Drug-Drug Interactions between COVID-19 and Tuberculosis Medications: A Comprehensive Review of CYP450 and Transporter-Mediated Effects
Abstract
:1. Introduction
2. COVID-19 and Tuberculosis Co-Infection
3. Role of Metabolic Enzymes and Transporters in Drug Disposition
4. FDA-Approved COVID-19 Treatments and Their Potential DDI with Anti-TB Drugs
4.1. Remdesivir (RDV)
4.1.1. A Brief on RDV
4.1.2. Potential DDI with Anti-TB Drugs
4.2. Paxlovid (Nirmatrelvir and Ritonavir Combination)
4.2.1. A Brief on Paxlovid
4.2.2. Potential DDI with Anti-TB Drugs
4.3. Molnupiravir
4.3.1. A Brief on Molnupiravir
4.3.2. Potential DDI with Anti-TB Drugs
5. Clinical Implications and Knowledge Gaps
6. Implication of PBPK Modeling Approach to Facilitate the Clinical Decision-Making
7. Recommendations for Frontline Healthcare Professionals
7.1. Stay Informed
7.2. Foster Interprofessional Collaboration
7.3. Create Comprehensive and Personalized Treatment Plans
7.4. Monitor and Report Adverse Events
7.5. Educate Patients
7.6. Engage in Shared Decision-Making
7.7. Pursue Continuous Learning
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Gupta, N.; Ish, P.; Gupta, A.; Malhotra, N.; Caminero, J.A.; Singla, R.; Kumar, R.; Yadav, S.R.; Dev, N.; Agrawal, S.; et al. A profile of a retrospective cohort of 22 patients with COVID-19 and active/treated tuberculosis. Eur. Respir. J. 2020, 56, 2003408. [Google Scholar] [CrossRef] [PubMed]
- Tadolini, M.; Codecasa, L.R.; García-García, J.M.; Blanc, F.X.; Borisov, S.; Alffenaar, J.W.; Andréjak, C.; Bachez, P.; Bart, P.A.; Belilovski, E.; et al. Active tuberculosis, sequelae and COVID-19 co-infection: First cohort of 49 cases. Eur. Respir. J. 2020, 56, 2001398. [Google Scholar] [CrossRef] [PubMed]
- Beigel, J.H.; Tomashek, K.M.; Dodd, L.E.; Mehta, A.K.; Zingman, B.S.; Kalil, A.C.; Hohmann, E.; Chu, H.Y.; Luetkemeyer, A.; Kline, S.; et al. Remdesivir for the Treatment of Covid-19—Final Report. N. Engl. J. Med. 2020, 383, 1813–1826. [Google Scholar] [CrossRef] [PubMed]
- Aleissa, M.M.; Silverman, E.A.; Acosta, L.M.P.; Nutt, C.T.; Richterman, A.; Marty, F.M. New Perspectives on Antimicrobial Agents: Remdesivir Treatment for COVID-19. Antimicrob. Agents Chemother. 2020, 65, e01814-20. [Google Scholar] [CrossRef] [PubMed]
- Fukunaga, R.; Glaziou, P.; Harris, J.B.; Date, A.; Floyd, K.; Kasaeva, T. Epidemiology of Tuberculosis and Progress toward Meeting Global Targets—Worldwide, 2019. MMWR Morb. Mortal. Wkly. Rep. 2021, 70, 427–430. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Lin, D.; Sun, X.; Curth, U.; Drosten, C.; Sauerhering, L.; Becker, S.; Rox, K.; Hilgenfeld, R. Crystal structure of SARS-CoV-2 main protease provides a basis for design of improved α-ketoamide inhibitors. Science 2020, 368, 409–412. [Google Scholar] [CrossRef]
- Barnard, D.L.; Hubbard, V.D.; Burton, J.; Smee, D.F.; Morrey, J.D.; Otto, M.J.; Sidwell, R.W. Inhibition of Severe Acute Respiratory Syndrome-Associated Coronavirus (SARSCoV) by Calpain Inhibitors and β-D-N4-Hydroxycytidine. Antivir. Chem. Chemother. 2004, 15, 15–22. [Google Scholar] [CrossRef] [PubMed]
- Painter, G.R.; Natchus, M.G.; Cohen, O.; Holman, W.; Painter, W.P. Developing a direct acting, orally available antiviral agent in a pandemic: The evolution of molnupiravir as a potential treatment for COVID-19. Curr. Opin. Virol. 2021, 50, 17–22. [Google Scholar] [CrossRef] [PubMed]
