Prioritization of Anti-SARS-Cov-2 Drug Repurposing Opportunities Based on Plasma and Target Site Concentrations Derived from their Established Human Pharmacokinetics

Usman Arshad, Henry Pertinez, Helen Box, Lee Tatham, Rajith K R Rajoli, Paul Curley, Megan Neary, Joanne Sharp, Neill J Liptrott, Anthony Valentijn, Christopher David, Steve P Rannard, Paul M O'Neill, Ghaith Aljayyoussi, Shaun H Pennington, Stephen A Ward, Andrew Hill, David J Back, Saye H Khoo, Patrick G Bray, Giancarlo A Biagini, Andrew Owen, Usman Arshad, Henry Pertinez, Helen Box, Lee Tatham, Rajith K R Rajoli, Paul Curley, Megan Neary, Joanne Sharp, Neill J Liptrott, Anthony Valentijn, Christopher David, Steve P Rannard, Paul M O'Neill, Ghaith Aljayyoussi, Shaun H Pennington, Stephen A Ward, Andrew Hill, David J Back, Saye H Khoo, Patrick G Bray, Giancarlo A Biagini, Andrew Owen

Abstract

There is a rapidly expanding literature on the in vitro antiviral activity of drugs that may be repurposed for therapy or chemoprophylaxis against severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2). However, this has not been accompanied by a comprehensive evaluation of the target plasma and lung concentrations of these drugs following approved dosing in humans. Accordingly, concentration 90% (EC90 ) values recalculated from in vitro anti-SARS-CoV-2 activity data was expressed as a ratio to the achievable maximum plasma concentration (Cmax ) at an approved dose in humans (Cmax /EC90 ratio). Only 14 of the 56 analyzed drugs achieved a Cmax /EC90 ratio above 1. A more in-depth assessment demonstrated that only nitazoxanide, nelfinavir, tipranavir (ritonavir-boosted), and sulfadoxine achieved plasma concentrations above their reported anti-SARS-CoV-2 activity across their entire approved dosing interval. An unbound lung to plasma tissue partition coefficient (Kp Ulung ) was also simulated to derive a lung Cmax /half-maximal effective concentration (EC50 ) as a better indicator of potential human efficacy. Hydroxychloroquine, chloroquine, mefloquine, atazanavir (ritonavir-boosted), tipranavir (ritonavir-boosted), ivermectin, azithromycin, and lopinavir (ritonavir-boosted) were all predicted to achieve lung concentrations over 10-fold higher than their reported EC50 . Nitazoxanide and sulfadoxine also exceeded their reported EC50 by 7.8-fold and 1.5-fold in lung, respectively. This analysis may be used to select potential candidates for further clinical testing, while deprioritizing compounds unlikely to attain target concentrations for antiviral activity. Future studies should focus on EC90 values and discuss findings in the context of achievable exposures in humans, especially within target compartments, such as the lungs, in order to maximize the potential for success of proposed human clinical trials.

Conflict of interest statement

D.J.B. has received honoraria or advisory board payments from AbbVie, Gilead, ViiV, Merck, Janssen, and educational grants from AbbVie, Gilead, ViiV, Merck, Janssen, and Novartis. A.O. and S.P.R. are Directors of Tandem Nano Ltd. A.O. has received research funding from ViiV, Merck, Janssen, and consultancy from Gilead, ViiV and Merck not related to the current paper. P.O.N. is currently engaged in a collaboration with Romark LLC but this interaction did not influence the prioritization or conclusions in the current paper. All other authors declared no competing interests for this work.

© 2020 The Authors. Clinical Pharmacology & Therapeutics published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
Assessment of the variation in reported half‐maximal effective concentration (EC50) values for severe acute respiratory syndrome‐coronavirus 2 across the drugs for which more than one value was available in the literature (a). The consequences of this variability in reported EC50 in terms of the peak plasma concentration (Cmax)/EC50 ratio is also provided (b). Amodiaquine and toremifene were estimated to exhibit subtherapeutic pharmacokinetics irrespective of which EC50 value was used. Similarly, nelfinavir was estimated to have Cmax value higher than its EC50 irrespective of which EC50 was used in the analysis. For the other drugs, interpretation was highly dependent upon which reported EC50 was utilised and this underscores the caution that should be taken in interpreting the available data.
