Á. F. Kiss, D. Vaskó, M. T. Déri, K. Tóth, and K. Monostory, Combination of CYP2C19 genotype with non-genetic factors evoking phenoconversion improves phenotype prediction, Pharmacol Rep, vol.70, issue.3, pp.525-557, 2018.

F. Stanke-labesque, E. Gautier-veyret, S. Chhun, and R. Guilhaumou, Inflammation is a major regulator of drug metabolizing enzymes and transporters: Consequences for the personalization of drug treatment, Pharmacol Ther, vol.215, p.107627, 2020.

T. Hirano and M. Murakami, COVID-19: A New Virus, but a Familiar Receptor and Cytokine Release Syndrome, Immunity, 2020.

O. Huet, G. Choukroun, and J. Mira, Récepteurs de type Toll, réponse inflammatoire et sepsis. Réanimation, vol.13, pp.167-75, 2004.

B. Beutler, Innate immunity: an overview, Mol Immunol, vol.40, issue.12, pp.845-59, 2004.

S. D. Wright, R. A. Ramos, P. S. Tobias, R. J. Ulevitch, and J. C. Mathison, CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science, vol.249, pp.1431-1434, 1990.

J. M. Cavaillon, Cytokines and macrophages, Biomed Pharmacother, vol.48, issue.10, pp.445-53, 1994.

D. L. Laskin and K. J. Pendino, Macrophages and Inflammatory Mediators in Tissue Injury, Annu Rev Pharmacol Toxicol, vol.35, issue.1, pp.655-77, 1995.

A. Koj, The role of interleukin-6 as the hepatocyte stimulating factor in the network of inflammatory cytokines, Ann N Y Acad Sci, vol.557, pp.1-8, 1989.

I. Cinel and S. M. Opal, Molecular biology of inflammation and sepsis: a primer, Crit Care Med, vol.37, issue.1, pp.291-304, 2009.

, Cytokines in context. J Cell Biol, vol.113, issue.5, pp.981-987, 1991.

F. R. Balkwill and F. Burke, The cytokine network, Immunol Today, vol.10, issue.9, pp.299-304, 1989.

H. Baumann and J. Gauldie, The acute phase response, Immunol Today, vol.15, issue.2, pp.74-80, 1994.

P. C. Heinrich, J. V. Castell, and T. Andus, Interleukin-6 and the acute phase response, Biochem J, vol.265, issue.3, pp.621-657, 1990.

A. Koj, Initiation of acute phase response and synthesis of cytokines, Biochim Biophys Acta BBA -Mol Basis Dis, vol.1317, issue.2, pp.84-94, 1996.

R. Eulenfeld, A. Dittrich, C. Khouri, P. J. Müller, B. Mütze et al., Interleukin-6 signalling: more than Jaks and STATs, Eur J Cell Biol, vol.91, issue.6-7, pp.486-95, 2012.

C. Garlanda, C. A. Dinarello, and A. Mantovani, The interleukin-1 family: back to the future. Immunity, vol.39, pp.1003-1021, 2013.

M. Bauer, A. T. Press, and M. Trauner, The liver in sepsis: patterns of response and injury, Curr Opin Crit Care, vol.19, issue.2, pp.123-130, 2013.

C. Gabay and I. Kushner, Acute-phase proteins and other systemic responses to inflammation, N Engl J Med, vol.340, issue.6, pp.448-54, 1999.

N. R. Sproston and J. J. Ashworth, Rôle de la protéine C-réactive sur les sites d'inflammation et d'infection. Immunologie, vol.754, 2018.

N. Moriya, H. Kataoka, H. Fujino, J. Nishikawa, and F. Kugawa, Effect of lipopolysaccharide on the xenobiotic-induced expression and activity of hepatic cytochrome P450 in mice, Biol Pharm Bull, vol.35, issue.4, pp.473-80, 2012.

A. E. Aitken, T. A. Richardson, and E. T. Morgan, Regulation of drug-metabolizing enzymes and transporters in inflammation, Annu Rev Pharmacol Toxicol, vol.46, pp.123-172, 2006.

