268 results on '"Plutzky J"'
Search Results
2. Bempedoic Acid and Cardiovascular Outcomes in Statin-Intolerant Patients
- Author
-
Nissen, SEE, Lincoff, AM, Brennan, D, Ray, KKK, Mason, D, Kastelein, JJP, Thompson, PDD, Libby, P, Cho, L, Plutzky, J, Bays, HEE, Moriarty, PMM, Menon, V, Grobbee, DEE, Louie, MJJ, Chen, C-F, Li, N, Bloedon, LA, Robinson, P, Horner, M, Sasiela, WJJ, McCluskey, J, Davey, D, Fajardo-Campos, P, Petrovic, P, Fedacko, J, Zmuda, W, Lukyanov, Y, Nicholls, SJJ, CLEAR, OI, Vascular Medicine, ACS - Atherosclerosis & ischemic syndromes, and ACS - Pulmonary hypertension & thrombosis
- Subjects
Coronary Disease/Myocardial Infarction ,Outpatient-Based Clinical Medicine ,Prevention ,Cardiology ,General Medicine ,Clinical Medicine General ,Clinical Medicine ,Lipids - Abstract
BACKGROUND Bempedoic acid, an ATP citrate lyase inhibitor, reduces low-density lipoprotein (LDL) cholesterol levels and is associated with a low incidence of muscle-related adverse events; its effects on cardiovascular outcomes remain uncertain. METHODS We conducted a double-blind, randomized, placebo-controlled trial involving patients who were unable or unwilling to take statins owing to unacceptable adverse effects (“statin-intolerant” patients) and had, or were at high risk for, cardiovascular disease. The patients were assigned to receive oral bempedoic acid, 180 mg daily, or placebo. The primary end point was a four-component composite of major adverse cardiovascular events, defined as death from cardiovascular causes, nonfatal myocardial infarction, nonfatal stroke, or coronary revascularization. RESULTS A total of 13,970 patients underwent randomization; 6992 were assigned to the bempedoic acid group and 6978 to the placebo group. The median duration of follow-up was 40.6 months. The mean LDL cholesterol level at baseline was 139.0 mg per deciliter in both groups, and after 6 months, the reduction in the level was greater with bempedoic acid than with placebo by 29.2 mg per deciliter; the observed difference in the percent reductions was 21.1 percentage points in favor of bempedoic acid. The incidence of a primary end-point event was significantly lower with bempedoic acid than with placebo (819 patients [11.7%] vs. 927 [13.3%]; hazard ratio, 0.87; 95% confidence interval [CI], 0.79 to 0.96; P=0.004), as were the incidences of a composite of death from cardiovascular causes, nonfatal stroke, or nonfatal myocardial infarction (575 [8.2%] vs. 663 [9.5%]; hazard ratio, 0.85; 95% CI, 0.76 to 0.96; P=0.006); fatal or nonfatal myocardial infarction (261 [3.7%] vs. 334 [4.8%]; hazard ratio, 0.77; 95% CI, 0.66 to 0.91; P=0.002); and coronary revascularization (435 [6.2%] vs. 529 [7.6%]; hazard ratio, 0.81; 95% CI, 0.72 to 0.92; P=0.001). Bempedoic acid had no significant effects on fatal or nonfatal stroke, death from cardiovascular causes, and death from any cause. The incidences of gout and cholelithiasis were higher with bempedoic acid than with placebo (3.1% vs. 2.1% and 2.2% vs. 1.2%, respectively), as were the incidences of small increases in serum creatinine, uric acid, and hepatic-enzyme levels. CONCLUSIONS Among statin-intolerant patients, treatment with bempedoic acid was associated with a lower risk of major adverse cardiovascular events (death from cardiovascular causes, nonfatal myocardial infarction, nonfatal stroke, or coronary revascularization). (Funded by Esperion Therapeutics; CLEAR Outcomes ClinicalTrials.gov number, NCT02993406. opens in new tab.)
- Published
- 2023
3. Perivascular Fibrosis Is Mediated by a KLF10-IL-9 Signaling Axis in CD4+ T Cells
- Author
-
Zhuang R, Chen J, Cheng H, Assa C, Jamaiyar A, Pandey A, Perez-Cremades D, Zhang B, Tzani A, Khyrul Wara A, Plutzky J, Barrera V, Bhetariya P, Mitchell R, Liu Z, and Feinberg M
- Abstract
BACKGROUND: Perivascular fibrosis, characterized by increased amount of connective tissue around vessels, is a hallmark for vascular disease. Ang II (angiotensin II) contributes to vascular disease and end-organ damage via promoting T-cell activation. Despite recent data suggesting the role of T cells in the progression of perivascular fibrosis, the underlying mechanisms are poorly understood.; METHODS: TF (transcription factor) profiling was performed in peripheral blood mononuclear cells of hypertensive patients. CD4-targeted KLF10 (Kruppel like factor 10)-deficient (Klf10fl/flCD4Cre+; [TKO]) and CD4-Cre (Klf10+/+CD4Cre+; (Cre)) control mice were subjected to Ang II infusion. End point characterization included cardiac echocardiography, aortic imaging, multiorgan histology, flow cytometry, cytokine analysis, aorta and fibroblast transcriptomic analysis, and aortic single-cell RNA-sequencing.; RESULTS: TF profiling identified increased KLF10 expression in hypertensive human subjects and in CD4+ T cells in Ang II-treated mice. TKO mice showed enhanced perivascular fibrosis, but not interstitial fibrosis, in aorta, heart, and kidney in response to Ang II, accompanied by alterations in global longitudinal strain, arterial stiffness, and kidney function compared with Cre control mice. However, blood pressure was unchanged between the 2 groups. Mechanistically, KLF10 bound to the IL (interleukin)-9 promoter and interacted with HDAC1 (histone deacetylase 1) inhibit IL-9 transcription. Increased IL-9 in TKO mice induced fibroblast intracellular calcium mobilization, fibroblast activation, and differentiation and increased production of collagen and extracellular matrix, thereby promoting the progression of perivascular fibrosis and impairing target organ function. Remarkably, injection of anti-IL9 antibodies reversed perivascular fibrosis in Ang II-infused TKO mice and C57BL/6 mice. Single-cell RNA-sequencing revealed fibroblast heterogeneity with activated signatures associated with robust ECM (extracellular matrix) and perivascular fibrosis in Ang II-treated TKO mice.; CONCLUSIONS: CD4+ T cell deficiency of Klf10 exacerbated perivascular fibrosis and multi-organ dysfunction in response to Ang II via upregulation of IL-9. Klf10 or IL-9 in T cells might represent novel therapeutic targets for treatment of vascular or fibrotic diseases.
- Published
- 2022
4. Osteopontin deficiency protects against obesity-induced hepatic steatosis and attenuates glucose production in mice
- Author
-
Kiefer, F. W., Neschen, S., Pfau, B., Legerer, B., Neuhofer, A., Kahle, M., Hrabé de Angelis, M., Schlederer, M., Mair, M., Kenner, L., Plutzky, J., Zeyda, M., and Stulnig, T. M.
- Published
- 2011
- Full Text
- View/download PDF
5. Liraglutide, a Once-Daily Human GLP-1 Analog, Reduces Systolic Blood Pressure with Minimal Impact from Weight Loss in Subjects with Type 2 Diabetes.
- Author
-
Henry, RR, primary, Fonseca, V, additional, Tabanera y Palacios, R, additional, Brett, J, additional, and Plutzky, J, additional
- Published
- 2010
- Full Text
- View/download PDF
6. RETINALDEHYDE DEHYDROGENASE 1 IS A NOVEL REGULATOR OF BROWN FAT FUNCTION IN WHITE ADIPOSE TISSUE: 169 accepted oral
- Author
-
Kiefer, F., Vernochet, C., OʼBrian, P., Spörl, S., Brown, J., Kahn, C. R., and Plutzky, J.
- Published
- 2012
7. THE POTENTIAL ROLE OF GLP-1 AT THE INTERSECTION OF METABOLISM AND CARDIOVASCULAR DISEASE
- Author
-
Plutzky, J.
- Published
- 2011
8. The selective peroxisome proliferator-activated receptor alpha modulator (SPPARMα) paradigm: conceptual framework and therapeutic potential : A consensus statement from the International Atherosclerosis Society (IAS) and the Residual Risk Reduction Initiative (R3i) Foundation
- Author
-
Fruchart, J.C., Santos, R.D., Aguilar-Salinas, C., Aikawa, M., Al Rasadi, K., Amarenco, P., Barter, P.J., Ceska, R., Corsini, A., Després, J.P., Duriez, P., Eckel, R.H., Ezhov, M.V., Farnier, M., Ginsberg, H.N., Hermans, M.P., Ishibashi, S., Karpe, F., Kodama, T., Koenig, W., Krempf, M., Lim, S., Lorenzatti, A.J., McPherson, R., Nuñez-Cortes, J.M., Nordestgaard, B.G., Ogawa, H., Packard, C.J., Plutzky, J., Ponte-Negretti, C.I., Pradhan, A., Ray, K.K., Reiner, Ž., Ridker, P.M., Ruscica, M., Sadikot, S., Shimano, H., Sritara, P., Stock, J.K., Su, T.C., Susekov, A.V., Tartar, A., Taskinen, M.R., Tenenbaum, A., Tokgözoğlu, L.S., Tomlinson, B., Tybjærg-Hansen, A., Valensi, P., Vrablík, M., Wahli, W., Watts, G.F., Yamashita, S., Yokote, K., Zambon, A., and Libby, P.
