30 results on '"John F Kellie"'
Search Results
2. Top-Down Characterization and Intact Mass Quantitation of a Monoclonal Antibody Drug from Serum by Use of a Quadrupole TOF MS System Equipped with Electron-Activated Dissociation
- Author
-
John F. Kellie, Nicole A. Schneck, Jason C. Causon, Takashi Baba, John T. Mehl, and Kerstin I. Pohl
- Subjects
Structural Biology ,Spectroscopy - Abstract
Time-of-flight MS systems for biopharmaceutical and protein characterization applications may play an even more pivotal role in the future as biotherapeutics increase in drug pipelines and as top-down MS approaches increase in use. Here, a recently developed TOF MS system is examined for monoclonal antibody (mAb) characterization from serum samples. After immunocapture, purified drug material spiked into monkey serum or dosed for an in-life study is analyzed by top-down MS. While characterization aspects are a distinct advantage of the MS platform, MS system and software capabilities are also shown regarding intact protein quantitation. Such applications are demonstrated to help enable comprehensive protein molecule quantitation and characterization by use of TOF MS instrumentation.
- Published
- 2022
- Full Text
- View/download PDF
3. 2021 White Paper on Recent Issues in Bioanalysis: Mass Spec of Proteins, Extracellular Vesicles, CRISPR, Chiral Assays, Oligos; Nanomedicines Bioanalysis; ICH M10 Section 7.1; Non-Liquid & Rare Matrices; Regulatory Inputs (<u>Part 1A</u> – Recommendations on Endogenous Compounds, Small Molecules, Complex Methods, Regulated Mass Spec of Large Molecules, Small Molecule, PoC & <u>Part 1B</u> - Regulatory Agencies' Inputs on Bioanalysis, Biomarkers, Immunogenicity, Gene & Cell Therapy and Vaccine)
- Author
-
Surinder Kaur, Stephen C Alley, Matt Szapacs, Amanda Wilson, Eugene Ciccimaro, Dian Su, Neil Henderson, Linzhi Chen, Fabio Garofolo, Shawna Hengel, Wenying Jian, John F Kellie, Anita Lee, John Mehl, Joe Palandra, Haibo Qiu, Natasha Savoie, Diaa Shakleya, Ludovicus Staelens, Hiroshi Sugimoto, Giane Sumner, Jan Welink, Robert Wheller, Y-J Xue, Jianing Zeng, Jinhui Zhang, Huiyu Zhou, Jian Wang, Scott Summerfield, Olga Kavetska, Lieve Dillen, Ragu Ramanathan, Mike Baratta, Arindam Dasgupta, Anna Edmison, Luca Ferrari, Sally Fischer, Daniela Fraier, Sam Haidar, Kathrin Heermeier, Christopher James, Allena Ji, Lina Luo, Gustavo Mendes Lima Santos, Noah Post, Anton I Rosenbaum, Sune Sporring, Sekhar Surapaneni, Stephen Vinter, Katty Wan, Eric Woolf, Seongeun (Julia) Cho, Elham Kossary, Sandra Prior, Mohsen Rajabi Abhari, Catherine Soo, Yow-Ming Wang, Abbas Bandukwala, Elana Cherry, Isabelle Cludts, Soma Ghosh, Shirley Hopper, Akiko Ishii-Watabe, Susan Kirshner, Kevin Maher, Kimberly Maxfield, Joao Pedras-Vasconcelos, Yoshiro Saito, Dean Smith, Therese Solstad, Daniela Verthelyi, Meenu Wadhwa, Leslie Wagner, Günter Waxenecker, Haoheng Yan, and Lucia Zhang
- Subjects
Medical Laboratory Technology ,Clinical Biochemistry ,General Medicine ,General Pharmacology, Toxicology and Pharmaceutics ,Analytical Chemistry - Abstract
The 15th edition of the Workshop on Recent Issues in Bioanalysis (15th WRIB) was held on 27 September to 1 October 2021. Even with a last-minute move from in-person to virtual, an overwhelmingly high number of nearly 900 professionals representing pharma and biotech companies, contract research organizations (CROs), and multiple regulatory agencies still eagerly convened to actively discuss the most current topics of interest in bioanalysis. The 15th WRIB included 3 Main Workshops and 7 Specialized Workshops that together spanned 1 week in order to allow exhaustive and thorough coverage of all major issues in bioanalysis, biomarkers, immunogenicity, gene therapy, cell therapy and vaccines. Moreover, in-depth workshops on biomarker assay development and validation (BAV) (focused on clarifying the confusion created by the increased use of the term “Context of Use – COU”); mass spectrometry of proteins (therapeutic, biomarker and transgene); state-of-the-art cytometry innovation and validation; and, critical reagent and positive control generation were the special features of the 15th edition. This 2021 White Paper encompasses recommendations emerging from the extensive discussions held during the workshop, and is aimed to provide the bioanalytical community with key information and practical solutions on topics and issues addressed, in an effort to enable advances in scientific excellence, improved quality and better regulatory compliance. Due to its length, the 2021 edition of this comprehensive White Paper has been divided into three parts for editorial reasons. This publication (Part 1A) covers the recommendations on Endogenous Compounds, Small Molecules, Complex Methods, Regulated Mass Spec of Large Molecules, Small Molecule, PoC. Part 1B covers the Regulatory Agencies' Inputs on Bioanalysis, Biomarkers, Immunogenicity, Gene & Cell Therapy and Vaccine. Part 2 (ISR for Biomarkers, Liquid Biopsies, Spectral Cytometry, Inhalation/Oral & Multispecific Biotherapeutics, Accuracy/LLOQ for Flow Cytometry) and Part 3 (TAb/NAb, Viral Vector CDx, Shedding Assays; CRISPR/Cas9 & CAR-T Immunogenicity; PCR & Vaccine Assay Performance; ADA Assay Comparabil ity & Cut Point Appropriateness) are published in volume 14 of Bioanalysis, issues 10 and 11 (2022), respectively.
