1
|
Liu J, Chen K, Tang M, Mu Q, Zhang S, Li J, Liao J, Jiang X, Wang C. Oxidative stress and inflammation mediate the adverse effects of cadmium exposure on all-cause and cause-specific mortality in patients with diabetes and prediabetes. Cardiovasc Diabetol 2025; 24:145. [PMID: 40158078 PMCID: PMC11954339 DOI: 10.1186/s12933-025-02698-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND The effect of cadmium exposure on mortality risk among individuals with diabetes and prediabetes remains unclear, particularly regarding potential mediation by oxidative stress and inflammation. This study aimed to investigate the associations of blood cadmium levels with all-cause, cardiovascular disease (CVD), and cancer mortality and the mediating effects of oxidative stress and inflammation biomarkers in patients with diabetes and prediabetes. METHODS In this prospective cohort study, we analyzed 17,687 adults with diabetes and prediabetes from the National Health and Nutrition Examination Survey (NHANES, 1999-2018). Nine biomarkers related to oxidative stress (gamma-glutamyl transferase [GGT], uric acid [UA], high-density lipoprotein [HDL], UA to HDL ratio [UHR]) and inflammation (neutrophil-lymphocyte ratio [NLR], monocyte-lymphocyte ratio [MLR], neutrophil-monocyte-lymphocyte ratio [NMLR], systemic inflammation response index [SIRI], systemic immune-inflammation index [SII]) were systematically assessed. Kaplan-Meier survival analysis, Cox proportional hazards models, and restricted cubic splines (RCS) were applied to evaluate the association of cadmium with mortality risk. Generalized linear models were used to assess the association of cadmium with oxidative stress and inflammation biomarkers, while Cox regression and RCS evaluated their effects on mortality. Causal mediation analysis identified biological pathways mediated by oxidative stress and inflammation. Stratified and sensitivity analyses were further employed to confirm the robustness of the results. RESULTS During 161,047.75 person-years of follow-up, 3562 deaths occurred, including 1214 from CVD and 680 from cancer. Higher blood cadmium levels were associated with increased risks of all-cause mortality (fully adjusted hazard ratio [HR]: 2.17; 95% confidence interval [CI] 1.69-2.79, comparing highest vs. lowest quartile), CVD mortality (HR 2.06; 95% CI 1.41-3.02), and cancer mortality (HR 2.38; 95% CI 1.47-3.85), without evidence of nonlinear relationship. Mediation analyses indicated that UA, NLR, MLR, NMLR, and SIRI partially mediated the associations of cadmium with all-cause and CVD mortality, although the mediated proportions were relatively modest (ranging from 1.4 to 4.8%). Additionally, GGT mediated a small fraction of the associations with all-cause and cancer mortality. CONCLUSION Cadmium exposure increases the risk of all-cause, CVD, and cancer mortality in patients with diabetes and prediabetes. Oxidative stress and inflammation appear to partially mediate this adverse effect. These findings emphasize the urgent need for targeted interventions to reduce cadmium-related mortality risks. RESEARCH INSIGHTS What is currently known about this topic? Cadmium exposure is linked to increased mortality. Oxidative stress and inflammation are critical in diabetes development and complications. What is the key research question? Does cadmium exposure increase mortality risk in patients with diabetes and prediabetes? Are oxidative stress and inflammation involved in mediating these effects? What is new? Cadmium exposure increases all-cause and cause-specific mortality in diabetes and prediabetes. Oxidative stress and inflammation mediate these associations. How might this study influence clinical practice? Monitor cadmium, oxidative stress, and inflammation to reduce mortality in diabetes and prediabetes.
Collapse
Affiliation(s)
- Jingqi Liu
- Department of Public Health Laboratory Sciences, West China School of Public Health, West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin South Road, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Kehan Chen
- Department of Public Health Laboratory Sciences, West China School of Public Health, West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin South Road, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Mingyuan Tang
- Department of Public Health Laboratory Sciences, West China School of Public Health, West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin South Road, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Qunzheng Mu
- Department of Public Health Laboratory Sciences, West China School of Public Health, West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin South Road, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Shirong Zhang
- Department of Public Health Laboratory Sciences, West China School of Public Health, West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin South Road, Wuhou District, Chengdu, 610041, Sichuan, China
| | - Jiayuan Li
- Department of Epidemiology and Health Statistics, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiaqiang Liao
- Department of Epidemiology and Health Statistics, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xia Jiang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chuan Wang
- Department of Public Health Laboratory Sciences, West China School of Public Health, West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin South Road, Wuhou District, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
2
|
Ujueta F, Lamas GA, Anstrom KJ, Navas-Acien A, Boineau R, Rosenberg Y, Stylianou M, Jones TLZ, Joubert BR, Yu Q, Wen J, Nemeth H, Huang Z, Fonseca V, Nathan DM, Uwaifo G, Arenas IA, Luo L, Baker J, Visentin D, Paixao A, Schmedtje JF, Mark DB. Multivitamins After Myocardial Infarction in Patients With Diabetes: A Randomized Clinical Trial. JAMA Intern Med 2025:2830883. [PMID: 40029647 DOI: 10.1001/jamainternmed.2024.8408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Importance In 2013, the Trial to Assess Chelation Therapy (TACT) reported that in 1708 patients with stable coronary disease and prior myocardial infarction (MI), oral multivitamins and multiminerals (OMVMs), in a factorial design with edetate disodium (EDTA) chelation therapy, did not reduce cardiovascular events relative to placebo OMVMs, but active EDTA combined with active OMVMs was superior to placebo OMVM/placebo EDTA. Objective To compare OMVM vs placebo in terms of efficacy for reducing major adverse cardiovascular events in patients with diabetes and prior MI. Design, Setting, and Participants The TACT2 randomized, multicenter double-masked 2 × 2 factorial clinical trial took place across 88 sites in the US and Canada. Participants were 50 years or older, had diabetes, and had an MI 6 weeks ago or more. TACT2 participants were enrolled between September 2016 and December 2020. Data were collected between October 2016 and June 2023. Interventions Six caplets daily of a 28 component OMVM or matching OMVM placebo, and 40 weekly infusions of an EDTA-based chelation solution or matching placebo, in a 1:1:1:1 allocation ratio. Main Outcomes and Measures The primary end point was the composite of all-cause mortality, MI, stroke, coronary revascularization, or hospitalization for unstable angina. Results A total of 1000 participants were randomized (500 in the active OMVM group and 500 in the placebo group). The median (IQR) age was 67 (60-72) years, and 730 (73%) were male. Median (IQR) follow-up was 48 (34-58) months. The primary end point occurred in 175 participants (35%) in the active OMVM group and 175 (35%) in the placebo group (hazard ratio [HR], 0.99 [95% CI, 0.80-1.22]; P = .92). The 5-year event rate for the primary end point in the EDTA chelation + active OMVM group was 34.0%; in the EDTA chelation + placebo OMVM group, 35.7%; in the placebo infusion + active OMVM group, 36.0%; and in the placebo infusion + placebo OMVM group, 34.3%. The comparison of the active infusion + active OMVM with the placebo infusion + placebo OMVM was not significant (HR, 0.91 [95% CI, 0.67-1.23]; P = .54). Although nonsignificant, there was a numerically higher event rate of MI, stroke, mortality from cardiovascular causes in the active OMVM compared to placebo OMVM group. Conclusions and Relevance The results of this randomized clinical trial demonstrated that, for participants with chronic coronary disease, diabetes, and a previous MI, high-dose OMVM alone or in conjunction with EDTA-based chelation did not reduce cardiovascular events. Trial Registration ClinicalTrials.gov Identifier: NCT02733185.
