Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Clin Transl Sci ; 17(2): e13687, 2024 02.
Article En | MEDLINE | ID: mdl-38362827

Co-administration of clesacostat (acetyl-CoA carboxylase inhibitor, PF-05221304) and ervogastat (diacylglycerol O-acyltransferase inhibitor, PF-06865571) in laboratory models improved non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH) end points and mitigated clesacostat-induced elevations in circulating triglycerides. Clesacostat is cleared via organic anion-transporting polypeptide-mediated hepatic uptake and cytochrome P450 family 3A (CYP3A); in vitro clesacostat is identified as a potential CYP3A time-dependent inactivator. In vitro ervogastat is identified as a substrate and potential inducer of CYP3A. Prior to longer-term efficacy trials in participants with NAFLD, safety and pharmacokinetics (PK) were evaluated in a phase I, non-randomized, open-label, fixed-sequence trial in healthy participants. In Cohort 1, participants (n = 7) received clesacostat 15 mg twice daily (b.i.d.) alone (Days 1-7) and co-administered with ervogastat 300 mg b.i.d. (Days 8-14). Mean systemic clesacostat exposures, when co-administered with ervogastat, decreased by 12% and 19%, based on maximum plasma drug concentration and area under the plasma drug concentration-time curve during the dosing interval, respectively. In Cohort 2, participants (n = 9) received ervogastat 300 mg b.i.d. alone (Days 1-7) and co-administered with clesacostat 15 mg b.i.d. (Days 8-14). There were no meaningful differences in systemic ervogastat exposures when administered alone or with clesacostat. Clesacostat 15 mg b.i.d. and ervogastat 300 mg b.i.d. co-administration was overall safe and well tolerated in healthy participants. Cumulative safety and no clinically meaningful PK drug interactions observed in this study supported co-administration of these two novel agents in additional studies exploring efficacy and safety in the management of NAFLD.


Non-alcoholic Fatty Liver Disease , Pyridines , Adult , Humans , Healthy Volunteers , Cytochrome P-450 CYP3A , Enzyme Inhibitors/adverse effects , Drug Interactions , Diacylglycerol O-Acyltransferase
2.
J Hepatol ; 80(2): 362-377, 2024 Feb.
Article En | MEDLINE | ID: mdl-37977245

The hepatic accumulation of excess triglycerides is a seminal event in the initiation and progression of non-alcoholic fatty liver disease (NAFLD). Hepatic steatosis occurs when the hepatic accrual of fatty acids from the plasma and de novo lipogenesis (DNL) is no longer balanced by rates of fatty acid oxidation and secretion of very low-density lipoprotein-triglycerides. Accumulating data indicate that increased rates of DNL are central to the development of hepatic steatosis in NAFLD. Whereas the main drivers in NAFLD are transcriptional, owing to both hyperinsulinemia and hyperglycaemia, the effectors of DNL are a series of well-characterised enzymes. Several have proven amenable to pharmacologic inhibition or oligonucleotide-mediated knockdown, with lead compounds showing liver fat-lowering efficacy in phase II clinical trials. In humans with NAFLD, percent reductions in liver fat have closely mirrored percent inhibition of DNL, thereby affirming the critical contributions of DNL to NAFLD pathogenesis. The safety profiles of these compounds have so far been encouraging. It is anticipated that inhibitors of DNL, when administered alone or in combination with other therapeutic agents, will become important agents in the management of human NAFLD.


Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/metabolism , Lipogenesis/physiology , Liver/pathology , Lipid Metabolism , Triglycerides/metabolism
3.
Diabetes Obes Metab ; 25(4): 992-1001, 2023 04.
Article En | MEDLINE | ID: mdl-36515213

AIM: To assess the safety, tolerability and pharmacodynamics (PD) of the ketohexokinase inhibitor PF-06835919 in participants with non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes (T2D). MATERIALS AND METHODS: This double-blind, placebo-controlled, parallel-group study enrolled adults with NAFLD (≥ 8% whole liver fat [WLF] using MRI proton density fat fraction [MRI-PDFF]) and T2D on stable doses of metformin (≥ 500 mg/day). Participants received once-daily placebo, PF-06835919 150 or 300 mg for 16 weeks. Randomization (1:1:1) was via an interactive response technology system. Endpoints included percentage change from baseline (CFB) in WLF using MRI-PDFF (primary endpoint) and CFB in HbA1c (co-primary endpoint) at 16 weeks, PD, safety and tolerability. RESULTS: Among 164 participants randomized and treated, 145 completed the treatment (placebo, n = 50; PF-06835919 150 mg, n = 46; PF-06835919 300 mg, n = 49). At week 16, least squares mean (90% confidence interval) percentage CFB in WLF was -5.26% (-12.86%, 2.99%), -17.05% (-24.01%, -9.46%) and -19.13% (-25.51%, -12.20%) in the placebo, PF-06835919 150-mg and 300-mg groups, respectively (PF-06835919 300-mg group vs. placebo, P = .0288). Modest numerical reductions in HbA1c were observed in all groups that did not reach statistical significance. Treatment-emergent adverse event incidence was similar across groups (40.7%, 45.5% and 32.7% in the placebo, PF-06835919 150-mg and 300-mg groups, respectively), with no apparent dose-related trend. CONCLUSIONS: PF-06835919 administration over 16 weeks was generally safe and well tolerated and resulted in reductions in WLF in participants with NAFLD and T2D.


