1
|
Kwon YS, Han Z. Advanced nanomedicines for the treatment of age-related macular degeneration. NANOSCALE 2024; 16:16769-16790. [PMID: 39177654 DOI: 10.1039/d4nr01917b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
The critical and unmet medical need for novel therapeutic advancements in the treatment of age-related macular degeneration (AMD) cannot be overstated, particularly given the aging global population and the increasing prevalence of this condition. Current AMD therapy involves intravitreal treatments that require monthly or bimonthly injections to maintain optimal efficacy. This underscores the necessity for improved approaches, prompting recent research into developing advanced drug delivery systems to prolong the intervals between treatments. Nanoparticle-based therapeutic approaches have enabled the controlled release of drugs, targeted delivery of therapeutic materials, and development of smart solutions for the harsh microenvironment of diseased tissues, offering a new perspective on ocular disease treatment. This review emphasizes the latest pre-clinical treatment options in ocular drug delivery to the retina and explores the advantages of nanoparticle-based therapeutic approaches, with a focus on AMD, the leading cause of irreversible blindness in the elderly.
Collapse
Affiliation(s)
- Yong-Su Kwon
- Department of Ophthalmology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
| | - Zongchao Han
- Department of Ophthalmology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, the University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
2
|
Yoshikawa E, Ueda K, Hakata R, Higashi K, Moribe K. Quantitative Investigation of Intestinal Drug Absorption Enhancement by Drug-Rich Nanodroplets Generated via Liquid-Liquid Phase Separation. Mol Pharm 2024; 21:1745-1755. [PMID: 38501717 DOI: 10.1021/acs.molpharmaceut.3c01078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Drug-rich droplets formed through liquid-liquid phase separation (LLPS) have the potential to enhance the oral absorption of drugs. This can be attributed to the diffusion of these droplets into the unstirred water layer (UWL) of the gastrointestinal tract and their reservoir effects on maintaining drug supersaturation. However, a quantitative understanding of the effect of drug-rich droplets on intestinal drug absorption is still lacking. In this study, the enhancement of intestinal drug absorption through the formation of drug-rich droplets was quantitatively evaluated on a mechanistic basis. To obtain fenofibrate (FFB)-rich droplets, an amorphous solid dispersion (ASD) of FFB/hypromellose (HPMC) was dispersed in an aqueous medium. Physicochemical characterization confirmed the presence of nanosized FFB-rich droplets in the supercooled liquid state within the FFB/HPMC ASD dispersion. An in situ single-pass intestinal perfusion (SPIP) assay in rats demonstrated that increased quantities of FFB-rich nanodroplets enhanced the intestinal absorption of FFB. The effective diffusion of FFB-rich nanodroplets through UWL would partially contribute to the improved FFB absorption. Additionally, confocal laser scanning microscopy (CLSM) of cross sections of the rat intestine after the administration of fluorescently labeled FFB-rich nanodroplets showed that these nanodroplets were directly taken up by small intestinal epithelial cells. Therefore, the direct uptake of drug-rich nanodroplets by the small intestine is a potential mechanism for improving FFB absorption in the intestine. To quantitatively evaluate the impact of FFB-rich droplets on the FFB absorption enhancement, we determined the apparent permeabilities of the FFB-rich nanodroplets and dissolved FFB based on the SPIP results. The apparent permeability of the FFB-rich nanodroplets was 110-130 times lower than that of dissolved FFB. However, when the FFB-rich nanodroplet concentration was several hundred times higher than that of dissolved FFB, the FFB-rich nanodroplets contributed significantly to FFB absorption improvement. The present study highlights that drug-rich nanodroplets play a direct role in enhancing drug absorption in the gastrointestinal tract, indicating their potential for further improvement of oral absorption from ASD formulations.
Collapse
Affiliation(s)
- Etsushi Yoshikawa
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Rei Hakata
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Kenjirou Higashi
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Kunikazu Moribe
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| |
Collapse
|
3
|
Tanaka Y, Takagi R, Mitou S, Shimmura M, Hasegawa T, Amarume J, Shinohara M, Kageyama Y, Sasase T, Ohta T, Muramatsu SI, Kakehashi A, Kaburaki T. Protective Effect of Pemafibrate Treatment against Diabetic Retinopathy in Spontaneously Diabetic Torii Fatty Rats. Biol Pharm Bull 2024:b23-00872. [PMID: 38432946 DOI: 10.1248/bpb.b23-00872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Diabetic retinopathy (DR) can cause visual impairment and blindness, and the increasing global prevalence of diabetes underscores the need for effective therapies to prevent and treat DR. Therefore, this study aimed to evaluate the protective effect of pemafibrate treatment against DR, using a Spontaneously Diabetic Torii (SDT) fatty rat model of obese type 2 diabetes. SDT fatty rats were fed either a diet supplemented with pemafibrate (0.3 mg/kg/day) for 16 weeks, starting at 8 weeks of age (Pf SDT fatty: study group), or normal chow (SDT fatty: controls). Normal chow was provided to Sprague-Dawley (SD) rats (SD: normal controls). Electroretinography (ERG) was performed at 8 and 24 weeks of age to evaluate the retinal neural function. After sacrifice, retinal thickness, number of retinal folds, and choroidal thickness were evaluated, and immunostaining was performed for aquaporin-4 (AQP4). No significant differences were noted in food consumption, body weight, or blood glucose level after pemafibrate administration. Triglyceride levels were reduced, and high-density lipoprotein cholesterol levels were increased. Extension of oscillatory potential (OP)1 and OP3 waves on ERG was suppressed in the Pf SDT fatty group. Retinal thickness at 1,500 microns from the optic disc improved in the Pf SDT fatty group. No significant improvements were noted in choroidal thickness or number of retinal folds. Quantitative analyses showed that AQP4-positive regions in the retinas were significantly larger in the Pf SDT fatty group than in the SDT fatty group. The findings suggest that pemafibrate treatment can exert protective effects against DR.
Collapse
Affiliation(s)
| | - Rina Takagi
- Department of Ophthalmology, Jichi Medical University
| | - Shingen Mitou
- Department of Ophthalmology, Jichi Medical University
| | | | | | | | | | | | - Tomohiko Sasase
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc
| | - Takeshi Ohta
- Laboratory of Animal Physiology and Functional Anatomy, Graduate School of Agriculture, Kyoto University
| | - Shin-Ichi Muramatsu
- Division of Neurological Gene Therapy, Center for Open Innovation, Jichi Medical University
| | | | | |
Collapse
|
4
|
Seo YB, Kim JH, Song JH, Jung W, Nam KY, Kim N, Choi YW, Cho S, Ki DH, Lee HJ, Moon J, Lee S, Kim J, Hong JH, Sunwoo J, Jung JG. Safety and pharmacokinetic comparison between fenofibric acid 135 mg capsule and 110 mg enteric-coated tablet in healthy volunteers. Transl Clin Pharmacol 2023; 31:95-104. [PMID: 37440778 PMCID: PMC10333648 DOI: 10.12793/tcp.2023.31.e7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/09/2023] [Accepted: 06/01/2023] [Indexed: 07/15/2023] Open
Abstract
This study aimed to compare the pharmacokinetic (PK) and safety profiles of 2 fenofibric acid formulations under fasting and fed conditions. The reference was a 135 mg capsule, while the test was a 110 mg enteric-coated tablet. This randomized, open-label, two-sequence, two-period crossover phase 1 clinical trial was conducted in healthy Korean men. Sixty participants were enrolled in each of the fasting and feeding groups. Blood samples were collected 72 hours after drug administration. PK parameters were calculated using a non-compartmental method with Phoenix WinNonlin®. A total of 53 and 51 participants from the fasting and feeding groups, respectively, completed the study. The geometric mean ratio and 90% confidence intervals of the maximum concentration (Cmax) and area under the concentration-time curve to the last measurable plasma concentration were 0.9195 (0.8795-0.9614) and 0.8630 (0.8472-0.8791) in the fasting study and 1.0926 (1.0102-1.1818) and 0.9998 (0.9675-1.0332) in the fed study, respectively. The time to reach Cmax of the enteric-coated tablet compared to that of the capsule was extended by 1 and 3 hours under fasting and fed conditions, respectively. In conclusion, enteric-coated tablets have a higher bioavailability than capsules. In addition, the enteric-coated tablet was smaller than the capsule, making it easier for patients to swallow. Trial Registration Clinical Research Information Service Identifier: KCT0007177, KCT0003304.
Collapse
Affiliation(s)
- Yu-Bin Seo
- Clinical Trials Center, Chungnam National University Hospital, Daejeon 35015, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea
| | - Jae Hoon Kim
- Clinical Trials Center, Chungnam National University Hospital, Daejeon 35015, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea
| | - Ji Hye Song
- Clinical Trials Center, Chungnam National University Hospital, Daejeon 35015, Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Korea
| | - WonTae Jung
- Korea United Pharm. Inc., Seoul 06116, Korea
| | | | - Nyung Kim
- Korea United Pharm. Inc., Seoul 06116, Korea
| | | | - SangMin Cho
- Korea United Pharm. Inc., Seoul 06116, Korea
| | - Do-Hyung Ki
- Korea United Pharm. Inc., Seoul 06116, Korea
| | | | | | | | - JaeHee Kim
- Caleb Multilab. Inc., Seoul 06745, Korea
| | - Jang Hee Hong
- Clinical Trials Center, Chungnam National University Hospital, Daejeon 35015, Korea
- Department of Pharmacology, Chungnam National University College of Medicine, Daejeon 35015, Korea
| | - Jung Sunwoo
- Clinical Trials Center, Chungnam National University Hospital, Daejeon 35015, Korea
| | - Jin-Gyu Jung
- Department of Family Medicine, Chungnam National University Hospital, Daejeon 35015, Korea
| |
Collapse
|
5
|
Tatsumi T. Current Treatments for Diabetic Macular Edema. Int J Mol Sci 2023; 24:ijms24119591. [PMID: 37298544 DOI: 10.3390/ijms24119591] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Diabetic retinopathy is a major retinal disorder and a leading cause of blindness. Diabetic macular edema (DME) is an ocular complication in patients with diabetes, and it can impair vision significantly. DME is a disorder of the neurovascular system, and it causes obstructions of the retinal capillaries, damage of the blood vessels, and hyperpermeability due to the expression and action of vascular endothelial growth factor (VEGF). These changes result in hemorrhages and leakages of the serous components of blood that result in failures of the neurovascular units (NVUs). Persistent edema of the retina around the macula causes damage to the neural cells that constitute the NVUs resulting in diabetic neuropathy of the retina and a reduction in vision quality. The macular edema and NVU disorders can be monitored by optical coherence tomography (OCT). Neuronal cell death and axonal degeneration are irreversible, and their development can result in permanent visual loss. Treating the edema before these changes are detected in the OCT images is necessary for neuroprotection and maintenance of good vision. This review describes the effective treatments for the macular edema that are therefore neuroprotective.
Collapse
Affiliation(s)
- Tomoaki Tatsumi
- Department of Ophthalmology and Visual Science, Chiba University Graduate School of Medicine, Inohana 1-8-1, Chuo-ku, Chiba 260-8670, Japan
| |
Collapse
|
6
|
Tomita Y, Usui-Ouchi A, Nilsson AK, Yang J, Ko M, Hellström A, Fu Z. Metabolism in Retinopathy of Prematurity. Life (Basel) 2021; 11:1119. [PMID: 34832995 PMCID: PMC8620873 DOI: 10.3390/life11111119] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/11/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Retinopathy of prematurity is defined as retinal abnormalities that occur during development as a consequence of disturbed oxygen conditions and nutrient supply after preterm birth. Both neuronal maturation and retinal vascularization are impaired, leading to the compensatory but uncontrolled retinal neovessel growth. Current therapeutic interventions target the hypoxia-induced neovessels but negatively impact retinal neurons and normal vessels. Emerging evidence suggests that metabolic disturbance is a significant and underexplored risk factor in the disease pathogenesis. Hyperglycemia and dyslipidemia correlate with the retinal neurovascular dysfunction in infants born prematurely. Nutritional and hormonal supplementation relieve metabolic stress and improve retinal maturation. Here we focus on the mechanisms through which metabolism is involved in preterm-birth-related retinal disorder from clinical and experimental investigations. We will review and discuss potential therapeutic targets through the restoration of metabolic responses to prevent disease development and progression.
