, Cuba
| | - Xeila Sobrino-Iglesias
- Departamento de Química-Física, Centro de Ingeniería Genética y Biotecnología, La Habana, Cuba
| | - Hilda Garay
- Departamento de Química-Física, Centro de Ingeniería Genética y Biotecnología, La Habana, Cuba
| | - Vladimir Besada
- Departamento de Química-Física, Centro de Ingeniería Genética y Biotecnología, La Habana, Cuba
| | - Celia Fernández-Ortega
- Departamento de Farmacéuticos, Centro de Ingeniería Genética y Biotecnología, La Habana, Cuba
| |
Collapse
|
14
|
Maritaz C, Combarel D, Dalban C, Blondel L, Broutin S, Marabelle A, Albiges L, Paci A. Nivolumab plasma concentration and clearance associated with overall survival in patients with renal cell carcinoma. J Immunother Cancer 2025; 13:e010059. [PMID: 39762076 PMCID: PMC11749330 DOI: 10.1136/jitc-2024-010059] [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: 07/09/2024] [Accepted: 11/23/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Nivolumab is an immune checkpoint inhibitor (ICI) that selectively inhibits programmed cell death protein 1 activation, restoring antitumor immunity. ICIs are indicated for various types of advanced solid tumors; however, not all patients benefit from them, and tools that could be used in the clinic to predict response to treatment represent an unmet need. Here we describe the development of a new population pharmacokinetic (PPK) model in patients treated with nivolumab in clinical trials. Applying the model to a patient population with renal cell carcinoma identified nivolumab clearance and plasma concentration as predictors of overall survival (OS). METHODS A custom liquid chromatography with tandem mass spectrometry method for quantifying nivolumab plasma concentration was developed and validated following the European Medicines Agency guidelines for bioanalytical method validation. The PPK model was developed using data from patients treated in the NIVIPIT (n=38) and NIVOREN (n=137) trials of nivolumab in metastatic melanoma and renal cell carcinoma, respectively. The PPK model was used to determine pharmacokinetic (PK) parameters such as baseline clearance and simulate individual clearance changes over time. The relationship between PK characteristics (including clearance at Cycle 1 (CLC1), plasma concentration at Cycle 3 and clinical outcomes was assessed in 137 patients treated in NIVOREN. Kaplan-Meier methodology was used in time-to-event analyses. RESULTS In 137 patients, the median nivolumab CLC1 was 6 mL/hour and the median plasma concentration at Cycle 3 was 48 µg/mL. Median follow-up was 21.0 months (95% CI 20.2 to 22.5 months) with a survival rate at 6 months of 91.2% and 77.9% at 12 months. In univariate analysis, OS was significantly higher in patients with CLC1<6 mL/hour versus ≥6 mL/hour (HR 2.2 (95% CI 1.2 to 4.1), p=0.0146). Shorter OS was observed in patients with plasma concentration at Cycle 3 below the median (48 µg/mL) versus those above the median (HR 0.4 (95% CI 0.2 to 0.8), p=0.0069). Multivariate analysis showed a trend towards lower clearance, but this did not reach statistical significance (p=0.0694). CONCLUSIONS Results of the study may potentially be used to predict outcomes of nivolumab therapy in patients with renal cell carcinoma. Additional applications may include guiding dose adjustments of nivolumab in those who are less likely to respond to the initial dose.
Collapse
Affiliation(s)
- Christophe Maritaz
- Paris-Saclay University, Gif-sur-Yvette, Île-de-France, France
- Gustave Roussy, Villejuif, Île-de-France, France
- Pharmacology Department, Gustave Roussy, Villejuif, Île-de-France, France
| | - David Combarel
- Paris-Saclay University, Gif-sur-Yvette, Île-de-France, France
- Gustave Roussy, Villejuif, Île-de-France, France
- Pharmacology Department, Gustave Roussy, Villejuif, Île-de-France, France
| | - Cécile Dalban
- Biostatistics Department, Centre Leon Berard, Lyon, Auvergne-Rhône-Alpes, France
| | | | | | | | | | - Angelo Paci
- Paris-Saclay University, Gif-sur-Yvette, Île-de-France, France
- Gustave Roussy, Villejuif, Île-de-France, France
- Pharmacology Department, Gustave Roussy, Villejuif, Île-de-France, France
| |
Collapse
|
15
|
Petroff BK, Eustace R, Thompson KA, Kozlowski C, Agnew D. Endocrine Diagnostics: Principles and Applications. Vet Clin North Am Exot Anim Pract 2025; 28:1-14. [PMID: 39414472 DOI: 10.1016/j.cvex.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Endocrine diagnostics currently depend on the ability to measure low and high concentrations of diagnostic hormones using immunoassays. This often is challenging in species other than humans, dogs, cats, and horses due to lack of validated assays and reference intervals. There are strategies to approach endocrine testing in zoo, wildlife, and zoologic companion animals but caution is needed in interpreting results. Newer techniques such as liquid chromatography-tandem mass spectrometry (LC-MS/MS) may be more useful for all species, although technical hurdles remain for this method too.
Collapse
Affiliation(s)
- Brian K Petroff
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, MI, USA; Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA.
| | - Ronan Eustace
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, MI, USA
| | - Kimberly A Thompson
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, MI, USA
| | - Corinne Kozlowski
- Department of Reproductive and Behavioral Sciences, Saint Louis Zoo, Saint Louis, MO, USA
| | - Dalen Agnew
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, MI, USA; Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
16
|
Zimmerman‐Federle H, Ren G, Dowling S, Warren C, Rusyniak D, Avera R, Manicke NE. Plasma drug screening using paper spray mass spectrometry with integrated solid phase extraction. Drug Test Anal 2025; 17:138-151. [PMID: 38584344 PMCID: PMC11729250 DOI: 10.1002/dta.3687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/04/2024] [Accepted: 03/14/2024] [Indexed: 04/09/2024]
Abstract
Drug overdoses have risen dramatically in recent years. We developed a simple nontargeted method using a disposable paper spray cartridge with an integrated solid phase extraction column. This method was used to screen for ~160 fentanyl analogs, synthetic cannabinoids, other synthetic drugs, and traditional drugs of abuse in over 300 authentic overdose samples collected at emergency departments in Indianapolis. A solid phase extraction step was implemented on the paper spray cartridge to enable subnanograms per milliliter synthetic drugs screening in plasma. Analysis was performed on a quadrupole orbitrap mass spectrometer using the sequential window acquisition of all theoretical fragment ion spectra approach in which tandem mass spectrometry was performed using 7 m/z isolation windows in the quadrupole. Calibration curves with isotopically labeled internal standards were constructed for 35 of the most frequently encountered synthetic and traditional illicit drugs by US toxicology labs. Additional qualitative-only drugs in a suspect screening list were also included. Limits of detection in plasma for synthetic cannabinoids ranged from 0.1 to 0.5 and 0.1 to 0.3 ng/mL for fentanyl and its analogs and between 1 and 5 ng/mL for most other drugs. Relative matrix effects were evaluated by determining the variation of the calibration slope in 10 different lots of biofluid and found to be between 3% and 20%. The method was validated on authentic overdose samples collected from two emergency departments in Indianapolis, Indiana, from suspected or known overdoses. Commonly detected synthetic drugs included fentanyl related substances, designer benzodiazepines such as flubromazolam, and the synthetic cannabinoid 5F-PB-22.
Collapse
Affiliation(s)
- Hannah Zimmerman‐Federle
- Department of Chemistry and Chemical BiologyIndiana University–Purdue University IndianapolisIndianapolisIndianaUSA
| | - Greta Ren
- Department of Chemistry and Chemical BiologyIndiana University–Purdue University IndianapolisIndianapolisIndianaUSA
| | - Sarah Dowling
- Department of Chemistry and Chemical BiologyIndiana University–Purdue University IndianapolisIndianapolisIndianaUSA
| | - Cassandra Warren
- Department of Chemistry and Chemical BiologyIndiana University–Purdue University IndianapolisIndianapolisIndianaUSA
| | | | - Robert Avera
- School of MedicineIndiana UniversityIndianapolisIndianaUSA
| | - Nicholas E. Manicke
- Department of Chemistry and Chemical BiologyIndiana University–Purdue University IndianapolisIndianapolisIndianaUSA
| |
Collapse
|
17
|
Remes PM, Jacob CC, Heil LR, Shulman N, MacLean BX, MacCoss MJ. Hybrid Quadrupole Mass Filter-Radial Ejection Linear Ion Trap and Intelligent Data Acquisition Enable Highly Multiplex Targeted Proteomics. J Proteome Res 2024; 23:5476-5486. [PMID: 39475161 PMCID: PMC11956834 DOI: 10.1021/acs.jproteome.4c00599] [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: 11/06/2024]
Abstract
Targeted mass spectrometry (MS) methods are powerful tools for the selective and sensitive analysis of peptides identified in global discovery experiments. Selected reaction monitoring (SRM) is the most widely accepted clinical MS method due to its reliability and performance. However, SRM and parallel reaction monitoring (PRM) are limited in throughput and are typically used for assays with around 100 targets or fewer. Here we introduce a new MS platform featuring a quadrupole mass filter, collision cell, and linear ion trap architecture, capable of targeting 5000-8000 peptides per hour. This high multiplexing capability is facilitated by acquisition rates of 70-100 Hz and real-time chromatogram alignment. We present a Skyline external software tool for building targeted methods based on data-independent acquisition chromatogram libraries or unscheduled analysis of heavy labeled standards. Our platform demonstrates ∼10× lower limits of quantitation (LOQs) than traditional SRM on a triple quadrupole instrument for highly multiplexed assays, due to parallel product ion accumulation. Finally, we explore how analytical figures of merit vary with method duration and the number of analytes, providing insights into optimizing assay performance.
Collapse
Affiliation(s)
- Philip M Remes
- Thermo Fisher Scientific, 355 River Oaks Parkway, San Jose, California 95134, United States
| | - Cristina C Jacob
- Thermo Fisher Scientific, 355 River Oaks Parkway, San Jose, California 95134, United States
| | - Lilian R Heil
- Thermo Fisher Scientific, 355 River Oaks Parkway, San Jose, California 95134, United States
| | - Nicholas Shulman
- Department of Genome Sciences, University of Washington, 3720 15th St. NE, Seattle, Washington 98195, United States
| | - Brendan X MacLean
- Department of Genome Sciences, University of Washington, 3720 15th St. NE, Seattle, Washington 98195, United States
| | - Michael J MacCoss
- Department of Genome Sciences, University of Washington, 3720 15th St. NE, Seattle, Washington 98195, United States
| |
Collapse
|
18
|
Petrovic I, LoPresti J, Fatemi S, Gianoukakis A, Burman K, Gomez-Lima CJ, Nguyen CT, Spencer CA. Influence of Thyroglobulin Autoantibodies on Thyroglobulin Levels Measured by Different Methodologies: IMA, LC-MS/MS, and RIA. J Clin Endocrinol Metab 2024; 109:3254-3263. [PMID: 38686504 PMCID: PMC11570386 DOI: 10.1210/clinem/dgae286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024]
Abstract
CONTEXT Serum thyroglobulin (Tg) measured by immunometric assay (IMA) is prone to underestimation due to Tg autoantibody (TgAb) interference, often prompting reflex Tg measurement by liquid chromatography/tandem mass spectrometry (MS) or radioimmunoassay (RIA). OBJECTIVE IMA, MS, and RIA methodologies were used to measure serum Tg in TgAb-negative (TgAb-) and TgAb-positive (TgAb+) patients with either distant metastatic differentiated thyroid cancer (DTC) or hyperthyroidism (HY)-patients in whom a detectable serum Tg would be expected. RESULTS When TgAb was absent, all methodologies detected Tg in the sera of all DTC and HY patients and reported appropriate Tg trends and treatment responses for DTC patients with progressive distant metastatic disease, albeit with high between-method variability (> 30% coefficient of variability). When TgAb was present, all methodologies reported lower serum Tg levels for both DTC and HY groups vs their respective TgAb- group. No Tg was detected by IMA or MS in ∼50% TgAb+ DTC patients (6% had no Tg detected by RIA). Surprisingly, 5% of TgAb+ HY patients also had no Tg detected by IMA or MS. The inverse log TgAb/log Tg correlations seen for the TgAb+ HY patient group with all methods suggested the presence of a TgAb-associated serum Tg-lowering effect. CONCLUSION (i) Between-method Tg variability necessitates method continuity when monitoring the Tg trends of TgAb- DTC patients. (ii) The presence and concentration of TgAb appeared to have a lowering effect on serum Tg measured by all methodologies (IMA, MS, and RIA). (iii) Since the reliability of Tg measured in the presence of TgAb is often uncertain, the TgAb trend (measured by the same method) may be a useful surrogate DTC tumor marker.