M. R. Chaluvadi, R. D. Kinloch, B. A. Nyagode, T. A. Richardson, M. J. Raynor et al., Regulation of Hepatic Cytochrome P450 Expression in Mice with Intestinal or Systemic Infections of Citrobacter rodentium, Drug Metab Dispos, vol.37, issue.2, pp.366-74, 2009.

S. M. Mimche, B. A. Nyagode, M. D. Merrell, C. Lee, N. S. Prasanphanich et al., Hepatic Cytochrome P450s, Phase II Enzymes and Nuclear Receptors Are Downregulated in a Th2 Environment during Schistosoma mansoni Infection, Drug Metab Dispos, vol.42, issue.1, pp.134-174, 2014.

S. M. Mimche, C. Lee, K. H. Liu, P. N. Mimche, R. D. Harvey et al., A non-lethal malarial infection results in reduced drug metabolizing enzyme expression and drug clearance in mice, Malar J, vol.18, issue.1, p.234, 2019.

A. De-oliveira, K. S. Poça, P. Totino, F. Paumgartten, P. N. Mimche et al., Modulation of Cytochrome P450 2A5 Activity by Lipopolysaccharide: Low-Dose Effects and Non-Monotonic Dose-Response Relationship, PLoS ONE, vol.10, issue.1, p.2532, 2015.

T. A. Richardson, M. Sherman, L. Antonovic, S. S. Kardar, H. W. Strobel et al., Hepatic and Renal Cytochrome P450 Gene Regulation During Citrobacter rodentium Infection in Wildtype and Toll-like Receptor 4 Mutant Mice, Drug Metab Dispos Biol Fate Chem, vol.34, issue.3, pp.354-60, 2006.

R. Jover, R. Bort, M. J. Gómez-lechón, and J. V. Castell, Down-regulation of human CYP3A4 by the inflammatory signal interleukin-6: molecular mechanism and transcription factors involved, FASEB J Off Publ Fed Am Soc Exp Biol, vol.16, issue.13, pp.1799-801, 2002.

L. J. Dickmann, S. K. Patel, D. A. Rock, L. C. Wienkers, and J. G. Slatter, Effects of Interleukin-6 (IL-6) and an Anti-IL-6 Monoclonal Antibody on Drug-Metabolizing Enzymes in Human Hepatocyte Culture, Drug Metab Dispos, vol.39, issue.8, pp.1415-1437, 2011.

M. Klein, B. Kandel, K. Klein, M. Thomas, W. Thasler et al., Influence of inflammatory processes on ADME gene expression in human liver and hepatocytes. Poster presented at 17th North American Regional ISSX Meeting, 2011.

B. A. Nyagode, R. Jahangardi, M. D. Merrell, M. G. Tansey, E. T. Morgan et al., Selective effects of a therapeutic protein targeting tumor necrosis factor-alpha on cytochrome P450 regulation during infectious colitis: implications for disease-dependent drug-drug interactions, Pharmacol Res Perspect, vol.2, issue.1, pp.312-333, 2011.

A. E. Aitken, C. Lee, and E. T. Morgan, Roles of Nitric Oxide in Inflammatory Down-Regulation of Human Cytochromes P450. Free Radic Biol Med, vol.44, pp.1161-1169, 2008.

Z. Abdel-razzak, P. Loyer, A. Fautrel, J. C. Gautier, L. Corcos et al., Cytokines downregulate expression of major cytochrome P-450 enzymes in adult human hepatocytes in primary culture, Mol Pharmacol, vol.44, issue.4, pp.707-722, 1993.

H. Ms, Effect of influenza immunization on CYP3A4 activity in vivo, J Clin Pharmacol, vol.43, issue.12, pp.1377-81, 2003.

A. E. Aitken and E. T. Morgan, Gene-Specific Effects of Inflammatory Cytokines on Cytochrome P4502C, 2B6 and 3A4 mRNA Levels in Human Hepatocytes, Drug Metab Dispos Biol Fate Chem, vol.35, issue.9, pp.1687-93, 2007.

B. A. Nyagode, C. Lee, and E. T. Morgan, Modulation of hepatic cytochrome P450s by Citrobacter rodentium infection in interleukin-6-and interferon-{gamma}-null mice, J Pharmacol Exp Ther, vol.335, issue.2, pp.480-488, 2010.