Collapse
Affiliation(s)
- Yohei Tomita
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Y.T.); (J.Y.); (M.K.)
| | - Ayumi Usui-Ouchi
- Department of Ophthalmology, Juntendo University Urayasu Hospital, Chiba 279-0021, Japan;
| | - Anders K. Nilsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 413 19 Gothenburg, Sweden; (A.K.N.); (A.H.)
| | - Jay Yang
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Y.T.); (J.Y.); (M.K.)
| | - Minji Ko
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Y.T.); (J.Y.); (M.K.)
| | - Ann Hellström
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 413 19 Gothenburg, Sweden; (A.K.N.); (A.H.)
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Y.T.); (J.Y.); (M.K.)
| |
Collapse
|
7
|
Grabacka M, Pierzchalska M, Płonka PM, Pierzchalski P. The Role of PPAR Alpha in the Modulation of Innate Immunity. Int J Mol Sci 2021; 22:10545. [PMID: 34638886 PMCID: PMC8508635 DOI: 10.3390/ijms221910545] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/25/2021] [Accepted: 09/26/2021] [Indexed: 12/14/2022] Open
Abstract
Peroxisome proliferator-activated receptor α is a potent regulator of systemic and cellular metabolism and energy homeostasis, but it also suppresses various inflammatory reactions. In this review, we focus on its role in the regulation of innate immunity; in particular, we discuss the PPARα interplay with inflammatory transcription factor signaling, pattern-recognition receptor signaling, and the endocannabinoid system. We also present examples of the PPARα-specific immunomodulatory functions during parasitic, bacterial, and viral infections, as well as approach several issues associated with innate immunity processes, such as the production of reactive nitrogen and oxygen species, phagocytosis, and the effector functions of macrophages, innate lymphoid cells, and mast cells. The described phenomena encourage the application of endogenous and pharmacological PPARα agonists to alleviate the disorders of immunological background and the development of new solutions that engage PPARα activation or suppression.
Collapse
Affiliation(s)
- Maja Grabacka
- Department of Biotechnology and General Technology of Foods, Faculty of Food Technology, University of Agriculture, ul. Balicka 122, 30-149 Cracow, Poland;
| | - Małgorzata Pierzchalska
- Department of Biotechnology and General Technology of Foods, Faculty of Food Technology, University of Agriculture, ul. Balicka 122, 30-149 Cracow, Poland;
| | - Przemysław M. Płonka
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Cracow, Poland;
| | - Piotr Pierzchalski
- Department of Medical Physiology, Faculty of Health Sciences, Jagiellonian University Medical College, ul. Michałowskiego 12, 31-126 Cracow, Poland;
| |
Collapse
|
8
|
Söğütlü I, Mahmood EA, Ahmadizadeh Shendy S, Ebrahimiasl S, Vessally E. Recent progress in application of nanocatalysts for carbonylative Suzuki cross-coupling reactions. RSC Adv 2021; 11:2112-2125. [PMID: 35424173 PMCID: PMC8693787 DOI: 10.1039/d0ra09846a] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 12/15/2020] [Indexed: 12/31/2022] Open
Abstract
In the past few decades, cross-coupling of aryl halides and arylboronic acids in the presence of carbon monoxide (CO), also called carbonylative Suzuki coupling, to form two new carbon-carbon bonds in the production of synthetically and biologically important biaryl ketones, has been widely studied. Consequently, various catalytic systems have been extensively investigated in order to maximize the efficiency of this appealing area of biaryl ketone synthesis. As evidenced in the literature, nanometal-based systems are among the most powerful catalysts for this transformation as their large surface area to volume ratio and reactive morphologies allow faster reaction rates under milder CO pressure even at very low catalyst loadings. This review aims to provide an overview of the recent advances and achievements in the application of nano-sized metal catalysts for carbonylative Suzuki cross-coupling reactions, which may serve as an inspiration to researchers in their future work.
Collapse
Affiliation(s)
- Inci Söğütlü
- Republic of Turkey Ministry of Agriculture and Forestry Ankara Turkey
| | - Evan Abdulkarim Mahmood
- College of Health Sciences, University of Human Development Sulaimaniyah Kurdistan region of Iraq Iraq
| | | | - Saeideh Ebrahimiasl
- Department of Chemistry, Ahar Branch, Islamic Azad University Ahar Iran
- Industrial Nanotechnology Research Center, Tabriz Branch, Islamic Azad University Tabriz Iran
| | - Esmail Vessally
- Department of Chemistry, Payame Noor University P.O. Box 19395-3697 Tehran Iran
| |
Collapse
|
9
|
Li X, Hu X, Pan T, Dong L, Ding L, Wang Z, Song R, Wang X, Wang N, Zhang Y, Wang J, Yang B. Kanglexin, a new anthraquinone compound, attenuates lipid accumulation by activating the AMPK/SREBP-2/PCSK9/LDLR signalling pathway. Biomed Pharmacother 2021; 133:110802. [PMID: 33202286 DOI: 10.1016/j.biopha.2020.110802] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/16/2020] [Accepted: 09/25/2020] [Indexed: 12/18/2022] Open
Abstract
Hyperlipidaemia is one of the major risk factors for atherosclerosis, coronary heart disease, stroke and diabetes. In the present study, we synthesized a new anthraquinone compound, 1,8-dihydroxy-3-succinic acid monoethyl ester-6-methylanthraquinone, and named it Kanglexin (KLX). The aim of this study was to evaluate whether KLX has a lipid-lowering effect and to explore the potential molecular mechanism. In this study, Sprague-Dawley rats were fed a high fat diet (HFD) for 5 weeks to establish a hyperlipidaemia model; then, the rats were orally administered KLX (20, 40, and 80 mg kg-1·d-1) or atorvastatin calcium (AT, 10 mg kg-1·d-1) once a day for 2 weeks. KLX had prominent effects on reducing blood lipids, hepatic lipid accumulation, body weight and the ratio of liver weight/body weight. Furthermore, KLXdramatically reduced the total cholesterol (TC) and triglyceride (TG) levels and lipid accumulation in a HepG2 cell model of dyslipidaemia induced by 1 mmol/L oleic acid (OA). KLX may decrease lipid levels by phosphorylating adenosine monophosphate-activated protein kinase (AMPK) and the downstream sterol regulatory element binding protein 2 (SREBP-2)/proprotein convertase subtilisin/kexin type 9 (PCSK9)/low-density lipoprotein receptor (LDLR) signalling pathway in the HFD rats and OA-treated HepG2 cells. The effects of KLX on the AMPK/SREBP-2/PCSK9/LDLR signalling pathway were abolished when AMPK was inhibited by compound C (a specific AMPK inhibitor) in HepG2 cells. In summary, KLX has an efficient lipid-lowering effect mediated by activation of the AMPK/SREBP-2/PCSK9/LDLR signalling pathway. Our findings may provide new insight into and evidence for the discovery of a new lipid-lowering drug for the prevention and treatment of hyperlipidaemia, fatty liver, and cardiovascular disease in the clinic.
Collapse
Affiliation(s)
- Xin Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Xueling Hu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Tengfei Pan
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| | - Lei Dong
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Lili Ding
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Zhenzhong Wang
- Jiangsu Kanion Pharmaceutical CO. LTD, Jiangsu, Lianyungang 222001, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu, Lianyungang 222001, China.
| | - Rui Song
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Xiuzhu Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Ning Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China; Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| | - Yan Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Jinhui Wang
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| | - Baofeng Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine - Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China; Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| |
Collapse
|
10
|
PPARα Agonist Oral Therapy in Diabetic Retinopathy. Biomedicines 2020; 8:biomedicines8100433. [PMID: 33086679 PMCID: PMC7589723 DOI: 10.3390/biomedicines8100433] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetic retinopathy (DR) is an eye condition that develops after chronically poorly-managed diabetes, and is presently the main cause for blindness on a global scale. Current treatments for DR such as laser photocoagulation, topical injection of corticosteroids, intravitreal injection of anti-vascular endothelial growth factor (VEGF) agents and vitreoretinal surgery are only applicable at the late stages of DR and there are possibilities of significant adverse effects. Moreover, the forms of treatment available for DR are highly invasive to the eyes. Safer and more effective pharmacological treatments are required for DR treatment, in particular at an early stage. In this review, we cover recently investigated promising oral pharmacotherapies, the methods of which are safer, easier to use, patient-friendly and pain-free, in clinical studies. We especially focus on peroxisome proliferator-activator receptor alpha (PPARα) agonists in which experimental evidence suggests PPARα activation may be closely related to the attenuation of vascular damages, including lipid-induced toxicity, inflammation, an excess of free radical generation, endothelial dysfunction and angiogenesis. Furthermore, oral administration of selective peroxisome proliferator-activated receptor alpha modulator (SPPARMα) agonists may induce hepatic fibroblast growth factor 21 expression, indirectly resulting in retinal protection in animal studies. Our review will enable more comprehensive approaches for understanding protective roles of PPARα for the prevention of DR development.
Collapse
|
11
|
Ihm SH, Chung WB, Lee JM, Hwang BH, Yoo KD, Her SH, Song WH, Chae IH, Park TH, Kim JH, Jeon DW, Cho BR, Kang SH, Park SD, Lee JB, Woo JT, Lee BW, Han KA, Won KH, Kim HS, Yu JM, Chung CH, Kim HJ, Cho HC, Seung KB. Efficacy and Tolerability of Pitavastatin Versus Pitavastatin/Fenofibrate in High-risk Korean Patients with Mixed Dyslipidemia: A Multicenter, Randomized, Double-blinded, Parallel, Therapeutic Confirmatory Clinical Trial. Clin Ther 2020; 42:2021-2035.e3. [PMID: 32891418 DOI: 10.1016/j.clinthera.2020.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/20/2020] [Accepted: 08/05/2020] [Indexed: 01/22/2023]
Abstract
PURPOSE Dyslipidemia is an important risk factor for cardiovascular disease (CVD). Statins are known to effectively reduce not only low-density lipoprotein cholesterol (LDL-C) level but also death and nonfatal myocardial infarction due to coronary heart disease. The risk for CVD from atherogenic dyslipidemia persists when elevated triglyceride (TG) and reduced high-density lipoprotein cholesterol (HDL-C) levels are not controlled with statin therapy. Therefore, statin/fenofibrate combination therapy is more effective in reducing CVD risk. Here, we assessed the efficacy and tolerability of pitavastatin/fenofibrate combination therapy in patients with mixed dyslipidemia and a high risk for CVD. METHODS This multicenter, randomized, double-blind, parallel-group, therapeutic-confirmatory clinical trial evaluated the efficacy and tolerability of fixed-dose combination therapy with pitavastatin/fenofibrate 2/160 mg in Korean patients with a high risk for CVD and a controlled LDL-C level (<100 mg/dL) and a TG level of 150-500 mg/dL after a run-in period with pitavastatin 2 mg alone. In the 8-week main study, 347 eligible patients were randomly assigned to receive pitavastatin 2 mg with or without fenofibrate 160 mg after a run-in period. In the extension study, patients with controlled LDL-C and non-HDL-C (<130 mg/dL) levels were included after the completion of the main study. All participants in the extension study received the pitavastatin/fenofibrate combination therapy for 16 weeks for the assessment of the tolerability of long-term treatment. FINDINGS The difference in the mean percentage change in non-HDL-C from baseline to week 8 between the combination therapy and monotherapy groups was -12.45% (95% CI, -17.18 to -7.72), and the combination therapy was associated with a greater reduction in non-HDL-C. The changes in lipid profile, including apolipoproteins, fibrinogen, and high-sensitivity C-reactive protein from baseline to weeks 4 and 8 were statistically significant with combination therapy compared to monotherapy at all time points. Furthermore, the rates of achievement of non-HDL-C and apolipoprotein B targets at week 8 in the combination therapy and monotherapy groups were 88.30% versus 77.98% (P = 0.0110) and 78.94% versus 68.45% (P = 0.0021), respectively. The combination therapy was well tolerated, with a safety profile similar to that of statin monotherapy. IMPLICATIONS In these Korean patients with mixed dyslipidemia and a high risk for CVD, combination therapy with pitavastatin/fenofibrate was associated with a greater reduction in non-HDL-C compared with that with pitavastatin monotherapy, and a significantly improvement in other lipid levels. Moreover, the combination therapy was well tolerated, with a safety profile similar to that of statin monotherapy. Therefore, pitavastatin/fenofibrate combination therapy could be effective and well tolerated in patients with mixed dyslipidemia. ClinicalTrials.gov identifier: NCT03618797.
