1
|
Sexton D, Faucette R, Rivera-Hernandez M, Kenniston JA, Papaioannou N, Cosic J, Kopacz K, Salmon G, Beauchemin C, Juethner S, Yeung D. A novel assay of excess plasma kallikrein-kinin system activation in hereditary angioedema. FRONTIERS IN ALLERGY 2024; 5:1436855. [PMID: 39391687 PMCID: PMC11464748 DOI: 10.3389/falgy.2024.1436855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/20/2024] [Indexed: 10/12/2024] Open
Abstract
Background Cleaved high-molecular-weight kininogen (HKa) is a disease state biomarker of kallikrein-kinin system (KKS) activation in patients with hereditary angioedema due to C1 inhibitor deficiency (HAE-C1INH), the endogenous inhibitor of plasma kallikrein (PKa). Objective Develop an HKa-specific enzyme-linked immunosorbent assay (ELISA) to monitor KKS activation in the plasma of HAE-C1INH patients. Methods A novel HKa-specific antibody was discovered by antibody phage display and used as a capture reagent to develop an HKa-specific ELISA. Results Specific HKa detection following KKS activation was observed in plasma from healthy controls but not in prekallikrein-, high-molecular-weight kininogen-, or coagulation factor XII (FXII)-deficient plasma. HKa levels in plasma collected from HAE-C1INH patients in a disease quiescent state were higher than in plasma from healthy controls and increased further in HAE-C1INH plasma collected during an angioedema attack. The specificity of the assay for PKa-mediated HKa generation in minimally diluted plasma activated with exogenous FXIIa was demonstrated using a specific monoclonal antibody inhibitor (lanadelumab, IC50 = 0.044 µM). Conclusions An ELISA was developed for the specific and quantitative detection of HKa in human plasma to support HAE-C1INH drug development. Improved quantification of the HKa biomarker may facilitate further pathophysiologic insight into HAE-C1INH and other diseases mediated by a dysregulated KKS and may enable the design of highly potent inhibitors targeting this pathway.
Collapse
Affiliation(s)
- Dan Sexton
- Takeda Development Center Americas Inc., Cambridge, MA, United States
| | - Ryan Faucette
- Takeda Development Center Americas Inc., Cambridge, MA, United States
| | | | - Jon A. Kenniston
- Takeda Development Center Americas Inc., Cambridge, MA, United States
| | | | - Janja Cosic
- Takeda Development Center Americas Inc., Cambridge, MA, United States
| | - Kris Kopacz
- Takeda Development Center Americas Inc., Cambridge, MA, United States
| | - Gary Salmon
- Charles River Laboratories, Harlow, United Kingdom
| | | | - Salomé Juethner
- Takeda Pharmaceuticals USA, Inc., Lexington, MA, United States
| | - Dave Yeung
- Takeda Development Center Americas Inc., Cambridge, MA, United States
| |
Collapse
|
2
|
Sexton D, Nguyen HQ, Juethner S, Luo H, Zhang Z, Jasper P, Zhu AZX. A quantitative systems pharmacology model of plasma kallikrein-kinin system dysregulation in hereditary angioedema. J Pharmacokinet Pharmacodyn 2024:10.1007/s10928-024-09919-6. [PMID: 38734778 DOI: 10.1007/s10928-024-09919-6] [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: 11/03/2023] [Accepted: 04/09/2024] [Indexed: 05/13/2024]
Abstract
Hereditary angioedema (HAE) due to C1-inhibitor deficiency is a rare, debilitating, genetic disorder characterized by recurrent, unpredictable, attacks of edema. The clinical symptoms of HAE arise from excess bradykinin generation due to dysregulation of the plasma kallikrein-kinin system (KKS). A quantitative systems pharmacology (QSP) model that mechanistically describes the KKS and its role in HAE pathophysiology was developed based on HAE attacks being triggered by autoactivation of factor XII (FXII) to activated FXII (FXIIa), resulting in kallikrein production from prekallikrein. A base pharmacodynamic model was constructed and parameterized from literature data and ex vivo assays measuring inhibition of kallikrein activity in plasma of HAE patients or healthy volunteers who received lanadelumab. HAE attacks were simulated using a virtual patient population, with attacks recorded when systemic bradykinin levels exceeded 20 pM. The model was validated by comparing the simulations to observations from lanadelumab and plasma-derived C1-inhibitor clinical trials. The model was then applied to analyze the impact of nonadherence to a daily oral preventive therapy; simulations showed a correlation between the number of missed doses per month and reduced drug effectiveness. The impact of reducing lanadelumab dosing frequency from 300 mg every 2 weeks (Q2W) to every 4 weeks (Q4W) was also examined and showed that while attack rates with Q4W dosing were substantially reduced, the extent of reduction was greater with Q2W dosing. Overall, the QSP model showed good agreement with clinical data and could be used for hypothesis testing and outcome predictions.