E. Siewert, R. Bort, R. Kluge, P. C. Heinrich, J. Castell et al., Hepatic cytochrome P450 downregulation during aseptic inflammation in the mouse is interleukin 6 dependent, Hepatol Baltim Md, vol.32, issue.1, pp.49-55, 2000.

T. Ashino, T. Oguro, S. Shioda, R. Horai, M. Asano et al., Involvement of interleukin-6 and tumor necrosis factor alpha in CYP3A11 and 2C29 down-regulation by Bacillus Calmette-Guerin and lipopolysaccharide in mouse liver, Drug Metab Dispos Biol Fate Chem, vol.32, issue.7, pp.707-721, 2004.

A. H. Tolson and H. Wang, Regulation of drug-metabolizing enzymes by xenobiotic receptors: PXR and CAR. Adv Drug Deliv Rev, vol.62, pp.1238-1287, 2010.

S. Teng and M. Piquette-miller, The involvement of the pregnane X receptor in hepatic gene regulation during inflammation in mice, J Pharmacol Exp Ther, vol.312, issue.2, pp.841-849, 2005.

P. Ruminy, C. Gangneux, S. Claeyssens, M. Scotte, M. Daveau et al., Gene transcription in hepatocytes during the acute phase of a systemic inflammation: from transcription factors to target genes, Inflamm Res, vol.50, issue.8, pp.383-90, 2001.

J. Yang, C. Hao, D. Yang, D. Shi, X. Song et al., Pregnane X receptor is required for interleukin-6 mediated down-regulation of cytochrome P450 3A4 in human hepatocytes, Toxicol Lett, vol.197, issue.3, pp.219-245, 2010.

A. P. Beigneux, A. H. Moser, J. K. Shigenaga, C. Grunfeld, and K. R. Feingold, Reduction in cytochrome P-450 enzyme expression is associated with repression of CAR (constitutive androstane receptor) and PXR (pregnane X receptor) in mouse liver during the acute phase response, Biochem Biophys Res Commun, vol.293, issue.1, pp.145-154, 2002.

T. A. Richardson and E. T. Morgan, Hepatic cytochrome P450 gene regulation during endotoxininduced inflammation in nuclear receptor knockout mice, J Pharmacol Exp Ther, vol.314, issue.2, pp.703-712, 2005.

K. , B. Co, and C. , Lipopolysaccharide and cecal ligation/puncture differentially affect the subcellular distribution of the pregnane X receptor but consistently cause suppression of its target genes CYP3A. Shock Augusta Ga, vol.19, pp.469-74, 2003.

Y. Tian, S. Ke, M. S. Denison, A. B. Rabson, and M. A. Gallo, Ah receptor and NF-kappaB interactions, a potential mechanism for dioxin toxicity, J Biol Chem, vol.274, issue.1, pp.510-515, 1999.

C. Zhou, M. M. Tabb, E. L. Nelson, F. Grün, S. Verma et al., Mutual repression between steroid and xenobiotic receptor and NF-kappaB signaling pathways links xenobiotic metabolism and inflammation, J Clin Invest, vol.116, issue.8, pp.2280-2289, 2006.

B. Zordoky and A. El-kadi, Role of NF-kappaB in the regulation of cytochrome P450 enzymes, Curr Drug Metab, vol.10, issue.2, pp.164-78, 2009.

R. Jover, M. Moya, and M. J. Gómez-lechón, Transcriptional regulation of cytochrome p450 genes by the nuclear receptor hepatocyte nuclear factor 4-alpha, Curr Drug Metab, vol.10, issue.5, pp.508-527, 2009.

R. E. Mcgehee, M. J. Ronis, and T. M. Badger, Regulation of the hepatic CYP 2E1 gene during chronic alcohol exposure: lack of an ethanol response element in the proximal 5'-flanking sequence, DNA Cell Biol, vol.16, issue.6, pp.725-761, 1997.

A. L. Roe, S. M. Poloyac, G. Howard, S. I. Shedlofsky, and R. A. Blouin, The effect of endotoxin on hepatocyte nuclear factor 1 nuclear protein binding: potential implications on CYP2E1 expression in the rat, J Pharm Pharmacol, vol.53, issue.10, pp.1365-71, 2001.