Collapse
Affiliation(s)
- Francisco Ujueta
- Brigham and Women's Hospital Division of Cardiovascular Medicine, Boston, Massachusetts
| | - Gervasio A Lamas
- Columbia University Division of Cardiology, Mount Sinai Medical Center, Miami Beach, Florida
| | - Kevin J Anstrom
- Gillings School of Global Public Health, University of North Carolina, Chapel Hill
| | - Ana Navas-Acien
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, New York
| | - Robin Boineau
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland
| | - Yves Rosenberg
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Mario Stylianou
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Teresa L Z Jones
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Bonnie R Joubert
- National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina
| | - Qilu Yu
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland
| | - Jun Wen
- Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | - Hayley Nemeth
- Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | - Zhen Huang
- Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | - Vivian Fonseca
- Tulane University School of Medicine, New Orleans, Louisiana
| | - David M Nathan
- Massachusetts General Hospital Diabetes Research Center, Harvard Medical School, Boston
| | - Gabriel Uwaifo
- Southern Illinois University School of Medicine, Springfield
| | | | - Lan Luo
- Central Florida Heart Center, Ocala
| | | | | | | | | | - Daniel B Mark
- Duke Clinical Research Institute, Duke University, Durham, North Carolina
| |
Collapse
|
3
|
Hasan S, Parikh MA, Trivedi DB, Frishman WH, Peterson SJ. Chelation Therapy in Coronary Artery Disease: Fact or Fiction? Cardiol Rev 2025:00045415-990000000-00418. [PMID: 39936901 DOI: 10.1097/crd.0000000000000871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Coronary artery disease (CAD) is responsible for 690,000 deaths a year, a leading cause of mortality worldwide. CAD results from cholesterol plaque buildup in arteries. Chelation therapy, which uses ethylenediaminetetraacetic acid to remove toxic metals from the bloodstream, has been explored as an alternative treatment for atherosclerotic CAD. While the 2013 Trial to Assess Chelation Therapy (TACT) trial showed modest cardiovascular benefits, particularly in patients with diabetes, subsequent studies such as TACT2 did not confirm its efficacy in reducing cardiovascular events in patients with diabetes. Adverse effects of chelation therapy could include renal dysfunction, electrolyte imbalances, and potential complications from heavy metal mobilization that could be fatal. Still, none of these were seen in TACT or TACT2.
Collapse
Affiliation(s)
- Sara Hasan
- From the Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
| | - Manish A Parikh
- From the Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
- Weill Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Dhaval B Trivedi
- From the Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
| | | | - Stephen J Peterson
- From the Department of Medicine, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY
- Weill Department of Medicine, Weill Cornell Medicine, New York, NY
| |
Collapse
|
4
|
Lamas GA, Anstrom KJ, Navas-Acien A, Boineau R, Nemeth H, Huang Z, Wen J, Rosenberg Y, Stylianou M, Jones TLZ, Joubert BR, Yu Q, Santella RM, Mon AC, Ujueta F, Escolar E, Nathan DM, Fonseca VA, Aude YW, Ehrman JK, Elliott T, Prashad R, Lewis EF, Lopes RD, Farkouh ME, Elliott AM, Newman JD, Mark DB. Edetate Disodium-Based Chelation for Patients With a Previous Myocardial Infarction and Diabetes: TACT2 Randomized Clinical Trial. JAMA 2024; 332:794-803. [PMID: 39141382 PMCID: PMC11325247 DOI: 10.1001/jama.2024.11463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/28/2024] [Indexed: 08/15/2024]
Abstract
Importance In 2013, the Trial to Assess Chelation Therapy (TACT) reported that edetate disodium (EDTA)-based chelation significantly reduced cardiovascular disease (CVD) events by 18% in 1708 patients with a prior myocardial infarction (MI). Objective To replicate the finding of TACT in individuals with diabetes and previous MI. Design, Setting, and Participants A 2 × 2 factorial, double-masked, placebo-controlled, multicenter trial at 88 sites in the US and Canada, involving participants who were 50 years or older, had diabetes, and had experienced an MI at least 6 weeks before recruitment compared the effect of EDTA-based chelation vs placebo infusions on CVD events and compared the effect of high doses of oral multivitamins and minerals with oral placebo. This article reports on the chelation vs placebo infusion comparisons. Interventions Eligible participants were randomly assigned to 40 weekly infusions of an EDTA-based chelation solution or matching placebo and to twice daily oral, high-dose multivitamin and mineral supplements or matching placebo for 60 months. This article addresses the chelation study. Main Outcomes and Measures The primary end point was the composite of all-cause mortality, MI, stroke, coronary revascularization, or hospitalization for unstable angina. Median follow-up was 48 months. Primary comparisons were made from patients who received at least 1 assigned infusion. Results Of the 959 participants (median age, 67 years [IQR, 60-72 years]; 27% females; 78% White, 10% Black, and 20% Hispanic), 483 received at least 1 chelation infusion and 476 at least 1 placebo infusion. A primary end point event occurred in 172 participants (35.6%) in the chelation group and in 170 (35.7%) in the placebo group (adjusted hazard ratio [HR], 0.93; 95% CI, 0.76-1.16; P = .53). The 5-year primary event cumulative incidence rates were 45.8% for the chelation group and 46.5% for the placebo group. CV death, MI, or stroke events occurred in 89 participants (18.4%) in the chelation group and in 94 (19.7%) in the placebo group (adjusted HR, 0.89; 95% CI, 0.66-1.19). Death from any cause occurred in 84 participants (17.4%) in the chelation group and in 84 (17.6%) in the placebo group (adjusted HR, 0.96; 95% CI, 0.71-1.30). Chelation reduced median blood lead levels from 9.03 μg/L at baseline to 3.46 μg/L at infusion 40 (P < .001). Corresponding levels in the placebo group were 9.3 μg/L and 8.7 μg/L, respectively. Conclusions and Relevance Despite effectively reducing blood lead levels, EDTA chelation was not effective in reducing cardiovascular events in stable patients with coronary artery disease who have diabetes and a history of MI. Trial Registration ClinicalTrials.gov Identifier: NCT02733185.