Collapse
Affiliation(s)
- Dan Sexton
- Takeda Development Center Americas, Inc., Lexington, MA, USA
| | - Hoa Q Nguyen
- Takeda Development Center Americas, Inc., Lexington, MA, USA
| | - Salomé Juethner
- Takeda Development Center Americas, Inc., Lexington, MA, USA
| | | | | | | | - Andy Z X Zhu
- Takeda Development Center Americas, Inc., Lexington, MA, USA.
- Preclinical and Translational Science Department, Takeda Pharmaceutical Company Limited, 35 Landsdowne Street, Cambridge, MA, 02139, USA.
| |
Collapse
|
3
|
Zuraw BL, Maurer M, Sexton DJ, Cicardi M. Therapeutic monoclonal antibodies with a focus on hereditary angioedema. Allergol Int 2023; 72:54-62. [PMID: 35787344 DOI: 10.1016/j.alit.2022.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 05/03/2022] [Accepted: 05/18/2022] [Indexed: 01/25/2023] Open
Abstract
Monoclonal antibodies (mAbs) have been shown to be effective and generally safe across a continually expanding list of therapeutic areas. We describe the advantages and limitations of mAbs as a therapeutic option compared with small molecules. Specifically, we discuss a novel mAb in the treatment of hereditary angioedema (HAE), a rare and potentially life-threatening condition characterized by recurrent unpredictable swelling attacks. HAE is mediated by dysregulation of plasma kallikrein activity leading to overproduction of bradykinin. Current prophylactic treatment for HAE includes androgens or replacement of the endogenous plasma kallikrein inhibitor, C1 inhibitor. However, there remains an unmet need for an effective, less burdensome treatment option. Lanadelumab is a fully human mAb targeting plasma kallikrein. Results from clinical trials, including a pivotal Phase 3 study and its ensuing open-label extension study, demonstrated that lanadelumab is associated with few treatment-related adverse events and reduced the rate of HAE attacks. This novel treatment option has the potential to significantly improve the lives of patients with HAE.
Collapse
Affiliation(s)
- Bruce L Zuraw
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, CA, USA.
| | - Marcus Maurer
- Institute of Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | | | - Marco Cicardi
- Department of Biomedical and Clinical Sciences, Luigi Sacco, University of Milan, IRCCS ICS Maugeri, Milano, Italy
| |
Collapse
|
4
|
Marceau F, Rivard GE, Hébert J, Gauthier J, Bachelard H, Gangnus T, Burckhardt BB. Picomolar Sensitivity Analysis of Multiple Bradykinin-Related Peptides in the Blood Plasma of Patients With Hereditary Angioedema in Remission: A Pilot Study. FRONTIERS IN ALLERGY 2022; 3:837463. [PMID: 35386662 PMCID: PMC8974669 DOI: 10.3389/falgy.2022.837463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/11/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundHereditary angioedema (HAE) is a rare autosomal dominant disease; the most well understood forms concern the haplodeficiency of C1 esterase inhibitor (C1INH) and a gain of function mutation of factor XII (FXII). The acute forms of these conditions are mediated by an excessive bradykinin (BK) formation by plasma kallikrein.MethodsA validated LC-MS/MS platform of picomolar sensitivity developed for the analysis of eleven bradykinin-related peptides was applied to the plasma of HAE-C1INH and HAE-FXII sampled during remission.ResultsIn HAE-C1INH plasma, the concentrations of the relatively stable BK1−5 fragment (mean ± S.E.M.: 12.0 ± 4.2 pmol/L), of BK2−9 (0.7 ± 0.2 pmol/L) and of the sums of BK and its tested fragments (18.0 ± 6.4 pmol/L) are significantly greater than those recorded in the plasma of healthy volunteers (1.9 ± 0.6, 0.03 ± 0.03 and 4.3 ± 0.8 pmol/L, respectively), consistent with the previous evidence of permanent plasma kallikrein activity in this disease. Kinin levels in the plasma of HAE-FXII patients did not differ from controls, suggesting that triggering factors for contact system activation are not active during remission.ConclusionBK1−5, BK2−9 and the sum of BK and its fragments determined by the sensitive LC-MS/MS technique are proposed as potential biomarkers of HAE-C1INH in remission while this was not applicable to HAE-FXII patients.
