Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 247
Filter
1.
Am Heart J ; 240: 101-113, 2021 10.
Article in English | MEDLINE | ID: mdl-34175315

ABSTRACT

BACKGROUND: The presence and extent of left ventricular hypertrophy (LVH) is a major determinant of symptoms in patients with hypertrophic cardiomyopathy (HCM). There is increasing evidence to suggest that myocardial energetic impairment represents a central mechanism leading to LVH in HCM. There is currently a significant unmet need for disease-modifying therapy that regresses LVH in HCM patients. Perhexiline, a potent carnitine palmitoyl transferase-1 (CPT-1) inhibitor, improves myocardial energetics in HCM, and has the potential to reduce LVH in HCM. OBJECTIVE: The primary objective is to evaluate the effects of perhexiline treatment on the extent of LVH, in symptomatic HCM patients with at least moderate LVH. METHODS/DESIGN: RESOLVE-HCM is a prospective, multicenter double-blind placebo-controlled randomized trial enrolling symptomatic HCM patients with at least moderate LVH. Sixty patients will be randomized to receive either perhexiline or matching placebo. The primary endpoint is change in LVH, assessed utilizing cardiovascular magnetic resonance (CMR) imaging, after 12-months treatment with perhexiline. SUMMARY: RESOLVE-HCM will provide novel information on the utility of perhexiline in regression of LVH in symptomatic HCM patients. A positive result would lead to the design of a Phase 3 clinical trial addressing long-term effects of perhexiline on risk of heart failure and mortality in HCM patients.


Subject(s)
Cardiomyopathy, Hypertrophic/drug therapy , Cardiomyopathy, Hypertrophic/physiopathology , Cardiovascular Agents/therapeutic use , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/prevention & control , Perhexiline/therapeutic use , Adult , Cardiomyopathy, Hypertrophic/diagnostic imaging , Double-Blind Method , Echocardiography , Female , Humans , Hypertrophy, Left Ventricular/diagnostic imaging , Magnetic Resonance Imaging , Male , Prospective Studies
2.
Bioorg Chem ; 102: 104067, 2020 09.
Article in English | MEDLINE | ID: mdl-32663671

ABSTRACT

Schistosomiasis is a neglected tropical disease mainly affecting the poorest tropical and subtropical areas of the world with the impressive number of roughly 200 million infections per year. Schistosomes are blood trematode flukes of the genus Schistosoma causing symptoms in humans and animals. Organ morbidity is caused by the accumulation of parasite eggs and subsequent development of fibrosis. If left untreated, schistosomiasis can result in substantial morbidity and even mortality. Praziquantel (PZQ) is the most effective and widely used compound for the treatment of the disease, in prevention and control programs in the last 30 years. Unfortunately, it has no effect on juvenile immature schistosomes and cannot prevent reinfection or interfere with the schistosome life cycle; moreover drug-resistance represents a serious threat. The search for an alternative or complementary treatment is urgent and drug repurposing could accelerate a solution. The anti-anginal drug perhexiline maleate (PHX) has been previously shown to be effective on larval, juvenile, and adult stages of S. mansoni and to impact egg production in vitro. Since PHX is a racemic mixture of R-(+)- and S-(-)-enantiomers, we designed and realized a stereoselective synthesis of both PHX enantiomers and developed an analytical procedure for the direct quantification of the enantiomeric excess also suitable for semipreparative separation of PHX enantiomers. We next investigated the impact of each enantiomer on viability of newly transformed schistosomula (NTS) and worm pairs of S. mansoni as well as on egg production and vitellarium morphology by in vitro studies. Our results indicate that the R-(+)-PHX is mainly driving the anti-schistosomal activity but that also the S-(-)-PHX possesses a significant activity towards S. mansoni in vitro.