Diabetes Mellitus, Type 2 , Metformin , Non-alcoholic Fatty Liver Disease , Adult , Humans , Diabetes Mellitus, Type 2/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Glycated Hemoglobin , Metformin/therapeutic use , Double-Blind Method , Treatment Outcome
4.
Nat Med ; 27(10): 1836-1848, 2021 10.
Article En | MEDLINE | ID: mdl-34635855

Alterations in lipid metabolism might contribute to the pathogenesis of non-alcoholic fatty liver disease (NAFLD). However, no pharmacological agents are currently approved in the United States or the European Union for the treatment of NAFLD. Two parallel phase 2a studies investigated the effects of liver-directed ACC1/2 inhibition in adults with NAFLD. The first study ( NCT03248882 ) examined the effects of monotherapy with a novel ACC1/2 inhibitor, PF-05221304 (2, 10, 25 and 50 mg once daily (QD)), versus placebo at 16 weeks of treatment; the second study ( NCT03776175 ) investigated the effects of PF-05221304 (15 mg twice daily (BID)) co-administered with a DGAT2 inhibitor, PF-06865571 (300 mg BID), versus placebo after 6 weeks of treatment. The primary endpoint in both studies was percent change from baseline in liver fat assessed by magnetic resonance imaging-proton density fat fraction. Dose-dependent reductions in liver fat reached 50-65% with PF-05221304 monotherapy doses ≥10 mg QD; least squares mean (LSM) 80% confidence interval (CI) was -7.2 (-13.9, 0.0), -17.1 (-22.7, -11.1), -49.9 (-53.3, -46.2), -55.9 (-59.0, -52.4) and -64.8 (-67.5, -62.0) with 16 weeks placebo and PF-05221304 2, 10, 25 and 50 mg QD, respectively. The overall incidence of adverse events (AEs) did not increase with increasing PF-05221304 dose, except for a dose-dependent elevation in serum triglycerides (a known consequence of hepatic acetyl-coenzyme A carboxylase (ACC) inhibition) in 23/305 (8%) patients, leading to withdrawal in 13/305 (4%), and a dose-dependent elevation in other serum lipids. Co-administration of PF-05221304 and PF-06865571 lowered liver fat compared to placebo (placebo-adjusted LSM (90% CI) -44.6% (-54.8, -32.2)). Placebo-adjusted LSM (90% CI) reduction in liver fat was -44.5% (-55.0, -31.7) and -35.4% (-47.4, -20.7) after 6 weeks with PF-05221304 or PF-06865571 alone. AEs were reported for 10/28 (36%) patients after co-administered PF-05221304 and PF-06865571, with no discontinuations due to AEs, and the ACC inhibitor-mediated effect on serum triglycerides was mitigated, suggesting that PF-05221304 and PF-06865571 co-administration has the potential to address some of the limitations of ACC inhibition alone.


Acetyl-CoA Carboxylase/antagonists & inhibitors , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Enzyme Inhibitors/administration & dosage , Liver/enzymology , Non-alcoholic Fatty Liver Disease/drug therapy , Acetyl-CoA Carboxylase/genetics , Diacylglycerol O-Acyltransferase/genetics , Double-Blind Method , Drug Synergism , Enzyme Inhibitors/adverse effects , Female , Humans , Lipid Metabolism/drug effects , Liver/drug effects , Liver/ultrastructure , Magnetic Resonance Imaging , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Placebos
5.
Mol Metab ; 48: 101196, 2021 06.
Article En | MEDLINE | ID: mdl-33667726

