1
|
Blume SY, Garg A, Martí-Mateos Y, Midha AD, Chew BTL, Lin B, Yu C, Dick R, Lee PS, Situ E, Sarwaikar R, Green E, Ramanan V, Grotenbreg G, Hoek M, Sinz C, Jain IH. HypoxyStat, a small-molecule form of hypoxia therapy that increases oxygen-hemoglobin affinity. Cell 2025; 188:1580-1588.e11. [PMID: 39965572 DOI: 10.1016/j.cell.2025.01.029] [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: 06/30/2023] [Revised: 12/11/2024] [Accepted: 01/19/2025] [Indexed: 02/20/2025]
Abstract
We have previously demonstrated that chronic inhaled hypoxia is remarkably therapeutic in the premier animal model of mitochondrial Leigh syndrome, the Ndufs4 knockout (KO) mouse. Subsequent work has extended this finding to additional mitochondrial diseases and more common conditions. However, challenges inherent to gas-based therapies have hindered the rapid translation of our findings to the clinic. Here, we tested a small molecule (hereafter termed HypoxyStat) that increases the binding affinity of hemoglobin for oxygen, thereby decreasing oxygen offloading to tissues. Daily oral dosing of HypoxyStat caused systemic hypoxia in mice breathing normoxic (21% O2) air. When administered prior to disease onset, this treatment dramatically extended the lifespan of Ndufs4 KO mice and rescued additional aspects of disease, including behavior, body weight, neuropathology, and body temperature. HypoxyStat was also able to reverse disease at a very late stage, thereby serving as a clinically tractable form of hypoxia therapy.
Collapse
Affiliation(s)
- Skyler Y Blume
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ankur Garg
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yolanda Martí-Mateos
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ayush D Midha
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Brandon T L Chew
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Baiwei Lin
- Maze Therapeutics, 171 Oyster Point Blvd STE 300, South San Francisco, CA 94080, USA
| | - Cecile Yu
- Maze Therapeutics, 171 Oyster Point Blvd STE 300, South San Francisco, CA 94080, USA
| | - Ryan Dick
- Maze Therapeutics, 171 Oyster Point Blvd STE 300, South San Francisco, CA 94080, USA
| | - Patrick S Lee
- Maze Therapeutics, 171 Oyster Point Blvd STE 300, South San Francisco, CA 94080, USA
| | - Eva Situ
- Maze Therapeutics, 171 Oyster Point Blvd STE 300, South San Francisco, CA 94080, USA
| | - Richa Sarwaikar
- Maze Therapeutics, 171 Oyster Point Blvd STE 300, South San Francisco, CA 94080, USA
| | - Eric Green
- Maze Therapeutics, 171 Oyster Point Blvd STE 300, South San Francisco, CA 94080, USA
| | - Vyas Ramanan
- Maze Therapeutics, 171 Oyster Point Blvd STE 300, South San Francisco, CA 94080, USA
| | - Gijsbert Grotenbreg
- Maze Therapeutics, 171 Oyster Point Blvd STE 300, South San Francisco, CA 94080, USA
| | - Maarten Hoek
- Maze Therapeutics, 171 Oyster Point Blvd STE 300, South San Francisco, CA 94080, USA
| | - Christopher Sinz
- Maze Therapeutics, 171 Oyster Point Blvd STE 300, South San Francisco, CA 94080, USA
| | - Isha H Jain
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Arc Institute, 3181 Porter Dr, Palo Alto, CA 94304, USA.
| |
Collapse
|
2
|
Patel ZV, Prajjwal P, Bethineedi LD, Patel DJ, Khullar K, Patel H, Khatri K, Marsool MDM, Gadam S, Aleti S, Amir O. Newer Modalities and Updates in the Management of Sickle Cell Disease: A Systematic Review. J Blood Med 2024; 15:435-447. [PMID: 39286637 PMCID: PMC11404495 DOI: 10.2147/jbm.s477507] [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: 06/04/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024] Open
Abstract
Sickle cell disease (SCD), the most common autosomal recessive genetic disorder, affects the hemoglobin (Hb) chains in human red blood cells. It is caused by mutations in the β-globin genes, leading to the production of hemoglobin S, which results in the formation of sickle-shaped red blood cells (RBCs). These abnormal cells cause hemolysis, endothelial damage, and small vessel occlusion, leading to both acute and long-term complications. According to the World Health Organization's 2008 estimates, SCD affects approximately 2.28 per 1000 individuals globally. Despite this high prevalence, therapeutic advancements have been slow. For many years, the only FDA-approved medications for managing SCD complications were hydroxyurea and deferiprone. However, recent years have seen the approval of several new therapies, including L-glutamine (2017), voxelotor and crizanlizumab (2019), as well as exagamglogene autotemcel (Casgevy) and lovotibeglogene autotemcel (Lyfgenia) (2023). These treatments have proven effective in managing both the acute and chronic effects of SCD, including hemolytic anemia, chronic pain, stroke, vaso-occlusive crises, and multiple organ damage syndromes. This review explores the mechanisms of action, practical considerations, and side effects of these emerging therapies, drawing from a comprehensive search of databases such as PubMed, Medline, and Cochrane.
Collapse
Affiliation(s)
| | | | | | - Divyakshi J Patel
- Internal Medicine, Smt. NHL Municipal Medical College, Ahmedabad, India
| | - Kaarvi Khullar
- Internal Medicine, Maharashtra University of Health Sciences, Government Medical College, Gondia, India
| | - Hinal Patel
- Internal Medicine, GMERS Medical College and Hospital Sola, Ahmedabad, India
| | - Kanishka Khatri
- Internal Medicine, Bharati Vidyapeeth University Medical College, Pune, India
| | | | - Srikanth Gadam
- Internal Medicine, NYC Health+ Hospitals, New York, NYC, USA
| | - Soumya Aleti
- Internal Medicine, Berkshire Medical Center, Pittsfield, MA, USA
| | - Omniat Amir
- Internal Medicine, Al Manhal Academy of Science, Sudan
| |
Collapse
|
3
|
Shu P, You G, Li W, Chen Y, Chu Z, Qin D, Wang Y, Zhou H, Zhao L. Cefmetazole sodium as an allosteric effector that regulates the oxygen supply efficiency of adult hemoglobin. J Biomol Struct Dyn 2024; 42:7442-7456. [PMID: 37555593 DOI: 10.1080/07391102.2023.2245043] [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: 03/15/2023] [Accepted: 07/17/2023] [Indexed: 08/10/2023]
Abstract
Allosteric effectors play an important role in regulating the oxygen supply efficiency of hemoglobin for blood storage and disease treatment. However, allosteric effectors that are approved by the US FDA are limited. In this study, cefmetazole sodium (CS) was found to bind adult hemoglobin (HbA) from FDA library (1338 compounds) using surface plasmon resonance imaging high-throughput screening. Using surface plasmon resonance (SPR), the interaction between CS and HbA was verified. The oxygen dissociation curve of HbA after CS interaction showed a significant increase in P50 and theoretical oxygen-release capacity. Acid-base sensitivity (SI) exhibited a decreasing trend, although not significantly different. An oxygen dissociation assay indicated that CS accelerated HbA deoxygenation. Microfluidic modulated spectroscopy showed that CS changed the ratio of the alpha-helix to the beta-sheet of HbA. Molecular docking suggested CS bound to HbA's β-chains via hydrogen bonds, with key amino acids being N282, K225, H545, K625, K675, and V544.The results of molecular dynamics simulations (MD) revealed a stable orientation of the HbA-CS complex. CS did not significantly affect the P50 of bovine hemoglobin, possibly due to the lack of Valβ1 and Hisβ2, indicating that these were the crucial amino acids involved in HbA's oxygen affinity. Competition between the 2,3-Diphosphoglycerate (2,3-DPG) and CS in the HbA interaction was also determined by SPR, molecular docking and MD. In summary, CS could interact with HbA and regulate the oxygen supply efficiency via forming stable hydrogen bonds with the β-chains of HbA, and showed competition with 2,3-DPG.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Peilin Shu
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| | - Guoxing You
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| | - Weidan Li
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| | - Yuzhi Chen
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| | - Zongtang Chu
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| | - Dong Qin
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| | - Ying Wang
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| | - Hong Zhou
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| | - Lian Zhao
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People's Liberation Army, Beijing, P.R. C
| |
Collapse
|
4
|
Gibson JS, Rees DC. Emerging drug targets for sickle cell disease: shedding light on new knowledge and advances at the molecular level. Expert Opin Ther Targets 2023; 27:133-149. [PMID: 36803179 DOI: 10.1080/14728222.2023.2179484] [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: 02/22/2023]
Abstract
INTRODUCTION In sickle cell disease (SCD), a single amino acid substitution at β6 of the hemoglobin (Hb) chain replaces glutamate with valine, forming HbS instead of the normal adult HbA. Loss of a negative charge, and the conformational change in deoxygenated HbS molecules, enables formation of HbS polymers. These not only distort red cell morphology but also have other profound effects so that this simple etiology belies a complex pathogenesis with multiple complications. Although SCD represents a common severe inherited disorder with life-long consequences, approved treatments remain inadequate. Hydroxyurea is currently the most effective, with a handful of newer treatments, but there remains a real need for novel, efficacious therapies. AREAS COVERED This review summarizes important early events in pathogenesis to highlight key targets for novel treatments. EXPERT OPINION A thorough understanding of early events in pathogenesis closely associated with the presence of HbS is the logical starting point for identification of new targets rather than concentrating on more downstream effects. We discuss ways of reducing HbS levels, reducing the impact of HbS polymers, and of membrane events perturbing cell function, and suggest using the unique permeability of sickle cells to target drugs specifically into those more severely compromised.
