1
|
Yuan X, Ruan W, Bobrow B, Carmeliet P, Eltzschig HK. Targeting hypoxia-inducible factors: therapeutic opportunities and challenges. Nat Rev Drug Discov 2024; 23:175-200. [PMID: 38123660 DOI: 10.1038/s41573-023-00848-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2023] [Indexed: 12/23/2023]
Abstract
Hypoxia-inducible factors (HIFs) are highly conserved transcription factors that are crucial for adaptation of metazoans to limited oxygen availability. Recently, HIF activation and inhibition have emerged as therapeutic targets in various human diseases. Pharmacologically desirable effects of HIF activation include erythropoiesis stimulation, cellular metabolism optimization during hypoxia and adaptive responses during ischaemia and inflammation. By contrast, HIF inhibition has been explored as a therapy for various cancers, retinal neovascularization and pulmonary hypertension. This Review discusses the biochemical mechanisms that control HIF stabilization and the molecular strategies that can be exploited pharmacologically to activate or inhibit HIFs. In addition, we examine medical conditions that benefit from targeting HIFs, the potential side effects of HIF activation or inhibition and future challenges in this field.
Collapse
Affiliation(s)
- Xiaoyi Yuan
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Wei Ruan
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Anaesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bentley Bobrow
- Department of Emergency Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Peter Carmeliet
- Laboratory of Angiogenesis & Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis & Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Holger K Eltzschig
- Department of Anaesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Outcomes Research Consortium, Cleveland, OH, USA.
| |
Collapse
|
2
|
A Nano Erythropoiesis Stimulating Agent (Nano-ESA) for the Treatment of Anemia and Associated Disorders. iScience 2022; 25:105021. [PMID: 36111254 PMCID: PMC9468392 DOI: 10.1016/j.isci.2022.105021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/20/2022] [Accepted: 08/19/2022] [Indexed: 11/24/2022] Open
|
3
|
Rodriguez-Labrada R, Ortega-Sanchez R, Hernández Casaña P, Santos Morales O, Padrón-Estupiñan MDC, Batista-Nuñez M, Jiménez Rodríguez D, Canales-Ochoa N, Peña Acosta A, Medrano Montero J, Labrada Aguilera PE, Estupiñán Rodriguez A, Vazquez-Mojena Y, Almaguer Gotay D, Aymed-García J, García-García I, Torres Vega R, Viada González C, Valenzuela Silva CM, Silva Ricardo Y, Columbié Ximelis J, Tribin Rivero K, Valle Cabrera R, García-Rodriguez JC, Crombet Ramos T, Amaro-González D, Rodriguez-Obaya T, Velázquez-Pérez L. Erythropoietin in Spinocerebellar Ataxia Type 2: Feasibility and Proof-of-Principle Issues from a Randomized Controlled Study. Mov Disord 2022; 37:1516-1525. [PMID: 35607776 DOI: 10.1002/mds.29045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 02/16/2022] [Accepted: 03/21/2022] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Several pieces of evidence have shown the neurotrophic effect of erythropoietin (EPO) and its introduction in the therapeutic practice of neurological diseases. However, its usefulness in the treatment of spinocerebellar ataxia type 2 (SCA2) has not been proven despite the fact that it is endogenously reduced in these patients. OBJECTIVE The study aims to investigate the safety, tolerability, and clinical effects of a nasally administered recombinant EPO in SCA2 patients. METHODS Thirty-four patients were enrolled in this double-blind, randomized, placebo-controlled, phase I-II clinical trial of the nasally administered human-recombinant EPO (NeuroEPO) for 6 months. The primary outcome was the change in the spinocerebellar ataxia functional index (SCAFI), while other motor, neuropsychological, and oculomotor measures were assessed. RESULTS The 6-month changes in SCAFI score were slightly higher in the patients allocated to NeuroEPO treatment than placebo in spite of the important placebo effect observed for this parameter. However, saccade latency was significantly decreased in the NeuroEPO group but not in placebo. The frequency and severity of adverse events were similar between both groups, without evidences of hematopoietic activity of the drug. CONCLUSIONS This study demonstrated the safety and tolerability of NeuroEPO in SCA2 patients after 6 months of treatments and suggested a small clinical effect of this drug on motor and cognitive abnormalities, but confirmatory studies are warranted. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Roberto Rodriguez-Labrada
- Center for Research and Rehabilitation of Hereditary Ataxias (CIRAH), Holguín, Cuba.,Cuban Centre for Neurosciences, Havana, Cuba
| | | | | | | | | | - Maricela Batista-Nuñez
- Otorhinolaryngology Department, "Octavio de la Concepción" Pediatric Hospital, Holguín, Cuba
| | | | - Nalia Canales-Ochoa
- Center for Research and Rehabilitation of Hereditary Ataxias (CIRAH), Holguín, Cuba
| | - Arnoy Peña Acosta
- Center for Research and Rehabilitation of Hereditary Ataxias (CIRAH), Holguín, Cuba
| | | | | | | | - Yaimee Vazquez-Mojena
- Center for Research and Rehabilitation of Hereditary Ataxias (CIRAH), Holguín, Cuba.,Cuban Centre for Neurosciences, Havana, Cuba
| | | | | | | | - Reydenis Torres Vega
- Center for Research and Rehabilitation of Hereditary Ataxias (CIRAH), Holguín, Cuba
| | | | | | | | | | | | | | | | | | | | | | - Luis Velázquez-Pérez
- Center for Research and Rehabilitation of Hereditary Ataxias (CIRAH), Holguín, Cuba.,Cuban Academy of Sciences, Havana, Cuba
| |
Collapse
|
4
|
Hoffmann A, de Souza LV, Seifert M, von Raffay L, Haschka D, Grubwieser P, Grander M, Mitterstiller AM, Nairz M, Poli M, Weiss G. Pharmacological Targeting of BMP6-SMAD Mediated Hepcidin Expression Does Not Improve the Outcome of Systemic Infections With Intra-Or Extracellular Gram-Negative Bacteria in Mice. Front Cell Infect Microbiol 2021; 11:705087. [PMID: 34368018 PMCID: PMC8342937 DOI: 10.3389/fcimb.2021.705087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/05/2021] [Indexed: 12/21/2022] Open
Abstract
Introduction Hepcidin is the systemic master regulator of iron metabolism as it degrades the cellular iron exporter ferroportin. In bacterial infections, hepcidin is upregulated to limit circulating iron for pathogens, thereby increasing iron retention in macrophages. This mechanism withholds iron from extracellular bacteria but could be of disadvantage in infections with intracellular bacteria. We aimed to understand the role of hepcidin in infections with intra- or extracellular bacteria using different hepcidin inhibitors. Methods For the experiments LDN-193189 and oversulfated heparins were used, which interact with the BMP6-SMAD pathway thereby inhibiting hepcidin expression. We infected male C57BL/6N mice with either the intracellular bacterium Salmonella Typhimurium or the extracellular bacterium Escherichia coli and treated these mice with the different hepcidin inhibitors. Results Both inhibitors effectively reduced hepcidin levels in vitro under steady state conditions and upon stimulation with the inflammatory signals interleukin-6 or lipopolysaccharide. The inhibitors also reduced hepcidin levels and increased circulating iron concentration in uninfected mice. However, both compounds failed to decrease liver- and circulating hepcidin levels in infected mice and did not affect ferroportin expression in the spleen or impact on serum iron levels. Accordingly, both BMP-SMAD signaling inhibitors did not influence bacterial numbers in different organs in the course of E.coli or S.Tm sepsis. Conclusion These data indicate that targeting the BMP receptor or the BMP-SMAD pathway is not sufficient to suppress hepcidin expression in the course of infection with both intra- or extracellular bacteria. This suggests that upon pharmacological inhibition of the central SMAD-BMP pathways during infection, other signaling cascades are compensatorily induced to ensure sufficient hepcidin formation and iron restriction to circulating microbes.