- Published
- 2022
- Full Text
- View/download PDF
4. 2021 White Paper on Recent Issues in Bioanalysis: Mass Spec of Proteins, Extracellular Vesicles, CRISPR, Chiral Assays, Oligos; Nanomedicines Bioanalysis; ICH M10 Section 7.1; Non-LiquidRare Matrices; Regulatory Inputs (
- Author
-
Surinder, Kaur, Stephen C, Alley, Matt, Szapacs, Amanda, Wilson, Eugene, Ciccimaro, Dian, Su, Neil, Henderson, Linzhi, Chen, Fabio, Garofolo, Shawna, Hengel, Wenying, Jian, John F, Kellie, Anita, Lee, John, Mehl, Joe, Palandra, Haibo, Qiu, Natasha, Savoie, Diaa, Shakleya, Ludovicus, Staelens, Hiroshi, Sugimoto, Giane, Sumner, Jan, Welink, Robert, Wheller, Y-J, Xue, Jianing, Zeng, Jinhui, Zhang, Huiyu, Zhou, Jian, Wang, Scott, Summerfield, Olga, Kavetska, Lieve, Dillen, Ragu, Ramanathan, Mike, Baratta, Arindam, Dasgupta, Anna, Edmison, Luca, Ferrari, Sally, Fischer, Daniela, Fraier, Sam, Haidar, Kathrin, Heermeier, Christopher, James, Allena, Ji, Lina, Luo, Gustavo Mendes, Lima Santos, Noah, Post, Anton I, Rosenbaum, Sune, Sporring, Sekhar, Surapaneni, Stephen, Vinter, Katty, Wan, Eric, Woolf, Seongeun Julia, Cho, Elham, Kossary, Sandra, Prior, Mohsen Rajabi, Abhari, Catherine, Soo, Yow-Ming, Wang, Abbas, Bandukwala, Elana, Cherry, Isabelle, Cludts, Soma, Ghosh, Shirley, Hopper, Akiko, Ishii-Watabe, Susan, Kirshner, Kevin, Maher, Kimberly, Maxfield, Joao, Pedras-Vasconcelos, Yoshiro, Saito, Dean, Smith, Therese, Solstad, Daniela, Verthelyi, Meenu, Wadhwa, Leslie, Wagner, Günter, Waxenecker, Haoheng, Yan, and Lucia, Zhang
- Subjects
Extracellular Vesicles ,Vaccines ,Nanomedicine ,Cell- and Tissue-Based Therapy ,Humans ,Biomarkers ,Mass Spectrometry - Abstract
The 15th edition of the Workshop on Recent Issues in Bioanalysis (15th WRIB) was held on 27 September to 1 October 2021. Even with a last-minute move from in-person to virtual, an overwhelmingly high number of nearly 900 professionals representing pharma and biotech companies, contract research organizations (CROs), and multiple regulatory agencies still eagerly convened to actively discuss the most current topics of interest in bioanalysis. The 15th WRIB included 3 Main Workshops and 7 Specialized Workshops that together spanned 1 week in order to allow exhaustive and thorough coverage of all major issues in bioanalysis, biomarkers, immunogenicity, gene therapy, cell therapy and vaccines. Moreover, in-depth workshops on biomarker assay development and validation (BAV) (focused on clarifying the confusion created by the increased use of the term "Context of Use - COU"); mass spectrometry of proteins (therapeutic, biomarker and transgene); state-of-the-art cytometry innovation and validation; and, critical reagent and positive control generation were the special features of the 15th edition. This 2021 White Paper encompasses recommendations emerging from the extensive discussions held during the workshop, and is aimed to provide the bioanalytical community with key information and practical solutions on topics and issues addressed, in an effort to enable advances in scientific excellence, improved quality and better regulatory compliance. Due to its length, the 2021 edition of this comprehensive White Paper has been divided into three parts for editorial reasons. This publication (Part 1A) covers the recommendations on Endogenous Compounds, Small Molecules, Complex Methods, Regulated Mass Spec of Large Molecules, Small Molecule, PoC. Part 1B covers the Regulatory Agencies' Inputs on Bioanalysis, Biomarkers, Immunogenicity, GeneCell Therapy and Vaccine. Part 2 (ISR for Biomarkers, Liquid Biopsies, Spectral Cytometry, Inhalation/OralMultispecific Biotherapeutics, Accuracy/LLOQ for Flow Cytometry) and Part 3 (TAb/NAb, Viral Vector CDx, Shedding Assays; CRISPR/Cas9CAR-T Immunogenicity; PCRVaccine Assay Performance; ADA Assay Comparabil ityCut Point Appropriateness) are published in volume 14 of Bioanalysis, issues 10 and 11 (2022), respectively.
- Published
- 2022
5. Determination of label efficiency and label degree of critical reagents by LC-MS and native MS
- Author
-
Katie Carle, John F. Kellie, George R. Gunn, and Yong Jiang
- Subjects
Biophysics ,Cell Biology ,Molecular Biology ,Biochemistry - Abstract
Degree of labeling and label efficiency are key factors for optimal characterization of critical reagents that are used in ligand binding assays. Here, three case studies are shown demonstrating how liquid chromatography-mass spectrometry (LC-MS) was utilized to characterize critical reagents using three unique methodologies. Critical reagent batches were prepared for LC-MS analysis by use of: 20 mM dithiothreitol (DTT) (Case 1), rapid PNGaseF (Case 2), and a mobile phase diluent (Case 3). LC-MS was run at three different MS method conditions in each troubleshooting case specific for reduced IgG, intact IgG, and native LC-MS, respectively. Specified LC-MS methods based on sample type and configuration elucidated clear MS profiles, allowing for degree of labeling and label efficiencies to be calculated. Ultimately the LC-MS analyses were fine-tuned for critical reagent characterization, and practices for analyzing similar reagents in the future can be established.
- Published
- 2023
- Full Text
- View/download PDF
6. A High Throughput Antibody-Free Platform for Multiplexed, Sensitive Quantification of Protein Biomarkers in Complex Biomatrices
- Author
-
Bo An, Timothy Sikorsiki, John F. Kellie, Zhuo Chen, Nicole Schneck, John Mehl, Huaping Tang, Jun Qu, Tujin Shi, Yuqian Gao, Jon M. Jacobs, Eshani Nandita, Remco van Soest, and Elliott Jones
- Subjects
History ,Polymers and Plastics ,Business and International Management ,Industrial and Manufacturing Engineering - Published
- 2022
- Full Text
- View/download PDF
7. Cloud solutions for GxP laboratories: considerations for data storage
- Author
-
Samuel O. Pine, Eric Woolf, Jeb Adams, Ryan Kelly, Jason Kentner, Michelle L. Dawes, Joel Usansky, Kimberly Honrine, John F. Kellie, Scott Davis, Shibani Mitra-Kaushik, John Evens, Hannes Hochreiner, and Sean M. Crawford
- Subjects
business.industry ,Computer science ,Clinical Biochemistry ,Information Storage and Retrieval ,Cloud computing ,General Medicine ,Cloud Computing ,Analytical Chemistry ,Medical Laboratory Technology ,Drug development ,Data integrity ,Computer data storage ,Systems engineering ,Humans ,General Pharmacology, Toxicology and Pharmaceutics ,business ,Laboratories ,Cloud storage ,Pharmaceutical industry - Abstract
Challenges for data storage during drug development have become increasingly complex as the pharmaceutical industry expands in an environment that requires on-demand availability of data and resources for users across the globe. While the efficiency and relative low cost of cloud services have become increasingly attractive, hesitancy toward the use of cloud services has decreased and there has been a significant shift toward real-world implementation. Within GxP laboratories, the considerations for cloud storage of data include data integrity and security, as well as access control and usage for users around the globe. In this review, challenges and considerations when using cloud storage options for the storage of laboratory-based GxP data are discussed and best practices are defined.