OBJECTIVE: Recent studies suggest that excess dietary fructose contributes to metabolic dysfunction by promoting insulin resistance, de novo lipogenesis (DNL), and hepatic steatosis, thereby increasing the risk of obesity, type 2 diabetes (T2D), non-alcoholic steatohepatitis (NASH), and related comorbidities. Whether this metabolic dysfunction is driven by the excess dietary calories contained in fructose or whether fructose catabolism itself is uniquely pathogenic remains controversial. We sought to test whether a small molecule inhibitor of the primary fructose metabolizing enzyme ketohexokinase (KHK) can ameliorate the metabolic effects of fructose. METHODS: The KHK inhibitor PF-06835919 was used to block fructose metabolism in primary hepatocytes and Sprague Dawley rats fed either a high-fructose diet (30% fructose kcal/g) or a diet reflecting the average macronutrient dietary content of an American diet (AD) (7.5% fructose kcal/g). The effects of fructose consumption and KHK inhibition on hepatic steatosis, insulin resistance, and hyperlipidemia were evaluated, along with the activation of DNL and the enzymes that regulate lipid synthesis. A metabolomic analysis was performed to confirm KHK inhibition and understand metabolite changes in response to fructose metabolism in vitro and in vivo. Additionally, the effects of administering a single ascending dose of PF-06835919 on fructose metabolism markers in healthy human study participants were assessed in a randomized placebo-controlled phase 1 study. RESULTS: Inhibition of KHK in rats prevented hyperinsulinemia and hypertriglyceridemia from fructose feeding. Supraphysiologic levels of dietary fructose were not necessary to cause metabolic dysfunction as rats fed the American diet developed hyperinsulinemia, hypertriglyceridemia, and hepatic steatosis, which were all reversed by KHK inhibition. Reversal of the metabolic effects of fructose coincided with reductions in DNL and inactivation of the lipogenic transcription factor carbohydrate response element-binding protein (ChREBP). We report that administering single oral doses of PF-06835919 was safe and well tolerated in healthy study participants and dose-dependently increased plasma fructose indicative of KHK inhibition. CONCLUSIONS: Fructose consumption in rats promoted features of metabolic dysfunction seen in metabolic diseases such as T2D and NASH, including insulin resistance, hypertriglyceridemia, and hepatic steatosis, which were reversed by KHK inhibition.


Enzyme Inhibitors/administration & dosage , Fructokinases/antagonists & inhibitors , Fructose/adverse effects , Hypertriglyceridemia/etiology , Hypertriglyceridemia/prevention & control , Metabolic Syndrome/etiology , Metabolic Syndrome/prevention & control , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/prevention & control , Adult , Animals , Cells, Cultured , Cohort Studies , Diet, Carbohydrate Loading/adverse effects , Fructose/administration & dosage , Fructose/metabolism , Healthy Volunteers , Hepatocytes/metabolism , Humans , Male , Middle Aged , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Treatment Outcome
6.
Toxicol Sci ; 179(2): 183-194, 2021 01 28.
Article En | MEDLINE | ID: mdl-33247737

Acetyl-CoA carboxylase (ACC) is an enzyme within the de novo lipogenesis (DNL) pathway and plays a role in regulating lipid metabolism. Pharmacologic ACC inhibition has been an area of interest for multiple potential indications including oncology, acne vulgaris, metabolic diseases such as type 2 diabetes mellitus, and nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. A critical role for ACC in de novo synthesis of long-chain fatty acids during fetal development has been demonstrated in studies in mice lacking Acc1, where the absence of Acc1 results in early embryonic lethality. Following positive predictions of developmental toxicity in the alternative in vitro assays (positive in murine embryonic stem cell [mESC] assay and rat whole embryo culture, but negative in zebrafish), developmental toxicity (growth retardation and dysmorphogenesis associated with disrupted midline fusion) was observed with the oral administration of the dual ACC1 and 2 inhibitors, PF-05175157, in Sprague Dawley rats and New Zealand White rabbits. The results of these studies are presented here to make comparisons across the assays, as well as mechanistic insights from the mESC assay demonstrating high ACC expression in the mESC and that ACC-induced developmental toxicity can be rescued with palmitic acid providing supportive evidence for DNL pathway inhibition as the underlying mechanism. Ultimately, while the battery of alternative approaches and weight-of-evidence case were useful for hazard identification, the embryo-fetal development studies were necessary to inform the risk assessment on the adverse fetal response, as malformations and/or embryo-fetal lethality were limited to doses that caused near-complete inhibition of DNL.


Acetyl-CoA Carboxylase , Diabetes Mellitus, Type 2 , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Animals , Lipogenesis , Mice , Rabbits , Rats , Rats, Sprague-Dawley , Zebrafish/metabolism
7.
Nat Metab ; 2(10): 1163-1178, 2020 10.
Article En | MEDLINE | ID: mdl-32929234

Acetyl-CoA carboxylase (ACC) catalyses the first step of de novo lipogenesis (DNL). Pharmacologic inhibition of ACC has been of interest for therapeutic intervention in a wide range of diseases. We demonstrate here that ACC and DNL are essential for platelet production in humans and monkeys, but in not rodents or dogs. During clinical evaluation of a systemically distributed ACC inhibitor, unexpected dose-dependent reductions in platelet count were observed. While platelet count reductions were not observed in rat and dog toxicology studies, subsequent studies in cynomolgus monkeys recapitulated these platelet count reductions with a similar concentration response to that in humans. These studies, along with ex vivo human megakaryocyte maturation studies, demonstrate that platelet lowering is a consequence of DNL inhibition likely to result in impaired megakaryocyte demarcation membrane formation. These observations demonstrate that while DNL is a minor quantitative contributor to global lipid balance in humans, DNL is essential to specific lipid pools of physiological importance.


