Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Am J Kidney Dis ; 82(1): 97-104, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36965827

RESUMEN

RATIONALE & OBJECTIVE: Patients with chronic kidney disease (CKD), hyperkalemia (serum potassium [sK+]>5.0 mEq/L), and hyperphosphatemia experience poor clinical outcomes. Patiromer, a potassium binder that uses calcium as the exchange ion, may also reduce serum phosphorus (sP). We characterized the effect of patiromer on sP in patients with CKD, hyperkalemia, and hyperphosphatemia. STUDY DESIGN: A post hoc pooled analysis of individual-level data from the AMETHYST-DN, OPAL-HK, and TOURMALINE trials of patiromer. SETTING & PARTICIPANTS: Patients with CKD and hyperkalemia. EXPOSURE: Patients treated with patiromer (8.4-33.6 g/day). OUTCOME: Mean changes from baseline in sP, sK+, serum calcium (sCa2+), and serum magnesium (sMg2+) after 2 and 4 weeks of treatment. ANALYTICAL APPROACH: Descriptive statistics to summarize pooled data on the study outcomes from the 3 studies. RESULTS: We included 578 patients in the analysis. Of these participants, 86 patients (14.9%) had baseline hyperphosphatemia of whom 75.6% (65 of 86) had CKD stage 4/5 and 31.1% (153 of 492) with sP≤4.5mg/dL had CKD stage 4/5. Among the patients with elevated sP and sK+at baseline, the mean±SD reduction in sP and sK+after 4 weeks of patiromer treatment was-0.62±1.09mg/dL and-0.71± 0.51 mEq/L, respectively. Additionally, the mean±SD reduction in sMg2+in these patients was -0.25±0.23mg/dL while sCa2+remained unchanged. Both sMg2+and sCa2+remained within the normal range. Patiromer was generally well tolerated, and no serious adverse events were considered related to patiromer. LIMITATIONS: These were post hoc analyses, no placebo comparison was performed due to the design of the original studies, and the follow-up period was limited to 4 weeks. CONCLUSIONS: Reductions in sP and sK+to the normal range were observed after 2 weeks of patiromer treatment, and the reduction was sustained during 4 weeks of treatment among patients with non-dialysis-dependent CKD, hyperkalemia, and hyperphosphatemia. Future controlled trials are needed to establish if patiromer is useful to reduce both sK+and sP in hyperkalemic patients with CKD and hyperphosphatemia.


Asunto(s)
Hiperpotasemia , Hiperfosfatemia , Insuficiencia Renal Crónica , Humanos , Hiperpotasemia/tratamiento farmacológico , Hiperfosfatemia/tratamiento farmacológico , Hiperfosfatemia/etiología , Calcio , Potasio , Fósforo
2.
Am J Kidney Dis ; 80(5): 658-666, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36057467

RESUMEN

Mineralocorticoid receptor (MR) activation is involved in propagating kidney injury, inflammation, and fibrosis and in the progression of chronic kidney disease (CKD). Multiple clinical studies have defined the efficacy of MR antagonism in attenuating progressive kidney disease, and the US Food and Drug Administration recently approved the nonsteroidal mineralocorticoid receptor antagonist (MRA) finerenone for this indication. In this review, we consider the basic science and clinical applicability of MR antagonism. Because hyperkalemia constitutes a constraint to implementing evidence-based MR blockade, we review MRA-associated hyperkalemia in the context of finerenone and discuss evolving mitigation strategies to enhance the safety and efficacy of this treatment. Although the FIDELIO-DKD and FIGARO-DKD clinical trials focused solely on patients with type 2 diabetes mellitus, we propose that MR activation and the resulting inflammation and fibrosis act as a substantive pathogenetic mediator not only in people with diabetic CKD but also in those with CKD without diabetes. We close by briefly discussing both recently initiated and future clinical trials that focus on extending the attributes of MR antagonism to a wider array of nondiabetic kidney disorders, such as patients with nonalbuminuric CKD.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hiperpotasemia , Insuficiencia Renal Crónica , Humanos , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Receptores de Mineralocorticoides/uso terapéutico , Aldosterona , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hiperpotasemia/etiología , Mineralocorticoides/uso terapéutico , Insuficiencia Renal Crónica/complicaciones , Fibrosis , Inflamación
3.
Nephrol Dial Transplant ; 37(2): 211-221, 2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-34459924