Figure 2
Figure 2
A bar chart displaying peak plasma concentration (Cmax)/effective concentration 90% (EC90) ratio for compounds studied for in vitro antiviral activity against severe acute respiratory syndrome‐coronavirus 2 for which data were available to recalculate an EC90. Drugs with a ratio below 1 were deemed not to provide plasma concentrations at their approved doses to exert sufficient systemic antiviral activity. Those drugs with a ratio above 1 (shown in orange) were deemed to have potential to provide plasma concentrations sufficient to exert at least some antiviral activity for at least some of their dosing interval at their approved dose. Drugs shown in green were predicted to exceed plasma concentrations over their EC90 by more than twofold.
Figure 3
Figure 3
Digitized pharmacokinetic (PK) interrogation of all drugs calculated to have a peak plasma concentration (Cmax)/half‐maximal effective concentration (EC50) ratio above 1. The lowest reported severe acute respiratory syndrome‐coronavirus 2 EC50 (dashed orange lines) and associated recalculated effective concentration 90% (EC90; dashed green lines) are also highlighted. References for the utilized data are nitazoxanide 500 mg b.i.d. and 1,000 mg b.i.d., tipranavir 500 mg b.i.d. with 200 mg ritonavir, sulfadoxine 1,500 mg with 75 mg pyrimethamine, nelfinavir 1,250 mg b.i.d., indomethacin 50 mg t.i.d., atazanavir 300 mg q.d. with 100 mg ritonavir, hydroxychloroquine 2,000 mg hydroxychloroquine sulfate/1,550 mg base administered over 3 days, eltrombopag 75 mg single dose, lopinavir 400 mg with 100 mg ritonavir, chloroquine 1,500 mg administered over 3 days, mefloquine 1,200 mg over 3 days, and anidulafungin 100 mg q.d. Robust PK data were unavailable for niclosamide 500 mg, ritonavir 600 mg, and merimepodib 300 mg in order to conduct this digitized interrogation of these molecules.
Figure 4
Figure 4
A bar chart displaying the simulated lung peak plasma concentration (Cmax)/half‐maximal effective concentration (EC50). Drugs with a ratio below 1 were deemed not to provide lung concentrations at their approved doses to exert sufficient pulmonary antiviral activity for treatment or prevention strategies. Those drugs with a ratio above 1 (shown in orange) were estimated to provide lung concentrations sufficient to exert at least some antiviral activity at their approved dose. Drugs shown in green were predicted to exceed lung concentrations over their EC50 by > 10‐fold.
Figure 5
Figure 5
A heatmap displaying the simulated tissue peak plasma concentration (Cmax)/half‐maximal effective concentration (EC50) values for all drugs with available data. Those drugs with a ratio above 1 (shown in orange) were estimated to provide tissue concentrations sufficient to exert at least some antiviral activity at their approved dose. Drugs shown in green were predicted to exceed tissue concentrations over their EC50 by > 10‐fold.

References

    1. Gu, J. , Han, B. & Wang, J. COVID‐19: Gastrointestinal manifestations and potential fecal‐oral transmission. Gastroenterology 158, 1518–1519 (2020).
    1. World Health Organisation . COVID‐19 Trials ‐ International Clinical Trials Registry Platform (ICTRP) <> (2020).
    1. Qin, C. et al. Dysregulation of immune response in patients with COVID‐19 in Wuhan, China. Clin. Infect. Dis. 10.1093/cid/ciaa248.
    1. Li, W. et al. Angiotensin‐converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature 426, 450–454 (2003).
    1. Zhao, Y. , Zhao, Z. , Wang, Y. , Zhou, Y. , Ma, Y. & Zuo, W. Single‐cell RNA expression profiling of ACE2, the receptor of SARS‐CoV‐2. J bioRxiv. 10.1101/2020.01.26.919985.
