1
|
Ekramzadeh M, Kalantar-Zadeh K, Kopple JD. The Relevance of Phytate for the Treatment of Chronic Kidney Disease. Clin J Am Soc Nephrol 2024; 19:1341-1355. [PMID: 39110986 PMCID: PMC11469791 DOI: 10.2215/cjn.0000000000000558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 07/30/2024] [Indexed: 10/13/2024]
Abstract
Diets high in plant-based foods are commonly recommended for people with CKD. One putative advantage of these diets is reduced intestinal phosphate absorption. This effect has been ascribed to phytic acid (myoinositol hexaphosphoric acid) and its anion, phytate, that are present in many plant foods, particularly in the seeds, nuts, grains, and fruits of plants. This article reviews the structure and many actions of phytate with particular reference to its potential effects on people with CKD. Phytate binds avidly to and can reduce gastrointestinal absorption of the phosphate anion and many macrominerals and trace elements including iron, zinc, calcium, and magnesium. This has led some opinion leaders to label phytate as an anti-nutrient. The human intestine lacks phytase; hence, phytate is essentially not degraded in the small intestine. A small amount of phytate is absorbed from the small intestine, although phytate bound to phosphate is poorly absorbed. Clinical trials in maintenance hemodialysis patients indicate that intravenously administered phytate may decrease hydroxyapatite formation, vascular calcification, and calciphylaxis. Orally administered phytate or in vitro studies indicate that phytate may also reduce osteoporosis, urinary calcium calculi formation, and dental plaque formation. Phytate seems to have anti-inflammatory and antioxidant effects, at least partly because of its ability to chelate iron. Other potential therapeutic roles for phytate, not definitively established, include suppression of cancer formation, reduction in cognitive decline that occurs with aging, and amelioration of certain neurodegenerative diseases and several gastrointestinal and metabolic disorders. These latter potential benefits of phytate are supported by cell or animal research or observational studies in humans. Many of the above disorders are particularly common in patients with CKD. Definitive clinical trials to identify potential therapeutic benefits of phytate in patients with CKD are clearly warranted.
Collapse
Affiliation(s)
- Maryam Ekramzadeh
- David Geffen School of Medicine at UCLA and the UCLA Fielding School of Public Health, Los Angeles, CA
| | - Kamyar Kalantar-Zadeh
- David Geffen School of Medicine at UCLA and the UCLA Fielding School of Public Health, Los Angeles, CA
| | - Joel D. Kopple
- David Geffen School of Medicine at UCLA and the UCLA Fielding School of Public Health, Los Angeles, CA
| |
Collapse
|
2
|
Zhu Y, Lai Y, Hu Y, Fu Y, Zhang Z, Lin N, Huang W, Zheng L. The mechanisms underlying acute myocardial infarction in chronic kidney disease patients undergoing hemodialysis. Biomed Pharmacother 2024; 177:117050. [PMID: 38968794 DOI: 10.1016/j.biopha.2024.117050] [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: 04/17/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/07/2024] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of death in chronic kidney disease (CKD). Hemodialysis is one of the main treatments for patients with end-stage kidney disease. Epidemiological data has shown that acute myocardial infarction (AMI) accounts for the main reason for death in patients with CKD under hemodialysis therapy. Immune dysfunction and changes in metabolism (including a high level of inflammatory cytokines, a disorder of lipid and mineral ion homeostasis, accumulation of uremic toxins et al.) during CKD can deteriorate stability of atherosclerotic plaque and promote vascular calcification, which are exactly the pathophysiological mechanisms underlying the occurrence of AMI. Meanwhile, the hemodialysis itself also has adverse effects on lipoprotein, the immune system and hemodynamics, which contribute to the high incidence of AMI in these patients. This review aims to summarize the mechanisms and further promising methods of prevention and treatment of AMI in CKD patients undergoing hemodialysis, which can provide an excellent paradigm for exploring the crosstalk between the kidney and cardiovascular system.
Collapse
Affiliation(s)
- Yujie Zhu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Yuchen Lai
- School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yuxuan Hu
- Hubei University of Science and Technology, Xianning 437100, China
| | - Yiwen Fu
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Zheng Zhang
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Nan Lin
- Department of Cardiology, Fujian Provincial Hospital, Fuzhou 350013, China
| | - Wei Huang
- Department of Cardiology, General Hospital of Central Theater Command, No.627, Wuluo Road, Wuhan 430070, China.
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China; Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, Beijing Institute of Brain Disorders, The Capital Medical University, Beijing 100050, China.
| |
Collapse
|
3
|
Olmaz R, Selen T, Gungor O. Vascular calcification inhibitors and cardiovascular events in peritoneal dialysis patients. Ther Apher Dial 2024; 28:169-181. [PMID: 38013624 DOI: 10.1111/1744-9987.14091] [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: 10/05/2023] [Revised: 11/08/2023] [Accepted: 11/16/2023] [Indexed: 11/29/2023]
Abstract
The prevalence of cardiovascular diseases is high among patients with chronic kidney disease (CKD) and peritoneal dialysis (PD) patients, which increases morbidity and mortality in this population and represents a significant financial burden for both the patients and the healthcare systems. Vascular calcification (VC) is associated with increased morbidity and mortality and VC risk is higher in patients with CKD than in healthy individuals. Calcification inhibitors, compounds that inhibit VC, were discovered as a result of efforts to explain why some patients are spared. It was found that certain proteins (e.g., fetuin-A, osteopontin, osteoprotegerin, bone morphogenetic protein-7) inhibit calcification in dialysis patients. In this narrative review, we provide an overview of known calcification inhibitors, describe the relevant regulatory mechanisms, and discuss their relation to VC development in PD patients.