S. H. Park and D. J. Waxman, Inhibitory cross-talk between STAT5b and liver nuclear factor HNF3beta: impact on the regulation of growth hormone pulse-stimulated, male-specific liver cytochrome P-450 gene expression, J Biol Chem, vol.276, issue.46, pp.43031-43040, 2001.

P. Cheng, M. Wang, and E. Morgan, Rapid Transcriptional Suppression of Rat Cytochrome P450 Genes by Endotoxin Treatment and Its Inhibition by Curcumin, J Pharmacol Exp Ther, vol.307, pp.1205-1217, 2004.

M. Murray, P. H. Cui, and F. Zhou, Roles of mitogen-activated protein kinases in the regulation of CYP genes, Curr Drug Metab, vol.11, issue.10, pp.850-858, 2010.

C. Koike, R. Moore, and M. Negishi, Extracellular Signal-Regulated Kinase Is an Endogenous Signal Retaining the Nuclear Constitutive Active/Androstane Receptor (CAR) in the Cytoplasm of Mouse Primary Hepatocytes, Mol Pharmacol, vol.71, issue.5, pp.1217-1238, 2007.

R. Keller, M. Klein, M. Thomas, A. Dräger, U. Metzger et al., Coordinating Role of RXR? in Downregulating Hepatic Detoxification during Inflammation Revealed by Fuzzy-Logic Modeling, PLOS Comput Biol, vol.12, issue.1, p.1004431, 2016.

J. L. Staudinger and K. Lichti, Cell Signaling and Nuclear Receptors: New Opportunities for Molecular Pharmaceuticals in Liver Disease, Mol Pharm, vol.5, issue.1, pp.17-34, 2008.

M. Febvre-james, A. Bruyère, L. Vée, M. Fardel, and O. , The JAK1/2 Inhibitor Ruxolitinib Reverses Interleukin-6-Mediated Suppression of Drug-Detoxifying Proteins in Cultured Human Hepatocytes, Drug Metab Dispos Biol Fate Chem, vol.46, issue.2, pp.131-171, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01699218

T. J. Carlson and R. E. Billings, Role of nitric oxide in the cytokine-mediated regulation of cytochrome P-450, Mol Pharmacol, vol.49, issue.5, pp.796-801, 1996.

C. Lee, B. Kim, L. Li, and E. T. Morgan, Nitric oxide-dependent proteasomal degradation of cytochrome P450 2B proteins, J Biol Chem, vol.283, issue.2, pp.889-98, 2008.

C. Lee, J. Pohl, and E. T. Morgan, Dual mechanisms of CYP3A protein regulation by proinflammatory cytokine stimulation in primary hepatocyte cultures, Drug Metab Dispos Biol Fate Chem, vol.37, issue.4, pp.865-72, 2009.

M. B. Sewer, T. B. Barclay, and E. T. Morgan, Down-Regulation of Cytochrome P450 mRNAs and Proteins in Mice Lacking a Functional NOS2 Gene, Mol Pharmacol, vol.54, issue.2, pp.273-282, 1998.

D. Xu, W. W. Sun, M. Wei, L. Wang, and J. , Melatonin attenuates lipopolysaccharideinduced down-regulation of pregnane X receptor and its target gene CYP3A in mouse liver, J Pineal Res, vol.38, issue.1, pp.27-34, 2005.

D. Xu, W. W. Sun, M. Wu, C. Wang, J. Wei et al., Kupffer cells and reactive oxygen species partially mediate lipopolysaccharide-induced downregulation of nuclear receptor pregnane x receptor and its target gene CYP3a in mouse liver, Free Radic Biol Med, vol.37, issue.1, pp.10-22, 2004.

C. Lee, B. Lee, S. L. Arnold, N. Isoherranen, and E. T. Morgan, Nitric oxide and interleukin-1? stimulate the proteasome-independent degradation of the retinoic acid hydroxylase CYP2C22 in primary rat hepatocytes, J Pharmacol Exp Ther, vol.348, issue.1, pp.141-52, 2014.

C. Lee, S. Tripathi, and E. T. Morgan, Nitric oxide-regulated proteolysis of human CYP2B6 via the ubiquitin-proteasome system, Free Radic Biol Med, vol.108, pp.478-86, 2017.