Collapse
Affiliation(s)
- François Marceau
- Axe Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- *Correspondence: François Marceau
| | - Georges-Etienne Rivard
- Division of Hematology/Oncology, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Jacques Hébert
- Service d'allergie, CHU de Québec-Université Laval, Québec, QC, Canada
| | - Julie Gauthier
- Molecular Diagnostic Laboratory, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, QC, Canada
- Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Hélène Bachelard
- Axe Endocrinologie et Néphrologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Tanja Gangnus
- Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany
| | - Bjoern B. Burckhardt
- Institute of Clinical Pharmacy and Pharmacotherapy, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
5
|
Grumach AS, Veronez CL, Csuka D, Farkas H. Angioedema Without Wheals: Challenges in Laboratorial Diagnosis. Front Immunol 2021; 12:785736. [PMID: 34956216 PMCID: PMC8694242 DOI: 10.3389/fimmu.2021.785736] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/03/2021] [Indexed: 11/13/2022] Open
Abstract
Angioedema is a prevailing symptom in different diseases, frequently occurring in the presence of urticaria. Recurrent angioedema without urticaria (AE) can be hereditary (HAE) and acquired (AAE), and several subtypes can be distinguished, although clinical presentation is quite similar in some of them. They present with subcutaneous and mucosal swellings, affecting extremities, face, genitals, bowels, and upper airways. AE is commonly misdiagnosed due to restricted access and availability of appropriate laboratorial tests. HAE with C1 inhibitor defect is associated with quantitative and/or functional deficiency. Although bradykinin-mediated disease results mainly from disturbance in the kallikrein–kinin system, traditionally complement evaluation has been used for diagnosis. Diagnosis is established by nephelometry, turbidimetry, or radial immunodiffusion for quantitative measurement of C1 inhibitor, and chromogenic assay or ELISA has been used for functional C1-INH analysis. Wrong handling of the samples can lead to misdiagnosis and, consequently, mistaken inappropriate approaches. Dried blood spot (DBS) tests have been used for decades in newborn screening for certain metabolic diseases, and there has been growing interest in their use for other congenital conditions. Recently, DBS is now proposed as an efficient tool to diagnose HAE with C1 inhibitor deficiency, and its use would improve the access to outbound areas and family members. Regarding HAE with normal C1 inhibitor, complement assays’ results are normal and the genetic sequencing of target genes, such as exon 9 of F12 and PLG, is the only available method. New methods to measure cleaved high-molecular-weight kininogen and activated plasma kallikrein have emerged as potential biochemical tests to identify bradykinin-mediated angioedema. Validated biomarkers of kallikrein–kinin system activation could be helpful in differentiating mechanisms of angioedema. Our aim is to focus on the capability to differentiate histaminergic AE from bradykinin-mediated AE. In addition, we will describe the challenges developing specific tests like direct bradykinin measurements. The need for quality tests to improve the diagnosis is well represented by the variability of results in functional assays.