Collapse
Affiliation(s)
- Refik Olmaz
- Department of Nephrology, Mersin City Hospital, Mersin, Turkey
| | - Tamer Selen
- Department of Nephrology, Duzce Ataturk State Hospital, Duzce, Turkey
| | - Ozkan Gungor
- Department of Nephrology, Faculty of Medicine, Kahramanmaras Sütcü Imam University, Kahramanmaras, Turkey
| |
Collapse
|
4
|
He M, Wei W, Zhang Y, Xiang Z, Peng D, Kasimumali A, Rong S. Gut microbial metabolites SCFAs and chronic kidney disease. J Transl Med 2024; 22:172. [PMID: 38369469 PMCID: PMC10874542 DOI: 10.1186/s12967-024-04974-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 02/11/2024] [Indexed: 02/20/2024] Open
Abstract
The global incidence of Chronic Kidney Disease (CKD) is steadily escalating, with discernible linkage to the intricate terrain of intestinal microecology. The intestinal microbiota orchestrates a dynamic equilibrium in the organism, metabolizing dietary-derived compounds, a process which profoundly impacts human health. Among these compounds, short-chain fatty acids (SCFAs), which result from microbial metabolic processes, play a versatile role in influencing host energy homeostasis, immune function, and intermicrobial signaling, etc. SCFAs emerge as pivotal risk factors influencing CKD's development and prognosis. This paper review elucidates the impact of gut microbial metabolites, specifically SCFAs, on CKD, highlighting their role in modulating host inflammatory responses, oxidative stress, cellular autophagy, the immune milieu, and signaling cascades. An in-depth comprehension of the interplay between SCFAs and kidney disease pathogenesis may pave the way for their utilization as biomarkers for CKD progression and prognosis or as novel adjunctive therapeutic strategies.
Collapse
Affiliation(s)
- Meng He
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Wenqian Wei
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yichen Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zhouxia Xiang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Dan Peng
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Ayijiaken Kasimumali
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Shu Rong
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
5
|
Dai Z, Zhang X. Pathophysiology and Clinical Impacts of Chronic Kidney Disease on Coronary Artery Calcification. J Cardiovasc Dev Dis 2023; 10:jcdd10050207. [PMID: 37233174 DOI: 10.3390/jcdd10050207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/27/2023] Open
Abstract
The global prevalence of chronic kidney disease (CKD) has increased in recent years. Adverse cardiovascular events have become the main cause of life-threatening events in patients with CKD, and vascular calcification is a risk factor for cardiovascular disease. Vascular calcification, especially coronary artery calcification, is more prevalent, severe, rapidly progressive, and harmful in patients with CKD. Some features and risk factors are unique to vascular calcification in patients with CKD; the formation of vascular calcification is not only influenced by the phenotypic transformation of vascular smooth muscle cells, but also by electrolyte and endocrine dysfunction, uremic toxin accumulation, and other novel factors. The study on the mechanism of vascular calcification in patients with renal insufficiency can provide a basis and new target for the prevention and treatment of this disease. This review aims to illustrate the impact of CKD on vascular calcification and to discuss the recent research data on the pathogenesis and factors involved in vascular calcification, mainly focusing on coronary artery calcification, in patients with CKD.
Collapse
Affiliation(s)
- Zhuoming Dai
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xiangyu Zhang
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| |
Collapse
|
6
|
Abstract
Patients with chronic kidney disease (CKD) exhibit tremendously elevated risk for cardiovascular disease, particularly ischemic heart disease, due to premature vascular and cardiac aging and accelerated ectopic calcification. The presence of cardiovascular calcification associates with increased risk in patients with CKD. Disturbed mineral homeostasis and diverse comorbidities in these patients drive increased systemic cardiovascular calcification in different manifestations with diverse clinical consequences, like plaque instability, vessel stiffening, and aortic stenosis. This review outlines the heterogeneity in calcification patterning, including mineral type and location and potential implications on clinical outcomes. The advent of therapeutics currently in clinical trials may reduce CKD-associated morbidity. Development of therapeutics for cardiovascular calcification begins with the premise that less mineral is better. While restoring diseased tissues to a noncalcified homeostasis remains the ultimate goal, in some cases, calcific mineral may play a protective role, such as in atherosclerotic plaques. Therefore, developing treatments for ectopic calcification may require a nuanced approach that considers individual patient risk factors. Here, we discuss the most common cardiac and vascular calcification pathologies observed in CKD, how mineral in these tissues affects function, and the potential outcomes and considerations for therapeutic strategies that seek to disrupt the nucleation and growth of mineral. Finally, we discuss future patient-specific considerations for treating cardiac and vascular calcification in patients with CKD-a population in need of anticalcification therapies.
Collapse
Affiliation(s)
- Joshua D. Hutcheson
- Department of Biomedical Engineering, Florida International University, Miami, FL (J.D.H.)
| | - Claudia Goettsch
- Department of Internal Medicine I, Division of Cardiology, Medical Faculty, RWTH Aachen University, Germany (C.G.)
| |
Collapse
|
7
|
Pan W, Jie W, Huang H. Vascular calcification: Molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2023; 4:e200. [PMID: 36620697 PMCID: PMC9811665 DOI: 10.1002/mco2.200] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 01/05/2023] Open
Abstract
Vascular calcification (VC) is recognized as a pathological vascular disorder associated with various diseases, such as atherosclerosis, hypertension, aortic valve stenosis, coronary artery disease, diabetes mellitus, as well as chronic kidney disease. Therefore, it is a life-threatening state for human health. There were several studies targeting mechanisms of VC that revealed the importance of vascular smooth muscle cells transdifferentiating, phosphorous and calcium milieu, as well as matrix vesicles on the progress of VC. However, the underlying molecular mechanisms of VC need to be elucidated. Though there is no acknowledged effective therapeutic strategy to reverse or cure VC clinically, recent evidence has proved that VC is not a passive irreversible comorbidity but an active process regulated by many factors. Some available approaches targeting the underlying molecular mechanism provide promising prospects for the therapy of VC. This review aims to summarize the novel findings on molecular mechanisms and therapeutic interventions of VC, including the role of inflammatory responses, endoplasmic reticulum stress, mitochondrial dysfunction, iron homeostasis, metabolic imbalance, and some related signaling pathways on VC progression. We also conclude some recent studies on controversial interventions in the clinical practice of VC, such as calcium channel blockers, renin-angiotensin system inhibitions, statins, bisphosphonates, denosumab, vitamins, and ion conditioning agents.