RESUMEN

The nexus of chronic kidney disease (CKD) and cardiovascular disease (CVD) amplifies the morbidity and mortality of CKD, emphasizing the need for defining and establishing therapeutic initiatives to modify and abrogate the progression of CKD and concomitant CV risks. In addition to the traditional CV risk factors, disturbances of mineral metabolism are specific risk factors that contribute to the excessive CV mortality in patients with CKD. These risk factors include dysregulations of circulating factors that modulate phosphate metabolism, including fibroblast growth factor 23 (FGF23) and soluble Klotho. Reduced circulating levels and suppressed renal Klotho expression may be associated with adverse outcomes in CKD patients. While elevated circulating concentrations or locally produced FGF23 in the strained heart exert prohypertrophic mechanisms on the myocardium, Klotho attenuates tissue fibrosis, progression of CKD, cardiomyopathy, endothelial dysfunction, vascular stiffness and vascular calcification. Mineralocorticoid receptor (MR) activation in nonclassical targets, mediated by aldosterone and other ligands, amplifies CVD in CKD. In concert, we detail how the interplay of elevated FGF23, activation of the MR and concomitant reductions of circulating Klotho in CKD may potentiate each other's deleterious effects on the kidney and heart, thereby contributing to the initiation and progression of kidney and cardiac functional deterioration, acting through multipronged, albeit complementary, mechanistic pathways.


Asunto(s)
Enfermedades Cardiovasculares , Factores de Crecimiento de Fibroblastos , Proteínas Klotho , Receptores de Mineralocorticoides , Insuficiencia Renal Crónica , Enfermedades Cardiovasculares/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/metabolismo , Humanos , Riñón/metabolismo , Proteínas Klotho/metabolismo , Receptores de Mineralocorticoides/metabolismo , Insuficiencia Renal Crónica/complicaciones
4.
Kidney Int ; 99(6): 1321-1330, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33781792

RESUMEN

The hepatorenal syndrome (HRS), a progressive but potentially reversible deterioration of kidney function, constitutes a serious complication of hepatic decompensation. Coexistence of liver/kidney damage, mentioned in the dropsy literature, was highlighted by Richard Bright in 1827 and confirmed in 1840 by his contemporary nephrology pioneer Pierre Rayer. Cholemic nephrosis was described in 1861 by Friedrich Frerichs, and the renal tubular lesions of HRS by Austin Flint in 1863. The term "acute hepato-nephritis" was introduced in 1916 by Paul Merklen, and its chronic form was designated HRS by Marcel Dérot in 1930s. HRS then was applied to renal failure in biliary tract surgery and to cases of coexistent renal and hepatic failure of diverse etiology. The pathogenesis of HRS was elucidated during the 1950 studies of renal physiology. Notably, studies of salt retention in edema and its relation to regulating the circulating plasma volume by John Peters and subsequently Otto Gauer defined the concept of "effective blood volume" and the consequent elucidation of ascites formation in liver failure. Parallel studies of intrarenal hemodynamics demonstrated severe renal vasoconstriction and preferential cortical ischemia to account for the functional renal dysfunction of HRS. Dialysis and liver or combined liver-kidney transplantation transformed the fatal HRS of old into a treatable disorder by the 1970s. Elucidation of the pathogenetic mechanisms of renal injury and refinements in definition, classification, and diagnosis of HRS since then have allowed for earlier therapeutic intervention with combined i.v. albumin and vasoconstrictor therapy, enabling the continued improvement of patient outcomes.