- Riccardi, N.; Canetti, D.; Rodari, P.; Besozzi, G.; Saderi, L.; Dettori, M.; Codecasa, L.R.; Sotgiu, G. Tuberculosis and pharmacological interactions: A narrative review. Curr. Res. Pharmacol. Drug Discov. 2021, 2, 100007. [Google Scholar] [CrossRef] [PubMed]
- Ogu, C.C.; Maxa, J.L. Drug interactions due to cytochrome P450. Bayl. Univ. Med. Cent. Proc. 2000, 13, 421–423. [Google Scholar] [CrossRef]
- Lynch, T.; Price, A. The effect of cytochrome P450 metabolism on drug response, interactions, and adverse effects. Am. Fam. Physician 2007, 76, 391–396. [Google Scholar] [PubMed]
- Bibi, Z. Role of cytochrome P450 in drug interactions. Nutr. Metab. 2008, 5, 27. [Google Scholar] [CrossRef] [PubMed]
- Rojas, J.C.; Aguilar, B.; Rodríguez-Maldonado, E.; Collados, M.T. Pharmacogenetics of oral anticoagulants. Blood Coagul. Fibrinolysis 2005, 16, 389–398. [Google Scholar] [CrossRef] [PubMed]
- Sproule, B.A.; Otton, S.V.; Cheung, S.W.; Zhong, X.H.; Romach, M.K.; Sellers, E.M. CYP2D6 inhibition in patients treated with sertraline. J. Clin. Psychopharmacol. 1997, 17, 102–106. [Google Scholar] [CrossRef] [PubMed]
- Koepsell, H. Organic Cation Transporters in Health and Disease. Pharmacol. Rev. 2020, 72, 253–319. [Google Scholar] [CrossRef] [PubMed]
- Yonezawa, A.; Inui, K.-I. Importance of the multidrug and toxin extrusion MATE/SLC47A family to pharmacokinetics, pharmacodynamics/toxicodynamics and pharmacogenomics. Br. J. Pharmacol. 2011, 164, 1817–1825. [Google Scholar] [CrossRef] [PubMed]
- Pedersen, J.M.; Matsson, P.; Bergström, C.A.S.; Hoogstraate, J.; Norén, A.; LeCluyse, E.L.; Artursson, P. Early Identification of Clinically Relevant Drug Interactions With the Human Bile Salt Export Pump (BSEP/ABCB11). Toxicol. Sci. 2013, 136, 328–343. [Google Scholar] [CrossRef] [PubMed]
- Järvinen, E.; Deng, F.; Kiander, W.; Sinokki, A.; Kidron, H.; Sjöstedt, N. The Role of Uptake and Efflux Transporters in the Disposition of Glucuronide and Sulfate Conjugates. Front. Pharmacol. 2021, 12, 802539. [Google Scholar] [CrossRef] [PubMed]
- Ho, R.H.; Tirona, R.G.; Leake, B.F.; Glaeser, H.; Lee, W.; Lemke, C.J.; Wang, Y.; Kim, R.B. Drug and Bile Acid Transporters in Rosuvastatin Hepatic Uptake: Function, Expression, and Pharmacogenetics. Gastroenterology 2006, 130, 1793–1806. [Google Scholar] [CrossRef]
- Garrison, D.A.; Talebi, Z.; Eisenmann, E.D.; Sparreboom, A.; Baker, S.D. Role of OATP1B1 and OATP1B3 in Drug-Drug Interactions Mediated by Tyrosine Kinase Inhibitors. Pharmaceutics 2020, 12, 856. [Google Scholar] [CrossRef]
- Telbisz, Á.; Ambrus, C.; Mózner, O.; Szabó, E.; Várady, G.; Bakos, É.; Sarkadi, B.; Özvegy-Laczka, C. Interactions of Potential Anti-COVID-19 Compounds with Multispecific ABC and OATP Drug Transporters. Pharmaceutics 2021, 13, 81. [Google Scholar] [CrossRef] [PubMed]
- Humeniuk, R.; Mathias, A.; Kirby, B.J.; Lutz, J.D.; Cao, H.; Osinusi, A.; Babusis, D.; Porter, D.; Wei, X.; Ling, J.; et al. Pharmacokinetic; Pharmacodynamic, and Drug-Interaction Profile of Remdesivir, a SARS-CoV-2 Replication Inhibitor. Clin. Pharmacokinet. 2021, 60, 569–583. [Google Scholar] [CrossRef] [PubMed]
- Zanger, U.M.; Schwab, M. Cytochrome P450 enzymes in drug metabolism: Regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol. Ther. 2013, 138, 103–141. [Google Scholar] [CrossRef] [PubMed]