Collapse
Affiliation(s)
- Ivana Petrovic
- Division of Endocrinology, Diabetes, & Metabolism, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jonathan LoPresti
- Division of Endocrinology, Diabetes, & Metabolism, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Shireen Fatemi
- Department of Endocrinology, Kaiser Permanente, Panorama City, CA 91402, USA
| | - Andrew Gianoukakis
- Division of Endocrinology, Lundquist Institute, Torrance, CA 90509, USA
- Division of Endocrinology, Diabetes and Metabolism, Harbor-UCLA Medical Center, Torrance, CA 90509, USA
| | - Kenneth Burman
- Endocrine Section, Medstar Washington Hospital Center, Washington, DC 20010, USA
| | | | - Caroline T Nguyen
- Division of Endocrinology, Diabetes, & Metabolism, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Carole Ann Spencer
- Division of Endocrinology, Diabetes, & Metabolism, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
19
|
Varga GM, Spendal M, Sigh J, Søeborg T, Nielsen NJ. Interference from anti-drug antibodies on the quantification of insulin: a comparison of an LC-MS/MS assay and immunoassays. Bioanalysis 2024; 16:1-11. [PMID: 39262387 PMCID: PMC11485705 DOI: 10.1080/17576180.2024.2389637] [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: 06/01/2024] [Accepted: 08/05/2024] [Indexed: 09/13/2024] Open
Abstract
Aim: This study aims to compare the anti-drug antibody (ADA) interference in four pharmacokinetic (PK) assays across different platforms (AlphaLISA, Gyrolab, LC-MS/MS) and to devise a strategy for ADA interference mitigation to improve the accuracy of measured drug in total PK assays.Materials & methods: Spiked test samples, created to achieve different ADA concentrations in human serum also containing an insulin analogue, were analyzed alongside pooled clinical samples using four assays.Results & conclusion: Interference was observed in all platforms. A novel approach using the Gyrolab mixing CD, including acid dissociation in the PK assay, significantly reduced interference and thereby improved relative error from >99% to ≤20% yielding measurements well within the acceptance criteria. Clinical sample results reinforced findings from the test samples.
Collapse
Affiliation(s)
- Georgina Marta Varga
- Analytical Chemistry Group, Department of Plant & Environmental Science, Faculty of Science, University of Copenhagen, Frederiksberg C, DK-1871, Denmark
- Non-clinical & Clinical Assay Sciences, Global Discovery & Development Sciences, Novo Nordisk A/S Måløv, DK-2760, Denmark
| | - Manca Spendal
- Analytical Chemistry Group, Department of Plant & Environmental Science, Faculty of Science, University of Copenhagen, Frederiksberg C, DK-1871, Denmark
- Non-clinical & Clinical Assay Sciences, Global Discovery & Development Sciences, Novo Nordisk A/S Måløv, DK-2760, Denmark
| | - Jens Sigh
- Non-clinical & Clinical Assay Sciences, Global Discovery & Development Sciences, Novo Nordisk A/S Måløv, DK-2760, Denmark
| | - Tue Søeborg
- Non-clinical & Clinical Assay Sciences, Global Discovery & Development Sciences, Novo Nordisk A/S Måløv, DK-2760, Denmark
| | - Nikoline Juul Nielsen
- Analytical Chemistry Group, Department of Plant & Environmental Science, Faculty of Science, University of Copenhagen, Frederiksberg C, DK-1871, Denmark
| |
Collapse
|
20
|
Johnson JS, Palandra J, Psychogios N, Walsh JM, Neubert H. Improving the Sensitivity of Protein Quantification by Immunoaffinity Liquid Chromatography─Triple Quadrupole Mass Spectrometry Using an Iterative Transition Summing Technique. Anal Chem 2024; 96:14326-14331. [PMID: 39186623 PMCID: PMC11391403 DOI: 10.1021/acs.analchem.3c04598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024]
Abstract
The desire to reach ever-diminishing lower limits of quantification (LLOQ) to probe changes in low abundance protein targets has led to enormous progress in sample preparation and liquid chromatography-tandem mass spectrometry (LC-MS/MS) instrumentation. To maximize signal and reduce noise, many approaches have been employed, including specific immunoaffinity (IA) enrichment and reducing the LC flow to the nanoflow (nLC) level; however, additional sensitivity gains may still be required. Recently, a technique termed "echo summing" has been described for small-molecular-weight analytes on a triple quadrupole (QqQ) MS where multiple iterations of the same, single selected reaction monitoring (SRM) transition are collected, summed, and integrated, yielding significant analyte dependent signal-to-noise (S/N) improvements. Herein, the direct applicability of echo summing to protein quantification by sequential IA combined with nLC-MS/MS (IA-nLC-MS/MS) is described for a beta nerve growth factor (NGF) and a soluble asialoglycoprotein receptor (sASGPR) assay from human serum. Five iterations of echo summing outperformed traditional collection in relative average accuracy (-1.5 ± 7.7 vs -41.7 ± 10.7% bias) and precision (7.8 vs 18.4% coefficient of variation (CV)) of the low-end quality control (QC) sample (N = 4) for NGF and improved functional sensitivity of serially diluted serum QC samples (N = 5 each population) approximately 2-fold (1.96 and 2.00-fold) for two peptides of sASGPR. Echo summing also extended the minimum quantifiable QC level for sASGPR 4-fold lower. Similar gains are believed to be achievable for most protein IA-nLC-MS/MS assays.
Collapse
Affiliation(s)
- Jay S. Johnson
- Pharmacokinetics, Dynamics
& Metabolism (PDM), Pfizer, Andover, Massachusetts 01810, United States
| | | | - Nikolaos Psychogios
- Pharmacokinetics, Dynamics
& Metabolism (PDM), Pfizer, Andover, Massachusetts 01810, United States
| | - Jason M. Walsh
- Pharmacokinetics, Dynamics
& Metabolism (PDM), Pfizer, Andover, Massachusetts 01810, United States
| | - Hendrik Neubert
- Pharmacokinetics, Dynamics
& Metabolism (PDM), Pfizer, Andover, Massachusetts 01810, United States
| |
Collapse
|
21
|
Lam KHB, Menlyadiev M, Buggs V, Parnprome S, Pesce A, Suhandynata RT, Fitzgerald RL, Song L, Metushi IG. A Comparative Analysis of Two Commonly Used FDA-Approved Immunoassays for Fentanyl Detection. J Appl Lab Med 2024; 9:905-912. [PMID: 38831664 DOI: 10.1093/jalm/jfae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 04/25/2024] [Indexed: 06/05/2024]
Abstract
BACKGROUND Given the opioid epidemic, fentanyl screening in urine has become increasingly important. Immunoassays remain the most common screening methodology due to the high throughput and ease of integration into automated chemistry systems. The fentanyl ARK II from Ark Diagnostics is a widely used immunoassay, while a novel fentanyl assay called FEN2 by Lin-Zhi has become available on the Roche platform. Here, we evaluate and compare their performance. METHODS Four hundred and thirty-four urine samples were analyzed for fentanyl across the Lin-Zhi FEN2 and ARK II assays on the Cobas c502 platform. Samples were analyzed immediately upon request for drug of abuse screening or frozen for subsequent analysis. For confirmation testing, a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method with a limit of detection of 1 ng/mL for fentanyl/norfentanyl was used. Any sample with either fentanyl or norfentanyl above the LC-MS/MS cutoff was deemed positive. RESULTS The ARK II had 11 false negatives and 7 false positives, while the Lin-Zhi FEN2 had 12 false negatives and 2 false positives. This resulted in ARK II having a sensitivity and specificity of 90.4% and 97.8% respectively, while Lin-Zhi FEN2 had a sensitivity and specificity of 89.5% and 99.4%. CONCLUSIONS Both the ARK II and Lin-Zhi FEN2 immunoassays detected fentanyl well. Overall, the Lin-Zhi assay had slightly better specificity than ARK II, in our data set. While some discrepant results were observed between the 2 immunoassay systems, most occurred near the immunoassay detection cutoffs.
Collapse
Affiliation(s)
- K H Brian Lam
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Marlen Menlyadiev
- Department of Pathology, Geisinger Commonwealth School of Medicine and Geisinger Health System, Danville, PA, United States
- Department of Pathology, Center for Advanced Laboratory Medicine, University of California, San Diego Health Systems, San Diego, CA, United States
| | - Vincent Buggs
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Suttida Parnprome
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Amadeo Pesce
- Precision Diagnostics (PDx), LLC, San Diego, CA, United States
| | - Raymond T Suhandynata
- Department of Pathology, Center for Advanced Laboratory Medicine, University of California, San Diego Health Systems, San Diego, CA, United States
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, San Diego, CA, United States
| | - Robert L Fitzgerald
- Department of Pathology, Center for Advanced Laboratory Medicine, University of California, San Diego Health Systems, San Diego, CA, United States
| | - Lu Song
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Imir G Metushi
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
22
|
Camilleri E, Kruijt M, den Exter PL, Cannegieter SC, van Rein N, Cobbaert CM, van Vlijmen BJM, Ruhaak LR. Quantitative protein mass spectrometry for multiplex measurement of coagulation and fibrinolytic proteins towards clinical application: What, why and how? Thromb Res 2024; 241:109090. [PMID: 39032389 DOI: 10.1016/j.thromres.2024.109090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/20/2024] [Accepted: 07/03/2024] [Indexed: 07/23/2024]
Abstract
Plasma proteins involved in coagulation and fibrinolysis are essential to hemostasis. Consequently, their circulating levels and functionality are critical in bleeding and thrombosis development. Well-established laboratory tests to assess these are available; however, said tests do not allow high multiplicity, require large volumes of plasma and are often costly. A novel technology to quantify plasma proteins is quantitative protein mass spectrometry (QPMS). Aided by stable isotope-labeled internal standards a large number of proteins can be quantified in one single analytical run requiring <30 μL of plasma. This provides an opportunity to improve insight in the etiology and prognosis of bleeding and thrombotic disorders, in which the balance between different proteins plays a crucial role. This manuscript aims to give an overview of the QPMS potential applications in thrombosis and hemostasis research (quantifying the 38 proteins assigned to coagulation and fibrinolysis by the KEGG database), but also to explore the potential and hurdles if designed for clinical practice. Advantages and limitations of QPMS are described and strategies for improved analysis are proposed, using as an example the test requirements for antithrombin. Application of this technology in the future could represent a step towards individualized patient care.