- Published
- 2021
8. IQ consortium perspective: complementary LBA and LC–MS in protein therapeutics bioanalysis and biotransformation assessment
- Author
-
Mark Jairaj, Jianing Zeng, Kevin P. Bateman, Jens Sydor, Jim Glick, John F. Kellie, and Surinder Kaur
- Subjects
Bioanalysis ,Protein therapeutics ,Computer science ,010401 analytical chemistry ,Clinical Biochemistry ,General Medicine ,Integrated approach ,Scientific expertise ,030226 pharmacology & pharmacy ,01 natural sciences ,Data science ,0104 chemical sciences ,Analytical Chemistry ,03 medical and health sciences ,Medical Laboratory Technology ,0302 clinical medicine ,Biotransformation ,Tandem Mass Spectrometry ,Lc ms ms ,Humans ,Biological Assay ,General Pharmacology, Toxicology and Pharmaceutics ,Chromatography, Liquid - Abstract
Increasingly diverse large molecule modalities have driven the need for complex bioanalysis and biotransformation assessment involving both traditional ligand-binding assays (LBA) and more recent hybrid immunoaffinity LC–MS platforms. Given the scientific expertise in LBA and LC–MS typically resides in different functions within the industry, this has presented operational challenges for an integrated approach for bioanalysis and biotransformation assessment. Encouragingly, over time, the industry has recognized the complementary value of the two platforms. This has not been an easy transition as organizational structures vary widely within the industry. However, there are tremendous benefits in adopting fully integrated strategies for biopharma. This IQ consortium paper presents current perspectives across the biopharma industry. It highlights the technical and operational challenges in current large molecule bioanalysis, the value of collaborations across LBA and LC–MS, and scientific expertise for fully integrated strategies for bioanalysis and biotransformation.
- Published
- 2020
- Full Text
- View/download PDF
9. An antibody-free platform for multiplexed, sensitive quantification of protein biomarkers in complex biomatrices
- Author
-
Bo An, Timothy W. Sikorski, John F. Kellie, Zhuo Chen, Nicole A. Schneck, John Mehl, Huaping Tang, Jun Qu, Tujin Shi, Yuqian Gao, Jon M. Jacobs, Eshani Nandita, Remco van Soest, and Elliott Jones
- Subjects
Solid Phase Extraction ,Organic Chemistry ,Proteins ,General Medicine ,Biochemistry ,Antibodies ,Mass Spectrometry ,Analytical Chemistry ,Dogs ,Animals ,Humans ,Peptides ,Biomarkers ,Chromatography, Liquid - Abstract
Sensitive, multiplexed protein quantification remains challenging despite recent advancements in LC-MS assays for targeted protein biomarker quantification. High-sensitivity protein biomarker measurements usually require immuno-affinity enrichment of target protein; a process which is highly dependent on capture reagent and limited in capability to measure multiple analytes. Herein, we report a novel antibody-free platform, which measures multiple biomarkers from complex matrices employing a strategically optimized solid-phase extraction cleanup and orthogonal multidimensional LC-MS. Eight human protein biomarkers with different specifications were spiked into canine plasma as a model investigation system. The developed strategy achieved the desired sensitivity, robustness, and throughput via the following steps: (1) post digestion mixed-mode cation exchange-reverse phase SPE enrichment cleaned up the sample initially; (2) rapid, high-pH peptide fractionation further eliminated background components efficiently while selectively enriched signature peptides (SP) to provide sufficient sensitivity for multiple targets; and (3) trapping-micro-LC-MS analysis delivered high sensitivity comparable to a nano-LC-MS method but with much better robustness and throughput for the final analysis. Compared with a conventional LC-MS assay with direct protein digestion and limited clean-up, analysis with this antibody-free platform improved the LLOQ by 1-2 orders of magnitude for the eight protein biomarkers, reaching as low as 5 ng/mL in plasma, with feasible robustness and throughput. This platform was applied for the quantification of biomarkers of respiratory conditions in patients with various lung diseases, demonstrating real-world applicability.
- Published
- 2022
- Full Text
- View/download PDF
10. Intact mAb LC-MS for drug concentration from pre-clinical studies: bioanalytical method performance and in-life samples
- Author
-
Nicole A Schneck, Matthew Szapacs, Yun Alelyunas, Ian Edwards, Henry Shion, Josh Albert, Caroline J. Sychterz, Zhuo Chen, Mark Wrona, and John F. Kellie
- Subjects
Bioanalysis ,medicine.drug_class ,Clinical Biochemistry ,Peptide ,Monoclonal antibody ,Mass spectrometry ,030226 pharmacology & pharmacy ,01 natural sciences ,Analytical Chemistry ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,Liquid chromatography–mass spectrometry ,Tandem Mass Spectrometry ,medicine ,Humans ,General Pharmacology, Toxicology and Pharmaceutics ,chemistry.chemical_classification ,Chromatography ,Molecular mass ,Chemistry ,010401 analytical chemistry ,Antibodies, Monoclonal ,General Medicine ,0104 chemical sciences ,Biological Therapy ,Medical Laboratory Technology ,Drug concentration ,Pharmaceutical Preparations ,Chromatography, Liquid - Abstract
Background: Antibody biotherapeutic measurement from pharmacokinetic studies has not been traditionally based on intact molecular mass as is the case for small molecules. However, recent advancements in protein capture and mass spectrometer technology have enabled intact mass detection and quantitation for dosed biotherapeutics. A bioanalytical method validation is part of the regulatory requirement for sample analysis to determine drug concentration from in-life study samples. Results/methodology: Here, an intact protein LC–MS assay is subjected to mock bioanalytical method validation, and unknown samples are compared between intact protein LC–MS and established bioanalytical assay formats: Ligand-binding assay and peptide LC–MS/MS. Discussion/conclusion: Results are presented from the intact and traditional bioanalytical method evaluations, where the in-life sample concentrations were comparable across method types with associated data analyses presented. Furthermore, for intact protein LC–MS, modification monitoring and evaluation of data processing parameters is demonstrated.
- Published
- 2020
11. Intact Protein Mass Spectrometry for Therapeutic Protein Quantitation, Pharmacokinetics, and Biotransformation in Preclinical and Clinical Studies: An Industry Perspective
- Author
-
Ying Ge, Kevin P. Bateman, Jack Henion, Barry R Jones, John C. Tran, John T Mehl, John F. Kellie, and Wenying Jian
- Subjects
Immunoconjugates ,Drug Evaluation, Preclinical ,Computational biology ,010402 general chemistry ,Mass spectrometry ,01 natural sciences ,Chromatography, Affinity ,Mass Spectrometry ,Specimen Handling ,Pharmacokinetics ,Biotransformation ,Structural Biology ,Animals ,Humans ,Spectroscopy ,Protein molecules ,Drug discovery ,Chemistry ,010401 analytical chemistry ,Therapeutic protein ,Intact protein ,Proteins ,Small molecule ,0104 chemical sciences ,Chromatography, Liquid - Abstract
Recent advancements in immunocapture methods and mass spectrometer technology have enabled intact protein mass spectrometry to be applied for the characterization of antibodies and other large biotherapeutics from in-life studies. Protein molecules have not been traditionally studied by intact mass or screened for catabolites in the same manner as small molecules, but the landscape has changed. Researchers have presented methods that can be applied to the drug discovery and development stages, and others are exploring the possibilities of the new approaches. However, a wide variety of options for assay development exists without clear recommendation on best practice, and data processing workflows may have limitations depending on the vendor. In this perspective, we share experiences and recommendations for current and future application of mass spectrometry for biotherapeutic molecule monitoring from preclinical and clinical studies.