- Subjects
Animals ,Benzoxazoles/adverse effects ,Benzoxazoles/therapeutic use ,Biomarkers/blood ,Butyrates/adverse effects ,Butyrates/therapeutic use ,Cardiovascular Diseases/blood ,Cardiovascular Diseases/diagnosis ,Cardiovascular Diseases/prevention & control ,Consensus ,Dyslipidemias/blood ,Dyslipidemias/diagnosis ,Dyslipidemias/drug therapy ,Humans ,Hypolipidemic Agents/adverse effects ,Hypolipidemic Agents/therapeutic use ,Lipids/blood ,Molecular Targeted Therapy ,PPAR alpha/agonists ,PPAR alpha/metabolism ,Patient Safety ,Risk Assessment ,Risk Factors ,Signal Transduction ,Treatment Outcome ,Atherogenic dyslipidemia ,Diabetes ,Inflammation ,PROMINENT ,Pemafibrate (K-877) ,Remnant cholesterol ,Residual cardiovascular risk ,SPPARMalpha ,Selective peroxisome proliferator-activated receptor alpha modulator ,Triglycerides ,Visceral obesity - Abstract
In the era of precision medicine, treatments that target specific modifiable characteristics of high-risk patients have the potential to lower further the residual risk of atherosclerotic cardiovascular events. Correction of atherogenic dyslipidemia, however, remains a major unmet clinical need. Elevated plasma triglycerides, with or without low levels of high-density lipoprotein cholesterol (HDL-C), offer a key modifiable component of this common dyslipidemia, especially in insulin resistant conditions such as type 2 diabetes mellitus. The development of selective peroxisome proliferator-activated receptor alpha modulators (SPPARMα) offers an approach to address this treatment gap. This Joint Consensus Panel appraised evidence for the first SPPARMα agonist and concluded that this agent represents a novel therapeutic class, distinct from fibrates, based on pharmacological activity, and, importantly, a safe hepatic and renal profile. The ongoing PROMINENT cardiovascular outcomes trial is testing in 10,000 patients with type 2 diabetes mellitus, elevated triglycerides, and low levels of HDL-C whether treatment with this SPPARMα agonist safely reduces residual cardiovascular risk.
- Published
- 2019
9. PPARs and their metabolic modulation: new mechanisms for transcriptional regulation?
- Author
-
Ahmed, W., Ziouzenkova, O., Brown, J., Devchand, P., Francis, S., Kadakia, M., Kanda, T., Orasanu, G., Sharlach, M., Zandbergen, F., and Plutzky, J.
- Published
- 2007
10. The residual risk reduction initiative: A call to action to reduce residual vascular risk in dyslipidaemic patients ( Review )
- Author
-
Fruchart, J. C., Sacks, F. M., Hermans, M. P., Assmann, G., Brown, W. V., Ceska, R., Chapman, M. J., Dodson, P. M., Fioretto, Paola, Ginsberg, H. N., Kadowaki, T., Lablanche, J. M., Marx, N., Plutzky, J., Reiner, Z., Rosenson, R. S., Staels, B., Stock, J. K., Sy, R., Wanner, C., Zambon, Alberto, and Zimmet, P.
- Published
- 2010
11. The Residual Risk Reduction Initiative: a call to action to reduce residual vascular risk in dyslipidaemic patient
- Author
-
Fruchart, Jc, Sacks, Fm, Hermans, Mp, Assmann, G, Brown, Wv, Ceska, R, Chapman, Mj, Dodson, Pm, Fioretto, Paola, Ginsberg, Hn, Kadowaki, T, Lablanche, Jm, Marx, N, Plutzky, J, Reiner, Z, Rosenson, Rs, Staels, B, Stock, Jk, Sy, R, Wanner, C, Zambon, Alberto, Zimmet, P, and Residual Risk Reduction Initiative
- Subjects
medicine.medical_specialty ,Health Knowledge, Attitudes, Practice ,medicine.drug_class ,Endocrinology, Diabetes and Metabolism ,Fibrate ,Type 2 diabetes ,Health Promotion ,chemistry.chemical_compound ,Pharmacotherapy ,Diabetes mellitus ,Internal medicine ,Internal Medicine ,medicine ,Humans ,Intensive care medicine ,Dyslipidemias ,Hypolipidemic Agents ,Evidence-Based Medicine ,business.industry ,Cholesterol ,Microcirculation ,nutritional and metabolic diseases ,medicine.disease ,Atherosclerosis ,Combined Modality Therapy ,Residual risk ,Endocrinology ,Treatment Outcome ,chemistry ,Cardiovascular Diseases ,Practice Guidelines as Topic ,lipids (amino acids, peptides, and proteins) ,Metabolic syndrome ,Cardiology and Cardiovascular Medicine ,business ,Risk Reduction Behavior ,Niacin - Abstract
Despite current standards of care aimed at achieving targets for low-density lipoprotein (LDL) cholesterol, blood pressure and glycaemia, dyslipidaemic patients remain at high residual risk of vascular events. Atherogenic dyslipidaemia, specifically elevated triglycerides and low levels of high-density lipoprotein (HDL) cholesterol, often with elevated apolipoprotein B and non-HDL cholesterol, is common in patients with established cardiovascular disease, type 2 diabetes, obesity or metabolic syndrome and is associated with macrovascular and microvascular residual risk. The Residual Risk Reduction Initiative (R3I) was established to address this important issue. This position paper aims to highlight evidence that atherogenic dyslipidaemia contributes to residual macrovascular risk and microvascular complications despite current standards of care for dyslipidaemia and diabetes, and to recommend therapeutic intervention for reducing this, supported by evidence and expert consensus. Lifestyle modification is an important first step. Additionally, pharmacotherapy is often required. Adding niacin, a fibrate or omega-3 fatty acids to statin therapy improves achievement of all lipid risk factors. Outcomes studies are evaluating whether these strategies translate to greater clinical benefit than statin therapy alone. In conclusion, the R3i highlights the need to address with lifestyle and/or pharmacotherapy the high level of residual vascular risk among dyslipidaemic patients who are treated in accordance with current standards of care.
- Published
- 2008
12. The Residual Risk Reduction Initiative: A call to action to reduce residual vascular risk in dyslipidemic patients. A position paper by the Residual Risk Reduction Initiative (R3I)
- Author
-
Fruchart, J-C., Sacks, F., Hermans, M.P., Assmann, G., Brown, V.W., Ceska, R., Chapman, M.J., Dodson, P.M., Fioretto, P., Ginsberg, H.N. Kadowaki, T. Lablanche, J-M., Marx, N., Plutzky, J., Reiner, Željko, Rosenson, R.S., Smith, S.C. Jr., Staels, B., Stock, J.Kk, Sy, R., Wanner, C., Zambon, A., and Zimmet, P.
- Subjects
dyslipidaemia ,residual risk ,nutritional and metabolic diseases ,lipids (amino acids, peptides, and proteins) - Abstract
Despite current standards of care aimed at achieving targets for low-density lipoprotein (LDL) cholesterol, blood pressure and glycaemia, dyslipidaemic patients remain at high residual risk of vascular events. Atherogenic dyslipidaemia, specifically elevated triglycerides and low levels of high-density lipoprotein (HDL) cholesterol, often with elevated apolipoprotein B and non-HDL cholesterol, is common in patients with established cardiovascular disease, type 2 diabetes, obesity or metabolic syndrome and is associated with macrovascular and microvascular residual risk.
- Published
- 2008
13. (Mis)interpreting studies on the adverse effects of statins
- Author
-
Zhang, H., primary, Plutzky, J., additional, and Turchin, A., additional
- Published
- 2014
- Full Text
- View/download PDF
14. Discontinuation of Statin in Routine Care Settings: A Cohort Study
- Author
-
Zhang, H., primary, Plutzky, J., additional, and Skentzos, S., additional
- Published
- 2013
- Full Text
- View/download PDF
15. Abstract: 511 THE RXR-PPAR COMPLEX IN THE REGULATION OF ENERGY BALANCE
- Author
-
Plutzky, J, primary
- Published
- 2009
- Full Text
- View/download PDF
16. Abstract: P640 PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR-Γ ACTIVATION REDUCES INFLAMMATION IN ATHEROSCLEROSIS AS ASSESSED BY SERIAL IN VIVO FLUORESCENCE MOLECULAR IMAGING
- Author
-
Chang, K, primary, Francis, S, additional, Aikawa, E, additional, Figueiredo, J, additional, Patel, P, additional, Weissleder, R, additional, Plutzky, J, additional, and Jaffer, F, additional
- Published
- 2009
- Full Text
- View/download PDF
17. Abstract: 124 PATHOPHYSIOLOGY OF MET SYN AND RELATED CARDIOMETABOLIC RISK: WHAT'S NEW?
- Author
-
Plutzky, J, primary and Magliaro, A, additional
- Published
- 2009
- Full Text
- View/download PDF
18. Reductions in lipids and CV risk markers in patients with type 2 diabetes treated with liraglutide: a meta-analysis
- Author
-
Plutzky, J., primary, Garber, A., additional, Falahati, A., additional, Toft, A.D., additional, and Poulter, N.R., additional
- Published
- 2009
- Full Text
- View/download PDF
19. Liraglutide, a once-daily human GLP-1 analogue, reduces systolic blood pressure within 2 weeks in patients with type 2 diabetes
- Author
-
Fonseca, V., primary, Falahati, A., additional, Zychma, M., additional, Madsbad, S., additional, and Plutzky, J., additional
- Published
- 2009
- Full Text
- View/download PDF
20. PPARα in atherosclerosis and inflammation
- Author
-
ZANDBERGEN, F, primary and PLUTZKY, J, additional
- Published
- 2007
- Full Text
- View/download PDF
21. PPARs in Diabetes and Cardiovascular Disease
- Author
-
Alexander, W., primary and Plutzky, J., additional
- Published
- 2006
- Full Text
- View/download PDF
22. Peroxisome Proliferator-Activated Receptors (PPARs) and Their Role in the Vessel Wall: Possible Mediators of Cardiovascular Risk?
- Author
-
Marx, N., primary, Libby, P., additional, and Plutzky, J., additional
- Published
- 2001
- Full Text
- View/download PDF
23. Identification of a human src homology 2-containing protein-tyrosine-phosphatase: a putative homolog of Drosophila corkscrew.