Collapse
Affiliation(s)
- Wei Pan
- Department of Cardiology, the Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Joint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic DiseaseSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Wei Jie
- Department of Cardiology, the Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Joint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic DiseaseSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Hui Huang
- Department of Cardiology, the Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Joint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic DiseaseSun Yat‐sen UniversityShenzhenGuangdongChina
| |
Collapse
|
8
|
Pujol A, Sanchis P, Grases F, Masmiquel L. Phytate Intake, Health and Disease: "Let Thy Food Be Thy Medicine and Medicine Be Thy Food". Antioxidants (Basel) 2023; 12:antiox12010146. [PMID: 36671007 PMCID: PMC9855079 DOI: 10.3390/antiox12010146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Phytate (myo-inositol hexakisphosphate or InsP6) is the main phosphorus reservoir that is present in almost all wholegrains, legumes, and oilseeds. It is a major component of the Mediterranean and Dietary Approaches to Stop Hypertension (DASH) diets. Phytate is recognized as a nutraceutical and is classified by the Food and Drug Administration (FDA) as Generally Recognized As Safe (GRAS). Phytate has been shown to be effective in treating or preventing certain diseases. Phytate has been shown to inhibit calcium salt crystallization and, therefore, to reduce vascular calcifications, calcium renal calculi and soft tissue calcifications. Moreover, the adsorption of phytate to the crystal faces can inhibit hydroxyapatite dissolution and bone resorption, thereby playing a role in the treatment/prevention of bone mass loss. Phytate has a potent antioxidation and anti-inflammatory action. It is capable of inhibiting lipid peroxidation through iron chelation, reducing iron-related free radical generation. As this has the effect of mitigating neuronal damage and loss, phytate shows promise in the treatment/prevention of neurodegenerative disease. It is reported that phytate improves lipid and carbohydrate metabolism, increases adiponectin, decreases leptin and reduces protein glycation, which is linked with macrovascular and microvascular diabetes complications. In this review, we summarize the benefits of phytate intake as seen in in vitro, animal model, epidemiological and clinical trials, and we also identify questions to answer in the future.
Collapse
Affiliation(s)
- Antelm Pujol
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands (IdISBa), 07198 Palma de Mallorca, Spain
- Correspondence: (A.P.); (L.M.)
| | - Pilar Sanchis
- Laboratory of Renal Lithiasis Research, Deptartment of Chemistry, University of Balearic Islands, Health Research Institute of Balearic Islands, (IdISBa), 07122 Palma de Mallorca, Spain
| | - Felix Grases
- Laboratory of Renal Lithiasis Research, Deptartment of Chemistry, University of Balearic Islands, Health Research Institute of Balearic Islands, (IdISBa), 07122 Palma de Mallorca, Spain
| | - Luis Masmiquel
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands (IdISBa), 07198 Palma de Mallorca, Spain
- Correspondence: (A.P.); (L.M.)
| |
Collapse
|
9
|
Kaur R, Singh R. Mechanistic insights into CKD-MBD-related vascular calcification and its clinical implications. Life Sci 2022; 311:121148. [DOI: 10.1016/j.lfs.2022.121148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/22/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
|
10
|
Ibrahim A, Rehman MEU, Khan MZ. Role of myoinositol hexaphosphate (SNF 472) in calciphylaxis: an update. Ther Apher Dial 2022; 26:848-849. [PMID: 35192233 DOI: 10.1111/1744-9987.13817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Atif Ibrahim
- The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | | | | |
Collapse
|
11
|
Finger M, Finger E, Bellucci A, Malieckal DA. Medical management for the prevention of kidney stones. Postgrad Med J 2021; 99:postgradmedj-2021-140971. [PMID: 34930814 DOI: 10.1136/postgradmedj-2021-140971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/23/2021] [Indexed: 12/17/2022]
Abstract
The alarming fact is that approximately one out of every 10 of us will have a kidney stone during our lifetime. The increasing prevalence and associated costs of kidney stones have resulted in it being one of the most commonly encountered and impactful medical conditions. Contributing factors include, but are not limited to, diet, climate, genetics, medications, activity and underlying medical conditions. Symptoms generally parallel stone size. Treatment varies from supportive to procedural (invasive and non-invasive). Prevention remains the best way to avoid this condition especially given the high recurrence rate. First time stone formers require counselling regarding dietary adjustments. Certain risk factors ultimately require a more in-depth metabolic investigation, especially if stones are recurrent. Ultimately, management is defined by stone composition. Where appropriate, we review both pharmacologic and non-pharmacologic options. Pivotal to successful prevention is patient education and the encouragement of compliance with the appropriate regimen.
Collapse
Affiliation(s)
- Mark Finger
- Medicine-Nephrology, Northwell Health, Great Neck, New York, USA
| | - Evan Finger
- Medicine, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
12
|
Zhao T, Yong X, Zhao Z, Dolce V, Li Y, Curcio R. Research status of Bacillus phytase. 3 Biotech 2021; 11:415. [PMID: 34485008 DOI: 10.1007/s13205-021-02964-9] [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: 05/28/2021] [Accepted: 07/27/2021] [Indexed: 10/20/2022] Open
Abstract
Phytic acid is abundant in seeds, roots and stems of plants, it acts as an anti-nutrient in food and feed industry, since it affects the absorption of nutrients by humans and monogastric animals. Furthermore, phosphorus produced through its decomposition by microorganisms can cause environmental pollution. Phytase degrades phytic acid generating precursors of inositol that can be used in clinical practice; in addition, phytase treatment can minimize the anti-nutritional effect of phytic acid. The use of phytase synthesized from Bacillus is more advantageous due to its high activity. Additionally, its good heat resistance under neutral conditions greatly fills the gap of commercial utilization of acid phytase. In this review, we summarize the latest research results on Bacillus phytase, including its physiological and biochemical characteristics, molecular structure information, calcium effects on its catalytic activity and stability, its catalytic mechanism and molecular modification.