Blood Platelets , Lipogenesis/physiology , Acetyl-CoA Carboxylase/antagonists & inhibitors , Acetyl-CoA Carboxylase/metabolism , Animals , Diabetes Mellitus, Type 2/drug therapy , Dogs , Dose-Response Relationship, Drug , Double-Blind Method , Enzyme Inhibitors/pharmacology , Gene Expression/drug effects , Humans , Lipid Metabolism , Macaca fascicularis , Megakaryocytes/physiology , Platelet Count , Rats
8.
J Med Chem ; 63(19): 10879-10896, 2020 10 08.
Article En | MEDLINE | ID: mdl-32809824

Preclinical and clinical data suggest that acetyl-CoA carboxylase (ACC) inhibitors have the potential to rebalance disordered lipid metabolism, leading to improvements in nonalcoholic steatohepatitis (NASH). Consistent with these observations, first-in-human clinical trials with our ACC inhibitor PF-05175157 led to robust reduction of de novo lipogenesis (DNL), albeit with concomitant reductions in platelet count, which were attributed to the inhibition of fatty acid synthesis within bone marrow. Herein, we describe the design, synthesis, and evaluation of carboxylic acid-based ACC inhibitors with organic anion transporting polypeptide (OATP) substrate properties, which facilitated selective distribution of the compounds at the therapeutic site of action (liver) relative to the periphery. These efforts led to the discovery of clinical candidate PF-05221304 (12), which selectively inhibits liver DNL in animals, while demonstrating considerable safety margins against platelet reduction in a nonhuman primate model.


Acetyl-CoA Carboxylase/antagonists & inhibitors , Drug Delivery Systems , Enzyme Inhibitors/pharmacology , Liver/drug effects , Acetyl-CoA Carboxylase/metabolism , Animals , Enzyme Inhibitors/therapeutic use , Humans , Lipogenesis , Non-alcoholic Fatty Liver Disease/drug therapy , Substrate Specificity
9.
Cell Mol Gastroenterol Hepatol ; 10(4): 829-851, 2020.
Article En | MEDLINE | ID: mdl-32526482

BACKGROUND & AIMS: Disordered metabolism, steatosis, hepatic inflammation, and fibrosis contribute to the pathogenesis of nonalcoholic steatohepatitis (NASH). Acetyl-CoA carboxylase (ACC) catalyzes the first committed step in de novo lipogenesis (DNL) and modulates mitochondrial fatty acid oxidation. Increased hepatic DNL flux and reduced fatty acid oxidation are hypothesized to contribute to steatosis. Some proinflammatory cells also show increased dependency on DNL, suggesting that ACC may regulate aspects of the inflammatory response in NASH. PF-05221304 is an orally bioavailable, liver-directed ACC1/2 inhibitor. The present studies sought to evaluate the effects of PF-05221304 on NASH pathogenic factors in experimental model systems. METHODS: The effects of PF-05221304 on lipid metabolism, steatosis, inflammation, and fibrogenesis were investigated in both primary human-derived in vitro systems and in vivo rodent models. RESULTS: PF-05221304 inhibited DNL, stimulated fatty acid oxidation, and reduced triglyceride accumulation in primary human hepatocytes, and reduced DNL and steatosis in Western diet-fed rats in vivo, showing the potential to reduce hepatic lipid accumulation and potentially lipotoxicity. PF-05221304 blocked polarization of human T cells to proinflammatory but not anti-inflammatory T cells, and suppressed activation of primary human stellate cells to myofibroblasts in vitro, showing direct effects on inflammation and fibrogenesis. Consistent with these observations, PF-05221304 also reduced markers of inflammation and fibrosis in the diethylnitrosamine chemical-induced liver injury model and the choline-deficient, high-fat-fed rat model. CONCLUSIONS: The liver-directed dual ACC1/ACC2 inhibitor directly improved multiple nonalcoholic fatty liver disease/NASH pathogenic factors including steatosis, inflammation, and fibrosis in both human-derived in vitro systems and rat models.


Acetyl-CoA Carboxylase/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Acetyl-CoA Carboxylase/metabolism , Animals , Humans , Lipogenesis/drug effects , Liver/metabolism , Liver/pathology , Male , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Rats, Sprague-Dawley
10.
Clin Pharmacol Drug Dev ; 9(4): 514-526, 2020 05.
Article En | MEDLINE | ID: mdl-32065514