- Author
-
Freeman, R M, primary, Plutzky, J, additional, and Neel, B G, additional
- Published
- 1992
- Full Text
- View/download PDF
24. Chromosomal localization of an SH2-containing tyrosine phosphatase (PTPN6)
- Author
-
Plutzky, J., primary, Neel, B.G., additional, Rosenberg, R.D., additional, Eddy, R.L., additional, Byers, M.G., additional, Jani-Sait, S., additional, and Shows, T.B., additional
- Published
- 1992
- Full Text
- View/download PDF
25. Isolation of a src homology 2-containing tyrosine phosphatase.
- Author
-
Plutzky, J, primary, Neel, B G, additional, and Rosenberg, R D, additional
- Published
- 1992
- Full Text
- View/download PDF
26. Modulating peroxisome proliferator-activated receptors for therapeutic benefit? Biology, clinical experience, and future prospects.
- Author
-
Rosenson RS, Wright RS, Farkouh M, Plutzky J, Rosenson, Robert S, Wright, R Scott, Farkouh, Michael, and Plutzky, Jorge
- Abstract
Clinical trials of cardiovascular disease (CVD) prevention in patients with type 2 diabetes mellitus primarily have been directed at the modification of a single major risk factor; however, in trials that enroll patients with and without diabetes, the absolute risk in CVD events remains higher in patients with diabetes. Efforts to reduce the macrovascular and microvascular residual risk have been directed toward a multifactorial CVD risk-factor modification; nonetheless, long-term complications remain high. Dual-peroxisome proliferator-activated receptor (PPAR) α/γ agonists may offer opportunities to lower macrovascular and microvascular complications of type 2 diabetes mellitus beyond the reductions achieved with conventional risk-factor modification. The information presented elucidates the differentiation of compound-specific vs class-effect properties of PPARs as the basis for future development of a new candidate molecule. Prior experience with thiazolidinediones, an approved class of PPARγ agonists, and glitazars, investigational class of dual-PPARα/γ agonists, also provides important lessons about the risks and benefits of targeting a nuclear receptor while revealing some of the future challenges for regulatory approval. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
27. Fifty shades of brown: perivascular fat, thermogenesis, and atherosclerosis.
- Author
-
Kiefer FW, Cohen P, Plutzky J, Kiefer, Florian W, Cohen, Paul, and Plutzky, Jorge
- Published
- 2012
- Full Text
- View/download PDF
28. The incretin axis in cardiovascular disease.
- Author
-
Plutzky J
- Published
- 2011
- Full Text
- View/download PDF
29. Deletion of the alpha-arrestin protein Txnip in mice promotes adiposity and adipogenesis while preserving insulin sensitivity.
- Author
-
Chutkow WA, Birkenfeld AL, Brown JD, Lee HY, Frederick DW, Yoshioka J, Patwari P, Kursawe R, Cushman SW, Plutzky J, Shulman GI, Samuel VT, Lee RT, Chutkow, William A, Birkenfeld, Andreas L, Brown, Jonathan D, Lee, Hui-Young, Frederick, David W, Yoshioka, Jun, and Patwari, Parth
- Abstract
Objective: Thioredoxin interacting protein (Txnip), a regulator of cellular oxidative stress, is induced by hyperglycemia and inhibits glucose uptake into fat and muscle, suggesting a role for Txnip in type 2 diabetes pathogenesis. Here, we tested the hypothesis that Txnip-null (knockout) mice are protected from insulin resistance induced by a high-fat diet.Research Design and Methods: Txnip gene-deleted (knockout) mice and age-matched wild-type littermate control mice were maintained on a standard chow diet or subjected to 4 weeks of high-fat feeding. Mice were assessed for body composition, fat development, energy balance, and insulin responsiveness. Adipogenesis was measured from ex vivo fat preparations, and in mouse embryonic fibroblasts (MEFs) and 3T3-L1 preadipocytes after forced manipulation of Txnip expression.Results: Txnip knockout mice gained significantly more adipose mass than controls due to a primary increase in both calorie consumption and adipogenesis. Despite increased fat mass, Txnip knockout mice were markedly more insulin sensitive than controls, and augmented glucose transport was identified in both adipose and skeletal muscle. RNA interference gene-silenced preadipocytes and Txnip(-/-) MEFs were markedly adipogenic, whereas Txnip overexpression impaired adipocyte differentiation. As increased adipogenesis and insulin sensitivity suggested aspects of augmented peroxisome proliferator-activated receptor-gamma (PPARgamma) response, we investigated Txnip's regulation of PPARgamma function; manipulation of Txnip expression directly regulated PPARgamma expression and activity.Conclusions: Txnip deletion promotes adiposity in the face of high-fat caloric excess; however, loss of this alpha-arrestin protein simultaneously enhances insulin responsiveness in fat and skeletal muscle, revealing Txnip as a novel mediator of insulin resistance and a regulator of adipogenesis. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
30. Peroxisome proliferator-activated receptors as transcriptional nodal points and therapeutic targets.
- Author
-
Brown JD and Plutzky J
- Published
- 2007
31. Primary prevention of cardiovascular diseases in people with diabetes mellitus: a scientific statement from the American Heart Association and the American Diabetes Association.
- Author
-
Buse JB, Ginsberg HN, Bakris GL, Clark NG, Costa F, Eckel R, Fonseca V, Gerstein HC, Grundy S, Nesto RW, Pignone MP, Plutzky J, Porte D, Redberg R, Stitzel KF, Stone NJ, and American Heart Association
- Published
- 2007
32. The cardiovascular disease continuum validated: clinical evidence of improved patient outcomes: part I: Pathophysiology and clinical trial evidence (risk factors through stable coronary artery disease)
- Author
-
Dzau VJ, Antman EM, Black HR, Hayes DL, Manson JE, Plutzky J, Popma JJ, and Stevenson W
- Published
- 2006
33. The cardiovascular disease continuum validated: clinical evidence of improved patient outcomes: part II: Clinical trial evidence (acute coronary syndromes through renal disease) and future directions.
- Author
-
Dzau VJ, Antman EM, Black HR, Hayes DL, Manson JE, Plutzky J, Popma JJ, and Stevenson W
- Published
- 2006
34. Liver X receptor activation and high-density lipoprotein biology: a reversal of fortune?
- Author
-
Lee CH, Plutzky J, Lee, Chih-Hao, and Plutzky, Jorge
- Published
- 2006
35. Effect of a peroxisome proliferator-activated receptor-gamma agonist on myocardial blood flow in type 2 diabetes.
- Author
-
McMahon GT, Plutzky J, Daher E, Bhattacharyya T, Grunberger G, DiCarli MF, McMahon, Graham T, Plutzky, Jorge, Daher, Edouard, Bhattacharyya, Tammy, Grunberger, George, and DiCarli, Marcelo F
- Abstract
Objective: The relationship between coronary endothelial function and insulin resistance remains speculative. We sought to determine whether pioglitazone, an insulin-sensitizing peroxisome proliferator-activated receptor (PPAR)-gamma agonist, improves cardiac endothelial function in individuals with type 2 diabetes.Research Design and Methods: Sixteen subjects with insulin-treated type 2 diabetes and without overt cardiovascular disease were randomly assigned to receive either 45 mg of pioglitazone or matching placebo for 3 months. Rest and adenosine-stimulated myocardial blood flow (MBF) were quantified with [(13)N]ammonia and positron emission tomography at baseline and study conclusion.Results: After 3 months, HbA(1c) levels dropped by 0.68% in the pioglitazone group and increased by 0.17% in the placebo group (P = 0.009 for difference between groups). Triglyceride (-93 vs. -39 mg/dl, P = 0.026) and HDL concentrations (+4.8 vs. -6.0 mg/dl, P = 0.014) improved significantly in the pioglitazone group compared with placebo. Despite these favorable changes, there was no demonstrable change in baseline MBF (-0.05 +/- 0.24 vs. -0.09 +/- 0.24 ml . min(-1) . g(-1), P = 0.45), adenosine-stimulated MBF (0.10 +/- 0.75 vs. 0.14 +/- 0.31 ml . min(-1) . g(-1), P = 0.25), or coronary flow reserve (0.45 +/- 1.22 vs. 0.35 +/- 0.72 ml . min(-1) . g(-1), P = 0.64) after 12 weeks of exposure to pioglitazone or placebo, respectively. Regression analysis revealed that lower glucose concentration at the time of the study was associated with higher coronary flow reserve (P = 0.012).Conclusions: Pioglitazone treatment for 12 weeks in subjects with insulin-requiring type 2 diabetes had no demonstrable effect on coronary flow reserve despite metabolic improvements. Higher ambient glucose levels contribute to impaired vascular reactivity in individuals with diabetes. [ABSTRACT FROM AUTHOR]- Published
- 2005
- Full Text
- View/download PDF
36. Inflammatory pathways in atherosclerosis and acute coronary syndromes.
- Author
-
Plutzky, Jorge and Plutzky, J
- Subjects
- *
ATHEROSCLEROSIS , *CARDIOVASCULAR system , *BLOOD proteins - Abstract
Evidence from a broad range of studies demonstrates that atherosclerosis is a chronic disease that, from its origins to its ultimate complications, involves inflammatory cells (T cells, monocytes, macrophages), inflammatory proteins (cytokines, chemokines), and inflammatory responses from vascular cells (endothelial cell expression of adhesion molecules). Investigators have identified a variety of proteins whose levels might predict cardiovascular risk. Of these candidates, C-reactive protein, tumor necrosis factor-alpha, and interleukin-6 have been most widely studied. There is also the prospect of inflammation as a therapeutic target, with investigators currently debating to what extent the decrease in cardiovascular risk seen with statins, angiotensin-converting enzyme inhibitors, and peroxisome proliferator-activated receptor ligands derives from changes in inflammatory parameters. These advances in basic and clinical science have placed us on a threshold of a new era in cardiovascular medicine. [ABSTRACT FROM AUTHOR]