Collapse
Affiliation(s)
- John S Gibson
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - David C Rees
- Department of Paediatric Haematology, King's College Hospital, London, UK
| |
Collapse
|
5
|
Böning D, Kuebler WM, Vogel D, Bloch W. The oxygen dissociation curve of blood in COVID-19-An update. Front Med (Lausanne) 2023; 10:1098547. [PMID: 36923010 PMCID: PMC10008909 DOI: 10.3389/fmed.2023.1098547] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/03/2023] [Indexed: 03/02/2023] Open
Abstract
An impressive effect of the infection with SARS-Co-19 is the impairment of oxygen uptake due to lung injury. The reduced oxygen diffusion may potentially be counteracted by an increase in oxygen affinity of hemoglobin. However, hypoxia and anemia associated with COVID-19 usually decrease oxygen affinity due to a rise in [2,3-bisphosphoglycerate]. As such, COVID-19 related changes in the oxygen dissociation curve may be critical for oxygen uptake and supply, but are hard to predict. A Pubmed search lists 14 publications on oxygen affinity in COVID-19. While some investigations show no changes, three large studies found an increased affinity that was related to a good prognosis. Exact causes remain unknown. The cause of the associated anemia in COVID-19 is under discussion. Erythrocytes with structural alterations of membrane and cytoskeleton have been observed, and virus binding to Band 3 and also to ACE2 receptors in erythroblasts has been proposed. COVID-19 presentation is moderate in many subjects suffering from sickle cell disease. A possible explanation is that COVID-19 counteracts the unfavorable large right shift of the oxygen dissociation curve in these patients. Under discussion for therapy are mainly affinity-increasing drugs.
Collapse
Affiliation(s)
- Dieter Böning
- Institute of Physiology, Charité Medical University of Berlin, Berlin, Germany
| | - Wolfgang M. Kuebler
- Institute of Physiology, Charité Medical University of Berlin, Berlin, Germany
| | - Dominik Vogel
- Klinik für Interdisziplinäre Intensivmedizin, Vivantes Humboldt-Klinikum, Berlin, Germany
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Cologne, Germany
| |
Collapse
|
6
|
Zheng D, Li Y, Song L, Xu T, Jiang X, Yin X, He Y, Xu J, Ma X, Chai L, Xu J, Hu J, Mi P, Jing J, Shi H. Improvement of radiotherapy with an ozone-carried liposome nano-system for synergizing cancer immune checkpoint blockade. NANO TODAY 2022; 47:101675. [DOI: 10.1016/j.nantod.2022.101675] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
|
7
|
Design, Synthesis, and Investigation of Novel Nitric Oxide (NO)-Releasing Aromatic Aldehyde as Drug Candidates for the Treatment of Sickle Cell Disease. Molecules 2022; 27:molecules27206835. [PMID: 36296435 PMCID: PMC9610770 DOI: 10.3390/molecules27206835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 11/29/2022] Open
Abstract
Sickle cell disease (SCD) is caused by a single-point mutation, and the ensuing deoxygenation-induced polymerization of sickle hemoglobin (HbS), and reduction in bioavailability of vascular nitric oxide (NO), contribute to the pathogenesis of the disease. In a proof-of-concept study, we successfully incorporated nitrate ester groups onto two previously studied potent antisickling aromatic aldehydes, TD7 and VZHE039, to form TD7-NO and VZHE039-NO hybrids, respectively. These compounds are stable in buffer but demonstrated the expected release of NO in whole blood in vitro and in mice. The more promising VZHE039-NO retained the functional and antisickling activities of the parent VZHE039 molecule. Moreover, VZHE039-NO, unlike VZHE039, significantly attenuated RBC adhesion to laminin, suggesting this compound has potential in vivo RBC anti-adhesion properties relevant to vaso-occlusive events. Crystallographic studies show that, as with VZHE039, VZHE039-NO also binds to liganded Hb to make similar protein interactions. The knowledge gained during these investigations provides a unique opportunity to generate a superior candidate drug in SCD with enhanced benefits.
Collapse
|
8
|
Oknińska M, Mackiewicz U, Zajda K, Kieda C, Mączewski M. New potential treatment for cardiovascular disease through modulation of hemoglobin oxygen binding curve: Myo-inositol trispyrophosphate (ITPP), from cancer to cardiovascular disease. Biomed Pharmacother 2022; 154:113544. [PMID: 35988421 DOI: 10.1016/j.biopha.2022.113544] [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: 07/04/2022] [Revised: 08/03/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022] Open
Abstract
The human body is a highly aerobic organism, which needs large amount of oxygen, especially in tissues characterized by high metabolic demand, such as the heart. Inadequate oxygen delivery underlies cardiovascular diseases, such as coronary artery disease, heart failure and pulmonary hypertension. Hemoglobin, the oxygen-transport metalloprotein in the red blood cells, gives the blood enormous oxygen carrying capacity; thus oxygen binding to hemoglobin in the lungs and oxygen dissociation in the target tissues are crucial points for oxygen delivery as well as potential targets for intervention. Myo-inositol trispyrophosphate (ITPP) acts as an effector of hemoglobin, shifting the oxygen dissociation curve to the right and increasing oxygen release in the target tissues, especially under hypoxic conditions. ITPP has been successfully used in cancer studies, demonstrating anti-cancer properties due to prevention of tumor hypoxia. Currently it is being tested in phase 2 clinical trials in humans with various tumors. First preclinical evidence also indicates that it can successfully alleviate myocardial hypoxia and prevent adverse left ventricular and right ventricular remodeling in post-myocardial infarction heart failure and pulmonary hypertension. The aim of the article is to summarize the current knowledge on ITTP, as well as to determine the prospects for its potential use in the treatment of many cardiovascular disorders.