Collapse
Affiliation(s)
- Alexander Hoffmann
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Lara Valente de Souza
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Seifert
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - Laura von Raffay
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| | - David Haschka
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Philipp Grubwieser
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Manuel Grander
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna-Maria Mitterstiller
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Maura Poli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
5
|
Chittepu VCSR, Kalhotra P, Osorio-Gallardo T, Jiménez-Martínez C, Torre RRRDL, Gallardo-Velazquez T, Osorio-Revilla G. New Molecular Insights into the Inhibition of Dipeptidyl Peptidase-4 by Natural Cyclic Peptide Oxytocin. Molecules 2019; 24:E3887. [PMID: 31661941 PMCID: PMC6864445 DOI: 10.3390/molecules24213887] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/23/2019] [Accepted: 10/23/2019] [Indexed: 11/21/2022] Open
Abstract
Protease inhibition has led to treating many diseases and has been successful in producing many commercial drugs by pharmaceutical companies. Among many proteases, serine protease has been attractive in treating metabolic disorder diabetes mellitus (DM). Gliptins have been proven to inhibit dipeptidyl peptidase-4 (DPP4), a serine protease, and are an emerging therapeutic drug target to reduce blood glucose levels, but until now there is no natural cyclic peptide proven to inhibit serine protease DPP4. This study demonstrates the potential mechanism of natural cyclic peptide oxytocin (OXT) as a DPP4 inhibitor. To achieve this, initially, activity atlas and field-based models of DPP4 inhibitors were utilized to predict the possible features of positive and negative electrostatic, hydrophobic, and activity shapes of DPP4 inhibition. Oxytocin binding mode, flexibility, and interacting residues were studied using molecular docking simulations studies. 3D-RISM calculations studies revealed that the stability of water molecules at the binding site are favorable. Finally, an experimental study using fluorescence assay revealed OXT inhibits DPP4 in a concentration-dependent manner in a significant way (p < 0.05) and possess IC50 of 110.7 nM. These new findings significantly expand the pharmaceutical application of cyclic peptides, and in specific OXT, and implicate further optimization of OXT inhibition capacity to understand the effect of DPP4 inhibition. This work highlights the development of natural cyclic peptides as future therapeutic peptides to reduce glucose levels and treat diabetes mellitus.
Collapse
Affiliation(s)
- Veera C S R Chittepu
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politecnico Nacional, Av. Wilfrido Massieu S/N, Col. Unidad Profesional Adolfo López Mateos, Zacatenco, Ciudad de Mexico 07738, Mexico.
| | - Poonam Kalhotra
- Departamento de Biofísica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Col. Santo Tomás, CP. Ciudad de Mexico 11340, Mexico.
| | - Tzayhri Osorio-Gallardo
- Departamento de Microbiologia e Immunologia, Facultad de Medicina Veterinaria Y Zootecnia, Universidad Nacional Autonoma de Mexico, Av. Universidad #3000, Delegacion Coyoacan, Col. Ciudad Universitaria, Ciudad de Mexico 04510, Mexico.
| | - Cristian Jiménez-Martínez
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politecnico Nacional, Av. Wilfrido Massieu S/N, Col. Unidad Profesional Adolfo López Mateos, Zacatenco, Ciudad de Mexico 07738, Mexico.
| | - Raúl René Robles-de la Torre
- Centro de Investigación en Biotecnología Aplicada CIBA, Instituto Politécnico Nacional, Carretera Estatal, Tecuexcomac-Tepetitla, Km 1.5, CP. Tlaxcala 90700, Mexico.
| | - Tzayhri Gallardo-Velazquez
- Departamento de Biofísica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Col. Santo Tomás, CP. Ciudad de Mexico 11340, Mexico.
| | - Guillermo Osorio-Revilla
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politecnico Nacional, Av. Wilfrido Massieu S/N, Col. Unidad Profesional Adolfo López Mateos, Zacatenco, Ciudad de Mexico 07738, Mexico.
| |
Collapse
|
6
|
Winzelberg Olson S, Hohenhaus AE. Feline non-regenerative anemia: Diagnostic and treatment recommendations. J Feline Med Surg 2019; 21:615-631. [PMID: 31234748 PMCID: PMC10814193 DOI: 10.1177/1098612x19856178] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PRACTICAL RELEVANCE Non-regenerative anemia, or anemia with reticulocytopenia, is a daily diagnosis in feline practice. CLINICAL CHALLENGES The disease processes underlying non-regenerative anemia are many and diverse. A major diagnostic evaluation may be required to correctly diagnose and treat the underlying cause. AUDIENCE All veterinarians caring for cats will face the diagnostic and therapeutic challenge of non-regenerative anemia. Readers will benefit from the review of diagnostic testing and therapeutic options for non-regenerative anemia. EVIDENCE BASE This review summarizes the currently available literature informing diagnostic and treatment recommendations related to non-regenerative anemia. The evidence available to support the recommendations in this review is graded as low and includes predominantly expert opinion, case reports and cases series, on which the authors' interpretation/consensus is based.
Collapse
Affiliation(s)
- Sarah Winzelberg Olson
- DVM, Diplomate ACVIM (Oncology and Small Animal Internal Medicine) Animal Medical Center, New York, NY, USA
| | - Ann E Hohenhaus
- DVM, Diplomate ACVIM (Oncology and Small Animal Internal Medicine) Animal Medical Center, New York, NY, USA
| |
Collapse
|
7
|
Heuberger JAAC, Cohen AF. Review of WADA Prohibited Substances: Limited Evidence for Performance-Enhancing Effects. Sports Med 2019; 49:525-539. [PMID: 30411235 PMCID: PMC6422964 DOI: 10.1007/s40279-018-1014-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The World Anti-Doping Agency is responsible for maintaining a Prohibited List that describes the use of substances and methods that are prohibited for athletes. The list currently contains 23 substance classes, and an important reason for the existence of this list is to prevent unfair competition due to pharmacologically enhanced performance. The aim of this review was to give an overview of the available evidence for performance enhancement of these substance classes. We searched the scientific literature through PubMed for studies and reviews evaluating the effects of substance classes on performance. Findings from double-blind, randomized controlled trials were considered as evidence for (the absence of) effects if they were performed in trained subjects measuring relevant performance outcomes. Only 5 of 23 substance classes show evidence of having the ability to enhance actual sports performance, i.e. anabolic agents, β2-agonists, stimulants, glucocorticoids and β-blockers. One additional class, growth hormone, has similar evidence but only in untrained subjects. The observed effects all relate to strength or sprint performance (and accuracy for β-blockers); there are no studies showing positive effects on reliable markers of endurance performance. For 11 classes, no well-designed studies are available, and, for the remaining six classes, there is evidence of an absence of a positive effect. In conclusion, for the majority of substance classes, no convincing evidence for performance enhancement is available, while, for the remaining classes, the evidence is based on a total of only 266 subjects from 11 studies.