- Published
- 2001
- Full Text
- View/download PDF
37. PPARalpha activators inhibit tissue factor expression and activity in human monocytes.
- Author
-
Marx, N, Mackman, N, Schönbeck, U, Yilmaz, N, Hombach, V, Libby, P, and Plutzky, J
- Published
- 2001
38. Emerging concepts in metabolic abnormalities associated with coronary artery disease.
- Author
-
Plutzky, Jorge and Plutzky, J
- Published
- 2000
- Full Text
- View/download PDF
39. Atherosclerotic plaque rupture: emerging insights and opportunities.
- Author
-
Plutzky, Jorge and Plutzky, J
- Subjects
- *
ATHEROSCLEROTIC plaque , *CORONARY heart disease risk factors , *THROMBOSIS - Abstract
Reduction in acute,coronary events requires interventions that affect the mechanisms leading to formation of atherosclerotic lesions, as well as the molecular events that precipitate acute myocardial infarction. Data from clinical trials indicate that it is the vulnerability of atherosclerotic plaque to rupture, rather than the degree of atherosclerosis, that is the primary determinant of thrombosis-mediated acute coronary events. The characteristics of a plaque that is vulnerable to rupture include a thin fibrous cap separating the circulation from procoagulants in the plaque's lipid core; increased numbers of inflammatory cells (e.g., macrophages and T cells); and a relative paucity of vascular smooth muscle cells (VSMC). Plaque stability reflects various dynamic factors: interaction of inflammatory cells, VSMC production of the extracellular matrix that is the bulwark of the fibrous cap, inhibition of this process by certain cytokines, and increased degradation of the matrix by matrix metalloproteinases. There is growing interest in the concept that intervention in the inflammatory processes of atherogenesis might reduce lesion formation and/or progression. There has also been substantial progress in understanding the transcriptional regulation of proteins that are critically involved in atherogenesis. Recently, peroxisomal proliferator-activated receptors (PPARs) have been identified as a potential link between insulin resistance and atherosclerosis. This concept is supported by the discovery through drug screening of thiazolidinediones (troglitazone, rosiglitazone), compounds that are not only ligands for PPARgamma, a nuclear receptor involved in adipogenesis, but also are antidiabetic agents. [ABSTRACT FROM AUTHOR]
- Published
- 1999
- Full Text
- View/download PDF
40. Evolution and organization of the human protein C gene.
- Author
-
Plutzky, J, Hoskins, J A, Long, G L, and Crabtree, G R
- Abstract
We have isolated overlapping phage genomic clones covering an area of 21 kilobases that encodes the human protein C gene. The gene is at least 11.2 kilobases long and is made up of nine exons and eight introns. Two regions homologous to epidermal growth factor and transforming growth factor are encoded by amino acids 46-91 and 92-136 and are precisely delimited by introns, as is a similar sequence in the genes for coagulation factor IX and tissue plasminogen activator. When homologous amino acids of factor IX and protein C are aligned, the positions of all eight introns correspond precisely, suggesting that these genes are the product of a relatively recent gene duplication. Nevertheless, the two genes are sufficiently distantly related that no nucleic acid homology remains in the intronic regions and that the size of the introns varies dramatically between the two genes. The similarity of the genes for factor IX and protein C suggests that they may be the most closely related members of the serine protease gene family involved in coagulation and fibrinolysis.
- Published
- 1986
- Full Text
- View/download PDF
41. Statins for stroke: the second story?
- Author
-
Plutzky, J and Ridker, P M
- Published
- 2001
- Full Text
- View/download PDF
42. Diabetes and Cardiovascular Disease in Older Adults: Current Status and Future Directions
- Author
-
Schmader, K. E., Musi, N., Huang, E. S., Harkless, L., Goldberg, A., Halter, J. B., Kirkman, M. S., McFarland Horne, F., Zieman, S., Crandall, J. P., Plutzky, J., Hazzard, W. R., and High, K. P.
- Subjects
3. Good health - Abstract
The prevalence of diabetes increases with age, driven in part by an absolute increase in incidence among adults aged 65 years and older. Individuals with diabetes are at higher risk for cardiovascular disease, and age strongly predicts cardiovascular complications. Inflammation and oxidative stress appear to play some role in the mechanisms underlying aging, diabetes, cardiovascular disease, and other complications of diabetes. However, the mechanisms underlying the age-associated increase in risk for diabetes and diabetes-related cardiovascular disease remain poorly understood. Moreover, because of the heterogeneity of the older population, a lack of understanding of the biology of aging, and inadequate study of the effects of treatments on traditional complications and geriatric conditions associated with diabetes, no consensus exists on the optimal interventions for older diabetic adults. The Association of Specialty Professors, along with the National Institute on Aging, the National Institute of Diabetes and Digestive and Kidney Diseases, the National Heart, Lung, and Blood Institute, and the American Diabetes Association, held a workshop, summarized in this Perspective, to discuss current knowledge regarding diabetes and cardiovascular disease in older adults, identify gaps, and propose questions to guide future research.
43. Residual macrovascular risk in 2013: what have we learned?
- Author
-
Fruchart, Jc, Davignon, J, Hermans, Mp, Al Rubeaan, K, Amarenco, P, Assmann, G, Barter, P, Betteridge, J, Bruckert, E, Cuevas, A, Farnier, M, Ferrannini, Eleuterio, Fioretto, P, Genest, J, Ginsberg, Hn, Gotto AM Jr, Hu, D, Kadowaki, T, Kodama, T, Krempf, M, Matsuzawa, Y, Núñez Cortés JM, Monfil, Cc, Ogawa, H, Plutzky, J, Rader, Dj, Sadikot, S, Santos, Rd, Shlyakhto, E, Sritara, P, Sy, R, Tall, A, Tan, Ce, Tokgözoğlu, L, Toth, Pp, Valensi, P, Wanner, C, Zambon, A, Zhu, J, Zimmet, P, Residual Risk Reduction Initiative, UCL - SSS/IREC/EDIN - Pôle d'endocrinologie, diabète et nutrition, UCL - (SLuc) Service d'endocrinologie et de nutrition, Residual Risk Reduction Initiative (R3i), and Kardiyoloji
- Subjects
Cardiovascular system--Diseases ,medicine.medical_specialty ,Statin ,Epidemiology ,medicine.drug_class ,Endocrinology, Diabetes and Metabolism ,Type 2 diabetes ,Disease ,Review ,Ezetimibe ,Risk Factors ,Internal medicine ,Diabetes mellitus ,medicine ,Non-insulin-dependent diabetes ,Animals ,Humans ,Learning ,ddc:610 ,Intensive care medicine ,Atherogenic dyslipidaemia ,Dyslipidemias ,business.industry ,medicine.disease ,Residual risk ,Endocrinology ,Diabetes Mellitus, Type 2 ,Cardiovascular Diseases ,Residual cardiovascular risk ,Therapeutic options ,Medicine ,lipids (amino acids, peptides, and proteins) ,Metabolic syndrome ,business ,Cardiology and Cardiovascular Medicine ,Niacin ,medicine.drug - Abstract
Cardiovascular disease poses a major challenge for the 21st century, exacerbated by the pandemics of obesity, metabolic syndrome and type 2 diabetes. While best standards of care, including high-dose statins, can ameliorate the risk of vascular complications, patients remain at high risk of cardiovascular events. The Residual Risk Reduction Initiative (R(3)i) has previously highlighted atherogenic dyslipidaemia, defined as the imbalance between proatherogenic triglyceride-rich apolipoprotein B-containing-lipoproteins and antiatherogenic apolipoprotein A-I-lipoproteins (as in high-density lipoprotein, HDL), as an important modifiable contributor to lipid-related residual cardiovascular risk, especially in insulin-resistant conditions. As part of its mission to improve awareness and clinical management of atherogenic dyslipidaemia, the R(3)i has identified three key priorities for action: i) to improve recognition of atherogenic dyslipidaemia in patients at high cardiometabolic risk with or without diabetes; ii) to improve implementation and adherence to guideline-based therapies; and iii) to improve therapeutic strategies for managing atherogenic dyslipidaemia. The R(3)i believes that monitoring of non-HDL cholesterol provides a simple, practical tool for treatment decisions regarding the management of lipid-related residual cardiovascular risk. Addition of a fibrate, niacin (North and South America), omega-3 fatty acids or ezetimibe are all options for combination with a statin to further reduce non-HDL cholesterol, although lacking in hard evidence for cardiovascular outcome benefits. Several emerging treatments may offer promise. These include the next generation peroxisome proliferator-activated receptor alpha agonists, cholesteryl ester transfer protein inhibitors and monoclonal antibody therapy targeting proprotein convertase subtilisin/kexin type 9. However, long-term outcomes and safety data are clearly needed. In conclusion, the R(3)i believes that ongoing trials with these novel treatments may help to define the optimal management of atherogenic dyslipidaemia to reduce the clinical and socioeconomic burden of residual cardiovascular risk.
- Full Text
- View/download PDF
44. The peroxisome proliferator-activated receptor-gamma agonist pioglitazone represses inflammation in a peroxisome proliferator-activated receptor-alpha-dependent manner in vitro and in vivo in mice.