- Published
- 2020
12. Antibody Subunit LC-MS Analysis for Pharmacokinetic and Biotransformation Determination from In-Life Studies for Complex Biotherapeutics
- Author
-
Yinghe Brenda Li, Andrew P Mayer, John F. Kellie, Caroline J. Sychterz, Kristen E. Pannullo, Matthew Szapacs, Kristy J Fraley, and Molly Z Karlinsey
- Subjects
Drug ,Immunoconjugates ,medicine.drug_class ,media_common.quotation_subject ,010402 general chemistry ,Monoclonal antibody ,01 natural sciences ,Mass Spectrometry ,Analytical Chemistry ,Pharmacokinetics ,Biotransformation ,In vivo ,medicine ,Animals ,Deamidation ,media_common ,Chemistry ,Drug discovery ,010401 analytical chemistry ,Antibodies, Monoclonal ,Fusion protein ,Macaca mulatta ,0104 chemical sciences ,Rats ,Biochemistry ,Chromatography, Liquid - Abstract
Complex biotherapeutics present challenges from drug discovery, screening, and development perspectives. While monoclonal antibody drugs are not monitored for metabolites in the same manner as small molecules, biotherapeutics such as fusion proteins, antibody-drug conjugates, or bispecific antibodies may undergo biotransformation (such as clipping, deamidation, or oxidation) in vivo, resulting in catabolites that can have a direct impact on drug safety or efficacy. Here antibody subunit LC-MS is utilized for evaluation of two classes of complex biotherapeutics: an antibody-drug conjugate and a mAb-fusion biotherapeutic. Pharmacokinetic concentration, biotransformation, and DAR data are collectively presented using the subunit LC-MS approach for the two molecules, and the methods shared in detail can be applied to any humanized IgG1 mAb biotherapeutic for preclinical study support. Overall, the data generated from antibody LC-MS analyses can provide key information in early phase development and deliver multiple study end points with a single data set.
- Published
- 2020
13. Intact protein LC–MS for pharmacokinetics
- Author
-
John F. Kellie
- Subjects
Bioanalysis ,Pharmacokinetics ,Chemistry ,Liquid chromatography–mass spectrometry ,Immunogenicity ,education ,Intact protein ,Pharmacology ,humanities - Abstract
Biography: John Kellie is currently a GlaxoSmithKline (GSK) fellow in the Bioanalysis, Immunogenicity, and Biomarkers group at GSK. John received his B.Sc. in Biochemistry from Indiana University (USA) and his PhD in Chemistry from Northwestern University (USA) studying under Dr Neil Kelleher. He was a post-doctoral scientist at Eli Lilly and Company, where he developed methods for intact protein quantitation of a Parkinson’s Disease biomarker from human brain tissue. At GSK, John utilizes mass spectrometry for development of novel bioanalytical methods for biotherapeutic and protein quantitation from pre-clinical and clinical samples, with a focus on intact protein and large mass quantitation for pharmacokinetics, catabolism, biotransformation and product quality attribute support. John Kellie speaks to the International Journal of Pharmacokinetics about intact protein LC–MS for pharmacokinetic application.
- Published
- 2019
- Full Text
- View/download PDF
14. A new era for proteomics
- Author
-
Zhuo Chen, Maria Busz, John F. Kellie, Thomas E. Angel, Ahmed H Moghieb, Timothy W. Sikorski, Matthew Szapacs, and Bo An
- Subjects
Proteomics ,Proteomics methods ,Chemistry ,Drug discovery ,Clinical Biochemistry ,General Medicine ,Computational biology ,Biomarkers, Pharmacological ,Mass Spectrometry ,Analytical Chemistry ,Medical Laboratory Technology ,Drug Discovery ,Humans ,General Pharmacology, Toxicology and Pharmaceutics - Published
- 2019
15. Review of approaches and examples for monitoring biotransformation in protein and peptide therapeutics by MS
- Author
-
Molly Z Karlinsey and John F. Kellie
- Subjects
0301 basic medicine ,Computer science ,010401 analytical chemistry ,Clinical Biochemistry ,General Medicine ,Computational biology ,01 natural sciences ,0104 chemical sciences ,Analytical Chemistry ,03 medical and health sciences ,Medical Laboratory Technology ,030104 developmental biology ,Drug concentration ,Biotransformation ,Drug development ,General Pharmacology, Toxicology and Pharmaceutics - Abstract
Biotherapeutic drugs have emerged in quantity in pharmaceutical pipelines, and increasingly diverse biomolecules are progressed through preclinical and clinical development. As purification, separation, mass spectrometer detection and data processing capabilities improve, there is opportunity to monitor drug concentration by traditional ligand-binding assay or MS measurement and to monitor metabolism, catabolism or other biomolecular mass variants present in circulation. This review highlights approaches and examples of monitoring biotransformation of biotherapeutics by MS as these techniques are poised to add value to drug development in years to come. The increased use of such approaches, and the successful quantitation of biotherapeutic structural modifications, will provide insightful data for the benefit of both researchers and patients.
- Published
- 2018
16. Biotherapeutic Antibody Subunit LC-MS and Peptide Mapping LC-MS Measurements to Study Possible Biotransformation and Critical Quality Attributes In Vivo
- Author
-
Shing H. Mai, Sarah L. Childs, Robert A. Biddlecombe, Molly Z Karlinsey, John R. White, John F. Kellie, Shugui Chen, and Andrew S. Thomson
- Subjects
Quality Control ,medicine.drug_class ,Chemistry, Pharmaceutical ,Pharmaceutical Science ,02 engineering and technology ,Monoclonal antibody ,030226 pharmacology & pharmacy ,Peptide Mapping ,03 medical and health sciences ,0302 clinical medicine ,Biotransformation ,Liquid chromatography–mass spectrometry ,In vivo ,Tandem Mass Spectrometry ,medicine ,Critical to quality ,Animals ,Chromatography, High Pressure Liquid ,Chemistry ,Ligand binding assay ,Antibodies, Monoclonal ,021001 nanoscience & nanotechnology ,Rats ,Biochemistry ,Drug development ,0210 nano-technology ,Critical quality attributes - Abstract
Biotransformation monitoring involves tracking drug modification occurring during in-life studies. Critical Quality Attribute monitoring from forced degraded drug material or in-life sample sets can provide an in-depth assessment of product quality for support in early- or late-stage drug development. For Critical Quality Attribute analysis, biotherapeutic monoclonal antibody (mAb) subunit analysis and peptide mapping liquid chromatography–mass spectrometry (LC-MS) approaches are used, although typically from an in vitro setting (e.g., formulation buffer) not involving biological samples or material. Here, samples from a high-dose rat study (in vivo) are subjected to analysis by ligand binding assay, mAb subunit LC-MS, and peptide mapping by LC-MS. Taken together, data from the 3 analytical approaches provide information regarding drug concentration in circulation, biotransformation, and biotherapeutic drug product quality. The concept of a multitier workflow for preclinical or clinical sample sets can be applied to other biotherapeutic mAb products such as bispecific mAbs, fusions proteins, or antibody-drug conjugates.