Collapse
Affiliation(s)
- Marta Oknińska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Urszula Mackiewicz
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Karolina Zajda
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
| | - Claudine Kieda
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland; Center for Molecular Biophysics, UPR 4301 CNRS, Orleans, France
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland.
| |
Collapse
|
9
|
Yu JJ, Non AL, Heinrich EC, Gu W, Alcock J, Moya EA, Lawrence ES, Tift MS, O'Brien KA, Storz JF, Signore AV, Khudyakov JI, Milsom WK, Wilson SM, Beall CM, Villafuerte FC, Stobdan T, Julian CG, Moore LG, Fuster MM, Stokes JA, Milner R, West JB, Zhang J, Shyy JY, Childebayeva A, Vázquez-Medina JP, Pham LV, Mesarwi OA, Hall JE, Cheviron ZA, Sieker J, Blood AB, Yuan JX, Scott GR, Rana BK, Ponganis PJ, Malhotra A, Powell FL, Simonson TS. Time Domains of Hypoxia Responses and -Omics Insights. Front Physiol 2022; 13:885295. [PMID: 36035495 PMCID: PMC9400701 DOI: 10.3389/fphys.2022.885295] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
The ability to respond rapidly to changes in oxygen tension is critical for many forms of life. Challenges to oxygen homeostasis, specifically in the contexts of evolutionary biology and biomedicine, provide important insights into mechanisms of hypoxia adaptation and tolerance. Here we synthesize findings across varying time domains of hypoxia in terms of oxygen delivery, ranging from early animal to modern human evolution and examine the potential impacts of environmental and clinical challenges through emerging multi-omics approaches. We discuss how diverse animal species have adapted to hypoxic environments, how humans vary in their responses to hypoxia (i.e., in the context of high-altitude exposure, cardiopulmonary disease, and sleep apnea), and how findings from each of these fields inform the other and lead to promising new directions in basic and clinical hypoxia research.
Collapse
Affiliation(s)
- James J. Yu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Amy L. Non
- Department of Anthropology, Division of Social Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Erica C. Heinrich
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, United States
| | - Wanjun Gu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
- Herbert Wertheim School of Public Health and Longevity Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Joe Alcock
- Department of Emergency Medicine, University of New Mexico, Albuquerque, MX, United States
| | - Esteban A. Moya
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Elijah S. Lawrence
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Michael S. Tift
- Department of Biology and Marine Biology, College of Arts and Sciences, University of North Carolina Wilmington, Wilmington, NC, United States
| | - Katie A. O'Brien
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
- Department of Physiology, Development and Neuroscience, Faculty of Biology, School of Biological Sciences, University of Cambridge, Cambridge, ENG, United Kingdom
| | - Jay F. Storz
- School of Biological Sciences, College of Arts and Sciences, University of Nebraska-Lincoln, Lincoln, IL, United States
| | - Anthony V. Signore
- School of Biological Sciences, College of Arts and Sciences, University of Nebraska-Lincoln, Lincoln, IL, United States
| | - Jane I. Khudyakov
- Department of Biological Sciences, University of the Pacific, Stockton, CA, United States
| | | | - Sean M. Wilson
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda, CA, United States
| | | | | | | | - Colleen G. Julian
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Lorna G. Moore
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, Aurora, CO, United States
| | - Mark M. Fuster
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Jennifer A. Stokes
- Department of Kinesiology, Southwestern University, Georgetown, TX, United States
| | - Richard Milner
- San Diego Biomedical Research Institute, San Diego, CA, United States
| | - John B. West
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Jiao Zhang
- Department of Medicine, UC San Diego School of Medicine, San Diego, CA, United States
| | - John Y. Shyy
- Department of Medicine, UC San Diego School of Medicine, San Diego, CA, United States
| | - Ainash Childebayeva
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - José Pablo Vázquez-Medina
- Department of Integrative Biology, College of Letters and Science, University of California, Berkeley, Berkeley, CA, United States
| | - Luu V. Pham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Omar A. Mesarwi
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - James E. Hall
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Zachary A. Cheviron
- Division of Biological Sciences, College of Humanities and Sciences, University of Montana, Missoula, MT, United States
| | - Jeremy Sieker
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Arlin B. Blood
- Department of Pediatrics Division of Neonatology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Jason X. Yuan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Graham R. Scott
- Department of Pediatrics Division of Neonatology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Brinda K. Rana
- Moores Cancer Center, UC San Diego, La Jolla, CA, United States
- Department of Psychiatry, UC San Diego, La Jolla, CA, United States
| | - Paul J. Ponganis
- Center for Marine Biotechnology and Biomedicine, La Jolla, CA, United States
| | - Atul Malhotra
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Frank L. Powell
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Tatum S. Simonson
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
10
|
Tarasev M, Ferranti M, Herppich A, Hines P. GBT1118, a voxelotor analog, protects red blood cells from damage during severe hypoxia. Am J Transl Res 2022; 14:240-251. [PMID: 35173841 PMCID: PMC8829590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 11/09/2021] [Indexed: 06/14/2023]
Abstract
A lack of objective metrics in Sickle Cell Disease (SCD) makes it difficult to assess individual patient therapy options or assess the effects of therapy. This is further complicated by mechanisms of action involving multiple interconnected effects, that combine to relieve SCD symptoms. In 2019, based on the increase in hemoglobin concentration observed in the HOPE trial, the Food and Drug Administration approved voxelotor (Oxbryta®, Global Blood Therapeutics) for SCD patients 12 years and older. The main mechanism of action for voxelotor was increased hemoglobin-oxygen affinity, but other mechanisms may apply. In this study, we assessed the effect of GBT1118, an Oxbryta analog, on hypoxia-induced lethal and sub-hemolytic red blood cell (RBC) membrane damage using RBC Mechanical Fragility (MF), a metric of existing membrane damage and prospective hemolysis. RBC MF was measured non-invasively using a proprietary system comprising an electromagnetic bead mill and fiberoptic spectrophotometry detection. Three cycles of severe hypoxia (<5% oxygenated hemoglobin) with follow-up reoxygenation resulted in a significant increase in RBC MF for all SCD (Hb-S >60%) samples. Supplementation with GBT1118 caused no significant changes in pre-hypoxia RBC MF. However, following GBT1118 treatment, cell stability showed significantly less degradation, as evidenced by a significantly smaller RBC MF increase after three cycles of hypoxia-reoxygenation. These findings indicate that GBT1118 prevents hypoxia-induced membrane damage in sickled RBC, in part by alternative mechanisms not associated with induced changes in hemoglobin-oxygen affinity.
Collapse
Affiliation(s)
| | | | | | - Patrick Hines
- Functional FluidicsDetroit, MI, USA
- Department of Pharmacology, Wayne State University School of MedicineDetroit, Michigan, MI, USA
| |
Collapse
|
11
|
Webb KL, Dominelli PB, Baker SE, Klassen SA, Joyner MJ, Senefeld JW, Wiggins CC. Influence of High Hemoglobin-Oxygen Affinity on Humans During Hypoxia. Front Physiol 2022; 12:763933. [PMID: 35095551 PMCID: PMC8795792 DOI: 10.3389/fphys.2021.763933] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/22/2021] [Indexed: 01/11/2023] Open
Abstract
Humans elicit a robust series of physiological responses to maintain adequate oxygen delivery during hypoxia, including a transient reduction in hemoglobin-oxygen (Hb-O2) affinity. However, high Hb-O2 affinity has been identified as a beneficial adaptation in several species that have been exposed to high altitude for generations. The observed differences in Hb-O2 affinity between humans and species adapted to high altitude pose a central question: is higher or lower Hb-O2 affinity in humans more advantageous when O2 availability is limited? Humans with genetic mutations in hemoglobin structure resulting in high Hb-O2 affinity have shown attenuated cardiorespiratory adjustments during hypoxia both at rest and during exercise, providing unique insight into this central question. Therefore, the purpose of this review is to examine the influence of high Hb-O2 affinity during hypoxia through comparison of cardiovascular and respiratory adjustments elicited by humans with high Hb-O2 affinity compared to those with normal Hb-O2 affinity.