Asunto(s)
Síndrome Hepatorrenal , Trasplante de Hígado , Síndrome Hepatorrenal/diagnóstico , Síndrome Hepatorrenal/etiología , Humanos , Pruebas de Función Renal , Cirrosis Hepática , Vasoconstrictores/uso terapéutico
5.
Am J Nephrol ; 52(3): 209-216, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33857953

RESUMEN

BACKGROUND: A full understanding of the mechanisms of action of aldosterone and its interaction with the mineralocorticoid receptor (MR) allows a theoretical framework to predict the therapeutic potential of MR antagonists (MRAs) in CKD, and heart failure with reduced ejection fraction. SUMMARY: The initial focus on the mechanisms of action of aldosterone was directed primarily on its role in modulating renal excretory function. In contrast, many recent studies have demonstrated a wider and expanded role for aldosterone in modulating inflammation, collagen formation, fibrosis, and necrosis. Increasing evidence has accrued that implicates the pathophysiological overactivation of the MR as a major determinant of progression of CKD. By promoting inflammation and fibrosis, MR overactivation constitutes a pivotal determinant of CKD progression and its associated morbidity and mortality. In accord with this mechanism of action, blockade of the MR is currently being investigated as a novel treatment regimen to slow the progression of CKD. The recently reported FIDELIO-DKD (FInerenone in reducing kiDnEy faiLure and dIsease prOgression in Diabetic Kidney Disease) study demonstrated that patients with CKD and type 2 diabetes who were treated with finerenone (a novel nonsteroidal MRA) manifested a lower risk of a composite primary outcome event compared with patients in the placebo arm (defined as kidney failure, or a sustained decrease of ≥40% in the estimated glomerular filtration rate from baseline, or death from renal causes). In addition, patients in the finerenone group also manifested a lower risk of a key secondary outcome event (defined as death from cardiovascular causes, nonfatal myocardial infarction, nonfatal stroke, or hospitalization for heart failure). Key Messages: Based on the success of the FIDELIO-DKD study, future studies should be implemented testing the hypothesis that a wide array of nondiabetic CKD is modulated by overactivation of the MR, and consequently may be amenable to treatment with novel nonsteroidal MRAs. Future studies are encouraged to elucidate the clinical implications of the interplay of nonsteroidal MRAs and the components of the renin-angiotensin cascade. The unique and recently reported interrelationship of fibroblast growth factor (FGF23) and aldosterone may also constitute a propitious subject for future investigation.


Asunto(s)
Aldosterona/fisiología , Enfermedades Cardiovasculares/etiología , Receptores de Mineralocorticoides/fisiología , Insuficiencia Renal Crónica/etiología , Humanos , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Naftiridinas/uso terapéutico , Insuficiencia Renal Crónica/tratamiento farmacológico
6.
Pharmacol Res ; 113(Pt A): 585-591, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27693804

RESUMEN

Hyperkalemia is a common electrolyte disorder, especially in chronic kidney disease, diabetes mellitus, or heart failure. Hyperkalemia can lead to potentially fatal cardiac dysrhythmias, and it is associated with increased mortality. Determining whether emergency therapy is warranted is largely based on subjective clinical judgment. The Investigator Network Initiative Cardiovascular and Renal Clinical Trialists (INI-CRCT) aimed to evaluate the current knowledge pertaining to the emergency treatment of hyperkalemia. The INI-CRCT developed a treatment algorithm reflecting expert opinion of best practices in the context of current evidence, identified gaps in knowledge, and set priorities for future research. We searched PubMed (to August 4, 2015) for consensus guidelines, reviews, randomized clinical trials, and observational studies, limited to English language but not by publication date. Treatment approaches are based on small studies, anecdotal experience, and traditional practice patterns. The safety and real-world effectiveness of standard therapies remain unproven. Prospective research is needed and should include studies to better characterize the population, define the serum potassium thresholds where life-threatening arrhythmias are imminent, assess the potassium and electrocardiogram response to standard interventions. Randomized, controlled trials are needed to test the safety and efficacy of new potassium binders for the emergency treatment of severe hyperkalemia in hemodynamically stable patients. Existing emergency treatments for severe hyperkalemia are not supported by a compelling body of evidence, and they are used inconsistently across institutions, with potentially significant associated side effects. Further research is needed to fill knowledge gaps, and definitive clinical trials are needed to better define optimal management strategies, and ultimately to improve outcomes in these patients.