Collapse
Affiliation(s)
- Eleonora Camilleri
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Mirjam Kruijt
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Paul L den Exter
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | - Suzanne C Cannegieter
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands; Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Nienke van Rein
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands; Department of Pharmacy, Leiden University Medical Center, Leiden, the Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Bart J M van Vlijmen
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| | - L Renee Ruhaak
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
23
|
Yoshijima C, Suzuki Y, Tanaka R, Ono H, Oda A, Ozaki T, Shibata H, Itoh H, Ohno K. Evaluation of intra- and inter-individual variations in plasma belimumab concentrations in adult patients with systemic lupus erythematosus. Pharmacol Res Perspect 2024; 12:e1255. [PMID: 39096038 PMCID: PMC11297282 DOI: 10.1002/prp2.1255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
In this study, plasma belimumab concentrations were measured over the course of treatment in systemic lupus erythematosus (SLE) patients on belimumab therapy, and intra- and interindividual variations in plasma belimumab concentration were evaluated. A single-center prospective study was conducted at Oita University Hospital to evaluate trough plasma concentrations over the course of treatment in 13 SLE patients treated with intravenous belimumab. Plasma belimumab concentrations were measured by a validated ultra-high performance liquid chromatography with tandem mass spectrometry method. The median age of the patients was 40 (interquartile range: 35-51) years and the median weight was 51.8 (47.0-58.1) kg. A mean of 9.4 (range: 1-13) blood samples was collected per patient at routine visits. The mean (± SD) plasma belimumab concentration was 33.4 ± 11.9 μg/mL in the patient with the lowest concentration and 170.0 ± 16.6 μg/mL in the patient with the highest concentration, indicating a 5-fold difference between patients. On the other hand, the within-patient coefficient of variation ranged from 7.1% to 35.7%, showing no large variations. No significant correlation was observed between plasma belimumab concentration and belimumab dose (mg/kg) (Spearman's rank correlation coefficient = 0.22, p = .54). Examinations of trough plasma belimumab concentrations over the course of treatment in patients with SLE showed small intraindividual variation but large interindividual variation. Plasma belimumab trough concentration varied widely among patients administered the approved dose.
Collapse
MESH Headings
- Humans
- Lupus Erythematosus, Systemic/drug therapy
- Lupus Erythematosus, Systemic/blood
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/blood
- Adult
- Female
- Middle Aged
- Prospective Studies
- Male
- Immunosuppressive Agents/pharmacokinetics
- Immunosuppressive Agents/blood
- Immunosuppressive Agents/administration & dosage
- Immunosuppressive Agents/therapeutic use
- Tandem Mass Spectrometry
- Chromatography, High Pressure Liquid
- Drug Monitoring/methods
Collapse
Affiliation(s)
- Chisato Yoshijima
- Department of Medication Use Analysis and Clinical ResearchMeiji Pharmaceutical UniversityTokyoJapan
| | - Yosuke Suzuki
- Department of Medication Use Analysis and Clinical ResearchMeiji Pharmaceutical UniversityTokyoJapan
| | - Ryota Tanaka
- Department of Clinical PharmacyOita University HospitalYufuOitaJapan
| | - Hiroyuki Ono
- Department of Clinical PharmacyOita University HospitalYufuOitaJapan
| | - Ayako Oda
- Department of Medication Use Analysis and Clinical ResearchMeiji Pharmaceutical UniversityTokyoJapan
| | - Takashi Ozaki
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of MedicineOita UniversityYufuOitaJapan
| | - Hirotaka Shibata
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of MedicineOita UniversityYufuOitaJapan
| | - Hiroki Itoh
- Department of Clinical PharmacyOita University HospitalYufuOitaJapan
| | - Keiko Ohno
- Department of Medication Use Analysis and Clinical ResearchMeiji Pharmaceutical UniversityTokyoJapan
| |
Collapse
|
24
|
Kreft IC, van Duijl TT, van Kwawegen C, Atiq F, Phan W, Schuller MBP, Boon-Spijker M, van der Zwaan C, Meijer AB, Hoogendijk AJ, Bierings R, Eikenboom JCJ, Leebeek FWG, van den Biggelaar M. Variant mapping using mass spectrometry-based proteotyping as a diagnostic tool in von Willebrand disease. J Thromb Haemost 2024; 22:1894-1908. [PMID: 38679335 DOI: 10.1016/j.jtha.2024.04.011] [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: 01/17/2024] [Revised: 03/20/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND von Willebrand disease (VWD) is the most common inherited bleeding disorder, characterized by either partial or complete von Willebrand factor (VWF) deficiency or by the occurrence of VWF proteoforms of altered functionality. The gene encoding VWF is highly polymorphic, giving rise to a variety of proteoforms with varying plasma concentrations and clinical significance. OBJECTIVES To address this complexity, we translated genomic variation in VWF to corresponding VWF proteoforms circulating in blood. METHODS VWF was characterized in VWD patients (n = 64) participating in the Willebrand in the Netherlands study by conventional laboratory testing, DNA sequencing and complementary discovery, and targeted mass spectrometry-based plasma proteomic strategies. RESULTS Unbiased plasma profiling combined with immune enrichment of VWF verified VWF and its binding partner factor VIII as key determinants of VWD and revealed a remarkable heterogeneity in VWF amino acid sequence coverage among patients. Subsequent VWF proteotyping enabled identification of both polymorphisms (eg, p.Thr789Ala, p.Gln852Arg, and p.Thr1381Ala), as well as pathogenic variants (n = 16) along with their corresponding canonical sequences. Targeted proteomics using stable isotope-labeled peptides confirmed unbiased proteotyping for 5 selected variants and suggested differential proteoform quantities in plasma. The variant-to-wild-type peptide ratio was determined in 6 type 2B patients heterozygous for p.Arg1306Trp, confirming the relatively low proteoform concentration of the pathogenic variant. The elevated VWF propeptide/VWF ratio indicated increased clearance of specific VWF proteoforms. CONCLUSION This study highlights how VWF proteotyping from plasma could be the first step to bridge the gap between genotyping and functional testing in VWD.
Collapse
Affiliation(s)
- Iris C Kreft
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands.
| | - Tirsa T van Duijl
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Calvin van Kwawegen
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ferdows Atiq
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Winny Phan
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Margo B P Schuller
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Mariëtte Boon-Spijker
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Carmen van der Zwaan
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Alexander B Meijer
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands; Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Arie J Hoogendijk
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Ruben Bierings
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jeroen C J Eikenboom
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | - Frank W G Leebeek
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Maartje van den Biggelaar
- Laboratory of Proteomics, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands.
| |
Collapse
|
25
|
Cui Z, Ayva CE, Liew YJ, Guo Z, Mutschler R, Dreier B, Fiorito MM, Walden P, Howard CB, Ely F, Plückthun A, Pretorius C, Ungerer JPJ, Buckle AM, Alexandrov K. mRNA Display Pipeline for Protein Biosensor Construction. ACS Sens 2024; 9:2846-2857. [PMID: 38807313 PMCID: PMC11218749 DOI: 10.1021/acssensors.3c02471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 05/01/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Despite the significant potential of protein biosensors, their construction remains a trial-and-error process. The most obvious approach for addressing this is to utilize modular biosensor architectures where specificity-conferring modalities can be readily generated to recognize new targets. Toward this goal, we established a workflow that uses mRNA display-based selection of hyper-stable monobody domains for the target of choice or ribosome display to select equally stable DARPins. These binders were integrated into a two-component allosteric biosensor architecture based on a calmodulin-reporter chimera. This workflow was tested by developing biosensors for liver toxicity markers such as cytosolic aspartate aminotransferase, mitochondrial aspartate aminotransferase, and alanine aminotransferase 1. We demonstrate that our pipeline consistently produced >103 unique binders for each target within a week. Our analysis revealed that the affinity of the binders for their targets was not a direct predictor of the binder's performance in a biosensor context. The interactions between the binding domains and the reporter module affect the biosensor activity and the dynamic range. We conclude that following binding domain selection, the multiplexed biosensor assembly and prototyping appear to be the most promising approach for identifying biosensors with the desired properties.
Collapse
Affiliation(s)
- Zhenling Cui
- ARC
Centre of Excellence in Synthetic Biology, Brisbane, Queensland 4001, Australia
- Centre
for Agriculture and the Bioeconomy, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
- School
of Biology and Environmental Science, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
| | - Cagla Ergun Ayva
- Centre
for Agriculture and the Bioeconomy, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
- School
of Biology and Environmental Science, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
| | - Yi Jin Liew
- CSIRO
Health & Biosecurity, Westmead, New South Wales 2145,Australia
| | - Zhong Guo
- ARC
Centre of Excellence in Synthetic Biology, Brisbane, Queensland 4001, Australia
- Centre
for Agriculture and the Bioeconomy, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
- School
of Biology and Environmental Science, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
| | - Roxane Mutschler
- Centre
for Agriculture and the Bioeconomy, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
- School
of Biology and Environmental Science, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
| | - Birgit Dreier
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, Zurich CH-8057, Switzerland
| | - Maria M Fiorito
- Centre
for Agriculture and the Bioeconomy, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
- School
of Biology and Environmental Science, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
| | - Patricia Walden
- Centre
for Agriculture and the Bioeconomy, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
- School
of Biology and Environmental Science, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
| | - Christopher B Howard
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | | | - Andreas Plückthun
- Department
of Biochemistry, University of Zurich, Winterthurerstrasse 190, Zurich CH-8057, Switzerland
| | - Carel Pretorius
- Department
of Chemical Pathology, Pathology Queensland, Brisbane, Queensland 4006, Australia
- Faculty
of Health and Behavioural Sciences, The
University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jacobus PJ Ungerer
- Department
of Chemical Pathology, Pathology Queensland, Brisbane, Queensland 4006, Australia
- Faculty
of Health and Behavioural Sciences, The
University of Queensland, Brisbane, Queensland 4072, Australia
| | | | - Kirill Alexandrov
- ARC
Centre of Excellence in Synthetic Biology, Brisbane, Queensland 4001, Australia
- Centre
for Agriculture and the Bioeconomy, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
- School
of Biology and Environmental Science, Queensland
University of Technology, Brisbane, Queensland 4001, Australia
| |
Collapse
|
26
|
Yu Z, Wang J, Xia W, Wang Y, Zhang Y, Tang J, Cui H, Yang X, Bao C, Ye Z. The Development of an Isotope Dilution Mass Spectrometry Method for Interleukin-6 Quantification. Int J Mol Sci 2024; 25:6777. [PMID: 38928482 PMCID: PMC11203838 DOI: 10.3390/ijms25126777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Inflammatory responses and tumor developments are closely related, with interleukin-6 (IL-6) playing important roles in both processes. IL-6 has been extensively identified as a potential tumor biomarker. This study developed an isotope dilution mass spectrometry (IDMS) method for quantifying IL-6 based on signature peptides. These peptides were screened by excluding those with missed cleavage or post-translational modification. The method's accuracy was verified using amino acid-based IDMS, in which purified IL-6 protein samples were quantified after hydrolyzing them into amino acids, and no significant difference was observed (p-value < 0.05). The method demonstrated good linearity and sensitivity upon testing. The specificity and matrix effect of the method were verified, and a precision study showed that the coefficient of variation was less than 5% for both the intra-day and inter-day tests. Compared to immunoassays, this method offers distinct advantages, such as the facilitation of multi-target analysis. Furthermore, the peptides used in this study are much more convenient for storage and operation than the antibodies or purified proteins typically used in immunoassays.