- Published
- 2018
17. Application of high-resolution MS for development of peptide and large-molecule drug candidates
- Author
-
John F. Kellie, Matthew Szapacs, and Jonathan R Kehler
- Subjects
Male ,Drug ,Bioanalysis ,media_common.quotation_subject ,Clinical Biochemistry ,High resolution ,Peptide ,Mass spectrometry ,030226 pharmacology & pharmacy ,01 natural sciences ,Mass Spectrometry ,Analytical Chemistry ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Nuclear magnetic resonance ,Limit of Detection ,Animals ,Molecule ,Tissue Distribution ,General Pharmacology, Toxicology and Pharmaceutics ,media_common ,chemistry.chemical_classification ,Chromatography ,010401 analytical chemistry ,General Medicine ,PK Parameters ,0104 chemical sciences ,Triple quadrupole mass spectrometer ,Medical Laboratory Technology ,Pharmaceutical Preparations ,chemistry ,Female ,Peptides - Abstract
Background: For quantitative bioanalysis utilizing MS, the instrument of choice is typically a triple quadruple mass spectrometer. However, advances in high-resolution MS have allowed sensitivity and dynamic ranges to approach that of triple quadrupole instruments. Results: A matrix-free protein digest, a digested therapeutic protein and the intact peptide therapeutic liraglutide were each analyzed on high-resolution and triple quadrupole mass spectrometers with data compared. Samples from a mouse PK study with liraglutide were analyzed using the two different instruments, and equivalent PK exposure data were demonstrated. Conclusion: High-resolution and triple quadrupole mass spectrometers can generate data resulting in identical PK parameters from an in-life sample set, thus giving confidence in either technique in support of biotherapeutic PK exposure studies.
- Published
- 2016
- Full Text
- View/download PDF
18. Mechanism of Inactivation of GABA Aminotransferase by (E)- and (Z)-(1S,3S)-3-Amino-4-fluoromethylenyl-1-cyclopentanoic Acid
- Author
-
Ruslan Sanishvili, Richard B. Silverman, John F. Kellie, Boobalan Pachaiyappan, Emma H. Doud, Hyunbeom Lee, Rui Wu, Dali Liu, Phillip D. Compton, Hoang V. Le, and Neil L. Kelleher
- Subjects
Models, Molecular ,Proline ,Swine ,Metabolite ,Central nervous system ,Pharmacology ,Crystallography, X-Ray ,Formyl group ,Biochemistry ,Article ,Vigabatrin ,chemistry.chemical_compound ,medicine ,Animals ,Enzyme Inhibitors ,Pyridoxal ,chemistry.chemical_classification ,General Medicine ,Inhibitory neurotransmitter ,Enzyme Activation ,medicine.anatomical_structure ,Enzyme ,nervous system ,chemistry ,4-Aminobutyrate Transaminase ,Molecular Medicine ,Salt bridge ,medicine.drug - Abstract
When γ-aminobutyric acid (GABA), the major inhibitory neurotransmitter in the mammalian central nervous system, falls below a threshold level, seizures occur. One approach to raise GABA concentrations is to inhibit GABA aminotransferase (GABA-AT), a pyridoxal 5'-phosphate-dependent enzyme that degrades GABA. We have previously developed (1S,3S)-3-amino-4-difluoromethylene-1-cyclopentanoic acid (CPP-115), which is 186 times more efficient in inactivating GABA-AT than vigabatrin, the only FDA-approved inactivator of GABA-AT. We also developed (E)- and (Z)-(1S,3S)-3-amino-4-fluoromethylenyl-1-cyclopentanoic acid (1 and 2, respectively), monofluorinated analogs of CPP-115, which are comparable to vigabatrin in inactivating GABA-AT. Here, we report the mechanism of inactivation of GABA-AT by 1 and 2. Both produce a metabolite that induces disruption of the Glu270-Arg445 salt bridge to accommodate interaction between the metabolite formyl group and Arg445. This is the second time that Arg445 has interacted with a ligand and is involved in GABA-AT inactivation, thereby confirming the importance of Arg445 in future inactivator design.
- Published
- 2015
- Full Text
- View/download PDF
19. Drug monitoring by volumetric absorptive microsampling: method development considerations to mitigate hematocrit effects
- Author
-
Chester L. Bowen, Molly Z Karlinsey, Christopher A. Evans, John F. Kellie, and Kasie Fang
- Subjects
Drug ,Bioanalysis ,media_common.quotation_subject ,Clinical Biochemistry ,Hematocrit ,030226 pharmacology & pharmacy ,01 natural sciences ,Analytical Chemistry ,Matrix (chemical analysis) ,03 medical and health sciences ,0302 clinical medicine ,Tandem Mass Spectrometry ,medicine ,Humans ,General Pharmacology, Toxicology and Pharmaceutics ,media_common ,Chromatography ,Human blood ,medicine.diagnostic_test ,Chemistry ,010401 analytical chemistry ,Extraction (chemistry) ,General Medicine ,Reference Standards ,Method development ,0104 chemical sciences ,Medical Laboratory Technology ,Pharmaceutical Preparations ,Calibration ,Drug Monitoring ,Quantitative analysis (chemistry) ,Chromatography, Liquid - Abstract
Aim: GSKA is a compound that was in development in clinical trials. A bioanalysis method to quantify GSKA using volumetric absorptive microsampling (VAMS) was developed and hematocrit (HCT) related assay bias was investigated. Methodology: After accurate sampling of 10 μl blood, VAMS tips were air dried approximately 18 h and desorbed by an aqueous solution containing internal standard. The recovered blood underwent liquid–liquid extraction in ethyl acetate to minimize matrix suppression. Assay accuracy, precision, linearity, carryover, selectivity, recovery, matrix effects, HCT effects and long-term quality control stability were evaluated. Conclusion: HCT-related assay bias was minimized in 30–60% blood HCT range, and all validation parameters met acceptance criteria. The method is suitable for quantitative analysis of GSKA in human blood.