Collapse
Affiliation(s)
| | - Adam F Cohen
- Centre for Human Drug Research, Zernikedreef 8, 2333 CL, Leiden, The Netherlands
- Department of Internal Medicine, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
8
|
Joharapurkar AA, Pandya VB, Patel VJ, Desai RC, Jain MR. Prolyl Hydroxylase Inhibitors: A Breakthrough in the Therapy of Anemia Associated with Chronic Diseases. J Med Chem 2018; 61:6964-6982. [PMID: 29712435 DOI: 10.1021/acs.jmedchem.7b01686] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Chronic kidney disease, cancer, chronic inflammatory disorders, nutritional, and genetic deficiency can cause anemia. Hypoxia causes induction of hypoxia-inducible factor (HIF), which stimulates erythropoietin (EPO) synthesis. Prolyl hydroxylase domain (PHD) enzyme inhibition can stabilize hypoxia-inducible factor (HIF). HIF stabilization also decreases hepcidin, a hormone of hepatic origin, which regulates iron homeostasis. PHD inhibitors represent a novel pharmacological treatment of anemia associated with chronic diseases. Many orally active PHD inhibitors like roxadustat, molidustat, vadadustat, and desidustat are in late phase clinical trials. This review discusses the role of PHD inhibitors in the treatment of anemia associated with chronic diseases.
Collapse
Affiliation(s)
- Amit A Joharapurkar
- Zydus Research Centre , Cadila Healthcare Limited , Sarkhej Bavla NH8A , Moraiya , Ahmedabad 382210 , India
| | - Vrajesh B Pandya
- Zydus Research Centre , Cadila Healthcare Limited , Sarkhej Bavla NH8A , Moraiya , Ahmedabad 382210 , India
| | - Vishal J Patel
- Zydus Research Centre , Cadila Healthcare Limited , Sarkhej Bavla NH8A , Moraiya , Ahmedabad 382210 , India
| | - Ranjit C Desai
- Zydus Research Centre , Cadila Healthcare Limited , Sarkhej Bavla NH8A , Moraiya , Ahmedabad 382210 , India
| | - Mukul R Jain
- Zydus Research Centre , Cadila Healthcare Limited , Sarkhej Bavla NH8A , Moraiya , Ahmedabad 382210 , India
| |
Collapse
|
9
|
Different effects of granulocyte colony-stimulating factor and erythropoietin on erythropoiesis. Stem Cell Res Ther 2018; 9:119. [PMID: 29720275 PMCID: PMC5930863 DOI: 10.1186/s13287-018-0877-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/08/2018] [Accepted: 04/16/2018] [Indexed: 11/24/2022] Open
Abstract
Background Red blood cells are the most abundant cells in the blood that deliver oxygen to the whole body. Erythropoietin (EPO), a positive regulator of erythropoiesis, is currently the major treatment for chronic anemia. Granulocyte colony-stimulating factor (G-CSF) is a multifunctional cytokine and a well-known regulator of hematopoietic stem cell proliferation, differentiation, and mobilization. The use of EPO in combination with G-CSF has been reported to synergistically improve erythroid responses in a group of patients with myelodysplastic syndromes who did not respond to EPO treatment alone; however, the mechanism remains unclear. Methods C57BL/6 J mice injected with G-CSF or EPO were used to compare the erythropoiesis status and the efficiency of erythroid mobilization by flow cytometry. Results In this study, we found that G-CSF induced more orthochromatophilic erythroblast production than did EPO in the bone marrow and spleen. In addition, in contrast to EPO treatments, G-CSF treatments enhanced the efficiency of the mobilization of newly synthesized reticulocytes into peripheral blood. Our results demonstrated that the effects of G-CSF on erythropoiesis and erythrocytic mobilization were independent of EPO secretion and, in contrast to EPO, G-CSF promoted progression of erythropoiesis through transition of early stage R2 (basophilic erythroblasts) to late stage R4 (orthochromatophilic erythroblasts). Conclusions We demonstrate for the first time that G-CSF treatments induce a faster erythropoiesis-enhancing response than that of EPO. These findings suggest an alternative approach to treating acute anemia, especially when patients are experiencing a clinical emergency in remote areas without proper blood bank supplies.
Collapse
|
10
|
Locatelli F, Mazzaferro S, Yee J. Iron Therapy Challenges for the Treatment of Nondialysis CKD Patients. Clin J Am Soc Nephrol 2016; 11:1269-1280. [PMID: 27185524 PMCID: PMC4934828 DOI: 10.2215/cjn.00080116] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The clinical consequences of untreated, severe anemia in patients with nondialysis CKD can be significant, but disparities exist in the anemia treatment guidelines and position papers issued from working groups and associations across the world. These differ in hemoglobin target and iron levels and their emphasis on various iron markers and other clinical outcomes. Not surprisingly, disparities are observed in anemia treatment strategies among patients with nondialysis CKD across different areas of the world. Over the past decade, the prescription and dosage of both iron therapies and erythropoiesis-stimulating agents have shifted, with notable regional differences observed. Moreover, there is ongoing debate regarding oral versus intravenous administration of iron. Compared with oral iron therapy, which often leads to gastrointestinal adverse events, low patient adherence, and low efficacy, intravenous iron administration has been associated with potential serious adverse events, such as anaphylaxis. New iron-based compounds and drugs currently under development are reviewed to describe their potential benefits in the treatment of anemia in patients with CKD. New oral compounds, including iron-based phosphate binders, heme iron polypeptide, and liposomal iron, show different rates of absorption with possibly different efficacy and improved tolerability. These new potential therapies offer health care providers additional anemia treatment options for their patients with CKD; however, the management of anemia in the CKD population continues to present challenges that require prospective studies to identify the optimal iron therapy for patients.
Collapse
Affiliation(s)
- Francesco Locatelli
- Department of Nephrology, Dialysis and Transplantation, Alessandro Manzoni Hospital, Lecco, Italy
| | - Sandro Mazzaferro
- Department of Cardiovascular, Respiratory, Nephrologic and Geriatric Sciences, Sapienza University, Rome, Italy; and
| | - Jerry Yee
- Division of Nephrology and Hypertension, Henry Ford Hospital, Detroit, Michigan
| |
Collapse
|
11
|
Kiers HD, Scheffer GJ, van der Hoeven JG, Eltzschig HK, Pickkers P, Kox M. Immunologic Consequences of Hypoxia during Critical Illness. Anesthesiology 2016; 125:237-49. [PMID: 27183167 PMCID: PMC5119461 DOI: 10.1097/aln.0000000000001163] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hypoxia and immunity are highly intertwined at clinical, cellular, and molecular levels. The prevention of tissue hypoxia and modulation of systemic inflammation are cornerstones of daily practice in the intensive care unit. Potentially, immunologic effects of hypoxia may contribute to outcome and represent possible therapeutic targets. Hypoxia and activation of downstream signaling pathways result in enhanced innate immune responses, aimed to augment pathogen clearance. On the other hand, hypoxia also exerts antiinflammatory and tissue-protective effects in lymphocytes and other tissues. Although human data on the net immunologic effects of hypoxia and pharmacologic modulation of downstream pathways are limited, preclinical data support the concept of tailoring the immune response through modulation of the oxygen status or pharmacologic modulation of hypoxia-signaling pathways in critically ill patients.