Collapse
Affiliation(s)
- Sang-Hyun Ihm
- Department of Internal Medicine, Bucheon St. Mary's Hospital, Bucheon, South Korea; College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Woo-Baek Chung
- Department of Internal Medicine, Seoul St. Mary's Hospital, Seoul, South Korea; College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jong-Min Lee
- Department of Internal Medicine, Uijeongbu St. Mary's Hospital, Uijeongbu, South Korea; College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Byung-Hee Hwang
- Department of Internal Medicine, Seoul St. Mary's Hospital, Seoul, South Korea; College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ki-Dong Yoo
- Department of Internal Medicine, St. Vincent's Hospital, Suwon, South Korea; College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Ho Her
- Department of Internal Medicine, St. Vincent's Hospital, Suwon, South Korea; College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Woo-Hyuk Song
- Department of Internal Medicine, Ansan Hospital, Korea University, Ansan, South Korea
| | - In-Ho Chae
- Department of Internal Medicine, Bundang Hospital, College of Medicine, Seoul National University, Seongnam, South Korea
| | - Tae-Ho Park
- Department of Internal Medicine, Dong-A University Hospital, Busan, South Korea
| | - Ju-Han Kim
- Department of Internal Medicine, Chonnam National University Hospital, Gwangju, South Korea
| | - Dong Woon Jeon
- Department of Internal Medicine, National Health Insurance Service, Ilsan Hospital, Goyang, South Korea
| | - Byung-Ryul Cho
- Department of Internal Medicine, Kangwon National University Hospital, Kangwon National University, Chuncheon, South Korea
| | - Seung-Ho Kang
- Department of Internal Medicine, Cheju Halla General Hospital, Jeju, South Korea
| | - Sang-Don Park
- Department of Internal Medicine, Inha University Hospital, Incheon, South Korea
| | - Jin-Bae Lee
- Department of Internal Medicine, Daegu Catholic University Medical Center, Daegu, South Korea
| | - Jeong-Taek Woo
- Department of Internal Medicine, Kyunghee University Medical Center, Seoul, South Korea
| | - Byung-Wan Lee
- Department of Internal Medicine, College of Medicine, Yonsei University, Seoul, South Korea
| | - Kyung-Ah Han
- Department of Internal Medicine, Nowon Eulji Medical Center, Eulji University, Seoul, South Korea
| | - Kyung-Heon Won
- Department of Internal Medicine, Seoul Medical Center, Seoul, South Korea
| | - Hyo-Soo Kim
- Department of Internal Medicine, Seoul National University Hospital, College of Medicine, Seoul National University, Seoul, South Korea
| | - Jae-Myung Yu
- Department of Internal Medicine, Kangnam Sacred Heart Hospital, Hallym University, Seoul, South Korea
| | - Choon Hee Chung
- Department of Internal Medicine, Wonju Severance Christian Hospital, Wonju, South Korea
| | - Hae-Jin Kim
- Department of Internal Medicine, Ajou University Hospital, Suwon, South Korea
| | - Ho-Chan Cho
- Department of Internal Medicine, Dongsan Hospital, Keimyung University, Daegu, South Korea
| | - Ki-Bae Seung
- Department of Internal Medicine, Seoul St. Mary's Hospital, Seoul, South Korea; College of Medicine, The Catholic University of Korea, Seoul, South Korea.
| |
Collapse
|
12
|
Sun C, Choi IY, Gonzalez YIR, Andersen P, Talbot CC, Iyer SR, Lovering RM, Wagner KR, Lee G. Duchenne muscular dystrophy hiPSC-derived myoblast drug screen identifies compounds that ameliorate disease in mdx mice. JCI Insight 2020; 5:134287. [PMID: 32343677 PMCID: PMC7308059 DOI: 10.1172/jci.insight.134287] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 04/23/2020] [Indexed: 12/18/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy. In the present study, when human induced pluripotent stem cells (hiPSCs) were differentiated into myoblasts, the myoblasts derived from DMD patient hiPSCs (DMD hiPSC-derived myoblasts) exhibited an identifiable DMD-relevant phenotype: myogenic fusion deficiency. Based on this model, we developed a DMD hiPSC-derived myoblast screening platform employing a high-content imaging (BD Pathway 855) approach to generate parameters describing morphological as well as myogenic marker protein expression. Following treatment of the cells with 1524 compounds from the Johns Hopkins Clinical Compound Library, compounds that enhanced myogenic fusion of DMD hiPSC-derived myoblasts were identified. The final hits were ginsenoside Rd and fenofibrate. Transcriptional profiling revealed that ginsenoside Rd is functionally related to FLT3 signaling, while fenofibrate is linked to TGF-β signaling. Preclinical tests in mdx mice showed that treatment with these 2 hit compounds can significantly ameliorate some of the skeletal muscle phenotypes caused by dystrophin deficiency, supporting their therapeutic potential. Further study revealed that fenofibrate could inhibit mitochondrion-induced apoptosis in DMD hiPSC-derived cardiomyocytes. We have developed a platform based on DMD hiPSC-derived myoblasts for drug screening and identified 2 promising small molecules with in vivo efficacy.
Collapse
Affiliation(s)
- Congshan Sun
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Genetic Muscle Disorders, Hugo W. Moser Research Institute at Kennedy Krieger Institute, Baltimore, Maryland, USA
| | | | - Yazmin I. Rovira Gonzalez
- Center for Genetic Muscle Disorders, Hugo W. Moser Research Institute at Kennedy Krieger Institute, Baltimore, Maryland, USA
- Cellular and Molecular Medicine Graduate Program, and
| | - Peter Andersen
- Institute for Cell Engineering
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - C. Conover Talbot
- The Johns Hopkins School of Medicine Institute for Basic Biomedical Sciences, Baltimore, Maryland, USA
| | | | - Richard M. Lovering
- Department of Orthopaedics and
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Kathryn R. Wagner
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Genetic Muscle Disorders, Hugo W. Moser Research Institute at Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Gabsang Lee
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for Cell Engineering
| |
Collapse
|
13
|
Ghosh MK, Wahed MII, Khan RI, Habib A, Barman RK. Pharmacological screening of fenofibrate-loaded solid dispersion in fructose-induced diabetic rat. J Pharm Pharmacol 2020; 72:909-915. [PMID: 32306394 DOI: 10.1111/jphp.13267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/14/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Hyperlipidaemia is a common phenomenon in diabetes mellitus. Fenofibrate (FF) is a good candidate for the treatment of lipid abnormalities in patients with type 2 diabetes. But the bioavailability as well as therapeutic efficacy of this drug is limited to its dissolution behaviour. Here, the authors assess the therapeutic efficacy of a newly formulated solid dispersion of fenofibrate (SDF) having enhanced dissolution profiles in contrast to pure FF using fructose-induced diabetic rat model. METHODS Fructose-induced diabetic rat model was developed to assess the pharmacological efficacy of the formulated SDF, and the results were compared with the effects of conventional FF therapy. KEY FINDINGS The 14 days treatment showed better improvement in lipid-lowering potency of SDF than pure FF. SDF containing one-third dose of pure FF showed similar effect in terms of triglyceride, total cholesterol and low-density lipoprotein lowering efficacy, whereas increased high-density lipoprotein at same extent. The similar dose of SDF produced more prominent effect than FF. Histological studies also demonstrated the enhanced lipid clearance from liver by SDF than FF that was concordant with the biochemical results. CONCLUSIONS This newly formulated SDF would be a promising alternative for conventional fenofibrate in treating hyperlipidaemia.
Collapse
Affiliation(s)
- Milon Kumar Ghosh
- Department of Pharmacy, University of Rajshahi, Rajshahi, Bangladesh.,Department of Pharmacy, Islamic University, Kushtia, Bangladesh
| | | | | | - Anwar Habib
- Department of Pharmacology, Rajshahi Medical College, Rajshahi, Bangladesh
| | | |
Collapse
|
14
|
Neurovascular protection by peroxisome proliferator-activated receptor α in ischemic stroke. Exp Neurol 2020; 331:113323. [PMID: 32320699 DOI: 10.1016/j.expneurol.2020.113323] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/14/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022]
Abstract
Ischemic stroke is a leading cause of death and disability worldwide. Currently, the only pharmacological therapy for ischemic stroke is thrombolysis with tissue plasminogen activator that has a narrow therapeutic window and increases the risk of intracerebral hemorrhage. New pharmacological treatments for ischemic stroke are desperately needed, but no neuroprotective drugs have successfully made it through clinical trials. Beneficial effects of peroxisome proliferator-activated receptor alpha (PPARα) activation on vascular integrity and function have been reported, and PPARα agonists have clinically been used for many years to manage cardiovascular disease. Thus, PPARα has gained interest in recent years as a target for neurovascular disease such as ischemic stroke. Accumulating preclinical evidence suggests that PPARα activation modulates several pathophysiological hallmarks of stroke such as oxidative stress, blood-brain barrier (BBB) dysfunction, and neuroinflammation to improve functional recovery. Therefore, this review summarizes the various actions PPARα exerts in neurovascular health and disease and the potential of employing exogenous PPARα agonists for future pharmacological treatment of ischemic stroke.
Collapse
|
15
|
Grabacka M, Plonka PM, Reiss K. Melanoma-Time to fast or time to feast? An interplay between PPARs, metabolism and immunity. Exp Dermatol 2020; 29:436-445. [PMID: 31957066 DOI: 10.1111/exd.14072] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 12/30/2019] [Accepted: 01/10/2020] [Indexed: 12/15/2022]
Abstract
Development and progression of melanoma can be accelerated by intensification of particular metabolic pathways, such as aerobic glycolysis and avid amino acid catabolism, and is accompanied by aberrant immune responses within the tumor microenvironment. Contrary to other cancer types, melanoma reveals some unique tissue-specific features, such as melanogenesis, which is intertwined with metabolism. Nuclear peroxisome proliferator-activated receptors (PPARs) take part in regulation of systemic and cellular metabolism, inflammation and melanogenesis. They appear as a focal regulatory point for these three distinct processes by occupying the intersection among AMP-dependent protein kinase (AMPK), mammalian target of rapamycin (mTOR) and PPAR gamma coactivator 1-alpha (PGC-1α) signalling pathways. When deregulated, they may accelerate melanoma malignant growth. Presenting the contribution of PPARα and PPARγ in melanoma biology, we attempt to ask how two contrasting metabolic states: obesity and fasting, can change progression of the disease and possible outcome of the treatment. This short essay is aimed to provoke a discussion about some practical implications for melanoma prevention and treatment, especially: how metabolic manipulation may be exploited to overcome immunosuppression and support immune checkpoint blockade efficacy.
Collapse
Affiliation(s)
- Maja Grabacka
- Department of Biotechnology and General Technology of Foods, Faculty of Food Technology, University of Agriculture, Kraków, Poland
| | - Przemyslaw M Plonka
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Krzysztof Reiss
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, USA
| |
Collapse
|
16
|
Takale BS, Thakore RR, Handa S, Gallou F, Reilly J, Lipshutz BH. A new, substituted palladacycle for ppm level Pd-catalyzed Suzuki-Miyaura cross couplings in water. Chem Sci 2019; 10:8825-8831. [PMID: 31803456 PMCID: PMC6849884 DOI: 10.1039/c9sc02528f] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 08/02/2019] [Indexed: 12/21/2022] Open
Abstract
A newly engineered palladacycle that contains substituents on the biphenyl rings along with the ligand HandaPhos is especially well-matched to an aqueous micellar medium, enabling valued Suzuki-Miyaura couplings to be run not only in water under mild conditions, but at 300 ppm of Pd catalyst. This general methodology has been applied to several targets in the pharmaceutical area. Multiple recyclings of the aqueous reaction mixture involving both the same as well as different coupling partners is demonstrated. Low temperature microscopy (cryo-TEM) indicates the nature and size of the particles acting as nanoreactors. Importantly, given the low loadings of Pd invested per reaction, ICP-MS analyses of residual palladium in the products shows levels to be expected that are well within FDA allowable limits.