    1. Wu, Y. et al. Prolonged presence of SARS‐CoV‐2 viral RNA in faecal samples. Lancet Gastroenterol. Hepatol. 10.1016/S2468-1253(20)30083-2.
    1. Diao, B.H. et al. Kidney is a target for novel severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) Infection. J medRxiv. 10.1101/2020.03.04.20031120
    1. Zhang, C. , Shi, L. & Wang, F.S. Liver injury in COVID‐19: management and challenges. Lancet Gastroenterol. Hepatol. 5, 428–430 (2020).
    1. Guo, Y.R. et al. The origin, transmission and clinical therapies on coronavirus disease 2019 (COVID‐19) outbreak ‐ an update on the status. Mil Med Res 7, 11 (2020).
    1. Hoffmann, M. et al. SARS‐CoV‐2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 181, 271–280.e8 (2020).
    1. Zhang, H. et al. The digestive system is a potential route of 2019‐nCov infection: a bioinformatics analysis based on single‐cell transcriptomes. J bioRxiv. 10.1101/2020.01.30.927806.
    1. Wong, S.H. , Lui, R.N. & Sung, J.J. COVID‐19 and the digestive system. J. Gastroenterol. Hepatol. 35, 744–748 (2020).
    1. Zhang, W. et al. Molecular and serological investigation of 2019‐nCoV infected patients: implication of multiple shedding routes. Emerg. Microbes Infect. 9, 386–389 (2020).
    1. Sun, C.B. , Wang, Y.‐Y. , Liu, G.‐H. , Liu, Z . Role of the Eye in Transmitting Human Coronavirus: What We Know and What We Do Not Know. Frontiers in Public Health 8, (2020). 10.3389/fpubh.2020.00155
    1. Lu, C.W. , Liu, X.F. & Jia, Z.F. 2019‐nCoV transmission through the ocular surface must not be ignored. Lancet 395, e39 (2020).
    1. Smith, C.A. , Kulkarni, U. , Chen, J. & Goldstein, D.R. Influenza virus inoculum volume is critical to elucidate age‐dependent mortality in mice. Aging Cell 18, e12893 (2019).
    1. Miller, D.S. , Kok, T. & Li, P. The virus inoculum volume influences outcome of influenza A infection in mice. Lab. Anim. 47, 74–77 (2013).
    1. Chen, X. et al. Detectable serum SARS‐CoV‐2 viral load (RNAaemia) is closely associated with drastically elevated interleukin 6 (IL‐6) level in critically ill COVID‐19 patients. Clin. Infect. Dis.10.1101/2020.02.29.20029520.
    1. Liu, Y. et al. Viral dynamics in mild and severe cases of COVID‐19. Lancet Infect. Dis. 10.1016/S473-3099(20)30232-2.
    1. Rodgers, T. , Leahy, D. & Rowland, M. Physiologically based pharmacokinetic modeling 1: predicting the tissue distribution of moderate‐to‐strong bases. J. Pharm. Sci. 94, 1259–1276 (2005).
    1. Rodgers, T. & Rowland, M. Physiologically based pharmacokinetic modelling 2: predicting the tissue distribution of acids, very weak bases, neutrals and zwitterions. J. Pharm. Sci. 95, 1238–1257 (2006).
    1. Rodgers, T. & Rowland, M. Mechanistic approaches to volume of distribution predictions: understanding the processes. Pharm. Res, 24, 918–933 (2007).
    1. Damle, B. , Stogniew, M. & Dowell, J. Pharmacokinetics and tissue distribution of anidulafungin in rats. Antimicrob. Agents Chemother. 52, 2673–2676 (2008).
    1. Chandrasekaran, A. , Ahmad, S. , Shen, L. , DeMaio, W. , Hultin, T. & Scatina, J. Disposition of bazedoxifene in rats. Xenobiotica 40, 578–585 (2010).
    1. Browning, D.J. Pharmacology of chloroquine and hydroxychloroquine. In: Hydroxychloroquine Chloroquine Retinopathy. 35–63 (Springer, New York, NY, 2014).