Collapse
Affiliation(s)
- Ting Zhao
- College of Life Science and Technology, Xinjiang University, Urumqi, China
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, China
| | - Xihao Yong
- College of Life Science and Technology, Xinjiang University, Urumqi, China
- Faculty of Bioengineering, Sichuan University of Science and Engineering, Zigong, People's Republic of China
| | - Ziming Zhao
- Faculty of Bioengineering, Sichuan University of Science and Engineering, Zigong, People's Republic of China
| | - Vincenza Dolce
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Yuan Li
- College of Life Science and Technology, Xinjiang University, Urumqi, China
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, Bari, Italy
| | - Rosita Curcio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| |
Collapse
|
13
|
Millán Á, Lanzer P, Sorribas V. The Thermodynamics of Medial Vascular Calcification. Front Cell Dev Biol 2021; 9:633465. [PMID: 33937234 PMCID: PMC8080379 DOI: 10.3389/fcell.2021.633465] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/18/2021] [Indexed: 12/14/2022] Open
Abstract
Medial vascular calcification (MVC) is a degenerative process that involves the deposition of calcium in the arteries, with a high prevalence in chronic kidney disease (CKD), diabetes, and aging. Calcification is the process of precipitation largely of calcium phosphate, governed by the laws of thermodynamics that should be acknowledged in studies of this disease. Amorphous calcium phosphate (ACP) is the key constituent of early calcifications, mainly composed of Ca2+ and PO4 3- ions, which over time transform into hydroxyapatite (HAP) crystals. The supersaturation of ACP related to Ca2+ and PO4 3- activities establishes the risk of MVC, which can be modulated by the presence of promoter and inhibitor biomolecules. According to the thermodynamic parameters, the process of MVC implies: (i) an increase in Ca2+ and PO4 3- activities (rather than concentrations) exceeding the solubility product at the precipitating sites in the media; (ii) focally impaired equilibrium between promoter and inhibitor biomolecules; and (iii) the progression of HAP crystallization associated with nominal irreversibility of the process, even when the levels of Ca2+ and PO4 3- ions return to normal. Thus, physical-chemical processes in the media are fundamental to understanding MVC and represent the most critical factor for treatments' considerations. Any pathogenetical proposal must therefore comply with the laws of thermodynamics and their expression within the medial layer.
Collapse
Affiliation(s)
- Ángel Millán
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza, Spain
| | - Peter Lanzer
- Division of Cardiovascular Disease, Department of Internal Medicine, Health Care Center Bitterfeld, Bitterfeld-Wolfen gGmbH, Bitterfeld-Wolfen, Germany
| | - Víctor Sorribas
- Molecular Toxicology Group, Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
14
|
Piccoli GB, Torreggiani M, Gendrot L, Nielsen L. Setting the clock back: new hope for dialysis patients. Sodium thiosulphate and the regression of vascular calcifications. J Nephrol 2021; 34:23-25. [PMID: 32418117 PMCID: PMC7229437 DOI: 10.1007/s40620-020-00744-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Giorgina Barbara Piccoli
- Nephrologie, Centre Hospitalier Le Mans, Avenue Roubillard 194, 72000, Le Mans, France. .,Department of Clinical and Biological Sceinces, University of Torino, Turin, Italy.
| | - Massimo Torreggiani
- Nephrologie, Centre Hospitalier Le Mans, Avenue Roubillard 194, 72000, Le Mans, France
| | - Lurlynis Gendrot
- Nephrologie, Centre Hospitalier Le Mans, Avenue Roubillard 194, 72000, Le Mans, France
| | - Louise Nielsen
- Nephrologie, Centre Hospitalier Le Mans, Avenue Roubillard 194, 72000, Le Mans, France
| |
Collapse
|
15
|
Bellasi A, Raggi P, Bover J, Bushinsky DA, Chertow GM, Ketteler M, Rodriguez M, Sinha S, Salcedo C, Garg R, Gold A, Perelló J. Trial design and baseline characteristics of CaLIPSO: a randomized, double-blind placebo-controlled trial of SNF472 in patients receiving haemodialysis with cardiovascular calcification. Clin Kidney J 2021; 14:366-374. [PMID: 33564440 PMCID: PMC7857813 DOI: 10.1093/ckj/sfz144] [Citation(s) in RCA: 6] [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: 07/22/2019] [Accepted: 09/13/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The objective of CaLIPSO, a Phase 2b, randomized, double-blind, placebo-controlled clinical trial, is to test the hypothesis that myo-inositol hexaphosphate (SNF472) attenuates the progression of cardiovascular calcification in patients receiving maintenance haemodialysis. Here we report the trial design and baseline characteristics of trial participants. METHODS Adult patients on maintenance haemodialysis (≥6 months) with an Agatston coronary artery calcium score, as measured by a multidetector computed tomography scanner, of 100-3500 U were enrolled. Patients were stratified by Agatston score (100-<400, 400-1000 or >1000 U) and randomized in a 1:1:1 ratio to receive placebo, SNF472 300 mg or SNF472 600 mg administered intravenously three times weekly during each haemodialysis session. RESULTS Overall, 274 patients were randomized. The mean age of trial participants was 63.6 (standard deviation 8.9) years and 39% were women. The coronary artery, aorta and aortic valve median (25th-75th percentile) Agatston scores at baseline were 730 U (315-1435), 1728 U (625-4978) and 103 U (31-262), respectively, and the median (25th-75th percentile) calcium volume scores at baseline were 666 (310-1234), 1418 (536-4052) and 107 (38-278), respectively. Older age and diabetes mellitus were associated with higher calcium scores at baseline. CONCLUSIONS The CaLIPSO trial enrolled patients on haemodialysis with pre-existent cardiovascular calcification to test the hypothesis that SNF472 attenuates its progression in the coronary arteries, aorta and aortic valve.