Subject(s)
Perhexiline/analogs & derivatives , Schistosoma mansoni/drug effects , Animals , Larva , Molecular Structure , Perhexiline/therapeutic use , Stereoisomerism , Structure-Activity Relationship
3.
Cancer Lett ; 473: 74-89, 2020 03 31.
Article in English | MEDLINE | ID: mdl-31904482

ABSTRACT

Gastrointestinal cancer causes countless deaths every year due to therapeutic resistance. However, whether metabolic alterations contribute to chemoresistance is not well understood. In this study, we report that fatty acid (FA) catabolism was activated in gastrointestinal cancer cells treated with oxaliplatin, which exhibited higher expression of the rate-limiting enzymes carnitine palmitoyltransferase 1B (CPT1B) and CPT2. The clinical analysis also showed that high expression of these enzymes was associated with poor oxaliplatin-based chemotherapy outcomes in patients. Furthermore, genetic or pharmacological inhibition of CPT2 with perhexiline disturbed NADPH and redox homeostasis and increased reactive oxygen species (ROS) generation and cell apoptosis in gastrointestinal cancer cells following oxaliplatin treatment. Specifically, the combination of oxaliplatin and perhexiline significantly suppressed the progression of gastrointestinal cancer in cell-based xenograft and patient-derived xenograft (PDX) models. Mechanistically, CPT2 was transcriptionally upregulated by nuclear factor of activated T cells 3 (NFATc3), which translocated to the nucleus in response to oxaliplatin treatment. In summary, our study suggests that the inhibition of CPT-mediated FA catabolism combined with conventional chemotherapy is a promising therapeutic strategy for patients with gastrointestinal cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Colorectal Neoplasms/drug therapy , Fatty Acids/metabolism , Stomach Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinogenesis/drug effects , Carcinogenesis/pathology , Carnitine O-Palmitoyltransferase/metabolism , Cell Line, Tumor , Colorectal Neoplasms/pathology , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , NADP/metabolism , NFATC Transcription Factors/metabolism , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Perhexiline/pharmacology , Perhexiline/therapeutic use , Reactive Oxygen Species , Stomach Neoplasms/pathology , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
4.
Cancer Res ; 79(23): 5907-5914, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31434645

ABSTRACT

Since Warburg's observation that most cancers exhibit elevated glycolysis, decades of research have attempted to reduce tumor glucose utilization as a therapeutic approach. Hexokinase (HK) activity is the first glycolytic enzymatic step; despite many attempts to inhibit HK activity, none has reached clinical application. Identification of HK isoforms, and recognition that most tissues express only HK1 while most tumors express HK1 and HK2, stimulated reducing HK2 activity as a therapeutic option. However, studies using HK2 shRNA and isogenic HK1+HK2- and HK1+HK2+ tumor cell pairs demonstrated that tumors expressing only HK1, while exhibiting reduced glucose consumption, progressed in vivo as well as tumors expressing both HK1 and HK2. However, HK1-HK2+ tumor subpopulations exist among many cancers. shRNA HK2 suppression in HK1-HK2+ liver cancer cells reduced xenograft tumor progression, in contrast to HK1+HK2+ cells. HK2 inhibition, and partial inhibition of both oxidative phosphorylation and fatty acid oxidation using HK2 shRNA and small-molecule drugs, prevented human liver HK1-HK2+ cancer xenograft progression. Using human multiple myeloma xenografts and mouse allogeneic models to identify potential clinical translational agents, triple therapies that include antisense HK2 oligonucleotides, metformin, and perhexiline prevent progression. These results suggest an agnostic approach for HK1-HK2+ cancers, regardless of tissue origin.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Glycolysis/drug effects , Hexokinase/antagonists & inhibitors , Hexokinase/genetics , Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Glycolysis/genetics , Hexokinase/metabolism , Humans , Liver/drug effects , Liver/metabolism , Metformin/pharmacology , Metformin/therapeutic use , Mice , Neoplasms/genetics , Neoplasms/pathology , Onium Compounds/pharmacology , Onium Compounds/therapeutic use , Oxidative Phosphorylation/drug effects , Perhexiline/pharmacology , Perhexiline/therapeutic use , RNA, Small Interfering/metabolism , Xenograft Model Antitumor Assays
5.
Eur J Heart Fail ; 18(12): 1420-1429, 2016 12.
Article in English | MEDLINE | ID: mdl-27813339

ABSTRACT

The failing heart has an increased metabolic demand and at the same time suffers from impaired energy efficiency, which is a detrimental combination. Therefore, therapies targeting the energy-deprived failing heart and rewiring cardiac metabolism are of great potential, but are lacking in daily clinical practice. Metabolic impairment in heart failure patients has been well characterized for patients with reduced ejection fraction, and is coming of age in patients with 'preserved' ejection fraction. Targeting cardiomyocyte metabolism in heart failure could complement current heart failure treatments that do improve cardiovascular haemodynamics, but not the energetic status of the heart. In this review, we discuss the hallmarks of normal cardiac metabolism, typical metabolic disturbances in heart failure, and past and present therapeutic targets that impact on cardiac metabolism.