PF-05221304 is a liver-targeted inhibitor of acetyl-CoA carboxylase, an enzyme that catalyzes the first committed step in de novo lipogenesis (DNL). This first-in-human study investigated safety/tolerability and pharmacokinetics of single and multiple ascending oral PF-05221304 doses, and fructose-stimulated DNL inhibition with repeated oral doses. Healthy subjects (n = 96) received single (1-240 mg) or repeated (2-200 mg daily) doses for 14 days or single 100-mg doses with and without food. PF-05221304 was well tolerated at all doses. Repeated PF-05221304 doses inhibited hepatic DNL in a dose-dependent manner, with near-complete inhibition seen at higher doses. With doses yielding ≥90% DNL inhibition, asymptomatic increases in fasting/postprandial serum triglyceride levels (≥40 mg/day) and declines in platelet count (≥60 mg/day) occurred; these were not observed at ≤80% DNL inhibition. Steady-state pharmacokinetics generally increased dose-proportionally, with a half-life of 14-18 hours and a minimal food effect on plasma exposure. The observed safety and tolerability, pharmacokinetics, and pharmacodynamics support the continued evaluation of PF-05221304 for the treatment of nonalcoholic steatohepatitis.


Acetyl-CoA Carboxylase/antagonists & inhibitors , Enzyme Inhibitors/administration & dosage , Food-Drug Interactions , Administration, Oral , Adult , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Female , Fructose/administration & dosage , Half-Life , Humans , Liver/metabolism , Male , Middle Aged , Young Adult
11.
Metabolism ; 97: 68-80, 2019 08.
Article En | MEDLINE | ID: mdl-31132381

PURPOSE: Exercise is recommended in addition to pharmacotherapies for the management of type 2 diabetes, but metformin and exercise training may have non-additive or even inhibitory effects on exercise-induced improvements in glycemic control and exercise capacity. The objectives of this report were to determine if co-treatment with a sodium-glucose cotransporter-2 inhibitor and exercise could (1) further improve glycemic control when compared to either monotherapy and (2) not worsen exercise capacity when compared to exercise alone. METHODS: A rodent model of type 2 diabetes (30 mg/kg streptozotocin and high-fat feeding in male Sprague-Dawley rats) was used to assess 12 weeks of co-treatment with a sodium-glucose cotransporter 2 inhibitor (SGLT2i) and exercise (EX; treadmill running) on glycemic control and exercise capacity. Animals were randomized to the following conditions (n = 7-10/group): vehicle (0.5% methyl cellulose) sedentary (VEH SED), VEH EX, canagliflozin (3 mg kg-1 d-1) SED (SGLT2i SED), or SGLT2i EX. RESULTS: Both EX and SGLT2i independently improved indices of glycemic control. The combination of SGLT2i and EX further improved glucose tolerance (glucose area under the curve 1109 ±â€¯51 vs 1427 ±â€¯82 mmol/ L 120 min-1 for SGLT2i EX vs. SGLT2i SED, respectively; p < 0.05) and insulin responses (insulin area under the curve 24,524 ±â€¯4126 vs. 41,208 ±â€¯2714 pmol L-1 120 min-1 for SGLT2i EX vs. VEH EX, respectively; p < 0.05) during an oral glucose tolerance test. Only the combination of SGLT2i EX lowered body weight compared to VEH SED (p < 0.01). SGLT2i caused several metabolic adaptations including increased ketone production and a greater reliance on fat as a source of energy during normal cage activity. Interestingly, animals that were given the SGLT2i and underwent exercise training (SGLT2i EX) had better submaximal exercise capacity than EX alone, as indicated by distance run prior to fatigue (882 ±â€¯183 vs.433 ±â€¯33 m for SGLT2i EX and VEH EX, respectively; p < 0.01), and this was accompanied by a greater reliance on fat as an energy source during exercise (p < 0.01). CONCLUSIONS: If these findings with the combination of SGLT2i and exercise translate to humans, they will have important clinical health implications.


Blood Glucose/drug effects , Diabetes Mellitus, Type 2/metabolism , Exercise Tolerance/drug effects , Physical Conditioning, Animal/physiology , Rodentia/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2/metabolism , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Exercise Tolerance/physiology , Glucose/metabolism , Glucose Tolerance Test/methods , Insulin/metabolism , Male , Metformin/pharmacology , Rats , Rats, Sprague-Dawley
12.
Sci Transl Med ; 11(492)2019 05 15.
Article En | MEDLINE | ID: mdl-31092695