Collapse
Affiliation(s)
- Harmke D. Kiers
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Department of Anesthesiology, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Centre for Infectious Diseases (RCI), Nijmegen, The Netherlands
| | - Gert-Jan Scheffer
- Department of Anesthesiology, Radboud university medical center, Nijmegen, The Netherlands
| | - Johannes G. van der Hoeven
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Centre for Infectious Diseases (RCI), Nijmegen, The Netherlands
| | - Holger K. Eltzschig
- Organ Protection Program; Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Centre for Infectious Diseases (RCI), Nijmegen, The Netherlands
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Department of Anesthesiology, Radboud university medical center, Nijmegen, The Netherlands
- Radboud Centre for Infectious Diseases (RCI), Nijmegen, The Netherlands
| |
Collapse
|
12
|
Schmid H, Jelkmann W. Investigational therapies for renal disease-induced anemia. Expert Opin Investig Drugs 2016; 25:901-16. [DOI: 10.1080/13543784.2016.1182981] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
13
|
Barbieri C, Bolzoni E, Mari F, Cattinelli I, Bellocchio F, Martin JD, Amato C, Stopper A, Gatti E, Macdougall IC, Stuard S, Canaud B. Performance of a Predictive Model for Long-Term Hemoglobin Response to Darbepoetin and Iron Administration in a Large Cohort of Hemodialysis Patients. PLoS One 2016; 11:e0148938. [PMID: 26939055 PMCID: PMC4777424 DOI: 10.1371/journal.pone.0148938] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 01/23/2016] [Indexed: 12/18/2022] Open
Abstract
Anemia management, based on erythropoiesis stimulating agents (ESA) and iron supplementation, has become an increasingly challenging problem in hemodialysis patients. Maintaining hemodialysis patients within narrow hemoglobin targets, preventing cycling outside target, and reducing ESA dosing to prevent adverse outcomes requires considerable attention from caregivers. Anticipation of the long-term response (i.e. at 3 months) to the ESA/iron therapy would be of fundamental importance for planning a successful treatment strategy. To this end, we developed a predictive model designed to support decision-making regarding anemia management in hemodialysis (HD) patients treated in center. An Artificial Neural Network (ANN) algorithm for predicting hemoglobin concentrations three months into the future was developed and evaluated in a retrospective study on a sample population of 1558 HD patients treated with intravenous (IV) darbepoetin alfa, and IV iron (sucrose or gluconate). Model inputs were the last 90 days of patients’ medical history and the subsequent 90 days of darbepoetin/iron prescription. Our model was able to predict individual variation of hemoglobin concentration 3 months in the future with a Mean Absolute Error (MAE) of 0.75 g/dL. Error analysis showed a narrow Gaussian distribution centered in 0 g/dL; a root cause analysis identified intercurrent and/or unpredictable events associated with hospitalization, blood transfusion, and laboratory error or misreported hemoglobin values as the main reasons for large discrepancy between predicted versus observed hemoglobin values. Our ANN predictive model offers a simple and reliable tool applicable in daily clinical practice for predicting the long-term response to ESA/iron therapy of HD patients.
Collapse
Affiliation(s)
| | | | - Flavio Mari
- Fresenius Medical Care, Bad Homburg, Germany
| | | | | | - José D. Martin
- Intelligent Data Analysis Laboratory, University of Valencia, Burjassot (Valencia), Spain
| | | | | | - Emanuele Gatti
- Center for Biomedical Technology at the Danube University, Krems, Austria
| | | | | | - Bernard Canaud
- Fresenius Medical Care, Bad Homburg, Germany
- UFR Medicine, University of Montpellier I, Montpellier, France
- * E-mail:
| |
Collapse
|
14
|
Vohwinkel CU, Hoegl S, Eltzschig HK. Hypoxia signaling during acute lung injury. J Appl Physiol (1985) 2015; 119:1157-63. [PMID: 25977449 PMCID: PMC4816417 DOI: 10.1152/japplphysiol.00226.2015] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/07/2015] [Indexed: 12/29/2022] Open
Abstract
Acute lung injury (ALI) is an inflammatory lung disease that manifests itself in patients as acute respiratory distress syndrome and thereby contributes significantly to the morbidity and mortality of patients experiencing critical illness. Even though it may seem counterintuitive, as the lungs are typically well-oxygenated organs, hypoxia signaling pathways have recently been implicated in the resolution of ALI. For example, functional studies suggest that transcriptional responses under the control of the hypoxia-inducible factor (HIF) are critical in optimizing alveolar epithelial carbohydrate metabolism, and thereby dampen lung inflammation during ALI. In the present review we discuss functional roles of oxygenation, hypoxia and HIFs during ALI, mechanisms of how HIFs are stabilized during lung inflammation, and how HIFs can mediate lung protection during ALI.
Collapse
Affiliation(s)
- Christine U Vohwinkel
- Organ Protection Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado; Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado; and
| | - Sandra Hoegl
- Organ Protection Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado; Department of Anesthesiology, University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Holger K Eltzschig
- Organ Protection Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
15
|
Sankaran VG, Weiss MJ. Anemia: progress in molecular mechanisms and therapies. Nat Med 2015; 21:221-30. [PMID: 25742458 DOI: 10.1038/nm.3814] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/04/2015] [Indexed: 12/12/2022]
Abstract
Anemia is a major source of morbidity and mortality worldwide. Here we review recent insights into how red blood cells (RBCs) are produced, the pathogenic mechanisms underlying various forms of anemia, and novel therapies derived from these findings. It is likely that these new insights, mainly arising from basic scientific studies, will contribute immensely to both the understanding of frequently debilitating forms of anemia and the ability to treat affected patients. Major worldwide diseases that are likely to benefit from new advances include the hemoglobinopathies (β-thalassemia and sickle cell disease); rare genetic disorders of RBC production; and anemias associated with chronic kidney disease, inflammation, and cancer. Promising new approaches to treatment include drugs that target recently defined pathways in RBC production, iron metabolism, and fetal globin-family gene expression, as well as gene therapies that use improved viral vectors and newly developed genome editing technologies.