Collapse
Affiliation(s)
- Antonio Bellasi
- Research, Innovation and Brand Reputation Unit, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Paolo Raggi
- Department of Medicine, Mazankowski Alberta Heart Institute and University of Alberta, Edmonton, AB, Canada
| | - Jordi Bover
- Department of Nephrology, Fundació Puigvert and Universitat Autònoma, IIB Sant Pau, REDinREN, Barcelona, Spain
| | - David A Bushinsky
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Glenn M Chertow
- Department of Medicine, Stanford University, Palo Alto, CA, USA
| | - Markus Ketteler
- Department of General Internal Medicine and Nephrology, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Mariano Rodriguez
- Nephrology Unit, Hospital Universitario Reina Sofia, IMIBIC, REDinREN, Córdoba, Spain
| | - Smeeta Sinha
- Department of Renal Medicine, Salford Royal NHS Foundation Trust, Salford, UK
| | | | - Rekha Garg
- Research and Development, Sanifit Therapeutics, San Diego, CA, USA
| | - Alex Gold
- Research and Development, Sanifit Therapeutics, San Diego, CA, USA
| | - Joan Perelló
- Research and Development, Sanifit Therapeutics, Palma, Spain
| |
Collapse
|
16
|
Perez MM, Ferrer MD, Lazo-Rodriguez M, Canals AZ, Banon-Maneus E, Campistol JM, Miller S, Garg R, Gold A, Salcedo C, Perelló J. A novel assay to measure calcification propensity: from laboratory to humans. Sci Rep 2020; 10:17578. [PMID: 33067536 PMCID: PMC7568551 DOI: 10.1038/s41598-020-74592-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 08/10/2020] [Indexed: 01/25/2023] Open
Abstract
Cardiovascular calcification (CVC) contributes to morbidity and mortality in patients undergoing dialysis. We examined the pharmacodynamic effects of SNF472, a calcification inhibitor, on plasma calcium phosphate crystallization using spectrometric measurements, and its correlations with effects on CVC in rats or humans. Rats (N = 38) injected with vitamin D (days 1–3) to induce CVC were infused with saline or SNF472 (days 1–12). Inhibition of CVC was 50–65% with SNF472 3 mg/kg and ~ 80% with SNF472 10 or 30 mg/kg. SNF472 dose-dependently inhibited calcium phosphate crystallization, which correlated with inhibition of CVC (r = 0.628, P = 0.005). In patients with calciphylaxis (N = 14), infusion of SNF472 (~ 7 mg/kg) during hemodialysis for 12 weeks inhibited calcium phosphate crystallization by nearly 70%. In patients with CVC (N = 274), infusion of SNF472 during hemodialysis for 52 weeks inhibited calcium phosphate crystallization (placebo: 15%; 300 mg: 61%; 600 mg: 75%), which correlated with inhibition of CVC (r = 0.401, P = 0.003). These findings show a direct correlation between inhibition of calcium phosphate crystallization in plasma and inhibition of CVC both in a rat model and in humans, supporting the use of the pharmacodynamic assay in clinical trials as a potentially predictive tool to evaluate the activity of calcification inhibitors.
Collapse
Affiliation(s)
- M Mar Perez
- Sanifit Therapeutics, Parc Bit - Europa Building, 2nd Floor, 07121, Palma de Mallorca, Spain
| | - Miguel D Ferrer
- Sanifit Therapeutics, Parc Bit - Europa Building, 2nd Floor, 07121, Palma de Mallorca, Spain.,Department of Fundamental Biology and Health Sciences, University of the Balearic Islands, Palma, Spain
| | - Marta Lazo-Rodriguez
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Fundació Clínic per a la Recerca Biomèdica, Barcelona, Spain.,Spanish Kidney Research Network, ISCIII-RETIC REDinREN RD016/0 009, Madrid, Spain
| | - Ana Zeralda Canals
- Sanifit Therapeutics, Parc Bit - Europa Building, 2nd Floor, 07121, Palma de Mallorca, Spain
| | - Elisenda Banon-Maneus
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Fundació Clínic per a la Recerca Biomèdica, Barcelona, Spain.,Spanish Kidney Research Network, ISCIII-RETIC REDinREN RD016/0 009, Madrid, Spain
| | - Josep M Campistol
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Fundació Clínic per a la Recerca Biomèdica, Barcelona, Spain.,Spanish Kidney Research Network, ISCIII-RETIC REDinREN RD016/0 009, Madrid, Spain
| | | | - Rekha Garg
- Sanifit Therapeutics, San Diego, CA, USA.,PharmaDRS Consulting, LLC, San Diego, USA
| | - Alex Gold
- Sanifit Therapeutics, San Diego, CA, USA.,Department of Medicine, Stanford University, Palo Alto, CA, USA
| | - Carolina Salcedo
- Sanifit Therapeutics, Parc Bit - Europa Building, 2nd Floor, 07121, Palma de Mallorca, Spain.
| | - Joan Perelló
- Sanifit Therapeutics, Parc Bit - Europa Building, 2nd Floor, 07121, Palma de Mallorca, Spain.,Laboratory of Renal Lithiasis Research, University of the Balearic Islands, Palma, Spain
| |
Collapse
|
17
|
Djuric P, Dimkovic N, Schlieper G, Djuric Z, Pantelic M, Mitrovic M, Jankovic A, Milanov M, Kuzmanovic Pficer J, Floege J. Sodium thiosulphate and progression of vascular calcification in end-stage renal disease patients: a double-blind, randomized, placebo-controlled study. Nephrol Dial Transplant 2020; 35:162-169. [PMID: 31764989 DOI: 10.1093/ndt/gfz204] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 07/18/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Sodium thiosulphate (NaTS) is mostly used in haemodialysis (HD) patients with calcific uraemic arteriolopathy. This double-blind, randomized, placebo-controlled study assessed the effect of NaTS on progression of cardiovascular calcifications in HD patients. METHODS From 65 screened patients, we recruited 60 patients with an abdominal aorta Agatston calcification score ≥100. Thirty patients were randomized to receive NaTS 25 g/1.73 m2 and 30 patients to receive 100 mL of 0.9% sodium chloride intravenously during the last 15 min of HD over a period of 6 months. The primary endpoint was the absolute change of the abdominal aortic calcification score. RESULTS The abdominal aortic calcification score and calcification volume of the abdominal aorta increased similarly in both treatment groups during the trial. As compared with the saline group, patients receiving NaTS exhibited a reduction of their iliac artery calcification score (-137 ± 641 versus 245 ± 755; P = 0.049), reduced pulse wave velocity (9.6 ± 2.7 versus 11.4 ± 3.6; P = 0.000) and a lower carotid intima-media thickness (0.77 ± 0.1 versus 0.83 ± 00.17; P = 0.033) and had better preservation of echocardiographic parameters of left ventricular hypertrophy. No patient of the NaTS group developed new cardiac valve calcifications during the trial as compared with 8 of 29 patients in the saline group. By univariate analysis, NaTS therapy was the only predictor of not developing new valvular calcifications. No adverse events possibly related to NaTS infusion were noted. CONCLUSIONS While NaTS failed to retard abdominal aortic calcification progress, it positively affected calcification progress in iliac arteries and heart valves as well as several other cardiovascular functional parameters.
