1
|
Shehab M, De Marco D, Lakatos PL, Bessissow T. The potential for medical therapies to address fistulizing Crohn's disease: a state-of-the-art review. Expert Opin Biol Ther 2024; 24:733-746. [PMID: 39045643 DOI: 10.1080/14712598.2024.2383882] [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] [Received: 02/27/2024] [Revised: 06/25/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Crohn's disease (CD) is a chronic, relapsing immune mediated disease, which is one of the two major types of inflammatory bowel disease (IBD). Fistulizing CD poses a significant clinical challenge for physicians. Effective management of CD requires a multidisciplinary approach, involving a gastroenterologist and a GI surgeon while tailoring treatment to each patient's unique risk factors, clinical representations, and preferences. AREAS COVERED This comprehensive review explores the intricacies of fistulizing CD including its manifestations, types, impact on quality of life, management strategies, and novel therapies under investigation. EXPERT OPINION Antibiotics are often used as first-line therapy to treat symptoms. Biologics that selectively target TNF-α, such infliximab (IFX), have shown high efficacy in randomized controlled trials. However, more than 50% of patients lose response to IFX, prompting them to explore alternative strategies. Current options include adalimumab and certolizumab pegol combination therapies, as well as small-molecule drugs targeting Janus kinases such as Upadacitinib. Furthermore, a promising treatment for complex fistulas is mesenchymal stem cells such as Darvadstrocel (Alofisel), an allogeneic stem cell-based therapy. However, surgical interventions are necessary for complex cases or intra-abdominal complications. Setons and LIFT procedures are the most common surgical options.
Collapse
Affiliation(s)
- Mohammad Shehab
- Division of Gastroenterology, Department of Internal Medicine, Mubarak Al-Kabeer University Hospital, Kuwait University, Kuwait City, Kuwait
| | - Davide De Marco
- Division of Gastroenterology and Hepatology, Department of Medicine, McGill University Health Center, Montreal, Canada
| | - Peter L Lakatos
- Division of Gastroenterology and Hepatology, Department of Medicine, McGill University Health Center, Montreal, Canada
- 1st Department of Medicine, Semmelweis University, Budapest, Hungary
| | - Talat Bessissow
- Division of Gastroenterology and Hepatology, Department of Medicine, McGill University Health Center, Montreal, Canada
| |
Collapse
|
2
|
David E, Hawkins K, Shokrian N, Del Duca E, Guttman-Yassky E. Monoclonal antibodies for moderate-to-severe atopic dermatitis: a look at phase III and beyond. Expert Opin Biol Ther 2024; 24:471-489. [PMID: 38888099 DOI: 10.1080/14712598.2024.2368192] [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] [Received: 04/10/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024]
Abstract
INTRODUCTION The understanding of atopic dermatitis (AD) pathogenesis has rapidly expanded in recent years, catalyzing the development of new targeted monoclonal antibody treatments for AD. AREAS COVERED This review aims to summarize the latest clinical and molecular data about monoclonal antibodies that are in later stages of development for AD, either in Phase 3 trials or in the pharmacopoeia for up to 5 years, highlighting the biologic underpinning of each drug's mechanism of action and the potential modulation of the AD immune profile. EXPERT OPINION The therapeutic pipeline of AD treatments is speedily progressing, introducing the potential for a personalized medical approach in the near future. Understanding how targeting pathogenic players in AD modifies disease progression and symptomatology is key in improving therapeutic choices for patients and identifying ideal patient candidates.
Collapse
Affiliation(s)
- Eden David
- Department of Dermatology, Icahn school of Medicine at Mount Sinai, New York, NY, USA
| | - Kelly Hawkins
- Department of Dermatology, Icahn school of Medicine at Mount Sinai, New York, NY, USA
- Department of Dermatology, Albert Einstein College of Medicine, New York, NY, USA
| | - Neda Shokrian
- Department of Dermatology, Icahn school of Medicine at Mount Sinai, New York, NY, USA
- Department of Dermatology, Albert Einstein College of Medicine, New York, NY, USA
| | - Ester Del Duca
- Department of Dermatology, Icahn school of Medicine at Mount Sinai, New York, NY, USA
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Emma Guttman-Yassky
- Department of Dermatology, Icahn school of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
3
|
Ayoub M, Mattay S, Yarur AJ, Deepak P. Managing Risks with Newer Oral Small Molecules in Patients with Inflammatory Bowel Diseases. Curr Gastroenterol Rep 2024; 26:145-156. [PMID: 38353899 DOI: 10.1007/s11894-024-00923-x] [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] [Accepted: 01/29/2024] [Indexed: 05/12/2024]
Abstract
PURPOSE OF REVIEW Treatment of Inflammatory Bowel Diseases (IBD) is challenging; thus, the need for newer therapeutic options with an oral route of administration has led to the development of novel small molecules drugs (SMDs). We aim to highlight the most common Adverse events (AEs) associated with SMDs and recommendations on monitoring for AEs before and during treatment. RECENT FINDINGS SMDs, such as Tofacitinib, a JAK inhibitor, have been associated with laboratory abnormalities, infections, and risk of thromboembolic events. Therefore, oral agents with greater selectivity in JAK inhibition, such as tofacitinib and upadacitinib, were later developed. Ozanimod and etrasimod, S1PR agonists, require closer safety profile monitoring by clinicians. Multiple therapies have been recently developed with variable efficacy. However, they have been associated with AEs, and some require close monitoring prior to and during therapy. Clinicians should highlight these adverse events to patients while reassuring the safety profile of these novel SMDs for IBD is favorable.
Collapse
Affiliation(s)
- Malek Ayoub
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Shivani Mattay
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Andres J Yarur
- Division of Gastroenterology and Hepatology, Cedars Sinai Medical Center, Inflammatory Bowel Disease Institute, Los Angeles, CA, USA
| | - Parakkal Deepak
- Division of Gastroenterology, John T. Milliken Department of Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, Campus, Box 8124, St. Louis, MO, 63110, USA.
| |
Collapse
|
4
|
Zou H, Hao P, Cao Y, Li L, Ding R, Bai X, Xue Y. Hippophae rhamnoides reverses decreased CYP2D6 expression in rats with BCG-induced liver injury. Sci Rep 2023; 13:17425. [PMID: 37833431 PMCID: PMC10575986 DOI: 10.1038/s41598-023-44590-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 10/10/2023] [Indexed: 10/15/2023] Open
Abstract
In this study, we investigated the effect of Hippophae rhamnoides L. (HRP) on the activity of CYP2D6 via the CAMP/PKA/NF-κB pathway in rats with Bacille Calmette-Guerin (BCG)-induced immunological liver injury. BCG (125 mg/kg) was injected to establish the rat model of liver injury. HRP was administered intragastrically for one week as the intervention drug. Proteomics techniques were used to analyze protein expression levels, obtaining a comprehensive understanding of the liver injury process. ELISA or western blotting was used to detect specific protein levels. Dextromethorphan was detected using high-performance liquid chromatography to reflect the metabolic activity of CYP2D6. BCG downregulated the expression of CYP2D6, cAMP, PKA, IκB, and P-CREB and upregulated that of NF-κB, IL-1β, TNF-α, and CREB in the liver; HRP administration reversed these effects. Therefore, HRP may restore the metabolic function of the liver by reversing the downregulation of CYP2D6 through inhibition of NF-κB signal transduction and regulation of the cAMP/PKA/CREB/CYP2D6 pathway. These findings highlight the role of HRP as an alternative clinical drug for treating hepatitis B and other immune-related liver diseases.
Collapse
Affiliation(s)
- Huiqiong Zou
- Institute of Pharmacokinetics and Liver Molecular Pharmacology, Department of Pharmacology, Baotou Medical College, No. 31 Jianshe Road, Donghe District, Baotou, 014060, China
| | - Peipei Hao
- Institute of Pharmacokinetics and Liver Molecular Pharmacology, Department of Pharmacology, Baotou Medical College, No. 31 Jianshe Road, Donghe District, Baotou, 014060, China
| | - Yingying Cao
- Institute of Pharmacokinetics and Liver Molecular Pharmacology, Department of Pharmacology, Baotou Medical College, No. 31 Jianshe Road, Donghe District, Baotou, 014060, China
| | - Li Li
- Institute of Pharmacokinetics and Liver Molecular Pharmacology, Department of Pharmacology, Baotou Medical College, No. 31 Jianshe Road, Donghe District, Baotou, 014060, China
| | - Ruifeng Ding
- Department of Gastroenterology, First Affiliated Hospital, Baotou Medical College, Baotou, China
| | - Xuefeng Bai
- Department of Pathology, Baotou Cancer Hospital, Baotou, China
| | - Yongzhi Xue
- Institute of Pharmacokinetics and Liver Molecular Pharmacology, Department of Pharmacology, Baotou Medical College, No. 31 Jianshe Road, Donghe District, Baotou, 014060, China.
| |
Collapse
|
5
|
Vishwakarma RK, Negi DS, Negi A. Abortitristoside A and desrhamnosylverbanscoside: the potential COX-2 inhibitor from the leaves of Nyctanthes arbor-tristis as anti-inflammatory agents based on the in vitro assay, molecular docking and ADMET prediction. CHEMICAL PAPERS 2023. [DOI: 10.1007/s11696-023-02686-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
6
|
Ceban F, Subramaniapillai M, Rosenblat JD, Mansur RB, McIntyre RS. Can COVID-19 have a clinically significant effect on drug metabolism? Expert Opin Drug Saf 2023; 22:1-3. [PMID: 36691834 DOI: 10.1080/14740338.2023.2172158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Felicia Ceban
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada.,Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada.,Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | | | - Joshua D Rosenblat
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Rodrigo B Mansur
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada.,Brain and Cognition Discovery Foundation, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
7
|
Zhao J, Yuan W, Wang S, Zhang H, Chen D, Niu X, Liu X, Liu L, Gao J. Comparative Pharmacokinetics and Tissue Distribution of M10 and Its Metabolite Myricetin in Normal and Dextran-Sodium-Sulfate-Induced Colitis Mice. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238140. [PMID: 36500233 PMCID: PMC9740244 DOI: 10.3390/molecules27238140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/09/2022] [Accepted: 11/20/2022] [Indexed: 11/25/2022]
Abstract
M10, a novel myricetin derivative, is an anti-inflammatory agent designed for treatment of colitis. Here, we aim to investigate its pharmacokinetic behavior and tissue distribution in a mouse model with colitis. Pharmacokinetics and tissue distribution of M10 and its metabolite myricetin were compared in normal mice and in dextran-sodium-sulfate (DSS)-induced colitis mice. The role of fecal microbiota was also analyzed during metabolism of M10 in vitro. After oral administration, M10 was very low in the plasma of both normal and diseased mice. However, both M10 and myricetin were mainly distributed in the gastrointestinal tract, including the stomach, colon and small intestine, in physiological and pathological conditions. Significantly, M10 and myricetin were found in higher levels in gastrointestinal tracts with inflamed tissues than in normal tissues of mice. An in vitro assay revealed that 80% of M10 was metabolized to myricetin via fecal microbiota. After oral administration, M10 was not absorbed into circulation but mainly distributed in the inflamed submucosal tissues of colitic mice, where it was metabolized into myricetin to prevent colitis development.
Collapse
Affiliation(s)
- Jianchun Zhao
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Marine Biomedical Research Institute of Qingdao, Qingdao 266073, China
- Correspondence:
| | - Wenmin Yuan
- Marine Biomedical Research Institute of Qingdao, Qingdao 266073, China
| | - Shixiao Wang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Hongwei Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Marine Biomedical Research Institute of Qingdao, Qingdao 266073, China
| | - Dan Chen
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Xiaochen Niu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Xiaochun Liu
- Marine Biomedical Research Institute of Qingdao, Qingdao 266073, China
| | - Li Liu
- Marine Biomedical Research Institute of Qingdao, Qingdao 266073, China
| | - Jiangming Gao
- Marine Biomedical Research Institute of Qingdao, Qingdao 266073, China
| |
Collapse
|
8
|
Development of potential COX inhibitor as anti-inflammatory agents from leaves of Lantana camara by in-vitro analysis, molecular docking and ADMET prediction. J INDIAN CHEM SOC 2022. [DOI: 10.1016/j.jics.2022.100694] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
9
|
Lenoir C, Niederer A, Rollason V, Desmeules JA, Daali Y, Samer CF. Prediction of cytochromes P450 3A and 2C19 modulation by both inflammation and drug interactions using physiologically based pharmacokinetics. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 11:30-43. [PMID: 34791831 PMCID: PMC8752107 DOI: 10.1002/psp4.12730] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/16/2021] [Accepted: 10/01/2021] [Indexed: 12/22/2022]
Abstract
Xenobiotics can interact with cytochromes P450 (CYPs), resulting in drug-drug interactions, but CYPs can also contribute to drug-disease interactions, especially in the case of inflammation, which downregulates CYP activities through pretranscriptional and posttranscriptional mechanisms. Interleukin-6 (IL-6), a key proinflammatory cytokine, is mainly responsible for this effect. The aim of our study was to develop a physiologically based pharmacokinetic (PBPK) model to foresee the impact of elevated IL-6 levels in combination with drug interactions with esomeprazole on CYP3A and CYP2C19. Data from a cohort of elective hip surgery patients whose CYP3A and CYP2C19 activities were measured before and after surgery were used to validate the accurate prediction of the developed models. Successive steps were to fit models for IL-6, esomeprazole, and omeprazole and its metabolite from the literature and to validate them. The models for midazolam and its metabolite were obtained from the literature. When appropriate, a correction factor was applied to convert drug concentrations from whole blood to plasma. Mean ratios between simulated and observed areas under the curve for omeprazole/5-hydroxy omeprazole, esomeprazole, and IL-6 were 1.53, 1.06, and 0.69, respectively, indicating an accurate prediction of the developed models. The impact of IL-6 and esomeprazole on the exposure to CYP3A and CYP2C19 probe substrates and respective metabolites were correctly predicted. Indeed, the ratio between predicted and observed mean concentrations were <2 for all observations (ranging from 0.51 to 1.7). The impact of IL-6 and esomeprazole on CYP3A and CYP2C19 activities after a hip surgery were correctly predicted with the developed PBPK models.
Collapse
Affiliation(s)
- Camille Lenoir
- Division of Clinical Pharmacology and Toxicology, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Amine Niederer
- Division of Clinical Pharmacology and Toxicology, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Victoria Rollason
- Division of Clinical Pharmacology and Toxicology, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jules Alexandre Desmeules
- Division of Clinical Pharmacology and Toxicology, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Youssef Daali
- Division of Clinical Pharmacology and Toxicology, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Caroline Flora Samer
- Division of Clinical Pharmacology and Toxicology, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
10
|
Texler B, Zollner A, Reinstadler V, Reider SJ, Macheiner S, Jelusic B, Pfister A, Watschinger C, Przysiecki N, Tilg H, Oberacher H, Moschen AR. Tofacitinib-Induced Modulation of Intestinal Adaptive and Innate Immunity and Factors Driving Cellular and Systemic Pharmacokinetics. Cell Mol Gastroenterol Hepatol 2021; 13:383-404. [PMID: 34624526 PMCID: PMC8688178 DOI: 10.1016/j.jcmgh.2021.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 09/12/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE By interfering with multiple cytokines, human Janus kinase inhibitors (JAKis) are of growing importance in the treatment of malignant and inflammatory conditions. Although tofacitinib has demonstrated efficacy as the first-in-class JAKi in ulcerative colitis many aspects concerning its mode of action and pharmacokinetics remain unresolved. DESIGN We studied tofacitinib's impact on various primary human innate and adaptive immune cells. In-depth in vivo studies were performed in dextran sodium sulfate-induced colitis in mice. Immune populations were characterized by flow cytometry and critical transcription factors and effector cytokines were analyzed. Pharmacokinetics of tofacitinib was studied by liquid chromatography-tandem mass spectrometry. RESULTS Tofacitinib inhibited proliferation in CD4+ and CD8+ T cells along with Th1 and Th17 differentiation, while Th2 and regulatory T cell lineages were largely unaffected. Monocytes and macrophages were directed toward an anti-inflammatory phenotype and cytokine production was suppressed in intestinal epithelial cells. These findings were largely reproducible in murine cells of the inflamed mucosa in dextran sulfate sodium colitis. Short-term treatment with tofacitinib had little impact on the mouse microbiota. Strikingly, the degree of inflammation and circulating tofacitinib levels showed a strong positive correlation. Finally, we identified inflammation-induced equilibrative nucleoside transporters as regulators of tofacitinib uptake into leukocytes. CONCLUSIONS We provide a detailed analysis of the cell-specific immune-suppressive effects of the JAKis tofacitinib on innate and adaptive immunity and reveal that intestinal inflammation critically impacts tofacitinib's pharmacokinetics in mice. Furthermore, we describe an unappreciated mechanism-namely induction of equilibrative nucleoside transporters-enhancing baseline cellular uptake that can be inhibited pharmaceutically.
Collapse
Affiliation(s)
- Bernhard Texler
- Christian Doppler Laboratory for Mucosal Immunology, Johannes Kepler University Linz, Linz, Austria; Daniel Swarovski Laboratory, Medical University Innsbruck, Innsbruck, Austria
| | - Andreas Zollner
- Christian Doppler Laboratory for Mucosal Immunology, Johannes Kepler University Linz, Linz, Austria; Division of Internal Medicine I (Gastroenterology, Hepatology, Endocrinology, and Metabolism), Department of Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Vera Reinstadler
- Institute of Legal Medicine and Core Facility Metabolomics, Medical University Innsbruck, Innsbruck, Austria
| | - Simon J Reider
- Christian Doppler Laboratory for Mucosal Immunology, Johannes Kepler University Linz, Linz, Austria; Internal Medicine 2 (Gastroenterology and Hepatology), Kepler University Hospital, Faculty of Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Sophie Macheiner
- Christian Doppler Laboratory for Mucosal Immunology, Johannes Kepler University Linz, Linz, Austria; Division of Internal Medicine I (Gastroenterology, Hepatology, Endocrinology, and Metabolism), Department of Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Barbara Jelusic
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Alexandra Pfister
- Christian Doppler Laboratory for Mucosal Immunology, Johannes Kepler University Linz, Linz, Austria; Division of Internal Medicine I (Gastroenterology, Hepatology, Endocrinology, and Metabolism), Department of Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Christina Watschinger
- Christian Doppler Laboratory for Mucosal Immunology, Johannes Kepler University Linz, Linz, Austria; Internal Medicine 2 (Gastroenterology and Hepatology), Kepler University Hospital, Faculty of Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Nicole Przysiecki
- Christian Doppler Laboratory for Mucosal Immunology, Johannes Kepler University Linz, Linz, Austria; Division of Internal Medicine I (Gastroenterology, Hepatology, Endocrinology, and Metabolism), Department of Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Division of Internal Medicine I (Gastroenterology, Hepatology, Endocrinology, and Metabolism), Department of Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Herbert Oberacher
- Institute of Legal Medicine and Core Facility Metabolomics, Medical University Innsbruck, Innsbruck, Austria
| | - Alexander R Moschen
- Christian Doppler Laboratory for Mucosal Immunology, Johannes Kepler University Linz, Linz, Austria; Internal Medicine 2 (Gastroenterology and Hepatology), Kepler University Hospital, Faculty of Medicine, Johannes Kepler University Linz, Linz, Austria.
| |
Collapse
|
11
|
Ben Ghezala I, Charkaoui M, Michiels C, Bardou M, Luu M. Small Molecule Drugs in Inflammatory Bowel Diseases. Pharmaceuticals (Basel) 2021; 14:ph14070637. [PMID: 34209234 PMCID: PMC8308576 DOI: 10.3390/ph14070637] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/22/2022] Open
Abstract
Inflammatory bowel diseases (IBDs), mainly represented by Crohn’s disease (CD) and Ulcerative Colitis (UC), are chronic disorders with an unclear pathogenesis. This incurable and iterative intestinal mucosal inflammation requires the life-long use of anti-inflammatory drugs to prevent flares or relapses, which are the major providers of complications, such as small bowel strictures and intestinal perforations. The introduction of tumor necrosis factor (TNF)-alpha inhibitors and other compounds, such as anti-IL12/23 and anti-alpha4/beta7 integrin monoclonal antibodies, has considerably improved the clinical management of IBDs. They are now the standard of care, being the first-line therapy in patients with aggressive disease and in patients with moderate to severe disease with an inadequate response to conventional therapy. However, for approximately one third of all patients, their efficacy remains insufficient by a lack or loss of response due to the formation of anti-drug antibodies or compliance difficulties with parenteral formulations. To address these issues, orally administered Small Molecules Drugs (SMDs) that use a broad range of novel pharmacological pathways, such as JAK inhibitors, sphingosine-1-phosphate receptor modulators, and phosphodiesterase 4 inhibitors, have been developed for CD and UC. This article provides an updated and complete review of the most recently authorized SMDs and SMDs in phase II/III development.
Collapse
Affiliation(s)
- Inès Ben Ghezala
- INSERM, CIC1432, Plurithematic Unit, 21079 Dijon, France; (I.B.G.); (M.B.)
- Clinical Investigation Center, Plurithematic Unit, Dijon Bourgogne University Hospital, 21079 Dijon, France
- Ophthalmology Department, Dijon Bourgogne University Hospital, 21079 Dijon, France
| | - Maëva Charkaoui
- Gastroenterology Department, Dijon Bourgogne University Hospital, 21079 Dijon, France; (M.C.); (C.M.)
| | - Christophe Michiels
- Gastroenterology Department, Dijon Bourgogne University Hospital, 21079 Dijon, France; (M.C.); (C.M.)
| | - Marc Bardou
- INSERM, CIC1432, Plurithematic Unit, 21079 Dijon, France; (I.B.G.); (M.B.)
- Clinical Investigation Center, Plurithematic Unit, Dijon Bourgogne University Hospital, 21079 Dijon, France
- Gastroenterology Department, Dijon Bourgogne University Hospital, 21079 Dijon, France; (M.C.); (C.M.)
| | - Maxime Luu
- INSERM, CIC1432, Plurithematic Unit, 21079 Dijon, France; (I.B.G.); (M.B.)
- Clinical Investigation Center, Plurithematic Unit, Dijon Bourgogne University Hospital, 21079 Dijon, France
- Correspondence:
| |
Collapse
|
12
|
White CM, Sicignano DJ, Smith K. Impact of Interferons and Biological Drug Inhibitors of IL-2 and IL-6 on Small-Molecule Drug Metabolism Through the Cytochrome P450 System. Ann Pharmacother 2021; 56:170-180. [PMID: 34078115 DOI: 10.1177/10600280211022281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE Assess the impact of interferons and interleukin (IL)-2 and IL-6 inhibitors on cytochrome P450 (CYP) drug metabolism in human subjects. DATA SOURCES PubMed search from 1980 to March 31, 2021, limited to human subjects and English language via search strategy: (biological drug names) [AND] (cytochrome [OR] CYP metabolism). STUDY SELECTION AND DATA EXTRACTION Narrative review of human studies assessing biological drugs in select classes that affect CYP drug metabolism. DATA SYNTHESIS Exogenous interferons suppress CYP1A2 (theophylline, caffeine, antipyrone) clearance by 20% to 49% in patients; have minimal impact on CYP3A4 (midazolam and dapsone), CYP2C9 (tolbutamide), or CYP2C19 (mephenytoin) metabolism; and increase CYP2D6 (debrisoquine, dextromethorphan) metabolism. Biological IL-2 inhibitors (basiliximab, daclizumab) have no effect on metabolism via CYP1A2 (caffeine), CYP2C9 (s-warfarin), CYP2C19 (omeprazole), CYP2D6 (dextromethorphan), and CYP3A4 (midazolam, tacrolimus) but may enhance CYP3A4 (cyclosporin) metabolism over time. IL-6 inhibitors (sirukumab, tocilizumab, sarilumab) significantly enhance metabolism via CYP2C9 (s-warfarin), CYP2C19 (omeprazole), and CYP3A4 (simvastatin, midazolam) and reduce metabolism via CYP1A2 (caffeine). RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE Patients using interferons, IL-2, or IL-6 blocking drugs at steady state with CYP substrates could have altered drug metabolism and experience adverse events. With interferons and biological anti-inflammatory drugs, some isoenzymes will be inhibited, whereas others will be enhanced, and the magnitude of the effect can sometimes be significant. In clinical practice, clinicians may consider these metabolic changes as an additive effect to a patient's entire disease and medication profile when determining risk/benefit of treatment. CONCLUSIONS Interferon therapy or inflammatory suppression via IL-2 or IL-6 can alter steady-state concentrations of CYP-metabolized small-molecule drugs.
Collapse
Affiliation(s)
- C Michael White
- University of Connecticut School of Pharmacy, Storrs, CT, USA.,Hartford Hospital, CT, USA
| | | | - Kimberly Smith
- University of Connecticut School of Pharmacy, Storrs, CT, USA
| |
Collapse
|
13
|
Nešković N, Marczi S, Mandić D, Mraovic B, Škiljić S, Kristek G, Vinković H, Kvolik S. ANALGESIC EFFECT OF TRAMADOL IS NOT ALTERED BY POSTOPERATIVE SYSTEMIC INFLAMMATION AFTER MAJOR ABDOMINAL SURGERY. Acta Clin Croat 2021; 60:268-275. [PMID: 34744277 PMCID: PMC8564835 DOI: 10.20471/acc.2021.60.02.13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/29/2020] [Indexed: 12/04/2022] Open
Abstract
Tramadol is a commonly used analgesic in intensive care units (ICUs) for acute postoperative pain. Conversion of tramadol into active metabolites may be impaired in inflammatory states. Catechol-O-methyltransferase may influence pain. The aim of the study was to examine differences in the analgesic effect of tramadol between ICU patients with and without signs of systemic inflammation. Forty-three patients were admitted to ICU after a major abdominal surgery. The patients received a dose of 100 mg of tramadol intravenously every 6 hours during the first 24 hours after surgical procedure. Pain scores were measured by the Numeric Rating Scale before and 30 minutes after tramadol administration in awake patients. Systemic inflammation was considered when at least two of the following postoperative parameters were present in the first 24 hours of ICU admission: fever or hypothermia, tachycardia, pCO2 <4.3 kPa, white blood cells >12000/mm3 or <4000/mm3, or preoperative value of C-reactive protein (CRP) >50 mg/L or/and procalcitonin (PCT) >0.5 mg/L. Catechol-O-methyltransferase was analyzed postoperatively. Fifteen (34.8%) patients met the criteria for systemic inflammation. Tramadol was proven to be an effective analgesic for the treatment of postoperative pain regardless of the presence of systemic inflammation (p<0.05). Lower perception of pain before tramadol application was observed in patients with systemic inflammation, but the difference was not significant. A negative correlation was observed between the preoperative values of CRP and PCT and the analgesic effect of tramadol assessed at the second measurement point (r=-0.358, p=0.03, and r=-0.364, p=0.02, respectively). Catechol-O-methyltransferase variants were not in correlation with pain and opioid consumption. Based on our findings, tramadol is effective in lowering pain scores after major abdominal surgery irrespective of the presence of systemic inflammation.
Collapse
Affiliation(s)
| | - Saška Marczi
- 1Osijek University Hospital Centre, Department of Anesthesiology, Resuscitation and Intensive Medicine, Osijek, Croatia; 2Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia; 3Osijek University Hospital Centre, Department of Transfusion Medicine, Laboratory of Molecular and HLA Diagnostics, Osijek, Croatia; 4Osijek University Hospital Centre, Department of Clinical and Laboratory Diagnostics, Osijek, Croatia; 5University of Missouri, Department of Anesthesiology and Perioperative Medicine, School of Medicine, Columbia, USA
| | - Dario Mandić
- 1Osijek University Hospital Centre, Department of Anesthesiology, Resuscitation and Intensive Medicine, Osijek, Croatia; 2Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia; 3Osijek University Hospital Centre, Department of Transfusion Medicine, Laboratory of Molecular and HLA Diagnostics, Osijek, Croatia; 4Osijek University Hospital Centre, Department of Clinical and Laboratory Diagnostics, Osijek, Croatia; 5University of Missouri, Department of Anesthesiology and Perioperative Medicine, School of Medicine, Columbia, USA
| | - Boris Mraovic
- 1Osijek University Hospital Centre, Department of Anesthesiology, Resuscitation and Intensive Medicine, Osijek, Croatia; 2Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia; 3Osijek University Hospital Centre, Department of Transfusion Medicine, Laboratory of Molecular and HLA Diagnostics, Osijek, Croatia; 4Osijek University Hospital Centre, Department of Clinical and Laboratory Diagnostics, Osijek, Croatia; 5University of Missouri, Department of Anesthesiology and Perioperative Medicine, School of Medicine, Columbia, USA
| | - Sonja Škiljić
- 1Osijek University Hospital Centre, Department of Anesthesiology, Resuscitation and Intensive Medicine, Osijek, Croatia; 2Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia; 3Osijek University Hospital Centre, Department of Transfusion Medicine, Laboratory of Molecular and HLA Diagnostics, Osijek, Croatia; 4Osijek University Hospital Centre, Department of Clinical and Laboratory Diagnostics, Osijek, Croatia; 5University of Missouri, Department of Anesthesiology and Perioperative Medicine, School of Medicine, Columbia, USA
| | - Gordana Kristek
- 1Osijek University Hospital Centre, Department of Anesthesiology, Resuscitation and Intensive Medicine, Osijek, Croatia; 2Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia; 3Osijek University Hospital Centre, Department of Transfusion Medicine, Laboratory of Molecular and HLA Diagnostics, Osijek, Croatia; 4Osijek University Hospital Centre, Department of Clinical and Laboratory Diagnostics, Osijek, Croatia; 5University of Missouri, Department of Anesthesiology and Perioperative Medicine, School of Medicine, Columbia, USA
| | - Hrvoje Vinković
- 1Osijek University Hospital Centre, Department of Anesthesiology, Resuscitation and Intensive Medicine, Osijek, Croatia; 2Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia; 3Osijek University Hospital Centre, Department of Transfusion Medicine, Laboratory of Molecular and HLA Diagnostics, Osijek, Croatia; 4Osijek University Hospital Centre, Department of Clinical and Laboratory Diagnostics, Osijek, Croatia; 5University of Missouri, Department of Anesthesiology and Perioperative Medicine, School of Medicine, Columbia, USA
| | - Slavica Kvolik
- 1Osijek University Hospital Centre, Department of Anesthesiology, Resuscitation and Intensive Medicine, Osijek, Croatia; 2Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia; 3Osijek University Hospital Centre, Department of Transfusion Medicine, Laboratory of Molecular and HLA Diagnostics, Osijek, Croatia; 4Osijek University Hospital Centre, Department of Clinical and Laboratory Diagnostics, Osijek, Croatia; 5University of Missouri, Department of Anesthesiology and Perioperative Medicine, School of Medicine, Columbia, USA
| |
Collapse
|
14
|
Darakjian L, Deodhar M, Turgeon J, Michaud V. Chronic Inflammatory Status Observed in Patients with Type 2 Diabetes Induces Modulation of Cytochrome P450 Expression and Activity. Int J Mol Sci 2021; 22:ijms22094967. [PMID: 34067027 PMCID: PMC8124164 DOI: 10.3390/ijms22094967] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 02/07/2023] Open
Abstract
Diabetes mellitus is a metabolic disease that causes a hyperglycemic status which leads, over time, to serious damage to the heart, blood vessels, eyes, kidneys and nerves. The most frequent form of diabetes is type 2 diabetes mellitus (T2DM) which is often part of a metabolic syndrome (hyperglycaemia, hypertension, hypercholesterolemia, abdominal obesity) that usually requires the use of several medications from different drug classes to bring each of these conditions under control. T2DM is associated with an increase in inflammatory markers such as interleukin-6 (IL-6) and the tumor necrosis factor alpha (TNF-α). Higher levels of IL-6 and TNF-α are associated with a downregulation of several drug metabolizing enzymes, especially the cytochrome P450 (P450) isoforms CYP3As and CYP2C19. A decrease in these P450 isoenzymes may lead to unexpected rise in plasma levels of substrates of these enzymes. It could also give rise to a mismatch between the genotypes determined for these enzymes, the predicted phenotypes based on these genotypes and the phenotypes observed clinically. This phenomenon is described as phenoconversion. Phenoconversion typically results from either a disease (such as T2DM) or concomitant administration of medications inducing or inhibiting (including competitive or non-competitive inhibition) a P450 isoenzyme used by other substrates for their elimination. Phenoconversion could have a significant impact on drug effects and genotypic-focused clinical outcomes. As the aging population is exposed to polypharmacy along with inflammatory comorbidities, consideration of phenoconversion related to drug metabolizing enzymes is of importance when applying pharmacogenomic results and establishing personalized and more precise drug regimens.
Collapse
Affiliation(s)
- Lucy Darakjian
- Tabula Rasa HealthCare, Precision Pharmacotherapy Research and Development Institute, Orlando, FL 32827, USA; (L.D.); (M.D.); (J.T.)
| | - Malavika Deodhar
- Tabula Rasa HealthCare, Precision Pharmacotherapy Research and Development Institute, Orlando, FL 32827, USA; (L.D.); (M.D.); (J.T.)
| | - Jacques Turgeon
- Tabula Rasa HealthCare, Precision Pharmacotherapy Research and Development Institute, Orlando, FL 32827, USA; (L.D.); (M.D.); (J.T.)
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Veronique Michaud
- Tabula Rasa HealthCare, Precision Pharmacotherapy Research and Development Institute, Orlando, FL 32827, USA; (L.D.); (M.D.); (J.T.)
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H3C 3J7, Canada
- Correspondence: ; Tel.: +1-407-454-9964
| |
Collapse
|
15
|
Jonsson-Schmunk K, Ghose R, Croyle MA. Immunization and Drug Metabolizing Enzymes: Focus on Hepatic Cytochrome P450 3A. Expert Rev Vaccines 2021; 20:623-634. [PMID: 33666138 DOI: 10.1080/14760584.2021.1899818] [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] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Infectious disease emergencies like the 2013-2016 Ebola epidemic and the 2009 influenza and current SARS-CoV-2 pandemics illustrate that vaccines are now given to diverse populations with preexisting pathologies requiring pharmacological management. Many natural biomolecules (steroid hormones, fatty acids, vitamins) and ~60% of prescribed medications are processed by hepatic cytochrome P450 (CYP) 3A4. The objective of this work was to determine the impact of infection and vaccines on drug metabolism. METHODS The impact of an adenovirus-based vaccine expressing Ebola glycoprotein (AdEBO) and H1N1 and H3N2 influenza viruses on hepatic CYP 3A4 and associated nuclear receptors was evaluated in human hepatocytes (HC-04 cells) and in mice. RESULTS CYP3A activity was suppressed by 55% in mice 24 h after administration of mouse-adapted H1N1, while ˂10% activity remained in HC-04 cells after infection with H1N1 and H3N2 due to global suppression of cellular translation capacity, indicated by reduction (70%, H1N1, 56%, H3N2) of phosphorylated eukaryotic translation initiation factor 4e (eIF4E). AdEBO suppressed CYP3A activity in vivo (44%) and in vitro (26%) 24 hours after infection. CONCLUSION As the clinical evaluation of vaccines for SARS-CoV-2 and other global pathogens rise, studies to evaluate the impact of new vaccines and emerging pathogens on CYP3A4 and other metabolic enzymes are warranted to avoid therapeutic failures that could further compromise the public health during infectious disease emergencies.
Collapse
Affiliation(s)
- Kristina Jonsson-Schmunk
- Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas, Austin, Texas, USA
| | - Romi Ghose
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Maria A Croyle
- Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas, Austin, Texas, USA.,LaMontagne Center for Infectious Disease, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
16
|
Fanni D, Pinna F, Gerosa C, Paribello P, Carpiniello B, Faa G, Manchia M. Anatomical distribution and expression of CYP in humans: Neuropharmacological implications. Drug Dev Res 2021; 82:628-667. [PMID: 33533102 DOI: 10.1002/ddr.21778] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022]
Abstract
The cytochrome P450 (CYP450) superfamily is responsible for the metabolism of most xenobiotics and pharmacological treatments generally used in clinical settings. Genetic factors as well as environmental determinants acting through fine epigenetic mechanisms modulate the expression of CYP over the lifespan (fetal vs. infancy vs. adult phases) and in diverse organs. In addition, pathological processes might alter the expression of CYP. In this selective review, we sought to summarize the evidence on the expression of CYP focusing on three specific aspects: (a) the anatomical distribution of the expression in body districts relevant in terms of drug pharmacokinetics (liver, gut, and kidney) and pharmacodynamics, focusing for the latter on the brain, since this is the target organ of psychopharmacological agents; (b) the patterns of expression during developmental phases; and (c) the expression of CYP450 enzymes during pathological processes such as cancer. We showed that CYP isoforms show distinct patterns of expression depending on the body district and the specific developmental phases. Of particular relevance for neuropsychopharmacology is the complex regulatory mechanisms that significantly modulate the complexity of the pharmacokinetic regulation, including the concentration of specific CYP isoforms in distinct areas of the brain, where they could greatly affect local substrate and metabolite concentrations of drugs.
Collapse
Affiliation(s)
- Daniela Fanni
- Unit of Anatomic Pathology, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.,Unit of Anatomic Pathology, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Federica Pinna
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.,Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Clara Gerosa
- Unit of Anatomic Pathology, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.,Unit of Anatomic Pathology, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Pasquale Paribello
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.,Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Bernardo Carpiniello
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.,Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Gavino Faa
- Unit of Anatomic Pathology, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.,Unit of Anatomic Pathology, University Hospital Agency of Cagliari, Cagliari, Italy
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy.,Unit of Clinical Psychiatry, University Hospital Agency of Cagliari, Cagliari, Italy.,Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
17
|
Yin J, Li F, Zhou Y, Mou M, Lu Y, Chen K, Xue J, Luo Y, Fu J, He X, Gao J, Zeng S, Yu L, Zhu F. INTEDE: interactome of drug-metabolizing enzymes. Nucleic Acids Res 2021; 49:D1233-D1243. [PMID: 33045737 PMCID: PMC7779056 DOI: 10.1093/nar/gkaa755] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/19/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022] Open
Abstract
Drug-metabolizing enzymes (DMEs) are critical determinant of drug safety and efficacy, and the interactome of DMEs has attracted extensive attention. There are 3 major interaction types in an interactome: microbiome-DME interaction (MICBIO), xenobiotics-DME interaction (XEOTIC) and host protein-DME interaction (HOSPPI). The interaction data of each type are essential for drug metabolism, and the collective consideration of multiple types has implication for the future practice of precision medicine. However, no database was designed to systematically provide the data of all types of DME interactions. Here, a database of the Interactome of Drug-Metabolizing Enzymes (INTEDE) was therefore constructed to offer these interaction data. First, 1047 unique DMEs (448 host and 599 microbial) were confirmed, for the first time, using their metabolizing drugs. Second, for these newly confirmed DMEs, all types of their interactions (3359 MICBIOs between 225 microbial species and 185 DMEs; 47 778 XEOTICs between 4150 xenobiotics and 501 DMEs; 7849 HOSPPIs between 565 human proteins and 566 DMEs) were comprehensively collected and then provided, which enabled the crosstalk analysis among multiple types. Because of the huge amount of accumulated data, the INTEDE made it possible to generalize key features for revealing disease etiology and optimizing clinical treatment. INTEDE is freely accessible at: https://idrblab.org/intede/.
Collapse
Affiliation(s)
- Jiayi Yin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhou
- The First Affiliated Hospital, Zhejiang University, Hangzhou 310000, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yinjing Lu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Kangli Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jia Xue
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yongchao Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianbo Fu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xu He
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| | - Su Zeng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| | - Lushan Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| |
Collapse
|
18
|
3D-Hepatocyte Culture Applied to Parasitology: Immune Activation of Canine Hepatic Spheroids Exposed to Leishmania infantum. Biomedicines 2020; 8:biomedicines8120628. [PMID: 33352885 PMCID: PMC7766187 DOI: 10.3390/biomedicines8120628] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/01/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
The application of innovative three-dimensional (3D) spheroids cell culture strategy to Parasitology offers the opportunity to closely explore host–parasite interactions. Here we present a first report on the application of 3D hepatic spheroids to unravel the immune response of canine hepatocytes exposed to Leishmania infantum. The liver, usually considered a major metabolic organ, also performs several important immunological functions and constitutes a target organ for L. infantum infection, the etiological agent of canine leishmaniasis (CanL), and a parasitic disease of major veterinary and public health concern. 3D hepatic spheroids were able to sense and immunologically react to L. infantum parasites, generating an innate immune response by increasing nitric oxide (NO) production and enhancing toll-like receptor (TLR) 2 and interleukin-10 gene expression. The immune response orchestrated by canine hepatocytes also lead to the impairment of several cytochrome P450 (CYP450) with possible implications for liver natural xenobiotic metabolization capacity. The application of meglumine antimoniate (MgA) increased the inflammatory response of 3D hepatic spheroids by inducing the expression of Nucleotide oligomerization domain (NOD) -like receptors 1 and NOD2 and TLR2, TLR4, and TLR9 and enhancing gene expression of tumour necrosis factor α. It is therefore suggested that hepatocytes are key effector cells and can activate and orchestrate the immune response to L. infantum parasites.
Collapse
|
19
|
Maillard M, Chevreau C, Le Louedec F, Cassou M, Delmas C, Gourdain L, Blay JY, Cupissol D, Bompas E, Italiano A, Isambert N, Delcambre-Lair C, Penel N, Bertucci F, Guillemet C, Plenecassagnes J, Foulon S, Chatelut É, Le Cesne A, Thomas F. Pharmacogenetic Study of Trabectedin-Induced Severe Hepatotoxicity in Patients with Advanced Soft Tissue Sarcoma. Cancers (Basel) 2020; 12:E3647. [PMID: 33291741 PMCID: PMC7761985 DOI: 10.3390/cancers12123647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/02/2020] [Accepted: 12/02/2020] [Indexed: 01/26/2023] Open
Abstract
Hepatotoxicity is an important concern for nearly 40% of the patients treated with trabectedin for advanced soft tissue sarcoma (ASTS). The mechanisms underlying these liver damages have not yet been elucidated but they have been suggested to be related to the production of reactive metabolites. The aim of this pharmacogenetic study was to identify genetic variants of pharmacokinetic genes such as CYP450 and ABC drug transporters that could impair the trabectedin metabolism in hepatocytes. Sixty-three patients with ASTS from the TSAR clinical trial (NCT02672527) were genotyped by next-generation sequencing for 11 genes, and genotype-toxicity association analyses were performed with R package SNPassoc. Among the results, ABCC2 c.1249A allele (rs2273697) and ABCG2 intron variant c.-15994T (rs7699188) were associated with an increased risk of severe cytolysis, whereas ABCC2 c.3563A allele had a protective effect, as well as ABCB1 variants rs2032582 and rs1128503 (p-value < 0.05). Furthermore, CYP3A5*1 rs776746 (c.6986A > G) increased the risk of severe overall hepatotoxicity (p = 0.012, odds ratio (OR) = 5.75), suggesting the implication of metabolites in the hepatotoxicity. However, these results did not remain significant after multiple analysis correction. These findings need to be validated on larger cohorts of patients, with mechanistic studies potentially being able to validate the functional consequences of these variants.
Collapse
Affiliation(s)
- Maud Maillard
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm UMR1037, 31059 Toulouse, France; (M.M.); (F.L.L.); (C.D.); (L.G.); (É.C.)
- Université Paul Sabatier—Toulouse III, 31400 Toulouse, France
- Institut Claudius Regaud, Institut Universitaire du Cancer (IUCT)—Oncopole, 31059 Toulouse, France; (C.C.); (M.C.); (J.P.)
| | - Christine Chevreau
- Institut Claudius Regaud, Institut Universitaire du Cancer (IUCT)—Oncopole, 31059 Toulouse, France; (C.C.); (M.C.); (J.P.)
| | - Félicien Le Louedec
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm UMR1037, 31059 Toulouse, France; (M.M.); (F.L.L.); (C.D.); (L.G.); (É.C.)
- Université Paul Sabatier—Toulouse III, 31400 Toulouse, France
- Institut Claudius Regaud, Institut Universitaire du Cancer (IUCT)—Oncopole, 31059 Toulouse, France; (C.C.); (M.C.); (J.P.)
| | - Manon Cassou
- Institut Claudius Regaud, Institut Universitaire du Cancer (IUCT)—Oncopole, 31059 Toulouse, France; (C.C.); (M.C.); (J.P.)
| | - Caroline Delmas
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm UMR1037, 31059 Toulouse, France; (M.M.); (F.L.L.); (C.D.); (L.G.); (É.C.)
- Institut Claudius Regaud, Institut Universitaire du Cancer (IUCT)—Oncopole, 31059 Toulouse, France; (C.C.); (M.C.); (J.P.)
| | - Laure Gourdain
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm UMR1037, 31059 Toulouse, France; (M.M.); (F.L.L.); (C.D.); (L.G.); (É.C.)
- Institut Claudius Regaud, Institut Universitaire du Cancer (IUCT)—Oncopole, 31059 Toulouse, France; (C.C.); (M.C.); (J.P.)
| | - Jean-Yves Blay
- Medical Oncology Department, Centre Léon Bérard, 69008 Lyon, France;
| | - Didier Cupissol
- Medical Oncology Department, Institut Régional du Cancer Val d’Aurelle, 34090 Montpellier, France;
| | - Emmanuelle Bompas
- Medical Oncology Department, Institut de Cancérologie de l’Ouest, 44800 Saint-Herblain, France;
| | - Antoine Italiano
- Medical Oncology Department, Institut Bergonié, 33000 Bordeaux, France;
| | - Nicolas Isambert
- Medical Oncology Department, Centre Georges François Leclerc, 21000 Dijon, France;
| | | | - Nicolas Penel
- Medical Oncology Department, Centre Oscar Lambret—Université de Lille, 59000 Lille, France;
| | - François Bertucci
- Medical Oncology Department, Institut Paoli-Calmettes, 13009 Marseille, France;
| | - Cécile Guillemet
- Medical Oncology Department, Centre Henri Becquerel, 76038 Rouen, France;
| | - Julien Plenecassagnes
- Institut Claudius Regaud, Institut Universitaire du Cancer (IUCT)—Oncopole, 31059 Toulouse, France; (C.C.); (M.C.); (J.P.)
| | - Stéphanie Foulon
- Department of Biostatistics and Epidemiology, Gustave Roussy, University Paris-Saclay, 94805 Villejuif, France;
- Oncostat U1018, Inserm, University Paris-Saclay, Labeled Ligue Contre le Cancer, 94805 Villejuif, France
| | - Étienne Chatelut
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm UMR1037, 31059 Toulouse, France; (M.M.); (F.L.L.); (C.D.); (L.G.); (É.C.)
- Université Paul Sabatier—Toulouse III, 31400 Toulouse, France
- Institut Claudius Regaud, Institut Universitaire du Cancer (IUCT)—Oncopole, 31059 Toulouse, France; (C.C.); (M.C.); (J.P.)
| | - Axel Le Cesne
- Medical Oncology Department, Gustave Roussy, 94805 Villejuif, France;
| | - Fabienne Thomas
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm UMR1037, 31059 Toulouse, France; (M.M.); (F.L.L.); (C.D.); (L.G.); (É.C.)
- Université Paul Sabatier—Toulouse III, 31400 Toulouse, France
- Institut Claudius Regaud, Institut Universitaire du Cancer (IUCT)—Oncopole, 31059 Toulouse, France; (C.C.); (M.C.); (J.P.)
| |
Collapse
|
20
|
Campesi I, Montella A, Franconi F. Letter to the Editor in response to the article 'Candidate drugs against SARS-CoV-2 and COVID-19'. Pharmacol Res 2020; 163:105285. [PMID: 33160068 PMCID: PMC7641521 DOI: 10.1016/j.phrs.2020.105285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 11/26/2022]
Affiliation(s)
- Ilaria Campesi
- Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, 07100 Sassari, Italy; Laboratorio Nazionale di Farmacologia e Medicina di Genere, Istituto Nazionale Biostrutture Biosistemi, 07100 Sassari, Italy.
| | - Andrea Montella
- Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, 07100 Sassari, Italy
| | - Flavia Franconi
- Laboratorio Nazionale di Farmacologia e Medicina di Genere, Istituto Nazionale Biostrutture Biosistemi, 07100 Sassari, Italy
| |
Collapse
|
21
|
Jarrar Y, Al-Doaiss A, Alfaifi M, Shati A, Al-Kahtani M, Jarrar B. The influence of five metallic nanoparticles on the expression of major drug-metabolizing enzyme genes with correlation of inflammation in mouse livers. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 80:103449. [PMID: 32593632 DOI: 10.1016/j.etap.2020.103449] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 06/11/2023]
Abstract
Metallic nanoparticles (NPs) are widely used in medical preparations. The present study aims to find out the influence of widely used five metallic NPs on the expression of major hepatic drug-metabolizing enzyme (DME) genes. Six groups of BALB/C mice, 7 mice each, were exposed to: Gold NPs, silver NPs, copper oxide NPs, silicon dioxide NPs and zinc oxide NPs, for 21 days. Liver biopsies from all mice were subjected to mouse cyp3a11, cyp2c29, ugt2b1 and interleukin-6 (il6) gene expression quantification using real-time polymerase chain reaction, in addition to inflammatory cell infiltration examination. All tested NPs caused a sharp and significant (ANOVA, p value <0.05) downregulation in the expression of DME genes, with the highest influence was observed in mice exposed to copper oxide NPs. Additionally, all NPs induced hepatic inflammation and upregulated the expression of il6 gene, which were inversely correlated with the expression of DMEs. It is concluded that all tested NPs downregulated the expression of DME genes, with the highest influence exhibited by copper oxide NPs, in correlation with inflammation and il6 gene induction in the liver. Further studies are needed to find out the effect of anti-inflammatory compounds against the alterations induced by metallic NPs exposure on hepatic DMEs.
Collapse
Affiliation(s)
- Yazun Jarrar
- Department of Pharmaceutical Science, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Jordan
| | - Amin Al-Doaiss
- Department of Biology, College of Science, King Khalid University, Saudi Arabia
| | - Mohammad Alfaifi
- Department of Biology, College of Science, King Khalid University, Saudi Arabia
| | - Ali Shati
- Department of Biology, College of Science, King Khalid University, Saudi Arabia
| | - Mohammed Al-Kahtani
- Department of Biology, College of Science, King Khalid University, Saudi Arabia
| | - Bashir Jarrar
- Nanobiology Unit, Department of Biological Sciences, College of Science, Jerash University, Jordan.
| |
Collapse
|
22
|
Penman SL, Kiy RT, Jensen RL, Beoku‐Betts C, Alfirevic A, Back D, Khoo SH, Owen A, Pirmohamed M, Park BK, Meng X, Goldring CE, Chadwick AE. Safety perspectives on presently considered drugs for the treatment of COVID-19. Br J Pharmacol 2020; 177:4353-4374. [PMID: 32681537 PMCID: PMC7404855 DOI: 10.1111/bph.15204] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
Intense efforts are underway to evaluate potential therapeutic agents for the treatment of COVID-19. In order to respond quickly to the crisis, the repurposing of existing drugs is the primary pharmacological strategy. Despite the urgent clinical need for these therapies, it is imperative to consider potential safety issues. This is important due to the harm-benefit ratios that may be encountered when treating COVID-19, which can depend on the stage of the disease, when therapy is administered and underlying clinical factors in individual patients. Treatments are currently being trialled for a range of scenarios from prophylaxis (where benefit must greatly exceed risk) to severe life-threatening disease (where a degree of potential risk may be tolerated if it is exceeded by the potential benefit). In this perspective, we have reviewed some of the most widely researched repurposed agents in order to identify potential safety considerations using existing information in the context of COVID-19.
Collapse
Affiliation(s)
- Sophie L. Penman
- MRC Centre for Drug Safety Science, Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
| | - Robyn T. Kiy
- MRC Centre for Drug Safety Science, Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
| | - Rebecca L. Jensen
- MRC Centre for Drug Safety Science, Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
| | | | - Ana Alfirevic
- MRC Centre for Drug Safety Science, Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
| | - David Back
- MRC Centre for Drug Safety Science, Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
| | - Saye H. Khoo
- MRC Centre for Drug Safety Science, Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
| | - Andrew Owen
- MRC Centre for Drug Safety Science, Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
| | - Munir Pirmohamed
- MRC Centre for Drug Safety Science, Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
| | - B. Kevin Park
- MRC Centre for Drug Safety Science, Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
| | - Xiaoli Meng
- MRC Centre for Drug Safety Science, Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
| | - Christopher E. Goldring
- MRC Centre for Drug Safety Science, Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
| | - Amy E. Chadwick
- MRC Centre for Drug Safety Science, Department of Pharmacology and TherapeuticsUniversity of LiverpoolLiverpoolUK
| |
Collapse
|
23
|
Wang J, Tripathy N, Chung EJ. Targeting and therapeutic peptide-based strategies for polycystic kidney disease. Adv Drug Deliv Rev 2020; 161-162:176-189. [PMID: 32866560 PMCID: PMC7736157 DOI: 10.1016/j.addr.2020.08.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/15/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022]
Abstract
Polycystic kidney disease (PKD) is characterized by progressive cyst growth and is a leading cause of renal failure worldwide. Currently, there are limited therapeutic options available to PKD patients, and only one drug, tolvaptan, has been FDA-approved to slow cyst progression. Similar to other small molecule drugs, however, tolvaptan is costly, only moderately effective, and causes adverse events leading to high patient dropout rates. Peptides may mitigate many drawbacks of small molecule drugs, as they can be highly tissue-specific, biocompatible, and economically scaled-up. Peptides can function as targeting ligands that direct therapies to diseased renal tissue, or be potent as therapeutic agents themselves. This review discusses various aberrant signaling pathways in PKD and renal receptors that can be potential targets of peptide-mediated strategies. Additionally, peptides utilized in other kidney applications, but may prove useful in the context of PKD, are highlighted. Insights into novel peptide-based solutions that have potential to improve clinical management of PKD are provided.
Collapse
Affiliation(s)
- Jonathan Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Nirmalya Tripathy
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA; Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA; Department of Medicine, Division of Nephrology and Hypertension, University of Southern California, Los Angeles, CA, USA; Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
24
|
Berman G, Croop R, Kudrow D, Halverson P, Lovegren M, Thiry AC, Conway CM, Coric V, Lipton RB. Safety of Rimegepant, an Oral CGRP Receptor Antagonist, Plus CGRP Monoclonal Antibodies for Migraine. Headache 2020; 60:1734-1742. [PMID: 32799325 PMCID: PMC7496574 DOI: 10.1111/head.13930] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 02/07/2023]
Abstract
Objective Evaluate the safety and tolerability of oral rimegepant when used for acute treatment concomitantly with a monoclonal antibody (mAb) targeting the calcitonin gene‐related peptide (CGRP) ligand or receptor (CGRP mAb) for the preventive treatment of migraine. Background The efficacy of CGRP mAbs for the preventive treatment of migraine and the small molecule CGRP receptor antagonist rimegepant for acute treatment has been demonstrated in randomized controlled clinical trials. Over the past few years, the US Food and Drug Administration has approved 4 CGRP mAbs for the preventive treatment of migraine and 2 small molecule CGRP receptor antagonists for the acute treatment of migraine. A previous case report of 2 patients receiving concomitant treatment with rimegepant and erenumab suggested that rimegepant may be safely used as acute treatment in patients who are also receiving a preventive regimen involving CGRP mAbs. We report here 13 additional patients with migraine who simultaneously used rimegepant and either erenumab, fremanezumab, or galcanezumab and assess the rate of on‐treatment adverse events (AEs). Methods This was a substudy nested within a multicenter, open‐label, long‐term safety study in adults with 2‐14 monthly migraine attacks of moderate to severe pain intensity. A subgroup experiencing 2‐8 monthly attacks and taking a stable dose of a CGRP mAb also took rimegepant 75 mg as needed up to once daily for acute treatment for 12 weeks. Results The 13 patients (11 women [85%]; mean age 49.9 years) enrolled in the substudy were being treated with CGRP mAbs (erenumab [n = 7], fremanezumab [n = 4], or galcanezumab [n = 2]). Mean (SD) time in the rimegepant treatment period was 9.6 (4.6) weeks. Mean (SD) 4‐week rimegepant exposure was 7.8 (5.5) doses; a total of 224 doses were taken. Five (38%) patients reported ≥1 on‐treatment AE. Of these, 2 (15%) patients had mild or moderate nasopharyngitis; no other AEs occurred in ≥2 patients. Three patients had AEs of mild or moderate severity that were considered potentially treatment‐related. No patients had serious AEs, AEs leading to discontinuation, or aminotransferase levels >3× the upper limit of normal. Conclusion Rimegepant, when used as an oral acute treatment in patients receiving CGRP mAbs as preventive treatment, was well tolerated; no safety issues were identified. Studies involving larger patient populations are needed to confirm these findings.
Collapse
Affiliation(s)
- Gary Berman
- Clinical Research Institute, Minneapolis, MN, USA
| | | | - David Kudrow
- California Medical Clinic for Headache, Santa Monica, CA, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Hepatocytic transcriptional signatures predict comparative drug interaction potential of rifamycin antibiotics. Sci Rep 2020; 10:12565. [PMID: 32724080 PMCID: PMC7387492 DOI: 10.1038/s41598-020-69228-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/08/2020] [Indexed: 12/16/2022] Open
Abstract
Current strategies to treat tuberculosis (TB) and co-morbidities involve multidrug combination therapies. Rifamycin antibiotics are a key component of TB therapy and a common source of drug–drug interactions (DDIs) due to induction of drug metabolizing enzymes (DMEs). Management of rifamycin DDIs are complex, particularly in patients with co-morbidities, and differences in DDI potential between rifamycin antibiotics are not well established. DME profiles induced in response to tuberculosis antibiotics (rifampin, rifabutin and rifapentine) were compared in primary human hepatocytes. We identified rifamycin induced DMEs, cytochrome P450 (CYP) 2C8/3A4/3A5, SULT2A, and UGT1A4/1A5 and predicted lower DDIs of rifapentine with 58 clinical drugs used to treat co-morbidities in TB patients. Transcriptional networks and upstream regulator analyses showed FOXA3, HNF4α, NR1I2, NR1I3, NR3C1 and RXRα as key transcriptional regulators of rifamycin induced DMEs. Our study findings are an important resource to design effective medication regimens to treat common co-conditions in TB patients.
Collapse
|
26
|
Liu F, Zhang Q, Lin C, Yao Y, Wang M, Liu C, Zhu C. A comparative study on pharmacokinetics and tissue distribution of 5-hydroxy-4-methoxycanthin-6-one and its metabolite in normal and dextran sodium sulfate-induced colitis rats by HPLC-MS/MS. J Pharm Pharmacol 2020; 72:1761-1770. [PMID: 32363585 DOI: 10.1111/jphp.13285] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 04/04/2020] [Indexed: 01/14/2023]
Abstract
OBJECTIVES This study aimed to investigate the existing form of 5-hydroxy-4-methoxycanthin-6-one (PQ-A) in vivo after oral administration and the effects on its pharmacokinetics and tissue distribution by colitis. METHODS A rapid HPLC-MS/MS method was established to simultaneously determine PQ-A and its main metabolite, 1-methoxicabony-β-carboline (PQ-B), in biological samples acquired from normal and dextran sodium sulfate (DSS)-induced colitic rats administered orally with PQ-A. Then, the pharmacokinetics of both PQ-A and PQ-B, and tissue distribution of PQ-A in the above two states were analysed. KEY FINDINGS The pharmacokinetic results showed that the prototype of PQ-A was the main existing form in both physiological and pathological conditions. And significant difference between the above two status in pharmacokinetics of PQ-A was observed, such as higher exposure and longer elimination in colitis than that in normal rats. It suggested that the pharmacokinetics of medications for colitis was affected by enteritis. The tissue distribution studies displayed that PQ-A mainly accumulated in intestinal tract. Especially, the distribution of PQ-A in intestinal tract was increased obviously in colitic rats. CONCLUSIONS These results contributed to further illuminate the ADME process of PQ-A in different status and were prospected to be the reference to the clinical application of similar medicines in pathological states.
Collapse
Affiliation(s)
- Fangle Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qiuyu Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chaozhan Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yufeng Yao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meiqi Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Changhui Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chenchen Zhu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
27
|
Neyshaburinezhad N, Rouini M, Shirzad N, Esteghamati A, Nakhjavani M, Namazi S, Ardakani YH. Evaluating the effect of type 2 diabetes mellitus on CYP450 enzymes and P-gp activities, before and after glycemic control: A protocol for a case-control pharmacokinetic study. MethodsX 2020; 7:100853. [PMID: 32337164 PMCID: PMC7176986 DOI: 10.1016/j.mex.2020.100853] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 02/29/2020] [Indexed: 12/18/2022] Open
Abstract
Cytochrome P450s (CYP450) family is one of the most critical factors in the metabolism process. Hence, the present study aims to characterize the activity of CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP3A4/5, and P-glycoprotein (P-gp) pump in patients with type 2 diabetes (T2DM). This characterization was performed before and after good glycemic control versus non-diabetic subjects following the administration of a substrate probe drug cocktail. This single-center clinical study proposes the characterization of T2DM impacts on major CYP450 drug-metabolizing enzyme and P-glycoprotein (P-gp) activities. The main propose of the present study is evaluating any alternation in major CYP450 enzymes and P-gp activities in patients with T2DM, before (A1C>7%) and after (A1C≤7%) good glycemic control along with comparing the activities versus non-diabetic subjects. The phenotypes will be assessed following the oral administration of a drug cocktail containing caffeine (CYP1A2), bupropion (CYP2B6), flurbiprofen (CYP2C9), omeprazole (CYP2C19), dextromethorphan (CYP2D6), midazolam (CYP3A4/5), and fexofenadine (P-gp) as probe substrates. Furthermore, the influence of variables such as glycemia, genetic polymorphisms, and inflammation on the metabolism process will be evaluated. The first patient has entered the study in Dec 2018.
Collapse
Affiliation(s)
- Navid Neyshaburinezhad
- Biopharmaceutics and Pharmacokinetic Division, Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Rouini
- Biopharmaceutics and Pharmacokinetic Division, Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nooshin Shirzad
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Esteghamati
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Manouchehr Nakhjavani
- Endocrinology and Metabolism Research Center (EMRC), Vali-Asr Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Soha Namazi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Yalda H. Ardakani
- Biopharmaceutics and Pharmacokinetic Division, Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Mimche SM, Lee CM, Liu KH, Mimche PN, Harvey RD, Murphy TJ, Nyagode BA, Jones DP, Lamb TJ, Morgan ET. A non-lethal malarial infection results in reduced drug metabolizing enzyme expression and drug clearance in mice. Malar J 2019; 18:234. [PMID: 31299982 PMCID: PMC6624958 DOI: 10.1186/s12936-019-2860-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/01/2019] [Indexed: 11/10/2022] Open
Abstract
Background Given the central importance of anti-malarial drugs in the treatment of malaria, there is a need to understand the effect of Plasmodium infection on the broad spectrum of drug metabolizing enzymes. Previous studies have shown reduced clearance of quinine, a treatment for Plasmodium infection, in individuals with malaria. Methods The hepatic expression of a large panel of drug metabolizing enzymes was studied in the livers of mice infected with the AS strain of Plasmodium chabaudi chabaudi, a nonlethal parasite in most strains of mice with several features that model human Plasmodium infections. C57BL/6J mice were infected with P. chabaudi by intraperitoneal injection of infected erythrocytes and sacrificed at different times after infection. Relative hepatic mRNA levels of various drug metabolizing enzymes, cytokines and acute phase proteins were measured by reverse transcriptase-real time PCR. Relative levels of cytochrome P450 proteins were measured by Western blotting with IR-dye labelled antibodies. Pharmacokinetics of 5 prototypic cytochrome P450 substrate drugs were measured by cassette dosing and high-resolution liquid chromatography-mass spectrometry. The results were analysed by MANOVA and post hoc univariate analysis of variance. Results The great majority of enzyme mRNAs were down-regulated, with the greatest effects occurring at the peak of parasitaemia 8 days post infection. Protein levels of cytochrome P450 enzymes in the Cyp 2b, 2c, 2d, 2e, 3a and 4a subfamilies were also down-regulated. Several distinct groups differing in their temporal patterns of regulation were identified. The cassette dosing study revealed that at the peak of parasitaemia, the clearances of caffeine, bupropion, tolbutamide and midazolam were markedly reduced by 60–70%. Conclusions These findings in a model of uncomplicated human malaria suggest that changes in drug clearance in this condition may be of sufficient magnitude to cause significant alterations in exposure and response of anti-malarial drugs and co-medications. Electronic supplementary material The online version of this article (10.1186/s12936-019-2860-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sylvie M Mimche
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Choon-Myung Lee
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ken H Liu
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Patrice N Mimche
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - R Donald Harvey
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Thomas J Murphy
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Beatrice A Nyagode
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Tracey J Lamb
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Edward T Morgan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
29
|
Taneja G, Maity S, Jiang W, Moorthy B, Coarfa C, Ghose R. Transcriptomic profiling identifies novel mechanisms of transcriptional regulation of the cytochrome P450 (Cyp)3a11 gene. Sci Rep 2019; 9:6663. [PMID: 31040347 PMCID: PMC6491424 DOI: 10.1038/s41598-019-43248-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 04/04/2019] [Indexed: 02/06/2023] Open
Abstract
Cytochrome P450 (CYP)3A is the most abundant CYP enzyme in the human liver, and a functional impairment of this enzyme leads to unanticipated adverse reactions and therapeutic failures; these reactions result in the early termination of drug development or the withdrawal of drugs from the market. The transcriptional regulation mechanism of the Cyp3a gene is not fully understood and requires a thorough investigation. We mapped the transcriptome of the Cyp3a gene in a mouse model. The Cyp3a gene was induced using the mPXR activator pregnenolone-16alpha-carbonitrile (PCN) and was subsequently downregulated using lipopolysaccharide (LPS). Our objective was to identify the transcription factors (TFs), epigenetic modulators and molecular pathways that are enriched or repressed by PCN and LPS based on a gene set enrichment analysis. Our analysis shows that 113 genes were significantly upregulated (by at least 1.5-fold) with PCN treatment, and that 834 genes were significantly downregulated (by at least 1.5-fold) with LPS treatment. Additionally, the targets of the 536 transcription factors were enriched by a combined treatment of PCN and LPS, and among these, 285 were found to have binding sites on Cyp3a11. Moreover, the repressed targets of the epigenetic markers HDAC1, HDAC3 and EZH2 were further suppressed by LPS treatment and were enhanced by PCN treatment. By identifying and contrasting the transcriptional regulators that are altered by PCN and LPS, our study provides novel insights into the transcriptional regulation of CYP3A in the liver.
Collapse
Affiliation(s)
- Guncha Taneja
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, 4849 Calhoun Rd., Houston, TX, 77204, USA
- DILIsym Services, A Simulations Plus Company, Research Triangle Park, North Carolina, 27709, USA
| | - Suman Maity
- Advanced Technology Cores, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Weiwu Jiang
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, 1102 Bates Avenue, Suite 530, Houston, TX, 77030, USA
| | - Bhagavatula Moorthy
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, 1102 Bates Avenue, Suite 530, Houston, TX, 77030, USA.
| | - Cristian Coarfa
- Dan L Duncan Comprehensive Cancer Center, Center for Precision Environmental Health, Molecular and Cellular Biology Department, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| | - Romi Ghose
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, 4849 Calhoun Rd., Houston, TX, 77204, USA.
| |
Collapse
|
30
|
Xu M, Tang H, Zhou X, Chen H, Dong Q, Zhang Y, Ye G, Shi F, Lv C, Jing B, He C, Zhao L, Li Y. Effects and mechanisms of sub-chronic exposure to copper nanoparticles on renal cytochrome P450 enzymes in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 63:135-146. [PMID: 30100346 DOI: 10.1016/j.etap.2018.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/03/2018] [Accepted: 08/04/2018] [Indexed: 06/08/2023]
Abstract
Copper nanoparticles (Cu NPs) have widespread application because of their special physicochemical characteristics, however we need to more clearly study the toxicity mechanism of Cu NPs to ensure its safe use in pharmaceutical and animal feed. Thus, the aim of this study was to evaluate the effects and mechanisms of sub-chronic exposure to Cu NPs on renal CYP450 s of rats. In this study, we investigated the effects of Cu NPs on renal oxidative stress, cytokines and histopathology of rats. We found that Cu NPs (200 mg/kg) significantly disordered the function and structure of the kidney and caused a dose-dependent increase in oxidative stress and cytokines, which significantly decreased the levels of mRNA, protein, and activity of CYP450 s. Micro-coppers (Cu MPs) and Cu ions have similar effects, but their effects on CYP450 s were weaker than Cu NPs. The expression of nuclear receptors were inhibited and the expression of Akt, STAT3/5, CREB, p70S6K, NF-κB, P38 and ERK1/2 were activated when the inhibition effects of CYP450 s activity were observed in renal of rats. Therefore, we believe that Cu NPs can activate the STAT, NF-κB and MAPK signaling pathways to down-regulate the expression and activity of CYP450 s by inducing oxidative stress and inflammatory response in rat kidney.
Collapse
Affiliation(s)
- Min Xu
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Huaqiao Tang
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - XueRong Zhou
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Helin Chen
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Qi Dong
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Yuanli Zhang
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Gang Ye
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Fei Shi
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Cheng Lv
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Bo Jing
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Changliang He
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Ling Zhao
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China
| | - Yinglun Li
- Sichuan Agricultural University, Department of Pharmacy, Sichuan, Chengdu, 611130, China.
| |
Collapse
|
31
|
Tang H, Xu M, Shi F, Ye G, Lv C, Luo J, Zhao L, Li Y. Effects and Mechanism of Nano-Copper Exposure on Hepatic Cytochrome P450 Enzymes in Rats. Int J Mol Sci 2018; 19:ijms19072140. [PMID: 30041454 PMCID: PMC6073330 DOI: 10.3390/ijms19072140] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/04/2018] [Accepted: 07/15/2018] [Indexed: 12/14/2022] Open
Abstract
Although nano-copper is currently used extensively, the adverse effects on liver cytochrome P450 (CYP450) enzymes after oral exposure are not clear. In this study, we determined the effects and mechanisms of action of nano- and micro-copper on the expression and activity of CYP450 enzymes in rat liver. Rats were orally exposed to micro-copper (400 mg/kg), Cu ion (100 mg/kg), or nano-copper (100, 200 and 400 mg/kg) daily for seven consecutive days. Histopathological, inflammatory and oxidative stress were measured in the livers of all rats. The mRNA levels and activity of CYP450 enzymes, as well as the mRNA levels of select nuclear receptors, were determined. Exposure to nano-copper (400 mg/kg) induced significant oxidative stress and inflammation relative to the controls, indicated by increased levels of interleukin (IL)-2, IL-6, interferon (IFN)-γ, macrophage inflammatory protein (MIP-1), total antioxidant capacity (T-AOC), malondialdehyde (MDA), inducible nitric oxide synthase (iNOS) and nitric oxide (NO) after exposure. The levels of mRNA expression of pregnane X receptor (PXR), constitutive androstane receptor (CAR) and aryl hydrocarbon receptor (AHR) were significantly decreased in 400 mg/kg nano-copper treated rats. Nano-copper activated the expression of the NF-kappa B (NF-κB), mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription (STAT)3 signaling pathways. Nano-copper decreased the mRNA expression and activity of CYP 1A2, 2C11, 2D6, 2E1 and 3A4 in a dose-dependent manner. The adverse effects of micro-copper are less severe than those of nano-copper on the CYP450 enzymes of rats after oral exposure. Ingestion of large amounts of nano-copper in animals severely affects the drug metabolism of the liver by inhibiting the expression of various CYP450 enzymes, which increases the risk of drug-drug interactions in animals.
Collapse
Affiliation(s)
- Huaqiao Tang
- Department of Pharmacy, School of Animal Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Min Xu
- Department of Pharmacy, School of Animal Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Fei Shi
- Department of Pharmacy, School of Animal Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Gang Ye
- Department of Pharmacy, School of Animal Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Cheng Lv
- Department of Pharmacy, School of Animal Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Jie Luo
- Department of Pharmacy, School of Animal Medicine, Sichuan Agricultural University, Chengdu 611130, China.
- School of Medicine, Tongren Polytechnic College, Guizhou 554300, China.
| | - Ling Zhao
- Department of Pharmacy, School of Animal Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Yinglun Li
- Department of Pharmacy, School of Animal Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
32
|
Clinical Efficacy of Trimetazidine and Holistic Management in the Treatment of Coronary Heart Disease. IRANIAN JOURNAL OF PUBLIC HEALTH 2018; 47:824-829. [PMID: 30087867 PMCID: PMC6077637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
BACKGROUND To investigate the clinical efficacy of trimetazidine and holistic management in the treatment of coronary heart disease. METHODS A total of 128 cases of patients with coronary heart disease were admitted in The Second Affiliated Hospital of Dalian Medical University from October 2014 to June 2017. These patients were divided into control group and experimental group, with 64 patients each. The patients in the control group underwent conventional treatment. On this basis, the patients in the experimental group were treated with trimetazidine. Both groups underwent holistic management. The clinical conditions, echocardiography indexes, life quality, and mental states of patients were compared between the two groups. RESULTS The total efficient rate of patients in the control group was significantly lower than that of the experimental group, and the difference was statistically significant (P<0.05). After treatment, the left ventricular ejection fraction of patients in the experimental group was higher than that of the control group; the left ventricular end diastolic diameter and left ventricular posterior wall thickness of patients in the experimental group were lower than those of the control group, and the differences were statistically significant (P<0.05). The physiological health score, mental health score, Hamilton Depression Scale and Hamilton Anxiety Rating Scale of patients were significantly decreased compared with the control group, and the differences were statistically significant (P<0.05). CONCLUSION The efficacy of trimetazidine in the treatment of coronary heart disease is definite. The assisting holistic management can significantly improve the mental status and life quality of patients by enhancing the cardiac function, which has clinical reference and promotion values.
Collapse
|