Subject(s)
Heart Failure/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Acetyl-CoA C-Acyltransferase/antagonists & inhibitors , Cardiovascular Agents/therapeutic use , Carnitine/analogs & derivatives , Carnitine/therapeutic use , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Dichloroacetic Acid/therapeutic use , Energy Metabolism , Enzyme Inhibitors/therapeutic use , Epoxy Compounds/therapeutic use , Fatty Acids/metabolism , Glucose/metabolism , Heart Failure/drug therapy , Heart Failure/physiopathology , Humans , Oxidation-Reduction , Perhexiline/therapeutic use , Ranolazine/therapeutic use , Stroke Volume , Trimetazidine/therapeutic use
6.
Cardiovasc Drugs Ther ; 30(4): 399-405, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27106834

ABSTRACT

Approaches to the pharmacotherapy of angina pectoris have previously centred on the concept that a transient imbalance between myocardial oxygen "demand" and supply within the myocardium can best be addressed by reducing demand (for example, with ß-adrenoceptor antagonist) or by increasing availability of blood (via coronary vasomotor reactivity adjustment or coronary revascularization). However, this principle is potentially challenged by the emergence of cases of angina unsuitable for such therapies (for example because of concomitant severe systolic heart failure) and by the recognition that impaired myocardial energetics may precipitate angina in the absence of fixed or variable coronary obstruction (for example in hypertrophic cardiomyopathy). The past 20 years have seen the re-emergence of a class of anti-anginal agents which act primarily by improving efficiency of myocardial oxygen utilization, and thus can correct impaired energetics, simultaneously treating angina and heart failure symptoms. We review the principles underlying the safe use of such agents, beginning with the prototype drug perhexiline maleate, which despite complex pharmacokinetics and potential hepato- or neuro-toxicity has emerged as an attractive management option in many "complicated" cases of angina pectoris.


Subject(s)
Angina Pectoris/drug therapy , Cardiovascular Agents/therapeutic use , Myocardium/metabolism , Perhexiline/analogs & derivatives , Angina Pectoris/metabolism , Animals , Cardiovascular Agents/pharmacology , Fatty Acids/metabolism , Glucose/metabolism , Humans , Mitochondria/metabolism , Perhexiline/pharmacology , Perhexiline/therapeutic use
7.
Br J Pharmacol ; 173(12): 1911-24, 2016 06.
Article in English | MEDLINE | ID: mdl-26993743

ABSTRACT

Many conditions culminate in heart failure (HF), a multi-organ systemic syndrome with an intrinsically poor prognosis. Pharmacotherapeutic agents that correct neurohormonal dysregulation and haemodynamic instability have occupied the forefront of developments within the treatment of HF in the past. Indeed, multiple trials aimed to validate these agents in the 1980s and early 1990s, resulting in a large and robust evidence-base supporting their use clinically. An established treatment paradigm now exists for the treatment of HF with reduced ejection fraction (HFrEF), but there have been very few notable developments in recent years. HF remains a significant health concern with an increasing incidence as the population ages. We may indeed be entering the surgical era for HF treatment, but these therapies remain expensive and inaccessible to many. Newer pharmacotherapeutic agents are slowly emerging, many targeting alternative therapeutic pathways, but with mixed results. Metabolic modulation and manipulation of the nitrate/nitrite/nitric oxide pathway have shown promise and could provide the answers to fill the therapeutic gap between medical interventions and surgery, but further definitive trials are warranted. We review the significant evidence base behind the current medical treatments for HFrEF, the physiology of metabolic impairment in HF, and discuss two promising novel agents, perhexiline and nitrite.