    1. McChesney, E.W. , Banks, W.F. Jr & Fabian, R.J. Tissue distribution of chloroquine, hydroxychloroquine, and desethylchloroquine in the rat. Toxicol. Appl. Pharmacol. 10, 501–513 (1967).
    1. Gupta, A. , Tulsankar, S.L. , Bhatta, R.S. & Misra, A. Pharmacokinetics, metabolism, and partial biodistribution of "pincer therapeutic" nitazoxanide in mice following pulmonary delivery of inhalable particles. Mol. Pharm. 14, 1204–1211 (2017).
    1. Lien, E.A. , Solheim, E. & Ueland, P.M. Distribution of tamoxifen and its metabolites in rat and human tissues during steady‐state treatment. Cancer Res. 51, 4837–4844 (1991).
    1. Rivulgo, V. et al. Comparative plasma exposure and lung distribution of two human use commercial azithromycin formulations assessed in murine model: a preclinical study. Biomed. Res. Int. 2013, 392010 (2013).
    1. Moss, L. , Wagner, D. , Kanaoka, E. , Olson, K. , Yueh, Y.L. & Bowers, G.D. The comparative disposition and metabolism of dolutegravir, a potent HIV‐1 integrase inhibitor, in mice, rats, and monkeys. Xenobiotica 45, 60–70 (2015).
    1. Kawai, R. , Mathew, D. , Tanaka, C. & Rowland, M. Physiologically based pharmacokinetics of cyclosporine A: extension to tissue distribution kinetics in rats and scale‐up to human. J. Pharmacol. Exp. Ther. 287, 457–468 (1998).
    1. Pharmaceuticals and Medical Devices Agency (PMDA) . Report on the deliberation results ‐ Xospata tablets 40 mg <> (2018). Accessed May 4, 2020.
    1. Bland, J.M. & Altman, D.G. Statistical methods for assessing agreement between two methods of clinical measurement. Lancet 1, 307–310 (1986).
    1. Weston, S. , Haupt, R. , Logue, J. , Matthews, K. & Frieman, M.B. FDA approved drugs with broad anti‐coronaviral activity inhibit SARS‐CoV‐2 in vitro. J bioRxiv. 10.1101/2020.03.25.008482.
    1. Ge, Y. et al. A data‐driven drug repositioning framework discovered a potential therapeutic agent targeting COVID‐19. J bioRxiv. 10.1101/2020.03.11.986836.
    1. Bojkova, D. et al. SARS‐CoV‐2 and SARS‐CoV differ in their cell tropism and drug sensitivity profiles. J bioRxiv . 10.1101/2020.04.03.024257.
    1. Touret, F. et al. In vitro screening of a FDA approved chemical library reveals potential inhibitors of SARS‐CoV‐2 replication. J bioRxiv. 10.1101/2020.04.03.023846.
    1. Wang, M. et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019‐nCoV) in vitro. Cell Res. 30, 269–271 (2020).
    1. Jeon, S. et al. Identification of antiviral drug candidates against SARS‐CoV‐2 from FDA‐approved drugs. J bioRxiv. 10.1101/2020.03.20.999730.
    1. Xu, T. , Gao, X. , Wu, Z. , Selinger, D.W. & Zhou, Z. Indomethacin has a potent antiviral activity against SARS CoV‐2 in vitro and canine coronavirus in vivo. J bioRxiv. 10.1101/2020.04.01.017624.
    1. Fintelman‐Rodrigues, N. et al. Atazanavir inhibits SARS‐CoV‐2 replication and pro‐inflammatory cytokine production. J bioRxiv. 10.1101/2020.04.04.020925.
    1. Yamamoto, N. , Matsuyama, S. , Hoshino, T. & Yamamoto, N. Nelfinavir inhibits replication of severe acute respiratory syndrome coronavirus 2 in vitro. J bioRxiv. 10.1101/2020.04.06.026476.