Collapse
Affiliation(s)
- Zetao Yu
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (Z.Y.); (J.W.); (Y.W.); (Y.Z.); (J.T.); (H.C.); (X.Y.); (C.B.)
| | - Jing Wang
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (Z.Y.); (J.W.); (Y.W.); (Y.Z.); (J.T.); (H.C.); (X.Y.); (C.B.)
| | - Wenqiang Xia
- Institute of Crop Science, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310012, China;
| | - Yuemin Wang
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (Z.Y.); (J.W.); (Y.W.); (Y.Z.); (J.T.); (H.C.); (X.Y.); (C.B.)
| | - Yafen Zhang
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (Z.Y.); (J.W.); (Y.W.); (Y.Z.); (J.T.); (H.C.); (X.Y.); (C.B.)
| | - Jintian Tang
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (Z.Y.); (J.W.); (Y.W.); (Y.Z.); (J.T.); (H.C.); (X.Y.); (C.B.)
| | - Haifeng Cui
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (Z.Y.); (J.W.); (Y.W.); (Y.Z.); (J.T.); (H.C.); (X.Y.); (C.B.)
| | - Xiaoying Yang
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (Z.Y.); (J.W.); (Y.W.); (Y.Z.); (J.T.); (H.C.); (X.Y.); (C.B.)
| | - Chenchen Bao
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (Z.Y.); (J.W.); (Y.W.); (Y.Z.); (J.T.); (H.C.); (X.Y.); (C.B.)
| | - Zihong Ye
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou 310018, China; (Z.Y.); (J.W.); (Y.W.); (Y.Z.); (J.T.); (H.C.); (X.Y.); (C.B.)
| |
Collapse
|
27
|
Rydberg M, Bruening ML, Manicke NE. Paper Spray Mass Spectrometry with On-Paper Electrokinetic Manipulations: Part-Per-Trillion Detection of Per/Polyfluoroalkyl Substances in Water and Opioids in Urine. Angew Chem Int Ed Engl 2024; 63:e202401729. [PMID: 38657037 PMCID: PMC11344455 DOI: 10.1002/anie.202401729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 04/26/2024]
Abstract
We developed a simple, paper-based device that enables sensitive detection by mass spectrometry (MS) without solid phase extraction or other sample preparation. Using glass fiber filter papers within a 3D printed holder, the device employs electrokinetic manipulations to stack, separate, and desalt charged molecules on paper prior to spray into the MS. Due to counter-balanced electroosmotic flow and electrophoresis, charged analytes stack on the paper and desalting occurs in minutes. One end of the paper strip was cut into a sharp point and positioned near the inlet of a MS. The stacked analyte bands move toward the paper tip with the EOF where they are ionized by paper spray. The device was applied to analysis of PFAS in tap water with sub part-per-trillion detection limits in less than ten minutes with no sample pretreatment. Analysis of opioids in urine also occurs in minutes. The crucial parameters to enable stacking, separation, and MS ionization of both positively and negatively charged analytes were determined and optimized. Experimental and computational modeling studies confirm the electrokinetic stacking and analyte transport mechanisms. On-paper separations were carried out by stacking analyte bands at different locations depending on their electrophoretic mobility, achieving baseline separation in some cases.
Collapse
Affiliation(s)
- Magnus Rydberg
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University Indianapolis
| | - Merlin L Bruening
- Department of Chemical & Biomolecular Engineering, University of Notre Dame
| | - Nicholas E Manicke
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University Indianapolis
| |
Collapse
|
28
|
Plubell DL, Remes PM, Wu CC, Jacob CC, Merrihew GE, Hsu C, Shulman N, MacLean BX, Heil L, Poston K, Montine T, MacCoss MJ. Development of highly multiplex targeted proteomics assays in biofluids using the Stellar mass spectrometer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597431. [PMID: 38895256 PMCID: PMC11185584 DOI: 10.1101/2024.06.04.597431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The development of targeted assays that monitor biomedically relevant proteins is an important step in bridging discovery experiments to large scale clinical studies. Targeted assays are currently unable to scale to hundreds or thousands of targets. We demonstrate the generation of large-scale assays using a novel hybrid nominal mass instrument. The scale of these assays is achievable with the Stellar™ mass spectrometer through the accommodation of shifting retention times by real-time alignment, while being sensitive and fast enough to handle many concurrent targets. Assays were constructed using precursor information from gas-phase fractionated (GPF) data-independent acquisition (DIA). We demonstrate the ability to schedule methods from an orbitrap and linear ion trap acquired GPF DIA library and compare the quantification of a matrix-matched calibration curve from orbitrap DIA and linear ion trap parallel reaction monitoring (PRM). Two applications of these proposed workflows are shown with a cerebrospinal fluid (CSF) neurodegenerative disease protein PRM assay and with a Mag-Net enriched plasma extracellular vesicle (EV) protein survey PRM assay.
Collapse
Affiliation(s)
- Deanna L Plubell
- Department of Genome Sciences, University of Washington, Seattle WA
| | | | - Christine C Wu
- Department of Genome Sciences, University of Washington, Seattle WA
| | | | | | - Chris Hsu
- Department of Genome Sciences, University of Washington, Seattle WA
| | - Nick Shulman
- Department of Genome Sciences, University of Washington, Seattle WA
| | | | | | - Kathleen Poston
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto CA, USA
| | - Tom Montine
- Department of Pathology, Stanford University, Palo Alto CA, USA
| | | |
Collapse
|
29
|
Moradian A, Goonatileke E, Lin TT, Hatten-Beck M, Emrick M, Schepmoes AA, Fillmore TL, MacCoss MJ, Sechi S, Sobhani K, Little R, Kabytaev K, van Eyk JE, Qian WJ, Hoofnagle AN. Interlaboratory Comparison of Antibody-Free LC-MS/MS Measurements of C-peptide and Insulin. Clin Chem 2024; 70:855-864. [PMID: 38549041 PMCID: PMC12052497 DOI: 10.1093/clinchem/hvae034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/29/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND The enhanced precision and selectivity of liquid chromatography-tandem mass spectrometry (LC-MS/MS) makes it an attractive alternative to certain clinical immunoassays. Easily transferrable work flows could help facilitate harmonization and ensure high-quality patient care. We aimed to evaluate the interlaboratory comparability of antibody-free multiplexed insulin and C-peptide LC-MS/MS measurements. METHODS The laboratories that comprise the Targeted Mass Spectrometry Assays for Diabetes and Obesity Research (TaMADOR) consortium verified the performance of a validated peptide-based assay (reproducibility, linearity, and lower limit of the measuring interval [LLMI]). An interlaboratory comparison study was then performed using shared calibrators, de-identified leftover laboratory samples, and reference materials. RESULTS During verification, the measurements were precise (2.7% to 3.7%CV), linear (4 to 15 ng/mL for C-peptide and 2 to 14 ng/mL for insulin), and sensitive (LLMI of 0.04 to 0.10 ng/mL for C-peptide and 0.03 ng/mL for insulin). Median imprecision across the 3 laboratories was 13.4% (inter-quartile range [IQR] 11.6%) for C-peptide and 22.2% (IQR 20.9%) for insulin using individual measurements, and 10.8% (IQR 8.7%) and 15.3% (IQR 14.9%) for C-peptide and insulin, respectively, when replicate measurements were averaged. Method comparison with the University of Missouri reference method for C-peptide demonstrated a robust linear correlation with a slope of 1.044 and r2 = 0.99. CONCLUSIONS Our results suggest that combined LC-MS/MS measurements of C-peptide and insulin are robust and adaptable and that standardization with a reference measurement procedure could allow accurate and precise measurements across sites, which could be important to diabetes research and help patient care in the future.
Collapse
Affiliation(s)
- Annie Moradian
- Precision Biomarker Laboratories, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Elisha Goonatileke
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Tai-Tu Lin
- Integrative Omics, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA
| | - Maya Hatten-Beck
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Michelle Emrick
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Athena A Schepmoes
- Integrative Omics, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA
| | - Thomas L. Fillmore
- Integrative Omics, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA
| | | | - Salvatore Sechi
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Kimia Sobhani
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Randie Little
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO
| | - Kuanysh Kabytaev
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO
| | - Jennifer E. van Eyk
- Precision Biomarker Laboratories, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Wei-Jun Qian
- Integrative Omics, Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA
| | - Andrew N. Hoofnagle
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
- Kidney Research Institute, Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
30
|
Thiery J, Fahrner M. Integration of proteomics in the molecular tumor board. Proteomics 2024; 24:e2300002. [PMID: 38143279 DOI: 10.1002/pmic.202300002] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/03/2023] [Accepted: 12/05/2023] [Indexed: 12/26/2023]
Abstract
Cancer remains one of the most complex and challenging diseases in mankind. To address the need for a personalized treatment approach for particularly complex tumor cases, molecular tumor boards (MTBs) have been initiated. MTBs are interdisciplinary teams that perform in-depth molecular diagnostics to cooperatively and interdisciplinarily advise on the best therapeutic strategy. Current molecular diagnostics are routinely performed on the transcriptomic and genomic levels, aiming to identify tumor-driving mutations. However, these approaches can only partially capture the actual phenotype and the molecular key players of tumor growth and progression. Thus, direct investigation of the expressed proteins and activated signaling pathways provide valuable complementary information on the tumor-driving molecular characteristics of the tissue. Technological advancements in mass spectrometry-based proteomics enable the robust, rapid, and sensitive detection of thousands of proteins in minimal sample amounts, paving the way for clinical proteomics and the probing of oncogenic signaling activity. Therefore, proteomics is currently being integrated into molecular diagnostics within MTBs and holds promising potential in aiding tumor classification and identifying personalized treatment strategies. This review introduces MTBs and describes current clinical proteomics, its potential in precision oncology, and highlights the benefits of multi-omic data integration.
Collapse
Affiliation(s)
- Johanna Thiery
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Matthias Fahrner
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and Cancer Research Center (DKFZ), Freiburg, Germany
| |
Collapse
|
31
|
Liu Z, Zhao X, Liu Y, Shi L, Wu L, Yuan H, Jin Y. Development of an insulin-like growth factor-1 certified reference material by SI-traceable isotope-dilution mass spectrometry. Talanta 2024; 273:125812. [PMID: 38452589 DOI: 10.1016/j.talanta.2024.125812] [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: 12/04/2023] [Revised: 01/29/2024] [Accepted: 02/17/2024] [Indexed: 03/09/2024]
Abstract
In this study, an insulin-like growth factor-1 (IGF-1) certified reference material (CRM) was developed by the National Institute of Metrology (NIM), and two different principles for evaluating the IGF-1 CRM were established. After optimisation of the acid hydrolysis conditions (110 °C, 36 h), quantitative determination of peptide purity, and chromatographic separation and mass spectrometric detection, amino acid analysis-based high-performance liquid chromatography combined with isotope-dilution tandem mass spectrometry (AAA-HPLC-IDMS/MS) and peptide analysis-based HPLC-IDMS/MS (Peptide-HPLC-IDMS/MS) were used for certified value assignment; the results obtained were 136.28 and 135.01 μg/g, respectively, which were in good agreement. These results were subjected to the normal distribution test, outlier test, and method consistency test. The homogeneity and stability of the reference materials were also examined, and the uncertainty introduced in the experimental process was calculated. The final certified value was (136 ± 15) μg g-1 (k = 2). The CRM was found to be stable for at least six months when stored at -70 °C and for 7 d when stored at higher temperatures (-20 °C, 4 °C, 25 °C, or 40 °C). The CRM is expected to be used as a primary calibrator for quality control in biopharmaceutical production and clinical diagnostics.