Collapse
Affiliation(s)
- Vijay G Sankaran
- 1] Division of Hematology and Oncology, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA. [2] Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA. [3] Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
16
|
Donck J, Gonzalez-Tabares L, Chanliau J, Martin H, Stamatelou K, Manamley N, Farouk M, Addison J. Preservation of anemia control and weekly ESA dosage after conversion from PEG-Epoetin beta to darbepoetin alfa in adult hemodialysis patients: the TRANSFORM study. Adv Ther 2014; 31:1155-68. [PMID: 25367412 PMCID: PMC4245491 DOI: 10.1007/s12325-014-0161-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Indexed: 12/31/2022]
Abstract
Introduction There is scant real-world information on switching treatment for anemia associated with chronic kidney disease (CKD) from methoxy polyethylene glycol-epoetin beta (PEG-Epo) to darbepoetin alfa (DA). TRANSFORM was a multi-center, observational study designed to describe the time course of hemoglobin (Hb) concentration (primary outcome measure) and other parameters of clinical management of anemia in European hemodialysis patients in clinical practice before and after a switch from PEG-Epo to DA. Methods Eligible subjects were adult patients with CKD dialyzed at European dialysis centers for ≥26 weeks and treated with PEG-Epo for ≥14 weeks immediately prior to being switched to DA and no earlier than January 2011. Erythropoiesis-stimulating agent doses and Hb values were recorded for the 14-week pre-switch and 26-week post-switch periods. Results Of the 1,027 eligible patients enrolled at 42 hemodialysis centers in 7 European countries, 785 were included in analyses. Mean (95% confidence interval [CI]) Hb was generally stable: 11.19 (11.11, 11.26), 11.48 (11.40, 11.57), and 11.29 (11.20, 11.37) g/dL at month −1 pre-switch and months 3 and 6 post-switch, respectively. The geometric mean (95% CI) PEG-Epo dose at month −1 was 27.4 (26.0, 28.8) µg/week; DA dose was 29.4 (27.9, 30.9), 23.3 (21.9, 24.9), and 25.6 (24.1, 27.1) µg/week at months 1, 4, and 6, respectively. The geometric mean (95% CI) dose ratio at switching was 1.06 (1.01, 1.11). When stratifying by dose-ratio categories <0.8, 0.8–1.2, and >1.2 at switching, mean DA dose and Hb converged within narrow ranges by month 6 post-switch: 23.9–27.0 µg/week and 11.1–11.5 g/dL, respectively. Hb excursions <10 g/dL were less frequent post-switch versus pre-switch. Conclusion Mean Hb values remained within a narrow range following switching from PEG-Epo to DA in this population of hemodialysis patients. Time trends of mean Hb and DA dose indicate that physicians titrated DA doses post-switch, to attain Hb concentrations comparable to those attained pre-switch with PEG-Epo. Funding Amgen (Europe) GmbH, Zug, Switzerland. Electronic supplementary material The online version of this article (doi:10.1007/s12325-014-0161-5) contains supplementary material, which is available to authorized users.
Collapse
|
17
|
Eltzschig HK, Bratton DL, Colgan SP. Targeting hypoxia signalling for the treatment of ischaemic and inflammatory diseases. Nat Rev Drug Discov 2014; 13:852-69. [PMID: 25359381 PMCID: PMC4259899 DOI: 10.1038/nrd4422] [Citation(s) in RCA: 277] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hypoxia-inducible factors (HIFs) are stabilized during adverse inflammatory processes associated with disorders such as inflammatory bowel disease, pathogen infection and acute lung injury, as well as during ischaemia-reperfusion injury. HIF stabilization and hypoxia-induced changes in gene expression have a profound impact on the inflamed tissue microenvironment and on disease outcomes. Although the mechanism that initiates HIF stabilization may vary, the final molecular steps that control HIF stabilization converge on a set of oxygen-sensing prolyl hydroxylases (PHDs) that mark HIFs for proteasomal degradation. PHDs are therefore promising therapeutic targets. In this Review, we discuss the emerging potential and associated challenges of targeting the PHD-HIF pathway for the treatment of inflammatory and ischaemic diseases.
Collapse
Affiliation(s)
- Holger K Eltzschig
- Organ Protection Program, Department of Anesthesiology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Donna L Bratton
- Department of Pediatrics, National Jewish Health, Denver, Colorado 80206, USA
| | - Sean P Colgan
- Mucosal Inflammation Program, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| |
Collapse
|
18
|
Ibrahim IA, Mohamad UM, Darweesh HA, Rashad AM. Impact of hepcidin, interleukin 6, and other inflammatory markers with respect to erythropoietin on anemia in chronic hemodialysis patients. THE EGYPTIAN JOURNAL OF INTERNAL MEDICINE 2014. [DOI: 10.4103/1110-7782.132882] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
|
19
|
Methoxy polyethylene glycol-epoetin beta versus darbepoetin alfa for anemia in non-dialysis-dependent CKD: a systematic review. Int J Clin Pharm 2014; 36:1115-25. [DOI: 10.1007/s11096-014-0023-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 09/25/2014] [Indexed: 01/14/2023]
|
20
|
Warady BA, Silverstein DM. Management of anemia with erythropoietic-stimulating agents in children with chronic kidney disease. Pediatr Nephrol 2014; 29:1493-505. [PMID: 24005791 DOI: 10.1007/s00467-013-2557-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 03/29/2013] [Accepted: 06/20/2013] [Indexed: 11/28/2022]
Abstract
Anemia management is an important component of the care provided to children with chronic kidney disease (CKD) and influences both morbidity and mortality risk. The introduction of recombinant human erythropoietin to the treatment regimen three decades ago revolutionized the therapy and significantly decreased the need for repeated blood transfusions and exposure to associated risks. Recent data on the efficacy and complications associated with erythropoietic-stimulating agent (ESA) usage has, however, prompted a reassessment of treatment-related recommendations. This review will address these recommendations, in addition to describing pediatric outcomes associated with current ESAs and presenting information on alternative ESAs, many of which will likely soon be incorporated into clinical practice.
Collapse
Affiliation(s)
- Bradley A Warady
- Children's Mercy Hospitals and Clinics, 2401 Gillham Road, Kansas City, MO, 64108, USA,
| | | |
Collapse
|
21
|
Abstract
Hepcidin, the liver-produced peptide hormone, is a principal regulator of iron homeostasis. Abnormal hepcidin production has emerged as a causative factor in several common iron disorders. Hepcidin insufficiency results in iron overload in hereditary hemochromatosis and iron-loading anemias, whereas hepcidin excess causes or contributes to the development of iron-restricted anemias in inflammatory diseases, infections, some cancers and chronic kidney disease. Not surprisingly, hepcidin and related pathways have become the target for the development of novel therapeutics for iron disorders. In this review, we will summarize the strategies and development programs that have been devised for agonizing or antagonizing hepcidin and its receptor ferroportin.
Collapse
|
22
|
Chand S, Ward DG, Ng ZYV, Hodson J, Kirby H, Steele P, Rooplal I, Bantugon F, Iqbal T, Tselepis C, Drayson MT, Whitelegg A, Chowrimootoo M, Borrows R. Serum hepcidin-25 and response to intravenous iron in patients with non-dialysis chronic kidney disease. J Nephrol 2014; 28:81-8. [PMID: 24687402 DOI: 10.1007/s40620-014-0083-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 03/18/2014] [Indexed: 01/15/2023]
Abstract
BACKGROUND Hepcidin-25 is an iron regulator which reduces iron absorption and promotes sequestration in the reticulo-endothelial system. We investigated hepcidin and traditional iron storage marker utility in predicting haemoglobin increment following bolus intravenous iron. METHODS The cohort included 129 consecutive non-dialysis chronic kidney disease patients that attended for intravenous iron over a 6-month period. Serum hepcidin-25 levels (determined by mass spectrometry) pre iron infusion and 6 weeks post were compared with ferritin and transferrin saturation in multivariate models. RESULTS Log10 ferritin [coefficient 0.559 (0.435-0.684) p < 0.001] and log10 high-sensitive C-reactive protein [coefficient 0.092 (0.000-0.184) p = 0.049] were significantly associated with baseline log10 hepcidin-25 levels. Log10 estimated glomerular filtration rate was the only independent determinant of pre-infusion haemoglobin [coefficient 1.37 (0.16-2.59) p = 0.027]. Log10 hepcidin-25 was an independent predictor of haemoglobin increment 6 weeks following iron infusion [coefficient -0.84 (-1.38 to -0.31) p = 0.002]. Ferritin, transferrin saturation and hepcidin had similar predictive utility for a 1 g/dl haemoglobin increase (c-statistics: 0.68, 0.70, 0.69). CONCLUSIONS Hepcidin is an iron sensor marker which predicts the magnitude of haemoglobin increment following protocolised intravenous iron infusion. Although displaying similar predictive performance to ferritin and transferrin saturation, hepcidin may also play a mechanistic role.