Collapse
Affiliation(s)
- Gervasio A. Lamas
- Columbia University Division of Cardiology, Mount Sinai Medical Center, Miami Beach, Florida
| | - Kevin J. Anstrom
- Gillings School of Global Public Health, University of North Carolina, Chapel Hill
| | - Ana Navas-Acien
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, New York
| | - Robin Boineau
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland
| | - Hayley Nemeth
- Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | - Zhen Huang
- Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | - Jun Wen
- Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | - Yves Rosenberg
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Mario Stylianou
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Teresa L. Z. Jones
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Bonnie R. Joubert
- National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina
| | - Qilu Yu
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland
| | - Regina M. Santella
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, New York
| | - Ana C. Mon
- Columbia University Division of Cardiology, Mount Sinai Medical Center, Miami Beach, Florida
| | - Francisco Ujueta
- Columbia University Division of Cardiology, Mount Sinai Medical Center, Miami Beach, Florida
| | - Esteban Escolar
- Columbia University Division of Cardiology, Mount Sinai Medical Center, Miami Beach, Florida
| | - David M. Nathan
- Massachusetts General Hospital Diabetes Research Center, Harvard Medical School, Boston
| | | | | | - Jonathan K. Ehrman
- Division of Cardiovascular Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Thomas Elliott
- BC Diabetes Research Institute, Vancouver, British Columbia, Canada
| | | | - Eldrin F. Lewis
- Stanford University School of Medicine, Palo Alto, California
| | - Renato D. Lopes
- Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | | | - Anne-Marie Elliott
- Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | | | - Daniel B. Mark
- Duke Clinical Research Institute, Duke University, Durham, North Carolina
| |
Collapse
|
5
|
Ordoñez-Sierra R, Domínguez-Cortinas G, Hernández-Paniagua IY, Expósito-Castillo JL, Gómez-Albores MA, Rodríguez-Reyes MG, Carrasco-Gallegos BV, Manzano-Solís LR. Regionalization of the Mortality Risk from Cardiomyopathy and Respiratory Diseases Based on the Maximum Entropy Model. Glob Health Epidemiol Genom 2024; 2024:6103589. [PMID: 39502913 PMCID: PMC11535265 DOI: 10.1155/2024/6103589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/07/2024] [Accepted: 07/31/2024] [Indexed: 11/08/2024] Open
Abstract
This research presents a time-series study in one of the most polluted regions in Mexico, the southern part of the Mezquital Valley. Three mortality causes related to areas highly contaminated by industrial activities were considered to carry out this model, namely, ischemic cardiomyopathy, mesothelioma, and pneumoconiosis. The pollutant exposure factors used in the maximum entropy modeling were distance to rivers, distance to industries, particulate matter less than 2.5 microns (PM < 2.5 µm), and the digital elevation model (DEM). A model that expresses the presence of the disease by areas of exposure to pollutants was also obtained. In addition, the odds ratio was calculated to evaluate the level of association of ischemic cardiomyopathy (OR = 3.37 and 95% CI: 3.05-3.6) and mesothelioma (OR = 4.79 and 95% CI: 3.5-6.08) by areas of exposure. In the case of pneumoconiosis, only cases in the very high exposure category were recorded, so it was not comparable with the remaining areas. It is important to mention that particulate matter in the municipalities of the Mezquital Valley presented values above 20 μg/m3 and that in accordance with the provisions of the Norma Oficial Mexicana de Salud Ambiental or NOM (translated as Mexican Official Standard for Environmental Health) and the Agency for Toxic Substances and the Disease Registry (ATSDR), high concentrations of particulate matter can have a severe impact on the development of some diseases. In the studied area, ischemic cardiomyopathy and mesothelioma were attributed to pollution in 70.3% and 79.1%, respectively; therefore, pollution mitigation could prevent the occurrence of these two diseases.
Collapse
Affiliation(s)
- Raymundo Ordoñez-Sierra
- Instituto Interamericano de Tecnología y Ciencias Del AguaUniversidad Autónoma Del Estado de México, Carretera Toluca-Atlacomulco, Km 14.5, Toluca, Estado de México, Mexico
- Facultad de GeografíaUniversidad Autónoma Del Estado de MéxicoCerro de Coatepec Sin NúmeroCiudad Universitaria, Toluca 50110, Estado de México, Mexico
| | | | - Iván Yassmany Hernández-Paniagua
- Instituto de Ciencias de la Atmósfera y Cambio ClimáticoCircuito de la Investigación Científica S/NUniversidad Nacional Autónoma de México, C.U., Coyoacán, Ciudad de México 04510, Mexico
| | - José Luis Expósito-Castillo
- Instituto Interamericano de Tecnología y Ciencias Del AguaUniversidad Autónoma Del Estado de México, Carretera Toluca-Atlacomulco, Km 14.5, Toluca, Estado de México, Mexico
| | - Miguel A. Gómez-Albores
- Instituto Interamericano de Tecnología y Ciencias Del AguaUniversidad Autónoma Del Estado de México, Carretera Toluca-Atlacomulco, Km 14.5, Toluca, Estado de México, Mexico
| | - María Guadalupe Rodríguez-Reyes
- Facultad de GeografíaUniversidad Autónoma Del Estado de MéxicoCerro de Coatepec Sin NúmeroCiudad Universitaria, Toluca 50110, Estado de México, Mexico
| | - Brisa Violeta Carrasco-Gallegos
- Facultad de GeografíaUniversidad Autónoma Del Estado de MéxicoCerro de Coatepec Sin NúmeroCiudad Universitaria, Toluca 50110, Estado de México, Mexico
| | - Luis Ricardo Manzano-Solís
- Facultad de GeografíaUniversidad Autónoma Del Estado de MéxicoCerro de Coatepec Sin NúmeroCiudad Universitaria, Toluca 50110, Estado de México, Mexico
| |
Collapse
|
6
|
Gruslova AB, Feldman MD. Can ethylene diamine tetra-acetic acid improve calcium fracture when combined with intravascular lithotripsy? Catheter Cardiovasc Interv 2024; 104:423-424. [PMID: 39011650 DOI: 10.1002/ccd.31075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 07/17/2024]
Abstract
Key points
Agents which dissolve hydroxyapatite may synergize with intravascular lithotripsy (IVL) to treat calcium plates more extensively, facilitating greater penetration of drug‐coated balloons delivered agents and maintain gain in lumen area without stenting.