Collapse
Affiliation(s)
- Zihan Liu
- National Institute of Metrology, Beijing, China; Department of Clinical Laboratory, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xu Zhao
- National Institute of Metrology, Beijing, China
| | - Yahui Liu
- National Institute of Metrology, Beijing, China
| | - Lianhua Shi
- National Institute of Metrology, Beijing, China
| | - Liqing Wu
- National Institute of Metrology, Beijing, China
| | - Hui Yuan
- Department of Clinical Laboratory, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Youxun Jin
- National Institute of Metrology, Beijing, China.
| |
Collapse
|
32
|
Remes PM, Jacob CC, Heil LR, Shulman N, MacLean BX, MacCoss MJ. Hybrid Quadrupole Mass Filter - Radial Ejection Linear Ion Trap and Intelligent Data Acquisition Enable Highly Multiplex Targeted Proteomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596848. [PMID: 38854069 PMCID: PMC11160808 DOI: 10.1101/2024.05.31.596848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Targeted mass spectrometry (MS) methods are powerful tools for selective and sensitive analysis of peptides identified by global discovery experiments. Selected reaction monitoring (SRM) is currently the most widely accepted MS method in the clinic, due to its reliability and analytical performance. However, due to limited throughput and the difficulty in setting up and analyzing large scale assays, SRM and parallel reaction monitoring (PRM) are typically used only for very refined assays of on the order of 100 targets or less. Here we introduce a new MS platform with a quadrupole mass filter, collision cell, linear ion trap architecture that has increased acquisition rates compared to the analogous hardware found in the Orbitrap™ Tribrid™ series instruments. The platform can target more analytes than existing SRM and PRM instruments - in the range of 5000 to 8000 peptides per hour. This capability for high multiplexing is enabled by acquisition rates of 70-100 Hz for peptide applications, and the incorporation of real-time chromatogram alignment that adjusts for retention time drift and enables narrow time scheduled acquisition windows. Finally, we describe a Skyline external software tool that implements the building of targeted methods based on data independent acquisition chromatogram libraries or unscheduled analysis of heavy labeled standards. We show that the platform delivers ~10x lower LOQs than traditional SRM analysis for a highly multiplex assay and also demonstrate how analytical figures of merit change while varying method duration with a constant number of analytes, or by keeping a constant time duration while varying the number of analytes.
Collapse
Affiliation(s)
- Philip M Remes
- Thermo Fisher Scientific, 355 River Oaks Parkway, San Jose, California 95134
| | - Cristina C Jacob
- Thermo Fisher Scientific, 355 River Oaks Parkway, San Jose, California 95134
| | - Lilian R Heil
- Thermo Fisher Scientific, 355 River Oaks Parkway, San Jose, California 95134
| | - Nicholas Shulman
- Department of Genome Sciences, University of Washington, 3720 15th St. NE, Seattle WA 98195
| | - Brendan X MacLean
- Department of Genome Sciences, University of Washington, 3720 15th St. NE, Seattle WA 98195
| | - Michael J MacCoss
- Department of Genome Sciences, University of Washington, 3720 15th St. NE, Seattle WA 98195
| |
Collapse
|
33
|
Chen J, Li Y, Chen J, Wang R, Lu M, Yu C. Miniature mass spectrometer-based point-of-care assay for quantification of metformin and sitagliptin in human blood and urine. Anal Bioanal Chem 2024; 416:3305-3312. [PMID: 38642098 DOI: 10.1007/s00216-024-05281-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/12/2024] [Accepted: 03/25/2024] [Indexed: 04/22/2024]
Abstract
Metformin (MET) and sitagliptin (STG) are widely used as the first-line and long-term oral hypoglycemic agents for managing type 2 diabetes mellitus (T2DM). However, the current lack of convenient and rapid measurement methods poses a challenge for individualized management. This study developed a point-of-care (POC) assay method utilizing a miniature mass spectrometer, enabling rapid and accurate quantification of MET and STG concentrations in human blood and urine. By combining the miniature mass spectrometer with paper spray ionization, this method simplifies the process into three to four steps, requires minimal amounts of bodily fluids (50 μL of blood and 2 μL of urine), and is able to obtain quantification results within approximately 2 min. Stable isotope-labeled internal standards were employed to enhance the accuracy and stability of measurement. The MS/MS responses exhibited good linear relationship with concentration, with relative standard deviations (RSDs) below 25%. It has the potential to provide immediate treatment feedback and decision support for patients and healthcare professionals in clinical practice.
Collapse
Affiliation(s)
- Jingying Chen
- Central Hospital Affiliated to Shandong First Medical University, Jinan, 250000, Shandong Province, China
| | - Yaohan Li
- Central Hospital Affiliated to Shandong First Medical University, Jinan, 250000, Shandong Province, China
| | - Jingjing Chen
- Central Hospital Affiliated to Shandong First Medical University, Jinan, 250000, Shandong Province, China
| | - Ruimin Wang
- Central Hospital Affiliated to Shandong First Medical University, Jinan, 250000, Shandong Province, China
| | - Miaoshan Lu
- Central Hospital Affiliated to Shandong First Medical University, Jinan, 250000, Shandong Province, China
| | - Changbin Yu
- Central Hospital Affiliated to Shandong First Medical University, Jinan, 250000, Shandong Province, China.
| |
Collapse
|
34
|
Zhang L, Nizhamuding X, Zheng H, Zeng J, Yuan X, Ma Z, Zhou W, Zhang C, Zhang T, Zhang C. An LC-MS/MS method for serum cystatin C quantification and its comparison with two commercial immunoassays. Clin Chem Lab Med 2024; 62:1092-1100. [PMID: 38253403 DOI: 10.1515/cclm-2023-0821] [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: 08/01/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024]
Abstract
OBJECTIVES The standardization of cystatin C (CysC) measurement has received increasing attention in recent years due to its importance in estimating glomerular filtration rate (GFR). Mass spectrometry-based assays have the potential to provide an accuracy base for CysC measurement. However, a precise, accurate and sustainable LC-MS/MS method for CysC is still lacking. METHODS The developed LC-MS/MS method quantified CysC by detecting signature peptide (T3) obtained from tryptic digestion. Stable isotope labeled T3 peptide (SIL-T3) was spiked to control matrix effects and errors caused by liquid handling. The protein denaturation, reduction and alkylation procedures were combined into a single step with incubation time of 1 h, and the digestion lasted for 3.5 h. In the method validation, digestion time-course, imprecision, accuracy, matrix effect, interference, limit of quantification (LOQ), carryover, linearity, and the comparability to two routine immunoassays were evaluated. RESULTS No significant matrix effect or interference was observed with the CysC measurement. The LOQ was 0.21 mg/L; the within-run and total imprecision were 1.33-2.05 % and 2.18-3.90 % for three serum pools (1.18-5.34 mg/L). The LC-MS/MS method was calibrated by ERM-DA471/IFCC and showed good correlation with two immunoassays traceable to ERM-DA471/IFCC. However, significant bias was observed for immunoassays against the LC-MS/MS method. CONCLUSIONS The developed LC-MS/MS method is robust and simpler and holds the promise to provide an accuracy base for routine immunoassays, which will promote the standardization of CysC measurement.
Collapse
Affiliation(s)
- Li Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Xiaerbanu Nizhamuding
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Hao Zheng
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| | - Jie Zeng
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| | - Xinyi Yuan
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Zijia Ma
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Weiyan Zhou
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| | - Chao Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| | - Tianjiao Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| | - Chuanbao Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing, P.R. China
- National Center of Gerontology, Beijing Engineering Research Center of Laboratory Medicine, Beijing, P.R. China
| |
Collapse
|
35
|
Pouw N, van der Linden J, Teuben S, Kos S. Clinically Relevant Laboratory Monitoring of Gender-Affirming Hormone Therapy in Transgender People-Experiences from a Teaching Hospital in the Netherlands. J Appl Lab Med 2024; 9:440-455. [PMID: 38452266 DOI: 10.1093/jalm/jfae006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/14/2023] [Indexed: 03/09/2024]
Abstract
BACKGROUND Transgender care is shifting from academic to nonacademic settings leading to use of common (immunoassay) compared to sophisticated (mass spectrometry) methods to monitor estradiol and testosterone during gender-affirming hormone therapy (GAHT). The type of assay can influence results and have significant implications for clinical decision making. An evidence gap is present in recommendations regarding the assay needed to monitor GAHT. The present study aimed to summarize current evidence and evaluate immunoassay estradiol and testosterone concentrations in transgender people visiting a nonacademic hospital for GAHT. METHODS Clinical practice guidelines on GAHT and scientific literature on assay methodologies were screened and summarized. Laboratory and medical data from 252 patients who visited the transgender outpatient clinic of the Maasstad Hospital for GAHT between 2020 and 2022 were retrospectively analyzed. RESULTS Our research showed that the most used clinical practice guidelines for GAHT provide hormonal target values without recommending a preferred method. A comprehensive literature search on agreement between immunoassay and mass spectrometry showed substantial heterogeneity in results. Retrospective analysis of our immunoassay measured data in transgender people showed hormonal changes during GAHT that are to be expected from the medication used. CONCLUSIONS We demonstrate that laboratory monitoring of GAHT in a nonacademic hospital can be done safely by immunoassay in most cases. Only in cases where clinical observation is discordant with the hormonal results do more sophisticated methods need to be deployed. A best practice model was proposed for transgender care in nonacademic hospitals.
Collapse
Affiliation(s)
- Nadine Pouw
- Laboratory of Clinical Chemistry, MaasstadLab, Maasstad Hospital, Rotterdam, the Netherlands
| | - Joke van der Linden
- Department of Internal Medicine, Maasstad Hospital, Rotterdam, the Netherlands
| | - Sonja Teuben
- Department of Internal Medicine, Maasstad Hospital, Rotterdam, the Netherlands
| | - Snježana Kos
- Laboratory of Clinical Chemistry, MaasstadLab, Maasstad Hospital, Rotterdam, the Netherlands
| |
Collapse
|
36
|
Zheng X, Yang N, Mao R, Hao Y, Teng D, Wang J. Pharmacokinetics and pharmacodynamics of antibacterial peptide NZX in Staphylococcus aureus mastitis mouse model. Appl Microbiol Biotechnol 2024; 108:260. [PMID: 38472422 PMCID: PMC11636681 DOI: 10.1007/s00253-024-13101-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024]
Abstract
Staphylococcus aureus is associated with dairy mastitis, which causes serious economic losses to dairy farming industry. Antibacterial peptide NZX showed good antibacterial activity against S. aureus. This study aimed to evaluate pharmacokinetics and pharmacodynamics of NZX against S. aureus-induced mouse mastitis. NZX exhibited potent in vitro antibacterial activity against the test S. aureus strains (minimal inhibitory concentration (MIC): 0.23-0.46 μM), low mutant prevention concentration (MPC: 1.18-3.68 μM), and a long post antibiotic effect (PAE: 2.20-8.84 h), which was superior to those of lincomycin and ceftiofur. Antibacterial mechanisms showed that NZX could penetrate the cell membrane, resulting in obvious cell membrane perforation and morphological changes, and bind to intracellular DNA. Furthermore, NZX had a good stability in milk environment (retention rate: 85.36%, 24 h) than that in mammary homogenate (47.90%, 24 h). In mouse mastitis model, NZX (25-400 μg/gland) could significantly reduce the bacterial load of mammary tissue in a dose-dependent manner. In addition, NZX (100 μg/gland) could relieve the inflammatory symptoms of mammary tissue, and significantly decreased its pathological scores. The concentration-time curve of NZX (100 μg/gland) in the mammary tissue was plotted and the corresponding pharmacokinetic parameters were obtained by non-compartment model calculation. Those parameters of Tmax, T1/2, Cmax and AUC were 0.5 h, 35.11 h, 32.49 μg/g and 391 μg·h/g, respectively. Therefore, these results suggest that NZX could act as a promising candidate for treating dairy mastitis disease caused by S. aureus. KEY POINTS: • NZX could kill S. aureus by dual mechanism involved in membrane and DNA disruption • NZX could relieve S. aureus-induced mouse mastitis • Pharmacokinetic parameters of NZX in mouse mammary gland were obtained.