H. Hara and T. Adachi, Contribution of hepatocyte nuclear factor-4 to down-regulation of CYP2D6 gene expression by nitric oxide, Mol Pharmacol, vol.61, issue.1, pp.194-200, 2002.

C. Vossen and M. Erard, Down-regulation of nuclear receptor DNA-binding activity by nitric oxide--HNF4 as a model system, Med Sci Monit Int Med J Exp Clin Res, vol.8, issue.10, pp.217-220, 2002.

S. Sabunciyan, B. Maher, S. Bahn, F. Dickerson, and R. H. Yolken, Association of DNA Methylation with Acute Mania and Inflammatory Markers, PloS One, vol.10, issue.7, p.132001, 2015.

A. Yu and Y. Pan, Noncoding microRNAs: small RNAs play a big role in regulation of ADME?, Acta Pharm Sin B, vol.2, issue.2, pp.93-101, 2012.

T. Yokoi and M. Nakajima, microRNAs as mediators of drug toxicity, Annu Rev Pharmacol Toxicol, vol.53, pp.377-400, 2013.

V. Lamba, Y. Ghodke, W. Guan, and T. S. Tracy, microRNA-34a is associated with expression of key hepatic transcription factors and cytochromes P450, Biochem Biophys Res Commun, vol.445, issue.2, pp.404-415, 2014.

J. K. Rieger, K. Klein, S. Winter, and U. M. Zanger, Expression variability of absorption, distribution, metabolism, excretion-related microRNAs in human liver: influence of nongenetic factors and association with gene expression, Drug Metab Dispos Biol Fate Chem, vol.41, issue.10, pp.1752-62, 2013.

J. K. Rieger, S. Reutter, U. Hofmann, M. Schwab, and U. M. Zanger, Inflammation-associated microRNA-130b down-regulates cytochrome P450 activities and directly targets CYP2C9, Drug Metab Dispos Biol Fate Chem, vol.43, issue.6, pp.884-892, 2015.

L. Zeng, Y. Chen, Y. Wang, L. Yu, B. Knox et al., MicroRNA hsa-miR-370-3p suppresses the expression and induction of CYP2D6 by facilitating mRNA degradation, Biochem Pharmacol, vol.140, pp.139-188, 2017.

E. G. Hrycay and S. M. Bandiera, Expression, function and regulation of mouse cytochrome P450 enzymes: comparison with human P450 enzymes, Curr Drug Metab, vol.10, issue.10, pp.1151-83, 2009.

T. Yasu, T. Konuma, S. Kato, Y. Kurokawa, S. Takahashi et al., Serum C-reactive protein levels affect the plasma voriconazole trough levels in allogeneic hematopoietic cell transplant recipients. Leuk Lymphoma, vol.58, pp.2731-2734, 2017.

E. Gautier-veyret, A. Truffot, S. Bailly, X. Fonrose, A. Thiebaut-bertrand et al., Inflammation is a potential risk factor of voriconazole overdose in hematological patients, Fundam Clin Pharmacol, vol.33, issue.2, pp.232-240, 2019.

E. Ventura, M. A. Van-wanrooy, M. Span, L. Rodgers, M. Van-den-heuvel et al., Longitudinal Analysis of the Effect of Inflammation on Voriconazole Trough Concentrations, Antimicrob Agents Chemother, vol.60, issue.5, pp.2727-2758, 2016.

M. Van-wanrooy, L. Span, M. Rodgers, E. R. Van-den-heuvel, D. Uges et al., Inflammation Is Associated with Voriconazole Trough Concentrations, Antimicrob Agents Chemother, vol.58, issue.12, pp.7098-101, 2014.

E. Ventura, M. A. Span, L. Van-den-heuvel, E. R. Groothuis, G. Alffenaar et al., Influence of inflammation on voriconazole metabolism, Antimicrob Agents Chemother, vol.59, issue.5, pp.2942-2945, 2015.

A. Veringa, M. Ter-avest, L. Span, E. R. Van-den-heuvel, D. J. Touw et al., Voriconazole metabolism is influenced by severe inflammation: a prospective study, J Antimicrob Chemother, vol.72, issue.1, pp.261-268, 2017.