Collapse
Affiliation(s)
- Petar Djuric
- Clinical Department for Nephrology, Zvezdara University Medical Center, Belgrade, Serbia
| | - Nada Dimkovic
- Clinical Department for Nephrology, Zvezdara University Medical Center, Belgrade, Serbia.,Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Georg Schlieper
- Division of Nephrology and Immunology, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany.,Center for Nephrology, Hypertension, and Metabolic Diseases, Hannover, Germany
| | - Zivka Djuric
- Clinical Department for Nephrology, Zvezdara University Medical Center, Belgrade, Serbia
| | - Milan Pantelic
- Center for Radiology, Zvezdara University Medical Center, Belgrade, Serbia
| | - Milica Mitrovic
- Center for Radiology and Magnetic Resonance Imaging, Clinical Center of Serbia, Beograd, Serbia
| | - Aleksandar Jankovic
- Clinical Department for Nephrology, Zvezdara University Medical Center, Belgrade, Serbia
| | - Marko Milanov
- Clinical Department for Cardiology, Zvezdara University Medical Center, Belgrade, Serbia
| | - Jovana Kuzmanovic Pficer
- Department for Medical Statistics and Informatics, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - Jürgen Floege
- Division of Nephrology and Immunology, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| |
Collapse
|
18
|
Thrivikraman G, Johnson SL, Syedain ZH, Hill RC, Hansen KC, Lee HS, Tranquillo RT. Biologically-engineered mechanical model of a calcified artery. Acta Biomater 2020; 110:164-174. [PMID: 32305446 DOI: 10.1016/j.actbio.2020.04.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/27/2020] [Accepted: 04/08/2020] [Indexed: 12/22/2022]
Abstract
Vascular calcification is a commonly occurring pathological process and is recognized as an independent prognostic marker for cardiovascular morbidity and mortality. Recent progress in developing novel therapies to modify vascular calcification is critically hampered due to the lack of reliable in vitro experimental models that recapitulate the structural and mechanical attributes of calcified arteries. In this study, we show the ability to model the behavior of diffuse vascular calcification in vitro using biologically-engineered grafts approximating the composition, structure, and mechanical properties of arteries. Transmural calcification was achieved by exposing the acellular grafts of collagenous ECM to complete medium containing elevated Calcium (Ca) and Phosphate (P) concentrations. It was found that increasing the serum concentration from 2% to 10% increased the extent and degree of calcification based on histochemical, ultrastructural, chemical and thermal analyses. The presence of variably-sized spherical calcific deposits within the matrix further confirmed its morphological similarity to pathologic calcification. Mechanical testing demonstrated up to a 16-fold decrease in compliance due to the calcification, consistent with prior reports for calcified arteries. The model developed thus has potential to improve the design and development of interventional devices and therapies for the diagnosis and treatment of arterial calcification. STATEMENT OF SIGNIFICANCE: The presence of extensive vascular calcification makes angiographic/interventional procedures difficult due to reduced arterial compliance. Current attempts to develop safe and effective non-surgical adjunctive techniques to treat calcified arteries are largely limited by the lack of a physiologically relevant testing platform that mimics the structural and mechanical features of vascular calcification. Herein, we developed an off-the-shelf calcified artery model, with the goal to accelerate the pre-clinical development of novel therapies for the management of arterial calcification. To the extent of our knowledge, this is the first report of an in vitro tissue-engineered model of diffuse arterial calcification.
Collapse
|
19
|
Raggi P, Bellasi A, Bushinsky D, Bover J, Rodriguez M, Ketteler M, Sinha S, Salcedo C, Gillotti K, Padgett C, Garg R, Gold A, Perelló J, Chertow GM. Slowing Progression of Cardiovascular Calcification With SNF472 in Patients on Hemodialysis. Circulation 2020; 141:728-739. [DOI: 10.1161/circulationaha.119.044195] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background:
The high cardiovascular morbidity and mortality in patients with end-stage kidney disease could be partially caused by extensive cardiovascular calcification. SNF472, intravenous myo-inositol hexaphosphate, selectively inhibits the formation and growth of hydroxyapatite.
Methods:
This double-blind, placebo-controlled phase 2b trial compared progression of coronary artery calcium volume score and other measurements of cardiovascular calcification by computed tomography scan during 52 weeks of treatment with SNF472 or placebo, in addition to standard therapy, in adult patients with end-stage kidney disease receiving hemodialysis. Patients were randomized 1:1:1 to SNF472 300 mg (n=92), SNF472 600 mg (n=91), or placebo (n=91) by infusion in the hemodialysis lines thrice weekly during hemodialysis sessions. The primary end point was change in log coronary artery calcium volume score from baseline to week 52. The primary efficacy analysis combined the SNF472 treatment groups and included all patients who received at least 1 dose of SNF472 or placebo and had an evaluable computed tomography scan after randomization.