Collapse
Affiliation(s)
- Xueling Zheng
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, People's Republic of China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China
| | - Na Yang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, People's Republic of China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China
| | - Ruoyu Mao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, People's Republic of China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China
| | - Ya Hao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, People's Republic of China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China
| | - Da Teng
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, People's Republic of China.
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China.
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China.
| | - Jianhua Wang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, People's Republic of China.
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China.
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China.
| |
Collapse
|
37
|
Reijnders E, van der Laarse A, Ruhaak LR, Cobbaert CM. Closing the gaps in patient management of dyslipidemia: stepping into cardiovascular precision diagnostics with apolipoprotein profiling. Clin Proteomics 2024; 21:19. [PMID: 38429638 PMCID: PMC10908091 DOI: 10.1186/s12014-024-09465-w] [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: 09/18/2023] [Accepted: 02/14/2024] [Indexed: 03/03/2024] Open
Abstract
In persons with dyslipidemia, a high residual risk of cardiovascular disease remains despite lipid lowering therapy. Current cardiovascular risk prediction mainly focuses on low-density lipoprotein cholesterol (LDL-c) levels, neglecting other contributing risk factors. Moreover, the efficacy of LDL-c lowering by statins resulting in reduced cardiovascular risk is only partially effective. Secondly, from a metrological viewpoint LDL-c falls short as a reliable measurand. Both direct and calculated LDL-c tests produce inaccurate test results at the low end under aggressive lipid lowering therapy. As LDL-c tests underperform both clinically and metrologically, there is an urging need for molecularly defined biomarkers. Over the years, apolipoproteins have emerged as promising biomarkers in the context of cardiovascular disease as they are the functional workhorses in lipid metabolism. Among these, apolipoprotein B (ApoB), present on all atherogenic lipoprotein particles, has demonstrated to clinically outperform LDL-c. Other apolipoproteins, such as Apo(a) - the characteristic apolipoprotein of the emerging risk factor lipoprotein(a) -, and ApoC-III - an inhibitor of triglyceride-rich lipoprotein clearance -, have attracted attention as well. To support personalized medicine, we need to move to molecularly defined risk markers, like the apolipoproteins. Molecularly defined diagnosis and molecularly targeted therapy require molecularly measured biomarkers. This review provides a summary of the scientific validity and (patho)physiological role of nine serum apolipoproteins, Apo(a), ApoB, ApoC-I, ApoC-II, ApoC-III, ApoE and its phenotypes, ApoA-I, ApoA-II, and ApoA-IV, in lipid metabolism, their association with cardiovascular disease, and their potential as cardiovascular risk markers when measured in a multiplex apolipoprotein panel.
Collapse
Affiliation(s)
- Esther Reijnders
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| | - Arnoud van der Laarse
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - L Renee Ruhaak
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
38
|
Zheng P, Raj P, Wu L, Mizutani T, Szabo M, Hanson WA, Barman I. Quantitative Detection of Thyroid-Stimulating Hormone in Patient Samples with a Nanomechanical Single-Antibody Spectro-Immunoassay. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305110. [PMID: 37752776 PMCID: PMC10922205 DOI: 10.1002/smll.202305110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 09/12/2023] [Indexed: 09/28/2023]
Abstract
Functional disorders of the thyroid remain a global challenge and have profound impacts on human health. Serving as the barometer for thyroid function, thyroid-stimulating hormone (TSH) is considered the single most useful test of thyroid function. However, the prevailing TSH immunoassays rely on two types of antibodies in a sandwich format. The requirement of repeated incubation and washing further complicates the issue, making it unable to meet the requirements of the shifting public health landscape that demands rapid, sensitive, and low-cost TSH tests. Herein, a systematic study is performed to investigate the clinical translational potential of a single antibody-based biosensing platform for the TSH test. The biosensing platform leverages Raman spectral variations induced by the interaction between a TSH antigen and a Raman molecule-conjugated TSH antibody. In conjunction with machine learning, it allows TSH concentrations in various patient samples to be predicted with high accuracy and precision, which is robust against substrate-to-substrate, intra-substrate, and day-to-day variations. It is envisioned that the simplicity and generalizability of this single-antibody immunoassay coupled with the demonstrated performance in patient samples pave the way for it to be widely applied in clinical settings for low-cost detection of hormones, other molecular biomarkers, DNA, RNA, and pathogens.
Collapse
Affiliation(s)
- Peng Zheng
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Piyush Raj
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Lintong Wu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Takayuki Mizutani
- Beckman Coulter Diagnostics – Immunoassay Business Unit, 1000 Lake Hazeltine Dr, Chaska, MN 55318
| | - Miklos Szabo
- Beckman Coulter Diagnostics – Immunoassay Business Unit, 1000 Lake Hazeltine Dr, Chaska, MN 55318
| | - William A. Hanson
- Beckman Coulter Diagnostics – Immunoassay Business Unit, 1000 Lake Hazeltine Dr, Chaska, MN 55318
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, United States
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| |
Collapse
|
39
|
Wenk D, Zuo C, Kislinger T, Sepiashvili L. Recent developments in mass-spectrometry-based targeted proteomics of clinical cancer biomarkers. Clin Proteomics 2024; 21:6. [PMID: 38287260 PMCID: PMC10826105 DOI: 10.1186/s12014-024-09452-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/14/2024] [Indexed: 01/31/2024] Open
Abstract
Routine measurement of cancer biomarkers is performed for early detection, risk classification, and treatment monitoring, among other applications, and has substantially contributed to better clinical outcomes for patients. However, there remains an unmet need for clinically validated assays of cancer protein biomarkers. Protein tumor markers are of particular interest since proteins carry out the majority of biological processes and thus dynamically reflect changes in cancer pathophysiology. Mass spectrometry-based targeted proteomics is a powerful tool for absolute peptide and protein quantification in biological matrices with numerous advantages that make it attractive for clinical applications in oncology. The use of liquid chromatography-tandem mass spectrometry (LC-MS/MS) based methodologies has allowed laboratories to overcome challenges associated with immunoassays that are more widely used for tumor marker measurements. Yet, clinical implementation of targeted proteomics methodologies has so far been limited to a few cancer markers. This is due to numerous challenges associated with paucity of robust validation studies of new biomarkers and the labor-intensive and operationally complex nature of LC-MS/MS workflows. The purpose of this review is to provide an overview of targeted proteomics applications in cancer, workflows used in targeted proteomics, and requirements for clinical validation and implementation of targeted proteomics assays. We will also discuss advantages and challenges of targeted MS-based proteomics assays for clinical cancer biomarker analysis and highlight some recent developments that will positively contribute to the implementation of this technique into clinical laboratories.
Collapse
Affiliation(s)
- Deborah Wenk
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Charlotte Zuo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
- Princess Margaret Cancer Research Tower, Room 9-807, 101 College Street, Toronto, ON, M5G 1L7, Canada.
| | - Lusia Sepiashvili
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, 555 University Ave, Rm 3606, Toronto, ON, M5G 1X8, Canada.
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada.
- Sickkids Research Institute, Toronto, ON, Canada.
| |
Collapse
|
40
|
Diederiks NM, Ruhaak LR, Romijn FPHTM, Pieterse MM, Smit NPM, Cobbaert CM. An LC-MS-based designated comparison method with similar performance to the Lp(a) reference measurement procedure to guide molar Lp(a) standardization. Clin Proteomics 2024; 21:5. [PMID: 38267848 PMCID: PMC10809433 DOI: 10.1186/s12014-023-09446-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/07/2023] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND The 2022 consensus statement of the European Atherosclerosis Society (EAS) on lipoprotein(a) (Lp(a)) recognizes the role of Lp(a) as a relevant genetically determined risk factor and recommends its measurement at least once in an individual's lifetime. It also strongly urges that Lp(a) test results are expressed as apolipoprotein (a) (apo(a)) amount of substance in molar units and no longer in confounded Lp(a) mass units (mg/dL or mg/L). Therefore, IVD manufacturers should transition to molar units. A prerequisite for this transition is the availability of an Lp(a) Reference Measurement Procedure (RMP) that allows unequivocal molecular detection and quantification of apo(a) in Lp(a). To that end an ISO 17511:2020 compliant LC-MS based and IFCC-endorsed RMP has been established that targets proteotypic peptides of apolipoprotein(a) (apo(a)) in Lp(a). The RMP is laborious and requires highly skilled operators. To guide IVD-manufacturers of immunoassay-based Lp(a) test kits in the transition from mass to molar units, a Designated Comparison Method (DCM) has been developed and evaluated. METHODS To assess whether the DCM provides equivalent results compared to the RMP, the procedural designs were compared and the analytical performance of DCM and RMP were first evaluated in a head-to-head comparison. Subsequently, apo(a) was quantified in 153 human clinical serum samples. Both DCM and RMP were calibrated using external native calibrators that produce results traceable to SRM2B. Measurement uncertainty (MU) was checked against predefined allowable MU. RESULTS The major difference in the design of the DCM for apo(a) is the use of only one enzymatic digestion step. The analytical performance of the DCM and RMP for apo(a) is highly similar. In a direct method comparison, equivalent results were obtained with a median regression slope 0.997 of and a median bias of - 0.2 nmol/L (- 0.2%); the intermediate imprecision of the test results was within total allowable error (TEa) (CVa of 10.2% at 90 nmol/L). CONCLUSIONS The semi-automated, higher throughput, LC-MS-based method for Lp(a) meets the predefined analytical performance specifications and allowable MU and is hence applicable as a higher order Designated Comparison Method, which is ideally suited to guide IVD manufacturers in the transition from Lp(a) mass to molar units.