    1. Bukreyeva, N. , Mantlo, E.K. , Sattler, R.A. , Huang, C. , Paessler, S. & Zeldis, J. The IMPDH inhibitor merimepodib suppresses SARS‐CoV‐2 replication in vitro. J bioRxiv. 10.1101/2020.04.07.028589.
    1. Jin, Z. et al. Structure of Mpro from COVID‐19 virus and discovery of its inhibitors. Nature. 10.1038/s41586-020-2223-y.
    1. Yao, X. et al. In vitro antiviral activity and projection of optimized dosing design of hydroxychloroquine for the treatment of severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2). Clin. Infect. Dis. 10.1093/cid/ciaa237.
    1. Choy, K.‐T. et al. Remdesivir, lopinavir, emetine, and homoharringtonine inhibit SARS‐CoV‐2 replication in vitro. Antiviral. Res. 178, 104786 (2020).
    1. Caly, L. , Druce, J.D. , Catton, M.G. , Jans, D.A. & Wagstaff, K.M. The FDA‐approved drug ivermectin inhibits the replication of SARS‐CoV‐2 in vitro. Antiviral Res. 178, 104787 (2020).
    1. Xu, Z. et al. Nelfinavir is active against SARS‐CoV‐2 in Vero E6. Cells. 10.26434/chemrxiv.12039888.
    1. Chen, C. et al. Favipiravir versus Arbidol for COVID‐19: a randomized clinical trial. J. medRxiv. 10.1101/2020.03.17.20037432.
    1. Grein, J. et al. Compassionate use of remdesivir for patients with severe Covid‐19. N. Engl. J. Med. 10.1056/NEJMoa2007016.
    1. Rossignol, J.F. Nitazoxanide, a new drug candidate for the treatment of Middle East respiratory syndrome coronavirus. J. Infect. Public Health 9, 227–230 (2016).
    1. Tilmanis, D. , van Baalen, C. , Oh, D.Y. , Rossignol, J.F. & Hurt, A.C. The susceptibility of circulating human influenza viruses to tizoxanide, the active metabolite of nitazoxanide. Antiviral Res. 147, 142–148 (2017).
    1. Haffizulla, J. et al. Effect of nitazoxanide in adults and adolescents with acute uncomplicated influenza: a double‐blind, randomised, placebo‐controlled, phase 2b/3 trial. Lancet Infect Dis. 14, 609–618 (2014).
    1. Gekonge, B. , Bardin, M.C. & Montaner, L.J. Short communication: Nitazoxanide inhibits HIV viral replication in monocyte‐derived macrophages. AIDS Res. Hum. Retroviruses 31, 237–241 (2015).
    1. Trabattoni, D. et al. Thiazolides elicit anti‐viral innate immunity and reduce HIV replication. Sci. Rep. 6, 27148 (2016).
    1. Rossignol, J.F. Nitazoxanide: a first‐in‐class broad‐spectrum antiviral agent. Antiviral Res. 110, 94–103 (2014).
    1. Rossignol, J.F. , La Frazia, S. , Chiappa, L. , Ciucci, A. & Santoro, M.G. Thiazolides, a new class of anti‐influenza molecules targeting viral hemagglutinin at the post‐translational level. J. Biol. Chem. 284, 29798–29808 (2009).
    1. Hickson, S.E. , Margineantu, D. , Hockenbery, D.M. , Simon, J.A. & Geballe, A.P. Inhibition of vaccinia virus replication by nitazoxanide. Virology 518, 398–405 (2018).
    1. Wang, Y.M. et al. Antiviral activities of niclosamide and nitazoxanide against chikungunya virus entry and transmission. Antiviral Res. 135, 81–90 (2016).
    1. Zhang, Y. et al. Site‐specific N‐glycosylation characterization of recombinant SARS‐CoV‐2 spike proteins using high‐resolution mass spectrometry. J bioRxiv. 10.1101/2020.03.28.013276.
    1. Clerici, M. , Trabattoni, D. , Pacei, M. , Biasin, M. & Rossignol, J.‐F. The anti‐infective nitazoxanide shows strong immumodulating effects. J. Immuno. 186, 155.21 (2011).
