Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
J Am Soc Nephrol ; 31(3): 469-482, 2020 03.
Article in English | MEDLINE | ID: mdl-31988269

ABSTRACT

Retrospective analyses and single-center prospective studies identify chronic metabolic acidosis as an independent and modifiable risk factor for progression of CKD. In patients with CKD, untreated chronic metabolic acidosis often leads to an accelerated reduction in GFR. Mechanisms responsible for this reduction include adaptive responses that increase acid excretion but lead to a decline in kidney function. Metabolic acidosis in CKD stimulates production of intrakidney paracrine hormones including angiotensin II, aldosterone, and endothelin-1 (ET-1) that mediate the immediate benefit of increased kidney acid excretion, but their chronic upregulation promotes inflammation and fibrosis. Chronic metabolic acidosis also stimulates ammoniagenesis that increases acid excretion but also leads to ammonia-induced complement activation and deposition of C3 and C5b-9 that can cause tubule-interstitial damage, further worsening disease progression. These effects, along with acid accumulation in kidney tissue, combine to accelerate progression of kidney disease. Treatment of chronic metabolic acidosis attenuates these adaptive responses; reduces levels of angiotensin II, aldosterone, and ET-1; reduces ammoniagenesis; and diminishes inflammation and fibrosis that may lead to slowing of CKD progression.


Subject(s)
Acidosis/complications , Aldosterone/metabolism , Angiotensin II/metabolism , Disease Progression , Endothelin-1/metabolism , Renal Insufficiency, Chronic/physiopathology , Acid-Base Equilibrium/drug effects , Acid-Base Equilibrium/physiology , Acidosis/diagnosis , Acidosis/drug therapy , Adaptation, Physiological/physiology , Biomarkers/blood , Female , Follow-Up Studies , Glomerular Filtration Rate/physiology , Humans , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/physiopathology , Male , Prospective Studies , Renal Insufficiency, Chronic/etiology , Retrospective Studies , Risk Assessment , Severity of Illness Index , Time Factors , Treatment Outcome
2.
J Pharmacol Exp Ther ; 375(3): 439-450, 2020 12.
Article in English | MEDLINE | ID: mdl-33033169

ABSTRACT

Current management of metabolic acidosis in patients with chronic kidney disease (CKD) relies on dietary intervention to reduce daily endogenous acid production or neutralization of retained acid with oral alkali (sodium bicarbonate, sodium citrate). Veverimer is being developed as a novel oral treatment for metabolic acidosis through removal of intestinal acid, resulting in an increase in serum bicarbonate. Veverimer is a free-amine polymer that combines high capacity and selectivity to bind and remove hydrochloric acid (HCl) from the gastrointestinal (GI) tract. In vitro studies demonstrated that veverimer had a binding capacity of 10.7 ± 0.4 mmol HCl per gram of polymer with significant binding capacity (>5 mmol/g) across the range of pH values found in the human GI tract (1.5-7). Upon protonation, veverimer bound chloride with high specificity but showed little or no binding of phosphate, citrate, or taurocholate (<1.5 mmol/g), which are all anions commonly found in the human GI tract. Administration of veverimer to rats with adenine-induced CKD and metabolic acidosis resulted in a significant increase in fecal chloride excretion and a dose-dependent increase in serum bicarbonate to within the normal range compared with untreated controls. Absorption, distribution, metabolism, and excretion studies in rats and dogs dosed with 14C-labeled veverimer showed that the polymer was not absorbed from the GI tract and was quantitatively eliminated in the feces. Acid removal by veverimer, an orally administered, nonabsorbed polymer, may provide a potential new treatment for metabolic acidosis in patients with CKD. SIGNIFICANCE STATEMENT: Metabolic acidosis is a complication of chronic kidney disease (CKD) as well as a cause of CKD progression. Veverimer is a high-capacity, selective, nonabsorbed, hydrochloric acid-binding polymer being developed as a treatment for metabolic acidosis. Veverimer binds and removes hydrochloric acid from the gastrointestinal tract, resulting in increased serum bicarbonate and the correction of metabolic acidosis. Veverimer is not an ion-exchange resin and does not deliver sodium or other counterions, and so it may be appropriate for patients with CKD with and without sodium-sensitive comorbidities.