T. Naito, T. Yamada, Y. Mino, and J. Kawakami, Impact of inflammation and concomitant glucocorticoid administration on plasma concentration of triazole antifungals in immunocompromised patients, Clin Chim Acta, vol.441, pp.127-159, 2015.

T. Niioka, N. Fujishima, M. Abumiya, T. Yamashita, K. Ubukawa et al., Relationship Between the CYP2C19 Phenotype Using the Voriconazole-to-Voriconazole N-Oxide Plasma Concentration Ratio and Demographic and Clinical Characteristics of Japanese Patients With Different CYP2C19 Genotypes, Ther Drug Monit, vol.39, issue.5, pp.514-535, 2017.

B. Vreugdenhil, W. Van-der-velden, T. Feuth, M. Kox, P. Pickkers et al., Moderate correlation between systemic IL-6 responses and CRP with trough concentrations of voriconazole, Br J Clin Pharmacol, vol.84, issue.9, pp.1980-1988, 2018.

M. Mafuru, S. Wu, S. He, X. Lu, J. Huang et al., The Influence of Proinflammatory Cytokines on Voriconazole Trough Concentration in Patients With Different Forms of Hematologic Disorders, J Clin Pharmacol, vol.59, issue.10, pp.1340-50, 2019.

A. Märtson, A. Veringa, M. Bakker, E. R. Van-den-heuvel, D. J. Touw et al., Posaconazole trough concentrations are not influenced by inflammation: A prospective study, Int J Antimicrob Agents, vol.53, issue.3, pp.325-334, 2019.

J. De-leon and F. J. Diaz, Serious respiratory infections can increase clozapine levels and contribute to side effects: a case report, Prog Neuropsychopharmacol Biol Psychiatry, vol.27, issue.6, pp.1059-63, 2003.

M. J. Haack, M. Bak, R. Beurskens, M. Maes, L. Stolk et al., Toxic rise of clozapine plasma concentrations in relation to inflammation, Eur Neuropsychopharmacol J Eur Coll Neuropsychopharmacol, vol.13, issue.5, pp.381-386, 2003.

J. Jecel, T. M. Michel, L. Gutknecht, D. Schmidt, B. Pfuhlmann et al., Toxic clozapine serum levels during acute urinary tract infection: a case report, Eur J Clin Pharmacol, vol.60, issue.12, pp.909-919, 2005.

C. Ruan, X. Zhang, W. Guo, W. Li, H. Zhuang et al., Two cases of high serum clozapine concentrations occurring during inflammation in Chinese patients, Int J Psychiatry Med, vol.53, issue.4, pp.292-305, 2018.

B. Pfuhlmann, C. Hiemke, S. Unterecker, R. Burger, A. Schmidtke et al., Toxic clozapine serum levels during inflammatory reactions, J Clin Psychopharmacol, vol.29, issue.4, pp.392-396, 2009.

G. Hefner, M. Shams, S. Unterecker, T. Falter, and C. Hiemke, Inflammation and psychotropic drugs: the relationship between C-reactive protein and antipsychotic drug levels, Psychopharmacology (Berl), vol.233, issue.9, pp.1695-705, 2016.

K. A. Espnes, K. O. Heimdal, and O. Spigset, A puzzling case of increased serum clozapine levels in a patient with inflammation and infection, Ther Drug Monit, vol.34, issue.5, pp.489-92, 2012.

A. Helland, S. Habib, L. Ulvestad, and O. Spigset, Systemic Inflammation Complicates the Interpretation of Therapeutic Drug Monitoring of Risperidone, J Clin Psychopharmacol, vol.38, issue.3, pp.263-268, 2018.

Y. Yamamoto, Y. Takahashi, A. Horino, N. Usui, T. Nishida et al., Influence of Inflammation on the Pharmacokinetics of Perampanel, Ther Drug Monit, vol.40, issue.6, pp.725-734, 2018.

N. J. Vet, J. M. Brussee, M. De-hoog, M. G. Mooij, C. Verlaat et al., Inflammation and Organ Failure Severely Affect Midazolam Clearance in Critically Ill Children, Am J Respir Crit Care Med, vol.194, issue.1, pp.58-66, 2016.