- Published
- 2018
20. Toward best practices in data processing and analysis for intact biotherapeutics by MS in quantitative bioanalysis
- Author
-
Scott G. Summerfield, Jonathan R Kehler, John F. Kellie, and Molly Z Karlinsey
- Subjects
Bioanalysis ,Chromatography ,Chemistry ,010401 analytical chemistry ,Clinical Biochemistry ,Intact protein ,Antibodies, Monoclonal ,General Medicine ,030226 pharmacology & pharmacy ,01 natural sciences ,0104 chemical sciences ,Analytical Chemistry ,Rats ,03 medical and health sciences ,Medical Laboratory Technology ,0302 clinical medicine ,Limit of Detection ,Tandem Mass Spectrometry ,Animals ,General Pharmacology, Toxicology and Pharmaceutics ,Peptides ,Chromatography, High Pressure Liquid - Abstract
Aim: Typically, quantitation of biotherapeutics from biological matrices by LC–MS is based on a surrogate peptide approach to determine molecule concentration. Recent efforts have focused on quantitation of the intact protein molecules or larger mass subunits of monoclonal antibodies. To date, there has been limited guidance for large or intact protein mass quantitation for quantitative bioanalysis. Methodology: Intact- and subunit-level analyses of biotherapeutics from biological matrices are performed at 12–25 kDa mass range with quantitation data presented. Results: Linearity, bias and other metrics are presented along with recommendations made on the viability of existing quantitation approaches. Conclusion: This communication is intended to start a discussion around intact protein data analysis and processing, recognizing that other published contributions will be required.
- Published
- 2017
21. A whole-molecule immunocapture LC-MS approach for the in vivo quantitation of biotherapeutics
- Author
-
Charles S Hottenstein, Jonathan R Kehler, John F. Kellie, Thomas Mencken, and Richard J Snell
- Subjects
Glycosylation ,medicine.drug_class ,Clinical Biochemistry ,Monoclonal antibody ,030226 pharmacology & pharmacy ,01 natural sciences ,Analytical Chemistry ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Liquid chromatography–mass spectrometry ,Tandem Mass Spectrometry ,medicine ,Humans ,General Pharmacology, Toxicology and Pharmaceutics ,Good practice ,Chromatography, High Pressure Liquid ,Immunoassay ,Chromatography ,Enzymatic digestion ,Chemistry ,010401 analytical chemistry ,Antibodies, Monoclonal ,General Medicine ,0104 chemical sciences ,Medical Laboratory Technology ,Drug development ,Human plasma ,Peptides ,Blood Chemical Analysis - Abstract
Aim: Large-molecule biotherapeutic quantitation in vivo by LC–MS has traditionally relied on enzymatic digestion followed by quantitation of a ‘surrogate peptide’ to infer whole-molecule concentration. MS methods presented here measure the whole molecule and provide a platform to better understand the various circulating drug forms by allowing for variant quantitation. Results: An immunocapture LC–MS method for quantitation of a biotherapeutic monoclonal antibody from human plasma is presented. Sensitivity, precision and accuracy for each molecular portion are presented along with an example of glycoform variant quantitation. Conclusion: The method is presented as a basic platform to be further developed for Good Practice (GxP) applications, critical quality attribute analysis or general understanding of molecular forms present as required for the wide range of drug development processes.
- Published
- 2016
22. Nano-LC FTICR Tandem Mass Spectrometry for Top-Down Proteomics: Routine Baseline Unit Mass Resolution of Whole Cell Lysate Proteins up to 72 kDa
- Author
-
Dorothy R. Ahlf, John C. Tran, Ji Eun Lee, Neil L. Kelleher, Kenneth R. Durbin, Adam D. Catherman, John F. Kellie, Jeremiah D. Tipton, Alan G. Marshall, and Christopher L. Hendrickson
- Subjects
Proteomics ,Spectrometry, Mass, Electrospray Ionization ,Chromatography ,Fourier Analysis ,Resolution (mass spectrometry) ,Protein mass spectrometry ,Chemistry ,Quantitative proteomics ,Proteins ,Tandem mass spectrometry ,Top-down proteomics ,Article ,Analytical Chemistry ,Molecular Weight ,Human proteome project ,Humans ,Nanotechnology ,Bottom-up proteomics ,Isoelectric Focusing ,Chromatography, High Pressure Liquid ,HeLa Cells - Abstract
Current high-throughput top-down proteomic platforms provide routine identification of proteins less than 25 kDa with 4-D separations. This short communication reports the application of technological developments over the past few years that improve protein identification and characterization for masses greater than 25 kDa. Advances in separation science have allowed increased numbers of proteins to be identified, especially by nanoliquid chromatography (nLC) prior to mass spectrometry (MS) analysis. Further, a goal of high-throughput top-down proteomics is to extend the mass range for routine nLC MS analysis up to 80 kDa because gene sequence analysis predicts that ~70% of the human proteome is transcribed to be less than 80 kDa. Normally, large proteins greater than 50 kDa are identified and characterized by top-down proteomics through fraction collection and direct infusion at relatively low throughput. Further, other MS-based techniques provide top-down protein characterization, however at low resolution for intact mass measurement. Here, we present analysis of standard (up to 78 kDa) and whole cell lysate proteins by Fourier transform ion cyclotron resonance mass spectrometry (nLC electrospray ionization (ESI) FTICR MS). The separation platform reduced the complexity of the protein matrix so that, at 14.5 T, proteins from whole cell lysate up to 72 kDa are baseline mass resolved on a nano-LC chromatographic time scale. Further, the results document routine identification of proteins at improved throughput based on accurate mass measurement (less than 10 ppm mass error) of precursor and fragment ions for proteins up to 50 kDa.
- Published
- 2012
- Full Text
- View/download PDF
23. Intact mass detection, interpretation, and visualization to automate Top-Down proteomics on a large scale
- Author
-
Neil L. Kelleher, Mingxi Li, Leonid Zamdborg, Steve M. M. Sweet, Kenneth R. Durbin, John F. Kellie, John C. Tran, Adam D. Catherman, and Ji Eun Lee
- Subjects
Proteomics ,Molecular Sequence Data ,Computational biology ,Biology ,Top-down proteomics ,Biochemistry ,Mass Spectrometry ,Article ,Fungal Proteins ,Histones ,Software ,Tubulin ,Humans ,Protein Isoforms ,Amino Acid Sequence ,Molecular Biology ,Data processing ,Chromatography ,business.industry ,Pipeline (software) ,Visualization ,Proteome ,Keratins ,Stathmin ,Ion trap ,business ,Algorithms ,HeLa Cells - Abstract
Applying high-throughput Top-Down MS to an entire proteome requires a yet-to-be-established model for data processing. Since Top-Down is becoming possible on a large scale, we report our latest software pipeline dedicated to capturing the full value of intact protein data in automated fashion. For intact mass detection, we combine algorithms for processing MS1 data from both isotopically resolved (FT) and charge-state resolved (ion trap) LC-MS data, which are then linked to their fragment ions for database searching using ProSight. Automated determination of human keratin and tubulin isoforms is one result. Optimized for the intricacies of whole proteins, new software modules visualize proteome-scale data based on the LC retention time and intensity of intact masses and enable selective detection of PTMs to automatically screen for acetylation, phosphorylation, and methylation. Software functionality was demonstrated using comparative LC-MS data from yeast strains in addition to human cells undergoing chemical stress. We further these advances as a key aspect of realizing Top-Down MS on a proteomic scale.