Subject(s)
Acidosis/complications , Acidosis/drug therapy , Hydrochloric Acid/metabolism , Polymers/administration & dosage , Polymers/pharmacology , Renal Insufficiency, Chronic/complications , Acidosis/metabolism , Administration, Oral , Animals , Biological Availability , Hydrogen-Ion Concentration , Male , Polymers/chemistry , Polymers/pharmacokinetics , Rats , Rats, Sprague-Dawley , Solubility
3.
Clin J Am Soc Nephrol ; 14(7): 1011-1020, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31196951

ABSTRACT

BACKGROUND AND OBJECTIVES: Metabolic acidosis is associated with progression of CKD and has significant adverse effects on muscle and bone. A systematic review and meta-analysis was conducted to evaluate the benefits and risks of metabolic acidosis treatment with oral alkali supplementation or a reduction of dietary acid intake in those with CKD. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: MEDLINE, Embase, and Cochrane CENTRAL were searched for relevant trials in patients with stage 3-5 CKD and metabolic acidosis (<22 mEq/L) or low-normal serum bicarbonate (22-24 mEq/L). Data were pooled in a meta-analysis with results expressed as weighted mean difference for continuous outcomes and relative risk for categorical outcomes with 95% confidence intervals (95% CIs), using a random effects model. Study quality and strength of evidence were assessed using Cochrane risk of bias and the Grading of Recommendations Assessment, Development and Evaluation criteria. RESULTS: Fourteen clinical trials were included (n=1394 participants). Treatment of metabolic acidosis with oral alkali supplementation or a reduction of dietary acid intake increased serum bicarbonate levels (14 studies, 1378 patients, mean difference 3.33 mEq/L, 95% CI, 2.37 to 4.29) and resulted in a slower decline in eGFR (13 studies, 1329 patients, mean difference -3.28 ml/min per 1.73 m2, 95% CI, -4.42 to -2.14; moderate certainty) and a reduction in urinary albumin excretion (very-low certainty), along with a reduction in the risk of progression to ESKD (relative risk, 0.32; 95% CI, 0.18 to 0.56; low certainty). Oral alkali supplementation was associated with worsening hypertension or the requirement for increased antihypertensive therapy (very-low certainty). CONCLUSIONS: Low-to-moderate certainty evidence suggest that oral alkali supplementation or a reduction in dietary acid intake may slow the rate of kidney function decline and potentially reduce the risk of ESKD in patients with CKD and metabolic acidosis.


Subject(s)
Acidosis/therapy , Renal Insufficiency, Chronic/complications , Alkalies/administration & dosage , Bicarbonates/blood , Humans , Kidney Failure, Chronic/prevention & control
4.
Clin J Am Soc Nephrol ; 13(1): 26-35, 2018 01 06.
Article in English | MEDLINE | ID: mdl-29102959

ABSTRACT

BACKGROUND AND OBJECTIVES: Metabolic acidosis is common in patients with CKD and has significant adverse effects on kidney, muscle, and bone. We tested the efficacy and safety of TRC101, a novel, sodium-free, nonabsorbed hydrochloric acid binder, to increase serum bicarbonate in patients with CKD and metabolic acidosis. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: One hundred thirty-five patients were enrolled in this randomized, double-blind, placebo-controlled, multicenter, in-unit study (designated the TRCA-101 Study). Patients had a mean baseline eGFR of 35 ml/min per 1.73 m2, a mean baseline serum bicarbonate of 17.7 mEq/L, and comorbidities, including hypertension (93%), diabetes (70%), and heart failure (21%). Patients ate a controlled diet and were treated for 14 days with placebo or one of four TRC101 dosing regimens (1.5, 3, or 4.5 g twice daily or 6 g once daily). After treatment, patients were discharged and followed for 7-14 days. RESULTS: All TRC101 treatment groups had a mean within-group increase in serum bicarbonate of ≥1.3 mEq/L (P<0.001) within 72 hours of the first dose and a mean increase in serum bicarbonate of 3.2-3.9 mEq/L (P<0.001) at the end of treatment compared with placebo, in which serum bicarbonate did not change. In the combined TRC101 treatment group, serum bicarbonate was normalized (22-29 mEq/L) at the end of treatment in 35% of patients and increased by ≥4 mEq/L in 39% of patients. After discontinuation of TRC101, serum bicarbonate decreased nearly to baseline levels within 2 weeks. All adverse events were mild or moderate, with gastrointestinal events most common. All patients completed the study. CONCLUSIONS: TRC101 safely and significantly increased the level of serum bicarbonate in patients with metabolic acidosis and CKD.