Dissolving hydroxyapatite could potentially allow more frequent use of high‐pressure balloons without IVL or atherectomy to fracture calcium plates, offering a lower cost approach.
Other mechanisms such as debonding between calcium plate and fibrous tissue junctions may also be enhanced with IVL and calcium‐modifying agents to improve calcified vessel compliance.
Collapse
Affiliation(s)
- Alexandra B Gruslova
- Department of Medicine, Division of Cardiology, University of Texas Health at San Antonio, San Antonio, Texas, USA
| | - Marc D Feldman
- Department of Medicine, Division of Cardiology, University of Texas Health at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
7
|
Navas-Acien A, Santella RM, Joubert BR, Huang Z, Lokhnygina Y, Ujueta F, Gurvich I, LoIacono NJ, Ravalli F, Ward CD, Jarrett JM, Salazar ADL, Boineau R, Jones TLZ, Mark DB, Newman JD, Nathan DM, Anstrom KJ, Lamas GA. Baseline characteristics including blood and urine metal levels in the Trial to Assess Chelation Therapy 2 (TACT2). Am Heart J 2024; 273:72-82. [PMID: 38621575 PMCID: PMC11162898 DOI: 10.1016/j.ahj.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/05/2024] [Accepted: 04/11/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND The reduction in cardiovascular disease (CVD) events with edetate disodium (EDTA) in the Trial to Assess Chelation Therapy (TACT) suggested that chelation of toxic metals might provide novel opportunities to reduce CVD in patients with diabetes. Lead and cadmium are vasculotoxic metals chelated by EDTA. We present baseline characteristics for participants in TACT2, a randomized, double-masked, placebo-controlled trial designed as a replication of the TACT trial limited to patients with diabetes. METHODS TACT2 enrolled 1,000 participants with diabetes and prior myocardial infarction, age 50 years or older between September 2016 and December 2020. Among 959 participants with at least one infusion, 933 had blood and/or urine metals measured at the Centers for Diseases Control and Prevention using the same methodology as in the National Health and Nutrition Examination Survey (NHANES). We compared metal levels in TACT2 to a contemporaneous subset of NHANES participants with CVD, diabetes and other inclusion criteria similar to TACT2's participants. RESULTS At baseline, the median (interquartile range, IQR) age was 67 (60, 72) years, 27% were women, 78% reported white race, mean (SD) BMI was 32.7 (6.6) kg/m2, 4% reported type 1 diabetes, 46.8% were treated with insulin, 22.3% with GLP1-receptor agonists or SGLT-2 inhibitors, 90.2% with aspirin, warfarin or P2Y12 inhibitors, and 86.5% with statins. Blood lead was detectable in all participants; median (IQR) was 9.19 (6.30, 13.9) µg/L. Blood and urine cadmium were detectable in 97% and median (IQR) levels were 0.28 (0.18, 0.43) µg/L and 0.30 (0.18, 0.51) µg/g creatinine, respectively. Metal levels were largely similar to those in the contemporaneous NHANES subset. CONCLUSIONS TACT2 participants were characterized by high use of medication to treat CVD and diabetes and similar baseline metal levels as in the general US population. TACT2 will determine whether chelation therapy reduces the occurrence of subsequent CVD events in this high-risk population. CLINICAL TRIALS REGISTRATION ClinicalTrials.gov. Identifier: NCT02733185. https://clinicaltrials.gov/study/NCT02733185.
Collapse
Affiliation(s)
- Ana Navas-Acien
- Department of Environmental Health Sciences Columbia University Mailman School of Public Health New York, NY.
| | - Regina M Santella
- Department of Environmental Health Sciences Columbia University Mailman School of Public Health New York, NY
| | - Bonnie R Joubert
- National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC
| | - Zhen Huang
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC
| | - Yuliya Lokhnygina
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC; Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC
| | - Francisco Ujueta
- Department of Medicine at Mount Sinai Medical Center, Miami Beach, FL
| | - Irina Gurvich
- Department of Environmental Health Sciences Columbia University Mailman School of Public Health New York, NY
| | - Nancy J LoIacono
- Department of Environmental Health Sciences Columbia University Mailman School of Public Health New York, NY
| | - Filippo Ravalli
- Department of Environmental Health Sciences Columbia University Mailman School of Public Health New York, NY
| | - Cynthia D Ward
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA
| | - Jeffery M Jarrett
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA
| | - Alfonsina De Leon Salazar
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA
| | - Robin Boineau
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD
| | - Teresa L Z Jones
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Daniel B Mark
- Duke Clinical Research Institute, Duke University Medical Center, Durham, NC
| | | | - David M Nathan
- Massachusetts General Hospital Diabetes Center, Harvard Medical School, Boston, MA
| | - Kevin J Anstrom
- Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Gervasio A Lamas
- Department of Medicine at Mount Sinai Medical Center, Miami Beach, FL; Columbia University Division of Cardiology at Mount Sinai Medical Center, Miami Beach, FL
| |
Collapse
|
8
|
Madonna R, Biondi F, Alberti M, Ghelardoni S, Mattii L, D'Alleva A. Cardiovascular outcomes and molecular targets for the cardiac effects of Sodium-Glucose Cotransporter 2 Inhibitors: A systematic review. Biomed Pharmacother 2024; 175:116650. [PMID: 38678962 DOI: 10.1016/j.biopha.2024.116650] [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: 02/19/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024] Open
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2i), a new class of glucose-lowering drugs traditionally used to control blood glucose levels in patients with type 2 diabetes mellitus, have been proven to reduce major adverse cardiovascular events, including cardiovascular death, in patients with heart failure irrespective of ejection fraction and independently of the hypoglycemic effect. Because of their favorable effects on the kidney and cardiovascular outcomes, their use has been expanded in all patients with any combination of diabetes mellitus type 2, chronic kidney disease and heart failure. Although mechanisms explaining the effects of these drugs on the cardiovascular system are not well understood, their effectiveness in all these conditions suggests that they act at the intersection of the metabolic, renal and cardiac axes, thus disrupting maladaptive vicious cycles while contrasting direct organ damage. In this systematic review we provide a state of the art of the randomized controlled trials investigating the effect of SGLT2i on cardiovascular outcomes in patients with chronic kidney disease and/or heart failure irrespective of ejection fraction and diabetes. We also discuss the molecular targets and signaling pathways potentially explaining the cardiac effects of these pharmacological agents, from a clinical and experimental perspective.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Department of Pathology, Cardiology Division, University of Pisa, Via Paradisa, Pisa 56124, Italy.