Collapse
Affiliation(s)
- Nina M Diederiks
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Postzone E2-P, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - L Renee Ruhaak
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Postzone E2-P, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Fred P H T M Romijn
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Postzone E2-P, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Mervin M Pieterse
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Postzone E2-P, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Nico P M Smit
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Postzone E2-P, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Postzone E2-P, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
41
|
Purohit K, Reddy N, Sunna A. Exploring the Potential of Bioactive Peptides: From Natural Sources to Therapeutics. Int J Mol Sci 2024; 25:1391. [PMID: 38338676 PMCID: PMC10855437 DOI: 10.3390/ijms25031391] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/18/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
Bioactive peptides, specific protein fragments with positive health effects, are gaining traction in drug development for advantages like enhanced penetration, low toxicity, and rapid clearance. This comprehensive review navigates the intricate landscape of peptide science, covering discovery to functional characterization. Beginning with a peptidomic exploration of natural sources, the review emphasizes the search for novel peptides. Extraction approaches, including enzymatic hydrolysis, microbial fermentation, and specialized methods for disulfide-linked peptides, are extensively covered. Mass spectrometric analysis techniques for data acquisition and identification, such as liquid chromatography, capillary electrophoresis, untargeted peptide analysis, and bioinformatics, are thoroughly outlined. The exploration of peptide bioactivity incorporates various methodologies, from in vitro assays to in silico techniques, including advanced approaches like phage display and cell-based assays. The review also discusses the structure-activity relationship in the context of antimicrobial peptides (AMPs), ACE-inhibitory peptides (ACEs), and antioxidative peptides (AOPs). Concluding with key findings and future research directions, this interdisciplinary review serves as a comprehensive reference, offering a holistic understanding of peptides and their potential therapeutic applications.
Collapse
Affiliation(s)
- Kruttika Purohit
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia;
- Australian Research Council Industrial Transformation Training Centre for Facilitated Advancement of Australia’s Bioactives (FAAB), Sydney, NSW 2109, Australia;
| | - Narsimha Reddy
- Australian Research Council Industrial Transformation Training Centre for Facilitated Advancement of Australia’s Bioactives (FAAB), Sydney, NSW 2109, Australia;
- School of Science, Parramatta Campus, Western Sydney University, Penrith, NSW 2751, Australia
| | - Anwar Sunna
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia;
- Australian Research Council Industrial Transformation Training Centre for Facilitated Advancement of Australia’s Bioactives (FAAB), Sydney, NSW 2109, Australia;
- Biomolecular Discovery Research Centre, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
42
|
Slusher GA, Kottke PA, Culberson AL, Chilmonczyk MA, Fedorov AG. Microfluidics enabled multi-omics triple-shot mass spectrometry for cell-based therapies. BIOMICROFLUIDICS 2024; 18:011302. [PMID: 38268742 PMCID: PMC10807926 DOI: 10.1063/5.0175178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/01/2024] [Indexed: 01/26/2024]
Abstract
In recent years, cell-based therapies have transformed medical treatment. These therapies present a multitude of challenges associated with identifying the mechanism of action, developing accurate safety and potency assays, and achieving low-cost product manufacturing at scale. The complexity of the problem can be attributed to the intricate composition of the therapeutic products: living cells with complex biochemical compositions. Identifying and measuring critical quality attributes (CQAs) that impact therapy success is crucial for both the therapy development and its manufacturing. Unfortunately, current analytical methods and tools for identifying and measuring CQAs are limited in both scope and speed. This Perspective explores the potential for microfluidic-enabled mass spectrometry (MS) systems to comprehensively characterize CQAs for cell-based therapies, focusing on secretome, intracellular metabolome, and surfaceome biomarkers. Powerful microfluidic sampling and processing platforms have been recently presented for the secretome and intracellular metabolome, which could be implemented with MS for fast, locally sampled screening of the cell culture. However, surfaceome analysis remains limited by the lack of rapid isolation and enrichment methods. Developing innovative microfluidic approaches for surface marker analysis and integrating them with secretome and metabolome measurements using a common analytical platform hold the promise of enhancing our understanding of CQAs across all "omes," potentially revolutionizing cell-based therapy development and manufacturing for improved efficacy and patient accessibility.
Collapse
Affiliation(s)
| | - Peter A. Kottke
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30318, USA
| | | | | | | |
Collapse
|
43
|
Lam KB, Song L, Buggs V, Vithlani G, Metushi IG. A case of false positive opiate immunoassay results from rifampin (rifampicin) treatment. Pract Lab Med 2023; 37:e00334. [PMID: 37711668 PMCID: PMC10498289 DOI: 10.1016/j.plabm.2023.e00334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/21/2023] [Accepted: 08/28/2023] [Indexed: 09/16/2023] Open
Abstract
The drug screen test on a 12-year-old male patient was positive for opiates by a kinetic interaction of microparticles in solution (KIMS) immunoassay method on the Roche Cobas C502. The positive opiates result was not confirmed by the liquid chromatography-tandem mass spectrometry (LC-MS/MS) method. A chart review revealed that the patient had tuberculosis and was on rifampin. We spiked rifampin into drug-free urine and tested opiates with the Cobas method. Once again, a positive result was obtained. This case showed that rifampin can still cause false positive opiate results measured with the KIMS method. We want to stress the importance of confirming positive screen results by more specific methods such as LC-MS/MS.
Collapse
Affiliation(s)
- K.H. Brian Lam
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, USA
| | - Lu Song
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, USA
| | - Vincent Buggs
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, USA
| | - Gopal Vithlani
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, USA
| | - Imir G. Metushi
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, USA
| |
Collapse
|
44
|
Diederiks NM, van der Burgt YEM, Ruhaak LR, Cobbaert CM. Developing an SI-traceable Lp(a) reference measurement system: a pilgrimage to selective and accurate apo(a) quantification. Crit Rev Clin Lab Sci 2023; 60:483-501. [PMID: 37128734 DOI: 10.1080/10408363.2023.2199353] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/14/2023] [Accepted: 04/01/2023] [Indexed: 05/03/2023]
Abstract
In the past decade a remarkable rebirth of serum/plasma lipoprotein(a) (Lp(a)) as an independent risk factor of cardiovascular disease (CVD) occurred. Updated evidence for a causal continuous association in different ethnic groups between Lp(a) concentrations and cardiovascular outcomes has been published in the latest European Atherosclerosis Society (EAS) Lp(a) consensus statement. Interest in measuring Lp(a) at least once in a person's lifetime moreover originates from the development of promising new Lp(a) lowering drugs. Accurate and clinically effective Lp(a) tests are of key importance for the timely detection of high-risk individuals and for future evaluation of the therapeutic effects of Lp(a) lowering medication. To this end, it is necessary to improve the performance and standardization of existing Lp(a) tests, as is also noted in the Lp(a) consensus statement. Consequently, a state-of-the-art internationally endorsed reference measurement system (RMS) must be in place that allows for performance evaluation of Lp(a) field tests in order to certify their validity and accuracy. An ELISA-based RMS from Northwest Lipid Research Laboratory (University of Washington, Seattle, USA) has been available since the 1990s. A next-generation apo(a)/Lp(a) RMS is now being developed by a working group from the International Federation of Clinical Chemistry and Laboratory Medicine (IFCC). The envisioned apo(a) RMS is based on the direct measurement of selected proteotypic fragments generated after proteolytic digestion using quantitative protein mass spectrometry (MS). The choice for an MS-based RMS enables selective measurement of the proteotypic peptides and is by design apo(a) isoform insensitive. Clearly, the equimolar conversion of apo(a) into the surrogate peptide measurands is required to obtain accurate Lp(a) results. The completeness of proteolysis under reaction conditions from the candidate reference measurement procedure (RMP) has been demonstrated for the quantifying apo(a) peptides. Currently, the candidate apo(a) RMP is endorsed by the IFCC and recommendations for suitable secondary reference materials have been made in a recent commutability study paper. Ongoing efforts toward a complete apo(a) RMS that is listed by the Joint Committee on Traceability in Laboratory Medicine (JCTLM) are focused on the peptide-based calibration and the establishment of a network of calibration laboratories running the apo(a) RMS in a harmonized way. Once completed, it will be the holy grail for evaluation and certification of Lp(a) field methods.
Collapse
Affiliation(s)
- Nina M Diederiks
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, ZA, The Netherlands
| | - Yuri E M van der Burgt
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, ZA, The Netherlands
| | - L Renee Ruhaak
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, ZA, The Netherlands
| | - Christa M Cobbaert
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, Leiden, ZA, The Netherlands
| |
Collapse
|
45
|
Chhun S, Trauchessec M, Melicine S, Nicolas F, Miele A, Lukic S, Vilain E, Amrouche L, Lebert D, Anglicheau D, Tartour E, Zuber J. A Validated LC-MS/MS Method for Performing Belatacept Drug Monitoring in Renal Transplantation. Biomedicines 2023; 11:2955. [PMID: 38001955 PMCID: PMC10669563 DOI: 10.3390/biomedicines11112955] [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: 09/22/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
Belatacept, a CTLA4-Ig, was designed to prevent rejection and graft loss in kidney transplant recipients. This immunotherapy showed a long-term clinical benefit mainly on renal function and better glycemic control but was also associated with a higher number of severe infectious diseases, particularly CMV disease, and lymphoproliferative disease. Therapeutic drug monitoring usually guides the benefit-risk assessment of long-term immunosuppression. In this study, an analytical method by LC-MS/MS was developed in 20 microL of plasma for the belatacept quantification. Intra- and inter-assay precision and accuracy were lower than 20% for the limit of quantification, and 15% for higher concentrations. The method was implemented in our lab and provided data about the inter-variability (N = 108) and intra-variability (N = 33) of belatacept concentrations in kidney transplant recipients with a stable renal function, after conversion from a CNI- to a belatacept-based regimen.
Collapse
Affiliation(s)
- Stéphanie Chhun
- Laboratory of Immunology, Georges Pompidou European Hospital and Necker Hospital, AP-HP, F-75015 Paris, France (E.T.)
- Faculty of Medecine, Université Paris-Cité, F-75006 Paris, France; (D.A.); (J.Z.)
| | | | - Sophie Melicine
- Laboratory of Immunology, Georges Pompidou European Hospital and Necker Hospital, AP-HP, F-75015 Paris, France (E.T.)
| | - Frédéric Nicolas
- Department of Kidney and Metabolic Diseases, Transplantation and Clinical Immunology, Necker Hospital, AP-HP, F-75015 Paris, France (L.A.)
| | - Agathe Miele
- Promise Proteomics, F-38040 Grenoble, France (D.L.)
| | - Srboljub Lukic
- Laboratory of Immunology, Georges Pompidou European Hospital and Necker Hospital, AP-HP, F-75015 Paris, France (E.T.)
| | - Estelle Vilain
- Department of Kidney and Metabolic Diseases, Transplantation and Clinical Immunology, Necker Hospital, AP-HP, F-75015 Paris, France (L.A.)
| | - Lucile Amrouche
- Department of Kidney and Metabolic Diseases, Transplantation and Clinical Immunology, Necker Hospital, AP-HP, F-75015 Paris, France (L.A.)
| | | | - Dany Anglicheau
- Faculty of Medecine, Université Paris-Cité, F-75006 Paris, France; (D.A.); (J.Z.)
- Department of Kidney and Metabolic Diseases, Transplantation and Clinical Immunology, Necker Hospital, AP-HP, F-75015 Paris, France (L.A.)
| | - Eric Tartour
- Laboratory of Immunology, Georges Pompidou European Hospital and Necker Hospital, AP-HP, F-75015 Paris, France (E.T.)
- Faculty of Medecine, Université Paris-Cité, F-75006 Paris, France; (D.A.); (J.Z.)
| | - Julien Zuber
- Faculty of Medecine, Université Paris-Cité, F-75006 Paris, France; (D.A.); (J.Z.)