Subject(s)
Acid-Base Equilibrium/drug effects , Acidosis/drug therapy , Bicarbonates/blood , Chelating Agents/therapeutic use , Polymers/therapeutic use , Renal Insufficiency, Chronic/complications , Acidosis/diagnosis , Acidosis/etiology , Acidosis/physiopathology , Adult , Aged , Biomarkers , Bulgaria , Chelating Agents/adverse effects , Double-Blind Method , Female , Georgia , Glomerular Filtration Rate , Humans , Kidney/physiopathology , Male , Middle Aged , Polymers/adverse effects , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/physiopathology , Time Factors , Treatment Outcome , United States
5.
J Cardiovasc Pharmacol Ther ; 21(5): 456-65, 2016 09.
Article in English | MEDLINE | ID: mdl-26856345

ABSTRACT

Hyperkalemia is a potentially life-threatening condition, and patients who have chronic kidney disease, who are diabetic, or who are taking renin-angiotensin-aldosterone system inhibitors are at increased risk. Therapeutic options for hyperkalemia tend to have limited effectiveness and can be associated with serious side effects. Colonic potassium secretion can increase to compensate when urinary potassium excretion decreases in patients with renal impairment, but this adaptation is insufficient and hyperkalemia still results. Patiromer is a novel, spherical, nonabsorbed polymer designed to bind and remove potassium, primarily in the colon, thereby decreasing serum potassium in patients with hyperkalemia. Patiromer has been found to decrease serum potassium in patients with hyperkalemia having chronic kidney disease who were on renin-angiotensin-aldosterone system inhibitors. Results of nonclinical studies and an early phase clinical study are reported here. Two studies with radiolabeled drug, one in rats and the other in dogs, confirmed that patiromer was not absorbed into the systemic circulation. Results of an in vitro study showed that patiromer was able to bind 8.5 to 8.8 mEq of potassium per gram of polymer at a pH similar to that found in the colon and had a much higher potassium-binding capacity compared with other resins, including polystyrene sulfonate. In a study in hyperkalemic rats, a decrease in serum potassium was observed via an increase in fecal potassium excretion. In a clinical study in healthy adult volunteers, a significant increase in fecal potassium excretion and a significant decrease in urinary potassium excretion were observed. Overall, patiromer is a high-capacity potassium binder, and the chemical and physical characteristics of patiromer may lead to good clinical efficacy, tolerability, and patient acceptance.


Subject(s)
Chelating Agents/therapeutic use , Hyperkalemia/drug therapy , Polymers/therapeutic use , Potassium/blood , Animals , Biomarkers/blood , Chelating Agents/adverse effects , Chelating Agents/pharmacokinetics , Colon/drug effects , Colon/metabolism , Disease Models, Animal , Feces/chemistry , Humans , Hyperkalemia/blood , Hyperkalemia/diagnosis , Intestinal Elimination , Polymers/adverse effects , Polymers/pharmacokinetics , Treatment Outcome
6.
Future Cardiol ; 8(1): 17-28, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22185443

ABSTRACT

The treatment of heart failure has seen considerable advances in the past decades. In particular, a therapeutic focus on the renin-angiotensin-aldosterone system has provided significant improvements in outcomes. Multiple inhibition points in the renin-angiotensin-aldosterone system, including direct renin inhibitors, angiotensin-converting enzyme inhibitors, angiotensin receptor blockers and mineralocorticoid receptor antagonists, have the common feature of either blocking aldosterone production (direct renin inhibitor, angiotensin-converting enzyme inhibitor, angiotensin receptor blocker) or the mineralocorticoid receptor. As a consequence of this inhibition, sodium and water reabsorption is blocked, and potassium (K(+)) excretion is reduced. Hyperkalemia may result from the use of multiple renin-angiotensin-aldosterone inhibitors or blockers, particularly in patients with heart failure and concomitant chronic kidney disease. Interventions to reliably control serum K(+) during renin-angiotensin-aldosterone inhibition have not been available to date, and would be of particular value with the use of mineralocorticoid receptor antagonists that have been shown to reduce mortality in patients with heart failure and a reduced left ventricular ejection fraction. In this review, we examine the PEARL-HF study, which has tested the combined use of RLY5016, a novel nonabsorbed K(+) binding polymer, with spironolactone in heart failure patients receiving standard care but with previous documented hyperkalemia or chronic kidney disease. RLY5016 significantly lowered serum K(+) levels from baseline relative to placebo, lowered the incidence of hyperkalemia and allowed a higher proportion of heart failure patients to receive spironolactone at a dose of 50 mg/day.