Collapse
Affiliation(s)
- Balaram S Takale
- Department of Chemistry and Biochemistry , University of California , Santa Barbara , California 93106 , USA .
| | - Ruchita R Thakore
- Department of Chemistry and Biochemistry , University of California , Santa Barbara , California 93106 , USA .
| | - Sachin Handa
- Department of Chemistry , University of Louisville , Louisville , KY 40292 , USA
| | | | - John Reilly
- Novartis Institute for Medical Research , Cambridge , MA 02139 , USA
| | - Bruce H Lipshutz
- Department of Chemistry and Biochemistry , University of California , Santa Barbara , California 93106 , USA .
| |
Collapse
|
17
|
Chemically Modified Variants of Fenofibrate with Antiglioblastoma Potential. Transl Oncol 2019; 12:895-907. [PMID: 31078963 PMCID: PMC6514324 DOI: 10.1016/j.tranon.2019.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/08/2019] [Accepted: 04/10/2019] [Indexed: 01/30/2023] Open
Abstract
Anticancer effects of a common lipid-lowering drug, fenofibrate, have been described in the literature for a quite some time; however, fenofibrate has not been used as a direct anticancer therapy. We have previously reported that fenofibrate in its unprocessed form (ester) accumulates in the mitochondria, inhibits mitochondrial respiration, and triggers a severe energy deficit and extensive glioblastoma cell death. However, fenofibrate does not cross the blood brain barrier and is quickly processed by blood and tissue esterases to form the PPARα agonist fenofibric acid, which is practically ineffective effective in triggering cancer cell death. To address these issues, we have made several chemical modifications in fenofibrate structure to increase its stability, water solubility, tissue penetration, and ultimately anticancer potential. Our data show that, in comparison to fenofibrate, four new compounds designated here as PP1, PP2, PP3, and PP4 have improved anticancer activity in vitro. Like fenofibrate, the compounds block mitochondrial respiration and trigger massive glioblastoma cell death in vitro. In addition, one of the lead compounds, PP1, has improved water solubility and is significantly more stable when exposed to human blood in comparison to fenofibrate. Importantly, mice bearing large intracranial glioblastoma tumors demonstrated extensive areas of tumor cell death within the tumor mass following oral administration of PP1, and the treated mice did not show any major signs of distress, and accumulated PP1 at therapeutically relevant concentrations in several tissues, including brain and intracranial tumors.
Collapse
|
18
|
Eleftheriadis GK, Mantelou P, Karavasili C, Chatzopoulou P, Katsantonis D, Irakli M, Mygdalia A, Vizirianakis IS, Fatouros DG. Development and Characterization of a Self-Nanoemulsifying Drug Delivery System Comprised of Rice Bran Oil for Poorly Soluble Drugs. AAPS PharmSciTech 2019; 20:78. [PMID: 30635752 DOI: 10.1208/s12249-018-1274-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/11/2018] [Indexed: 11/30/2022] Open
Abstract
Poor aqueous solubility and low bioavailability are limiting factors in the oral delivery of lipophilic drugs. In a formulation approach to overcome these limitations, rice bran (RB) oil was evaluated as drug carrier in the development of self-nanoemulsifying drug delivery systems (SNEDDS). The performance of RB in formulations incorporating Kolliphor RH40 or Kolliphor EL as surfactants and Transcutol HP as cosolvent was compared to a common oil vehicle, corn oil (CO). Serial dilutions of the preconcentrates were performed in various media [distilled water and simulated intestinal fluids mimicking fasted state (FaSSIF) and fed state (FeSSIF)] and at different dilution ratios to simulate the in vivo droplets' behavior. The developed SNEDDS were assessed by means of phase separation, droplet size, polydispersity index, and ζ-potential. Complex ternary diagrams were constructed to identify compositions exhibiting monophasic behavior, droplet size < 100 nm, and polydispersity index (PDI) < 0.25. Multifactor analysis and response surface areas intended to determine the factors significantly affecting droplet size. The oil capacity to accommodate lipophilic drugs was assessed via fluorescence spectroscopy based on the solvatochromic behavior of Nile Red. Solubility studies were performed to prepare fenofibrate- and itraconazole-loaded SNEDDS and assess their droplet size, whereas dissolution experiments were conducted in simulated intestinal fluids. Caco-2 cell viability studies confirmed the safety of the SNEDDS formulations at 1:100 and 1:1000 dilutions after cell exposure in culture for 4 h. The obtained results showed similar performance between RB and CO supporting the potential of RB as oil vehicle for the effective oral delivery of lipophilic compounds.
Collapse
|
19
|
Alshamsan A, Kazi M, Badran MM, Alanazi FK. Role of Alternative Lipid Excipients in the Design of Self-Nanoemulsifying Formulations for Fenofibrate: Characterization, in vitro Dispersion, Digestion and ex vivo Gut Permeation Studies. Front Pharmacol 2018; 9:1219. [PMID: 30455642 PMCID: PMC6232043 DOI: 10.3389/fphar.2018.01219] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/08/2018] [Indexed: 11/30/2022] Open
Abstract
Background: The choice of lipid excipients and their origin are crucial determinant factors in the design of self-nanoemulsifying drug delivery system (SNEDDS). Aim: To investigate the aspects of alternative excipients which can influence the development of efficient SNEDDS and determine the fate of fenofibrate in aqueous media. Methods: SNEDDS of two groups (a and b) were developed using Cremercoor MCT/Capmul MCM and Kollisolv MCT/Imwitor 742 blended oils and water soluble surfactants (to improve lipid polarity) for the model anti-cholesterol drug fenofibrate. Visual assessment was employed and droplet size measurement was taken into initial consideration for optimized SNEDDS. Further SNEDDS optimizations were done on the basis of maximum drug loading by equilibrium solubility studies and maximum solubilized drug upon aqueous dispersion by dynamic dispersion studies. In vitro lipolysis was examined under simulated Fed and Fasted conditions. Intestinal permeability study of the optimal SNEDDS formulation was compared with the raw fenofibrate dispersion using non- everted "intestinal sac technique." Results: Initial characterization and solubility studies showed that mixed glycerides of Kollisolv MCT/Imwitor 742 (group b) containing formulations generated highly efficient SNEDDS as they are stable and produced lower nanodroplets with higher drug loading (group b) as compared to mixed glycerides of Cremercoor MCT/Capmul MCM (group a). In vitro dispersion and digestion studies confirmed that SNEDDS of group b (polar mixed glycerides) can retain high amount of drug (99% drug in solution for more than 24 h time) in dispersion media and have high recovery after digestion. The results from the permeability assessment confirmed that fenofibrate had 4.3-fold increase with F3b SNEDDS compared with the control. Conclusion: SNEDDS formulations containing alternative excipients (Kollisolv MCT/Imwitor 742 blend) could be a potential oral pharmaceutical product in taking anti-hyperlipidaemic agent fenofibrate to the systemic circulation as solubilized form.
Collapse
Affiliation(s)
- Aws Alshamsan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Nanobiotechnology Unit, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed M. Badran
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Fars Kaed Alanazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
20
|
Li T, Liu J, Zheng Y, Yang S, Liu X, Li X. Effects of triptolide on pharmacokinetics of fenofibrate in rats and its potential mechanism. Xenobiotica 2018; 49:211-215. [PMID: 29412757 DOI: 10.1080/00498254.2018.1438685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Triptolide and fenofibrate are often used together for the treatment of nephrotic syndrome in Chinese clinics. This study investigates the effects of triptolide on the pharmacokinetics of fenofibrate in rats and it potential mechanism. The pharmacokinetics of fenofibrate (20 mg/kg) with or without triptolide pretreatment (2 mg/kg/day for seven days) were investigated. Additionally, the inhibitory effects of triptolide on the metabolic stability of fenofibrate were investigated using rat liver microsome incubation systems. The results indicated that the Cmax (35.34 ± 7.52 vs. 30.43 ± 6.45 μg/mL), t1/2 (6.17 ± 1.15 vs. 4.90 ± 0.82 h) and AUC(0-t) (468.12 ± 35.84 vs. 416.35 ± 32.68 mg h L-1) of fenofibric acid decreased significantly (p < .05). The Tmax of fenofibric acid increased significantly (p < .05) from 5.12 ± 0.36 to 6.07 ± 0.68 h. Additionally, the metabolic stability of fenofibrate was prolonged from 35.8 ± 6.2 to 48.6 ± 7.5 min (p < .05) with the pretreatment of triptolide. In conclusion, these results indicated that triptolide could affect the pharmacokinetics of fenofibric acid, possibly by inhibiting the metabolism of fenofibrate in rat liver when they were co-administered.
Collapse
Affiliation(s)
- Tonghui Li
- a Department of Pharmacy , the Third Hospital of Hebei Medical University , Shijiazhuang , China
| | - Jijun Liu
- b Department of Pharmacy , the Second Hospital of Hebei Medical University , Shijiazhuang , China
| | - Yingying Zheng
- a Department of Pharmacy , the Third Hospital of Hebei Medical University , Shijiazhuang , China
| | - Shengchang Yang
- c Department of Physiology , Hebei University of Chinese Medicine , Shijiazhuang , China
| | - Xun Liu
- d Department of General Surgery , the Fourth Hospital of Hebei Medical University , Shijiazhuang , China
| | - Xuejing Li
- a Department of Pharmacy , the Third Hospital of Hebei Medical University , Shijiazhuang , China
| |
Collapse
|
21
|
Liu H, Kurtoglu M, León-Annicchiarico CL, Munoz-Pinedo C, Barredo J, Leclerc G, Merchan J, Liu X, Lampidis TJ. Combining 2-deoxy-D-glucose with fenofibrate leads to tumor cell death mediated by simultaneous induction of energy and ER stress. Oncotarget 2017; 7:36461-36473. [PMID: 27183907 PMCID: PMC5095013 DOI: 10.18632/oncotarget.9263] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 04/16/2016] [Indexed: 11/25/2022] Open
Abstract
Unregulated growth and replication as well as an abnormal microenvironment, leads to elevated levels of stress which is a common trait of cancer. By inducing both energy and endoplasmic reticulum (ER) stress, 2-Deoxy-glucose (2-DG) is particularly well-suited to take advantage of the therapeutic window that heightened stress in tumors provides. Under hypoxia, blocking glycolysis with 2-DG leads to significant lowering of ATP resulting in energy stress and cell death in numerous carcinoma cell types. In contrast, under normoxia, 2-DG at a low-concentration is not toxic in most carcinomas tested, but induces growth inhibition, which is primarily due to ER stress. Here we find a synergistic toxic effect in several tumor cell lines in vitro combining 2-DG with fenofibrate (FF), a drug that has been safely used for over 40 years to lower cholesterol in patients. This combination induces much greater energy stress than either agent alone, as measured by ATP reduction, increased p-AMPK and downregulation of mTOR. Inhibition of mTOR results in blockage of GRP78 a critical component of the unfolded protein response which we speculate leads to greater ER stress as observed by increased p-eIF2α. Moreover, to avoid an insulin response and adsorption by the liver, 2-DG is delivered by slow-release pump yielding significant anti-tumor control when combined with FF. Our results provide promise for developing this combination clinically and others that combine 2-DG with agents that act synergistically to selectively increase energy and ER stress to a level that is toxic to numerous tumor cell types.
Collapse
Affiliation(s)
- Huaping Liu
- Department of Cell Biology, University of Miami, Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | | | - Clara Lucia León-Annicchiarico
- Cell Death Regulation Group, Bellvitage Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Cristina Munoz-Pinedo
- Cell Death Regulation Group, Bellvitage Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Julio Barredo
- Department of Pediatrics, University of Miami, Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Guy Leclerc
- Department of Pediatrics, University of Miami, Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Jaime Merchan
- Department of Medicine, University of Miami, Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Xiongfei Liu
- Department of Cell Biology, University of Miami, Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Theodore J Lampidis
- Department of Cell Biology, University of Miami, Miller School of Medicine and Sylvester Comprehensive Cancer Center, Miami, FL, USA
| |
Collapse
|
22
|
Diane A, Pierce WD, Kelly SE, Sokolik S, Borthwick F, Jacome-Sosa M, Mangat R, Pradillo JM, Allan SM, Ruth MR, Field CJ, Hutcheson R, Rocic P, Russell JC, Vine DF, Proctor SD. Mechanisms of Comorbidities Associated With the Metabolic Syndrome: Insights from the JCR:LA-cp Corpulent Rat Strain. Front Nutr 2016; 3:44. [PMID: 27777929 PMCID: PMC5056323 DOI: 10.3389/fnut.2016.00044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 09/23/2016] [Indexed: 01/08/2023] Open
Abstract
Obesity and its metabolic complications have emerged as the epidemic of the new millennia. The use of obese rodent models continues to be a productive component of efforts to understand the concomitant metabolic complications of this disease. In 1978, the JCR:LA-cp rat model was developed with an autosomal recessive corpulent (cp) trait resulting from a premature stop codon in the extracellular domain of the leptin receptor. Rats that are heterozygous for the cp trait are lean-prone, while those that are homozygous (cp/cp) spontaneously display the pathophysiology of obesity as well as a metabolic syndrome (MetS)-like phenotype. Over the years, there have been formidable scientific contributions that have originated from this rat model, much of which has been reviewed extensively up to 2008. The premise of these earlier studies focused on characterizing the pathophysiology of MetS-like phenotype that was spontaneously apparent in this model. The purpose of this review is to highlight areas of recent advancement made possible by this model including; emerging appreciation of the "thrifty gene" hypothesis in the context of obesity, the concept of how chronic inflammation may drive obesogenesis, the impact of acute forms of inflammation to the brain and periphery during chronic obesity, the role of dysfunctional insulin metabolism on lipid metabolism and vascular damage, and the mechanistic basis for altered vascular function as well as novel parallels between the human condition and the female JCR:LA-cp rat as a model for polycystic ovary disease (PCOS).