Subject(s)
Cardiovascular Agents/therapeutic use , Heart Failure/drug therapy , Nitrites/therapeutic use , Perhexiline/therapeutic use , Heart Failure/metabolism , Humans
10.
Diab Vasc Dis Res ; 12(4): 239-48, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25941161

ABSTRACT

Type 2 diabetes mellitus escalates the risk of heart failure partly via its ability to induce a cardiomyopathic state that is independent of coronary artery disease and hypertension. Although the pathogenesis of diabetic cardiomyopathy has yet to be fully elucidated, aberrations in cardiac substrate metabolism and energetics are thought to be key drivers. These aberrations include excessive fatty acid utilisation and storage, suppressed glucose oxidation and impaired mitochondrial oxidative phosphorylation. An appreciation of how these abnormalities arise and synergise to promote adverse cardiac remodelling is critical to their effective amelioration. This review focuses on disturbances in myocardial fuel (fatty acids and glucose) flux and energetics in type 2 diabetes, how these disturbances relate to the development of diabetic cardiomyopathy and the potential therapeutic agents that could be used to correct them.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetic Cardiomyopathies/metabolism , Fatty Acids/metabolism , Glucose/metabolism , Mitochondria, Heart/metabolism , Myocardium/metabolism , Oxidative Phosphorylation , Antioxidants/therapeutic use , Cardiovascular Agents/therapeutic use , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/etiology , Energy Metabolism , Humans , Organophosphorus Compounds/therapeutic use , Oxidative Stress , Perhexiline/therapeutic use , Ranolazine/therapeutic use , Trimetazidine/therapeutic use , Ubiquinone/analogs & derivatives , Ubiquinone/therapeutic use
11.
Blood ; 125(18): 2806-14, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25784680

ABSTRACT

Oncogenic activation of NOTCH1 signaling plays a central role in the pathogenesis of T-cell acute lymphoblastic leukemia, with mutations on this signaling pathway affecting more than 60% of patients at diagnosis. However, the transcriptional regulatory circuitries driving T-cell transformation downstream of NOTCH1 remain incompletely understood. Here we identify Hairy and Enhancer of Split 1 (HES1), a transcriptional repressor controlled by NOTCH1, as a critical mediator of NOTCH1-induced leukemogenesis strictly required for tumor cell survival. Mechanistically, we demonstrate that HES1 directly downregulates the expression of BBC3, the gene encoding the PUMA BH3-only proapoptotic factor in T-cell acute lymphoblastic leukemia. Finally, we identify perhexiline, a small-molecule inhibitor of mitochondrial carnitine palmitoyltransferase-1, as a HES1-signature antagonist drug with robust antileukemic activity against NOTCH1-induced leukemias in vitro and in vivo.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation, Leukemic , Gene Targeting/methods , Homeodomain Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Animals , Antineoplastic Agents/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Gene Expression Regulation, Leukemic/drug effects , Gene Silencing , HEK293 Cells , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/physiology , Humans , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Molecular Targeted Therapy , Perhexiline/therapeutic use , Receptor, Notch1/genetics , Transcription Factor HES-1
13.
JACC Heart Fail ; 3(3): 202-11, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25650370

ABSTRACT

OBJECTIVES: The aim of this study was to determine whether short-term treatment with perhexiline improves cardiac energetics, left ventricular function, and symptoms of heart failure by altering cardiac substrate utilization. BACKGROUND: Perhexiline improves exercise capacity and left ventricular ejection fraction (LVEF) in patients with heart failure (HF). (31)P cardiac magnetic resonance spectroscopy can be used to quantify the myocardial phosphocreatine/adenosine triphosphate ratio. Because improvement of HF syndrome can improve cardiac energetics secondarily, we investigated the effects of short-term perhexiline therapy. METHODS: Patients with systolic HF of nonischemic etiology (n = 50, 62 ± 1.8 years of age, New York Heart Association functional class II to IV, LVEF: 27.0 ± 1.44%) were randomized to receive perhexiline 200 mg or placebo for 1 month in a double-blind fashion. Clinical assessment, echocardiography, and (31)P cardiac magnetic resonance spectroscopy were performed at baseline and after 1 month. A substudy of 22 patients also underwent cross-heart blood sampling at completion of the study to quantify metabolite utilization. RESULTS: Perhexiline therapy was associated with a 30% increase in the phosphocreatine/adenosine triphosphate ratio (from 1.16 ± 0.39 to 1.51 ± 0.51; p < 0.001) versus a 3% decrease with placebo (from 1.36 ± 0.31 to 1.34 ± 0.31; p = 0.37). Perhexiline therapy also led to an improvement in New York Heart Association functional class compared with placebo (p = 0.036). Short-term perhexiline therapy did not change LVEF. Cross-heart measures of cardiac substrate uptake and respiratory exchange ratio (which reflects the ratio of substrates used) did not differ between patients who received perhexiline versus placebo. CONCLUSIONS: Perhexiline improves cardiac energetics and symptom status with no evidence of altered cardiac substrate utilization. No change in LVEF is seen at this early stage. (Metabolic Manipulation in Chronic Heart Failure; NCT00841139).