Subject(s)
Heart Failure/complications , Hyperkalemia/prevention & control , Polymers/pharmacology , Potassium/blood , Aged , Analysis of Variance , Female , Glomerular Filtration Rate , Health Status Indicators , Humans , Hyperkalemia/chemically induced , Hyperkalemia/etiology , Male , Mineralocorticoid Receptor Antagonists/therapeutic use , Polymers/therapeutic use , Renin-Angiotensin System/drug effects , Spironolactone/therapeutic use
7.
J Bacteriol ; 186(24): 8478-89, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15576798

ABSTRACT

Gene products required for in vivo growth and survival of microbial pathogens comprise a unique functional class and may represent new targets for antimicrobial chemotherapy, vaccine construction, or diagnostics. Although some factors governing Staphylococcus aureus pathogenicity have been identified and studied, a comprehensive genomic analysis of virulence functions will be a prerequisite for developing a global understanding of interactions between this pathogen and its human host. In this study, we describe a genetic screening strategy and demonstrate its use in screening a collection of 6,300 S. aureus insertion mutants for virulence attenuation in a murine model of systemic infection. Ninety-five attenuated mutants were identified, reassembled into new pools, and rescreened using the same murine model. This effort identified 24 highly attenuated mutants, each of which was further characterized for virulence attenuation in vivo and for growth phenotypes in vitro. Mutants were recovered in numbers up to 1,200-fold less than wild type in the spleens of systemically infected animals and up to 4,000-fold less than wild type in localized abscess infections. Genetic analysis of the mutants identified insertions in 23 unique genes. The largest gene classes represented by these mutants encoded enzymes involved in small-molecule biosynthesis and cell surface transmembrane proteins involved in small-molecule binding and transport. Additionally, three insertions defined two histidine kinase sensor-response regulator gene pairs important for S. aureus in vivo survival. Our findings extend the understanding of pathogenic mechanisms employed by S. aureus to ensure its successful growth and survival in vivo. Many of the gene products we have identified represent attractive new targets for antibacterial chemotherapy.


Subject(s)
Bacteremia/microbiology , Bacterial Proteins/genetics , Genes, Bacterial , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity , Virulence/genetics , Animals , DNA Transposable Elements , Disease Models, Animal , Female , Gene Library , Genetic Markers , Humans , Mice , Mice, Inbred BALB C , Mutagenesis, Insertional , Staphylococcus aureus/genetics
8.
Antimicrob Agents Chemother ; 47(7): 2273-82, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12821479

ABSTRACT

Compounds that selectively disrupt fungal mitosis have proven to be effective in controlling agricultural pests, but no specific mitotic inhibitor is available for the treatment of systemic mycoses in mammalian hosts. In an effort to identify novel mitotic inhibitors, we used a cell-based screening strategy that exploited the hypersensitivity of a yeast alpha-tubulin mutant strain to growth inhibition by antimitotic agents. The compounds identified inhibited yeast nuclear division and included one structural class of compounds shown to be fungus specific. MC-305904 and structural analogs inhibited fungal cell mitosis and inhibited the in vitro polymerization of fungal tubulin but did not block mammalian cell microtubule function or mammalian tubulin polymerization. Extensive analysis of yeast mutations that specifically alter sensitivity to MC-305904 structural analogs suggested that compounds in the series bind to a site on fungal beta-tubulin near amino acid 198. Features of the proposed binding site explain the observed fungal tubulin specificity of the series and are consistent with structure-activity relationships among a library of related compounds.


Subject(s)
Antifungal Agents/pharmacology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Tubulin/genetics , Antifungal Agents/chemistry , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Binding Sites , Drug Design , Microbial Sensitivity Tests , Mutation , Polymers
SELECTION OF CITATIONS
SEARCH DETAIL