- Author
-
Orasanu G, Ziouzenkova O, Devchand PR, Nehra V, Hamdy O, Horton ES, Plutzky J, Orasanu, Gabriela, Ziouzenkova, Ouliana, Devchand, Pallavi R, Nehra, Vedika, Hamdy, Osama, Horton, Edward S, and Plutzky, Jorge
- Abstract
Objectives: Our aim was to investigate if the peroxisome proliferator-activated receptor (PPAR)-gamma agonist pioglitazone modulates inflammation through PPARalpha mechanisms.Background: The thiazolidinediones (TZDs) pioglitazone and rosiglitazone are insulin-sensitizing PPARgamma agonists used to treat type 2 diabetes (T2DM). Despite evidence for TZDs limiting inflammation and atherosclerosis, questions exist regarding differential responses to TZDs. In a double-blinded, placebo-controlled 16-week trial among recently diagnosed T2DM subjects (n = 34), pioglitazone-treated subjects manifested lower triglycerides and lacked the increase in soluble vascular cell adhesion molecules (sVCAM)-1 evident in the placebo group. Previously we reported PPARalpha but not PPARgamma agonists could repress VCAM-1 expression. Since both triglyceride-lowering and VCAM-1 repression characterize PPARalpha activation, we studied pioglitazone's effects via PPARalpha.Methods: Pioglitazone effects on known PPARalpha responses--ligand binding domain activation and PPARalpha target gene expression--were tested in vitro and in vivo, including in wild-type and PPARalpha-deficient cells and mice, and compared with the effects of other PPARgamma (rosiglitazone) and PPARalpha (WY14643) agonists.Results: Pioglitazone repressed endothelial TNFalpha-induced VCAM-1 messenger ribonucleic acid expression and promoter activity, and induced hepatic IkappaBalpha in a manner dependent on both pioglitazone exposure and PPARalpha expression. Pioglitazone also activated the PPARalpha ligand binding domain and induced PPARalpha target gene expression, with in vitro effects that were most pronounced in endothelial cells. In vivo, pioglitazone administration modulated sVCAM-1 levels and IkappaBalpha expression in wild-type but not PPARalpha-deficient mice.Conclusions: Pioglitazone regulates inflammatory target genes in hepatic (IkappaBalpha) and endothelial (VCAM-1) settings in a PPARalpha-dependent manner. These data offer novel mechanisms that may underlie distinct TZD responses. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
45. Semaglutide Effects on Cardiovascular Outcomes in People With Overweight or Obesity (SELECT): Outcomes by Sex.
- Author
-
Verma S, Colhoun HM, Dicker D, Hovingh GK, Kahn SE, Kautzky-Willer A, Lingvay I, Plutzky J, Rasmussen S, Rathor N, Hoff ST, and Lincoff AM
- Abstract
Competing Interests: Funding Support and Author Disclosures This study was funded by Novo Nordisk A/S. Dr Verma has received research grants and/or speaking honoraria from Amarin, Amgen, AstraZeneca, Bayer, Boehringer Ingelheim, Canadian Medical and Surgical Knowledge Translation Research Group, Eli Lilly, HLS Therapeutics, Janssen, Novartis, Novo Nordisk, Pfizer, PhaseBio, S & L Solutions Event Management Inc, and Sanofi. Dr Colhoun holds the AXA Research Fund endowed Chair in Medical Informatics and Epidemiology at the University of Edinburgh. Dr Dicker has received consulting honoraria from Novo Nordisk, Eli Lilly, Boehringer Ingelheim, and AstraZeneca; and has received research grants through his affiliation from Novo Nordisk, Eli Lilly, Boehringer Ingelheim, and Rhythm. Dr Hovingh is an employee of and stakeholder in Novo Nordisk. Dr Kahn has received advisory board/consulting fees from AltPep, Bayer, Boehringer Ingelheim, Casma Therapeutics, Eli Lilly, Intarcia, Merck, Novo Nordisk, Oramed, Pfizer, and Third Rock Ventures. Dr Kautzky-Willer has received speaker honoraria from Boehringer Ingelheim, Novartis, Novo Nordisk, and Merck. Dr Lingvay has received research grants from Boehringer Ingelheim, Merck, Mylan Pharmaceuticals Inc, Novo Nordisk, Pfizer, and Sanofi U.S. Services Inc; has served as a consultant for AstraZeneca, Bayer Healthcare Pharmaceuticals Inc, Biomea, Boehringer Ingelheim, Carmot, Eli Lilly and Company, Intarcia, Intercept Pharmaceuticals, Inc, Janssen Global Services, LLC, Johnson & Johnson Medical Devices & Diagnostics Group–Latin America, LLC, MannKind Corporation, Merck, Novo Nordisk, Pfizer Inc, Sanofi U.S. Services Inc, Shionogi Inc, Structure Therapeutics, Target Pharma, Valeritas, Inc, and Zealand Pharma; and has received travel expenses from Boehringer Ingelheim, Eli Lilly and Company, Johnson & Johnson Medical Devices & Diagnostics Group–Latin America, LLC, Novo Nordisk, Sanofi U.S. Services Inc, and Zealand Pharma A/S. Dr Plutzky has received consulting honoraria from Altimmune, Amgen, Esperion Therapeutics, Inc, Merck, MJH Life Sciences, Novartis, and Novo Nordisk; has received a grant, paid to his institution, from Boehringer Ingelheim; and holds the position of Director, Preventive Cardiology, at Brigham and Women's Hospital. Drs Rasmussen, Rathor, and Hoff are employees of, and shareholder in, Novo Nordisk. Dr Lincoff has received research grants from AbbVie Inc, AstraZeneca, CSL Behring, Eli Lilly and Company, Esperion Therapeutics, Inc, and Novartis, paid to his institution; and has served as a consultant for Akebia Therapeutics Inc, Alnylam Pharmaceuticals Inc, Ardelyx, Eli Lilly and Company, FibroGen, GlaxoSmithKline, Intarcia, Medtronic Vascular, Inc, Novartis Pharmaceuticals Corporation, Novo Nordisk, Provention Bio, Entity, and ReCor Medical.
- Published
- 2024
- Full Text
- View/download PDF
46. DCRM 2.0: Multispecialty practice recommendations for the management of diabetes, cardiorenal, and metabolic diseases.
- Author
-
Handelsman Y, Anderson JE, Bakris GL, Ballantyne CM, Bhatt DL, Bloomgarden ZT, Bozkurt B, Budoff MJ, Butler J, Cherney DZI, DeFronzo RA, Del Prato S, Eckel RH, Filippatos G, Fonarow GC, Fonseca VA, Garvey WT, Giorgino F, Grant PJ, Green JB, Greene SJ, Groop PH, Grunberger G, Jastreboff AM, Jellinger PS, Khunti K, Klein S, Kosiborod MN, Kushner P, Leiter LA, Lepor NE, Mantzoros CS, Mathieu C, Mende CW, Michos ED, Morales J, Plutzky J, Pratley RE, Ray KK, Rossing P, Sattar N, Schwarz PEH, Standl E, Steg PG, Tokgözoğlu L, Tuomilehto J, Umpierrez GE, Valensi P, Weir MR, Wilding J, and Wright EE Jr
- Subjects
- Humans, Diabetes Mellitus, Type 2 therapy, Metabolic Diseases therapy
- Abstract
The spectrum of cardiorenal and metabolic diseases comprises many disorders, including obesity, type 2 diabetes (T2D), chronic kidney disease (CKD), atherosclerotic cardiovascular disease (ASCVD), heart failure (HF), dyslipidemias, hypertension, and associated comorbidities such as pulmonary diseases and metabolism dysfunction-associated steatotic liver disease and metabolism dysfunction-associated steatohepatitis (MASLD and MASH, respectively, formerly known as nonalcoholic fatty liver disease and nonalcoholic steatohepatitis [NAFLD and NASH]). Because cardiorenal and metabolic diseases share pathophysiologic pathways, two or more are often present in the same individual. Findings from recent outcome trials have demonstrated benefits of various treatments across a range of conditions, suggesting a need for practice recommendations that will guide clinicians to better manage complex conditions involving diabetes, cardiorenal, and/or metabolic (DCRM) diseases. To meet this need, we formed an international volunteer task force comprising leading cardiologists, nephrologists, endocrinologists, and primary care physicians to develop the DCRM 2.0 Practice Recommendations, an updated and expanded revision of a previously published multispecialty consensus on the comprehensive management of persons living with DCRM. The recommendations are presented as 22 separate graphics covering the essentials of management to improve general health, control cardiorenal risk factors, and manage cardiorenal and metabolic comorbidities, leading to improved patient outcomes., Competing Interests: Declaration of competing interest None of the Task Force members received monetary renumeration for their contributions to the creation or writing of this consensus document. See the Supplementary Appendix for full declarations of competing interests for each author., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
47. Semaglutide versus placebo in patients with heart failure and mildly reduced or preserved ejection fraction: a pooled analysis of the SELECT, FLOW, STEP-HFpEF, and STEP-HFpEF DM randomised trials.