Collapse
Affiliation(s)
- Abdoulaye Diane
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| | - W. David Pierce
- Department of Sociology, University of Alberta, Edmonton, AB, Canada
| | - Sandra E. Kelly
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| | - Sharon Sokolik
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| | - Faye Borthwick
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| | - Miriam Jacome-Sosa
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| | - Rabban Mangat
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| | | | - Stuart McRae Allan
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Megan R. Ruth
- Department of Agricultural Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Catherine J. Field
- Department of Agricultural Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | | | | | - James C. Russell
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| | - Donna F. Vine
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| | - Spencer D. Proctor
- Metabolic and Cardiovascular Diseases Laboratory, Division of Human Nutrition, Alberta Diabetes and Mazakowski Heart Institutes, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
23
|
Mohsin K, Alamri R, Ahmad A, Raish M, Alanazi FK, Hussain MD. Development of self-nanoemulsifying drug delivery systems for the enhancement of solubility and oral bioavailability of fenofibrate, a poorly water-soluble drug. Int J Nanomedicine 2016; 11:2829-38. [PMID: 27366063 PMCID: PMC4914069 DOI: 10.2147/ijn.s104187] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Background Self-nanoemulsifying drug delivery systems (SNEDDS) have become a popular formulation option as nanocarriers for poorly water-soluble drugs. The objective of this study was to investigate the factor that can influence the design of successful lipid formulation classification system (LFCS) Type III SNEDDS formulation and improve the oral bioavailability (BA) of fenofibrate. Materials and methods LFCS Type III SNEDDS were designed using various oils, water-soluble surfactants, and/or cosolvents (in considering the polarity of the lipids) for the model anticholesterol drug, fenofibrate. The developed SNEDDS were assessed visually and by measurement of the droplet size. Equilibrium solubility of fenofibrate in the SNEDDS was conducted to find out the maximum drug loading. Dynamic dispersion studies were carried out (1/100 dilution) in water to investigate how much drug stays in solution after aqueous dispersion of the formulation. The BA of SNEDDS formulation was evaluated in the rat. Results The results from the characterization and solubility studies showed that formulations containing mixed glycerides were highly efficient SNEDDS as they had higher solubility of the drug and produced nanosized droplets. The dispersion studies confirmed that SNEDDS (containing polar mixed glycerides) can retain >98% drug in solution for >24 hours in aqueous media. The in vivo pharmacokinetics parameters of SNEDDS formulation in comparison with pure drug showed significant increase in Cmax and AUC0–t, ~78% and 67%, respectively. The oral BA of fenofibrate from SNEDDS in rats was ~1.7-fold enhanced as compared with the BA from pure drug. Conclusion Fenofibrate-loaded LFCS Type III SNEDDS formulations could be a potential oral pharmaceutical product for administering the poorly water-soluble drug, fenofibrate, with an enhanced oral BA.
Collapse
Affiliation(s)
- Kazi Mohsin
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, King Saud University, Riyadh, Saudi Arabia
| | - Rayan Alamri
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, King Saud University, Riyadh, Saudi Arabia
| | - Ajaz Ahmad
- Department of Clinical Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad Raish
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Fars K Alanazi
- Kayyali Chair for Pharmaceutical Industry, Department of Pharmaceutics, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad Delwar Hussain
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Health Science University, Clovis, CA, USA
| |
Collapse
|
24
|
Alamri RG, Mohsin K, Ahmad A, Raish M, Alanazi FK. Development and validation of bioanalytical UHPLC-UV method for simultaneous analysis of unchanged fenofibrate and its metabolite fenofibric acid in rat plasma: Application to pharmacokinetics. Saudi Pharm J 2016; 25:128-135. [PMID: 28223873 PMCID: PMC5310137 DOI: 10.1016/j.jsps.2016.05.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 05/19/2016] [Indexed: 11/26/2022] Open
Abstract
A simple, precise, selective and fast ultra-high performance liquid chromatography (UHPLC-UV) method has been developed and validated for the simultaneous determination of a lipid regulating agent fenofibrate and its metabolite fenofibric acid in rat plasma. The chromatographic separation was carried out on a reversed-phase Acquity® BEH C18 column using methanol–water (65:35, v/v) as the mobile phase. The isocratic flow was 0.3 ml/min with rapid run time of 2.5 min and UV detection was at 284 nm. The method was validated over a concentration range of 100–10000 ng/ml (r2 ⩾ 0.9993). The selectivity, specificity, recovery, accuracy and precision were validated for determination of fenofibrate/fenofibric acid in rat plasma. The lower limits of detection and quantitation of the method were 30 and 90 ng/ml for fenofibrate and 40 and 100 ng/ml for fenofibric acid, respectively. The within and between-day coefficients of variation were less than 5%. The validated method has been successfully applied to measure the plasma concentrations in pharmacokinetics study of fenofibrate in an animal model to illustrate the scope and application of the method.
Collapse
Affiliation(s)
- Rayan G Alamri
- Kayyali Chair for Pharmaceutical Industries, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Kazi Mohsin
- Kayyali Chair for Pharmaceutical Industries, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ajaz Ahmad
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Raish
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fars K Alanazi
- Kayyali Chair for Pharmaceutical Industries, Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
25
|
Mathur M, Kusum Devi V. Potential of novel drug delivery strategies for the treatment of hyperlipidemia. J Drug Target 2016; 24:916-926. [PMID: 27029893 DOI: 10.3109/1061186x.2016.1172586] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Emergence of hyperlipidemia in urban population of India and the world at large is very high and accounts to several fatal diseases. This condition is known to manifest elevated levels of lipids and/or lipoproteins. Serious limitations like inadequate solubility, less absorption, less bioavailability, ineffectiveness in lowering of cholesterol levels, patient incompliance and so on are noticed with majority of anti-hyperlipidemic drugs and dosage forms, which are used conventionally. To overcome these shortcomings, building technology platforms for development of appropriate dosage forms is the need of the hour. These efforts are required to maximize patient acceptability while maintaining safety, efficacy, accessibility and affordability. Hyperlipidemia, its types, etiology, pathophysiology and conventional dosage forms are discussed here. The current approaches and novel developments which illustrate controlled drug release and sustained therapeutic effect along with site specific and target oriented drug delivery with better patient compliance are also reviewed critically. Despite the incentives provided by the efforts of formulation scientists, there is still a need for implementation of pharmaceutical technologies that enable to combat limitations of anti-hyperlipidemic drugs and conventional dosage forms associated with it. The present review emphasize on applications of novel drug delivery systems in pharmacotherapy of anti-hyperlipidemic drugs demonstrating the advantages and disadvantages.
Collapse
Affiliation(s)
- Mahima Mathur
- a Department of Pharmaceutics, Al-Ameen College of Pharmacy , Bangalore , Karnataka , India
| | - V Kusum Devi
- a Department of Pharmaceutics, Al-Ameen College of Pharmacy , Bangalore , Karnataka , India
| |
Collapse
|
26
|
Massaro M, Scoditti E, Pellegrino M, Carluccio MA, Calabriso N, Wabitsch M, Storelli C, Wright M, De Caterina R. Therapeutic potential of the dual peroxisome proliferator activated receptor (PPAR)α/γ agonist aleglitazar in attenuating TNF-α-mediated inflammation and insulin resistance in human adipocytes. Pharmacol Res 2016; 107:125-136. [PMID: 26976796 DOI: 10.1016/j.phrs.2016.02.027] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 02/25/2016] [Accepted: 02/25/2016] [Indexed: 12/13/2022]
Abstract
Adipose tissue inflammation is a mechanistic link between obesity and its related sequelae, including insulin resistance and type 2 diabetes. Dual ligands of peroxisome proliferator activated receptor (PPAR)α and γ, combining in a single molecule the metabolic and inflammatory-regulatory properties of α and γ agonists, have been proposed as a promising therapeutic strategy to antagonize adipose tissue inflammation. Here we investigated the effects of the dual PPARα/γ agonist aleglitazar on human adipocytes challenged with inflammatory stimuli. Human Simpson-Golabi-Behmel syndrome (SGBS) adipocytes were treated with aleglitazar or - for comparison - the selective agonists for PPARα or γ fenofibrate or rosiglitazone, respectively, for 24h before stimulation with TNF-α. Aleglitazar, at concentrations as low as 10nmol/L, providing the half-maximal transcriptional activation of both PPARα and PPARγ, reduced the stimulated expression of several pro-inflammatory mediators including interleukin (IL)-6, the chemokine CXC-L10, and monocyte chemoattractant protein (MCP)-1. Correspondingly, media from adipocytes treated with aleglitazar reduced monocyte migration, consistent with suppression of MCP-1 secretion. Under the same conditions, aleglitazar also reversed the TNF-α-mediated suppression of insulin-stimulated ser473 Akt phosphorylation and decreased the TNF-α-induced ser312 IRS1 phosphorylation, two major switches in insulin-mediated metabolic activities, restoring glucose uptake in insulin-resistant adipocytes. Such effects were similar to those obtainable with a combination of single PPARα and γ agonists. In conclusion, aleglitazar reduces inflammatory activation and dysfunction in insulin signaling in activated adipocytes, properties that may benefit diabetic and obese patients. The effect of aleglitazar was consistent with dual PPARα and γ agonism, but with no evidence of synergism.
Collapse
Affiliation(s)
- Marika Massaro
- National Research Council (CNR) Institute of Clinical Physiology, Lecce, Italy
| | - Egeria Scoditti
- National Research Council (CNR) Institute of Clinical Physiology, Lecce, Italy
| | - Mariangela Pellegrino
- National Research Council (CNR) Institute of Clinical Physiology, Lecce, Italy; Department of Biological and Environmental Science and Technology (DISTEBA), University of Salento, Lecce, Italy
| | | | - Nadia Calabriso
- National Research Council (CNR) Institute of Clinical Physiology, Lecce, Italy
| | - Martin Wabitsch
- Division of Pediatric Endocrinology, Diabetes and Obesity, Department of Pediatrics and Adolescent Medicine, University of Ulm, Germany
| | - Carlo Storelli
- Department of Biological and Environmental Science and Technology (DISTEBA), University of Salento, Lecce, Italy
| | | | - Raffaele De Caterina
- G. dAnnunzio University and Center of Excellence on Aging, Chieti, Italy; G. Monasterio Foundation for Clinical Research, Pisa, Italy.
| |
Collapse
|
27
|
Pallebage-Gamarallage M, Takechi R, Lam V, Elahy M, Mamo J. Pharmacological modulation of dietary lipid-induced cerebral capillary dysfunction: Considerations for reducing risk for Alzheimer's disease. Crit Rev Clin Lab Sci 2015; 53:166-83. [PMID: 26678521 DOI: 10.3109/10408363.2015.1115820] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
An increasing body of evidence suggests that cerebrovascular dysfunction and microvessel disease precede the evolution of hallmark pathological features that characterise Alzheimer's disease (AD), consistent with a causal association for onset or progression. Recent studies, principally in genetically unmanipulated animal models, suggest that chronic ingestion of diets enriched in saturated fats and cholesterol may compromise blood-brain barrier (BBB) integrity resulting in inappropriate blood-to-brain extravasation of plasma proteins, including lipid macromolecules that may be enriched in amyloid-β (Aβ). Brain parenchymal retention of blood proteins and lipoprotein bound Aβ is associated with heightened neurovascular inflammation, altered redox homeostasis and nitric oxide (NO) metabolism. Therefore, it is a reasonable proposition that lipid-lowering agents may positively modulate BBB integrity and by extension attenuate risk or progression of AD. In addition to their robust lipid lowering properties, reported beneficial effects of lipid-lowering agents were attributed to their pleiotropic properties via modulation of inflammation, oxidative stress, NO and Aβ metabolism. The review is a contemporary consideration of a complex body of literature intended to synthesise focussed consideration of mechanisms central to regulation of BBB function and integrity. Emphasis is given to dietary fat driven significant epidemiological evidence consistent with heightened risk amongst populations consuming greater amounts of saturated fats and cholesterol. In addition, potential neurovascular benefits associated with the use of hypolipidemic statins, probucol and fenofibrate are also presented in the context of lipid-lowering and pleiotropic properties.