Collapse
Affiliation(s)
- Kevin L. Webb
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | | | - Sarah E. Baker
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Stephen A. Klassen
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
| | - Michael J. Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Jonathon W. Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Chad C. Wiggins
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
- *Correspondence: Chad C. Wiggins,
| |
Collapse
|
12
|
Kanne CK, Nebor D, Pochron M, Oksenberg D, Sheehan VA. Rheological Impact of GBT1118 Cessation in a Sickle Mouse Model. Front Physiol 2021; 12:742784. [PMID: 34630162 PMCID: PMC8497897 DOI: 10.3389/fphys.2021.742784] [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: 07/16/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
In sickle cell disease (SCD), higher whole blood viscosity is a risk factor for vaso-occlusive crisis, avascular necrosis, and proliferative retinopathy. Blood viscosity is strongly impacted by hemoglobin (Hb) levels and red blood cell (RBC) deformability. Voxelotor is a hemoglobin S (HbS) polymerization inhibitor with anti-sickling properties that increases the Hb affinity for oxygen, thereby reducing HbS polymerization. In clinical trials, voxelotor increased Hb by an average of 1g/dl, creating concern that this rise in Hb could increase viscosity, particularly when the drug was cleared. To investigate this potential rebound hyperviscosity effect, we treated SCD mice with GBT1118, a voxelotor analog, and stopped the treatment to determine the effect on blood viscosity and RBC deformability under a range of oxygen concentrations. GBT1118 treatment increased Hb, improved RBC deformability by increasing the elongation index under normoxic (EImax) and hypoxic conditions (EImin), and decreased the point of sickling (PoS) without increasing blood viscosity. The anti-sickling effects and improvement of RBC deformability balanced the effect of increased Hb such that there was no increase in blood viscosity. Forty-eight hours after ceasing GBT1118, Hb declined from the rise induced by treatment, viscosity did not increase, and EImin remained elevated compared to control animals. Hb and PoS were not different from control animals, suggesting a return to native oxygen affinity and clearance of the drug. RBC deformability did not return to baseline, suggesting some residual rheological improvement. These data suggest that concerns regarding viscosity rise above pre-treatment levels upon sudden cessation of voxelotor are not warranted.
Collapse
Affiliation(s)
- Celeste K. Kanne
- Aflac Cancer & Blood Disorders Center Children’s Healthcare of Atlanta, School of Medicine, Emory University, Atlanta, GA, United States
- Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, TX, United States
| | - Danitza Nebor
- Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, TX, United States
| | - Mira Pochron
- Global Blood Therapeutics, South, San Francisco, CA, United States
| | - Donna Oksenberg
- Global Blood Therapeutics, South, San Francisco, CA, United States
| | - Vivien A. Sheehan
- Aflac Cancer & Blood Disorders Center Children’s Healthcare of Atlanta, School of Medicine, Emory University, Atlanta, GA, United States
- Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
13
|
Ahmed TA, El-Say KM, Abd-Allah FI, Omar AM, El-Araby ME, Muhammad YA, Pagare PP, Zhang Y, Mohmmad KA, Abdulmalik O, Safo MK. Improving the Solubility and Oral Bioavailability of a Novel Aromatic Aldehyde Antisickling Agent (PP10) for the Treatment of Sickle Cell Disease. Pharmaceutics 2021; 13:1148. [PMID: 34452107 PMCID: PMC8401948 DOI: 10.3390/pharmaceutics13081148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/15/2021] [Accepted: 07/23/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Aromatic aldehydes, with their ability to increase the oxygen affinity of sickle hemoglobin, have become important therapeutic agents for sickle cell disease (SCD). One such compound, voxelotor, was recently approved for SCD treatment. Methyl 6-((2-formyl-3-hydroxyphenoxy)methyl) picolinate (PP10) is another promising aromatic aldehyde, recently reported by our group. Like voxelotor, PP10 exhibits O2-dependent antisickling activity, but, unlike voxelotor, PP10 shows unique O2-independent antisickling effect. PP10, however, has limited solubility. This study therefore aimed to develop oral and parenteral formulations to improve PP10 solubility and bioavailability. METHODS Oral drug tablets with 2-hydroxypropyl beta cyclodextrin (HP-β-CD), polyvinylpyrrolidone, or Eudragit L100-55 PP10-binary system, and an intravenous (IV) formulation with d-α-tocopherol polyethylene glycol 1000 succinate (TPGS) or HP-β-CD, were developed. The pharmacokinetic behavior of the formulations was studied in Sprague-Dawley rats. PP10, a methylester, and its acid metabolite were also studied in vitro with sickle whole blood to determine their effect on Hb modification, Hb oxygen affinity, and sickle red blood cell inhibition. RESULTS Aqueous solubility of PP10 was enhanced ~5 times with the HP-β-CD binary system, while the TPGS aqueous micelle formulation was superior, with a drug concentration of 0.502 ± 0.01 mg/mL and a particle size of 26 ± 3 nm. The oral tablets showed relative and absolute bioavailabilities of 173.4% and 106.34%, respectively. The acid form of PP10 appeared to dominate in vivo, although both PP10 forms demonstrated pharmacologic effect. CONCLUSION Oral and IV formulations of PP10 were successfully developed using HP-β-CD binary system and TPGS aqueous micelles, respectively, resulting in significantly improved solubility and bioavailability.
Collapse
Affiliation(s)
- Tarek A. Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt;
| | - Khalid M. El-Say
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt;
| | - Fathy I. Abd-Allah
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt;
| | - Abdelsattar M. Omar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.M.O.); (M.E.E.-A.); (Y.A.M.); (K.A.M.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt
| | - Moustafa E. El-Araby
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.M.O.); (M.E.E.-A.); (Y.A.M.); (K.A.M.)
| | - Yosra A. Muhammad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.M.O.); (M.E.E.-A.); (Y.A.M.); (K.A.M.)
| | - Piyusha P. Pagare
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA; (P.P.P.); (Y.Z.); (M.K.S.)
| | - Yan Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA; (P.P.P.); (Y.Z.); (M.K.S.)
| | - Khadijah A. Mohmmad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.M.O.); (M.E.E.-A.); (Y.A.M.); (K.A.M.)
| | - Osheiza Abdulmalik
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Martin K. Safo
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA; (P.P.P.); (Y.Z.); (M.K.S.)
- Development, School of Pharmacy, The Institute for Structural Biology, Drug Discovery, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
14
|
Dufu K, Williams AT, Muller CR, Walser CM, Lucas A, Eaker AM, Alt C, Cathers BE, Oksenberg D, Cabrales P. Increased hemoglobin affinity for oxygen with GBT1118 improves hypoxia tolerance in sickle cell mice. Am J Physiol Heart Circ Physiol 2021; 321:H400-H411. [PMID: 34213392 DOI: 10.1152/ajpheart.00048.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Therapeutic agents that increase the Hb affinity for oxygen (O2) could, in theory, lead to decreased O2 release from Hb and impose a hypoxic risk to tissues. In this study, GBT1118, an allosteric modifier of Hb affinity for O2, was used to assess the impact of increasing Hb affinity for O2 on brain tissue oxygenation, blood pressure, heart rate, O2 delivery, and tolerance to hypoxia in Townes transgenic sickle cell disease (SCD) mice. Brain oxygenation and O2 delivery were studied during normoxia and severe hypoxic challenges. Chronic treatment with GBT1118 increased Hb affinity for O2, reducing the Po2 for 50% HbO2 saturation (P50) in SCD mice from 31 mmHg to 18 mmHg. This treatment significantly reduced anemia, increasing hematocrit by 33%, improved cardiac output (CO), and O2 delivery and extraction. Chronically increasing Hb affinity for O2 with GBT1118 preserved cortical O2 tension during normoxia, improved cortical O2 tension during hypoxia, and increased tolerance to severe hypoxia in SCD mice. Independent of hematological changes induced by chronic treatment, a single dose of GBT1118 significantly improved tolerance to hypoxia, highlighting the benefits of increasing Hb affinity for O2 and consequently reducing sickling of RBCs in blood during hypoxia in SCD.NEW & NOTEWORTHY Chronic pharmacologically increased hemoglobin affinity for oxygen in sickle cell disease mice alleviated hematological consequences of sickle cell disease, increasing RBC half-life, hematocrit, and hemoglobin concentration, while also decreasing reticulocyte count. Additionally, chronically increased hemoglobin affinity for oxygen significantly improved survival as well as cortical tissue oxygenation in sickle cell disease mice during hypoxia, suggesting that oxygen delivery and utilization is improved by increased hemoglobin affinity for oxygen.