Subject(s)
Cardiomyopathy, Dilated/complications , Energy Metabolism/drug effects , Heart Failure/drug therapy , Myocardium/metabolism , Perhexiline/therapeutic use , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/physiopathology , Cardiovascular Agents/therapeutic use , Double-Blind Method , Echocardiography , Female , Follow-Up Studies , Heart Failure/etiology , Heart Failure/metabolism , Humans , Male , Middle Aged , Retrospective Studies , Stroke Volume , Time Factors , Treatment Outcome , Ventricular Function, Left
14.
Mol Biosyst ; 11(2): 564-73, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25437646

ABSTRACT

Energy depletion has been highlighted as an important contributor to the pathology of hypertrophic cardiomyopathy (HCM), a common inherited cardiac disease. Pharmacological reversal of energy depletion appears an attractive approach and the use of perhexiline has been proposed as it is thought to shift myocardial metabolism from fatty acid to glucose utilisation, increasing ATP production and myocardial efficiency. We used the Mybpc3-targeted knock-in mouse model of HCM to investigate changes in the cardiac metabolome following perhexiline treatment. Echocardiography indicated that perhexiline induced partial improvement of some, but not all hypertrophic parameters after six weeks. Non-targeted metabolomics, applying ultra-high performance liquid chromatography-mass spectrometry, described a phenotypic modification of the cardiac metabolome with 272 unique metabolites showing a statistically significant change (p < 0.05). Changes in fatty acids and acyl carnitines indicate altered fatty acid transport into mitochondria, implying reduction in fatty acid beta-oxidation. Increased glucose utilisation is indirectly implied through changes in the glycolytic, glycerol, pentose phosphate, tricarboxylic acid and pantothenate pathways. Depleted reduced glutathione and increased production of NADPH suggest reduction in oxidative stress. These data delineate the metabolic changes occurring during improvement of the HCM phenotype and indicate the requirements for further targeted interventions.


Subject(s)
Cardiomyopathy, Hypertrophic/drug therapy , Cardiomyopathy, Hypertrophic/metabolism , Metabolome , Myocardium/metabolism , Perhexiline/therapeutic use , Animals , Cardiomyopathy, Hypertrophic/diagnostic imaging , Cardiomyopathy, Hypertrophic/pathology , Chromatography, High Pressure Liquid , Disease Models, Animal , Male , Mass Spectrometry , Metabolome/drug effects , Metabolomics , Mice, Inbred C57BL , Myocardium/pathology , Perhexiline/pharmacology , Phenotype , Principal Component Analysis , Ultrasonography
15.
Eur J Cardiothorac Surg ; 48(3): 354-62, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25538197