L. G. Franken, B. De-winter, A. D. Masman, M. Van-dijk, F. Baar et al., Population pharmacodynamic modelling of midazolam induced sedation in terminally ill adult patients, Br J Clin Pharmacol, vol.84, issue.2, pp.320-350, 2018.

E. L. Swart, K. P. Zuideveld, D. Jongh, J. Danhof, M. Thijs et al., Comparative population pharmacokinetics of lorazepam and midazolam during long-term continuous infusion in critically ill patients, Br J Clin Pharmacol, vol.57, issue.2, pp.135-180, 2004.

M. A. Sikma, E. M. Van-maarseveen, E. A. Van-de-graaf, J. H. Kirkels, M. C. Verhaar et al., Pharmacokinetics and Toxicity of Tacrolimus Early After Heart and Lung Transplantation

, Am J Transplant Off J Am Soc Transplant Am Soc Transpl Surg, vol.15, issue.9, pp.2301-2314, 2015.

A. Nakamura, N. Amada, I. Haga, K. Tokodai, and T. Kashiwadate, Effects of elevated tacrolimus trough levels in association with infectious enteritis on graft function in renal transplant recipients, Transplant Proc, vol.46, issue.2, pp.592-596, 2014.

K. Sato, A. N. Sato, T. Miura, S. Ohashi, Y. Sekiguchi et al., Severe elevations of FK506 blood concentration due to diarrhea in renal transplant recipients, Clin Transplant, vol.18, issue.5, pp.585-90, 2004.

W. Lemahieu, B. Maes, K. Verbeke, P. Rutgeerts, K. Geboes et al., Cytochrome P450 3A4 and P-glycoprotein activity and assimilation of tacrolimus in transplant patients with persistent diarrhea, Am J Transplant Off J Am Soc Transplant Am Soc Transpl Surg, vol.5, issue.6, pp.1383-91, 2005.

S. Maezono, K. Sugimoto, K. Sakamoto, M. Ohmori, S. Hishikawa et al., Elevated blood concentrations of calcineurin inhibitors during diarrheal episode in pediatric liver transplant recipients: involvement of the suppression of intestinal cytochrome P450 3A and P-glycoprotein, Pediatr Transplant, vol.9, issue.3, pp.315-338, 2005.

E. Bonneville, E. Gautier-veyret, C. Ihl, M. Hilleret, M. Baudrant et al., Unexpected overdose blood concentration of tacrolimus: Keep in mind the role of inflammation, Br J Clin Pharmacol

,

M. A. Sikma, C. C. Hunault, E. M. Van-maarseveen, A. Huitema, E. A. Van-de-graaf et al., High Variability of Whole-Blood Tacrolimus Pharmacokinetics Early After Thoracic Organ Transplantation, Eur J Drug Metab Pharmacokinet, vol.45, issue.1, pp.123-157, 2020.

M. Rayar, C. Tron, C. Jézéquel, J. M. Beaurepaire, A. Petitcollin et al., High Intrapatient Variability of Tacrolimus Exposure in the Early Period After Liver Transplantation Is Associated With Poorer Outcomes, Transplantation, vol.102, issue.3, pp.108-122, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02454671

Y. L. Chen, L. Vraux, V. Leneveu, A. Dreyfus, F. Stheneur et al., Acute-phase response, interleukin-6, and alteration of cyclosporine pharmacokinetics, Clin Pharmacol Ther, vol.55, issue.6, pp.649-60, 1994.

T. Mizuno, M. M. O'brien, and A. A. Vinks, Significant effect of infection and food intake on sirolimus pharmacokinetics and exposure in pediatric patients with acute lymphoblastic leukemia, Eur J Pharm Sci, vol.128, pp.209-223, 2019.

K. C. Chang, T. D. Bell, B. A. Lauer, and H. Chai, Altered theophylline pharmacokinetics during acute respiratory viral illness, Lancet Lond Engl, vol.1, issue.8074, pp.1132-1135, 1978.

M. J. Kraemer, C. T. Furukawa, J. R. Koup, G. G. Shapiro, W. E. Pierson et al., Altered Theophylline Clearance During an Influenza B Outbreak, Pediatrics, vol.69, issue.4, pp.476-80, 1982.