Results:
The mean change in coronary artery calcium volume score was 11% (95% CI, 7–15) for the combined SNF472 dose group and 20% (95% CI, 14–26) for the placebo group (
P
=0.016). SNF472 compared with placebo attenuated progression of calcium volume score in the aortic valve (14% [95% CI, 5–24] versus 98% [95% CI, 77–123];
P
<0.001) but not in the thoracic aorta (23% [95% CI, 16–30] versus 28% [95% CI, 19–38];
P
=0.40). Death occurred in 7 patients (4%) who received SNF472 and 5 patients (6%) who received placebo. At least 1 treatment-emergent adverse event occurred in 86%, 92%, and 87% of patients treated with SNF472 300 mg, SNF472 600 mg, and placebo, respectively. Most adverse events were mild. Adverse events resulted in discontinuation of SNF472 300 mg, SNF472 600 mg, and placebo for 14%, 29%, and 20% of patients, respectively.
Conclusions:
Compared with placebo, SNF472 significantly attenuated the progression of coronary artery calcium and aortic valve calcification in patients with end-stage kidney disease receiving hemodialysis in addition to standard care. Future studies are needed to determine the effects of SNF472 on cardiovascular events.
Registration:
URL:
https://www.clinicaltrials.gov
; Unique identifier: NCT02966028.
Collapse
Affiliation(s)
- Paolo Raggi
- Department of Medicine, Mazankowski Alberta Heart Institute and University of Alberta, Edmonton, Canada (P.R.)
| | - Antonio Bellasi
- Research, Innovation and Brand Reputation Unit, ASST Papa Giovanni XXIII, Bergamo, Italy (A.B.)
| | - David Bushinsky
- Department of Medicine, University of Rochester Medical Center, NY (D.B.)
| | - Jordi Bover
- Department of Nephrology, Fundació Puigvert and Universitat Autònoma, IIB Sant Pau, REDinREN, Barcelona, Spain (J.B.)
| | - Mariano Rodriguez
- Nephrology Unit, Hospital Universitario Reina Sofia, IMIBIC, REDinREN, Córdoba, Spain (M.R.)
| | - Markus Ketteler
- Department of General Internal Medicine and Nephrology, Robert-Bosch-Krankenhaus, Stuttgart, Germany (M.K.)
| | - Smeeta Sinha
- Department of Renal Medicine, Salford Royal NHS Foundation Trust, UK (S.S.)
| | - Carolina Salcedo
- Research and Development, Sanifit Therapeutics, Palma, Spain (C.S., J.P.)
| | - Kristen Gillotti
- Research and Development, Sanifit Therapeutics, San Diego, CA (K.G., C.P. R.G., A.G.)
| | - Claire Padgett
- Research and Development, Sanifit Therapeutics, San Diego, CA (K.G., C.P. R.G., A.G.)
| | - Rekha Garg
- Research and Development, Sanifit Therapeutics, San Diego, CA (K.G., C.P. R.G., A.G.)
| | - Alex Gold
- Research and Development, Sanifit Therapeutics, San Diego, CA (K.G., C.P. R.G., A.G.)
- Department of Medicine, Stanford University, Palo Alto, CA (A.G., G.M.C.)
| | - Joan Perelló
- Research and Development, Sanifit Therapeutics, Palma, Spain (C.S., J.P.)
- University of the Balearic Islands, Palma, Spain (J.P.)
| | - Glenn M. Chertow
- Department of Medicine, Stanford University, Palo Alto, CA (A.G., G.M.C.)
| |
Collapse
|
20
|
Schantl AE, Verhulst A, Neven E, Behets GJ, D'Haese PC, Maillard M, Mordasini D, Phan O, Burnier M, Spaggiari D, Decosterd LA, MacAskill MG, Alcaide-Corral CJ, Tavares AAS, Newby DE, Beindl VC, Maj R, Labarre A, Hegde C, Castagner B, Ivarsson ME, Leroux JC. Inhibition of vascular calcification by inositol phosphates derivatized with ethylene glycol oligomers. Nat Commun 2020; 11:721. [PMID: 32024848 PMCID: PMC7002685 DOI: 10.1038/s41467-019-14091-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 12/18/2019] [Indexed: 12/14/2022] Open
Abstract
Myo-inositol hexakisphosphate (IP6) is a natural product known to inhibit vascular calcification (VC), but with limited potency and low plasma exposure following bolus administration. Here we report the design of a series of inositol phosphate analogs as crystallization inhibitors, among which 4,6-di-O-(methoxy-diethyleneglycol)-myo-inositol-1,2,3,5-tetrakis(phosphate), (OEG2)2-IP4, displays increased in vitro activity, as well as more favorable pharmacokinetic and safety profiles than IP6 after subcutaneous injection. (OEG2)2-IP4 potently stabilizes calciprotein particle (CPP) growth, consistently demonstrates low micromolar activity in different in vitro models of VC (i.e., human serum, primary cell cultures, and tissue explants), and largely abolishes the development of VC in rodent models, while not causing toxicity related to serum calcium chelation. The data suggest a mechanism of action independent of the etiology of VC, whereby (OEG2)2-IP4 disrupts the nucleation and growth of pathological calcification.