ABSTRACT

OBJECTIVES: Patients undergoing cardiac surgery require adequate myocardial protection. Manipulating myocardial metabolism may improve the extent of myocardial protection. Perhexiline has been shown to be an effective anti-anginal agent due to its metabolic modulation properties by inhibiting the uptake of free fatty acids into the mitochondrion, and thereby promoting a more efficient carbohydrate-driven myocardial metabolism. Metabolic modulation may augment myocardial protection, particularly in patients with left ventricular hypertrophy (LVH) known to have a deranged metabolic state and are at risk of poor postoperative outcomes. This study aimed to evaluate the role of perhexiline as an adjunct in myocardial protection in patients with LVH secondary to aortic stenosis (AS), undergoing an aortic valve replacement (AVR). METHODS: In a multicentre double-blind randomized controlled trial of patients with AS undergoing AVR ± coronary artery bypass graft surgery, patients were randomized to preoperative oral therapy with either perhexiline or placebo. The primary end point was incidence of inotrope use to improve haemodynamic performance due to a low cardiac output state during the first 6 h of reperfusion, judged by a blinded end points committee. Secondary outcome measures included haemodynamic measurements, electrocardiographic and biochemical markers of new myocardial injury and clinical safety outcome measures. RESULTS: The trial was halted early on the advice of the Data Safety and Monitoring Board. Sixty-two patients were randomized to perhexiline and 65 to placebo. Of these, 112 (54 perhexiline and 48 placebo) patients received the intervention, remained in the trial at the time of the operation and were analysed. Of 110 patients who achieved the primary end point, 30 patients (16 perhexiline and 14 placebo) had inotropes started appropriately; there was no difference in the incidence of inotrope usage OR of 1.65 [confidence interval (CI): 0.67-4.06] P = 0.28. There was no difference in myocardial injury as evidenced by electrocardiogram odds ratio (OR) of 0.36 (CI: 0.07-1.97) P = 0.24 or postoperative troponin release. Gross secondary outcome measures were comparable between the groups. CONCLUSIONS: Perhexiline as a metabolic modulator to enhance standard myocardial protection does not provide an additional benefit in haemodynamic performance or attenuate myocardial injury in the hypertrophied heart secondary to AS. The role of perhexiline in cardiac surgery is limited.


Subject(s)
Cardiotonic Agents/therapeutic use , Hypertrophy, Left Ventricular/therapy , Perhexiline/therapeutic use , Aged , Combined Modality Therapy , Coronary Artery Bypass/methods , Double-Blind Method , Electrocardiography , Female , Hemodynamics/drug effects , Humans , Hypertrophy, Left Ventricular/drug therapy , Hypertrophy, Left Ventricular/surgery , Male , Middle Aged
16.
Eur J Cardiothorac Surg ; 47(3): 464-72, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24948413

ABSTRACT

OBJECTIVES: Perhexiline is thought to modulate metabolism by inhibiting mitochondrial carnitine palmitoyltransferase-1, reducing fatty acid uptake and increasing carbohydrate utilization. This study assessed whether preoperative perhexiline improves markers of myocardial protection in patients undergoing coronary artery bypass graft surgery and analysed its effect on the myocardial metabolome. METHODS: In a prospective, randomized, double-blind, placebo-controlled trial, patients at two centres were randomized to receive either oral perhexiline or placebo for at least 5 days prior to surgery. The primary outcome was a low cardiac output episode in the first 6 h. All pre-specified analyses were conducted according to the intention-to-treat principle with a statistical power of 90% to detect a relative risk of 0.5 and a conventional one-sided α-value of 0.025. A subset of pre-ischaemic left ventricular biopsies was analysed using mass spectrometry-based metabolomics. RESULTS: Over a 3-year period, 286 patients were randomized, received the intervention and were included in the analysis. The incidence rate of a low cardiac output episode in the perhexiline arm was 36.7% (51/139) vs 34.7% (51/147) in the control arm [odds ratio (OR) 0.92, 95% confidence interval (CI) 0.56-1.50, P = 0.74]. Perhexiline was associated with a reduction in the cardiac index at 6 h [difference in means 0.19, 95% CI 0.07-0.31, P = 0.001] and an increase in inotropic support in the first 12 h (OR 0.55, 95% CI 0.34-0.89, P = 0.015). There were no significant differences in myocardial injury with troponin-T or electrocardiogram, reoperation, renal dysfunction or length of stay. No difference in the preischaemic left ventricular metabolism was identified between groups on metabolomics analysis. CONCLUSIONS: Preoperative perhexiline does not improve myocardial protection in patients undergoing coronary surgery and in fact reduced perioperative cardiac output, increasing the need for inotropic support. Perhexiline has no significant effect on the mass spectrometry-visible polar myocardial metabolome in vivo in humans, supporting the suggestion that it acts via a pathway that is independent of myocardial carnitine palmitoyltransferase inhibition and may explain the lack of clinical benefit observed following surgery. CLINICALTRIALSGOV ID: NCT00845364.