- Lemaitre, F.; Solas, C.; Grégoire, M.; Lagarce, L.; Elens, L.; Polard, E.; Saint-Salvi, B.; Sommet, A.; Tod, M.; Guellec, C.B.-L. Potential drug-drug interactions associated with drugs currently proposed for COVID-19 treatment in patients receiving other treatments. Fundam. Clin. Pharmacol. 2020, 34, 530–547. [Google Scholar] [CrossRef] [PubMed]
- Atmar, R.L.; Finch, N. New Perspectives on Antimicrobial Agents: Molnupiravir and Nirmatrelvir/Ritonavir for Treatment of COVID-19. Antimicrob. Agents Chemother. 2022, 66, e0240421. [Google Scholar] [CrossRef]
- Mousquer, G.T.; Peres, A.; Fiegenbaum, M. Pathology of TB/COVID-19 Co-Infection: The phantom menace. Tuberculosis 2021, 126, 102020. [Google Scholar] [CrossRef] [PubMed]
- Foti, R.S. Utility of PBPK Modeling in Predicting and Characterizing Clinical Drug Interactions. Drug Metab. Dispos. 2024, 52, DMD-MR-2023-001384. [Google Scholar] [CrossRef] [PubMed]
- Owen, D.R.; Allerton, C.M.N.; Anderson, A.S.; Aschenbrenner, L.; Avery, M.; Berritt, S.; Boras, B.; Cardin, R.D.; Carlo, A.; Coffman, K.J.; et al. An oral SARS-CoV-2 M(pro) inhibitor clinical candidate for the treatment of COVID-19. Science 2021, 374, 1586–1593. [Google Scholar] [CrossRef] [PubMed]
- Chatterjee, B.; Thakur, S.S. Remdesivir and Its Combination With Repurposed Drugs as COVID-19 Therapeutics. Front. Immunol. 2022, 13, 830990. [Google Scholar] [CrossRef] [PubMed]
- Thomas, L.; Birangal, S.R.; Ray, R.; Miraj, S.S.; Munisamy, M.; Varma, M.; Sanju S.V., C.; Banerjee, M.; Shenoy, G.G.; Rao, M. Prediction of potential drug interactions between repurposed COVID-19 and antitubercular drugs: An integrational approach of drug information software and computational techniques data. Ther. Adv. Drug Saf. 2021, 12, 20420986211041277. [Google Scholar] [CrossRef]
- Rowland, M.; Peck, C.; Tucker, G. Physiologically-based pharmacokinetics in drug development and regulatory science. Annu. Rev. Pharmacol. Toxicol. 2011, 51, 45–73. [Google Scholar] [CrossRef]
- Jones, H.; Rowland-Yeo, K. Basic Concepts in Physiologically Based Pharmacokinetic Modeling in Drug Discovery and Development. CPT Pharmacomet. Syst. Pharmacol. 2013, 2, 63. [Google Scholar] [CrossRef] [PubMed]
- Luzon, E.; Blake, K.; Cole, S.; Nordmark, A.; Versantvoort, C.; Berglund, E.G. Physiologically based pharmacokinetic modeling in regulatory decision-making at the European Medicines Agency. Clin. Pharmacol. Ther. 2017, 102, 98–105. [Google Scholar] [CrossRef]
- Jamei, M.; Dickinson, G.L.; Rostami-Hodjegan, A. A framework for assessing inter-individual variability in pharmacokinetics using virtual human populations and integrating general knowledge of physical chemistry, biology, anatomy, physiology and genetics: A tale of ‘bottom-up’ vs ‘top-down’ recognition of covariates. Drug Metab. Pharmacokinet. 2009, 24, 53–75. [Google Scholar] [PubMed]
- Grimstein, M.; Yang, Y.; Zhang, X.; Grillo, J.; Huang, S.M.; Zineh, I.; Wang, Y. Physiologically Based Pharmacokinetic Modeling in Regulatory Science: An Update From the U.S. Food and Drug Administration’s Office of Clinical Pharmacology. J. Pharm. Sci. 2019, 108, 21–25. [Google Scholar] [CrossRef]
- European Medicines Agency, Regulatory Guidelines on the Reporting of Physiologically Based Pharmacokinetic (Pbpk) Modeling Analysis, Physiologically Based Pharmacokinetic (PBPK) Modeling and Simulations; European Medicines Agency: Amsterdam, The Netherlands, 2021; pp. 457–467.