Collapse
Affiliation(s)
- Kobina Dufu
- Global Blood Therapeutics, Inc., San Francisco, California
| | - Alexander T Williams
- Department of Bioengineering, University of California San Diego, La Jolla, California
| | - Cynthia R Muller
- Department of Bioengineering, University of California San Diego, La Jolla, California
| | - Cynthia M Walser
- Department of Bioengineering, University of California San Diego, La Jolla, California
| | - Alfredo Lucas
- Department of Bioengineering, University of California San Diego, La Jolla, California
| | - Allyn M Eaker
- Department of Bioengineering, University of California San Diego, La Jolla, California
| | - Carsten Alt
- Global Blood Therapeutics, Inc., San Francisco, California
| | | | | | - Pedro Cabrales
- Department of Bioengineering, University of California San Diego, La Jolla, California
| |
Collapse
|
15
|
Stewart GM, Cross TJ, Joyner MJ, Chase SC, Curry T, Lehrer-Graiwer J, Dufu K, Vlahakis NE, Johnson BD. Impact of Pharmacologically Left Shifting the Oxygen-Hemoglobin Dissociation Curve on Arterial Blood Gases and Pulmonary Gas Exchange During Maximal Exercise in Hypoxia. High Alt Med Biol 2021; 22:249-262. [PMID: 34152867 DOI: 10.1089/ham.2020.0159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Stewart, Glenn M., Troy J. Cross, Michael J. Joyner, Steven C. Chase, Timothy Curry, Josh Lehrer-Graiwer, Kobina Dufu, Nicholas E. Vlahakis, and Bruce D. Johnson. Impact of pharmacologically left shifting the oxygen-hemoglobin dissociation curve on arterial blood gases and pulmonary gas exchange during maximal exercise in hypoxia. High Alt Med Biol. 22:249-262, 2021. Introduction: Physiological and pathological conditions, which reduce the loading of oxygen onto hemoglobin (Hb), can impair exercise capacity and cause debilitating symptoms. Accordingly, this study examined the impact of pharmacologically left shifting the oxygen-hemoglobin dissociation curve (ODC) on arterial oxygen saturation (SaO2) and exercise capacity. Methods: Eight healthy subjects completed a maximal incremental exercise test in hypoxia (FIO2: 0.125) and normoxia (FIO2: 0.21) before (Day 1) and after (Day 15) daily ingestion of 900 mg of voxelotor (an oxygen/Hb affinity modulator). Pulmonary gas exchange and arterial blood gases were assessed throughout exercise and at peak. Data for a 1,500 mg daily drug dose are reported in a limited cohort (n = 3). Results: Fourteen days of drug administration left shifted the ODC (p50 measured under standard conditions, Day 1: 28.0 ± 2.1 mmHg vs. Day 15: 26.1 ± 1.8 mmHg, p < 0.05). Throughout incremental exercise in hypoxia, SaO2 was systematically higher after drug (peak exercise SaO2 on Day 1: 71 ± 2 vs. Day 15: 81% ± 2%, p < 0.001), whereas oxygen extraction (Ca-vO2 diff) and consumption (VO2) were similar (peak exercise Ca-vO2 diff on Day 1: 11.5 ± 1.7 vs. Day 15: 11.0 ± 1.8 ml/100 ml blood, p = 0.417; peak VO2 on Day 1: 2.59 ± 0.39 vs. Day 15: 2.47 ± 0.43 l/min, p = 0.127). Throughout incremental exercise in normoxia, SaO2 was systematically higher after drug, whereas peak VO2 was reduced (peak exercise SaO2 on Day 1: 93.9 ± 1.8 vs. Day 15: 95.8% ± 1.0%, p = 0.008; peak VO2 on Day 1: 3.62 ± 0.55 vs. Day 15: 3.26 ± 52 l/min, p = 0.001). Conclusion: Pharmacologically increasing the affinity of Hb for oxygen improved SaO2 during hypoxia without impacting exercise capacity; however, left shifting the ODC in healthy individuals appears detrimental to exercise capacity in normoxia. Left shifting the ODC to different magnitudes and under more chronic forms of hypoxia warrants further study.
Collapse
Affiliation(s)
- Glenn M Stewart
- Human Integrative and Environmental Physiology Laboratory, Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Troy J Cross
- Human Integrative and Environmental Physiology Laboratory, Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA.,Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Michael J Joyner
- Department of Anesthesia and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Steven C Chase
- Human Integrative and Environmental Physiology Laboratory, Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Timothy Curry
- Department of Anesthesia and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Kobina Dufu
- Global Blood Therapeutics, South San Francisco, California, USA
| | | | - Bruce D Johnson
- Human Integrative and Environmental Physiology Laboratory, Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
16
|
Glaros AK, Razvi R, Shah N, Zaidi AU. Voxelotor: alteration of sickle cell disease pathophysiology by a first-in-class polymerization inhibitor. Ther Adv Hematol 2021; 12:20406207211001136. [PMID: 33796238 PMCID: PMC7983433 DOI: 10.1177/20406207211001136] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 12/16/2022] Open
Abstract
Sickle cell disease, despite its recognition as a severely debilitating genetic condition affecting hundreds of thousands of neonates throughout the world each year, was not a target of pharmaceutical research focus for most of its 100-year existence in the medical consciousness. This has changed in recent years as many novel therapeutics are currently under investigation, with three new disease-modifying drugs achieving FDA approval in the last 4 years. One of these drugs, voxelotor, is especially encouraging as an inhibitor of sickling for its ability to safely improve the chronic hemolytic anemia of sickle cell disease. This was demonstrated during all clinical phases of investigation by an average improvement in hemoglobin of greater than 1 g/dL, as well as statistically significant improvements in established markers of hemolysis. While anemia itself represents a potential cause of morbidity, it is more importantly a marker of the hemolysis known to cause the long-term vascular and organ damage that makes sickle cell disease so debilitating and frequently fatal early in life. Given the recency of the approval, there has not been sufficient long-term follow-up to demonstrate improvement in the chronic sequelae of sickle cell disease as a result of voxelotor-induced improvements in hemolytic anemia. There is hope, however, based on the experience with hydroxyurea improving morbidity and mortality via reductions in sickling and improved rheology, that voxelotor may have similar long-term benefits by positively manipulating the kinetics of hemoglobin polymerization. This review aims to summarize the targeted pathobiology of sickle cell disease, the mechanism of action of voxelotor, and the safety and efficacy data from preclinical to late clinical stage investigations of this long-awaited medication, in the hopes of better informing the decision-making process behind prescribing or not prescribing it for patients in need of intervention.
Collapse
Affiliation(s)
- Alexander K. Glaros
- Central Michigan University, Mt. Pleasant, MI, USA
- Children’s Hospital of Michigan, Detroit, MI, USA
| | - Reza Razvi
- Children’s Hospital of Michigan, Detroit, MI, USA
| | | | - Ahmar U. Zaidi
- Central Michigan University, Mt. Pleasant, MI, USA
- Children’s Hospital of Michigan, Detroit, MI, USA
| |
Collapse
|
17
|
Abstract
Oxygen affinity to haemoglobin is indicated by the p50 value (pO2 at 50% O2Hb) and critically determines cellular oxygen availability. Although high Hb-O2 affinity can cause tissue hypoxia under conditions of well O2 saturated blood, individual differences in p50 are commonly not considered in clinical routine. Here, we investigated the diversity in Hb-O2 affinity in the context of physiological relevance. Oxyhaemoglobin dissociation curves (ODCs) of 60 volunteers (18–40 years, both sexes, either endurance trained or untrained) were measured at rest and after maximum exercise (VO2max) test. At rest, p50 values of all participants ranged over 7 mmHg. For comparison, right shift of ODC after VO2max test, representing the maximal physiological range to release oxygen to the tissue, indicated a p50 difference of up to 10 mmHg. P50 at rest differs significantly between women and men, with women showing lower Hb-O2 affinity that is determined by higher 2,3-BPG and BPGM levels. Regular endurance exercise did not alter baseline Hb-O2 affinity. Thus, p50 diversity is already high at baseline level and needs to be considered under conditions of impaired tissue oxygenation. For fast prediction of Hb-O2 affinity by blood gas analysis, only venous but not capillary blood samples can be recommended.