| | - Filippo Biondi
- Department of Pathology, Cardiology Division, University of Pisa, Via Paradisa, Pisa 56124, Italy
| | - Mattia Alberti
- Department of Pathology, Cardiology Division, University of Pisa, Via Paradisa, Pisa 56124, Italy
| | - Sandra Ghelardoni
- Department of Pathology, Laboratory of Biochemistry, University of Pisa, Italy
| | - Letizia Mattii
- Department of Clinical and Experimental Medicine, Histology Division, University of Pisa, Pisa, Italy
| | - Alberto D'Alleva
- Cardiac Intensive Care and Interventional Cardiology Unit, Santo Spirito Hospital, Pescara, Italy
| |
Collapse
|
9
|
Abstract
Heavy metals are harmful environmental pollutants that have attracted widespread attention due to their health hazards to human cardiovascular disease. Heavy metals, including lead, cadmium, mercury, arsenic, and chromium, are found in various sources such as air, water, soil, food, and industrial products. Recent research strongly suggests a connection between cardiovascular disease and exposure to toxic heavy metals. Epidemiological, basic, and clinical studies have revealed that heavy metals can promote the production of reactive oxygen species, which can then exacerbate reactive oxygen species generation and induce inflammation, resulting in endothelial dysfunction, lipid metabolism distribution, disruption of ion homeostasis, and epigenetic changes. Over time, heavy metal exposure eventually results in an increased risk of hypertension, arrhythmia, and atherosclerosis. Strengthening public health prevention and the application of chelation or antioxidants, such as vitamins and beta-carotene, along with minerals, such as selenium and zinc, can diminish the burden of cardiovascular disease attributable to metal exposure.
Collapse
Affiliation(s)
- Ziwei Pan
- Key Laboratory of Combined Multi Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China (Z.P., P.L.)
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China (Z.P., P.L.)
| | - Tingyu Gong
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China (T.G.)
| | - Ping Liang
- Key Laboratory of Combined Multi Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China (Z.P., P.L.)
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China (Z.P., P.L.)
| |
Collapse
|
10
|
Abstract
Ubiquitous environmental exposures increase cardiovascular disease risk via diverse mechanisms. This review examines personal strategies to minimize this risk. With regard to fine particulate air pollution exposure, evidence exists to recommend the use of portable air cleaners and avoidance of outdoor activity during periods of poor air quality. Other evidence may support physical activity, dietary modification, omega-3 fatty acid supplementation, and indoor and in-vehicle air conditioning as viable strategies to minimize adverse health effects. There is currently insufficient data to recommend specific personal approaches to reduce the adverse cardiovascular effects of noise pollution. Public health advisories for periods of extreme heat or cold should be observed, with limited evidence supporting a warm ambient home temperature and physical activity as strategies to limit the cardiovascular harms of temperature extremes. Perfluoroalkyl and polyfluoroalkyl substance exposure can be reduced by avoiding contact with perfluoroalkyl and polyfluoroalkyl substance-containing materials; blood or plasma donation and cholestyramine may reduce total body stores of perfluoroalkyl and polyfluoroalkyl substances. However, the cardiovascular impact of these interventions has not been examined. Limited utilization of pesticides and safe handling during use should be encouraged. Finally, vasculotoxic metal exposure can be decreased by using portable air cleaners, home water filtration, and awareness of potential contaminants in ground spices. Chelation therapy reduces physiological stores of vasculotoxic metals and may be effective for the secondary prevention of cardiovascular disease.
Collapse
Affiliation(s)
- Luke J Bonanni
- Grossman School of Medicine (L.J.B.), NYU Langone Health, New York, NY
| | | |
Collapse
|
11
|
Bautista CJ, Arango N, Plata C, Mitre-Aguilar IB, Trujillo J, Ramírez V. Mechanism of cadmium-induced nephrotoxicity. Toxicology 2024; 502:153726. [PMID: 38191021 DOI: 10.1016/j.tox.2024.153726] [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: 11/07/2023] [Revised: 12/21/2023] [Accepted: 01/03/2024] [Indexed: 01/10/2024]
Abstract
Heavy metals are found naturally in our environment and have many uses and applications in daily life. However, high concentrations of metals may be a result of pollution due to industrialization. In particular, cadmium (Cd), a white metal abundantly distributed in the terrestrial crust, is found in mines together with zinc, which accumulates after volcanic eruption or is found naturally in the sea and earth. High levels of Cd have been associated with disease. In the human body, Cd accumulates in two ways: via inhalation or consumption, mainly of plants or fish contaminated with high concentrations. Several international organizations have been working to establish the limit values of heavy metals in food, water, and the environment to avoid their toxic effects. Increased Cd levels may induce kidney, liver, or neurological diseases. Cd mainly accumulates in the kidney, causing renal disease in people exposed to moderate to high levels, which leads to the development of end-stage chronic kidney disease or death. The aim of this review is to provide an overview of Cd-induced nephrotoxicity, the mechanisms of Cd damage, and the current treatments used to reduce the toxic effects of Cd exposure.
Collapse
Affiliation(s)
- Claudia J Bautista
- Departamento de Biología de la Reproducción del Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México 14080, Mexico
| | - Nidia Arango
- Departamento de Cirugía Experimental del Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México 14080, Mexico
| | - Consuelo Plata
- Departamento de Nefrología del Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México 14080, Mexico
| | - Irma B Mitre-Aguilar
- Unidad de Bioquímica del Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México 14080, Mexico
| | - Joyce Trujillo
- Consejo Nacional de Humanidades, Ciencia y Tecnología, Instituto Potosino de Investigación Científica y Tecnológica A. C. División de Materiales Avanzados (CONAHCYT-IPICYT-DMA), San Luis Potosí, Mexico
| | - Victoria Ramírez
- Departamento de Cirugía Experimental del Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México 14080, Mexico.