Collapse
Affiliation(s)
- Sourabh Chand
- Renal Department, First Floor, Centre for Translational Inflammation Research, Queen Elizabeth Hospital Birmingham, University of Birmingham, Birmingham, B15 2WB, UK,
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Anemia has remained one of the most characteristic and visible manifestations of chronic renal failure for over 150 years. The pathogenesis of anemia of chronic kidney disease (CKD) is multifactorial with inadequate production of erythropoietin being the leading factor. The development of recombinant human erythropoietin (epoetin) in the late 1980s was a milestone in treatment of renal anemia. Despite new drugs, our 'good old friend' erythropoietin-stimulating agents are our everyday life in nephrology practice. It seems that peginesatide would not become a new approach for treating anemia of CKD patients, but rather a falling star. Several new strategies for treating the anemia of CKD are currently being investigated in clinical trials, including prolyl hydroxylase inhibitors and modulators of hepcidin activity, but their role in the management of this condition remains to be established. As shown by the expert in this review, we have to take into account not only the safety and convenience of administration but also cost-effectiveness, while biosimilars are consequently knocking at the doors of dialysis units more and more, particularly in Europe.
Collapse
Affiliation(s)
- Jolanta Malyszko
- Medical University, 2nd Department of Nephrology and Hypertension with Dialysis Unit , ul. M. Sklodowskiej-Curie 24a, 15-276 Bialystok , Poland
| |
Collapse
|
24
|
Choi P, Farouk M, Manamley N, Addison J. Dose conversion ratio in hemodialysis patients switched from darbepoetin alfa to PEG-epoetin beta: AFFIRM study. Adv Ther 2013; 30:1007-17. [PMID: 24173670 PMCID: PMC3898130 DOI: 10.1007/s12325-013-0063-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Indexed: 11/26/2022]
Abstract
INTRODUCTION There is limited information published on switching erythropoiesis-stimulating agent (ESA) treatment for anemia associated with chronic kidney disease (CKD) from darbepoetin alfa (DA) to methoxy polyethylene glycol-epoetin beta (PEG-Epo) outside the protocol of interventional clinical studies. AFFIRM (Aranesp Efficiency Relative to Mircera) was a retrospective, multi-site, observational study designed to estimate the population mean maintenance dose conversion ratio [DCR; dose ratio achieving comparable hemoglobin level (Hb) between two evaluation periods] in European hemodialysis patients whose treatment was switched from DA to PEG-Epo. METHODS Eligible patients had received hemodialysis for ≥ 12 months and DA for ≥ 7 months. Data were collected from 7 months before until 7 months after switching treatment. DCR was calculated for patients with Hb and ESA data available in both evaluation periods (EP; Months 1 and 2 were defined as the pre-switch EP, and Months 6 and 7 as the post-switch EP). Red blood cell transfusions pre- and post-switch were quantified. RESULTS Of 302 patients enrolled, 206 had data available for DCR analysis. The geometric mean DCR was 1.17 (95% CI 1.05, 1.29). Regression analysis indicated a non-linear relationship between pre- and post-switch ESA doses; DCR decreased with increasing pre-switch DA dose. The geometric mean weekly ESA doses were 24.1 μg DA in the pre-switch EP and 28.6 μg PEG-Epo in the post-switch EP. Mean Hb was 11.5 g/dL in the pre-switch EP and 11.4 g/dL in the post-switch EP. There were 16 transfusions and 34 units transfused in the pre-switch period, versus 48 transfusions and 95 units transfused post-switch. Excluding patients receiving a transfusion within 90 days of or during either EP, the DCR was 1.21 (95% CI 1.09, 1.35). CONCLUSION In these hemodialysis patients switched from DA to PEG-Epo the DCR was 1.17 and 1.21 after accounting for the effect of transfusions. The number of transfusions and units transfused increased approximately threefold from the pre-switch to the post-switch period.
Collapse
Affiliation(s)
- Peter Choi
- Department of Nephrology, John Hunter Hospital, Lookout Road, New Lambton Heights, NSW, 2305, Australia,
| | | | | | | |
Collapse
|
25
|
Magwood JS, Lebby A, Chen B, Kessler S, Norris L, Bennett CL. Emerging drugs for treatment of anemia of chronic kidney disease. Expert Opin Emerg Drugs 2013; 18:421-9. [DOI: 10.1517/14728214.2013.836490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
26
|
Valliant A, Hofmann RM. Managing dialysis patients who develop anemia caused by chronic kidney disease: focus on peginesatide. Int J Nanomedicine 2013; 8:3297-307. [PMID: 24023516 PMCID: PMC3767492 DOI: 10.2147/ijn.s44944] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Anemia in chronic kidney disease is a prevalent and expensive problem in the United States, and it is well documented that anemia worsens as glomerular filtration rates decline. The complications of severe anemia in this patient population contribute significantly to their overall morbidity with increased cardiovascular complications, decreased quality of life, and increased dependence on transfusions to maintain adequate hemoglobin levels. Erythropoietin-stimulating agents (ESAs) have revolutionized the treatment of anemia in this population, but there has been a great deal of controversy surrounding the quest for the ideal hemoglobin target. In addition, there are economic and practice management implications where anemia treatment is concerned, with ongoing refinement of Centers for Medicare and Medicaid Services-bundled payments. One of the newest additions to the arsenal used to fight anemia in end-stage renal disease patients is peginesatide (Omontys), a synthetic, PEGylated, peptide-based ESA that acts by stimulating the erythropoietin receptor. The role of peginesatide in the future treatment of anemia in chronic kidney disease remains uncertain, with new safety concerns being brought to attention as it emerges on the market, prompting a national recall.
Collapse
Affiliation(s)
- Amanda Valliant
- Division of Nephrology, University of Wisconsin, Madison, WI, USA
| | | |
Collapse
|
27
|
Shi X, Yang J, Zhu H, Ye L, Feng M, Li J, Huang H, Tao Q, Ye D, Sun LHK, Sun BNC, Sun CRY, Han G, Liu Y, Yao M, Zhou P, Ju D. Pharmacokinetics and pharmacodynamics of recombinant human EPO-Fc fusion protein in vivo. PLoS One 2013; 8:e72673. [PMID: 23977338 PMCID: PMC3747110 DOI: 10.1371/journal.pone.0072673] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 07/11/2013] [Indexed: 01/01/2023] Open
Abstract
In this study, the in vivo pharmacokinetics and pharmacodynamics
of a novel recombinant human erythropoietin (rhEPO) Fc fusion protein, rhEPO-Fc,
were studied in both rodents and rhesus monkeys. Animal models of anemia induced
by irradiation, cyclophosphamide and partial renal ablation were used to
evaluate therapeutic effects of rhEPO-Fc. We have demonstrated that serum
half-life of rhEPO-Fc was 29.5 to 38.9 h at doses of 8, 25, 80 µg/kg in
rhesus monkeys and 35.5 to 43.5 h at doses of 16, 50, 160 µg/kg in rats.