- Author
-
Kosiborod MN, Deanfield J, Pratley R, Borlaug BA, Butler J, Davies MJ, Emerson SS, Kahn SE, Kitzman DW, Lingvay I, Mahaffey KW, Petrie MC, Plutzky J, Rasmussen S, Rönnbäck C, Shah SJ, Verma S, Weeke PE, and Lincoff AM
- Subjects
- Aged, Female, Humans, Male, Middle Aged, Diabetes Mellitus, Type 2 drug therapy, Diabetes Mellitus, Type 2 complications, Hospitalization statistics & numerical data, Obesity drug therapy, Treatment Outcome, Glucagon-Like Peptides therapeutic use, Heart Failure drug therapy, Heart Failure mortality, Stroke Volume, Glucagon-Like Peptide-1 Receptor Agonists therapeutic use
- Abstract
Background: Heart failure with mildly reduced or preserved ejection fraction (hereafter referred to as HFpEF) is the most common type of heart failure and is associated with a high risk of hospitalisation and death, especially in patients with overweight, obesity, or type 2 diabetes. In the STEP-HFpEF and STEP-HFpEF DM trials, semaglutide improved heart failure-related symptoms and physical limitations in participants with HFpEF. Whether semaglutide also reduces clinical heart failure events in this group remains to be established., Methods: We conducted a post-hoc pooled, participant-level analysis of four randomised, placebo-controlled trials (SELECT, FLOW, STEP-HFpEF, and STEP-HFpEF DM) to examine the effects of once-weekly subcutaneous semaglutide (2·4 mg in SELECT, STEP-HFpEF, and STEP-HFpEF DM; 1·0 mg in FLOW) on heart failure events. The STEP-HFpEF and STEP-HFpF DM trials enrolled participants with obesity-related HFpEF, the SELECT trial enrolled participants with atherosclerotic cardiovascular disease and overweight or obesity, and the FLOW trial enrolled participants with type 2 diabetes and chronic kidney disease. Hence, for this analysis, we include all participants from the STEP-HFpEF trials and those with an investigator-reported history of HFpEF from SELECT and FLOW. The main outcomes for this analysis were the composite endpoint of time to cardiovascular death or first worsening heart failure event (defined as hospitalisation or urgent visit due to heart failure), time to first worsening heart failure event, and time to cardiovascular death. Efficacy and safety endpoints were analysed with the full analysis set (ie, all participants randomly assigned to treatment, according to the intention-to-treat principle). The SELECT, FLOW, STEP-HFpEF, and STEP-HFpEF DM trials are registered at ClinicalTrials.gov, NCT03574597, NCT03819153, NCT04788511, and NCT04916470, respectively, and all are complete., Findings: Across the four trials, 3743 (16·8%) of 22 282 participants had a history of HFpEF (1914 assigned to semaglutide and 1829 assigned to placebo). In this group of participants with HFpEF, semaglutide reduced the risk of the combined endpoint of cardiovascular death or heart failure events (103 [5·4%] of 1914 in the semaglutide group had events vs 138 [7·5%] of 1829 in the placebo group; hazard ratio [HR] 0·69 [95% CI 0·53-0·89]; p=0·0045). Semaglutide also reduced the risk of worsening heart failure events (54 [2·8%] vs 86 [4·7%]; HR 0·59 [0·41-0·82]; p=0·0019). No significant effect on cardiovascular death alone was seen (59 [3·1%] vs 67 [3·7%]; HR 0·82 [0·57-1·16]; p=0·25). A lower proportion of patients treated with semaglutide had serious adverse events than did those who were treated with placebo (572 [29·9%] vs 708 [38·7%])., Interpretation: In patients with HFpEF, semaglutide reduced the risk of the combined endpoint of cardiovascular death or worsening heart failure events, and worsening heart failure events alone, whereas its effect on cardiovascular death alone was not significant. These data support the use of semaglutide as an efficacious therapy to reduce the risk of clinical heart failure events in patients with HFpEF, for whom few treatment options are currently available., Funding: Novo Nordisk., Competing Interests: Declaration of interests MNK has served as a consultant for 35Pharma, Alnylam, Amgen, AstraZeneca, Bayer, Boehringer Ingelheim, Cytokinetics, Corcept Therapeutics, Dexcom, Eli Lilly, Esperion Therapeutics, Imbria Pharmaceuticals, Janssen, Lexicon Pharmaceuticals, Merck (Diabetes and Cardiovascular), Novo Nordisk, Pharmacosmos, Pfizer, Regeneron, scPharmaceuticals, Structure Therapeutics, Vifor Pharma, and Youngene Therapeutics; has served on a data monitoring board for Applied Therapeutics; has served on an advisory board for Alnylam, Amgen, Arrowhead Pharmaceuticals, AstraZeneca, Bayer, Boehringer Ingelheim, Corcept Therapeutics, Eli Lilly, Esperion Therapeutics, Merck (Diabetes and Cardiovascular), Novo Nordisk, and Sanofi; has received research grants from AstraZeneca, Boehringer Ingelheim, and Pfizer; holds stocks in Artera Health and Saghmos Therapeutics; has received honoraria from AstraZeneca, Boehringer Ingelheim, and Novo Nordisk; and has received support for data analyses from AstraZeneca and Vifor. JD has received consulting honoraria from Aegerion, Amgen, AstraZeneca, Bayer, Boehringer Ingelheim, Merck, Novartis, Novo Nordisk, Pfizer, Sanofi, and Takeda and research grants from Aegerion, British Heart Foundation, Colgate, UK Medical Research Council, MSD, National Institute for Health and Care Research, Pfizer, Public Health England, and Roche. RP reports speaker fees from Eli Lilly, Lilly USA, and Novo Nordisk; consulting fees from AbbVie, AstraZeneca, Bayer, Bayer HealthCare Pharmaceuticals, Corcept Therapeutics, Dexcom, Endogenex, Gasherbrum Bio, Genprex, Getz Pharma, Hanmi Pharmaceutical, Hengrui (USA), Intas Pharmaceuticals, Eli Lilly, Lilly USA, Merck, Novo Nordisk, Pfizer, Rivus Pharmaceuticals, Scholar Rock, and Sun Pharmaceutical Industries; and grants from Biomea Fusion, Carmot Therapeutics, Dompe, Endogenex, Fractyl, Eli Lilly, Novo Nordisk, and Sanofi. All grants are directed to RP's institution. Up to Dec 31, 2023, payment for RP's consultant and speaker services was made directly to his employer, AdventHealth, a nonprofit entity; as of Jan 1, 2024, payment was directed to RP personally. BAB receives research support from the US National Institutes of Health and the US Department of Defense, as well as research grant funding from AstraZeneca, Axon Therapies, GlaxoSmithKline, Medtronic, Mesoblast, Novo Nordisk, Rivus, and Tenax Therapeutics; he has served as a consultant for Actelion, Amgen, Aria, Axon Therapies, BD, Boehringer Ingelheim, Cytokinetics, Edwards Lifesciences, Eli Lilly, Imbria, Janssen, Merck, NGM, Novo Nordisk, NXT, and VADovations; and is named inventor (US patent number 10,307,179) for the tools and approach for a minimally invasive pericardial modification procedure to treat heart failure. JB is a consultant to Abbott, American Regent, Amgen, Applied Therapeutics, AskBio, Astellas, AstraZeneca, Bayer, Boehringer Ingelheim, Boston Scientific, Bristol Myers Squibb, Cardiac Dimension, CardioCell, Cardior, CSL Behring, CVRx, Cytokinetics, Daxor, Edwards Lifesciences, Element Science, Faraday, Foundry, G3P, Imbria, Impulse Dynamics, Innolife, Inventiva, Ionis, Levator, Lexicon, Lilly, LivaNova, Janssen, Medtronics, Merck, Occlutech, Owkin, Novartis, Novo Nordisk, Pfizer, Pharmacosmos, PharmaIN, Prolaio, Pulnovo, Regeneron, Renibus, Roche, Salamandra, Salubris, Sanofi, scPharmaceuticals, Secretome, Sequana, SQ Innovation, Tenex, Tricog, Ultromics, Vifor, and Zoll. MJD has acted as a consultant, advisory board member, and speaker for Boehringer Ingelheim, Eli Lilly, Novo Nordisk, and Sanofi; an advisory board member for AstraZeneca, Carmot/Roche, Medtronic, Pfizer, and Zealand Pharma; and a speaker for Amgen and AstraZeneca; and has received grants from AstraZeneca, Boehringer Ingelheim, Eli Lilly, Janssen, Novo Nordisk, and Sanofi-Aventis. SSE has received consulting fees from Amylyx, AstraZeneca, Avillion, Ayala, Bayer, BeiGene, Boehringer Ingelheim, 89 Bio, BioAge, BioAtla, Bristol Meyer Squibb, BridgeBio, Daiichi Sankyo, Denovo, Fore Therapeutics, GlaxoSmithKline, Inovio, Insmed, Ipsen, Karuna, Lilly, Lundbeck, Mirati, Moderna, Novartis, Novavax, Novo Nordisk, National Surgical Adjuvant Breast and Bowel Project, Pfizer, Principia, Reata, Rebiotx, Roche, Sanofi, solvd, Sutro Biopharma, and TG Therapeutics. SEK received consulting fees from Casma Therapeutics, Oramed, and Third Rock Ventures; and served on an advisory board for AltPep, Anji Pharmaceuticals, Bayer, Boehringer Ingelheim, Eli Lilly, Intarcia, Merck, and Novo Nordisk. He has received stock options from AltPep. DK was supported in part by the Kermit Glenn Phillips II Chair in Cardiovascular Medicine and US National Institutes of Health grants U01AG076928, R01AG078153, R01AG045551, R01AG18915, P30AG021332, U24AG059624, and U01HL160272; and received honoraria as a consultant for AstraZeneca, Bayer, Boehringer Ingelheim, Corvia Medical, Ketyo, Novartis, Novo Nordisk, Pfizer, and Rivus; grant funding from AstraZeneca, Bayer, Novartis, Novo Nordisk, Pfizer, and Rivus; and has stock ownership in Gilead Sciences. IL has received research grants from Boehringer Ingelheim, Merck, Mylan Pharmaceuticals, Novo Nordisk, Pfizer, and Sanofi; served as a consultant for AstraZeneca, Bayer Healthcare Pharmaceuticals, Biomea, Boehringer Ingelheim, Carmot, Eli Lilly, Intarcia, Intercept Pharmaceuticals, Janssen Global Services, Johnson & Johnson Medical Devices & Diagnostics Group—Latin America, MannKind, Merck, Novo Nordisk, Pfizer, Sanofi, Shionogi, Structure Therapeutics, Target Pharma, Valeritas, and Zealand Pharma; and received travel expenses from Boehringer Ingelheim, Eli Lilly, Johnson & Johnson Medical Devices & Diagnostics Group—Latin America, Novo Nordisk, Sanofi, and Zealand Pharma. KWM has received grants from AHA, Apple, Bayer, California Institute for Regenerative Medicine, CSL Behring, Eidos, Ferring, Gilead, Google, Idorsia, Johnson & Johnson, Luitpold, Novartis, PAC-12, Precordior, and Sanifit; and consulting fees from Applied Therapeutics, Bayer, BMS, BridgeBio, CSL Behring, Elsevier, Fosun Pharma, Human, Johnson & Johnson, Moderna, Myokardia, Novartis, Novo Nordisk, Otsuka, Phasebio, Portola, Quidel, and Theravance; and holds stock options for Human, Medeloop, Precordior, and Regencor. MCP was supported by the British Heart Foundation Centre of Research Excellence Grant RE/18/6/34217; has received research funding from AstraZeneca, Boehringer Ingelheim, Boston Scientific, Medtronic, Novartis, Novo Nordisk, Pharmacosmos, Roche, and SQ Innovations; and served on committees or consulted for AbbVie, Akero, AnaCardio, Applied Therapeutics, AstraZeneca, Bayer, Biosensors, Boehringer Ingelheim, Cardiorentis, Corvia, Eli Lilly, Horizon Therapeutics, LIB Therapeutics, Moderna, New Amsterdam, Novartis, Novo Nordisk, Pharmacosmos, Siemens, SQ Innovations, Takeda, Teikoku, and Vifor. JP has received consulting fees from Altimmune, Amgen, Esperion Therapeutics, Merck, MJH Life Sciences, Novartis, and Novo Nordisk and received a grant, paid to his institution, from Boehringer Ingelheim. CR, SR, and PEW are employees and SR and PEW are shareholders of Novo Nordisk. SJS has received research grants from AstraZeneca, Corvia, and Pfizer; and consulting fees from Abbott, Alleviant, Amgen, Aria CV, AstraZeneca, Axon Therapies, Bayer, Boehringer Ingelheim, Boston Scientific, Bristol Myers Squibb, Cyclerion, Cytokinetics, Edwards Lifesciences, Eidos, Imara, Impulse Dynamics, Intellia, Ionis, Lilly, Merck, MyoKardia, Novartis, Novo Nordisk, Pfizer, Prothena, ReCor, Regeneron, Rivus, Sardocor, Shifamed, Tenax, Tenaya, and Ultromics. SV reports speaking honoraria or consulting fees from Abbott, Amarin, AstraZeneca, Bayer, Boehringer Ingelheim, Canadian Medical and Surgical Knowledge Translation Research Group, Eli Lilly, HLS Therapeutics, Janssen, Merck, Novartis, Novo Nordisk, Pfizer, PhaseBio, and TIMI. AML has received research grants from AbbVie, AstraZeneca, CSL Behring, Eli Lilly, Esperion Therapeutics, and Novartis, paid to his institution; and consulting fees from Akebia Therapeutics, Alnylam Pharmaceuticals, Ardelyx, Eli Lilly, FibroGen, GlaxoSmithKline, Intarcia, Medtronic Vascular, Novartis, Novo Nordisk, Provention Bio, Amgen, and ReCor Medical. All other authors declare no competing interests., (Copyright © 2024 Elsevier Ltd. All rights reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
48. Semaglutide and cardiovascular outcomes in patients with obesity and prevalent heart failure: a prespecified analysis of the SELECT trial.