- Published
- 2010
- Full Text
- View/download PDF
24. Quantitative immunocapture MS: current status and challenges in drug discovery
- Author
-
John F. Kellie and Matthew Szapacs
- Subjects
Immunoassay ,Bioanalysis ,medicine.diagnostic_test ,Drug discovery ,Chemistry ,Clinical Biochemistry ,Quantitative proteomics ,Ms analysis ,Therapeutic protein ,General Medicine ,Computational biology ,Reference Standards ,Molecular biology ,Mass Spectrometry ,Analytical Chemistry ,Medical Laboratory Technology ,Drug Discovery ,medicine ,Humans ,General Pharmacology, Toxicology and Pharmaceutics ,Chromatography, Liquid - Abstract
Immunocapture is poised to play an increasingly vital role in therapeutic protein quantitation. In recent years, MS has emerged as an alternative to immunoassay for quantitative work. MS analysis can be achieved with or without an immunocapture step; however, the inherent selectivity of the technique typically affords LLOQ coupling immunocapture-MS methods [1]. With an ever-growing need to develop high-sensitivity assays while retaining selectivity, immunocapture followed by digestion before analysis by MS is a viable approach for protein quantitation [2,3]. Here, a brief summary on the status and challenges of protein quantitation by immunocapture followed by MS are discussed.
- Published
- 2014
25. Quantitative Measurement of Intact Alpha-Synuclein Proteoforms from Post-Mortem Control and Parkinson's Disease Brain Tissue by Intact Protein Mass Spectrometry
- Author
-
John F. Kellie, Anthony Major, Thomas G. Beach, Charles H. Adler, Richard E. Higgs, Michael D. Knierman, Kalpana M. Merchant, and John W. Ryder
- Subjects
Spectrometry, Mass, Electrospray Ionization ,Pathology ,medicine.medical_specialty ,Parkinson's disease ,Serial dilution ,Blotting, Western ,Top-down proteomics ,Mass spectrometry ,Tandem mass spectrometry ,Article ,chemistry.chemical_compound ,Tandem Mass Spectrometry ,Cadaver ,medicine ,Humans ,Aged, 80 and over ,Alpha-synuclein ,Multidisciplinary ,Brain ,Parkinson Disease ,Human brain ,medicine.disease ,Blot ,medicine.anatomical_structure ,chemistry ,Case-Control Studies ,Data Interpretation, Statistical ,alpha-Synuclein ,Female ,Biomarkers ,Chromatography, Liquid - Abstract
A robust top down proteomics method is presented for profiling alpha-synuclein species from autopsied human frontal cortex brain tissue from Parkinson's cases and controls. The method was used to test the hypothesis that pathology associated brain tissue will have a different profile of post-translationally modified alpha-synuclein than the control samples. Validation of the sample processing steps, mass spectrometry based measurements, and data processing steps were performed. The intact protein quantitation method features extraction and integration of m/z data from each charge state of a detected alpha-synuclein species and fitting of the data to a simple linear model which accounts for concentration and charge state variability. The quantitation method was validated with serial dilutions of intact protein standards. Using the method on the human brain samples, several previously unreported modifications in alpha-synuclein were identified. Low levels of phosphorylated alpha synuclein were detected in brain tissue fractions enriched for Lewy body pathology and were marginally significant between PD cases and controls (p = 0.03).
- Published
- 2014
- Full Text
- View/download PDF
26. Robust Analysis of the Yeast Proteome under 50 kDa by Molecular-Mass-Based Fractionation and Top-Down Mass Spectrometry
- Author
-
Adam D. Catherman, John C. Tran, Neil L. Kelleher, Paul M. Thomas, Jeremiah D. Tipton, Charles E. Witkowski, Kenneth R. Durbin, Jeremy L. Norris, and John F. Kellie
- Subjects
Chromatography ,Saccharomyces cerevisiae Proteins ,Molecular mass ,Protein mass spectrometry ,Proteome ,Chemistry ,Quantitative proteomics ,Tandem mass spectrometry ,Mass spectrometry ,Proteomics ,Article ,Mass Spectrometry ,Analytical Chemistry ,Molecular Weight ,Biochemistry ,Tandem Mass Spectrometry ,Electrophoresis, Polyacrylamide Gel ,Bottom-up proteomics ,Chromatography, Liquid - Abstract
As the process of top-down mass spectrometry continues to mature, we benchmark the next installment of an improving methodology that incorporates a tube-gel electrophoresis (TGE) device to separate intact proteins by molecular mass. Top-down proteomics is accomplished in a robust fashion to yield the identification of hundreds of unique proteins, many of which correspond to multiple protein forms. The TGE platform separates 0–50 kDa proteins extracted from the yeast proteome into 12 fractions prior to automated nanocapillary LC–MS/MS in technical triplicate. The process may be completed in less than 72 h. From this study, 530 unique proteins and 1103 distinct protein species were identified and characterized, thus representing the highest coverage to date of the Saccharomyces cerevisiae proteome using top-down proteomics. The work signifies a significant step in the maturation of proteomics based on direct measurement and fragmentation of intact proteins.
- Published
- 2011
27. Mapping intact protein isoforms in discovery mode using top-down proteomics
- Author
-
Cong Wu, Adam D. Catherman, Dorothy R. Ahlf, John C. Tran, Ji Eun Lee, Steve M. M. Sweet, Mingxi Li, Paul M. Thomas, Kenneth R. Durbin, Leonid Zamdborg, John F. Kellie, Philip D. Compton, Adaikkalam Vellaichamy, Neil L. Kelleher, Nertila Siuti, Bryan P. Early, Richard D. LeDuc, and Jeremiah D. Tipton
- Subjects
Proteomics ,Proteome ,Chemical biology ,Biology ,Top-down proteomics ,01 natural sciences ,Cell Line ,03 medical and health sciences ,Human proteome project ,Humans ,Protein Isoforms ,HMGA1a Protein ,HMGA1b Protein ,Databases, Protein ,Cellular Senescence ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,010401 analytical chemistry ,Alternative splicing ,0104 chemical sciences ,Cell biology ,Alternative Splicing ,Phenotype ,Proteolysis ,Cell aging ,Protein Processing, Post-Translational ,DNA Damage ,HeLa Cells - Abstract
A full description of the human proteome relies on the challenging task of detecting mature and changing forms of protein molecules in the body. Large-scale proteome analysis has routinely involved digesting intact proteins followed by inferred protein identification using mass spectrometry. This 'bottom-up' process affords a high number of identifications (not always unique to a single gene). However, complications arise from incomplete or ambiguous characterization of alternative splice forms, diverse modifications (for example, acetylation and methylation) and endogenous protein cleavages, especially when combinations of these create complex patterns of intact protein isoforms and species. 'Top-down' interrogation of whole proteins can overcome these problems for individual proteins, but has not been achieved on a proteome scale owing to the lack of intact protein fractionation methods that are well integrated with tandem mass spectrometry. Here we show, using a new four-dimensional separation system, identification of 1,043 gene products from human cells that are dispersed into more than 3,000 protein species created by post-translational modification (PTM), RNA splicing and proteolysis. The overall system produced greater than 20-fold increases in both separation power and proteome coverage, enabling the identification of proteins up to 105 kDa and those with up to 11 transmembrane helices. Many previously undetected isoforms of endogenous human proteins were mapped, including changes in multiply modified species in response to accelerated cellular ageing (senescence) induced by DNA damage. Integrated with the latest version of the Swiss-Prot database, the data provide precise correlations to individual genes and proof-of-concept for large-scale interrogation of whole protein molecules. The technology promises to improve the link between proteomics data and complex phenotypes in basic biology and disease research.