In anemia animal models, rhEPO-Fc dose-dependently (7.5–30.0 µg/kg
in mice, 5.4–21.4 µg/kg in rats and 5.0–10.0 µg/kg in
rhesus monkeys) increased reticulocyte level, followed by an increase of RBC
count, hemoglobin and hematocrit levels. At reduced intervention frequency of
weekly treatments, rhEPO-Fc showed similar hematopoietic effects as compared
with rhEPO given three times a week. These results indicated that rhEPO-Fc could
potentially be used in treatment of anemia and warrants future clinical
trials.
Collapse
Affiliation(s)
- Xunlong Shi
- Department of Biosynthesis, School of
Pharmacy, Fudan University, Shanghai, China
| | - Jianjun Yang
- Shanghai Meiye Biotech Institute, Shanghai,
China
| | - Haiyan Zhu
- Department of Biosynthesis, School of
Pharmacy, Fudan University, Shanghai, China
| | - Li Ye
- Department of Biosynthesis, School of
Pharmacy, Fudan University, Shanghai, China
| | - Meiqing Feng
- Department of Biosynthesis, School of
Pharmacy, Fudan University, Shanghai, China
| | - Jiyang Li
- Department of Biosynthesis, School of
Pharmacy, Fudan University, Shanghai, China
| | - Hai Huang
- Department of Biosynthesis, School of
Pharmacy, Fudan University, Shanghai, China
| | - Qun Tao
- Shanghai Meiye Biotech Institute, Shanghai,
China
| | - Dan Ye
- Shanghai Meiye Biotech Institute, Shanghai,
China
| | | | | | | | - Guizhen Han
- Department of Pharmacology, School of
Medicine, Fudan University, Shanghai, China
| | - Yuanyuan Liu
- Department of Pharmacology, School of
Medicine, Fudan University, Shanghai, China
| | - Minghui Yao
- Department of Pharmacology, School of
Medicine, Fudan University, Shanghai, China
| | - Pei Zhou
- Department of Biosynthesis, School of
Pharmacy, Fudan University, Shanghai, China
| | - Dianwen Ju
- Department of Biosynthesis, School of
Pharmacy, Fudan University, Shanghai, China
- * E-mail:
| |
Collapse
|
28
|
Jelkmann I, Jelkmann W. Impact of erythropoietin on intensive care unit patients. ACTA ACUST UNITED AC 2013; 40:310-8. [PMID: 24273484 DOI: 10.1159/000354128] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 08/06/2013] [Indexed: 12/13/2022]
Abstract
Anemia is common in intensive care unit (ICU) patients. Red blood cell (RBC) transfusions are mainstays of their treatment and can be life-saving. Allogeneic blood components inherently bear risks of infection and immune reactions. Although these risks are rare in developed countries, recombinant human erythropoietin (rhEpo) and other erythropoiesis-stimulating agents (ESAs) have been considered alternative anti-anemia treatment options. As summarized herein, however, most of the clinical studies suggest that ESAs are not usually advisable in ICU patients unless approved indications exist (e.g., renal disease). First, ESAs act in a delayed way, inducing an increase in reticulocytes only after a lag of 3-4 days. Second, many critically ill patients present with ESA resistance as inflammatory mediators impair erythropoietic cell proliferation and iron availability. Third, the ESA doses used for treatment of ICU patients are very high. Fourth, ESAs are not legally approved for general use in ICU patients. Solely in distinct cases, such as Jehovah's Witnesses who refuse allogeneic blood transfusions due to religious beliefs, ESAs may be considered an exceptional therapy.
Collapse
Affiliation(s)
- Ines Jelkmann
- Department of Surgery, University of Lübeck, Germany
| | | |
Collapse
|
29
|
Hörl WH. Differentiating factors between erythropoiesis-stimulating agents: an update to selection for anaemia of chronic kidney disease. Drugs 2013; 73:117-30. [PMID: 23338536 DOI: 10.1007/s40265-012-0002-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Erythropoiesis-stimulating agents (ESAs) have become a hallmark of anaemia therapy in patients with chronic kidney disease (CKD). Although different ESAs are available for the treatment of renal anaemia, each nephrologist should select a single ESA for an individual patient. Epoetin alfa and epoetin beta have been used 1-3 times weekly but extended-interval dosing up to every 4 weeks is also effective in a substantial majority of CKD patients. However, the epoetin dose necessary to achieve or maintain target haemoglobin (Hb) levels increases substantially as the dosing interval increases. Subcutaneous administration of short-acting ESAs is more effective than the intravenous route of administration. Darbepoetin alfa and the continuous erythropoietin receptor activator (CERA) have been developed as a treatment for anaemia with extended administration intervals (every 2 weeks and every 4 weeks, respectively). Dose requirements for these long-acting ESAs are independent of the route of administration. Patents of short-acting ESAs have expired, which has opened the field for biosimilars. Epoetin biosimilars approved by the European Medicines Agency (EMA) or the US Food and Drug Administration (FDA) have been shown to have a comparable efficacy and safety profile to their originators. An alarming increase in pure red cell aplasia (PRCA) in Thailand with follow-on epoetins manufactured in Asia (but also those manufactured in Latin America) indicates that stringent country-specific approval and pharmacovigilance protocols for ESAs manufactured in non-North American and non-EU European countries are urgently needed. Two PRCA cases occurring with subcutaneous HX575 (one certain, one likely) indicate that chances of inducing a more immunogenic product are unpredictable, even with a biosimilar epoetin approved under the EMA biosimilar approval pathway. Phase III clinical trials with peginesatide, a pegylated synthetic peptide-based ESA without any homology to erythropoietin raised safety concerns in non-dialysis CKD patients but not in dialysis patients.
Collapse
Affiliation(s)
- Walter H Hörl
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
30
|
Jelkmann W. Physiology and pharmacology of erythropoietin. ACTA ACUST UNITED AC 2013; 40:302-9. [PMID: 24273483 DOI: 10.1159/000356193] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 07/03/2013] [Indexed: 12/13/2022]
Abstract
Human erythropoietin (Epo) is a 30.4 kDa glycoprotein hormone composed of a single 165 amino acid residues chain to which four glycans are attached. The kidneys are the primary sources of Epo, its synthesis is controlled by hypoxia-inducible transcription factors (HIFs). Epo is an essential factor for the viability and proliferation of erythrocytic progenitors. Whether Epo exerts cytoprotection outside the bone marrow still needs to be clarified. Epo deficiency is the primary cause of the anemia in chronic kidney disease (CKD). Treatment with recombinant human Epo (rhEpo, epoetin) can be beneficial not only in CKD but also for other indications, primarily anemia in cancer patients receiving chemotherapy. Considering unwanted events, the administration of rhEpo or its analogs may increase the incidence of thromboembolism. The expiry of the patents for the original epoetins has initiated the production of similar biological medicinal products ('biosimilars'). Furthermore, analogs (darbepoetin alfa, methoxy PEG-epoetin beta) with prolonged survival in circulation have been developed ('biobetter'). New erythropoiesis-stimulating agents are in clinical trials. These include compounds that augment erythropoiesis directly (e.g. Epo mimetic peptides or activin A binding protein) and chemicals that act indirectly by stimulating endogenous Epo synthesis (HIF stabilizers).