W. Stephenj, B. Juditha, and F. Geoffreyc, INHIBITION OF THEOPHYLLINE METABOLISM BY INTERFERON. The Lancet, vol.330, pp.939-980, 1987.

E. T. Morgan, J. L. Dempsey, S. M. Mimche, T. J. Lamb, S. Kulkarni et al., Physiological Regulation of Drug Metabolism and Transport: Pregnancy, Microbiome, Inflammation, Infection, and Fasting, Drug Metab Dispos, vol.46, issue.5, pp.503-516, 2018.

L. Elens, L. J. Langman, D. A. Hesselink, S. Bergan, D. Moes et al., Pharmacologic Treatment of Transplant Recipients Infected With SARS-CoV-2: Considerations Regarding Therapeutic Drug Monitoring and Drug-Drug Interactions, Ther Drug Monit
URL : https://hal.archives-ouvertes.fr/hal-02563361

Q. Ye, B. Wang, and J. Mao, The pathogenesis and treatment of the `Cytokine Storm' in COVID-19, J Infect, vol.80, issue.6, pp.607-620, 2020.

C. Huang, Y. Wang, X. Li, L. Ren, J. Zhao et al., Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. The Lancet, vol.395, pp.497-506, 2020.

P. Conti, G. Ronconi, A. Caraffa, C. Gallenga, R. Ross et al., Induction of proinflammatory cytokines (IL-1 and IL-6) and lung inflammation by Coronavirus-19 (COVI-19 or SARS-CoV-2): anti-inflammatory strategies, J Biol Regul Homeost Agents, vol.34, issue.2, 202014.

B. Cao, Y. Wang, D. Wen, W. Liu, J. Wang et al., A Trial of Lopinavir-Ritonavir in Adults Hospitalized with Severe Covid-19, N Engl J Med, vol.382, issue.19, pp.1787-99, 2020.

C. Schoergenhofer, B. Jilma, T. Stimpfl, M. Karolyi, and A. Zoufaly, Pharmacokinetics of Lopinavir and Ritonavir in Patients Hospitalized With Coronavirus Disease 2019 (COVID-19), Ann Intern Med, 2020.

M. P. Lê, P. Jaquet, J. Patrier, P. Wicky, L. Hingrat et al., Pharmacokinetics of lopinavir/ritonavir oral solution to treat COVID-19 in mechanically ventilated ICU patients, J Antimicrob Chemother, 2020.

I. Ofotokun, J. L. Lennox, M. E. Eaton, J. C. Ritchie, K. A. Easley et al., Immune Activation Mediated Change in Alpha-1-Acid Glycoprotein: Impact on Total and Free Lopinavir Plasma Exposure, J Clin Pharmacol, vol.51, issue.11, pp.1539-1587, 2011.

K. Choy, A. Wong, P. Kaewpreedee, S. F. Sia, D. Chen et al., Remdesivir, lopinavir, emetine, and homoharringtonine inhibit SARS-CoV-2 replication in vitro, Antiviral Res, vol.178, p.104786, 2020.

S. Meziyerh, T. C. Zwart, R. W. Van-etten, J. A. Janson, T. Van-gelder et al., Severe COVID-19 in a renal transplant recipient: A focus on pharmacokinetics, Am J Transplant, 2020.

S. Testa, P. Prandoni, O. Paoletti, R. Morandini, M. Tala et al., Direct oral anticoagulant plasma levels' striking increase in severe COVID-19 respiratory syndrome patients treated with antiviral agents: The Cremona experience, J Thromb Haemost, 2020.

J. De-leon, C. Ruan, H. Verdoux, and C. Wang, Clozapine is strongly associated with the risk of pneumonia and inflammation, Gen Psychiatry, vol.33, issue.2, 2020.
URL : https://hal.archives-ouvertes.fr/inserm-02632133

T. Cranshaw and T. Harikumar, COVID-19 Infection May Cause Clozapine Intoxication: Case Report and Discussion, 2020.

R. Kajani, A. Apramian, A. Vega, N. Ubhayakar, P. Xu et al., Neuroleptic malignant syndrome in a COVID-19 patient, Brain Behav Immun, vol.88, pp.28-37, 2020.