Collapse
|
18
|
Polymerized human hemoglobin facilitated modulation of tumor oxygenation is dependent on tumor oxygenation status and oxygen affinity of the hemoglobin-based oxygen carrier. Sci Rep 2020; 10:11372. [PMID: 32647211 PMCID: PMC7347553 DOI: 10.1038/s41598-020-68190-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/17/2020] [Indexed: 12/18/2022] Open
Abstract
Administration of hemoglobin-based oxygen carriers (HBOCs) into the systemic circulation is a potential strategy to relieve solid tumor hypoxia in order to increase the effectiveness of chemotherapeutics. Previous computational analysis indicated that the oxygen (O2) status of the tumor and HBOC O2 affinity may play a role in increased O2 delivery to the tumor. However, no study has experimentally investigated how low- and high-affinity HBOCs would perform in normoxic and hypoxic tumors. In this study, we examined how the HBOC, polymerized human hemoglobin (PolyhHb), in the relaxed (R) or tense (T) quaternary state modulates O2 delivery to hypoxic (FME) and normoxic (LOX) human melanoma xenografts in a murine window chamber model. We examined microcirculatory fluid flow via video shearing optical microscopy, and O2 distributions via phosphorescence quenching microscopy. Additionally, we examined how weekly infusion of a 20% top-load dose of PolyhHb influences growth rate, vascularization, and regional blood flow in the FME and LOX tumor xenografts. Infusion of low-affinity T-state PolyhHb led to increased tissue oxygenation, decreased blood flow, decreased tumor growth, and decreased vascularization in hypoxic tumors. However, infusion of both T-state and R-state PolyhHbs led to worse outcomes in normoxic tumors. Of particular concern was the high-affinity R-state PolyhHb, which led to no improvement in hypoxic tumors and significantly worsened outcomes in normoxic tumors. Taken together, the results of this study indicate that the tumor O2 status is a primary determinant of the potency and outcomes of infused PolyhHb.
Collapse
|
19
|
Belcher DA, Munoz C, Pires IS, Williams AT, Cabrales P, Palmer AF. Apohemoglobin-haptoglobin complexes attenuate the hypertensive response to low-molecular-weight polymerized hemoglobin. Blood Adv 2020; 4:2739-2750. [PMID: 32559292 PMCID: PMC7322967 DOI: 10.1182/bloodadvances.2020002045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 05/15/2020] [Indexed: 01/21/2023] Open
Abstract
Polymerized hemoglobin (PolyHb) is a promising hemoglobin (Hb)-based oxygen carrier currently undergoing development as a red blood cell substitute. Unfortunately, commercially developed products are composed of low-molecular-weight (LMW) PolyHb molecules, which extravasate, scavenge nitric oxide, and result in vasoconstriction and hypertension. The naturally occurring Hb-scavenging species haptoglobin (Hp), combined with the purified heme-scavenging species apohemoglobin (apoHb), is a potential candidate to alleviate the pressor effect of PolyHb. This study evaluated the protective activity of administering the apoHb-Hp complex to mitigate the vasoactive response induced by the transfusion of LMW PolyHb. Hp binding to PolyHb was characterized in vitro. The effectiveness of apoHb-Hp administration on reducing the vasoconstriction and pressor effects of PolyHb was assessed by measuring systemic and microcirculatory hemodynamics. Transfusion of LMW PolyHb to vehicle control pretreated animals increased mean arterial pressure while decreasing arteriole diameter and functional capillary density. However, transfusion of LMW PolyHb to apoHb-Hp pretreated animals prevented changes in mean arterial pressure, heart rate, arteriole diameter, blood flow, and functional capillary density relative to before transfusion. These results indicate that the increased size of PolyHb after binding to the apoHb-Hp complex may help compartmentalize PolyHb in the vascular space and thus reduce extravasation, nitric oxide scavenging, and toxicity responsible for vasoconstriction and systemic hypertension.
Collapse
Affiliation(s)
- Donald A Belcher
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH; and
| | - Carlos Munoz
- Department of Bioengineering, University of California San Diego, La Jolla, CA
| | - Ivan S Pires
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH; and
| | | | - Pedro Cabrales
- Department of Bioengineering, University of California San Diego, La Jolla, CA
| | - Andre F Palmer
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH; and
| |
Collapse
|
20
|
Chu Z, Wang Y, You G, Wang Q, Ma N, Li B, Zhao L, Zhou H. The P50 value detected by the oxygenation-dissociation analyser and blood gas analyser. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:867-874. [DOI: 10.1080/21691401.2020.1770272] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Zongtang Chu
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People’s Liberation Army, Beijing, P.R. China
| | - Ying Wang
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People’s Liberation Army, Beijing, P.R. China
| | - Guoxing You
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People’s Liberation Army, Beijing, P.R. China
| | - Quan Wang
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People’s Liberation Army, Beijing, P.R. China
| | - Ning Ma
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People’s Liberation Army, Beijing, P.R. China
| | - Bingting Li
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People’s Liberation Army, Beijing, P.R. China
| | - Lian Zhao
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People’s Liberation Army, Beijing, P.R. China
| | - Hong Zhou
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Academy of Military Science of the Chinese People’s Liberation Army, Beijing, P.R. China
| |
Collapse
|
21
|
Stewart GM, Chase S, Cross TJ, Wheatley-Guy CM, Joyner MJ, Curry T, Lehrer-Graiwer J, Dufu K, Vlahakis NE, Johnson BD. Effects of an allosteric hemoglobin affinity modulator on arterial blood gases and cardiopulmonary responses during normoxic and hypoxic low-intensity exercise. J Appl Physiol (1985) 2020; 128:1467-1476. [PMID: 32324473 DOI: 10.1152/japplphysiol.00185.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Numerous pathophysiological conditions induce hypoxemia-related cardiopulmonary perturbations, decrements in exercise capacity, and debilitating symptoms. Accordingly, this study investigated the efficacy of an allosteric hemoglobin modulator (voxelotor) to enhance arterial oxygen saturation during low-intensity exercise in hypoxia. Eight normal healthy subjects (36 ± 7 yr; 73.8 ± 9.5 kg; 3 women) completed a submaximal cycling test (60 W) under normoxic ([Formula: see text]: 0.21; O2 partial pressure: 144 mmHg) and hypoxic ([Formula: see text]: 0.125; O2 partial pressure: 82 mmHg) conditions before (day 1) and after (day 15) 14 days of oral drug administration. While stationary on a cycle ergometer and during exercise, ratings of perceived exertion (RPE) and dyspnea, oxygen consumption (V̇o2), and cardiac output (Q) were measured noninvasively, while arterial blood pressure (MAP) and blood gases ([Formula: see text], [Formula: see text], and [Formula: see text]) were measured invasively. The 14-day drug administration left shifted the oxygen-hemoglobin dissociation curve (ODC; p50 measured at standard pH and Pco2; day 1: 28.0 ± 2.1 mmHg vs. day 15: 26.1 ± 1.8 mmHg, P < 0.05). RPE, dyspnea, V̇o2, Q, and MAP were not different between day 1 and day 15. [Formula: see text] was similar during normoxia on day 1 and day 15 while stationary but higher during exercise (day 1: 95.2 ± 0.4% vs. day 15: 96.6 ± 0.3%, P < 0.05). [Formula: see text] was higher during hypoxia on day 15 while stationary (day 1: 82.9 ± 3.4% vs. day 15: 90.9 ± 1.8%, P < 0.05) and during exercise (day 1: 73.6 ± 2.5% vs. day 15: 84.8 ± 2.7%, P < 0.01). [Formula: see text] and [Formula: see text]were systematically higher and lower, respectively, after drug (P < 0.01), while the alveolar-arterial oxygen difference was unchanged suggesting hyperventilation contributed to the rise in [Formula: see text]. Oral administration of voxelotor left shifted the ODC and stimulated a mild hyperventilation, leading to improved arterial oxygen saturation without altering V̇o2 and central hemodynamics during rest and low-intensity exercise. This effect was more pronounced during submaximal hypoxic exercise, when arterial desaturation was more evident. Additional studies are needed to determine the effects of voxelotor during maximal exercise and under chronic forms of hypoxia.NEW & NOTEWORTHY In humans, a novel allosteric hemoglobin-oxygen affinity modulator was administered to comprehensively examine the cardiopulmonary consequences of stabilizing a portion of the available hemoglobin in a high-oxygen affinity state during submaximal exercise in normoxia and hypoxia. Oral administration of voxelotor enhanced arterial oxygen saturation during submaximal exercise without altering oxygen consumption and central hemodynamics; however, the partial pressure of arterial carbon dioxide was reduced and the partial pressure of arterial oxygen was increased implying that hyperventilation also contributed to the increase in oxygen saturation. The preservation of arterial oxygen saturation and content was particularly evident during hypoxic submaximal exercise, when arterial desaturation typically occurs, but this did not influence arterial-venous oxygen difference.