Sebum plays important physiological roles in human skin. Excess sebum production contributes to the pathogenesis of acne vulgaris, and suppression of sebum production reduces acne incidence and severity. We demonstrate that sebum production in humans depends on local flux through the de novo lipogenesis (DNL) pathway within the sebocyte. About 80 to 85% of sebum palmitate (16:0) and sapienate (16:1n10) were derived from DNL, based on stable isotope labeling, much higher than the contribution of DNL to triglyceride palmitate in circulation (~20%), indicating a minor contribution by nonskin sources to sebum lipids. This dependence on local sebocyte DNL was not recapitulated in two widely used animal models of sebum production, Syrian hamsters and Göttingen minipigs. Confirming the importance of DNL for human sebum production, an acetyl-CoA carboxylase inhibitor, ACCi-1, dose-dependently suppressed DNL and blocked synthesis of fatty acids, triglycerides, and wax esters but not free sterols in human sebocytes in vitro. ACCi-1 dose-dependently suppressed facial sebum excretion by ~50% (placebo adjusted) in human individuals dosed orally for 2 weeks. Sebum triglycerides, wax esters, and free fatty acids were suppressed by ~66%, whereas non-DNL-dependent lipid species, cholesterol, and squalene were not reduced, confirming selective modulation of DNL-dependent lipids. Last, individuals with acne vulgaris exhibited increased sebum production rates relative to individuals with normal skin, with >80% of palmitate and sapienate derived from DNL. These findings highlight the importance of local sebocyte DNL for human skin sebaceous gland biology and illuminate a potentially exploitable therapeutic target for the treatment of acne vulgaris.


Acetyl-CoA Carboxylase/antagonists & inhibitors , Acne Vulgaris/enzymology , Enzyme Inhibitors/pharmacology , Lipogenesis , Sebum/metabolism , Acetyl-CoA Carboxylase/metabolism , Adolescent , Adult , Animals , Cells, Cultured , Cricetinae , Enzyme Inhibitors/chemistry , Female , Humans , Lipogenesis/drug effects , Male , Malonyl Coenzyme A/metabolism , Middle Aged , Rats, Wistar , Sebaceous Glands/drug effects , Sebaceous Glands/metabolism , Sebaceous Glands/pathology , Sebum/drug effects , Swine , Swine, Miniature , Triglycerides/biosynthesis , Young Adult
13.
Emerg Microbes Infect ; 8(1): 624-636, 2019.
Article En | MEDLINE | ID: mdl-30999821

Flaviviruses are (re)-emerging RNA viruses strictly dependent on lipid metabolism for infection. In the search for host targeting antivirals, we explored the effect of pharmacological modulation of fatty acid metabolism during flavivirus infection. Considering the central role of acetyl-Coenzyme A carboxylase (ACC) on fatty acid metabolism, we analyzed the effect of three small-molecule ACC inhibitors (PF-05175157, PF-05206574, and PF-06256254) on the infection of medically relevant flaviviruses, namely West Nile virus (WNV), dengue virus, and Zika virus. Treatment with these compounds inhibited the multiplication of the three viruses in cultured cells. PF-05175157 induced a reduction of the viral load in serum and kidney in WNV-infected mice, unveiling its therapeutic potential for the treatment of chronic kidney disease associated with persistent WNV infection. This study constitutes a proof of concept of the reliability of ACC inhibitors to become viable antiviral candidates. These results support the repositioning of metabolic inhibitors as broad-spectrum antivirals.


Acetyl-CoA Carboxylase/antagonists & inhibitors , Dengue Virus/physiology , Dengue/enzymology , Enzyme Inhibitors/administration & dosage , West Nile Fever/enzymology , West Nile virus/physiology , Zika Virus Infection/enzymology , Zika Virus/physiology , Acetyl-CoA Carboxylase/metabolism , Animals , Antiviral Agents/administration & dosage , Dengue/drug therapy , Dengue/virology , Dengue Virus/drug effects , Dengue Virus/genetics , Disease Models, Animal , Female , Humans , Male , Mice , Virus Replication/drug effects , West Nile Fever/drug therapy , West Nile Fever/virology , West Nile virus/drug effects , West Nile virus/genetics , Zika Virus/drug effects , Zika Virus/genetics , Zika Virus Infection/drug therapy , Zika Virus Infection/virology
14.
Cell Mol Gastroenterol Hepatol ; 8(2): 247-267, 2019.
Article En | MEDLINE | ID: mdl-31004828

The prevalence and diagnosis of nonalcoholic fatty liver disease (NAFLD) is on the rise worldwide and currently has no FDA-approved pharmacotherapy. The increase in disease burden of NAFLD and a more severe form of this progressive liver disease, nonalcoholic steatohepatitis (NASH), largely mirrors the increase in obesity and type 2 diabetes (T2D) and reflects the hepatic manifestation of an altered metabolic state. Indeed, metabolic syndrome, defined as a constellation of obesity, insulin resistance, hyperglycemia, dyslipidemia and hypertension, is the major risk factor predisposing the NAFLD and NASH. There are multiple potential pharmacologic strategies to rebalance aspects of disordered metabolism in NAFLD. These include therapies aimed at reducing hepatic steatosis by directly modulating lipid metabolism within the liver, inhibiting fructose metabolism, altering delivery of free fatty acids from the adipose to the liver by targeting insulin resistance and/or adipose metabolism, modulating glycemia, and altering pleiotropic metabolic pathways simultaneously. Emerging data from human genetics also supports a role for metabolic drivers in NAFLD and risk for progression to NASH. In this review, we highlight the prominent metabolic drivers of NAFLD pathogenesis and discuss the major metabolic targets of NASH pharmacotherapy.