Asunto(s)
Hiperpotasemia/tratamiento farmacológico , Arritmias Cardíacas/sangre , Arritmias Cardíacas/tratamiento farmacológico , Electrocardiografía/métodos , Humanos , Hiperpotasemia/sangre , Estudios Observacionales como Asunto , Potasio/sangre , Ensayos Clínicos Controlados Aleatorios como Asunto
7.
Eur Heart J ; 2020 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-32984871
8.
Eur Heart J ; 41(47): 4468-4470, 2020 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-32735314
9.
Eur Heart J ; 41(16): 1540-1541, 2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32318742
10.
Am J Physiol Renal Physiol ; 309(2): F90-7, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25995107

RESUMEN

This article reviews the pioneering and visionary contributions of the Catalan surgeon Josep Trueta (1897­1977) to the changes in renal circulation that contribute to the pathogenesis of acute renal failure (ARF). An erudite scientist with eclectic interests in physiology, orthopedics, politics, and medical history, Trueta's initial involvement in wound healing as a trauma surgeon during the Spanish Civil War and the London Blitz is what prompted him to postulate that a trauma-induced "neural effect" on the renal vasculature, with resultant renal arterial constriction could cause ARF. To test his hypothesis, Trueta assembled an experienced radiologist, a renowned physiologist, and a renal pathologist to study ARF in Oxford. They investigated the renal circulation of rabbits in response to diverse traumatic conditions by injecting a radio-opaque substance, using cine-radiography to visualize the flow of blood through the renal vasculature. Trueta's suggestion of renal cortical ischemia and diversion of blood to the less resistant medullary circulation (Trueta shunt) was criticized by Homer Smith and coworkers. In contrast to Homer Smith's data, which were derived from clearance studies and renal arteriovenous oxygen, Trueta used the diametrical opposite method of "direct" observation of the renal circulation. Their differing methodologies, direct visualization of the renal circulation as opposed to inferred computations from clearance studies, accounts for some of their conflicting theories. Nevertheless, the proposal of disparate renal flow compartments focused attention on intrarenal hemodynamics. Trueta's focus on renal cortical ischemia was ultimately validated by the studies of Barger in the dog and Hollenberg and Epstein in human subjects.


Asunto(s)
Lesión Renal Aguda/etiología , Nefrología/historia , Circulación Renal , Animales , Investigación Biomédica/historia , Historia del Siglo XVII , Historia del Siglo XVIII , Historia del Siglo XIX , Historia del Siglo XX , Humanos
12.
Kidney Int ; 81(10): 955-968, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22336987

RESUMEN

The past two decades have witnessed a striking paradigm shift with respect to our understanding of the widespread effects of aldosterone. There is substantive evidence that mineralocorticoid receptor (MR) activation promotes myriad 'off target' effects on the heart, the vasculature, and importantly the kidney. In the present review, we summarize the expanding role of MR activation in promoting both vascular and renal injury. We review the recent clinical studies that investigated the efficacy of MR antagonism (MRA) in reducing proteinuria and attenuating progressive renal disease. We also review in-depth both the utility and safety of MRA in the end-stage renal disease (ESRD) patient undergoing dialysis. Because the feasibility of add-on MRA is critically dependent on our ability to minimize or avoid hyperkalemia, and because controversy centers on the incidence of hyperkalemia, we critically review the risk of hyperkalemia with add-on MRA. Our present analysis suggests that hyperkalemia supervening in MRA-treated patients is overstated. Furthermore, recent studies demonstrating the efficacy of new non-absorbed, orally administered, potassium [K+]-binding polymers suggest that a multi-pronged approach encompassing adequate surveillance, moderate or low-dose MRA, and K-binding polymers may adequately control serum K in both chronic kidney disease and ESRD patients.