| |
Collapse
|
12
|
Lieberman‐Cribbin W, Li Z, Lewin M, Ruiz P, Jarrett JM, Cole SA, Kupsco A, O'Leary M, Pichler G, Shimbo D, Devereux RB, Umans JG, Navas‐Acien A, Nigra AE. The Contribution of Declines in Blood Lead Levels to Reductions in Blood Pressure Levels: Longitudinal Evidence in the Strong Heart Family Study. J Am Heart Assoc 2024; 13:e031256. [PMID: 38205795 PMCID: PMC10926826 DOI: 10.1161/jaha.123.031256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 11/21/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Chronic lead exposure is associated with both subclinical and clinical cardiovascular disease. We evaluated whether declines in blood lead were associated with changes in systolic and diastolic blood pressure in adult American Indian participants from the SHFS (Strong Heart Family Study). METHODS AND RESULTS Lead in whole blood was measured in 285 SHFS participants in 1997 to 1999 and 2006 to 2009. Blood pressure and measures of cardiac geometry and function were obtained in 2001 to 2003 and 2006 to 2009. We used generalized estimating equations to evaluate the association of declines in blood lead with changes in blood pressure; cardiac function and geometry measures were considered secondary. Mean blood lead was 2.04 μg/dL at baseline. After ≈10 years, mean decline in blood lead was 0.67 μg/dL. In fully adjusted models, the mean difference in systolic blood pressure comparing the highest to lowest tertile of decline (>0.91 versus <0.27 μg/dL) in blood lead was -7.08 mm Hg (95% CI, -13.16 to -1.00). A significant nonlinear association between declines in blood lead and declines in systolic blood pressure was detected, with significant linear associations where blood lead decline was 0.1 μg/dL or higher. Declines in blood lead were nonsignificantly associated with declines in diastolic blood pressure and significantly associated with declines in interventricular septum thickness. CONCLUSIONS Declines in blood lead levels in American Indian adults, even when small (0.1-1.0 μg/dL), were associated with reductions in systolic blood pressure. These findings suggest the need to further study the cardiovascular impacts of reducing lead exposures and the importance of lead exposure prevention.
Collapse
Affiliation(s)
- Wil Lieberman‐Cribbin
- Department of Environmental Health SciencesColumbia University Mailman School of Public HealthNew YorkNYUSA
| | - Zheng Li
- Office of Capacity Development and Applied Prevention Science, Agency for Toxic Substances and Disease RegistryAtlantaGAUSA
| | - Michael Lewin
- Office of Community Health and Hazard Assessment, Agency for Toxic Substances and Disease RegistryAtlantaGAUSA
| | - Patricia Ruiz
- Office of Innovation and Analytics, Agency for Toxic Substances and Disease RegistryAtlantaGAUSA
| | - Jeffery M. Jarrett
- Division for Laboratory SciencesCenters for Disease Control and PreventionAtlantaGAUSA
| | - Shelley A. Cole
- Population Health ProgramTexas Biomedical Research InstituteSan AntonioTXUSA
| | - Allison Kupsco
- Department of Environmental Health SciencesColumbia University Mailman School of Public HealthNew YorkNYUSA
| | - Marcia O'Leary
- Missouri Breaks Research Industries Research, Inc.Eagle ButteSDUSA
| | - Gernot Pichler
- Department of CardiologyKarl Landsteiner Institute for Cardiovascular and Critical Care Research, Clinic FloridsdorfViennaAustria
| | - Daichi Shimbo
- Division of CardiologyColumbia University Irving Medical CenterNew YorkNYUSA
| | | | - Jason G. Umans
- MedStar Health Research InstituteHyattsvilleMDUSA
- Georgetown‐Howard Universities Center for Clinical and Translational ScienceWashingtonDCUSA
| | - Ana Navas‐Acien
- Department of Environmental Health SciencesColumbia University Mailman School of Public HealthNew YorkNYUSA
| | - Anne E. Nigra
- Department of Environmental Health SciencesColumbia University Mailman School of Public HealthNew YorkNYUSA
| |
Collapse
|
13
|
Aaseth JO, Alehagen U, Opstad TB, Alexander J. Vitamin K and Calcium Chelation in Vascular Health. Biomedicines 2023; 11:3154. [PMID: 38137375 PMCID: PMC10740993 DOI: 10.3390/biomedicines11123154] [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: 10/10/2023] [Revised: 11/17/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
The observation that the extent of artery calcification correlates with the degree of atherosclerosis was the background for the alternative treatment of cardiovascular disease with chelator ethylenediamine tetraacetate (EDTA). Recent studies have indicated that such chelation treatment has only marginal impact on the course of vascular disease. In contrast, endogenous calcium chelation with removal of calcium from the cardiovascular system paralleled by improved bone mineralization exerted, i.e., by matrix Gla protein (MGP) and osteocalcin, appears to significantly delay the development of cardiovascular diseases. After post-translational vitamin-K-dependent carboxylation of glutamic acid residues, MGP and other vitamin-K-dependent proteins (VKDPs) can chelate calcium through vicinal carboxyl groups. Dietary vitamin K is mainly provided in the form of phylloquinone from green leafy vegetables and as menaquinones from fermented foods. Here, we provide a review of clinical studies, addressing the role of vitamin K in cardiovascular diseases, and an overview of vitamin K kinetics and biological actions, including vitamin-K-dependent carboxylation and calcium chelation, as compared with the action of the exogenous (therapeutic) chelator EDTA. Consumption of vitamin-K-rich foods and/or use of vitamin K supplements appear to be a better preventive strategy than EDTA chelation for maintaining vascular health.
Collapse
Affiliation(s)
- Jan O. Aaseth
- Research Department, Innlandet Hospital Trust, P.O. Box 104, N-2381 Brumunddal, Norway
- Faculty of Health and Social Sciences, Inland Norway University of Applied Sciences, P.O. Box 400, N-2418 Elverum, Norway
| | - Urban Alehagen
- Division of Cardiovascular Medicine, Department of Medical and Health Sciences, Linköping University, SE-581 85 Linköping, Sweden;
| | - Trine Baur Opstad
- Oslo Centre for Clinical Heart Research Laboratory, Department of Cardiology, Oslo University Hospital Ullevål, P.O. Box 4950, Nydalen, N-0424 Oslo, Norway;
- Faculty of Medicine, University of Oslo, N-0370 Oslo, Norway
| | - Jan Alexander
- Norwegian Institute of Public Health, P.O. Box 222, N-0213 Oslo, Norway;
| |
Collapse
|
14
|
Feldmann A, Nitschke Y, Linß F, Mulac D, Stücker S, Bertrand J, Buers I, Langer K, Rutsch F. Improved Reversion of Calcifications in Porcine Aortic Heart Valves Using Elastin-Targeted Nanoparticles. Int J Mol Sci 2023; 24:16471. [PMID: 38003660 PMCID: PMC10671589 DOI: 10.3390/ijms242216471] [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: 09/29/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Calcified aortic valve disease in its final stage leads to aortic valve stenosis, limiting cardiac function. To date, surgical intervention is the only option for treating calcific aortic valve stenosis. This study combined controlled drug delivery by nanoparticles (NPs) and active targeting by antibody conjugation. The chelating agent diethylenetriaminepentaacetic acid (DTPA) was covalently bound to human serum albumin (HSA)-based NP, and the NP surface was modified using conjugating antibodies (anti-elastin or isotype IgG control). Calcification was induced ex vivo in porcine aortic valves by preincubation in an osteogenic medium containing 2.5 mM sodium phosphate for five days. Valve calcifications mainly consisted of basic calcium phosphate crystals. Calcifications were effectively resolved by adding 1-5 mg DTPA/mL medium. Incubation with pure DTPA, however, was associated with a loss of cellular viability. Reversal of calcifications was also achieved with DTPA-coupled anti-elastin-targeted NPs containing 1 mg DTPA equivalent. The addition of these NPs to the conditioned media resulted in significant regression of the valve calcifications compared to that in the IgG-NP control without affecting cellular viability. These results represent a step further toward the development of targeted nanoparticular formulations to dissolve aortic valve calcifications.