- Published
- 2011
28. The emerging process of Top Down mass spectrometry for protein analysis: biomarkers, protein-therapeutics, and achieving high throughput†
- Author
-
Adam D. Catherman, Neil L. Kelleher, Haylee M. Thomas, Kenneth R. Durbin, John F. Kellie, Ji Eun Lee, Ioanna Ntai, John C. Tran, Paul M. Thomas, Adaikkalam Vellaichamy, Leonid Zamdborg, and Dorothy R. Ahlf
- Subjects
Proteomics ,Chromatography ,Protein therapeutics ,Chemistry ,Proteins ,Computational biology ,Mass spectrometry ,Article ,Mass Spectrometry ,Humans ,Protein identification ,Instrumentation (computer programming) ,Molecular Biology ,Throughput (business) ,Biomarkers ,Biotechnology ,HeLa Cells - Abstract
Top Down mass spectrometry (MS) has emerged as an alternative to common Bottom Up strategies for protein analysis. In the Top Down approach, intact proteins are fragmented directly in the mass spectrometer to achieve both protein identification and characterization, even capturing information on combinatorial post-translational modifications. Just in the past two years, Top Down MS has seen incremental advances in instrumentation and dedicated software, and has also experienced a major boost from refined separations of whole proteins in complex mixtures that have both high recovery and reproducibility. Combined with steadily advancing commercial MS instrumentation and data processing, a high-throughput workflow covering intact proteins and polypeptides up to 70 kDa is directly visible in the near future.
- Published
- 2010
29. Size-sorting combined with improved nanocapillary liquid chromatography-mass spectrometry for identification of intact proteins up to 80 kDa
- Author
-
Neil L. Kelleher, John C. Tran, Adam D. Catherman, Paul M. Thomas, Adaikkalam Vellaichamy, Kenneth R. Durbin, John F. Kellie, Steve M. M. Sweet, Ji Eun Lee, Leonid Zamdborg, Dorothy R. Ahlf, and Gary A. Valaskovic
- Subjects
Saccharomyces cerevisiae Proteins ,Time Factors ,Proteome ,Polymers ,Molecular Sequence Data ,Top-down proteomics ,Proteomics ,Mass spectrometry ,Fourier transform spectroscopy ,Mass Spectrometry ,Article ,Analytical Chemistry ,Liquid chromatography–mass spectrometry ,Mole ,Animals ,Humans ,Nanotechnology ,Amino Acid Sequence ,Quadrupole ion trap ,Chromatography ,Chemistry ,Proteins ,Molecular Weight ,Cattle ,Ion trap ,Porosity ,Chromatography, Liquid ,HeLa Cells - Abstract
Despite the availability of ultra-high-resolution mass spectrometers, methods for separation and detection of intact proteins for proteome-scale analyses are still in a developmental phase. Here we report robust protocols for online LC-MS to drive high-throughput top-down proteomics in a fashion similar to that of bottom-up proteomics. Comparative work on protein standards showed that a polymeric stationary phase led to superior sensitivity over a silica-based medium in reversed-phase nanocapillary LC, with detection of proteins50 kDa routinely accomplished in the linear ion trap of a hybrid Fourier transform mass spectrometer. Protein identification was enabled by nozzle-skimmer dissociation and detection of fragment ions with10 ppm mass accuracy for highly specific database searching using tailored software. This overall approach led to identification of proteins up to 80 kDa, with 10-60 proteins identified in single LC-MS runs of samples from yeast and human cell lines prefractionated by their molecular mass using a gel-based sieving system.
- Published
- 2010
30. A robust two-dimensional separation for top-down tandem mass spectrometry of the low-mass proteome
- Author
-
John C. Tran, Ioanna Ntai, Alan G. Marshall, Jeremiah D. Tipton, Dorothy R. Ahlf, Ji Eun Lee, Haylee M. Thomas, Neil L. Kelleher, Adaikkalam Vellaichamy, Kenneth R. Durbin, John F. Kellie, and Adam D. Catherman
- Subjects
Protein mass spectrometry ,Proteome ,Analytical chemistry ,Fractionation ,Chemical Fractionation ,Mass spectrometry ,Top-down proteomics ,Ion cyclotron resonance spectrometry ,Tandem mass spectrometry ,01 natural sciences ,Fourier transform ion cyclotron resonance ,Article ,Specimen Handling ,03 medical and health sciences ,Structural Biology ,Spectroscopy, Fourier Transform Infrared ,Biomarkers, Tumor ,Humans ,Electrophoresis, Gel, Two-Dimensional ,Spectroscopy ,030304 developmental biology ,0303 health sciences ,Chromatography ,Chemistry ,010401 analytical chemistry ,0104 chemical sciences ,Neoplasm Proteins ,HeLa Cells - Abstract
For fractionation of intact proteins by molecular weight (MW), a sharply improved two-dimensional (2D) separation is presented to drive reproducible and robust fractionation before top-down mass spectrometry of complex mixtures. The “GELFrEE” (i.e., gel-eluted liquid fraction entrapment electrophoresis) approach is implemented by use of Tris-glycine and Tris-tricine gel systems applied to human cytosolic and nuclear extracts from HeLa S3 cells, to achieve a MW-based fractionation of proteins from 5 to >100 kDa in 1 h. For top-down tandem mass spectroscopy (MS/MS) of the low-mass proteome (5–25 kDa), between 5 and 8 gel-elution (GE) fractions are sampled by nanocapillary-LC-MS/MS with 12 or 14.5 tesla Fourier transform ion cyclotron resonance (FT-ICR) mass spectrometers. Single injections give about 40 detectable proteins, about half of which yield automated ProSight identifications. Reproducibility metrics of the system are presented, along with comparative analysis of protein targets in mitotic versus asynchronous cells. We forward this basic 2D approach to facilitate wider implementation of top-down mass spectrometry and a variety of other protein separation and/or characterization approaches.
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.