Asunto(s)
Aldosterona/metabolismo , Fallo Renal Crónico/terapia , Riñón/efectos de los fármacos , Antagonistas de Receptores de Mineralocorticoides , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Diálisis Renal , Aldosterona/sangre , Animales , Quelantes/uso terapéutico , Terapia Combinada , Progresión de la Enfermedad , Humanos , Hiperpotasemia/sangre , Hiperpotasemia/etiología , Hiperpotasemia/prevención & control , Riñón/metabolismo , Riñón/fisiopatología , Fallo Renal Crónico/tratamiento farmacológico , Fallo Renal Crónico/metabolismo , Fallo Renal Crónico/fisiopatología , Antagonistas de Receptores de Mineralocorticoides/efectos adversos , Potasio/sangre , Receptores de Mineralocorticoides/metabolismo , Diálisis Renal/efectos adversos , Medición de Riesgo , Factores de Riesgo , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento
14.
Kidney Int Suppl (2011) ; 12(1): 69-75, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35529085

RESUMEN

The recent successful demonstrations that the nonsteroidal mineralocorticoid receptor (MR) antagonist finerenone provides effective kidney and cardiovascular (CV) protection in patients with chronic kidney disease (CKD) and type 2 diabetes constitutes a platform for considering and implementing an array of future clinical trials in patients with nondiabetic CKD. Activation of the MR, with consequent inflammation and fibrosis, should be operative as a pathogenetic mediator not only in patients with diabetic CKD but also in those with nondiabetic kidney disease. Consequently, it is proposed that MR antagonism therapy will be equally efficacious in patients with nondiabetic CKD. Recently, a major new clinical trial has been initiated testing finerenone in patients with nondiabetic kidney disease (FIND-CKD; NCT05047263). A second clinical development program, FIONA, is dedicated to studies of finerenone in children with glomerular and nonglomerular CKD. Finally, the interrelationship of fibroblast growth factor 23 (FGF23), membrane αKlotho (hereafter called Klotho), and aldosterone may be a propitious subject for future investigation. The interplay and intersection of these seemingly disparate yet intricate relationships may unmask novel, and indeed compelling, opportunities for therapeutic interventions that are capable of interrupting the vicious cycle of excess aldosterone/MR activation and FGF23 secretion with concomitant Klotho insufficiency characteristically present in patients with CKD.

15.
Diabetes Ther ; 13(4): 583-588, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35294746

RESUMEN

The classic focus on the mechanisms of action of aldosterone was directed primarily on its role in modulating renal excretory function and maintaining volume homeostasis. In contrast, many recent studies have demonstrated a much wider and expanded role for aldosterone and for the mineralocorticoid receptor (MR). Activation of the MR promotes inflammation, collagen formation, fibrosis, and necrosis with consequent renal injury. Increasing evidence has accrued that implicates the pathophysiological overactivation of the MR as a major determinant of progression of both diabetic and nondiabetic chronic kidney disease (CKD). By promoting cascades of injury encompassing inflammation and fibrosis, MR overactivation constitutes a pivotal determinant of CKD progression and consequently its associated morbidity and mortality. Based on this mechanism of action, blockade of the MR with the nonsteroidal MR antagonist finerenone is currently being investigated as a novel treatment regimen to slow the progression of CKD. The recently reported FIDELIO-DKD (FInerenone in reducing kiDnEy faiLure and dIsease prOgression in Diabetic Kidney Disease) study demonstrated that patients with CKD and type 2 diabetes (T2D) who were treated with finerenone manifested a lower risk of a composite primary outcome event compared with patients in the placebo arm (defined as kidney failure or a sustained decrease of ≥ 40% in the estimated glomerular filtration rate from baseline, or death from renal causes). In addition, patients in the finerenone group also manifested a lower risk of a key secondary outcome event (defined as death from cardiovascular causes, nonfatal myocardial infarction, nonfatal stroke, or hospitalization for heart failure). Based on the success of these major clinical trials, finerenone was approved by the FDA on 9 July 2021 as a novel treatment for retarding CKD progression in patients with T2D ( https://www.fda.gov/drugs/news-events-human-drugs/fda-approves-drug-reduce-risk-serious-kidney-and-heart-complications-adults-chronic-kidney-disease ). Podcast Video (MP4 258973 KB).