Collapse
Affiliation(s)
- Anete S Grumach
- Clinical Immunology, Faculdade de Medicina, Centro Universitario Faculdade de Medicina ABC (FMABC), Santo Andre, Brazil
| | - Camila L Veronez
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, San Diego, CA, United States.,Research Service, San Diego Veterans Affairs Healthcare, San Diego, CA, United States
| | - Dorottya Csuka
- Hungarian Angioedema Center of Reference and Excellence, Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
| | - Henriette Farkas
- Hungarian Angioedema Center of Reference and Excellence, Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
6
|
Förster TM, Magerl M, Maurer M, Zülbahar S, Zielke S, Inhaber N, Crocetta D, Rolfs A, Skrahina V. HAE patient self-sampling for biomarker establishment. Orphanet J Rare Dis 2021; 16:399. [PMID: 34583739 PMCID: PMC8478266 DOI: 10.1186/s13023-021-02021-x] [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: 02/22/2021] [Accepted: 09/20/2021] [Indexed: 11/30/2022] Open
Abstract
Background Hereditary Angioedema (HAE) is a genetic disorder that leads to frequent angioedema attacks in various parts of the body. In most cases it is caused by pathogenic variants in the SERPING1 gene, coding for C1-Inhibitor (C1-INH). The pathogenic variants in the gene result in reduced C1-INH levels and/or activity, which causes aberrant bradykinin production and enhanced vascular permeability. The standard-of-care diagnostic test is performed biochemically via measuring C1-INH level and activity as well as the C4 level. This, however, does not allow for the diagnosis of HAE types with normal C1-INH. There is an urgent need to identify and characterize HAE biomarkers for facilitating diagnostics and personalizing the treatment. The Hereditary Angioedema Kininogen Assay (HAEKA) study aims to measure the dynamics of cleaved High Molecular Weight Kininogen (HKa) and other metabolite levels during the angioedema and non-angioedema state of the disease. The metabolites will be analyzed and verified by liquid chromatography ion mobility high resolution mass spectrometry (LC/IM-QToF MS) of dried blood spot (DBS) cards upon the study completion. The study design is truly innovative: 100 enrolled participants provide blood samples via DBS: (1) every 3 months within 2 years during regular study site visits and (2) by at-home self-sampling during HAE attacks via finger pricking. We are presenting a project design that permits clinical study activities during pandemic contact restrictions and opens the door for other clinical studies during COVID-19. Results As of October 2020, there are 41 patients from 5 sites in Germany enrolled. 90 blood samples were collected during the regular visits, and 19 of the participants also performed self-sampling during the HAE attacks from which a total of 286 attack blood samples were collected. Participating patients rate the study procedures as easy to implement in their daily lives. The concept of home self-sampling is effective, reproducible, and convenient especially in times of contact restrictions due to the COVID-19 pandemic. Conclusions It is the hope that the HAEKA study will complete in 2023, reveal biomarker(s) for monitoring HAE disease activity, and may help to avoid HAE attacks via applying medication prior to the symptom onset. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-02021-x.
Collapse
Affiliation(s)
- Toni M Förster
- CENTOGENE GmbH, Rostock, Germany. .,Arcensus GmbH, Goethestrasse 20, 18055, Rostock, Germany.
| | - Markus Magerl
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marcus Maurer
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | | | - Neil Inhaber
- Takeda Pharmaceutical Company Limited, Lexington, MA, USA
| | | | - Arndt Rolfs
- CENTOGENE GmbH, Rostock, Germany.,Universität Rostock, Medizinische Fakultät, Rostock, Germany.,Arcensus GmbH, Goethestrasse 20, 18055, Rostock, Germany
| | - Volha Skrahina
- CENTOGENE GmbH, Rostock, Germany.,Arcensus GmbH, Goethestrasse 20, 18055, Rostock, Germany
| |
Collapse
|
7
|
Porebski G, Kwitniewski M, Reshef A. Biomarkers in Hereditary Angioedema. Clin Rev Allergy Immunol 2021; 60:404-415. [PMID: 33560480 PMCID: PMC8272698 DOI: 10.1007/s12016-021-08845-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2021] [Indexed: 12/20/2022]
Abstract
A biomarker is a defined characteristic measured as an indicator of normal, biologic, pathogenic processes, or biological responses to an exposure or intervention. Diagnostic biomarkers are used to detect a disease or a subtype of a disease; monitoring biomarkers are measured serially to assess a medical condition; response biomarkers are used to check biologic response following a medical intervention; predictive biomarkers are used to identify patients who are more likely to respond to a medical intervention; and prognostic biomarkers are used to assess the future likelihood of a clinical event. Although biomarkers have been extensively investigated and validated in many diseases and pathologies, very few are currently useful for the diagnosis, evaluation of disease activity, and treatment of hereditary angioedema (HAE). Pathophysiologic pathways involved in HAE reveal a plethora of molecules from the complement, coagulation, and fibrinolysis systems or from the vascular endothelium, which may serve as biomarkers. The most promising candidates, together with their laboratory readout systems, should be evaluated with regard to their analytical and clinical validity and utility. To be highly specific, such biomarkers should be linked to the pathomechanisms of HAE, particularly the bradykinin-generating cascade. Additionally, major advances in high-throughput omics-based technologies may facilitate the discovery of new candidate biomarkers in the future. This review will cover the existing as well as future potential biomarkers that will support the diagnosis, monitor disease activity, and can be used to assess the efficacy of new avenues of therapy of HAE and other forms of angioedema.
Collapse
Affiliation(s)
- Grzegorz Porebski
- Department of Clinical and Environmental Allergology, Jagiellonian University Medical College, Krakow, Poland.
| | - Mateusz Kwitniewski
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Avner Reshef
- Barzilai University Medical Centre, Ashkelon, Israel
| |
Collapse
|
8
|
Lai Y, Zhang G, Inhaber N, Bernstein JA, Cwik M, Zhou Z, Chockalingam PS, Wu J. A robust multiplexed assay to quantify C1-inhibitor, C1q, and C4 proteins for in vitro diagnosis of hereditary angioedema from dried blood spot. J Pharm Biomed Anal 2020; 195:113844. [PMID: 33388640 DOI: 10.1016/j.jpba.2020.113844] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 10/22/2022]
Abstract
Hereditary angioedema (HAE) is a rare genetic disease caused by deficiency or dysfunction of C1 esterase inhibitor (C1-INH). Plasma C1-INH activity and concentrations of C1-INH and complement components 1q and 4 (C1q, C4) are critical to the HAE diagnosis. We describe a novel multiplexed assay to simultaneously measure C1-INH, C1q, and C4 levels in dried blood spot (DBS) of HAE patients. The blood proteins were extracted from 3 mm punches of DBS samples and were subsequently digested by trypsin. The signature peptide derived from each protein was quantified by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Analyte-depleted blood was generated as a surrogate matrix for the preparation of calibration curves to overcome the interference of endogenous proteins, and the assay reproducibility was further monitored by assessing the signal of plasma transferrin as a house-keeping protein. The assay was fully validated following regulatory guideline, with a quantification range of 12.5-800 μg/mL for C1-INH and C4 and 3.13-200 μg/mL for C1q. The precision and accuracy ranged from 3.3%-9.8% and -8.2%-12.6%, respectively. All the patient samples exhibited C1-INH levels lower than normal range except the Type II patient and the C4 and C1q concentrations were as expected. Results from the DBS-based LC-MS assay were highly correlated with the ELISA data measured in plasma of the same subjects. The method described here offers unique advantages such as less invasive sampling, minimal blood processing, and easy transportation and sample storage, allowing, for the first time, C1-INH, C4, and C1q levels to be simultaneously determined in a drop of dried blood.
Collapse
Affiliation(s)
- Yongquan Lai
- Clinical Biomarker Innovation and Development, Takeda Pharmaceuticals International Co., 125 Binney Street, Cambridge, MA, USA
| | - Guodong Zhang
- Clinical Biomarker Innovation and Development, Takeda Pharmaceuticals International Co., 125 Binney Street, Cambridge, MA, USA
| | - Neil Inhaber
- Global Medical Affairs, Takeda Pharmaceuticals International Co., 300 Shire Way, Lexington, MA, USA
| | - Jonathan A Bernstein
- Department of Internal Medicine, Division of Immunology, University of Cincinnati College of Medicine, Bernstein Allergy Group and Bernstein Clinical Research Center, Cincinnati, OH, USA
| | - Michael Cwik
- Clinical Biomarker Innovation and Development, Takeda Pharmaceuticals International Co., 125 Binney Street, Cambridge, MA, USA
| | - Zhiwei Zhou
- Clinical Biomarker Innovation and Development, Takeda Pharmaceuticals International Co., 125 Binney Street, Cambridge, MA, USA
| | - Priya S Chockalingam
- Clinical Biomarker Innovation and Development, Takeda Pharmaceuticals International Co., 125 Binney Street, Cambridge, MA, USA
| | - Jiang Wu
- Clinical Biomarker Innovation and Development, Takeda Pharmaceuticals International Co., 125 Binney Street, Cambridge, MA, USA.
| |
Collapse
|
9
|
Angioedema without urticaria: novel findings which must be measured in clinical setting. Curr Opin Allergy Clin Immunol 2020; 20:253-260. [DOI: 10.1097/aci.0000000000000633] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
10
|
Lai Y, Zhang G, Zhou Z, Inhaber N, Bernstein JA, Chockalingam PS, Wu J. A novel functional C1 inhibitor activity assay in dried blood spot for diagnosis of Hereditary angioedema. Clin Chim Acta 2020; 504:155-162. [PMID: 32059947 DOI: 10.1016/j.cca.2020.02.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/29/2020] [Accepted: 02/11/2020] [Indexed: 10/25/2022]
Abstract
BACKGROUND Hereditary angioedema (HAE) is a rare genetic disease caused by deficiency or dysfunction of C1 esterase inhibitor (C1-INH). Timely and accurate diagnosis is an ongoing challenge. Measurement of plasma C1-INH activity is currently the critical standard test. We describe a novel and highly robust point-of-care assay to quantify C1-INH activity in dried blood spot (DBS). METHODS C1-INH was extracted from 3 mm punches of DBS samples and incubated with excess amount of C1 esterase (C1s). The mixture was subsequentially incubated with C1s substrate, followed by liquid chromatography-tandem mass spectrometry (LC-MS/MS) quantitation of the enzyme reaction product. RESULTS The assay was validated within a quantification range from 100 to 1500 mU/mL. The intra-day precision and accuracy ranged from 4.0% to 11.6% and -11.1% to -2.1%, and the inter-day precision and accuracy were 8.1-13.1% and -10.3% to 0.9%, respectively. Normal C1-INH activity (n = 103) ranged from 311 to 1090 mU/mL, whereas 23 out of 24 HAE patients exhibited C1-INH activity lower than 100 mU/mL. CONCLUSION DBS specimen collection for measurement of functional C1-INH activity in a physician's office is straightforward and not limited by logistic considerations and therefore, appropriate for the diagnosis of HAE in high throughput diagnostic laboratories.
Collapse
Affiliation(s)
- Yongquan Lai
- Clinical Biomarker Innovation and Development, Takeda Pharmaceuticals International Co., 125 Binney Street, Cambridge, MA, USA
| | - Guodong Zhang
- Clinical Biomarker Innovation and Development, Takeda Pharmaceuticals International Co., 125 Binney Street, Cambridge, MA, USA
| | - Zhiwei Zhou
- Clinical Biomarker Innovation and Development, Takeda Pharmaceuticals International Co., 125 Binney Street, Cambridge, MA, USA
| | - Neil Inhaber
- Global Medical Affairs, Takeda Pharmaceuticals International Co., 300 Shire Way, Lexington, MA, USA
| | - Jonathan A Bernstein
- Department of Internal Medicine, Division of Immunology, University of Cincinnati College of Medicine, Bernstein Allergy Group and Bernstein Clinical Research Center, Cincinnati, OH, USA
| | - Priya S Chockalingam
- Clinical Biomarker Innovation and Development, Takeda Pharmaceuticals International Co., 125 Binney Street, Cambridge, MA, USA
| | - Jiang Wu
- Clinical Biomarker Innovation and Development, Takeda Pharmaceuticals International Co., 125 Binney Street, Cambridge, MA, USA.
| |
Collapse
|
11
|
A sensitive antibody-free 2D-LC–MS/MS assay for the quantitation of myostatin in the serum of different species. Bioanalysis 2019; 11:957-970. [DOI: 10.4155/bio-2018-0311] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Aim: Myostatin (MSTN) is an attractive therapeutic target for the treatment of muscle degeneration-related diseases and is being evaluated as a target engagement biomarker. Methods: A sensitive 2D-LC–MS/MS assay was developed to quantify MSTN in different animal species. Sample preparation involved SDS denaturation of serum proteins followed by tryptic digestion and peptide enrichment by SPE. Results: The assay was validated with LLOQ of 2.5 ng/ml in rat and monkey serum. The precision was within 13.7%, and the bias was within ±12.6% for all quality control samples in authentic matrices. Conclusion: This new assay was successfully applied to measure MSTN in mouse, rat, monkey and human serum. The total MSTN in rat and monkey serum was elevated following administration of an MSTN inhibitor.