- Department of Kidney and Metabolic Diseases, Transplantation and Clinical Immunology, Necker Hospital, AP-HP, F-75015 Paris, France (L.A.)
| |
Collapse
|
46
|
Tölke SA, Masetto T, Reuschel T, Grimmler M, Bindila L, Schneider K. Immunoaffinity LC-MS/MS Quantification of the Sepsis Biomarker Procalcitonin Using Magnetic- and Polystyrene-Bead Immobilized Polyclonal Antibodies. J Proteome Res 2023; 22:3135-3148. [PMID: 37672672 DOI: 10.1021/acs.jproteome.3c00082] [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: 09/08/2023]
Abstract
Procalcitonin (PCT) is a biomarker for bacterial sepsis, and accurate quantification of PCT is critical for sepsis diagnosis and treatment. Immunological PCT quantification methods are routinely used in clinical laboratories, yet there is a need for harmonization of PCT quantification protocols. An orthogonal method to clinical immunological assays, such as LC-MS/MS, is required. In this study, a highly sensitive and robust immunoaffinity LC-MRM quantitative method for detecting procalcitonin in human serum has been developed. An initial comparison of immunocapture of PCT with a polyclonal anti-PCT antibody immobilized on polystyrene nanoparticles (Latex) and magnetic beads demonstrated superior performance with magnetic beads. Three tryptic PCT peptides from the N- and C-terminal regions of PCT were selected for LC-MS/MS quantification. For PCT quantification, an LLOQ of 0.25 ng/mL of PCT in human serum was achieved using a sample volume of 1 mL. The method's trueness and precision consistently lie within the 15% margin. The parallel measurement of three PCT peptides may allow future differentiation of intact PCT vs other PCT forms originating from potential degradation, processing, or polymorphisms. An established and validated LC-MRM-based quantification of PCT will be relevant as an orthogonal method for harmonization and standardization of clinical assays for PCT.
Collapse
Affiliation(s)
- Sebastian-Alexander Tölke
- Institute for Biomolecular Research, Hochschule Fresenius, University of Applied Sciences, Limburger Straße 2, 65510 Idstein, Germany
- Clinical Lipidomics Unit, Institute of Physiological Chemistry, University Medical Center, 55131 Mainz, Germany
| | - Thomas Masetto
- Institute of Molecular Medicine I, Medical Faculty,, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
- DiaSys Diagnostic Systems GmbH, Alte Straße 9, 65558 Holzheim, Germany
| | - Thomas Reuschel
- Institute for Biomolecular Research, Hochschule Fresenius, University of Applied Sciences, Limburger Straße 2, 65510 Idstein, Germany
| | - Matthias Grimmler
- Institute for Biomolecular Research, Hochschule Fresenius, University of Applied Sciences, Limburger Straße 2, 65510 Idstein, Germany
- DiaSys Diagnostic Systems GmbH, Alte Straße 9, 65558 Holzheim, Germany
- DiaServe Laboratories GmbH, Seeshaupter Straße 27, 82393 Iffeldorf, Germany
| | - Laura Bindila
- Clinical Lipidomics Unit, Institute of Physiological Chemistry, University Medical Center, 55131 Mainz, Germany
| | - Klaus Schneider
- Institute for Biomolecular Research, Hochschule Fresenius, University of Applied Sciences, Limburger Straße 2, 65510 Idstein, Germany
| |
Collapse
|
47
|
Sleumer B, Kema IP, van de Merbel NC. Quantitative bioanalysis of proteins by digestion and LC-MS/MS: the use of multiple signature peptides. Bioanalysis 2023; 15:1203-1216. [PMID: 37724471 DOI: 10.4155/bio-2023-0129] [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: 09/20/2023] Open
Abstract
The use of multiple signature peptides for the quantification of proteins by digestion and LC-MS/MS is reviewed and evaluated here. A distinction is made based on the purpose of the use of multiple peptides: confirmation of the protein concentration, discrimination between different protein forms or species and in vivo biotransformation. Most reports that describe methods with at least two peptides use these for confirmation, but it is not always mentioned how the peptides are used and how possible differences in concentration between the peptides are handled. Differences in concentration are often reported in the case of monitoring different protein forms or in vivo biotransformation, and this offers insight into the biological fate of the protein.
Collapse
Affiliation(s)
- Bas Sleumer
- ICON Bioanalytical Laboratories, Amerikaweg 18, 9407 TK, Assen, The Netherlands
- Department of Analytical Biochemistry, University of Groningen, A Deusinglaan 1, 9700 AV Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, EA61, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, EA61, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Nico C van de Merbel
- ICON Bioanalytical Laboratories, Amerikaweg 18, 9407 TK, Assen, The Netherlands
- Department of Analytical Biochemistry, University of Groningen, A Deusinglaan 1, 9700 AV Groningen, The Netherlands
| |
Collapse
|
48
|
Agostini M, Traldi P, Hamdan M. Mass Spectrometry-Based Proteomics: Analyses Related to Drug-Resistance and Disease Biomarkers. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1722. [PMID: 37893440 PMCID: PMC10608342 DOI: 10.3390/medicina59101722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023]
Abstract
Mass spectrometry-based proteomics is a key player in research efforts to characterize aberrant epigenetic alterations, including histone post-translational modifications and DNA methylation. Data generated by this approach complements and enrich datasets generated by genomic, epigenetic and transcriptomics approaches. These combined datasets can provide much-needed information on various mechanisms responsible for drug resistance, the discovery and validation of potential biomarkers for different diseases, the identification of signaling pathways, and genes and enzymes to be targeted by future therapies. The increasing use of high-resolution, high-accuracy mass spectrometers combined with more refined protein labeling and enrichment procedures enhanced the role of this approach in the investigation of these epigenetic modifications. In this review, we discuss recent MS-based studies, which are contributing to current research efforts to understand certain mechanisms behind drug resistance to therapy. We also discuss how these MS-based analyses are contributing to biomarkers discovery and validation.
Collapse
Affiliation(s)
| | - Pietro Traldi
- Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti 4, 35100 Padova, Italy; (M.A.); (M.H.)
| | | |
Collapse
|
49
|
Abdelkader Y, Perez-Davalos L, LeDuc R, Zahedi RP, Labouta HI. Omics approaches for the assessment of biological responses to nanoparticles. Adv Drug Deliv Rev 2023; 200:114992. [PMID: 37414362 DOI: 10.1016/j.addr.2023.114992] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/08/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Nanotechnology has enabled the development of innovative therapeutics, diagnostics, and drug delivery systems. Nanoparticles (NPs) can influence gene expression, protein synthesis, cell cycle, metabolism, and other subcellular processes. While conventional methods have limitations in characterizing responses to NPs, omics approaches can analyze complete sets of molecular entities that change upon exposure to NPs. This review discusses key omics approaches, namely transcriptomics, proteomics, metabolomics, lipidomics and multi-omics, applied to the assessment of biological responses to NPs. Fundamental concepts and analytical methods used for each approach are presented, as well as good practices for omics experiments. Bioinformatics tools are essential to analyze, interpret and visualize large omics data, and to correlate observations in different molecular layers. The authors envision that conducting interdisciplinary multi-omics analyses in future nanomedicine studies will reveal integrated cell responses to NPs at different omics levels, and the incorporation of omics into the evaluation of targeted delivery, efficacy, and safety will improve the development of nanomedicine therapies.
Collapse
Affiliation(s)
- Yasmin Abdelkader
- Unity Health Toronto - St. Michael's Hospital, University of Toronto, 209 Victoria St., Toronto, Ontario M5B 1T8, Canada; College of Pharmacy, Apotex Centre, University of Manitoba, 750 McDermot Av. W, Winnipeg, Manitoba R3E 0T5, Canada; Department of Cell Biology, Biotechnology Research Institute, National Research Centre, 33 El Buhouth St., Cairo 12622, Egypt
| | - Luis Perez-Davalos
- Unity Health Toronto - St. Michael's Hospital, University of Toronto, 209 Victoria St., Toronto, Ontario M5B 1T8, Canada; College of Pharmacy, Apotex Centre, University of Manitoba, 750 McDermot Av. W, Winnipeg, Manitoba R3E 0T5, Canada
| | - Richard LeDuc
- Children's Hospital Research Institute of Manitoba, 513 - 715 McDermot Av. W, Winnipeg, Manitoba R3E 3P4, Canada; Department of Biochemistry and Medical Genetics, University of Manitoba, 745 Bannatyne Av., Winnipeg, Manitoba R3E 0J9, Canada
| | - Rene P Zahedi
- Department of Biochemistry and Medical Genetics, University of Manitoba, 745 Bannatyne Av., Winnipeg, Manitoba R3E 0J9, Canada; Department of Internal Medicine, 715 McDermot Av., Winnipeg, Manitoba R3E 3P4, Canada; Manitoba Centre for Proteomics and Systems Biology, 799 JBRC, 715 McDermot Av., Winnipeg, Manitoba R3E 3P4, Canada; CancerCare Manitoba Research Institute, 675 McDermot Av., Manitoba R3E 0V9, Canada
| | - Hagar I Labouta
- Unity Health Toronto - St. Michael's Hospital, University of Toronto, 209 Victoria St., Toronto, Ontario M5B 1T8, Canada; College of Pharmacy, Apotex Centre, University of Manitoba, 750 McDermot Av. W, Winnipeg, Manitoba R3E 0T5, Canada; Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, Ontario M5S 3M2, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, M5S 3G9, Canada; Faculty of Pharmacy, Alexandria University, 1 Khartoum Square, Azarita, Alexandria, Egypt, 21521.
| |
Collapse
|
50
|
Sleumer B, Zwerwer J, van Faassen M, Vos MJ, Bischoff R, Kema IP, van de Merbel NC. An antibody-free LC-MS/MS method for the quantification of sex hormone binding globulin in human serum and plasma. Clin Chem Lab Med 2023; 61:1266-1274. [PMID: 36773321 DOI: 10.1515/cclm-2022-1225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/26/2023] [Indexed: 02/13/2023]
Abstract
OBJECTIVES Sex hormone binding globulin (SHBG) is a hormone binding protein which plays an important role in regulating the transport and availability of biologically active androgens and estradiol to target cells and used to calculate free testosterone concentrations. METHODS A liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed, featuring an albumin removal step followed by a tryptic digestion. After a reduction step with dithiothreitol and alkylation with iodoacetamide three signature peptides were used for the quantification of SHBG. RESULTS The method enables the quantification of serum and plasma SHBG over the clinically relevant range of 200-20,000 ng/mL and was validated according to the most recent guidelines. The LC-MS/MS method correlates well with the Abbott Alinity immunoassay (R2>0.95), but the LC-MS/MS results are on average 16-17% lower than the immunoassay results, which is consistent for all three signature peptides. CONCLUSIONS The LC-MS/MS method which includes an albumin depletion step allows quantification of SHBG in serum and plasma without an immunocapture step at clinically relevant SHBG levels, thus contributing to better lab-to-lab consistency of results.
Collapse
Affiliation(s)
- Bas Sleumer
- ICON Bioanalytical Laboratories, Assen, The Netherlands
- Department of Analytical Biochemistry, University of Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jordan Zwerwer
- ICON Bioanalytical Laboratories, Assen, The Netherlands
- Department of Analytical Biochemistry, University of Groningen, Groningen, The Netherlands
| | - Martijn van Faassen
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Michel J Vos
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rainer Bischoff
- Department of Analytical Biochemistry, University of Groningen, Groningen, The Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Nico C van de Merbel
- ICON Bioanalytical Laboratories, Assen, The Netherlands
- Department of Analytical Biochemistry, University of Groningen, Groningen, The Netherlands
| |
Collapse
|