Collapse
Affiliation(s)
- Glenn M Stewart
- Human Integrative and Environmental Physiology Laboratory, Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Steven Chase
- Human Integrative and Environmental Physiology Laboratory, Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Troy J Cross
- Human Integrative and Environmental Physiology Laboratory, Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Courtney M Wheatley-Guy
- Human Integrative and Environmental Physiology Laboratory, Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Michael J Joyner
- Department of Anesthesia and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Timothy Curry
- Department of Anesthesia and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Kobina Dufu
- Global Blood Therapeutics, South San Francisco, California
| | | | - Bruce D Johnson
- Human Integrative and Environmental Physiology Laboratory, Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
22
|
LI X, LI W, FENG S, WANG R. [Research progress on mechanism in adaptation of hemoglobin to plateau hypoxia]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2019; 48:674-681. [PMID: 31955543 PMCID: PMC8800677 DOI: 10.3785/j.issn.1008-9292.2019.12.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/08/2019] [Indexed: 06/10/2023]
Abstract
Low oxygen partial pressure is the main cause of acute mountain sickness.Hemoglobin plays a crucial physiological role in the binding, utilization, transportation and release of oxygen in the body. To increase the capacity of oxygen binding of hemoglobin or the capacity of oxygen supply in tissues can help alleviate altitude sickness. However, increasing hemoglobin content has certain limitations. Using techniques from molecular biology, researchers are looking for endogenous or exogenous substances that can regulate the conformation of hemoglobin to increase oxygen uptake in the alveoli, or the availability of alveolar oxygen in the tissues. At present, the research on allosteric modulators to improve the affinity of hemoglobin has made some progress, and research on applying this mechanism to plateau hypoxia is also underway. This article reviews the relationship between hemoglobin and hypoxia, the structure of hemoglobin and the role of various allosteric modulators in hypoxia, which would provide information for finding new substances regulating the conformation of hemoglobin.
Collapse
Affiliation(s)
| | | | - Shilan FENG
- 封士兰(1957-), 女, 教授, 博士生导师, 主要从事中药中化学成分分离分析研究, E-mail:
| | - Rong WANG
- 王荣(1969-), 男, 博士, 主任药师, 教授, 博士生导师, 主要从事高原药代动力学等研究, E-mail:
;
https://orcid.org/0000-0001-9139-7311
| |
Collapse
|
23
|
Lucas A, Ao-ieong ESY, Williams AT, Jani VP, Muller CR, Yalcin O, Cabrales P. Increased Hemoglobin Oxygen Affinity With 5-Hydroxymethylfurfural Supports Cardiac Function During Severe Hypoxia. Front Physiol 2019; 10:1350. [PMID: 31736778 PMCID: PMC6831744 DOI: 10.3389/fphys.2019.01350] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 10/10/2019] [Indexed: 12/20/2022] Open
Abstract
Acclimatization to hypoxia or high altitude involves physiological adaptation processes, to influence oxygen (O2) transport and utilization. Several natural products, including aromatic aldehydes and isothiocyanates stabilize the R-state of hemoglobin (Hb), increasing Hb-O2 affinity and Hb-O2 saturation. These products are a counter intuitive therapeutic strategy to increase O2 delivery during hypoxia. 5-Hydroxymethylfurfural (5-HMF) is well known Amadori compound formed during the Maillard reaction (the non-enzymatic browning and caramelization of carbohydrate-containing foods after thermal treatment), with well documented effects in Hb-O2 affinity. This study explores the therapeutic potential of 5-HMF on left ventricular (LV) cardiac function (LVCF) during hypoxia. Anesthetized Golden Syrian hamsters received 5-HMF i.v., at 100 mg/kg and were subjected to stepwise increased hypoxia (15, 10, and 5%) every 30 min. LVCF was assessed using a closed chest method with a miniaturized conductance catheter via continuous LV pressure-volume (PV) measurements. Heart hypoxic areas were studied using pimonidazole staining. 5-HMF improved cardiac indices, including stroke volume (SV), cardiac output (CO), ejection fraction (EF), and stroke work (SW) compared to the vehicle group. At 5% O2, SV, CO, EF, and SW were increased by 53, 42, 33, and 51% with 5-HMF relative to vehicle. Heart chronotropic activity was not statistically changed, suggesting that differences in LV-CF during hypoxia by 5-HMF were driven by volume dependent effects. Analysis of coronary blood flow and cardiac muscle metabolism suggest no direct pharmacological effects from 5-HMF, therefore these results can be attributed to 5-HMF-dependent increase in Hb-O2 affinity. These studies establish that naturally occurring aromatic aldehydes, such as 5-HMF, produce modification of hemoglobin oxygen affinity with promising therapeutic potential to increase O2 delivery during hypoxic hypoxia.
Collapse
Affiliation(s)
- Alfredo Lucas
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Eilleen S. Y. Ao-ieong
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Alexander T. Williams
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Vivek P. Jani
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Cynthia R. Muller
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Ozlem Yalcin
- School of Medicine, Koç University, Istanbul, Turkey
| | - Pedro Cabrales
- Department of Bioengineering, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
24
|
Mahon RT, Ciarlone GE, Roney NG, Swift JM. Cardiovascular Parameters in a Swine Model of Normobaric Hypoxia Treated With 5-Hydroxymethyl-2-Furfural (5-HMF). Front Physiol 2019; 10:395. [PMID: 31057414 PMCID: PMC6482156 DOI: 10.3389/fphys.2019.00395] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 03/21/2019] [Indexed: 11/24/2022] Open
Abstract
Introduction: The consequences of low partial pressure of O2 include low arterial O2 saturations (SaO2), low blood O2 content (CaO2), elevated mean pulmonary artery pressure (PAP), and decreased O2 consumption VO2. 5-hydroxymethyl-2-furfural (5-HMF) binds to the N-terminal valine of hemoglobin (HgB) and increases its affinity to O2. We used an instrumented, sedated swine model to study the effect of 5-HMF on cardiovascular parameters during exposure to acute normobaric hypoxia (NH). Methods Twenty-three sedated and instrumented swine were randomly assigned to one of three treatment groups and received equal volume of normal saline (VEH), 20 mg/kg 5-HMF (5-HMF-20) or 40 mg/kg 5-HMF (5-HMF-40). Animals then breathed 10% FiO2 for 120 min. Parameters recorded were Cardiac Output (CO), Mean Arterial Blood Pressure (MAP), Heart Rate (HR), Mean Pulmonary Artery Pressure (PAP), SaO2 and saturation of mixed venous blood (SvO2). The P50 was measured at fixed time intervals prior to and during NH. Results 5-HMF decreased P50. In the first 30 min of NH, treatment with 5-HMF-20 and 5-HMF-40 resulted in a (1) significantly smaller decrement in SaO2 and SvO2, (2) significantly lower HR and CO, and (3) smaller increase in PAP compared to VEH. In the 120 min of NH there was a trend toward improved mortality with 5-HMF treatment. Conclusion 5-HMF treatment decreased P50, improved SaO2, and mitigated increases in PAP in this swine model of NH.