Collapse
Affiliation(s)
- Anja Feldmann
- Department of General Pediatrics, Muenster University Children’s Hospital, D-48149 Muenster, Germany; (A.F.); (Y.N.); (I.B.)
- International Network of Ectopic Calcification (INTEC), 9000 Ghent, Belgium; (F.L.); (S.S.); (J.B.)
| | - Yvonne Nitschke
- Department of General Pediatrics, Muenster University Children’s Hospital, D-48149 Muenster, Germany; (A.F.); (Y.N.); (I.B.)
- International Network of Ectopic Calcification (INTEC), 9000 Ghent, Belgium; (F.L.); (S.S.); (J.B.)
| | - Franziska Linß
- International Network of Ectopic Calcification (INTEC), 9000 Ghent, Belgium; (F.L.); (S.S.); (J.B.)
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, D-48149 Muenster, Germany; (D.M.); (K.L.)
| | - Dennis Mulac
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, D-48149 Muenster, Germany; (D.M.); (K.L.)
| | - Sina Stücker
- International Network of Ectopic Calcification (INTEC), 9000 Ghent, Belgium; (F.L.); (S.S.); (J.B.)
- Department of Orthopaedic Surgery, Otto-von-Guericke-University Magdeburg, D-39120 Magdeburg, Germany
| | - Jessica Bertrand
- International Network of Ectopic Calcification (INTEC), 9000 Ghent, Belgium; (F.L.); (S.S.); (J.B.)
- Department of Orthopaedic Surgery, Otto-von-Guericke-University Magdeburg, D-39120 Magdeburg, Germany
| | - Insa Buers
- Department of General Pediatrics, Muenster University Children’s Hospital, D-48149 Muenster, Germany; (A.F.); (Y.N.); (I.B.)
- International Network of Ectopic Calcification (INTEC), 9000 Ghent, Belgium; (F.L.); (S.S.); (J.B.)
| | - Klaus Langer
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, D-48149 Muenster, Germany; (D.M.); (K.L.)
| | - Frank Rutsch
- Department of General Pediatrics, Muenster University Children’s Hospital, D-48149 Muenster, Germany; (A.F.); (Y.N.); (I.B.)
- International Network of Ectopic Calcification (INTEC), 9000 Ghent, Belgium; (F.L.); (S.S.); (J.B.)
| |
Collapse
|
15
|
Virani SS, Newby LK, Arnold SV, Bittner V, Brewer LC, Demeter SH, Dixon DL, Fearon WF, Hess B, Johnson HM, Kazi DS, Kolte D, Kumbhani DJ, LoFaso J, Mahtta D, Mark DB, Minissian M, Navar AM, Patel AR, Piano MR, Rodriguez F, Talbot AW, Taqueti VR, Thomas RJ, van Diepen S, Wiggins B, Williams MS. 2023 AHA/ACC/ACCP/ASPC/NLA/PCNA Guideline for the Management of Patients With Chronic Coronary Disease: A Report of the American Heart Association/American College of Cardiology Joint Committee on Clinical Practice Guidelines. J Am Coll Cardiol 2023; 82:833-955. [PMID: 37480922 DOI: 10.1016/j.jacc.2023.04.003] [Citation(s) in RCA: 163] [Impact Index Per Article: 81.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
AIM The "2023 AHA/ACC/ACCP/ASPC/NLA/PCNA Guideline for the Management of Patients With Chronic Coronary Disease" provides an update to and consolidates new evidence since the "2012 ACCF/AHA/ACP/AATS/PCNA/SCAI/STS Guideline for the Diagnosis and Management of Patients With Stable Ischemic Heart Disease" and the corresponding "2014 ACC/AHA/AATS/PCNA/SCAI/STS Focused Update of the Guideline for the Diagnosis and Management of Patients With Stable Ischemic Heart Disease." METHODS A comprehensive literature search was conducted from September 2021 to May 2022. Clinical studies, systematic reviews and meta-analyses, and other evidence conducted on human participants were identified that were published in English from MEDLINE (through PubMed), EMBASE, the Cochrane Library, Agency for Healthcare Research and Quality, and other selected databases relevant to this guideline. STRUCTURE This guideline provides an evidenced-based and patient-centered approach to management of patients with chronic coronary disease, considering social determinants of health and incorporating the principles of shared decision-making and team-based care. Relevant topics include general approaches to treatment decisions, guideline-directed management and therapy to reduce symptoms and future cardiovascular events, decision-making pertaining to revascularization in patients with chronic coronary disease, recommendations for management in special populations, patient follow-up and monitoring, evidence gaps, and areas in need of future research. Where applicable, and based on availability of cost-effectiveness data, cost-value recommendations are also provided for clinicians. Many recommendations from previously published guidelines have been updated with new evidence, and new recommendations have been created when supported by published data.