16.
Am J Med ; 135(5): 576-580, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34979095

RESUMEN

Chronic kidney disease affects approximately 10% of the population or 800 million people globally, with diabetes being the leading cause. The presence of chronic kidney disease with impaired kidney function or with albuminuria is associated with an increased risk of a progressive loss of renal function and increased risk of cardiovascular disease and excess mortality. Screening for chronic kidney disease is critically important because during the initial stages patients often have no symptoms and because we now have available recently approved multiple interventions that can reduce the high risks dramatically. Screening should be performed with regular measurement of albumin in the urine and creatinine or cystatin C in blood for estimation of kidney function. Regretfully recent data indicates that screening for albuminuria is conducted in only 20%-50% of people at risk depending on the setting. Clinicians need to perform regular screening and concomitant management of risk factors. Recent therapeutic options must be implemented to improve outcomes. Finally, a reduction in albuminuria after initiation of intervention constitutes a treatment target because it indicates improved prognosis.


Asunto(s)
Enfermedades Cardiovasculares , Insuficiencia Renal Crónica , Albuminuria/epidemiología , Creatinina , Femenino , Humanos , Masculino , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/epidemiología , Factores de Riesgo
17.
Kidney Int Suppl (2011) ; 12(1): 48-62, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35316977

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic, causing considerable mortality and morbidity worldwide, has fully engaged the biomedical community in attempts to elucidate the pathophysiology of COVID-19 and develop robust therapeutic strategies. To this end, the predominant research focus has been on the adaptive immune response to COVID-19 infections stimulated by mRNA and protein vaccines and on the duration and persistence of immune protection. In contrast, the role of the innate immune response to the viral challenge has been underrepresented. This overview focuses on the innate immune response to COVID-19 infection, with an emphasis on the roles of extracellular proteases in the tissue microenvironment. Proteinase-mediated signaling caused by enzymes in the extracellular microenvironment occurs upstream of the increased production of inflammatory cytokines that mediate COVID-19 pathology. These enzymes include the coagulation cascade, kinin-generating plasma kallikrein, and the complement system, as well as angiotensin-generating proteinases of the renin-angiotensin system. Furthermore, in the context of several articles in this Supplement elucidating and detailing the trajectory of diverse profibrotic pathways, we extrapolate these insights to explore how fibrosis and profibrotic pathways participate importantly in the pathogenesis of COVID-19. We propose that the lessons garnered from understanding the roles of microenvironment proteinases in triggering the innate immune response to COVID-19 pathology will identify potential therapeutic targets and inform approaches to the clinical management of COVID-19. Furthermore, the information may also provide a template for understanding the determinants of COVID-19-induced tissue fibrosis that may follow resolution of acute infection (so-called "long COVID"), which represents a major new challenge to our healthcare systems.

20.
Am J Med ; 134(2): 166-175, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32950502

RESUMEN

The current Coronavirus Disease 2019 (COVID-19) pandemic has exerted an unprecedented impact across the globe. As a consequence of this overwhelming catastrophe, long-established prevailing medical and scientific paradigms have been disrupted. The response of the scientific community, medical journals, media, and some politicians has been far from ideal. The present manuscript discusses the failure of the scientific enterprise in its initiatives to address the COVID-19 outbreak as a consequence of the disarray attributable to haste and urgency. To enhance conveying our message, this manuscript is organized into 3 interrelated sections: 1) the accelerated pace of publications coupled with a dysfunctional review process; 2) failure of the clinical trial enterprise; 3) propagation of misleading information by the media. In response we propose a template comprising a focus on randomized controlled clinical trials, and an insistence on responsible journal publication, and enumeration of policies to deal with social media-propagated news. We conclude with a reconsideration of the appropriate role of academic medicine and journals.


Asunto(s)
COVID-19 , Investigación Biomédica , Comunicación , Predicción , Humanos , Edición/normas , Edición/estadística & datos numéricos , Ensayos Clínicos Controlados Aleatorios como Asunto , Facultades de Medicina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA