1
|
Cattaneo D, Ridolfo AL, Giacomelli A, Castagna A, Dolci A, Antinori S, Gervasoni C. The Case of Dolutegravir Plus Darunavir Antiretroviral Regimens: Is It Always Useful to Double the Drug Doses? Ther Drug Monit 2025; 47:309-312. [PMID: 39235337 DOI: 10.1097/ftd.0000000000001259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/26/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Antiretroviral drug combinations affect dolutegravir trough concentrations. Here, the authors focused on dolutegravir plus booster darunavir antiretroviral regimens to investigate the effect of the booster and/or timing of drug administration on dolutegravir and darunavir plasma trough concentrations. METHODS This retrospective observational study included consecutive people with HIV (PWH) receiving dolutegravir plus booster darunavir antiretroviral regimens for at least 3 months, with at least one assessment of dolutegravir and darunavir plasma trough concentrations. RESULTS A total of 200 drug therapeutic drug monitoring results from 116 PWH were included. Dolutegravir and darunavir trough concentrations ranged, respectively, from 70 to 3648 mcg/L and from 102 to 11,876 mcg/L. The antiretroviral drug combination associated with the highest dolutegravir trough concentration was dolutegravir plus darunavir/cobicistat, both once daily (1410 ± 788 mcg/L), whereas dolutegravir once daily plus darunavir/ritonavir twice daily had the lowest trough concentrations (686 ± 481 mcg/L). Doubling the dose of dolutegravir did not significantly increase drug trough concentrations compared with that of once-daily regimens. Instead, the highest darunavir trough concentrations were with ritonavir (2850 ± 1456 mcg/L, P < 0.05 versus cobicistat-based regimens). Doubling the drug dose resulted in a significant increase in the darunavir trough concentration (4445 ± 2926 mcg/L, P < 0.05). CONCLUSIONS Dolutegravir trough concentrations were significantly reduced in PWH receiving darunavir/ritonavir twice daily. This evidence should be carefully considered in clinical conditions requiring higher dolutegravir exposure, such as in the presence of drug-drug interactions with drugs known to reduce dolutegravir bioavailability or in highly experienced PWH.
Collapse
Affiliation(s)
- Dario Cattaneo
- Clinical Pathology Unit, ASST Fatebenefratelli Sacco
- Gestione Ambulatoriale Politerapie (GAP) Outpatient Clinic, ASST Fatebenefratelli Sacco
| | | | - Andrea Giacomelli
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco
- Department of Biomedical and Clinical Sciences, University of Milan; and
| | - Antonella Castagna
- Department of Infectious Diseases, IRCSS San Raffaele Scientific Institute, Milan, Italy
| | - Alberto Dolci
- Clinical Pathology Unit, ASST Fatebenefratelli Sacco
- Department of Biomedical and Clinical Sciences, University of Milan; and
| | - Spinello Antinori
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco
- Department of Biomedical and Clinical Sciences, University of Milan; and
| | - Cristina Gervasoni
- Gestione Ambulatoriale Politerapie (GAP) Outpatient Clinic, ASST Fatebenefratelli Sacco
- Department of Infectious Diseases, ASST Fatebenefratelli Sacco
| |
Collapse
|
2
|
Avedissian SN, Mu Y, McCarthy C, Bosch RJ, Spudich S, Gandhi RT, McMahon DK, Eron JJ, Mellors JW, Liu J, Podany AT, Fletcher CV. Pharmacokinetic approaches to standardize antiviral exposure in cerebrospinal fluid. Pharmacotherapy 2025. [PMID: 40152440 DOI: 10.1002/phar.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVES HIV has been shown to persist in the central nervous system (CNS) in persons on antiretroviral therapy (ART). Our objective was to use pharmacokinetic (PK) modeling to estimate cerebrospinal fluid (CSF) exposure from time-variant concentrations of various antiretrovirals of ART regimens and to standardize CSF metrics, including maximum concentration [CMAX], area under the curve [AUC], and trough [CTrough]. METHODS Advancing Clinical Therapeutics Globally (ACTG) A5321 is a prospective cohort study of HIV-1 reservoirs in persons with HIV. Plasma and CSF antiretroviral (ARV) concentrations were measured in 74 participants who were receiving ART. PK modeling (Pmetrics) was performed for nine ARVs. Relative CSF penetration for each ARV was estimated by comparing CSF CMAX and AUC to plasma CMAX and AUC (i.e., CMAXmethod and AUCmethod). The CSF CTrough for each ARV was compared with in vitro literature values of HIV inhibitory concentration values (IC50, 90, or 95). RESULTS Emtricitabine exhibited the highest median relative CSF penetration (CMAXmethod, 46.3%; AUCmethod, 72%) and dolutegravir had the lowest CSF penetration (CMAXmethod, 0.57%; AUCmethod, 0.57%). Tenofovir, lamivudine, atazanavir, and raltegravir had median estimated CSF CTrough concentrations less than IC50, 90, or 95. Interparticipant variability of relative CSF penetration based on exposures ranged from 160% for lamivudine to approximately 9% for dolutegravir. CONCLUSIONS PK modeling successfully standardized ARV CSF concentrations to a given time point (i.e., CMAX or CTrough) to allow estimation of CSF penetration. This approach provides uniformity for the assessment of exposure, for the estimation of whether desired therapeutic drug goals are obtained in the CSF, and for further studies to investigate whether CSF exposure metrics calculated using this method are associated with measures of HIV persistence.
Collapse
Affiliation(s)
- Sean N Avedissian
- Antiviral Pharmacology Laboratory, UNMC Center for Drug Discovery, Omaha, Nebraska, USA
| | - Ying Mu
- Antiviral Pharmacology Laboratory, UNMC Center for Drug Discovery, Omaha, Nebraska, USA
| | - Caitlyn McCarthy
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Ronald J Bosch
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | | | | | | | - Joseph J Eron
- University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Jiajun Liu
- Division of Pharmacometrics, Office of Clinical Pharmacology, Office of Translational Sciences, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Anthony T Podany
- Antiviral Pharmacology Laboratory, UNMC Center for Drug Discovery, Omaha, Nebraska, USA
| | - Courtney V Fletcher
- Antiviral Pharmacology Laboratory, UNMC Center for Drug Discovery, Omaha, Nebraska, USA
| |
Collapse
|
3
|
Gatti M, Campoli C, Muratore E, Belotti T, Masetti R, Lanari M, Viale P, Pea F. Impact of Inflammatory Burden on Voriconazole Exposure in Oncohematological Pediatric Patients Receiving Antifungal Prophylaxis after Allogeneic HCT. Microorganisms 2024; 12:1388. [PMID: 39065156 PMCID: PMC11278995 DOI: 10.3390/microorganisms12071388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/04/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
(1) Background: The impact of inflammation on voriconazole exposure in oncohematological pediatric patients represents a debated issue. We aimed to investigate the impact of serum C-reactive protein (CRP), procalcitonin (PCT), and interleukin-6 (IL-6) levels on voriconazole exposure in oncohematological pediatric patients requiring allogeneic hematopoietic stem cell transplantation (HCT). (2) Methods: Pediatric patients undergoing allogeneic HCT and receiving therapeutic drug monitoring (TDM)-guided voriconazole as primary antifungal prophylaxis between January 2021 and December 2023 were included. The ratio between concentration and dose (C/D) of voriconazole was used as a surrogate marker of total clearance. A receiving operating characteristic curve analysis was performed by using CRP, PCT, or IL-6 values as the test variable and voriconazole C/D ratio > 0.188 or >0.375 (corresponding to a trough concentration value [Cmin] of 3 mg/L normalized to the maintenance dose of 16 mg/kg/day in patients of age < 12 years and of 8 mg/kg/day in those ≥12 years, respectively) as the state variable. Area under the curve (AUC) and 95% confidence interval (CI) were calculated. (3) Results: Overall, 39 patients were included. The median (IQR) voriconazole Cmin was 1.7 (0.7-3.0) mg/L. A CRP value > 8.49 mg/dL (AUC = 0.72; 95%CI 0.68-0.76; p < 0.0001), a PCT value > 2.6 ng/mL (AUC = 0.71; 95%CI 0.63-0.77; p < 0.0001), and an IL-6 value > 27.9 pg/mL (AUC = 0.80; 95%CI 0.71-0.88; p < 0.0001) were significantly associated with voriconazole overexposure. Consistent results were found in patients aged <12 and ≥12 years. (4) Conclusions: A single specific threshold of inflammatory biomarkers may be linked to a significantly higher risk of voriconazole exposure in oncohematological pediatric patients after HCT, irrespective of age. Adopting a TDM-guided strategy could be useful for minimizing the risk of voriconazole overexposure.
Collapse
Affiliation(s)
- Milo Gatti
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (R.M.); (M.L.); (P.V.); (F.P.)
- Clinical Pharmacology Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Caterina Campoli
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Edoardo Muratore
- Pediatric Hematology and Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (E.M.); (T.B.)
| | - Tamara Belotti
- Pediatric Hematology and Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (E.M.); (T.B.)
| | - Riccardo Masetti
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (R.M.); (M.L.); (P.V.); (F.P.)
- Pediatric Hematology and Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (E.M.); (T.B.)
| | - Marcello Lanari
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (R.M.); (M.L.); (P.V.); (F.P.)
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Pierluigi Viale
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (R.M.); (M.L.); (P.V.); (F.P.)
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Federico Pea
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy; (R.M.); (M.L.); (P.V.); (F.P.)
- Clinical Pharmacology Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
4
|
Giamarellos-Bourboulis EJ. Adjunctive treatment in COVID-19 and sepsis-What did we learn? Med Klin Intensivmed Notfmed 2023; 118:80-85. [PMID: 37966529 PMCID: PMC10733190 DOI: 10.1007/s00063-023-01089-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/24/2023] [Indexed: 11/16/2023]
Abstract
The introduction of anakinra, baricitinib and tocilizumab into the treatment armamentarium of severe coronavirus disease 2019 (COVID-19) reinforced the concept of immunotherapy for bacterial sepsis. The current review investigates how the example of COVID-19 may be extrapolated to sepsis using a three-step approach. In the first step, the clinical evidence on how the immunotherapy of COVID-19 assisted viral clearance is presented. In a second step, the indications acquired from human and animal studies on the need to employ strategies with primary effective phagocytosis in sepsis are presented. In a final step, lessons learnt from COVID-19 immunotherapy are applied for sepsis. The end result is that sepsis immunotherapy should rely on the use of biomarkers which provide information on the activation of a specific prevailing mechanism in order to enable the selection of the appropriate drug.
Collapse
Affiliation(s)
- Evangelos J Giamarellos-Bourboulis
- Fourth Department of Internal Medicine, ATTIKON University Hospital, National and Kapodistrian University of Athens, Medical School, 1 Rimini Street, 124 62 Athens, Greece.
| |
Collapse
|
5
|
Toledo T, Castro T, Oliveira VG, Veloso VG, Grinsztejn B, Cardoso SW, Torres TS, Estrela R. Pharmacokinetics of Antiretroviral Drugs in Older People Living with HIV: A Systematic Review. Clin Pharmacokinet 2023; 62:1219-1230. [PMID: 37561283 DOI: 10.1007/s40262-023-01291-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND AND OBJECTIVE The life expectancy of people living with HIV (PLWHIV) has significantly improved in recent decades, mostly due to antiretroviral (ARV) therapy. Aging can affect the pharmacokinetics of drugs and, as a consequence, increase the risk of drug interactions and toxicity that may impact treatment. The aim of this study was to carry out a systematic review of the literature on the effect of aging on ARV pharmacokinetics. METHODS Searches were performed in the BVS, EMBASE and PUBMED databases until November 2022. All studies available in English, Spanish and Portuguese investigating the pharmacokinetics of ARV approved by the US Food and Drug Administration (FDA) from 2005 to 2020 were selected. Peer-reviewed publications were included if they met all criteria: adults (≥ 18 years of age) living with or without HIV; report any pharmacokinetic parameter or plasma concentration of at least one of the following ARVs: tenofovir alafenamide fumarate (TAF); doravirine (DOR), rilpivirine (RIL) and etravirine (ETR); darunavir (DRV), tipranavir (TPV) and fostemsavir (FTR); dolutegravir (DTG), raltegravir (RAL), bictegravir (BIC) and elvitegravir (EVG); maraviroc (MVC); ibalizumab (IBA); cobicistat (COBI). Pharmacokinetic parameters were reported stratified per age group: young adults (aged 18-49 years) or older (age ≥ 50 years) and all studies were evaluated for quality. The review protocol was registered in the PROSPERO database (registration number CRD42021236432). RESULTS Among 97 studies included, 20 reported pharmacokinetic evaluation in older individuals (age ≥ 50 years). Twenty five percent of the articles were phase I randomized clinical trials with HIV-negative participants and non-compartmental pharmacokinetic analysis presenting the parameters area under the curve (AUC) and peak drug concentration (Cmax). Seven age-stratified studies evaluated BIC, ETR, DRV, DTG, DOR and RAL. We found publications with discordant results for ETR and DTG pharmacokinetics in different age groups. DRV exposure was highly variable but modestly increased in aging PLWHIV. In contrast, no influence of age on BIC, DOR and RAL exposure was observed. A variability in pharmacokinetic parameters could be observed for the other ARVs (TAF and MVC) in different age groups. CONCLUSION Exposure to DRV increases modestly with age, while exposure to BIC, DOR and RAL appears to be unaffected by age. As the available evidence to confirm a potential effect of aging on ARV pharmacokinetics is limited, further studies are necessary.
Collapse
Affiliation(s)
- Thainá Toledo
- Sérgio Arouca National School of Public Health, ENSP Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Thales Castro
- Evandro Chagas National Institute of Infectious Diseases, INI Fiocruz, Rio de Janeiro, Brazil
| | - Vanessa G Oliveira
- Evandro Chagas National Institute of Infectious Diseases, INI Fiocruz, Rio de Janeiro, Brazil
| | | | - Beatriz Grinsztejn
- Evandro Chagas National Institute of Infectious Diseases, INI Fiocruz, Rio de Janeiro, Brazil
| | - Sandra Wagner Cardoso
- Evandro Chagas National Institute of Infectious Diseases, INI Fiocruz, Rio de Janeiro, Brazil
| | - Thiago S Torres
- Evandro Chagas National Institute of Infectious Diseases, INI Fiocruz, Rio de Janeiro, Brazil
| | - Rita Estrela
- Sérgio Arouca National School of Public Health, ENSP Fiocruz, Rio de Janeiro, RJ, Brazil.
- Evandro Chagas National Institute of Infectious Diseases, INI Fiocruz, Rio de Janeiro, Brazil.
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
6
|
Liu C, Cojutti PG, Giannella M, Roberto M, Casadei B, Cristiano G, Papayannidis C, Vianelli N, Zinzani PL, Viale P, Bonifazi F, Pea F. Does Cytokine-Release Syndrome Induced by CAR T-Cell Treatment Have an Impact on the Pharmacokinetics of Meropenem and Piperacillin/Tazobactam in Patients with Hematological Malignancies? Findings from an Observational Case-Control Study. Pharmaceutics 2023; 15:pharmaceutics15031022. [PMID: 36986882 PMCID: PMC10059857 DOI: 10.3390/pharmaceutics15031022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a promising approach for some relapse/refractory hematological B-cell malignancies; however, in most patients, cytokine release syndrome (CRS) may occur. CRS is associated with acute kidney injury (AKI) that may affect the pharmacokinetics of some beta-lactams. The aim of this study was to assess whether the pharmacokinetics of meropenem and piperacillin may be affected by CAR T-cell treatment. The study included CAR T-cell treated patients (cases) and oncohematological patients (controls), who were administered 24-h continuous infusion (CI) meropenem or piperacillin/tazobactam, optimized by therapeutic drug monitoring, over a 2-year period. Patient data were retrospectively retrieved and matched on a 1:2 ratio. Beta-lactam clearance (CL) was calculated as CL = daily dose/infusion rate. A total of 38 cases (of whom 14 and 24 were treated with meropenem and piperacillin/tazobactam, respectively) was matched with 76 controls. CRS occurred in 85.7% (12/14) and 95.8% (23/24) of patients treated with meropenem and piperacillin/tazobactam, respectively. CRS-induced AKI was observed in only 1 patient. CL did not differ between cases and controls for both meropenem (11.1 vs. 11.7 L/h, p = 0.835) and piperacillin (14.0 vs. 10.4 L/h, p = 0.074). Our findings suggest that 24-h CI meropenem and piperacillin dosages should not be reduced a priori in CAR T-cell patients experiencing CRS.
Collapse
Affiliation(s)
- Chun Liu
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, 40138 Bologna, Italy
| | - Pier Giorgio Cojutti
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, 40138 Bologna, Italy
- Clinical Pharmacology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Maddalena Giannella
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, 40138 Bologna, Italy
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Marcello Roberto
- Istituto di Ematologia "Seràgnoli", IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Beatrice Casadei
- Istituto di Ematologia "Seràgnoli", IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Gianluca Cristiano
- Istituto di Ematologia "Seràgnoli", IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Cristina Papayannidis
- Istituto di Ematologia "Seràgnoli", IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Nicola Vianelli
- Istituto di Ematologia "Seràgnoli", IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Pier Luigi Zinzani
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, 40138 Bologna, Italy
- Istituto di Ematologia "Seràgnoli", IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Pierluigi Viale
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, 40138 Bologna, Italy
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesca Bonifazi
- Istituto di Ematologia "Seràgnoli", IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Federico Pea
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, 40138 Bologna, Italy
- Clinical Pharmacology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
7
|
McColl ER, Croyle MA, Zamboni WC, Honer WG, Heise M, Piquette-Miller M, Goralski KB. COVID-19 Vaccines and the Virus: Impact on Drug Metabolism and Pharmacokinetics. Drug Metab Dispos 2023; 51:130-141. [PMID: 36273826 PMCID: PMC11022893 DOI: 10.1124/dmd.122.000934] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 09/07/2022] [Accepted: 09/30/2022] [Indexed: 01/08/2023] Open
Abstract
This article reports on an American Society of Pharmacology and Therapeutics, Division of Drug Metabolism and Disposition symposium held at Experimental Biology on April 2, 2022, in Philadelphia. As of July 2022, over 500 million people have been infected with SARS-CoV-2 (the virus causing COVID-19) and over 12 billion vaccine doses have been administered. Clinically significant interactions between viral infections and hepatic drug metabolism were first recognized over 40 years ago during a cluster of pediatric theophylline toxicity cases attributed to reduced hepatic drug metabolism amid an influenza B outbreak. Today, a substantive body of research supports that the activated innate immune response generally decreases hepatic cytochrome P450 activity. The interactions extend to drug transporters and other organs and have the potential to impact drug absorption, distribution, metabolism, and excretion (ADME). Based on this knowledge, altered ADME is predicted with SARS-CoV-2 infection or vaccination. The report begins with a clinical case exploring the possibility of SARS-CoV-2 vaccination increasing clozapine levels. This is followed by discussions of how SARS-CoV-2 infection or vaccines alter the metabolism and disposition of complex drugs, such as nanoparticles and biologics and small molecule therapies. The review concludes with a discussion of the effects of viral infections on placental amino acid transport and their potential to impact fetal development. The session improved our understanding of the impact of emerging viral infections and vaccine technologies on drug metabolism and disposition, which will help mitigate drug toxicity and improve drug and vaccine safety and effectiveness. SIGNIFICANCE STATEMENT: Altered pharmacokinetics of small molecule and complex molecule drugs and fetal brain distribution of amino acids following SARS-CoV-2 infection or immunization are possible. The proposed mechanisms involve decreased liver cytochrome P450 metabolism of small molecules, enhanced innate immune system metabolism of complex molecules, and altered placental and fetal blood-brain barrier amino acid transport, respectively. Future research is needed to understand the effects of these interactions on adverse drug responses, drug and vaccine safety, and effectiveness and fetal neurodevelopment.
Collapse
Affiliation(s)
- Eliza R McColl
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - Maria A Croyle
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - William C Zamboni
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - William G Honer
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - Mark Heise
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - Micheline Piquette-Miller
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| | - Kerry B Goralski
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada (E.R.M., M.P-M.); Department of Molecular Pharmaceutics and Drug Delivery and LaMontagne Center for Infectious Disease, University of Texas at Austin, College of Pharmacy, Austin, Texas (M.A.C.); Eshelman School of Pharmacy (W.C.Z.) and Department of Genetics, Department of Microbiology and Immunology, and The Rapidly Emerging Antiviral Drug Development Initiative (READDI) (M.H.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry, University of British Columbia and British Columbia Mental Health and Substance Use Services Research Institute, Vancouver, British Columbia, Canada (W.G.H.); and College of Pharmacy, Faculty of Health and Department of Pharmacology and Department of Pediatrics, Faculty of Medicine, Dalhousie University (K.B.G.); Division of Pediatric Hematology and Oncology, Department of Pediatrics, IWK Health Centre (K.B.G.); and Beatrice Hunter Cancer Research Institute (K.B.G.), Halifax, Nova Scotia, Canada
| |
Collapse
|
8
|
Coutant DE, Boulton DW, Dahal UP, Deslandes A, Grimaldi C, Pereira JNS, Säll C, Sarvaiya H, Schiller H, Tai G, Umehara K, Yuan Y, Dallas S. Therapeutic Protein Drug Interactions: A White Paper From the International Consortium for Innovation and Quality in Pharmaceutical Development. Clin Pharmacol Ther 2022; 113:1185-1198. [PMID: 36477720 DOI: 10.1002/cpt.2814] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022]
Abstract
Typically, therapeutic proteins (TPs) have a low risk for eliciting meaningful drug interactions (DIs). However, there are select instances where TP drug interactions (TP-DIs) of clinical concern can occur. This white paper discusses the various types of TP-DIs involving mechanisms such as changes in disease state, target-mediated drug disposition, neonatal Fc receptor (FcRn), or antidrug antibodies formation. The nature of TP drug interaction being investigated should determine whether the examination is conducted as a standalone TP-DI study in healthy participants, in patients, or assessed via population pharmacokinetic analysis. DIs involving antibody-drug conjugates are discussed briefly, but the primary focus here will be DIs involving cytokine modulation. Cytokine modulation can occur directly by certain TPs, or indirectly due to moderate to severe inflammation, infection, or injury. Disease states that have been shown to result in indirect disease-DIs that are clinically meaningful have been listed (i.e., typically a twofold change in the systemic exposure of a coadministered sensitive cytochrome P450 substrate drug). Type of disease and severity of inflammation should be the primary drivers for risk assessment for disease-DIs. While more clinical inflammatory marker data needs to be collected, the use of two or more clinical inflammatory markers (such as C-reactive protein, albumin, or interleukin 6) may help broadly categorize whether the predicted magnitude of inflammatory disease-DI risk is negligible, weak, or moderate to strong. Based on current knowledge, clinical DI studies are not necessary for all TPs, and should no longer be conducted in certain disease patient populations such as psoriasis, which do not have sufficient systemic inflammation to cause a meaningful indirect disease-DI.
Collapse
Affiliation(s)
- David E Coutant
- Drug Disposition Department, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - David W Boulton
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, Research & Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - Upendra P Dahal
- Pharmacokinetics and Drug Metabolism, Amgen, Inc., South San Francisco, California, USA
| | - Antoine Deslandes
- Translational Medicine and Early Development, Sanofi Research & Development, Chilly-Mazarin, France
| | - Christine Grimaldi
- Formerly of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut, USA
| | - Joao N S Pereira
- Drug Disposition & Design, Merck Healthcare KGaA, Darmstadt, Germany
| | - Carolina Säll
- Development Absorption, Distribution, Metabolism, and Elimination, Novo Nordisk A/S, Måløv, Denmark
| | - Hetal Sarvaiya
- Drug Metabolism, Pharmacokinetics, and Bioanalytical, AbbVie Inc., California, South San Francisco, USA
| | - Hilmar Schiller
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Guoying Tai
- Department of Metabolism and Pharmacokinetics, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Kenichi Umehara
- Pharmaceutical Sciences, Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Yang Yuan
- Formerly of Department of Metabolism and Pharmacokinetics, Bristol Myers Squibb Pharmaceutical Research and Development, Princeton, New Jersey, USA
| | - Shannon Dallas
- Preclinical Sciences & Translational Safety, Janssen Research & Development, Springhouse, Pennsylvania, USA
| |
Collapse
|
9
|
Klyushova LS, Perepechaeva ML, Grishanova AY. The Role of CYP3A in Health and Disease. Biomedicines 2022; 10:2686. [PMID: 36359206 PMCID: PMC9687714 DOI: 10.3390/biomedicines10112686] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
CYP3A is an enzyme subfamily in the cytochrome P450 (CYP) superfamily and includes isoforms CYP3A4, CYP3A5, CYP3A7, and CYP3A43. CYP3A enzymes are indiscriminate toward substrates and are unique in that these enzymes metabolize both endogenous compounds and diverse xenobiotics (including drugs); almost the only common characteristic of these compounds is lipophilicity and a relatively large molecular weight. CYP3A enzymes are widely expressed in human organs and tissues, and consequences of these enzymes' activities play a major role both in normal regulation of physiological levels of endogenous compounds and in various pathological conditions. This review addresses these aspects of regulation of CYP3A enzymes under physiological conditions and their involvement in the initiation and progression of diseases.
Collapse
Affiliation(s)
| | - Maria L. Perepechaeva
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Timakova Str. 2, 630117 Novosibirsk, Russia
| | | |
Collapse
|
10
|
Gao K, Wang R, Chen J, Cheng L, Frishcosy J, Huzumi Y, Qiu Y, Schluckbier T, Wei X, Wei GW. Methodology-Centered Review of Molecular Modeling, Simulation, and Prediction of SARS-CoV-2. Chem Rev 2022; 122:11287-11368. [PMID: 35594413 PMCID: PMC9159519 DOI: 10.1021/acs.chemrev.1c00965] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite tremendous efforts in the past two years, our understanding of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), virus-host interactions, immune response, virulence, transmission, and evolution is still very limited. This limitation calls for further in-depth investigation. Computational studies have become an indispensable component in combating coronavirus disease 2019 (COVID-19) due to their low cost, their efficiency, and the fact that they are free from safety and ethical constraints. Additionally, the mechanism that governs the global evolution and transmission of SARS-CoV-2 cannot be revealed from individual experiments and was discovered by integrating genotyping of massive viral sequences, biophysical modeling of protein-protein interactions, deep mutational data, deep learning, and advanced mathematics. There exists a tsunami of literature on the molecular modeling, simulations, and predictions of SARS-CoV-2 and related developments of drugs, vaccines, antibodies, and diagnostics. To provide readers with a quick update about this literature, we present a comprehensive and systematic methodology-centered review. Aspects such as molecular biophysics, bioinformatics, cheminformatics, machine learning, and mathematics are discussed. This review will be beneficial to researchers who are looking for ways to contribute to SARS-CoV-2 studies and those who are interested in the status of the field.
Collapse
Affiliation(s)
- Kaifu Gao
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Rui Wang
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Jiahui Chen
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Limei Cheng
- Clinical
Pharmacology and Pharmacometrics, Bristol
Myers Squibb, Princeton, New Jersey 08536, United States
| | - Jaclyn Frishcosy
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Yuta Huzumi
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Yuchi Qiu
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Tom Schluckbier
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Xiaoqi Wei
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Guo-Wei Wei
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Biochemistry and Molecular Biology, Michigan
State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
11
|
Critical clinical gaps in cancer precision nanomedicine development. J Control Release 2022; 345:811-818. [PMID: 35378214 DOI: 10.1016/j.jconrel.2022.03.055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 12/18/2022]
Abstract
Active targeting strategy is adopted in nanomedicine for cancer treatment. Personalizing the nanomedicine in accordance with patients' omics, under the precision medicine platform, is met with challenges in targeting ligand and matrix material selection at nanoformulation stage. The past 5-year literatures show that the nanoparticulate targeting ligand and matrix material are not selected based upon the cancer omics profiles of patients. The expression of cancer cellular target receptors and metabolizing enzymes is primarily influenced by age, gender, race/ethnic group and geographical origin of patients. The personalized perspective of a nanomedicine cannot be realised with premature digestion of matrix and targeting ligand by specific metabolizing enzymes that are overexpressed by the patients, and unmatched targeting ligand to the majority of cell surface receptors overexpressed in cancer. Omics analysis of individual metabolizing enzyme and cancer cell surface receptor expressed in cancer facilitates targeting ligand and matrix material selection in nanomedicine development.
Collapse
|
12
|
Elfaki I. The Impact of the Coronavirus (COVID-19) Infection on the Drug-Metabolizing Enzymes Cytochrome P450s. Drug Metab Lett 2022; 15:DML-EPUB-122095. [PMID: 35362390 DOI: 10.2174/1872312815666220331142046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/10/2022] [Accepted: 02/21/2022] [Indexed: 11/22/2022]
Abstract
Coronaviruses cause disease in human and animals. In 2019 a novel coronavirus was first characterized in Wuhan, China. It causes acute respiratory disease and designated the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or COVID-19. The COVID-19 spread to all cities of China, and in 2020 to the whole world. Patients with COVID-19 may recover without medical treatment. However, some patients need medical care. The Cytochrome p450s (CYP450s) are large superfamily of enzymes catalyze the metabolism of endogenous substrates and xenobiotics. CYP450s catalyze the biotransformation of 80% of the drug in clinical use. The CYP450 present in liver, lungs, intestine and other tissues. COVID-19 has been reported to decrease the activity of certain isoforms of CYP450s in an isoform specific manner. Furthermore, the COVID-19 infection decreases the liver functions including the drug clearance or detoxification medicated by the CYP450s. The healthcare providers should be aware of this disease-drug interaction when prescribing drugs for treatment of COVID-19 and other comorbidities.
Collapse
Affiliation(s)
- Imadeldin Elfaki
- Department of Biochemistry, Faculty of Science, University of Tabuk, Kingdom of Saudi Arabia
| |
Collapse
|
13
|
Gatti M, Pea F. The Cytokine Release Syndrome and/or the Proinflammatory Cytokines as Underlying Mechanisms of Downregulation of Drug Metabolism and Drug Transport: A Systematic Review of the Clinical Pharmacokinetics of Victim Drugs of this Drug-Disease Interaction Under Different Clinical Conditions. Clin Pharmacokinet 2022; 61:1519-1544. [PMID: 36059001 PMCID: PMC9441320 DOI: 10.1007/s40262-022-01173-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND AND OBJECTIVE An ever-growing body of evidence supports the impact of cytokine modulation on the patient's phenotypic drug response. The aim of this systematic review was to analyze the clinical studies that assessed the pharmacokinetics of victim drugs of this drug-disease interaction in the presence of different scenarios of cytokine modulation in comparison with baseline conditions. METHODS We conducted a systematic review by searching the PubMed-MEDLINE database from inception until February 2022 to retrieve prospective and/or retrospective observational studies, population pharmacokinetic studies, phase I studies, and/or case series/reports that investigated the impact of cytokine modulation on the pharmacokinetic behavior of victim drugs. Only studies providing quantitative pharmacokinetic data of victim drugs by comparing normal status versus clinical conditions with documented cytokine modulation or by assessing the influence of anti-inflammatory biological agents on metabolism and/or transport of victim drugs were included. RESULTS Overall, 26 studies were included. Rheumatoid arthritis (6/26; 23.1%) and sepsis (5/26; 19.2%) were the two most frequently investigated pro-inflammatory clinical scenarios. The victim drug most frequently assessed was midazolam (14/26; 53.8%; as a probe for cytochrome P450 [CYP] 3A4). Cytokine modulation showed a moderate inhibitory effect on CYP3A4-mediated metabolism (area under the concentration-time curve increase and/or clearance decrease between 1.98-fold and 2.59-fold) and a weak-to-moderate inhibitory effect on CYP1A2, CYP2C9, and CYP2C19-mediated metabolism (in the area under the concentration-time curve increase or clearance decrease between 1.29-fold and 1.97-fold). Anti-interleukin-6 agents showed remarkable activity in counteracting downregulation of CYP3A4-mediated activity (increase in the area under the concentration-time curve between 1.75-fold and 2.56-fold). CONCLUSIONS Cytokine modulation may cause moderate or weak-to-moderate downregulation of metabolism/transport of victim drugs, and this may theoretically have relevant clinical consequences.
Collapse
Affiliation(s)
- Milo Gatti
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy ,Clinical Pharmacology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | - Federico Pea
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy ,Clinical Pharmacology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Massarenti 9, 40138 Bologna, Italy
| |
Collapse
|
14
|
Lenoir C, Rollason V, Desmeules JA, Samer CF. Influence of Inflammation on Cytochromes P450 Activity in Adults: A Systematic Review of the Literature. Front Pharmacol 2021; 12:733935. [PMID: 34867341 PMCID: PMC8637893 DOI: 10.3389/fphar.2021.733935] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/13/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Available in-vitro and animal studies indicate that inflammation impacts cytochromes P450 (CYP) activity via multiple and complex transcriptional and post-transcriptional mechanisms, depending on the specific CYP isoforms and the nature of inflammation mediators. It is essential to review the current published data on the impact of inflammation on CYP activities in adults to support drug individualization based on comorbidities and diseases in clinical practice. Methods: This systematic review was conducted in PubMed through 7th January 2021 looking for articles that investigated the consequences of inflammation on CYP activities in adults. Information on the source of inflammation, victim drugs (and CYPs involved), effect of disease-drug interaction, number of subjects, and study design were extracted. Results: The search strategy identified 218 studies and case reports that met our inclusion criteria. These articles were divided into fourteen different sources of inflammation (such as infection, autoimmune diseases, cancer, therapies with immunomodulator…). The impact of inflammation on CYP activities appeared to be isoform-specific and dependent on the nature and severity of the underlying disease causing the inflammation. Some of these drug-disease interactions had a significant influence on drug pharmacokinetic parameters and on clinical management. For example, clozapine levels doubled with signs of toxicity during infections and the concentration ratio between clopidogrel's active metabolite and clopidogrel is 48-fold lower in critically ill patients. Infection and CYP3A were the most cited perpetrator of inflammation and the most studied CYP, respectively. Moreover, some data suggest that resolution of inflammation results in a return to baseline CYP activities. Conclusion: Convincing evidence shows that inflammation is a major factor to be taken into account in drug development and in clinical practice to avoid any efficacy or safety issues because inflammation modulates CYP activities and thus drug pharmacokinetics. The impact is different depending on the CYP isoform and the inflammatory disease considered. Moreover, resolution of inflammation appears to result in a normalization of CYP activity. However, some results are still equivocal and further investigations are thus needed.
Collapse
Affiliation(s)
- Camille Lenoir
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care, and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Victoria Rollason
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care, and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jules A Desmeules
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care, and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Caroline F Samer
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care, and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
15
|
Cattaneo D, Pasina L, Conti F, Giacomelli A, Oreni L, Pezzati L, Bonazzetti C, Piscaglia M, Carrozzo G, Antinori S, Gervasoni C. Risks of potential drug-drug interactions in COVID-19 patients treated with corticosteroids: a single-center experience. J Endocrinol Invest 2021; 44:2849-2851. [PMID: 34053008 PMCID: PMC8164688 DOI: 10.1007/s40618-021-01604-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 05/22/2021] [Indexed: 02/07/2023]
Affiliation(s)
- D Cattaneo
- Gestione Ambulatoriale Politerapie (GAP) Outpatient Clinic, ASST Fatebenefratelli-Sacco University Hospital, Milan, Italy
- Unit of Clinical Pharmacology, ASST Fatebenefratelli-Sacco University Hospital, Milan, Italy
| | - L Pasina
- Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - F Conti
- Department of Infectious Diseases, ASST Fatebenefratelli-Sacco University Hospital, Via GB Grassi 74, 20157, Milano, Italy
| | - A Giacomelli
- Department of Infectious Diseases, ASST Fatebenefratelli-Sacco University Hospital, Via GB Grassi 74, 20157, Milano, Italy
| | - L Oreni
- Department of Infectious Diseases, ASST Fatebenefratelli-Sacco University Hospital, Via GB Grassi 74, 20157, Milano, Italy
| | - L Pezzati
- Department of Infectious Diseases, ASST Fatebenefratelli-Sacco University Hospital, Via GB Grassi 74, 20157, Milano, Italy
| | - C Bonazzetti
- Department of Infectious Diseases, ASST Fatebenefratelli-Sacco University Hospital, Via GB Grassi 74, 20157, Milano, Italy
| | - M Piscaglia
- Department of Infectious Diseases, ASST Fatebenefratelli-Sacco University Hospital, Via GB Grassi 74, 20157, Milano, Italy
| | - G Carrozzo
- Department of Infectious Diseases, ASST Fatebenefratelli-Sacco University Hospital, Via GB Grassi 74, 20157, Milano, Italy
| | - S Antinori
- Department of Infectious Diseases, ASST Fatebenefratelli-Sacco University Hospital, Via GB Grassi 74, 20157, Milano, Italy
| | - C Gervasoni
- Gestione Ambulatoriale Politerapie (GAP) Outpatient Clinic, ASST Fatebenefratelli-Sacco University Hospital, Milan, Italy.
- Department of Infectious Diseases, ASST Fatebenefratelli-Sacco University Hospital, Via GB Grassi 74, 20157, Milano, Italy.
| |
Collapse
|
16
|
Fujino C, Sanoh S, Katsura T. Variation in Expression of Cytochrome P450 3A Isoforms and Toxicological Effects: Endo- and Exogenous Substances as Regulatory Factors and Substrates. Biol Pharm Bull 2021; 44:1617-1634. [PMID: 34719640 DOI: 10.1248/bpb.b21-00332] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The CYP3A subfamily, which includes isoforms CYP3A4, CYP3A5, and CYP3A7 in humans, plays important roles in the metabolism of various endogenous and exogenous substances. Gene and protein expression of CYP3A4, CYP3A5, and CYP3A7 show large inter-individual differences, which are caused by many endogenous and exogenous factors. Inter-individual differences can cause negative outcomes, such as adverse drug events and disease development. Therefore, it is important to understand the variations in CYP3A expression caused by endo- and exogenous factors, as well as the variation in the metabolism and kinetics of endo- and exogenous substrates. In this review, we summarize the factors regulating CYP3A expression, such as bile acids, hormones, microRNA, inflammatory cytokines, drugs, environmental chemicals, and dietary factors. In addition, variations in CYP3A expression under pathological conditions, such as coronavirus disease 2019 and liver diseases, are described as examples of the physiological effects of endogenous factors. We also summarize endogenous and exogenous substrates metabolized by CYP3A isoforms, such as cholesterol, bile acids, hormones, arachidonic acid, vitamin D, and drugs. The relationship between the changes in the kinetics of these substrates and the toxicological effects in our bodies are discussed. The usefulness of these substrates and metabolites as endogenous biomarkers for CYP3A activity is also discussed. Notably, we focused on discrimination between CYP3A4, CYP3A5, and CYP3A7 to understand inter-individual differences in CYP3A expression and function.
Collapse
Affiliation(s)
- Chieri Fujino
- Laboratory of Clinical Pharmaceutics and Therapeutics, College of Pharmaceutical Sciences, Ritsumeikan University
| | - Seigo Sanoh
- Graduate School of Biomedical and Health Sciences, Hiroshima University.,School of Pharmaceutical Sciences, Wakayama Medical University
| | - Toshiya Katsura
- Laboratory of Clinical Pharmaceutics and Therapeutics, College of Pharmaceutical Sciences, Ritsumeikan University
| |
Collapse
|
17
|
Bartelink IH, Bet PM, Widmer N, Guidi M, Duijvelaar E, Grob B, Honeywell R, Evelo A, Tielbeek IPE, Snape SD, Hamer H, Decosterd LA, Jan Bogaard H, Aman J, Swart EL. Elevated acute phase proteins affect pharmacokinetics in COVID-19 trials: Lessons from the CounterCOVID - imatinib study. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:1497-1511. [PMID: 34608769 PMCID: PMC8646516 DOI: 10.1002/psp4.12718] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/09/2021] [Accepted: 09/15/2021] [Indexed: 12/04/2022]
Abstract
This study aimed to determine whether published pharmacokinetic (PK) models can adequately predict the PK profile of imatinib in a new indication, such as coronavirus disease 2019 (COVID‐19). Total (bound + unbound) and unbound imatinib plasma concentrations obtained from 134 patients with COVID‐19 participating in the CounterCovid study and from an historical dataset of 20 patients with gastrointestinal stromal tumor (GIST) and 85 patients with chronic myeloid leukemia (CML) were compared. Total imatinib area under the concentration time curve (AUC), maximum concentration (Cmax) and trough concentration (Ctrough) were 2.32‐fold (95% confidence interval [CI] 1.34–3.29), 2.31‐fold (95% CI 1.33–3.29), and 2.32‐fold (95% CI 1.11–3.53) lower, respectively, for patients with CML/GIST compared with patients with COVID‐19, whereas unbound concentrations were comparable among groups. Inclusion of alpha1‐acid glycoprotein (AAG) concentrations measured in patients with COVID‐19 into a previously published model developed to predict free imatinib concentrations in patients with GIST using total imatinib and plasma AAG concentration measurements (AAG‐PK‐Model) gave an estimated mean (SD) prediction error (PE) of −20% (31%) for total and −7.0% (56%) for unbound concentrations. Further covariate modeling with this combined dataset showed that in addition to AAG; age, bodyweight, albumin, CRP, and intensive care unit admission were predictive of total imatinib oral clearance. In conclusion, high total and unaltered unbound concentrations of imatinib in COVID‐19 compared to CML/GIST were a result of variability in acute phase proteins. This is a textbook example of how failure to take into account differences in plasma protein binding and the unbound fraction when interpreting PK of highly protein bound drugs, such as imatinib, could lead to selection of a dose with suboptimal efficacy in patients with COVID‐19.
Collapse
Affiliation(s)
- Imke H Bartelink
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Pierre M Bet
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Nicolas Widmer
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.,Specialised Centre for Emergency and Disaster Pharmacy, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland.,Pharmacy of the Eastern Vaud Hospitals, Rennaz, Switzerland
| | - Monia Guidi
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.,Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Erik Duijvelaar
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Bram Grob
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Richard Honeywell
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Amanda Evelo
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Ivo P E Tielbeek
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | | | - Henrike Hamer
- Department of Clinical Chemistry, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Laurent A Decosterd
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Harm Jan Bogaard
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Jurjan Aman
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| | - Eleonora L Swart
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC, location VUmc, Amsterdam, The Netherlands
| |
Collapse
|
18
|
White CM. Inflammation Suppresses Patients' Ability to Metabolize Cytochrome P450 Substrate Drugs. Ann Pharmacother 2021; 56:809-819. [PMID: 34590872 DOI: 10.1177/10600280211047864] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE To assess the impact of inflammation on cytochrome P450 (CYP) drug metabolism in human subjects. DATA SOURCES A PubMed search was done from 1980 to July 2021 limited to human subjects and English language using a search strategy of (((phase I metabolism) OR (CYP) OR (cytochrome P450)) AND (inflammatory OR inflammation)). STUDY SELECTION AND DATA EXTRACTION Narrative review of human studies assessing the impact of inflammation or inflammatory suppression with biologic drugs on CYP drug metabolism were used. DATA SYNTHESIS Patients with inflammatory conditions ranging from fungal, viral, or bacterial infections to noninfectious causes (critical illness, surgical procedure, cancer, or transplantation of stem cells or organs) have suppressed drug metabolism. Markers of inflammation such as C-reactive protein or α-1-acid glycoprotein are correlated with reduced clearance through CYP3A4, CYP1A2, and CYP2C19. Elevated interleukin-6 concentrations are also associated or correlated with reduced clearance for CYP3A4 and CYP2C-19 isoenzymes. There was insufficient information to properly assess CYP2D6. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE Health professionals should appreciate that patients with acute or chronic inflammation from infectious or noninfectious causes could have suppressed drug metabolism through CYP3A4, CYP1A2, and CYP2C19. For narrow therapeutic index drugs, such as many of the drugs assessed in this review, that means more judicious drug monitoring to prevent adverse events. CONCLUSIONS Like other types of drug-drug or drug-disease interactions, inflammation can alter the steady-state concentration of CYP metabolized drugs.
Collapse
Affiliation(s)
- C Michael White
- University of Connecticut School of Pharmacy, Storrs, CT, USA.,Hartford Hospital, Hartford, CT, USA
| |
Collapse
|
19
|
Stader F, Battegay M, Sendi P, Marzolini C. Physiologically Based Pharmacokinetic Modelling to Investigate the Impact of the Cytokine Storm on CYP3A Drug Pharmacokinetics in COVID-19 Patients. Clin Pharmacol Ther 2021; 111:579-584. [PMID: 34496043 PMCID: PMC8652944 DOI: 10.1002/cpt.2402] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/03/2021] [Indexed: 12/16/2022]
Abstract
Patients with coronavirus disease 2019 (COVID‐19) may experience a cytokine storm with elevated interleukin‐6 (IL‐6) levels in response to severe acute respiratory syndrome‐coronavirus 2 (SARS‐CoV‐2). IL‐6 suppresses hepatic enzymes, including CYP3A; however, the effect on drug exposure and drug‐drug interaction magnitudes of the cytokine storm and resulting elevated IL‐6 levels have not been characterized in patients with COVID‐19. We used physiologically‐based pharmacokinetic (PBPK) modeling to simulate the effect of inflammation on the pharmacokinetics of CYP3A metabolized drugs. A PBPK model was developed for lopinavir boosted with ritonavir (LPV/r), using clinically observed data from people living with HIV (PLWH). The inhibition of CYPs by IL‐6 was implemented by a semimechanistic suppression model and verified against clinical data from patients with COVID‐19, treated with LPV/r. Subsequently, the verified model was used to simulate the effect of various clinically observed IL‐6 levels on the exposure of LPV/r and midazolam, a CYP3A model drug. Clinically observed LPV/r concentrations in PLWH and patients with COVID‐19 were predicted within the 95% confidence interval of the simulation results, demonstrating its predictive capability. Simulations indicated a twofold higher LPV exposure in patients with COVID‐19 compared with PLWH, whereas ritonavir exposure was predicted to be comparable. Varying IL‐6 levels under COVID‐19 had only a marginal effect on LPV/r pharmacokinetics according to our model. Simulations showed that a cytokine storm increased the exposure of the CYP3A paradigm substrate midazolam by 40%. Our simulations suggest that CYP3A metabolism is altered in patients with COVID‐19 having increased cytokine release. Caution is required when prescribing narrow therapeutic index drugs particularly in the presence of strong CYP3A inhibitors.
Collapse
Affiliation(s)
- Felix Stader
- Departments of Medicine and Clinical Research, University Hospital Basel, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Manuel Battegay
- Departments of Medicine and Clinical Research, University Hospital Basel, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Parham Sendi
- Departments of Medicine and Clinical Research, University Hospital Basel, Basel, Switzerland.,University of Basel, Basel, Switzerland.,Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Catia Marzolini
- Departments of Medicine and Clinical Research, University Hospital Basel, Basel, Switzerland.,University of Basel, Basel, Switzerland.,Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
20
|
Lenoir C, Terrier J, Gloor Y, Curtin F, Rollason V, Desmeules JA, Daali Y, Reny JL, Samer CF. Impact of SARS-CoV-2 Infection (COVID-19) on Cytochromes P450 Activity Assessed by the Geneva Cocktail. Clin Pharmacol Ther 2021; 110:1358-1367. [PMID: 34473836 PMCID: PMC8653122 DOI: 10.1002/cpt.2412] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/20/2021] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019 (COVID‐19), caused by severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) infection, is a severe acute respiratory syndrome with an underlying inflammatory state. We have previously demonstrated that acute inflammation modulates cytochromes P450 (CYPs) activity in an isoform‐specific manner. We therefore hypothesized that COVID‐19 might also impact CYP activity, and thus aimed to evaluate the impact of acute inflammation in the context of SARS‐CoV‐2 infection on the six main human CYPs activity. This prospective observational study was conducted in 28 patients hospitalized at the Geneva University Hospitals (Switzerland) with a diagnosis of moderate to severe COVID‐19. They received the Geneva phenotyping cocktail orally during the first 72 hours of hospitalization and after 3 months. Capillary blood samples were collected 2 hours after cocktail administration to assess the metabolic ratios (MRs) of CYP1A2, 2B6, 2C9, 2C19, 2D6, and 3A. C‐reactive protein (CRP), interleukin 6 (IL‐6), and tumor necrosis factor‐α (TNF‐α) levels were also measured in blood. CYP1A2, CYP2C19, and CYP3A MRs decreased by 52.6% (P = 0.0001), 74.7% (P = 0.0006), and 22.8% (P = 0.045), respectively, in patients with COVID‐19. CYP2B6 and CYP2C9 MRs increased by 101.1% (P = 0.009) and 55.8% (P = 0.0006), respectively. CYP2D6 MR variation did not reach statistical significance (P = 0.072). As expected, COVID‐19 was a good acute inflammation model as mean serum levels of CRP, IL‐6, and TNF‐α were significantly (P < 0.001) higher during SARS‐CoV‐2 infection. CYP activity are modulated in an isoform‐specific manner by SARS‐CoV‐2 infection. The pharmacokinetics of CYP substrates, whether used to treat the disease or as the usual treatment of patients, could be therefore clinically impacted.
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
| | - Jean Terrier
- Division of Clinical Pharmacology and Toxicology, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland.,Division of General Internal Medicine, Department of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Yvonne Gloor
- Division of Clinical Pharmacology and Toxicology, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - François Curtin
- Division of Clinical Pharmacology and Toxicology, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Geneva, Switzerland.,Personalized Health and Related Technologies, Swiss Federal Institute of Technology in Zurich, Zurich, 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.,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.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jean-Luc Reny
- Division of General Internal Medicine, Department of Medicine, Geneva University Hospitals, 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
|
21
|
Calcagno A, Trunfio M, D'Avolio A, Di Perri G, Bonora S. The impact of age on antiretroviral drug pharmacokinetics in the treatment of adults living with HIV. Expert Opin Drug Metab Toxicol 2021; 17:665-676. [PMID: 33913394 DOI: 10.1080/17425255.2021.1915285] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION People living with HIV (PLWH) are aging and will receive life-long treatment: despite substantial improvement in drug efficacy and tolerability, side effects still occur and they can blunt antiretroviral treatment effectiveness. Since age may affect drug exposure and may be associated with side-effects we aimed at reviewing available data on the effect of age on antiretrovirals' pharmacokinetics in adult patients. AREAS COVERED We searched public databases and major conference proceedings for data on age and pharmacokinetics/pharmacodynamics in PLWH. We limited our review to currently used drugs and focused on population pharmacokinetics and physiologically-based pharmacokinetic modeling studies. EXPERT OPINION Available evidence of a potential detrimental effect in elderly PLWH is limited by study design and small sample sizes. Careful consideration of undoubtful benefits and potential harms is advised when prescribing ARVs to geriatric patients and the knowledge of pharmacokinetics changes need to be included in the process. With the 'greying' of the pandemic we need studies with a specific focus on geriatric patients living with HIV that will consider specific phenotypes and associated changes (including sarcopenia).
Collapse
Affiliation(s)
- Andrea Calcagno
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Mattia Trunfio
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Antonio D'Avolio
- Laboratory and Clinical Pharmacology and Pharmacogenetics, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Giovanni Di Perri
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Stefano Bonora
- Unit of Infectious Diseases, Department of Medical Sciences, University of Torino, Torino, Italy
| |
Collapse
|
22
|
Cojutti PG, Londero A, Della Siega P, Givone F, Fabris M, Biasizzo J, Tascini C, Pea F. Authors' Reply to Cattaneo et al.: "Comment on: Comparative Population Pharmacokinetics of Darunavir in SARS-CoV-2 Patients vs. HIV Patients: The Role of Interleukin6". Clin Pharmacokinet 2021; 60:833-834. [PMID: 33864624 PMCID: PMC8052551 DOI: 10.1007/s40262-021-00996-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Pier Giorgio Cojutti
- Department of Medicine, University of Udine, Udine, Italy.,Institute of Clinical Pharmacology, Santa Maria della Misericordia University Hospital of Udine, ASUFC, Udine, Italy
| | - Angela Londero
- Clinic of Infectious Diseases, Santa Maria della Misericordia University Hospital of Udine, ASUFC, Udine, Italy
| | - Paola Della Siega
- Clinic of Infectious Diseases, Santa Maria della Misericordia University Hospital of Udine, ASUFC, Udine, Italy
| | - Filippo Givone
- Clinic of Infectious Diseases, Santa Maria della Misericordia University Hospital of Udine, ASUFC, Udine, Italy
| | - Martina Fabris
- Institute of Clinical Pathology, Santa Maria della Misericordia University Hospital of Udine, ASUFC, Udine, Italy
| | - Jessica Biasizzo
- Institute of Clinical Pathology, Santa Maria della Misericordia University Hospital of Udine, ASUFC, Udine, Italy
| | - Carlo Tascini
- Clinic of Infectious Diseases, Santa Maria della Misericordia University Hospital of Udine, ASUFC, Udine, Italy
| | - Federico Pea
- Department of Medicine, University of Udine, Udine, Italy. .,Institute of Clinical Pharmacology, Santa Maria della Misericordia University Hospital of Udine, ASUFC, Udine, Italy. .,Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy. .,University Hospital IRCCS Policlinico Sant'Orsola, Bologna, Italy.
| |
Collapse
|
23
|
Comment on "Comparative Population Pharmacokinetics of Darunavir in SARS-CoV-2 Patients vs. HIV Patients: The Role of Interleukin-6". Clin Pharmacokinet 2021; 60:829-831. [PMID: 33864625 PMCID: PMC8052541 DOI: 10.1007/s40262-021-00992-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2021] [Indexed: 11/25/2022]
|
24
|
Deb S, Arrighi S. Potential Effects of COVID-19 on Cytochrome P450-Mediated Drug Metabolism and Disposition in Infected Patients. Eur J Drug Metab Pharmacokinet 2021; 46:185-203. [PMID: 33538960 PMCID: PMC7859725 DOI: 10.1007/s13318-020-00668-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Coronavirus Disease 2019 (COVID-19) has been a global health crisis since it was first identified in December 2019. In addition to fever, cough, headache, and shortness of breath, an intense increase in immune response-based inflammation has been the hallmark of Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2) virus infection. This narrative review summarizes and critiques pathophysiology of COVID-19 and its plausible effects on drug metabolism and disposition. The release of inflammatory cytokines (e.g., interleukins, tumor necrosis factor α), also known as 'cytokine storm', leads to altered molecular pathophysiology and eventually organ damage in the lung, heart, and liver. The laboratory values for various liver function tests (e.g., alanine aminotransferase, aspartate aminotransferase, total bilirubin, albumin) have indicated potential hepatocellular injury in COVID-19 patients. Since the liver is the powerhouse of protein synthesis and the primary site of cytochrome P450 (CYP)-mediated drug metabolism, even a minor change in the liver function status has the potential to affect the hepatic clearance of xenobiotics. It has now been well established that extreme increases in cytokine levels are common in COVID-19 patients, and previous studies with patients infected with non-SARS-CoV-2 virus have shown that CYP enzymes can be suppressed by an infection-related cytokine increase and inflammation. Alongside the investigational COVID-19 drugs, the patients may also be on therapeutics for comorbidities; especially epidemiological studies have indicated that individuals with hypertension, hyperglycemia, and obesity are more vulnerable to COVID-19 than the average population. This complicates the drug-disease interaction profile of the patients as both the investigational drugs (e.g., remdesivir, dexamethasone) and the agents for comorbidities can be affected by compromised CYP-mediated hepatic metabolism. Overall, it is imperative that healthcare professionals pay attention to the COVID-19 and CYP-driven drug metabolism interactions with the goal to adjust the dose or discontinue the affected drugs as appropriate.
Collapse
Affiliation(s)
- Subrata Deb
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL, 33169, USA.
| | - Scott Arrighi
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL, 33169, USA
| |
Collapse
|
25
|
Clinically Significant Drug Interactions Between Psychotropic Agents and Repurposed COVID-19 Therapies. CNS Drugs 2021; 35:345-384. [PMID: 33866523 PMCID: PMC8053373 DOI: 10.1007/s40263-021-00811-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/26/2021] [Indexed: 02/07/2023]
Abstract
As many patients with underlying psychiatric disorders may be infected with COVID-19, and COVID-19-affected subjects may frequently experience a new onset of psychiatric manifestations, concomitant use of psychotropic medications and COVID-19 therapies is expected to be highly likely and raises concerns of clinically relevant drug interactions. In this setting, four major mechanisms responsible for drug interactions involving psychotropic agents and COVID-19 therapies may be identified: (1) pharmacokinetic drug-drug interactions mainly acting on cytochrome P450; (2) pharmacodynamic drug-drug interactions resulting in additive or synergistic toxicity; (3) drug-disease interactions according to stage and severity of the disease; and (4) pharmacogenetic issues associated with polymorphisms of cytochrome P450 isoenzymes. In this review, we summarise the available literature on relevant drug interactions between psychotropic agents and COVID-19 therapies, providing practical clinical recommendations and potential management strategies according to severity of illness and clinical scenario.
Collapse
|
26
|
Cobicistat/darunavir/cobicistat/darunavir/emtricitabine/tenofovir-alafenamide/darunavir/ritonavir. REACTIONS WEEKLY 2020. [PMCID: PMC7726297 DOI: 10.1007/s40278-020-86953-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
27
|
Guaraldi G, Milic J, Cozzi-Lepri A, Pea F, Mussini C. Tocilizumab in COVID-19: finding the optimal route and dose - Authors' reply. LANCET RHEUMATOLOGY 2020; 2:e739-e740. [PMID: 32964210 PMCID: PMC7498220 DOI: 10.1016/s2665-9913(20)30333-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Giovanni Guaraldi
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena 41124, Italy.,Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Jovana Milic
- Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandro Cozzi-Lepri
- Centre for Clinical Research, Epidemiology, Modelling and Evaluation, Institute for Global Health, University College London, London, UK
| | - Federico Pea
- Institute of Clinical Pharmacology, Azienda Ospedaliero-Universitaria Santa Maria Della Misericordia, University of Udine, Udine, Italy
| | - Cristina Mussini
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria Policlinico of Modena, Modena 41124, Italy.,Department of Surgical, Medical, Dental and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|