Collapse
Affiliation(s)
- Richard Thomas Mahon
- Department of Undersea Medicine, Naval Medical Research Center, Silver Spring, MD, United States
| | - Geoffrey E Ciarlone
- Department of Undersea Medicine, Naval Medical Research Center, Silver Spring, MD, United States
| | - Nicholas G Roney
- Department of Undersea Medicine, Naval Medical Research Center, Silver Spring, MD, United States
| | - Joshua M Swift
- Department of Undersea Medicine, Naval Medical Research Center, Silver Spring, MD, United States
| |
Collapse
|
25
|
Al Balushi H, Dufu K, Rees DC, Brewin JN, Hannemann A, Oksenberg D, Lu DC, Gibson JS. The effect of the antisickling compound GBT1118 on the permeability of red blood cells from patients with sickle cell anemia. Physiol Rep 2019; 7:e14027. [PMID: 30916477 PMCID: PMC6436144 DOI: 10.14814/phy2.14027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 02/21/2019] [Accepted: 02/23/2019] [Indexed: 12/27/2022] Open
Abstract
Sickle cell anemia (SCA) is one of the commonest severe inherited disorders. Nevertheless, effective treatments remain inadequate and novel ones are avidly sought. A promising advance has been the design of novel compounds which react with hemoglobin S (HbS) to increase oxygen (O2 ) affinity and reduce sickling. One of these, voxelotor (GBT440), is currently in advanced clinical trials. A structural analogue, GBT1118, was investigated in the current work. As RBC dehydration is important in pathogenesis of SCA, the effect of GBT1118 on RBC cation permeability was also studied. Activities of Psickle , the Gardos channel and the KCl cotransporter (KCC) were all reduced. Gardos channel and KCC activities were also inhibited in RBCs treated with Ca2+ ionophore or the thiol reagent N-ethylmaleimide, indicative of direct effects on these two transport systems. Consistent with its action on RBC membrane transporters, GBT1118 significantly increased RBC hydration. RBC hemolysis was reduced in a nonelectrolyte lysis assay. Further to its direct effects on O2 affinity, GBT1118 was therefore found to reduce RBC shrinkage and fragility. Findings reveal important effects of GBT1118 on protecting sickle cells and suggest that this is approach may represent a useful therapy for amelioration of the clinical complications of SCA.
Collapse
Affiliation(s)
- Halima Al Balushi
- Department of Veterinary MedicineUniversity of CambridgeCambridgeUnited Kingdom
| | - Kobina Dufu
- Global Blood TherapeuticsSouth San FranciscoCalifornia
| | - David C. Rees
- Department of Paediatric HaematologyKing's College London School of MedicineKing's College Hospital NHS Foundation TrustLondonUnited Kingdom
| | - John N. Brewin
- Department of Paediatric HaematologyKing's College London School of MedicineKing's College Hospital NHS Foundation TrustLondonUnited Kingdom
| | - Anke Hannemann
- Department of Veterinary MedicineUniversity of CambridgeCambridgeUnited Kingdom
| | | | - David C.‐Y. Lu
- Department of Veterinary MedicineUniversity of CambridgeCambridgeUnited Kingdom
| | - John S. Gibson
- Department of Veterinary MedicineUniversity of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
26
|
Lindsey ML, Gray GA, Wood SK, Curran-Everett D. Statistical considerations in reporting cardiovascular research. Am J Physiol Heart Circ Physiol 2018; 315:H303-H313. [PMID: 30028200 PMCID: PMC6139626 DOI: 10.1152/ajpheart.00309.2018] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The problem of inadequate statistical reporting is long standing and widespread in the biomedical literature, including in cardiovascular physiology. Although guidelines for reporting statistics have been available in clinical medicine for some time, there are currently no guidelines specific to cardiovascular physiology. To assess the need for guidelines, we determined the type and frequency of statistical tests and procedures currently used in the American Journal of Physiology-Heart and Circulatory Physiology. A PubMed search for articles published in the American Journal of Physiology-Heart and Circulatory Physiology between January 1, 2017, and October 6, 2017, provided a final sample of 146 articles evaluated for methods used and 38 articles for indepth analysis. The t-test and ANOVA accounted for 71% (212 of 300 articles) of the statistical tests performed. Of six categories of post hoc tests, Bonferroni and Tukey tests were used in 63% (62 of 98 articles). There was an overall lack in details provided by authors publishing in the American Journal of Physiology-Heart and Circulatory Physiology, and we compiled a list of recommended minimum reporting guidelines to aid authors in preparing manuscripts. Following these guidelines could substantially improve the quality of statistical reports and enhance data rigor and reproducibility.
Collapse
Affiliation(s)
- Merry L Lindsey
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi.,Research Service, G. V. (Sonny) Montgomery Veterans Affairs Medical Center , Jackson, Mississippi
| | - Gillian A Gray
- British Heart Foundation/University Centre for Cardiovascular Science, Edinburgh Medical School, University of Edinburgh , Edinburgh , United Kingdom
| | - Susan K Wood
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine , Columbia, South Carolina
| | - Douglas Curran-Everett
- Division of Biostatistics and Bioinformatics, National Jewish Health , Denver, Colorado.,Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Denver , Denver, Colorado
| |
Collapse
|
27
|
Patel MP, Siu V, Silva-Garcia A, Xu Q, Li Z, Oksenberg D. Development and validation of an oxygen dissociation assay, a screening platform for discovering, and characterizing hemoglobin-oxygen affinity modifiers. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:1599-1607. [PMID: 29910606 PMCID: PMC5989706 DOI: 10.2147/dddt.s157570] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Introduction Hemoglobin (Hb) is a critical molecule necessary for all vertebrates to maintain aerobic metabolism. Hb–oxygen (O2) affinity modifiers have been studied to address various diseases including sickle cell disease, hypoxemia, tumor hypoxia, and wound healing. However, drug development of exogenous Hb modifiers has been hindered by the lack of a technique to rapidly screen compounds for their ability to alter Hb–O2 affinity. We have developed a novel screening assay based upon the spectral changes observed during Hb deoxygenation and termed it the oxygen dissociation assay (ODA). Methodology ODA allows for the quantitation of oxygenated Hb at given time points during Hb deoxygenation on a 96-well plate. This assay was validated by comparing the ability of 500 Hb modifiers to alter the Hb–O2 affinity in the ODA vs the oxygen equilibrium curves obtained using the industry standard Hemox Analyzer instrument. Results A correlation (R2) of 0.7 indicated that the ODA has the potential to screen and identify potent exogenous Hb modifiers. In addition, it allows for concurrent comparison of compounds, concentrations, buffers, or pHs on the level of Hb oxygenation. Conclusion With a cost-effective, simple, rapid, and highly adaptable assay, the ODA will allow researchers to rapidly characterize Hb–O2 affinity modifiers.
Collapse
Affiliation(s)
- Mira P Patel
- Biology Department, Global Blood Therapeutics Inc., South San Francisco, CA, USA
| | - Vincent Siu
- Biology Department, Global Blood Therapeutics Inc., South San Francisco, CA, USA
| | - Abel Silva-Garcia
- Biology Department, Global Blood Therapeutics Inc., South San Francisco, CA, USA
| | - Qing Xu
- Chemistry Department, Global Blood Therapeutics Inc., South San Francisco, CA, USA
| | - Zhe Li
- Chemistry Department, Global Blood Therapeutics Inc., South San Francisco, CA, USA
| | - Donna Oksenberg
- Biology Department, Global Blood Therapeutics Inc., South San Francisco, CA, USA
| |
Collapse
|