Subject(s)
Cardiotonic Agents/therapeutic use , Coronary Artery Bypass/methods , Coronary Vessels/surgery , Myocardial Reperfusion Injury/prevention & control , Perhexiline/therapeutic use , Aged , Cardiac Output/drug effects , Coronary Artery Bypass/adverse effects , Double-Blind Method , Female , Heart Ventricles/chemistry , Heart Ventricles/metabolism , Humans , Male , Metabolome/drug effects , Middle Aged , Myocardial Reperfusion Injury/metabolism , Placebos , Postoperative Complications , Prospective Studies
17.
Future Cardiol ; 10(6): 693-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25495811

ABSTRACT

Recently heart failure with preserved ejection fraction (HFpEF) has emerged as a huge epidemic. Increasing evidence shows the role of energy deficiency in the pathophysiology of HFpEF. In the current study, we hypothesize that the use of metabolic modulator perhexiline would correct myocardial energy deficiency and improve exercise capacity and diastolic abnormalities in patients with this syndrome.


Subject(s)
Cardiovascular Agents/therapeutic use , Heart Failure/drug therapy , Heart Failure/physiopathology , Perhexiline/therapeutic use , Stroke Volume/physiology , Clinical Protocols , Double-Blind Method , Humans , Outcome Assessment, Health Care , Syndrome
18.
Orphanet J Rare Dis ; 8: 163, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-24131551

ABSTRACT

BACKGROUND: Currently, there are no effective medical treatment options to prevent the formation of heterotopic bones in fibrodysplasia ossificans progressiva (FOP). By the drug repositioning strategy, we confirmed that perhexiline maleate (Pex) potentially ameliorates heterotopic ossification in model cells and mice. Here, we conducted a prospective study to assess the efficacy and safety of Pex in the treatment of FOP patients. METHODS: FOP patients in this open-label single-center study were treated with Pex for a total of 12 months, and followed up for 12 consecutive months after medication discontinuation. The safety of the treatment was assessed regularly by physical and blood examinations. The efficacy of Pex for preventing heterotopic ossifications was evaluated by the presence of flare-ups, measurements of serum bone markers, and changes in the total bone volume calculated by the three-dimensional computed tomography (3D-CT) images. RESULTS: Five patients with an average age of 23.4 years were enrolled. Within safe doses of Pex administration in each individual, there were no drug-induced adverse effects during the medication phase. Three patients showed no intense inflammatory reactions during the study period, while two patients had acute flare-ups around the hip joint without evidence of trauma during the medication phase. In addition, one of them became progressively incapable of opening her mouth over the discontinuation phase. Serum levels of alkaline phosphatase (ALP) and bone specific ALP (BAP) were significantly and synchronously increased with the occurrence of flare-ups. Volumetric 3D-CT analysis demonstrated a significant increase in the total bone volume of Case 2 (378 cm(3)) and Case 3 (833 cm(3)) during the two-year study period. CONCLUSIONS: We could not prove the efficacy of oral Pex administration in the prevention of heterotopic ossifications in FOP. Serum levels of ALP and BAP appear to be promising biomarkers for monitoring the development of ectopic ossifications and efficacy of the therapy. Quantification of change in the total bone volume by whole body CT scanning could be a reliable evaluation tool for disease progression in forthcoming clinical trials of FOP.


Subject(s)
Myositis Ossificans/drug therapy , Perhexiline/analogs & derivatives , Adolescent , Adult , Alkaline Phosphatase/blood , Female , Humans , Male , Myositis Ossificans/blood , Myositis Ossificans/diagnostic imaging , Osteocalcin/blood , Perhexiline/therapeutic use , Radiography , Young Adult
19.
Ther Drug Monit ; 34(2): 227-31, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22322401