Collapse
Affiliation(s)
- Menuka Pallebage-Gamarallage
- a Faculty of Health Sciences , School of Public Health Curtin University , Perth , WA , Australia and.,b Curtin Health Innovation Research Institute of Aging and Chronic Disease, Curtin University , Perth , WA , Australia
| | - Ryusuke Takechi
- a Faculty of Health Sciences , School of Public Health Curtin University , Perth , WA , Australia and.,b Curtin Health Innovation Research Institute of Aging and Chronic Disease, Curtin University , Perth , WA , Australia
| | - Virginie Lam
- a Faculty of Health Sciences , School of Public Health Curtin University , Perth , WA , Australia and.,b Curtin Health Innovation Research Institute of Aging and Chronic Disease, Curtin University , Perth , WA , Australia
| | - Mina Elahy
- a Faculty of Health Sciences , School of Public Health Curtin University , Perth , WA , Australia and.,b Curtin Health Innovation Research Institute of Aging and Chronic Disease, Curtin University , Perth , WA , Australia
| | - John Mamo
- a Faculty of Health Sciences , School of Public Health Curtin University , Perth , WA , Australia and.,b Curtin Health Innovation Research Institute of Aging and Chronic Disease, Curtin University , Perth , WA , Australia
| |
Collapse
|
28
|
Molecular mechanisms of fenofibrate-induced metabolic catastrophe and glioblastoma cell death. Mol Cell Biol 2014; 35:182-98. [PMID: 25332241 DOI: 10.1128/mcb.00562-14] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Fenofibrate (FF) is a common lipid-lowering drug and a potent agonist of the peroxisome proliferator-activated receptor alpha (PPARα). FF and several other agonists of PPARα have interesting anticancer properties, and our recent studies demonstrate that FF is very effective against tumor cells of neuroectodermal origin. In spite of these promising anticancer effects, the molecular mechanism(s) of FF-induced tumor cell toxicity remains to be elucidated. Here we report a novel PPARα-independent mechanism explaining FF's cytotoxicity in vitro and in an intracranial mouse model of glioblastoma. The mechanism involves accumulation of FF in the mitochondrial fraction, followed by immediate impairment of mitochondrial respiration at the level of complex I of the electron transport chain. This mitochondrial action sensitizes tested glioblastoma cells to the PPARα-dependent metabolic switch from glycolysis to fatty acid β-oxidation. As a consequence, prolonged exposure to FF depletes intracellular ATP, activates the AMP-activated protein kinase-mammalian target of rapamycin-autophagy pathway, and results in extensive tumor cell death. Interestingly, autophagy activators attenuate and autophagy inhibitors enhance FF-induced glioblastoma cytotoxicity. Our results explain the molecular basis of FF-induced glioblastoma cytotoxicity and reveal a new supplemental therapeutic approach in which intracranial infusion of FF could selectively trigger metabolic catastrophe in glioblastoma cells.
Collapse
|
29
|
Stillhart C, Imanidis G, Griffin BT, Kuentz M. Biopharmaceutical modeling of drug supersaturation during lipid-based formulation digestion considering an absorption sink. Pharm Res 2014; 31:3426-44. [PMID: 24962509 DOI: 10.1007/s11095-014-1432-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 06/03/2014] [Indexed: 11/29/2022]
Abstract
PURPOSE In vitro lipolysis is widely utilized for predicting in vivo performance of oral lipid-based formulations (LBFs). However, evaluation of LBFs in the absence of an absorption sink may have limited in vivo relevance. This study aimed at employing biopharmaceutical modeling to simulate LBF digestion and drug supersaturation in a continuous absorptive environment. METHODS Three fenofibrate-loaded LBFs were characterized in vitro (dispersion and lipolysis) and drug precipitation was monitored using in-line Raman spectroscopy. In vitro data were combined with pharmacokinetic data derived from an in vivo study in pigs to simulate intestinal LBF transit. This biopharmaceutical model allowed calculation of lipolysis-triggered drug supersaturation while drug and lipolysis products are absorbed from the intestine. RESULTS The biopharmaceutical model predicted that, in a continuous absorption environment, fenofibrate supersaturation was considerably lower compared to in vitro lipolysis (non-sink). Hence, the extensive drug precipitation observed in vitro was predicted to be unlikely in vivo. The absorption of lipolysis products increased drug supersaturation, but drug precipitation was unlikely for highly permeable drugs. CONCLUSIONS Biopharmaceutical modeling is a valuable approach for predicting LBFs performance in vivo. In the absence of in vitro tools simulating absorptive conditions, modeling strategies should be further considered.
Collapse
Affiliation(s)
- Cordula Stillhart
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | | | | | | |
Collapse
|
30
|
Nita C, Bala C, Porojan M, Hancu N. Fenofibrate improves endothelial function and plasma myeloperoxidase in patients with type 2 diabetes mellitus: an open-label interventional study. Diabetol Metab Syndr 2014; 6:30. [PMID: 24594096 PMCID: PMC3974011 DOI: 10.1186/1758-5996-6-30] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 02/13/2014] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Fenofibrate offers a number of benefits on the cardiovascular system and it is plausible that its anti-inflammatory, anti-oxidant and anti-fibrotic effects and enhancement of cardiac metabolic performances may account for its direct cardioprotective effects.In this study we aimed to investigate the effect of fenofibrate on endothelial function assesed by vascular studies and levels of soluble E-selectin (sE-selectin) as well as the effect on plasma myeloperoxidase (MPO) in patients with type 2 diabetes mellitus (T2DM) without previous use of lipid-lowering medication. METHODS 27 patients (14 men and 13 women) with T2DM and good glycemic control (HbA1c: min 5.9%, max: 7.1%) treated with metformin monotherapy, without previous use of lipid-lowering medication were enrolled in this study. Vascular studies included measures of brachial artery diameter before and after release of a suprasystolic ischemia. FMD was calculated as the percent (%) change in arterial diameter following reactive hyperemia. Student's paired t test and Wilcoxon Signed Ranks Test were used to compare values before and after fenofibrate therapy. RESULTS Fenofibrate therapy significantly increased post ischemia mean brachial artery diameter at 60 s (from 4.7 [4.4; 5.0] mm to 4.9 [4.6; 5.2] mm, p = 0.01) and at 90 s (from 4.7 [4.4; 5.0] mm to 4.9 [4.6; 5.1], p = 0.02). FMD response to hyperaemia at 60 s increased with 4.5 ± 13.7% (median value pre- treatment: 22.2%, median value post- treatment 25.0%, z = -2.9, p = 0.004). After 8 weeks of fenofibrate therapy, plasma MPO levels decreased to 49.5 [30.3; 71.5] ng/ml (% change from baseline = 4.6%, z = -2.2, p = 0.03) and mean plasma sE-selectin levels decreased to 67.1 [54.4; 79.8] ng/ml, (% change from baseline = 2.6%, p = 0.03). CONCLUSION In patients with T2DM without previous treatment for dyslipidemia, short-term treatment with fenofibrate improved vascular endothelial function as demonstrated by increased post ischemia mean brachial artery diameter, increased FMD and decreased plasma sE-selectin and favorably affected plasma MPO levels. Therefore, fenofibrate may be considered a protective cardiovascular drug in this group of patients. TRIAL REGISTRATION (Australian New Zealand Clinical Trials Registry ANZCTR12612000734864).
Collapse
Affiliation(s)
- Cristina Nita
- Department of Diabetes, Nutrition and Metabolic Diseases, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 2-4 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Cornelia Bala
- Department of Diabetes, Nutrition and Metabolic Diseases, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 2-4 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Mihai Porojan
- Department of Internal Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 4-6 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Nicolae Hancu
- Department of Diabetes, Nutrition and Metabolic Diseases, “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, 2-4 Clinicilor Street, 400006 Cluj-Napoca, Romania
| |
Collapse
|
31
|
|
32
|
Cho YD, Park YJ. In vitro and in vivo evaluation of a self-microemulsifying drug delivery system for the poorly soluble drug fenofibrate. Arch Pharm Res 2013; 37:193-203. [DOI: 10.1007/s12272-013-0169-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 05/16/2013] [Accepted: 05/28/2013] [Indexed: 11/28/2022]
|
33
|
Fei Y, Kostewicz ES, Sheu MT, Dressman JB. Analysis of the enhanced oral bioavailability of fenofibrate lipid formulations in fasted humans using an in vitro-in silico-in vivo approach. Eur J Pharm Biopharm 2013; 85:1274-84. [PMID: 23500116 DOI: 10.1016/j.ejpb.2013.03.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 02/14/2013] [Accepted: 03/02/2013] [Indexed: 12/21/2022]
Abstract
Lipid-based formulations have established a significant role in the formulation of poorly soluble drugs for oral administration. In order to better understand their potential advantages over solid oral dosage forms, we studied the solubility and dissolution/precipitation characteristics of three self-microemulsifying drug delivery system (SMEDDS) formulations and one suspension of micronized fenofibrate in lipid excipients, for which pharmacokinetic studies had already been reported in the open literature. The in vitro dispersion/dissolution studies were carried out in biorelevant media using USP II apparatus. These were followed up by in silico simulations using STELLA® software, in which not only dispersion/dissolution, but also the precipitation and re-dissolution of fenofibrate was taken into account. While unformulated drug exhibited poor solubility (0.22 μg/mL in FaSSGF and 4.31 μg/mL in FaSSIF-V2(PO4)) and dissolved less than 2% in dissolution tests, the solubility of fenofibrate in the presence of the lipid excipients increased dramatically (e.g., to 65.44 μg/mL in the presence of the Myritol 318/TPGS/Tween 80 SMEDDS) and there was an attendant increase in the dissolution (over 80% from capsules containing the Myritol 318/TPGS/Tween 80 SMEDDS and about 20% from the dispersion of fenofibrate in lipid excipients). For the four lipid-based fenofibrate formulations studied, combining in vitro data in biorelevant media with in silico simulation resulted in accurate prediction of the in vivo human plasma profiles. The point estimates of C(max) and AUC ratio calculated from the in silico and in vivo plasma profiles fell within the 0.8-1.25 range for the SMEDDS solution and capsule formulations, suggesting an accurate simulation of the in vivo profiles. This similarity was confirmed by calculation of the respective f2 factors. Sensitivity analysis of the simulation profiles revealed that the SMEDDS formulations had virtually removed any dependency of absorption on the dissolution rate in the small intestine, whereas for the dispersion in lipid excipients, this barrier remained. Such results pave the way to optimizing the performance of oral lipid-based formulations via an in vitro-in silico-in vivo approach.
Collapse
Affiliation(s)
- Yang Fei
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany.
| | | | | | | |
Collapse
|
34
|
Premature drug release of polymeric micelles and its effects on tumor targeting. Int J Pharm 2013; 445:117-24. [DOI: 10.1016/j.ijpharm.2013.01.059] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 01/18/2013] [Accepted: 01/21/2013] [Indexed: 11/22/2022]
|
35
|
Mohsin K. Design of lipid-based formulations for oral administration of poorly water-soluble drug fenofibrate: effects of digestion. AAPS PharmSciTech 2012; 13:637-46. [PMID: 22547370 DOI: 10.1208/s12249-012-9787-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Accepted: 04/04/2012] [Indexed: 11/30/2022] Open
Abstract
Lipid-based drug carriers are likely to have influence on bioavailability through enhanced solubilization of the drug in the gastrointestinal tract. The study was designed to investigate the lipid formulation digestibility in the simulated gastro intestinal media. Fenofibrate was formulated in representative Type II, IIIA, IIIB and IV self-emulsifying/microemulsifying lipid delivery systems (SEDDS and SMEDDS designed for oral administration) using various medium-chain glyceride components, non-ionic surfactants and cosolvents as excipients. Soybean oil was used only as an example of long-chain triglycerides to compare the effects of formulation with their counterparts. The formulations were subjected to in vitro digestion specifically to predict the fate of the drug in the gastro intestinal tract after exposure of the formulation to pancreatic enzymes and bile. In vitro digestion experiments were carried out using a pH-stat maintained at pH 7.5 for 30 min using intestinal fluids simulating the fed and fasted states. The digestion rate was faster and almost completed in Type II and IIIA systems. Most of the surfactants used in the studies are digestible. However, the high concentration of surfactant and/or cosolvent used in Type IIIB or IV systems lowered the rate of digestion. The digestion of medium-chain triglycerides was faster than long-chain triglycerides, but kept comparatively less drug in the post digestion products. Medium-chain mixed glycerides are good solvents for fenofibrate as rapidly digested but to improve fenofibrate concentration in post digestion products the use of long-chain mixed glycerides are suggested for further investigations.