Non-alcoholic Fatty Liver Disease/metabolism , Animals , Diabetes Mellitus, Type 2 , Humans , Metabolic Syndrome , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Obesity
15.
PLoS One ; 12(1): e0169566, 2017.
Article En | MEDLINE | ID: mdl-28081256

Tumor cell proliferation and migration processes are regulated by multiple metabolic pathways including glycolysis and de novo lipogenesis. Since acetyl-CoA carboxylase (ACC) is at the junction of lipids synthesis and oxidative metabolic pathways, we investigated whether use of a dual ACC inhibitor would provide a potential therapy against certain lipogenic cancers. The impact of dual ACC1/ACC2 inhibition was investigated using a dual ACC1/ACC2 inhibitor as well as dual siRNA knock down on the cellular viability and metabolism of two glioblastoma multiform cancer cell lines, U87 and a more aggressive form, U87 EGFRvIII. We first demonstrated that while ACCi inhibited DNL in both cell lines, ACCi preferentially blunted the U87 EGFRvIII cellular proliferation capacity. Metabolically, chronic treatment with ACCi significantly upregulated U87 EGFRvIII cellular respiration and extracellular acidification rate, a marker of glycolytic activity, but impaired mitochondrial health by reducing maximal respiration and decreasing mitochondrial ATP production efficiency. Moreover, ACCi treatment altered the cellular lipids content and increased apoptotic caspase activity in U87 EGFRvIII cells. Collectively these data indicate that ACC inhibition, by reducing DNL and increasing cellular metabolic rate, may have therapeutic utility for the suppression of lipogenic tumor growth and warrants further investigation.


Acetyl-CoA Carboxylase/antagonists & inhibitors , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , ErbB Receptors , Glioblastoma/drug therapy , Glioblastoma/enzymology , Lipogenesis/drug effects , Neoplasm Proteins/antagonists & inhibitors , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Lipogenesis/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oxygen Consumption/drug effects , Oxygen Consumption/genetics
16.
Bioorg Med Chem Lett ; 25(22): 5352-6, 2015 Nov 15.
Article En | MEDLINE | ID: mdl-26411795

A novel series of spirocyclic-diamine based, isoform non-selective inhibitors of acetyl-CoA carboxylase (ACC) is described. These spirodiamine derivatives were discovered by design of a library to mimic the structural rigidity and hydrogen-bonding pattern observed in the co-crystal structure of spirochromanone inhibitor I. The lead compound 3.5.1 inhibited de novo lipogenesis in rat hepatocytes, with an IC50 of 0.30 µM.


Acetyl Coenzyme A/metabolism , Acetyl-CoA Carboxylase/antagonists & inhibitors , Drug Discovery , Hepatocytes/drug effects , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Animals , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hepatocytes/enzymology , Humans , Inhibitory Concentration 50 , Models, Biological , Molecular Structure , Rats , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
17.
J Med Chem ; 57(24): 10512-26, 2014 Dec 26.
Article En | MEDLINE | ID: mdl-25423286

Acetyl-CoA carboxylase (ACC) inhibitors offer significant potential for the treatment of type 2 diabetes mellitus (T2DM), hepatic steatosis, and cancer. However, the identification of tool compounds suitable to test the hypothesis in human trials has been challenging. An advanced series of spirocyclic ketone-containing ACC inhibitors recently reported by Pfizer were metabolized in vivo by ketone reduction, which complicated human pharmacology projections. We disclose that this metabolic reduction can be greatly attenuated through introduction of steric hindrance adjacent to the ketone carbonyl. Incorporation of weakly basic functionality improved solubility and led to the identification of 9 as a clinical candidate for the treatment of T2DM. Phase I clinical studies demonstrated dose-proportional increases in exposure, single-dose inhibition of de novo lipogenesis (DNL), and changes in indirect calorimetry consistent with increased whole-body fatty acid oxidation. This demonstration of target engagement validates the use of compound 9 to evaluate the role of DNL in human disease.


Acetyl-CoA Carboxylase/antagonists & inhibitors , Hepatocytes/drug effects , Ketones/metabolism , Lipogenesis/drug effects , Microsomes/drug effects , Acetyl-CoA Carboxylase/metabolism , Adult , Animals , Area Under Curve , Cells, Cultured , Cross-Over Studies , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Dogs , Double-Blind Method , Hepatocytes/cytology , Humans , Male , Malonyl Coenzyme A/metabolism , Microsomes/metabolism , Middle Aged , Models, Molecular , Molecular Structure , Rats , Rats, Sprague-Dawley , Rats, Wistar , Structure-Activity Relationship , Young Adult
18.
J Med Chem ; 50(21): 5202-16, 2007 Oct 18.
Article En | MEDLINE | ID: mdl-17887659