- Palleria, C.; Di Paolo, A.; Giofrè, C.; Caglioti, C.; Leuzzi, G.; Siniscalchi, A.; De Sarro, G.; Gallelli, L. Pharmacokinetic drug-drug interaction and their implication in clinical management. J. Res. Med. Sci. 2013, 18, 601–610. [Google Scholar]
- Li, C.; Chen, L.; Li, L.; Chen, W. Drug-drug interactions and dose management of BTK inhibitors when initiating nirmatrelvir/ritonavir (paxlovid) based on physiologically-based pharmacokinetic models. Eur. J. Pharm. Sci. 2023, 189, 106564. [Google Scholar] [CrossRef] [PubMed]
- Sagawa, K.; Lin, J.; Jaini, R.; Di, L. Physiologically-Based Pharmacokinetic Modeling of PAXLOVID™ with First-Order Absorption Kinetics. Pharm. Res. 2023, 40, 1927–1938. [Google Scholar] [CrossRef]
- Wang, Z.; Chan, E.C.Y. Physiologically-Based Pharmacokinetic Modeling-Guided Dose Management of Oral Anticoagulants when Initiating Nirmatrelvir/Ritonavir (Paxlovid) for COVID-19 Treatment. Clin. Pharmacol. Ther. 2022, 112, 803–807. [Google Scholar] [CrossRef]
- Deb, S.; Reeves, A.A. Simulation of Remdesivir Pharmacokinetics and Its Drug Interactions. J. Pharm. Pharm. Sci. 2021, 24, 277–291. [Google Scholar] [CrossRef]
- Jamei, M. Where Do PBPK Models Stand in Pharmacometrics and Systems Pharmacology? CPT Pharmacomet. Syst. Pharmacol. 2020, 9, 75–76. [Google Scholar] [CrossRef] [PubMed]
- Min, J.S.; Bae, S.K. Prediction of drug-drug interaction potential using physiologically based pharmacokinetic modeling. Arch. Pharm. Res. 2017, 40, 1356–1379. [Google Scholar] [CrossRef] [PubMed]
- Lin, W.; Chen, Y.; Unadkat, J.D.; Zhang, X.; Wu, D.; Heimbach, T. Applications, Challenges, and Outlook for PBPK Modeling and Simulation: A Regulatory, Industrial and Academic Perspective. Pharm. Res. 2022, 39, 1701–1731. [Google Scholar] [CrossRef] [PubMed]
- Mitra, A.; Parrott, N.; Miller, N.; Lloyd, R.; Tistaert, C.; Heimbach, T.; Ji, Y.; Kesisoglou, F. Prediction of pH-Dependent Drug-Drug Interactions for Basic Drugs Using Physiologically Based Biopharmaceutics Modeling: Industry Case Studies. J. Pharm. Sci. 2020, 109, 1380–1394. [Google Scholar] [CrossRef] [PubMed]
- Salem, F.; Nimavardi, A.; Mudunuru, J.; Tompson, D.; Bloomer, J.; Turner, D.B.; Taskar, K.S. Physiologically based pharmacokinetic modeling for development and applications of a virtual celiac disease population using felodipine as a model drug. CPT Pharmacomet. Syst. Pharmacol. 2023, 12, 808–820. [Google Scholar] [CrossRef] [PubMed]
- Gill, J.; Moullet, M.; Martinsson, A.; Miljković, F.; Williamson, B.; Arends, R.H.; Reddy, V.P. Evaluating the performance of machine-learning regression models for pharmacokinetic drug-drug interactions. CPT Pharmacomet. Syst. Pharmacol. 2023, 12, 122–134. [Google Scholar] [CrossRef]
- Taskar, K.S.; Reddy, V.P.; Burt, H.; Posada, M.M.; Varma, M.; Zheng, M.; Ullah, M.; Riedmaier, A.E.; Umehara, K.I.; Snoeys, J.; et al. Physiologically-Based Pharmacokinetic Models for Evaluating Membrane Transporter Mediated Drug-Drug Interactions: Current Capabilities, Case Studies, Future Opportunities, and Recommendations. Clin. Pharmacol. Ther. 2020, 107, 1082–1115. [Google Scholar] [CrossRef]
TB-Drugs (Perpetrator) | COVID Drug (Victim) | Possible Pathway | Possible Mechanism |
---|---|---|---|
Rifampin | Remdesivir | Transporter/enzyme interplay | Inhibition of CYP3A4, P-gp and OATP transporters or induction of metabolic enzymes and P-gp transporters |
Rifabutin | Transporter/enzyme interplay | Inhibition of P-gp efflux pump and induction of CYP3A4 | |
Clofazimine | Transporter/enzyme interplay | Inhibition of CYP3A4 enzyme and P-gp efflux pump | |
Linezolid | Transporter/enzyme interplay | Inhibition of CYP3A4 enzyme and OATP uptake transporters | |
Clarithromycin | Transporter mediated | Inhibition of P-gp efflux pump | |
Azithromycin | Transporter mediated |
COVID Drug (Perpetrator) | Anti-TB Drugs (Victim) | Possible Pathway | Possible Mechanism |
---|---|---|---|
Remdesivir | Rifampin | Transporter/enzyme interplay | Inhibition of CYP3A4 enzyme and OATP uptake transporters |
PAS | Transporter mediated | Inhibition of OATP1B1 uptake transporter | |
Clofazimine | Enzyme mediated | Inhibition of CYP1A2 enzyme | |
Clarithromycin | Enzyme mediated | Inhibition of CYP3A4 enzyme | |
Roxithromycin | Enzyme mediated | Inhibition of CYP3A4 enzyme | |
Azithromycin | Enzyme mediated | Inhibition of OATP uptake transporters | |
Bedaquiline | Enzyme mediated | Inhibition of CYP2C8 and CYP3A4 enzyme |
COVID Drug (Perpetrator) | Anti-TB Drugs (Victim) | Possible Pathway | Possible Mechanism |
---|---|---|---|
Paxlovid | Rifampin | Transporter/enzyme interplay | Inhibition of CYP3A4 enzyme and P-gp efflux transporters |
Rifabutin | Transporter/enzyme interplay | Inhibition of CYP3A4 enzyme and P-gp efflux transporters | |
Rifapentine | Transporter/enzyme interplay | Inhibition of CYP3A4 enzyme and P-gp efflux transporters | |
Clarithromycin | Enzyme mediated | Inhibition of CYP3A4 enzyme | |
Roxithromycin | Enzyme mediated | Inhibition of CYP3A4 enzyme | |
Clofazimine | Transporter mediated | Inhibition of P-gp uptake transporters | |
Bedaquiline | Enzyme mediated | Inhibition of CYP3A4 enzyme |
COVID Drug (Victim) | Anti-TB Drugs (Perpetrator) | Possible Pathway | Possible Mechanism |
---|---|---|---|
Paxlovid | Rifampin | Transporter/enzyme interplay | Inhibition/induction of CYP3A4 enzyme and P-gp efflux transporters |
Prothionamide | Enzyme mediated | Inhibition of CYP3A4 enzyme | |
Clofazimine | Transporter/enzyme interplay | Inhibition of CYP3A4, CYP2D6 enzyme and P-gp efflux transporters | |
Clarithromycin | Enzyme mediated | Inhibition of CYP3A4 enzyme and P-gp efflux transporters | |
Azithromycin | Transporter mediated | Inhibition of P-gp efflux transporters | |
Linezolid | Enzyme mediated | Inhibition of CYP3A4 enzyme |
Parameters | Description | Data Source/Experimentation | Significance |
---|---|---|---|
Administration Route | |||
Intravenous, oral, dermal, etc., and associated parameters | Route and method of drug administration | Clinical trials | Determines the initial distribution and absorption characteristics of a drug |
Formulation | |||
Dosage form | Physical form of the drug product | Pharmaceutical development studies | Influences drug release and absorption characteristics |
Chemical Properties | |||
Molecular weight (MW) | Mass of a molecule | Analytical chemistry | Influences drug absorption, distribution, and elimination |
Solubility/dissolution | Ability to dissolve in a solvent | Solubility assays, shake-flask method | Determines how well the drug dissolves in bodily fluids |
Lipophilicity (LogP) | Measure of a compound’s affinity for lipid vs. water phases | LogP determination, HPLC, octanol-water partitioning | Affects membrane permeability and tissue distribution |
pKa | Acid dissociation constant | pKa determination assays, potentiometric titration | Impacts ionization state, influencing absorption and distribution |
Absorption Parameters | |||
Fraction unbound in plasma (fu,p) | Proportion of drug remains unbound in plasma | In vitro plasma binding studies or in silico modeling | Determines the PD and PK effects |
Fraction unbound in gut (fu,gut) | Proportion of drug remains unbound in the gut | In vitro plasma binding studies or in silico modeling | Determines how much of the orally administered dose is available for absorption. |
Absorption rate constants (Ka) | Rate at which a drug enters systemic circulation | In vitro absorption studies (e.g., Caco-2 cells) | Determines the rate of drug absorption |
Permeability coefficients (Papp) | Measure of the drug’s ability to cross biological membranes | In vitro assays (e.g., Caco-2 cells, PAMPA) | Indicates the ability of the drug to cross biological membranes |
Distribution Parameters | |||
Blood to Plasma ratio (B:P) | Ratio of concentrations of a compound in blood and plasma | Whole blood, equilibrium dialysis | Indicates how a drug distributes between plasma and blood |
Tissue-to-plasma concentration ratios (Kp) | Ratio of drug concentration in tissue to plasma | Imaging studies, literature data | Helps predict drug levels in different tissues |
Metabolism Parameters | |||
Enzyme kinetic parameters (Vmax, Km) | Maximum rate of reaction and Michaelis constant | In vitro enzyme assays (e.g., microsomes, hepatocytes, recombinant enzyme) | Determines the rate of drug metabolism |
Metabolic clearance rates | Rate at which a drug is metabolized | In vitro metabolism studies | Affects overall drug elimination from the body |
Metabolism (Phase 1) | |||
Oxidation, reduction, hydrolysis (Vmax, Km) | Phase 1 metabolic reactions | In vitro enzyme assays, liver microsomes | Modifies drug structure to enhance excretion |
Metabolism (Phase 2) | |||
Conjugation reactions (Vmax, Km) | Phase 2 metabolic reactions (e.g., glucuronidation, sulfation) | In vitro enzyme assays, liver microsomes | Increases drug solubility for easier excretion |
Elimination Parameters | |||
Renal clearance (CLr) | Rate at which a drug is removed via the kidneys | Urine analysis | Determines the rate of drug removal via the kidneys |
Hepatic clearance (CLhep) | Rate at which a drug is removed via the liver | Bile analysis | Determines the rate of drug removal via the liver |
Clearance pathways | Routes of drug excretion (e.g., renal, biliary) | In vivo studies, literature data | Identifies routes of drug excretion |
Transporters | |||
Membrane transporters (Vmax, Km) | Transport proteins that move drugs across cell membranes (e.g., P-gp, OATPs) | In vitro assays (e.g., cell lines expressing transporters) | Influence drug absorption, distribution, and elimination |
DDI Model Parameters | |||
Inhibition constants (Ki or IC50) | Measure of a drug’s potency to inhibit enzymes or transporters | In vitro inhibition studies | Measures the potency of a drug to inhibit enzymes or transporters |
Induction parameters (Emax, EC50) | Maximum effect and concentration for half-maximal effect | In vitro induction studies, clinical studies | Measures the capacity of a drug to induce enzyme or transporter expression |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Jony, M.R.; Ahn, S. Drug-Drug Interactions between COVID-19 and Tuberculosis Medications: A Comprehensive Review of CYP450 and Transporter-Mediated Effects. Pharmaceuticals 2024, 17, 1035. https://doi.org/10.3390/ph17081035
Jony MR, Ahn S. Drug-Drug Interactions between COVID-19 and Tuberculosis Medications: A Comprehensive Review of CYP450 and Transporter-Mediated Effects. Pharmaceuticals. 2024; 17(8):1035. https://doi.org/10.3390/ph17081035
Chicago/Turabian StyleJony, M. Rasheduzzaman, and Sangzin Ahn. 2024. "Drug-Drug Interactions between COVID-19 and Tuberculosis Medications: A Comprehensive Review of CYP450 and Transporter-Mediated Effects" Pharmaceuticals 17, no. 8: 1035. https://doi.org/10.3390/ph17081035