Collapse
|
12
|
Protein quantification by LC–MS: a decade of progress through the pages of Bioanalysis. Bioanalysis 2019; 11:629-644. [DOI: 10.4155/bio-2019-0032] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Over the past 10 years, there has been a remarkable increase in the use of LC–MS for the quantitative determination of proteins, and this technique can now be considered an established bioanalytical platform for the quantification of macromolecular drugs and biomarkers, next to the traditional ligand-binding assays. Many researchers have contributed to the field and helped improve both the technical possibilities of LC–MS-based workflows and our understanding of the meaning of the results that are obtained. As a tribute to Bioanalysis, which has published many important contributions, this report gives a high-level overview of the most important trends in the field of protein LC–MS, as published in this journal since its inauguration a decade ago. It describes the major technical developments with regard to sample handling, separation and MS detection of both digested and intact protein analysis. In addition, the relevance of the complex structure and in vivo behavior of proteins is discussed and the effect of protein–protein interactions, biotransformation and the occurrence of isoforms on the analytical result is addressed.
Collapse
|
13
|
An LC–MS/MS approach to assess total and free protein target in the serum of cynomolgus monkey. Bioanalysis 2019; 11:393-406. [DOI: 10.4155/bio-2018-0294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: Develop LC–MS/MS-based assays to measure total and free complement C5 in cynomolgus monkey serum as a target engagement biomarker for pharmacokinetic/pharmacodynamic correlation study. Materials & methods/results: The C5-specific signature peptide derived from pellet digestion of serum proteins with and without prior immunodepletion of the drug-bound C5 by protein A beads was quantified to assess free and total C5 levels, respectively. Conditions for immunodepletion by protein A were optimized to ensure complete depletion of IgGs (and drug-bound C5). The effect of sample dilution on drug-target dissociation and thus free C5 measurement was evaluated by applying a mathematical simulation. Conclusion: The procedure described here allows for the assessment of protein target engagement, aiding in pharmacokinetic/pharmacodynamic correlation analysis and human dose projection.
Collapse
|
14
|
2018 White Paper on Recent Issues in Bioanalysis: focus on immunogenicity assays by hybrid LBA/LCMS and regulatory feedback (Part 2 - PK, PD & ADA assays by hybrid LBA/LCMS & regulatory agencies' inputs on bioanalysis, biomarkers and immunogenicity). Bioanalysis 2018; 10:1897-1917. [PMID: 30488729 DOI: 10.4155/bio-2018-0285] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The 2018 12th Workshop on Recent Issues in Bioanalysis took place in Philadelphia, PA, USA on April 9-13, 2018 with an attendance of over 900 representatives from pharmaceutical/biopharmaceutical companies, biotechnology companies, contract research organizations and regulatory agencies worldwide. WRIB was once again a 5-day, week-long event - a full immersion week of bioanalysis, biomarkers and immunogenicity. As usual, it was specifically designed to facilitate sharing, reviewing, discussing and agreeing on approaches to address the most current issues of interest including both small- and large-molecule bioanalysis involving LCMS, hybrid LBA/LCMS and LBA/cell-based assays approaches. This 2018 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 2018 edition of this comprehensive White Paper has been divided into three parts for editorial reasons. This publication (Part 2) covers the recommendations for PK, PD and ADA assays by hybrid LBA/LCMS and regulatory agencies' input. Part 1 (LCMS for small molecules, peptides, oligonucleotides and small molecule biomarkers) and Part 3 (LBA/cell-based assays: immunogenicity, biomarkers and PK assays) are published in volume 10 of Bioanalysis, issues 22 and 24 (2018), respectively.
Collapse
|
15
|
Selectivity for quantitation of biomarkers using liquid chromatography and mass spectrometry. Bioanalysis 2018; 10:1461-1465. [DOI: 10.4155/bio-2018-0148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
16
|
Hubers SA, Kohm K, Wei S, Yu C, Nian H, Grabert R, Sexton DJ, Brown NJ. Endogenous bradykinin and B1-B5 during angiotensin-converting enzyme inhibitor-associated angioedema. J Allergy Clin Immunol 2018; 142:1636-1639.e5. [PMID: 30036596 DOI: 10.1016/j.jaci.2018.06.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 06/01/2018] [Accepted: 06/15/2018] [Indexed: 11/19/2022]
Affiliation(s)
- Scott A Hubers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn
| | - Kevin Kohm
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn
| | - Shouzuo Wei
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn
| | - Chang Yu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tenn
| | - Hui Nian
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tenn
| | - Ryan Grabert
- Global Nonclinical Development, Shire, Lexington, Mass
| | | | - Nancy J Brown
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tenn.
| |
Collapse
|