The peptide hormone ghrelin is the endogenous ligand for the type 1a growth hormone secretagogue receptor (GHS-R1a) and the only currently known circulating appetite stimulant. GHS-R1a antagonism has therefore been proposed as a potential approach for obesity treatment. More recently, ghrelin has been recognized to also play a role in controlling glucose-induced insulin secretion, which suggests another possible benefit for a GHS-R1a antagonist, namely, the role as an insulin secretagogue with potential value for diabetes treatment. In our laboratories, piperidine-substituted quinazolinone derivatives were identified as a new class of small-molecule GHS-R1a antagonists. Starting from an agonist with poor oral bioavailability, optimization led to potent, selective, and orally bioavailable antagonists. In vivo efficacy evaluation of selected compounds revealed suppression of food intake and body weight reduction as well as glucose-lowering effects mediated by glucose-dependent insulin secretion.


Diabetes Mellitus/drug therapy , Obesity/drug therapy , Quinazolinones/chemical synthesis , Receptors, Ghrelin/antagonists & inhibitors , Administration, Oral , Animals , Binding, Competitive , Blood Glucose/analysis , Cell Line , Eating/drug effects , Glucose Tolerance Test , Humans , Male , Mice , Mice, Inbred C57BL , Quinazolinones/chemistry , Quinazolinones/pharmacology , Radioligand Assay , Rats , Rats, Wistar , Stereoisomerism , Structure-Activity Relationship , Weight Loss/drug effects
19.
Endocrinology ; 148(11): 5175-85, 2007 Nov.
Article En | MEDLINE | ID: mdl-17656463

Ghrelin, through action on its receptor, GH secretagogue receptor type 1a (GHS-R1a), exerts a variety of metabolic functions including stimulation of appetite and weight gain and suppression of insulin secretion. In the present study, we examined the effects of novel small-molecule GHS-R1a antagonists on insulin secretion, glucose tolerance, and weight loss. Ghrelin dose-dependently suppressed insulin secretion from dispersed rat islets. This effect was fully blocked by a GHS-R1a antagonist. Consistent with this observation, a single oral dose of a GHS-R1a antagonist improved glucose homeostasis in an ip glucose tolerance test in rat. Improvement in glucose tolerance was attributed to increased insulin secretion. Daily oral administration of a GHS-R1a antagonist to diet-induced obese mice led to reduced food intake and weight loss (up to 15%) due to selective loss of fat mass. Pair-feeding experiments indicated that weight loss was largely a consequence of reduced food intake. The impact of a GHS-R1a antagonist on gastric emptying was also examined. Although the GHS-R1a antagonist modestly delayed gastric emptying at the highest dose tested (10 mg/kg), delayed gastric emptying does not appear to be a requirement for weight loss because lower doses produced weight loss without an effect on gastric emptying. Consistent with the hypothesis that ghrelin regulates feeding centrally, the anorexigenic effects of potent GHS-R1a antagonists in mice appeared to correspond with their brain exposure. These observations demonstrate that GHS-R1a antagonists have the potential to improve the diabetic condition by promoting glucose-dependent insulin secretion and promoting weight loss.


Appetite Depressants/therapeutic use , Appetite/drug effects , Glucose Intolerance/drug therapy , Receptors, Ghrelin/antagonists & inhibitors , Weight Loss/drug effects , Animals , Appetite Depressants/pharmacology , Cells, Cultured , Drug Evaluation, Preclinical , Ghrelin/antagonists & inhibitors , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Male , Mice , Mice, Inbred C57BL , Models, Biological , Piperidines/pharmacology , Piperidines/therapeutic use , Quinazolinones/pharmacology , Quinazolinones/therapeutic use , Rats , Rats, Sprague-Dawley , Rats, Wistar
20.
Bioorg Med Chem Lett ; 14(8): 1935-8, 2004 Apr 19.
Article En | MEDLINE | ID: mdl-15050631

(Hydroxyethyl)urea peptidomimetics are potent inhibitors of gamma-secretase that are accessible in a few synthetic steps. Systematic alteration of P2-P4' revealed that the corresponding S2-S4' active site pockets accommodate a variety of substituents, consistent with the fact that this protease cleaves a variety of single-pass membrane proteins; however, phenylalanine is not well tolerated at P2'. A compound spanning P2-P3' was identified as a low nM inhibitor of gamma-secretase activity both in cells and under cell-free conditions.


Biomimetic Materials/pharmacology , Endopeptidases/metabolism , Hydroxyurea/analogs & derivatives , Hydroxyurea/pharmacology , Peptide Fragments/pharmacology , Protease Inhibitors/pharmacology , Amyloid Precursor Protein Secretases , Animals , Binding Sites/drug effects , Biomimetic Materials/chemical synthesis , Biomimetic Materials/chemistry , CHO Cells , Cricetinae , Endopeptidases/drug effects , Hydroxyurea/chemical synthesis , Molecular Structure , Peptide Fragments/chemical synthesis , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Structure-Activity Relationship
...