Collapse
|
31
|
|
32
|
Myllyharju J. Prolyl 4-hydroxylases, master regulators of the hypoxia response. Acta Physiol (Oxf) 2013; 208:148-65. [PMID: 23489300 DOI: 10.1111/apha.12096] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 11/07/2012] [Accepted: 03/08/2013] [Indexed: 12/13/2022]
Abstract
A decrease in oxygenation is a life-threatening situation for most organisms. An evolutionarily conserved efficient and rapid hypoxia response mechanism activated by a hypoxia-inducible transcription factor (HIF) is present in animals ranging from the simplest multicellular phylum Placozoa to humans. In humans, HIF induces the expression of more than 100 genes that are required to increase oxygen delivery and to reduce oxygen consumption. As its name indicates HIF is found at protein level only in hypoxic cells, whereas in normoxia, it is degraded by the proteasome pathway. Prolyl 4-hydroxylases, enzymes that require oxygen in their reaction, are the cellular oxygen sensors regulating the stability of HIF. In normoxia, 4-hydroxyproline residues formed in the α-subunit of HIF by these enzymes lead to its ubiquitination by the von Hippel-Lindau E3 ubiquitin ligase and immediate destruction in proteasomes thus preventing the formation of a functional HIF αβ dimer. Prolyl 4-hydroxylation is inhibited in hypoxia, facilitating the formation of the HIF dimer and activation of its target genes, such as those for erythropoietin and vascular endothelial growth factor. This review starts with a summary of the molecular and catalytic properties and individual functions of the four HIF prolyl 4-hydroxylase isoenzymes. Induction of the hypoxia response via inhibition of the HIF prolyl 4-hydroxylases may provide a novel therapeutic target in the treatment of hypoxia-associated diseases. The current status of studies aiming at such therapeutic approaches is introduced in the final part of this review.
Collapse
Affiliation(s)
- J. Myllyharju
- Oulu Center for Cell-Matrix Research; Biocenter Oulu and Department of Medical Biochemistry and Molecular Biology; University of Oulu; Oulu; Finland
| |
Collapse
|
33
|
Abstract
INTRODUCTION Erythropoiesis-stimulating agents (ESAs) are the mainstay of treatment in anemic chronic kidney disease (CKD) patients. A tailored ESA therapy should combine maximal efficacy and safety with greatest convenience in dosing. Peginesatide, recently approved in the US for once-monthly dosing in adult patients on dialysis, is a promising novel PEGylated erythropoietin-mimetic peptide for the treatment of renal disease-induced anemia. AREAS COVERED Published animal and human studies that evaluated the pharmacodynamics, pharmacokinetics, clinical efficacy and safety of peginesatide were critically analyzed. EXPERT OPINION Peginesatide has a well-studied pharmacological and immunological profile, and latest published data favor the use of peginesatide in place of epoetin in dialysis patients. A more detailed evaluation of its safety profile particularly in trials with CKD patients not requiring dialysis is urgently needed, as peginesatide could be a perfect treatment solution for these patients. In addition, clinical long-term data and results from supplemental studies, e.g., with the PEGylated continuous erythropoietin receptor activator as comparator, should briefly follow. The fate of peginesatide on the highly competitive ESA market is currently not predictable and depends on safety and efficacy results of upcoming trials as well as finally on market and price policy.
Collapse
Affiliation(s)
- Holger Schmid
- University of Munich, Department of Internal Medicine, Munich, D-80336, Germany.
| |
Collapse
|
34
|
|
35
|
Dewamitta SR, Russell MR, Nandurkar H, Walkley CR. Darbepoietin-alfa has comparable erythropoietic stimulatory effects to recombinant erythropoietin whilst preserving the bone marrow microenvironment. Haematologica 2012; 98:686-90. [PMID: 23242598 DOI: 10.3324/haematol.2012.078709] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Erythropoiesis stimulating agents are widely used for the treatment of anemia. Recently, we reported erythroid expansion with impaired B lymphopoiesis and loss of trabecular bone in C57BL/6 mice following ten days of treatment with low-dose short acting recombinant human erythropoietin. We have assessed erythropoietin against longer-acting darbepoietin-alfa at a comparable erythroid stimulatory dosage regime. Darbepoietin-alfa and erythropoietin induced similar in vivo erythropoietic expansion. Both agents induced an expansion of the colony-forming unit-erythroid populations. However, unlike erythropoietin, darbepoietin-alfa did not impair bone marrow B lymphopoiesis. Strikingly the bone loss observed with erythropoietin was not apparent following darbepoietin-alfa treatment. This analysis demonstrates that whilst darbepoietin-alfa has similar in vivo erythropoietic potency to erythropoietin, it preserves the bone marrow microenvironment. Thus erythropoietin and darbepoietin-alfa manifest different action showing that erythropoiesis stimulating agents have differential non-erythroid effects dependent on their duration of action.
Collapse
Affiliation(s)
- Sita R Dewamitta
- St Vincent’s Institute of Medical Research and Department of Medicine at St. Vincent’s Hospital, University of Melbourne, Fitzroy,Victoria, Australia
| | | | | | | |
Collapse
|
36
|
Mass spectrometric detection of peginesatide in human urine in doping control analysis. J Pharm Biomed Anal 2012; 70:512-7. [DOI: 10.1016/j.jpba.2012.07.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 07/19/2012] [Accepted: 07/22/2012] [Indexed: 11/19/2022]
|
37
|
Möller I, Thomas A, Geyer H, Schänzer W, Thevis M. Development and validation of a mass spectrometric detection method of peginesatide in dried blood spots for sports drug testing. Anal Bioanal Chem 2012; 403:2715-24. [DOI: 10.1007/s00216-012-6043-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 03/28/2012] [Accepted: 04/10/2012] [Indexed: 11/28/2022]
|
38
|
Möller I, Thomas A, Wingender A, Machnik M, Schänzer W, Thevis M. Detection of peginesatide in equine serum using liquid chromatography-tandem mass spectrometry for doping control purposes. EUROPEAN JOURNAL OF MASS SPECTROMETRY (CHICHESTER, ENGLAND) 2012; 18:407-412. [PMID: 22971699 DOI: 10.1255/ejms.1189] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Erythropoietin (EPO) and its recombinant analogues are suspected to be illicitly administered to horses for performance enhancing purposes and, consequently, prohibited in equine sports. Recently, a new erythropoiesis-stimulating agent, peginesatide (Omontys, formerly referred to as Hematide), belonging to the upcoming class of EPO-mimetic peptides, received approval for the treatment of anaemia in humans with chronic kidney disease on dialysis. As the pegylated dimeric peptide of approximately 45 kDa without sequence homology to EPO is not detectable by conventional EPO detection assays, specific methods are bound to be established for horse sports drug testing. Thus, by fortifying equine serum with peginesatide, an approach consisting of a proteolytic digestion with subtilisin after protein precipitation was developed, eventually targeting a proteotypic and xenobiotic pentapeptide which is easily accessible to liquid chromatography- tandem mass spectrometry analysis. The method was validated for qualitative purposes and demonstrated to be specific, precise (relative standard deviations below 14%), sensitive (limit of detection 10 ng mL(-1)) and linear. Being simple, cost-effective and readily transferable to other doping control laboratories, a mass spectrometric assay for the detection of therapeutic concentrations of peginesatide in equine serum is, in terms of preventive doping research, applicable to routine analysis shortly after approval of the drug.
Collapse
Affiliation(s)
- Ines Möller
- Institute of Biochemistry-Center for Preventive Doping Research, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933 Cologne, Germany
| | | | | | | | | | | |
Collapse
|