ABSTRACT

BACKGROUND: Concomitant treatment with amiodarone and perhsexiline has been considered to be relatively contraindicated because of the hypothetical risk of potentiated adverse effects mediated by additive inhibition of carnitine palmitoyl transferase 1. AIM: To study the prevalence of adverse effects associated with the concomitant use of perhexiline and amiodarone. METHODS: A retrospective analysis of a single hospital database of patients receiving perhexiline and amiodarone between July 2009 and April 2011. Files were reviewed for short- and long-term adverse effects requiring drug cessation. Glucose concentration, gamma glutamyl transferase activity. and perhexiline blood concentrations were recorded. RESULTS: We identified 26 patients concomitantly treated with perhexiline and amiodarone, 20 on a long-term basis. In 6 cases, amiodarone was introduced on top of preceding perhexiline. In none of the cases were drugs stopped because of adverse effects. Although blood glucose concentrations fell significantly 48 hours postadmission to hospital, this seems to reflect the resolution of "admission hyperglycemia" rather than onset of hypoglycemia; the latter was rare (5 patients), mild, and clinically silent. In 4 patients, gamma glutamyl transferase approximately doubled. CONCLUSIONS: Traditionally, concomitant treatment with amiodarone and perhexiline has been considered to be relatively contraindicated on the basis of the theoretical potential for synergistic toxicity. This cohort of 26 patients received this concomitant treatment without any excess of major adverse reactions. Our findings suggest that concomitant treatment with perhexiline and amiodarone may be safe in the setting of (1) previous tolerance of either agent, and (2) titration of plasma perhexiline concentrations to guide therapy.


Subject(s)
Amiodarone/adverse effects , Perhexiline/analogs & derivatives , Vasodilator Agents/adverse effects , Adult , Aged , Aged, 80 and over , Amiodarone/administration & dosage , Amiodarone/therapeutic use , Blood Glucose/drug effects , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Databases, Factual , Drug Interactions , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Perhexiline/administration & dosage , Perhexiline/adverse effects , Perhexiline/therapeutic use , Prevalence , Retrospective Studies , Vasodilator Agents/administration & dosage , Vasodilator Agents/therapeutic use , gamma-Glutamyltransferase/drug effects , gamma-Glutamyltransferase/metabolism
20.
Trials ; 12: 140, 2011 Jun 06.
Article in English | MEDLINE | ID: mdl-21645332

ABSTRACT

BACKGROUND: Heart failure is a major cause of morbidity and mortality in society. Current medical therapy centres on neurohormonal modulation with angiotensin converting enzyme inhibitors and ß-blockers. There is growing evidence for the use of metabolic manipulating agents as adjunctive therapy in patients with heart failure. We aim to determine the effect of perhexiline on cardiac energetics and alterations in substrate utilisation in patients with non-ischaemic dilated cardiomyopathy. METHODS: A multi-centre, prospective, randomised double-blind, placebo-controlled trial of 50 subjects with non-ischaemic dilated cardiomyopathy recruited from University Hospital Birmingham NHS Foundation Trust and Cardiff and Vale NHS Trust. Baseline investigations include magnetic resonance spectroscopy to assess cardiac energetic status, echocardiography to assess left ventricular function and assessment of symptomatic status. Subjects are then randomised to receive 200 mg perhexiline maleate or placebo daily for 4 weeks with serum drug level monitoring. All baseline investigations will be repeated at the end of the treatment period. A subgroup of patients will undergo invasive investigations with right and left heart catheterisation to calculate respiratory quotient, and mechanical efficiency. The primary endpoint is an improvement in the phosphocreatine to adenosine triphosphate ratio at 4 weeks. Secondary end points are: i) respiratory quotient; ii) mechanical efficiency; iii) change in left ventricular (LV) function. TRIAL REGISTRATION: ClinicalTrials.gov: NCT00841139 ISRCTN: ISRCTN72887836.


Subject(s)
Cardiomyopathy, Dilated/drug therapy , Cardiovascular Agents/therapeutic use , Energy Metabolism/drug effects , Heart Failure/drug therapy , Myocardium/metabolism , Perhexiline/therapeutic use , Research Design , Adenosine Triphosphate/metabolism , Cardiac Catheterization , Cardiomyopathy, Dilated/complications , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/physiopathology , Cardiovascular Agents/blood , Double-Blind Method , Drug Monitoring , Echocardiography, Doppler , Heart Failure/diagnosis , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/physiopathology , Humans , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Perhexiline/blood , Phosphocreatine/metabolism , Placebo Effect , Prospective Studies , Recovery of Function , Stroke Volume/drug effects , Time Factors , Treatment Outcome , United Kingdom , Ventricular Function, Left/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...