Collapse
|
36
|
Hugo M, Kunath K, Dressman J. Selection of excipient, solvent and packaging to optimize the performance of spray-dried formulations: case example fenofibrate. Drug Dev Ind Pharm 2012; 39:402-12. [PMID: 22591213 DOI: 10.3109/03639045.2012.685176] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Along with other options, solid dispersions prepared by spray drying offer the possibility of formulating poorly soluble drugs in a rapidly dissolving format. As a wide range of potential excipients and solvents is available for spray drying, it is usually necessary to carry out a comprehensive array of studies to arrive at an optimal formulation. OBJECTIVE To study the influence of formulation parameters such as co-sprayed excipients, solvents and packaging on the manufacture, in vitro performance and stability of spray-dried oral drug products using fenofibrate as a model drug. MATERIALS AND METHODS Solid dispersions of fenofibrate with different amorphous polymers were manufactured from two solvent systems by spray drying. These were characterized in terms of physicochemical properties, crystalline content and dissolution behavior in biorelevant media upon production and after storage in two packaging systems (Glass and Activ-Vials(™)). RESULTS AND DISCUSSION Spray drying the same formulation from two different solvents led to different physicochemical properties, dissolution behavior and long-term stability. The dissolution behavior and long-term stability also varied significantly among excipients. The viscosity of the polymer and the packaging material proved to be important to the long-term stability. CONCLUSION For spray-dried products containing fenofibrate, the excipients were ranked according to dissolution and stability performance as follows: PVP derivatives >> HPMC 2910/15, HPMCAS-MF, HP-β-CD >> PVP:PVA 2:8. EtOH 96% proved superior to acetone/water for spray drying with polymers. The results were used to propose a general approach to developing spray-dried formulations of poorly soluble drugs.
Collapse
Affiliation(s)
- Marc Hugo
- Institute of Pharmaceutical Technology, Johann Wolfgang Goethe University, Frankfurt am Main, Germany
| | | | | |
Collapse
|
37
|
The role of ruminant trans fat as a potential nutraceutical in the prevention of cardiovascular disease. Food Res Int 2012. [DOI: 10.1016/j.foodres.2011.08.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
38
|
Lin YM, Wu JY, Chen YC, Su YD, Ke WT, Ho HO, Sheu MT. In situ formation of nanocrystals from a self-microemulsifying drug delivery system to enhance oral bioavailability of fenofibrate. Int J Nanomedicine 2011; 6:2445-57. [PMID: 22072880 PMCID: PMC3205139 DOI: 10.2147/ijn.s25339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVES In situ formation of nanocrystals and dissolution profiles of fenofibrate (FFB) from a self-microemulsifying drug delivery system (SMEDDS) were characterized. METHODS SMEDDS formulated with Myritol and surfactant mixture (Smix) of D-α-Tocopheryl polyethylene glycol 1000 succinate (TPGS) and either Tween 20 (A, C, E, G, M, S, N, T, O) or Tween 80 (B, D, F, H, P, U, Q, V, R) at various oil/Smix ratios (Group I: A and B of 0.42, C and D of 0.25, E and F of 0.11; Group II: G and H of 1.38, M and P of 1.11, S and U of 0.9, N and Q of 0.73, T and V of 0.58, and O and R of 0.46) and water contents (1: 9.5%, 2: 5.0%, 3: 0.0%, G-V: 4.5%). Their dissolutions were conducted at different rotation speeds. Two optimal SMEDDSs containing Tween 80(B2) or a higher oil/Smix ratio(Q) and B2(solution) were selected for pharmacokinetic study. RESULTS FFB particles formed within the nanosize range from Group I gradually increased with time but decreased with increasing stirring rates. However, the mean size of FFB formed by B series was as low as 200 nm, which was smaller than that of A series at three stirring rates. The release rate from both groups obviously increased with increasing stirring rate. However, incomplete release was observed for S and N in Tween 20 series, whereas a faster release rate and complete release were observed for Tween 80 series with an insignificant difference among them. Results of pharmacokinetic study demonstrated that the highest-ranked area under the curve and Cmax values were for Q(SMEDDS) and B2(solution), respectively. The relative bioavailability of Q(SMEDDS) with respect to Tricor was enhanced by about 1.14-1.22-fold. CONCLUSION SMEDDS, consisting of Myritol 318 and TPGS combined with Tween 80 at 4:1, was able to enhance the oral bioavailability of FFB.
Collapse
Affiliation(s)
- You-Meei Lin
- Department of Pharmacy, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan, ROC
| | | | | | | | | | | | | |
Collapse
|
39
|
Godfrey AR, Digiacinto J, Davis MW. Single-dose bioequivalence of 105-mg fenofibric acid tablets versus 145-mg fenofibrate tablets under fasting and fed conditions: a report of two phase I, open-label, single-dose, randomized, crossover clinical trials. Clin Ther 2011; 33:766-75. [PMID: 21704241 DOI: 10.1016/j.clinthera.2011.05.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2011] [Indexed: 11/27/2022]
Abstract
BACKGROUND Fenofibrate is used to treat primary hypercholesterolemia, mixed lipidemia, and hypertriglyceridemia in adults who do not respond to nonpharmacologic measures. Fenofibrate is a prodrug that is rapidly and completely hydrolyzed to fenofibric acid, the active moiety. A new orally administered agent, fenofibric acid, was developed as an alternative to fenofibrate. OBJECTIVE Two separate studies were conducted to evaluate the bioequivalence of fenofibric acid relative to fenofibrate under fasted and fed (standard breakfast) conditions, characterize the pharmacokinetic profile, and assess the safety and tolerability of fenofibric acid. METHODS In study 1 (fasted), during each study period, volunteers received a single 105-mg dose of fenofibric acid or single 145-mg dose of fenofibrate (depending on their randomization scheme) after an overnight fast (a minimum fast of 10 hours). A 7-day washout period followed the first treatment period, after which the volunteers received the alternate treatment. Study 2 followed a similar dosing scheme and differed only in that volunteers received their single dose after being fed a standard meal (575 calories, of which 36% were contributed by fat). Serial blood samples in both studies were collected up to 72 hours after drug administration. The pharmacokinetic parameters of interest for assessing bioequivalence were AUC(0-t), AUC(0-∞), C(max), and T(max). The criterion for a lack of difference between products was a 90% CI between 0.80 and 1.25 for the fenofibric acid:fenofibrate ratios for AUC(0-t), AUC(0-∞), and C(max.) Tolerability was assessed by adverse events (AEs), laboratory parameters, vital signs, and physical examinations. RESULTS Volunteers in study 1 (fasted; n = 54) were aged 18 to 43 years; 19 (35%) were men and 35 (65%) were women; mean weight was 155.2 pounds (range, 103.0-267.0 pounds); and 48 (89%) were white, 1 (2%) was black, and 5 (9%) were white/American Indian/Alaskan native/Asian. Volunteers in study 2 (fed; n = 54) were aged 18 to 43 years; 27 (50%) were men and 27 (50%) were women; mean weight was 161.9 pounds (range, 112.0-225.0 pounds); and 51 (94%) were white (including 2 Hispanic) and 3 (6%) were black. The 90% CIs about the ratio of the fenofibric acid geometric mean to the fenofibrate geometric mean were within the 80% and 125% limits for the pharmacokinetic parameters C(max), AUC(0-t), and AUC(0-∞) of the ln-transformed data in both study 1 (fasted) and study 2. In study 1 (fasted), 14 volunteers (26%) experienced a total of 29 AEs; the most common nonlaboratory AEs were dizziness (6%) and headache (4%). In study 2, 12 volunteers (22%) experienced a total of 19 AEs; the most common nonlaboratory AEs were headache (17%) and dry throat (4%). AEs were generally mild or moderate in intensity. CONCLUSIONS In these 2 single-dose studies, these healthy volunteers administered a single oral dose of 105-mg fenofibric acid met the US Food and Drug Administration regulatory criteria for assuming bioequivalence to a single oral dose of 145-mg fenofibrate tablets with respect to the rate and extent of fenofibric acid absorption in both fed and fasted states. Fenofibric acid at the dose studied was well tolerated in this population.
Collapse
|
40
|
Design of fenofibrate microemulsion for improved bioavailability. Int J Pharm 2011; 420:251-5. [PMID: 21907776 DOI: 10.1016/j.ijpharm.2011.08.043] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 07/04/2011] [Accepted: 08/28/2011] [Indexed: 11/21/2022]
Abstract
The objective of the present study was to formulate a microemulsion system for oral administration to improve the solubility and bioavailability of fenofibrate. Various formulations were prepared using different ratios of oils, surfactants and co-surfactants (S&CoS). Pseudo-ternary phase diagrams were constructed to evaluate the microemulsification existence area. The formulations were characterized by solubility of the drug in the vehicles, mean droplet size, and drug content. The stability was also investigated by store for 3 months under 4°C, 25°C and 40°C and diluted 100 times for 3 days. The optimal formulation consists of 25% Capryol 90, 27.75% Cremophore EL, 9.25% Transcutol P and 38% water (w/w), with a maximum solubility of fenofibrate up to ∼40.96 mg/mL. The microemulsion was physicochemical stable and mean droplet size was about 32.5-41.7 nm. The pharmacokinetic study was performed in dogs and compared with Lipanthy capsule. The result showed that microemulsion has significantly increased the C(max) and AUC compared to that of Lipanthy capsule (p<0.05). The oral bioavailability of fenofibrate microemulsions (FEN-MEs) in ME-3 and ME-4 were 1.63 and 1.30-fold higher than that of the capsule. Our results indicated that the microemulsions could be used as an effective formulation for enhancing the oral bioavailability of fenofibrate.
Collapse
|
41
|
The peroxisome proliferator-activated receptor-α agonist, BAY PP1, attenuates renal fibrosis in rats. Kidney Int 2011; 80:1182-97. [PMID: 21814170 DOI: 10.1038/ki.2011.254] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Recent studies have shown renoprotective effects of the peroxisome proliferator-activated receptor-α (PPAR-α), but its role in kidney fibrosis is unknown. In order to gain insight into this, we examined the effect of a novel PPAR-α agonist, BAY PP1, in two rat models of renal fibrosis: unilateral ureteral obstruction and the 5/6 nephrectomy. In healthy animals, PPAR-α was expressed in tubular but not in interstitial cells. Upon induction of fibrosis, PPAR-α was significantly downregulated, and treatment with BAY PP1 significantly restored its expression. During ureteral obstruction, treatment with BAY PP1 significantly reduced tubulointerstitial fibrosis, proliferation of interstitial fibroblasts, and TGF-β(1) expression. Treatment with a less potent PPAR-α agonist, fenofibrate, had no effects. Treatment with BAY PP1, initiated in established disease in the 5/6 nephrectomy, halted the decline of renal function and significantly ameliorated renal fibrosis. In vitro, BAY PP1 had no direct effect on renal fibroblasts but reduced collagen, fibronectin, and TGF-β(1) expression in tubular cells. Conditioned media of BAY PP1-treated tubular cells reduced fibroblast proliferation. Thus, renal fibrosis is characterized by a reduction of PPAR-α expression, and treatment with BAY PP1 restores PPAR-α expression and ameliorates renal fibrosis by modulating the cross-talk between tubular cells and fibroblasts. Hence, potent PPAR-α agonists might be useful in the treatment of renal fibrosis.
Collapse
|
42
|
Preparation and Evaluation of Novel Fenofibrate-loaded Self-Microemulsifying Drug Delivery System (SMEDDS). JOURNAL OF PHARMACEUTICAL INVESTIGATION 2010. [DOI: 10.4333/kps.2010.40.6.339] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
43
|
Wei JD, Ho HO, Chen CH, Ke WT, Chen ETH, Sheu MT. Characterisation of fenofibrate dissolution delivered by a self-microemulsifying drug-delivery system. J Pharm Pharmacol 2010; 62:1685-96. [DOI: 10.1111/j.2042-7158.2010.01182.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Abstract
Objectives
This study attempted to characterise the in-vitro release profiles of fenofibrate (FFB) from a self-microemulsifying drug-delivery system (SMEDDS) for optimising formulation factors and dissolution conditions for in-vivo absorption.
Methods
The study was conducted by profiling the release of FFB formulated with either a complete solution or a micronised dispersion system (MDS) in a SMEDDS composed of medium-chain triglyceride (MCT) oil and surfactant mixtures (Smix) of TPGS and Tweens at different ratios (Km = TPGS/Tweens), with and without adding water. Optimised FFB SMEDDS formulations were then selected for in-vivo bioavailability study.
Key findings
The release rates of FFB from TPGS/Tween 20 systems were faster than those from TPGS/Tween 80 systems at the same Km value. In both systems, the release rates of FFB increased with a decrease in the Km value. Furthermore, both the release rates and the amounts of FFB from MDS in the water medium decreased with an increasing percentage of Smix added to both water contents. However, the release rates and amounts of FFB from MDSs increased with an increasing percentage of Smix in a 0.025 M sodium lauryl sulfate (SLS) solution. It was further illustrated that the release of FFB from SMEDDSs was complete within 30 min in both the 0.025 M SLS solution and water medium, but the release of FFB from Tricor® or MDSs was limited in water medium. An optimised FFB SMEDDS with either Tween 20(E5(20)) or Tween 80(E5(80)) and one MDS were selected for a pharmacokinetic study to compare with Tricor®. The results demonstrated that the area under the receiver operating curve and Cmax values were in the order of Tricor® > E5(80) ≅ E5(20) > MDS and Tricor® ≅ E5(80) > E5(20) > MDS, respectively.