- Author
-
Deanfield J, Verma S, Scirica BM, Kahn SE, Emerson SS, Ryan D, Lingvay I, Colhoun HM, Plutzky J, Kosiborod MN, Hovingh GK, Hardt-Lindberg S, Frenkel O, Weeke PE, Rasmussen S, Goudev A, Lang CC, Urina-Triana M, Pietilä M, and Lincoff AM
- Subjects
- Humans, Male, Female, Middle Aged, Double-Blind Method, Aged, Stroke Volume drug effects, Treatment Outcome, Cardiovascular Diseases mortality, Cardiovascular Diseases prevention & control, Hypoglycemic Agents therapeutic use, Hypoglycemic Agents adverse effects, Injections, Subcutaneous, Glucagon-Like Peptides therapeutic use, Glucagon-Like Peptides administration & dosage, Glucagon-Like Peptides adverse effects, Heart Failure mortality, Heart Failure drug therapy, Obesity complications, Obesity drug therapy
- Abstract
Background: Semaglutide, a GLP-1 receptor agonist, reduces the risk of major adverse cardiovascular events (MACE) in people with overweight or obesity, but the effects of this drug on outcomes in patients with atherosclerotic cardiovascular disease and heart failure are unknown. We report a prespecified analysis of the effect of once-weekly subcutaneous semaglutide 2·4 mg on ischaemic and heart failure cardiovascular outcomes. We aimed to investigate if semaglutide was beneficial in patients with atherosclerotic cardiovascular disease with a history of heart failure compared with placebo; if there was a difference in outcome in patients designated as having heart failure with preserved ejection fraction compared with heart failure with reduced ejection fraction; and if the efficacy and safety of semaglutide in patients with heart failure was related to baseline characteristics or subtype of heart failure., Methods: The SELECT trial was a randomised, double-blind, multicentre, placebo-controlled, event-driven phase 3 trial in 41 countries. Adults aged 45 years and older, with a BMI of 27 kg/m
2 or greater and established cardiovascular disease were eligible for the study. Patients were randomly assigned (1:1) with a block size of four using an interactive web response system in a double-blind manner to escalating doses of once-weekly subcutaneous semaglutide over 16 weeks to a target dose of 2·4 mg, or placebo. In a prespecified analysis, we examined the effect of semaglutide compared with placebo in patients with and without a history of heart failure at enrolment, subclassified as heart failure with preserved ejection fraction, heart failure with reduced ejection fraction, or unclassified heart failure. Endpoints comprised MACE (a composite of non-fatal myocardial infarction, non-fatal stroke, and cardiovascular death); a composite heart failure outcome (cardiovascular death or hospitalisation or urgent hospital visit for heart failure); cardiovascular death; and all-cause death. The study is registered with ClinicalTrials.gov, NCT03574597., Findings: Between Oct 31, 2018, and March 31, 2021, 17 604 patients with a mean age of 61·6 years (SD 8·9) and a mean BMI of 33·4 kg/m2 (5·0) were randomly assigned to receive semaglutide (8803 [50·0%] patients) or placebo (8801 [50·0%] patients). 4286 (24·3%) of 17 604 patients had a history of investigator-defined heart failure at enrolment: 2273 (53·0%) of 4286 patients had heart failure with preserved ejection fraction, 1347 (31·4%) had heart failure with reduced ejection fraction, and 666 (15·5%) had unclassified heart failure. Baseline characteristics were similar between patients with and without heart failure. Patients with heart failure had a higher incidence of clinical events. Semaglutide improved all outcome measures in patients with heart failure at random assignment compared with those without heart failure (hazard ratio [HR] 0·72, 95% CI 0·60-0·87 for MACE; 0·79, 0·64-0·98 for the heart failure composite endpoint; 0·76, 0·59-0·97 for cardiovascular death; and 0·81, 0·66-1·00 for all-cause death; all pinteraction >0·19). Treatment with semaglutide resulted in improved outcomes in both the heart failure with reduced ejection fraction (HR 0·65, 95% CI 0·49-0·87 for MACE; 0·79, 0·58-1·08 for the composite heart failure endpoint) and heart failure with preserved ejection fraction groups (0·69, 0·51-0·91 for MACE; 0·75, 0·52-1·07 for the composite heart failure endpoint), although patients with heart failure with reduced ejection fraction had higher absolute event rates than those with heart failure with preserved ejection fraction. For MACE and the heart failure composite, there were no significant differences in benefits across baseline age, sex, BMI, New York Heart Association status, and diuretic use. Serious adverse events were less frequent with semaglutide versus placebo, regardless of heart failure subtype., Interpretation: In patients with atherosclerotic cardiovascular diease and overweight or obesity, treatment with semaglutide 2·4 mg reduced MACE and composite heart failure endpoints compared with placebo in those with and without clinical heart failure, regardless of heart failure subtype. Our findings could facilitate prescribing and result in improved clinical outcomes for this patient group., Funding: Novo Nordisk., Competing Interests: Declaration of interests JD declares having received consulting honoraria from Amgen, Boehringer Ingelheim, Merck, Pfizer, Aegerion, Novartis, Sanofi, Takeda, Novo Nordisk, and Bayer, and research grants from British Heart Foundation, Medical Research Council (UK), National Institute for Health and Care Research, Public Health England, MSD, Pfizer, Aegerion, Colgate, and Roche. BMS declares having received institutional research grants to Brigham and Women's Hospital from Better Therapeutics, Merck, Novo Nordisk, and Pfizer, and consulting fees from Allergan, Boehringer Ingelheim, Better Therapeutics, Elsevier PracticeUpdate Cardiology, Esperion, Hanmi, Lexicon, Novo Nordisk, and equity in health at Scale and Doximity. SV reports speaking honoraria or consulting fees from Abbott, Amarin, AstraZeneca, Bayer, Boehringer Ingelheim, Canadian Medical and Surgical Knowledge Translation Research Group, Eli Lilly, HLS Therapeutics, Janssen, Merck, Novartis, Novo Nordisk, Pfizer, PhaseBio, and TIMI. DR declares having received consulting honoraria from Altimmune, Amgen, Biohaven, Boehringer Ingelheim, Calibrate, Carmot Therapeutics, CinRx, Eli Lilly, Epitomee, Gila Therapeutics, Ifa Celtic, Novo Nordisk, Pfizer, Rhythm, Scientific Intake, Wondr Health, and Zealand. DR also declares having received stock options from Calibrate, Epitomee, Scientific Intake, and Xeno Bioscience. IL declares having received research funding (paid to institution) from Novo Nordisk, Sanofi, Mylan, and Boehringer Ingelheim. IL received advisory or consulting fees or other support from Altimmune, AstraZeneca, Bayer, Biomea, Boehringer Ingelheim, Carmot, Cytoki Pharma, Eli Lilly, Intercept, Janssen/Johnson & Johnson, Mannkind, Mediflix, Merck, Metsera, Novo Nordisk, Pharmaventures, Pfizer, Regeneron, Sanofi, Shionogi, Structure Therapeutics, Target RWE, Terns Pharma, The Comm Group, Valeritas, WebMD, and Zealand Pharma. HMC declares being a stockholder and serving on an advisory panel for Bayer; receiving research grants from Chief Scientist Office, Diabetes UK, European Commission, IQVIA, Juvenile Diabetes Research Foundation, and Medical Research Council; serving on an advisory board and speakers bureau for Novo Nordisk; and holding stock in Roche Pharmaceuticals. SEK declares having received consulting honoraria from Anii Pharmaceuticals, Boehringer Ingelheim, Eli Lilly, Merck, Novo Nordisk, and Oramed and stock options from Altpep. JP declares having received consulting honoraria from Altimmune, Amgen, Esperion Therapeutics, Merck, MJH Life Sciences, Novartis, and Novo Nordisk; he has received a grant, paid to his institution, from Boehringer Ingelheim, and holds the position of Director, Preventive Cardiology, at Brigham and Women's Hospital. MNK declares having served as a consultant or on an advisory board for 35Pharma, Alnylam, Amgen, Applied Therapeutics, AstraZeneca, Bayer, Boehringer Ingelheim, Cytokinetics, Dexcom, Eli Lilly, Esperion Therapeutics, Imbria Pharmaceuticals, Janssen, Lexicon Pharmaceuticals, Merck (Diabetes and Cardiovascular), Novo Nordisk, Pharmacosmos, Pfizer, Sanofi, scPharmaceuticals, Structure Therapeutics, Vifor Pharma, and Youngene Therapeutics; has received research grants from AstraZeneca, Boehringer Ingelheim, and Pfizer; holds stocks in Artera Health and Saghmos Therapeutics; and has received honoraria from