Collapse
Affiliation(s)
- Antonia E Schantl
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Anja Verhulst
- Laboratory of Pathophysiology, University of Antwerp, Antwerp, Belgium
| | - Ellen Neven
- Laboratory of Pathophysiology, University of Antwerp, Antwerp, Belgium
| | - Geert J Behets
- Laboratory of Pathophysiology, University of Antwerp, Antwerp, Belgium
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, University of Antwerp, Antwerp, Belgium
| | - Marc Maillard
- Service of Nephrology and Hypertension, Lausanne University Hospital, Lausanne, Switzerland
| | - David Mordasini
- Service of Nephrology and Hypertension, Lausanne University Hospital, Lausanne, Switzerland
| | - Olivier Phan
- Service of Nephrology and Hypertension, Lausanne University Hospital, Lausanne, Switzerland
| | - Michel Burnier
- Service of Nephrology and Hypertension, Lausanne University Hospital, Lausanne, Switzerland
| | - Dany Spaggiari
- Division of Clinical Pharmacology, Lausanne University Hospital, Lausanne, Switzerland
| | - Laurent A Decosterd
- Division of Clinical Pharmacology, Lausanne University Hospital, Lausanne, Switzerland
| | - Mark G MacAskill
- University-BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Carlos J Alcaide-Corral
- University-BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Adriana A S Tavares
- University-BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - David E Newby
- University-BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Victoria C Beindl
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | | | - Anne Labarre
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Canada
| | - Chrismita Hegde
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Canada
| | - Bastien Castagner
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Canada
| | | | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
21
|
Pasquali M, De Martini N, Mazzaferro S. Calciphylaxis: a conundrum for patients and nephrologists? J Nephrol 2019; 32:677-680. [DOI: 10.1007/s40620-019-00639-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 08/10/2019] [Indexed: 01/15/2023]
|
22
|
Zabirnyk A, Ferrer MD, Bogdanova M, Pérez MM, Salcedo C, Kaljusto ML, Kvitting JPE, Stensløkken KO, Perelló J, Vaage J. SNF472, a novel anti-crystallization agent, inhibits induced calcification in an in vitro model of human aortic valve calcification. Vascul Pharmacol 2019; 122-123:106583. [PMID: 31437530 DOI: 10.1016/j.vph.2019.106583] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 08/03/2019] [Accepted: 08/18/2019] [Indexed: 12/27/2022]
Abstract
The purpose of the present study was to investigate whether SNF472, the hexasodium salt of myo-inositol hexaphosphate (IP6 or phytate): 1. Inhibits induced calcification in cultured aortic valve interstitial cells (VIC) as an in vitro model of aortic valve stenosis and 2. Whether inhibition is different in VIC obtained from healthy and calcified aortic valves. VIC from healthy (n = 5) and calcified (n = 7) human aortic valves were seeded in basic growth medium, osteogenic differentiation medium alone, or in osteogenic medium with SNF472 (3, 10, and 30 μM) and cultivated for 3 weeks. Calcification was quantified spectrophotometrically after Alizarin Red staining. In VIC from calcified valves, a complete inhibition of calcification was observed with SNF472 concentrations of 10 and 30 μM (p < .01), significantly stronger than in VIC from healthy valves. When SNF472 was added to VIC after 1 week in osteogenic medium, 30 and 100 μM SNF472 inhibited the progression of ongoing calcification by 81 and 100% (p < .01), respectively. The same concentrations of SNF472 given after 2 weeks reduced calcification by 35 and 40% respectively (not significant). SNF472 inhibited both the formation and the progression of calcification with the strongest effect in VIC from calcified valves.
Collapse
Affiliation(s)
- A Zabirnyk
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| | - M D Ferrer
- Sanifit Therapeutics, Palma, Spain; Department of Fundamental Biology and Health Sciences, University of the Balearic Islands, Palma, Spain
| | - M Bogdanova
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | | | - M-L Kaljusto
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - J-P E Kvitting
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - K-O Stensløkken
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - J Perelló
- Sanifit Therapeutics, Palma, Spain; Department of Chemistry, University of the Balearic Islands, Palma, Spain
| | - J Vaage
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
23
|
Improvement in wound healing, pain, and quality of life after 12 weeks of SNF472 treatment: a phase 2 open-label study of patients with calciphylaxis. J Nephrol 2019; 32:811-821. [PMID: 31401795 DOI: 10.1007/s40620-019-00631-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 07/13/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Calciphylaxis in end-stage renal disease is characterized by painful necrotic skin ulcers and high mortality. There are no approved therapies. SNF472, an intravenous formulation of myo-inositol hexaphosphate, inhibits the formation and growth of hydroxyapatite crystals, the final common pathway in the pathogenesis of vascular calcification. METHODS In this open-label, single-arm study, calciphylaxis patients on thrice-weekly hemodialysis and standard care, received intravenous SNF472 3 times per week for 12 weeks. The primary endpoint was wound healing assessed using the quantitative Bates-Jensen Wound Assessment Tool (BWAT). Pain visual analog scale (VAS), quality of life (wound-QoL), and qualitative wound image review were secondary endpoints. Quantitative changes from baseline were analyzed by paired t-tests using multiple imputation to account for missing observations. RESULTS Fourteen patients received SNF472. Improvements from baseline to week 12 were observed for mean BWAT score (- 8.1; P < 0.001), pain VAS (- 23.6 mm; P = 0.015) and wound-QoL global score (- 0.90; P = 0.003). Of the 9 patients with ulcerated lesions at baseline who completed treatment, wound image review showed improvement for 7. SNF472 was well tolerated with no serious treatment-related adverse events. The most common adverse events were infections which occur frequently in patients on hemodialysis. None of these were considered as treatment-related. CONCLUSIONS SNF472 was well-tolerated and improvements from baseline to week 12 in wound healing, pain, and quality of life were observed. A randomized, double-blind, placebo-controlled trial is planned to evaluate SNF472 in patients with calciphylaxis.
Collapse
|
24
|
Salcedo C, Joubert PH, Ferrer MD, Canals AZ, Maduell F, Torregrosa V, Campistol JM, Ojeda R, Perelló J. A phase 1b randomized, placebo-controlled clinical trial with SNF472 in haemodialysis patients. Br J Clin Pharmacol 2019; 85:796-806. [PMID: 30632182 DOI: 10.1111/bcp.13863] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/20/2018] [Accepted: 12/23/2018] [Indexed: 12/19/2022] Open
Affiliation(s)
| | - Pieter H Joubert
- Laboratoris Sanifit, Palma, Spain.,Institute of Pharmaceutical Science, King's College, London, UK
| | - Miguel D Ferrer
- Laboratoris Sanifit, Palma, Spain.,Departament de Biologia Fonamental i Ciències de la Salut, Universitat de les Illes Balears, Palma, Spain
| | | | | | | | | | - Raquel Ojeda
- Nephrology Department, Hospital Clinic, Barcelona, Spain
| | - Joan Perelló
- Laboratoris Sanifit, Palma, Spain.,Laboratori d'Investigació en Litiasi Renal, Universitat de les Illes Balears, Palma, Spain
| |
Collapse
|