Conclusions
The absorption of drug carried by SMEDDS might not be enhanced as a result of the smaller volume of water taken with oral administration of SMEDDSs and the agitation rate of the gastrointestinal tract not being strong enough to efficiently promote the self-microemulsification process to facilitate the in-vivo dissolution rate.
Collapse
Affiliation(s)
- Jyh-Ding Wei
- Department of Orthopedics, Shin Kong Wu Ho-Su Memorial Hospital, 95 Wenchang Road, Taiwan
| | - Hsiu-O Ho
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan
| | - Chien-Ho Chen
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan
| | - Wen-Tin Ke
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan
| | - Eric Tsu-Hsin Chen
- Department of Orthopedics, Shin Kong Wu Ho-Su Memorial Hospital, 95 Wenchang Road, Taiwan
| | - Ming-Thau Sheu
- School of Pharmacy, College of Pharmacy, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan
| |
Collapse
|
44
|
Long-term follow-up of bezafibrate treatment in patients with the myopathic form of carnitine palmitoyltransferase 2 deficiency. Clin Pharmacol Ther 2010; 88:101-8. [PMID: 20505667 DOI: 10.1038/clpt.2010.55] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Carnitine palmitoyltransferase 2 (CPT2) deficiency is a rare mitochondrial fatty acid oxidation (FAO) disorder characterized by myalgia, exercise intolerance, and rhabdomyolysis. We evaluate the efficacy of bezafibrate (BZ), a hypolipidemic drug, as a treatment for this form of CPT2 deficiency. A pilot trial was conducted with BZ in six patients for 6 months. There was a follow-up period of 3 years. The oxidation rates of the long-chain fatty acid derivative palmitoyl-CoA, measured in the mitochondria of the patients' muscles, were markedly lower than normal before treatment and increased significantly (+39 to +206%; P = 0.028) in all patients after BZ treatment. The evaluation of the therapeutic effects by the patients themselves (using the Short Form Health Survey (SF-36)), as well as by the physicians, indicated an improvement in the condition of the patients; there was an increase in physical activity and a decline in muscular pain. The results suggest that BZ has a therapeutic effect in the muscular form of CPT2 deficiency.
Collapse
|
45
|
Han Y, Do MH, Kim MS, Seo E, Park MK, Kim DK, Lee HJ, Seo SY. Fenofibrate Reduces Age-related Hypercholesterolemia in Normal Rats on a Standard Diet. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2010; 14:77-81. [PMID: 20473378 DOI: 10.4196/kjpp.2010.14.2.77] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 04/09/2010] [Accepted: 04/16/2010] [Indexed: 01/22/2023]
Abstract
Plasma cholesterol is increased in normal aging in both rodents and humans. This is associated with reduced elimination of cholesterol and decreased receptor-mediated clearance of plasma low-density lipoprotein (LDL) cholesterol. The aims of this study were: (1) to determine age-related changes in plasma lipid profiles, and (2) to determine the effect of fenofibrate, an activator of peroxisome proliferator activated receptor alpha (PPAR alpha), on plasma lipid profiles in normal rats on a standard diet. Male Sprague-Dawley (SD) rats (n=15) were fed standard chow and water from 10 to 25 weeks of age. During that period, we measured daily food intake, body weight, fasting and random blood glucose levels, plasma total cholesterol (TC), triglycerides (TG), and free fatty acid (FFA) levels. At 20 weeks of age, all rats were randomly divided into two groups: a fenofibrate group (in which rats were gavaged with 300 mg/kg/day of fenofibrate) and a control group (gavaged with water). Fenofibrate treatment lasted 5 weeks. There were no significant changes in daily food intake, blood glucose, and plasma TG level with age. Body weight, plasma TC, and FFA levels were significantly increased with age. Fenofibrate significantly decreased plasma concentrations of TC and FFA, which had been increased with age. However, fenofibrate did not influence the plasma concentration of TG, which had not increased with age. These results suggest that fenofibrate might have a novel role in preventing age-related hypercholesterolemia in SD rats on a normal diet.
Collapse
Affiliation(s)
- Ying Han
- Department of Pharmacology, Dong-A University College of Medicine, Medical Science Research Center, Busan 602-714, Korea
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Fenofibrate causes regression of endometriotic implants: a rat model. Fertil Steril 2009; 92:2100-2. [DOI: 10.1016/j.fertnstert.2009.05.065] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2007] [Revised: 05/20/2009] [Accepted: 05/27/2009] [Indexed: 11/21/2022]
|
47
|
Filippatos T, Milionis HJ. Treatment of hyperlipidaemia with fenofibrate and related fibrates. Expert Opin Investig Drugs 2008; 17:1599-614. [PMID: 18808320 DOI: 10.1517/13543784.17.10.1599] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Fenofibrate is the most widely used fibrate. Its efficacy and tolerability in the treatment of hypertriglyceridaemia and combined hyperlipidaemia have been demonstrated in several clinical trials. OBJECTIVE To review the pharmacology, lipid-lowering and extra-lipid effects of fenofibrate and to preview ABT-335, an investigational new fenofibric acid molecule. RESULTS The effects of fenofibrate are mediated through the active metabolite fenofibric acid, and are described in detail in the paper. ABT-335 is a salt of fenofibric acid and, unlike fenofibrate, does not require first pass metabolism to the active moiety. ABT-335 is being developed for combination use with statins, and has recently completed three large Phase III randomised controlled trials in which the efficacy and safety of ABT-335 in combination with the three most commonly prescribed statins, atorvastatin, simvastatin and rosuvastatin, was evaluated in patients with mixed dyslipidaemia. CONCLUSION ABT-335 in combination with statins may provide a safe and efficacious treatment modality that enables achievement of several therapeutic goals in patients with mixed dyslipidaemia who have high cardiovascular risk.
Collapse
Affiliation(s)
- Theodosios Filippatos
- University of Ioannina, School of Medicine, Department of Internal Medicine, 451 10 Ioannina, Greece
| | | |
Collapse
|
48
|
Cree MG, Newcomer BR, Read LK, Sheffield-Moore M, Paddon-Jones D, Chinkes D, Aarsland A, Wolfe RR. Plasma triglycerides are not related to tissue lipids and insulin sensitivity in elderly following PPAR-alpha agonist treatment. Mech Ageing Dev 2007; 128:558-65. [PMID: 17904198 PMCID: PMC3503242 DOI: 10.1016/j.mad.2007.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2006] [Revised: 08/05/2007] [Accepted: 08/09/2007] [Indexed: 12/25/2022]
Abstract
Increases in plasma lipids, tissue triglycerides and decreases in mitochondrial function have been linked to insulin resistance and aging. In animals, peroxisome proliferator-activated receptor-alpha (PPAR-alpha) agonists decrease plasma lipids, intramyocellular fat (IMCL) and liver fat (LFAT) and improve mitochondrial beta-oxidative function and insulin sensitivity, but the effects in elderly were not known. Insulin sensitivity was assessed with a 2-h oral glucose tolerance test, magnetic resonance spectroscopy was used to asses IMCL, LFAT and plasma lipids were measured before and after 6, 11 and 61 days of PPAR-alpha agonist (fenofibrate) administration in 19 elderly (age 70+/-1 years) volunteers. Volunteers were stratified into healthy (N=7) and insulin resistant (N=12) groups. The baseline insulin sensitivity index (8.1+/-1.2 vs. 3.8+/-0.5, healthy vs. insulin resistant; P<0.001) was significantly higher in the healthy group. Fenofibrate treatment induced significant reductions in plasma triglycerides (P<0.001) and total cholesterol (P<0.001) in both groups. Nonetheless, neither fasted free fatty acids, glucose, insulin, nor insulin sensitivity improved in either group (day 1 vs. day 61, 8.1+/-1.2 vs. 8.1+/-0.9, healthy; and 3.8+/-0.5 vs. 4.2+/-0.05, insulin resistant). Furthermore, there was no change in IMCL or LFAT. These results indicate that whereas fenofibrate significantly lowers plasma lipids it neither affects insulin sensitivity nor intracellular lipids in elderly.
Collapse
Affiliation(s)
- Melanie G. Cree
- Dept of Preventive Medicine and Community Health, The University of Texas Medical Branch Galveston, TX 77555
| | - Bradley R. Newcomer
- Department of Diagnostic and Therapeutic Sciences, School of Health Related Professions, Univ. of Alabama at Birmingham, Birmingham, AL 35294
| | - Laura K. Read
- Metabolism Unit, Dept. of Surgery, The University of Texas Medical Branch Galveston, TX 77555
| | - Melinda Sheffield-Moore
- Metabolism Unit, Dept. of Surgery, The University of Texas Medical Branch Galveston, TX 77555
| | - Douglas Paddon-Jones
- Metabolism Unit, Dept. of Surgery, The University of Texas Medical Branch Galveston, TX 77555
| | - David Chinkes
- Dept of Preventive Medicine and Community Health, The University of Texas Medical Branch Galveston, TX 77555
- Metabolism Unit, Dept. of Surgery, The University of Texas Medical Branch Galveston, TX 77555
| | - Asle Aarsland
- Dept. of Anesthesiology, The University of Texas Medical Branch Galveston, TX 77555
| | - Robert R. Wolfe
- Dept of Preventive Medicine and Community Health, The University of Texas Medical Branch Galveston, TX 77555
- Metabolism Unit, Dept. of Surgery, The University of Texas Medical Branch Galveston, TX 77555
- Dept. of Anesthesiology, The University of Texas Medical Branch Galveston, TX 77555
| |
Collapse
|
49
|
Hong XZ, Li LD, Wu LM. Effects of fenofibrate and xuezhikang on high-fat diet-induced non-alcoholic fatty liver disease. Clin Exp Pharmacol Physiol 2007; 34:27-35. [PMID: 17201732 DOI: 10.1111/j.1440-1681.2007.04547.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
1. Fenofibrate and xuezhikang are two types of drugs widely used in the treatment of dyslipidaemia in China. The main purpose of present study was to test the efficacies and explore the potential mechanisms of action of the two lipid-lowering agents on high-fat diet-induced non-alcoholic fatty liver disease (NAFLD). 2. Rats were randomly divided into four groups, with eight rats per group. One group was given normal diet, whereas the other three groups were fed a high-fat diet. Forty-two days later, two of the high-fat diet-fed groups were administered fenofibrate (100 mg/kg, p.o.) and xuezhikang (300 mg/kg, p.o.) for another 42 consecutive days. The other two groups were administered placebo (saline) by gavage. 3. Typical pathological symptoms of NAFLD occurred in the high-fat diet groups. Fenofibrate and xuezhikang treatment markedly improved NAFLD, ameliorating dyslipidaemia and fat accumulation in the liver, improving insulin resistance and ameliorating oxidative stress. Hepatic steatosis, necro-inflammation and collagen deposition were lessened in the drug-treated groups. However, both xuezhikang and fenofibrate failed to reverse hepatomegaly and fenofibrate even aggravated it. Xuezhikang reversed aminotransferase abnormalities, but fenofibrate had less of an effect. 4. The common therapeutic mechanism of action of fenofibate and xuezhikang likely involves inhibition of the hepatic expression of tumour necrosis factor-alpha. Fenofibrate upregulated mRNA levels of peroxisome proliferator-activated receptor (PPAR) alpha in the liver, whereas xuezhikang had no effect on the hepatic expression of PPARalpha and this may explain, in part, their different effects on the NAFLD rats. 5. The results suggest that fenofibrate and xuezhikang may have potential clinical application in the treatment of NAFLD. However, the side-effects of fenofibrate and the underlying constituents of xuezhikang need to be determined and investigated further.
Collapse
Affiliation(s)
- Xue Zhi Hong
- Institute of Chinese Herb Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | | | | |
Collapse
|
50
|
Serisier S, Briand F, Ouguerram K, Siliart B, Magot T, Nguyen P. Fenofibrate lowers lipid parameters in obese dogs. J Nutr 2006; 136:2037S-2040S. [PMID: 16772490 DOI: 10.1093/jn/136.7.2037s] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Samuel Serisier
- Endocrinology and Nutrition Unit, National Veterinary School of Nantes, France
| | | | | | | | | | | |
Collapse
|