Collapse
|
16
|
Virani SS, Newby LK, Arnold SV, Bittner V, Brewer LC, Demeter SH, Dixon DL, Fearon WF, Hess B, Johnson HM, Kazi DS, Kolte D, Kumbhani DJ, LoFaso J, Mahtta D, Mark DB, Minissian M, Navar AM, Patel AR, Piano MR, Rodriguez F, Talbot AW, Taqueti VR, Thomas RJ, van Diepen S, Wiggins B, Williams MS. 2023 AHA/ACC/ACCP/ASPC/NLA/PCNA Guideline for the Management of Patients With Chronic Coronary Disease: A Report of the American Heart Association/American College of Cardiology Joint Committee on Clinical Practice Guidelines. Circulation 2023; 148:e9-e119. [PMID: 37471501 DOI: 10.1161/cir.0000000000001168] [Citation(s) in RCA: 436] [Impact Index Per Article: 218.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
AIM The "2023 AHA/ACC/ACCP/ASPC/NLA/PCNA Guideline for the Management of Patients With Chronic Coronary Disease" provides an update to and consolidates new evidence since the "2012 ACCF/AHA/ACP/AATS/PCNA/SCAI/STS Guideline for the Diagnosis and Management of Patients With Stable Ischemic Heart Disease" and the corresponding "2014 ACC/AHA/AATS/PCNA/SCAI/STS Focused Update of the Guideline for the Diagnosis and Management of Patients With Stable Ischemic Heart Disease." METHODS A comprehensive literature search was conducted from September 2021 to May 2022. Clinical studies, systematic reviews and meta-analyses, and other evidence conducted on human participants were identified that were published in English from MEDLINE (through PubMed), EMBASE, the Cochrane Library, Agency for Healthcare Research and Quality, and other selected databases relevant to this guideline. STRUCTURE This guideline provides an evidenced-based and patient-centered approach to management of patients with chronic coronary disease, considering social determinants of health and incorporating the principles of shared decision-making and team-based care. Relevant topics include general approaches to treatment decisions, guideline-directed management and therapy to reduce symptoms and future cardiovascular events, decision-making pertaining to revascularization in patients with chronic coronary disease, recommendations for management in special populations, patient follow-up and monitoring, evidence gaps, and areas in need of future research. Where applicable, and based on availability of cost-effectiveness data, cost-value recommendations are also provided for clinicians. Many recommendations from previously published guidelines have been updated with new evidence, and new recommendations have been created when supported by published data.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dave L Dixon
- Former Joint Committee on Clinical Practice Guideline member; current member during the writing effort
| | - William F Fearon
- Society for Cardiovascular Angiography and Interventions representative
| | | | | | | | - Dhaval Kolte
- AHA/ACC Joint Committee on Clinical Data Standards
| | | | | | | | - Daniel B Mark
- Former Joint Committee on Clinical Practice Guideline member; current member during the writing effort
| | | | | | | | - Mariann R Piano
- Former Joint Committee on Clinical Practice Guideline member; current member during the writing effort
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Kontoghiorghes GJ. The Vital Role Played by Deferiprone in the Transition of Thalassaemia from a Fatal to a Chronic Disease and Challenges in Its Repurposing for Use in Non-Iron-Loaded Diseases. Pharmaceuticals (Basel) 2023; 16:1016. [PMID: 37513928 PMCID: PMC10384919 DOI: 10.3390/ph16071016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/13/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
The iron chelating orphan drug deferiprone (L1), discovered over 40 years ago, has been used daily by patients across the world at high doses (75-100 mg/kg) for more than 30 years with no serious toxicity. The level of safety and the simple, inexpensive synthesis are some of the many unique properties of L1, which played a major role in the contribution of the drug in the transition of thalassaemia from a fatal to a chronic disease. Other unique and valuable clinical properties of L1 in relation to pharmacology and metabolism include: oral effectiveness, which improved compliance compared to the prototype therapy with subcutaneous deferoxamine; highly effective iron removal from all iron-loaded organs, particularly the heart, which is the major target organ of iron toxicity and the cause of mortality in thalassaemic patients; an ability to achieve negative iron balance, completely remove all excess iron, and maintain normal iron stores in thalassaemic patients; rapid absorption from the stomach and rapid clearance from the body, allowing a greater frequency of repeated administration and overall increased efficacy of iron excretion, which is dependent on the dose used and also the concentration achieved at the site of drug action; and its ability to cross the blood-brain barrier and treat malignant, neurological, and microbial diseases affecting the brain. Some differential pharmacological activity by L1 among patients has been generally shown in relation to the absorption, distribution, metabolism, elimination, and toxicity (ADMET) of the drug. Unique properties exhibited by L1 in comparison to other drugs include specific protein interactions and antioxidant effects, such as iron removal from transferrin and lactoferrin; inhibition of iron and copper catalytic production of free radicals, ferroptosis, and cuproptosis; and inhibition of iron-containing proteins associated with different pathological conditions. The unique properties of L1 have attracted the interest of many investigators for drug repurposing and use in many pathological conditions, including cancer, neurodegenerative conditions, microbial conditions, renal conditions, free radical pathology, metal intoxication in relation to Fe, Cu, Al, Zn, Ga, In, U, and Pu, and other diseases. Similarly, the properties of L1 increase the prospects of its wider use in optimizing therapeutic efforts in many other fields of medicine, including synergies with other drugs.
Collapse
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol 3021, Cyprus
| |
Collapse
|
18
|
Baccarelli A, Dolinoy DC, Walker CL. A precision environmental health approach to prevention of human disease. Nat Commun 2023; 14:2449. [PMID: 37117186 PMCID: PMC10147599 DOI: 10.1038/s41467-023-37626-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/24/2023] [Indexed: 04/30/2023] Open
Abstract
Human health is determined by the interaction of our environment with the genome, epigenome, and microbiome, which shape the transcriptomic, proteomic, and metabolomic landscape of cells and tissues. Precision environmental health is an emerging field leveraging environmental and system-level ('omic) data to understand underlying environmental causes of disease, identify biomarkers of exposure and response, and develop new prevention and intervention strategies. In this article we provide real-life illustrations of the utility of precision environmental health approaches, identify current challenges in the field, and outline new opportunities to promote health through a precision environmental health framework.
Collapse
Affiliation(s)
- Andrea Baccarelli
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA.
| | - Dana C Dolinoy
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Cheryl Lyn Walker
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
19
|
Yen TH, Yen JS. Chelation trial in patients with cardiovascular disease. Am Heart J 2023; 256:1. [PMID: 36336079 DOI: 10.1016/j.ahj.2022.10.084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Affiliation(s)
- Tzung-Hai Yen
- Department of Nephrology and Clinical Poison Center, Chang Gung Memorial Hospital, Linkou Branch, and College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Ju-Shao Yen
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| |
Collapse
|
20
|
Lamas GA. Response to: Letter to the editor by Yen. Am Heart J 2023; 256:158. [PMID: 36336080 DOI: 10.1016/j.ahj.2022.10.085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Affiliation(s)
- Gervasio A Lamas
- Principal Investigator, TACT2, Mount Sinai Medical Center, Miami Beach, FL.
| |
Collapse
|