AstraZeneca, Boehringer Ingelheim, and Novo Nordisk. MNK has also received other research support from AstraZeneca. SSE declares having received consulting honoraria from Amylyx, AstraZeneca, Avillion, Ayala, Bayer, BeiGene, Boehringer Ingelheim, 89 Bio, BioAge, BioAtla, Bristol Myers Squibb, BridgeBio, Daiichi Sankyo, Denovo, Fore Therapeutics, GlaxoSmithKline, Inovio, Insmed, Ipsen, Karuna, Lilly, Lundbeck, Mirati, Moderna, Novartis, Novavax, Novo Nordisk, National Surgical Adjuvant Breast and Bowel Project, Pfizer, Principia, Reata, Rebiotx, Roche, Sanofi, Solvd, Sutro Biopharma, and TG Therapeutics. AG declares having received honoraria from Novo Nordisk, AstraZeneca, Novartis, Boehringer Ingelheim, and Bayer. CCL declares having received consulting and research honoraria from Amarin, Applied Therapeutics, AstraZeneca, Boehringer Ingelheim, Bristol Myers Squibb, Moderna, Novartis, Novo Nordisk, Pfizer, and Roche Diagnostics. MU-T declares having received consulting and research honoraria from Abbott, AstraZeneca, Bayer, Boehringer Ingelheim, Bristol Myers Squibb, Frosst Laboratories, Johnson and Johnson, Menarini, Novartis, Novo Nordisk, Pfizer, Procaps, Sanofi-Aventis, Servier, and Tecnofarma. MP declares having received honoraria from Novo Nordisk. AML declares having received honoraria from Novo Nordisk, Eli Lilly, Akebia, Amgen, Ardelyx, Becton Dickson, Endologix, Fibrogen, GlaxoSmithkline, Medtronic, Neovasc, Provention Bio, ReCor, Brainstorm Cell, Alnylam, and Intarcia for consulting activities and research funding to his institution from AbbVie, Esperion, AstraZeneca, CSL Behring, Novartis, and Eli Lilly. All other authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
49. Semaglutide and Cardiovascular Outcomes by Baseline HbA1c and Change in HbA1c in People With Overweight or Obesity but Without Diabetes in SELECT.
- Author
-
Lingvay I, Deanfield J, Kahn SE, Weeke PE, Toplak H, Scirica BM, Rydén L, Rathor N, Plutzky J, Morales C, Lincoff AM, Lehrke M, Jeppesen OK, Gajos G, Colhoun HM, Cariou B, and Ryan D
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Cardiovascular Diseases prevention & control, Cardiovascular Diseases mortality, Hypoglycemic Agents therapeutic use, Glucagon-Like Peptides therapeutic use, Glycated Hemoglobin metabolism, Obesity complications, Obesity drug therapy, Overweight complications, Overweight drug therapy
- Abstract
Objective: To evaluate the cardiovascular effects of semaglutide by baseline glycated hemoglobin (HbA1c) and change in HbA1c in a prespecified analysis of Semaglutide Effects on Cardiovascular Outcomes in People With Overweight or Obesity (SELECT)., Research Design and Methods: In SELECT, people with overweight or obesity and atherosclerotic cardiovascular disease without diabetes were randomized to weekly semaglutide 2.4 mg or placebo. The primary end point of first major adverse cardiovascular event (MACE) (cardiovascular mortality, nonfatal myocardial infarction, or stroke) was reduced by 20% with semaglutide versus placebo. Analysis of outcomes included first MACE, its individual components, expanded MACE (cardiovascular mortality, nonfatal myocardial infarction, or stroke; coronary revascularization; or hospitalization for unstable angina), a heart failure composite (heart failure hospitalization or urgent medical visit or cardiovascular mortality), coronary revascularization, and all-cause mortality by baseline HbA1c subgroup and categories of HbA1c change (<-0.3, -0.3 to 0.3, and >0.3 percentage points) from baseline to 20 weeks using the intention-to-treat principle with Cox proportional hazards., Results: Among 17,604 participants (mean age 61.6 years, 72.3% male), baseline HbA1c was <5.7% for 33.5%, 5.7% to <6.0% for 34.6%, and 6.0% to <6.5% for 31.9%. Cardiovascular risk reduction with semaglutide versus placebo was not shown to be different across baseline HbA1c groups and was consistent with that of the overall study for all end points, except all-cause mortality. Cardiovascular outcomes were also consistent across subgroups of HbA1c change., Conclusions: In people with overweight or obesity and established atherosclerotic cardiovascular disease but not diabetes, semaglutide reduced cardiovascular events irrespective of baseline HbA1c or change in HbA1c. Thus, semaglutide is expected to confer cardiovascular benefits in people with established atherosclerotic cardiovascular disease who are normoglycemic at baseline and/or in those without HbA1c improvements., (© 2024 by the American Diabetes Association.)
- Published
- 2024
- Full Text
- View/download PDF
50. Effect of Pemafibrate on Diabetic Foot Ulceration and Gangrene: An Exploratory Analysis From PROMINENT.
- Author
-
Marinho LL, Everett BM, Aday AW, Visseren FLJ, MacFadyen JG, Zaharris E, Plutzky J, Santos RD, Libby P, Fruchart JC, Ridker PM, and Pradhan AD
- Subjects
- Humans, Butyrates pharmacology, Butyrates therapeutic use, Male, Female, Middle Aged, Treatment Outcome, Diabetic Foot drug therapy, Diabetic Foot complications, Gangrene etiology, Benzoxazoles therapeutic use
- Abstract
Competing Interests: Funding Support and Author Disclosures Dr Marinho is supported by the Lemann Foundation Cardiovascular Research Fellowship. Dr Everett has received grants from PCORI and Novo Nordisk; has received royalties from UpToDate; and has been a consultant for Circulation/American Heart Association, Eli Lilly and Company, U.S. Food and Drug Administration, Novo Nordisk, Gilead Pharmaceuticals, Ipsen Biosciences Inc, Johnson & Johnson, Provention Bio, and Roche Diagnostics. Dr Aday has received compensation for consultant services from Aeglea. Ms MacFadyen has received salary support from a research grant to BWH from Kowa Research Institute. Dr Santos has received honoraria related to consulting and/or speaker activities from Aché, Abbott, Amryt, Amgen, Biolab, EMS, Eli Lilly, Getz Pharma, Pfizer, PTC Therapeutics, Novartis, Novo Nordisk, and Sanofi/Regeneron; and has received research grants from Amgen, Esperion, Kowa, Novartis, and Sanofi/Regeneron. Dr Libby is an unpaid consultant to or involved in clinical trials for Amgen, AstraZeneca, Baim Institute, Beren Therapeutics, Esperion Therapeutics, Genentech, Kancera, Kowa, Medimmune, Merck, Moderna, Novo Nordisk, Novartis, Pfizer, and Sanofi-Regeneron; and is a member of the scientific advisory board for Amgen, Caristo Diagnostics, Cartesian Therapeutics, CSL Behring, DalCor Pharmaceuticals, Elucid Bioimaging, Kancera, Kowa Pharmaceuticals, Olatec Therapeutics, Medimmune, Novartis, PlaqueTec, TenSixteen Bio, Soley Therapeutics, and XBiotech, Inc. Dr Libby’s laboratory has received research funding in the last 2 years from Novartis, Novo Nordisk, and Genentech; is on the Board of Directors of XBiotech, Inc; and has a financial interest in Xbiotech (a company developing therapeutic human antibodies), in TenSixteen Bio (a company targeting somatic mosaicism and clonal hematopoiesis of indeterminate potential to discover and develop novel therapeutics to treat age-related diseases), and in Soley Therapeutics (a biotechnology company that is combining artificial intelligence with molecular and cellular response detection for discovering and developing new drugs, currently focusing on cancer therapeutics). Dr Fruchart is a consultant to Kowa. Dr Ridker has received institutional research grant support from Kowa Research Institute, Novartis, Amarin, Pfizer, Esperion, Novo Nordisk, and the National Heart, Lung, and Blood Institute; has served as a consultant to Novartis, Flame, Agepha, Ardelyx, AstraZeneca, Janssen, CiVi Biopharma, GlaxoSmithKline, SOCAR, Novo Nordisk, Health Outlook, Montai Health, Eli Lilly, New Amsterdam, Boehringer Ingelheim, RTI, Zomagen, Cytokinetics, Horizon Therapeutics, and Cardiol Therapeutics; has minority shareholder equity positions in Uppton, Bitterroot Bio, and ANGIOWave; and receives compensation for service on the Peter Munk Advisory Board (University of Toronto), the Leducq Foundation, Paris FR, and the Baim Institute. Dr Pradhan has received research grants from Kowa Research Europe and Denka, LTD; has received compensation for consultant services from Optum, Novo Nordisk, and Reliant Medical Foundation; receives compensation for lectures from Medtelligence and NACE; and is currently employed by Bristol Myers Squibb. All other authors have reported that they have no relationships relevant to the contents of this paper to disclose.
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.