    1. Jurgeit, A. , McDowell, R. , Moese, S. , Meldrum, E. , Schwendener, R. & Greber, U.F. Niclosamide is a proton carrier and targets acidic endosomes with broad antiviral effects. PLoS Pathog. 8, e1002976 (2012).
    1. Zhang, X.W. & Yap, Y.L. Old drugs as lead compounds for a new disease? Binding analysis of SARS coronavirus main proteinase with HIV, psychotic and parasite drugs. Bioorg. Med. Chem. 12, 2517–2521 (2004).
    1. Gassen, N.C. et al. SKP2 attenuates autophagy through Beclin1‐ubiquitination and its inhibition reduces MERS‐Coronavirus infection. Nat. Commun. 10, 5770 (2019).
    1. Li, R. et al. Inhibition of STAT3 by niclosamide synergizes with erlotinib against head and neck cancer. PLoS One 8, e74670 (2013).
    1. Lin, C.K. et al. Preclinical evaluation of a nanoformulated antihelminthic, niclosamide, in ovarian cancer. Oncotarget 7, 8993–9006 (2016).
    1. Miner, K. et al. Drug repurposing: the anthelmintics niclosamide and nitazoxanide are potent TMEM16A antagonists that fully bronchodilate airways. Front. Pharmacol. 10, 51 (2019).
    1. Lv, Z. , Chu, Y. & Wang, Y. HIV protease inhibitors: a review of molecular selectivity and toxicity. HIV AIDS (Auckl) 7, 95–104 (2015).
    1. Streeck, H. & Rockstroh, J.K. Review of tipranavir in the treatment of drug‐resistant HIV. Ther. Clin. Risk Manag. 3, 641–651 (2007).
    1. Sanders, J.M. , Monogue, M.L. , Jodlowski, T.Z. , Cutrell, J.B. pharmacologic treatments for coronavirus disease 2019 (COVID‐19). JAMA. 10.1001/jama.2020.6019.
    1. Justice, A.C. et al. Drug toxicity, HIV progression, or comorbidity of aging: does tipranavir use increase the risk of intracranial hemorrhage? Clin. Infect. Dis. 47, 1226–1230 (2008).
    1. Flexner, C. , Bate, G. & Kirkpatrick, P. Tipranavir. Nat. Rev. Drug Discovery 4, 955–956 (2005).
    1. Chan‐Tack, K.M. , Struble, K.A. & Birnkrant, D.B. Intracranial hemorrhage and liver‐associated deaths associated with tipranavir/ritonavir: review of cases from the FDA's Adverse Event Reporting System. AIDS Patient Care STDS 22, 843–850 (2008).
    1. Hirani, V.N. , Raucy, J.L. & Lasker, J.M. Conversion of the HIV protease inhibitor nelfinavir to a bioactive metabolite by human liver CYP2C19. Drug Metab. Dispos. 32, 1462–1467 (2004).
    1. Zhang, K.E. et al. Circulating metabolites of the human immunodeficiency virus protease inhibitor nelfinavir in humans: structural identification, levels in plasma, and antiviral activities. Antimicrob. Agents Chemother. 45, 1086–1093 (2001).
    1. Motoya, T. et al. Characterization of nelfinavir binding to plasma proteins and the lack of drug displacement interactions. HIV Med. 7, 122–128 (2006).
    1. King, J.R. & Acosta, E.P. Tipranavir. Clin. Pharmacokinet. 45, 665–682 (2006).
    1. Markowitz, M. et al. Long‐term efficacy and safety of tipranavir boosted with ritonavir in HIV‐1‐infected patients failing multiple protease inhibitor regimens: 80‐week data from a phase 2 study. J. Acquir. Immune Defic. Syndr. 45, 401–410 (2007).
    1. Justice, A.C. et al. Drug toxicity, HIV progression, or comorbidity of aging: does tipranavir use increase the risk of intracranial hemorrhage? Clin. Infect. Dis. 47, 1226–1230 (2008).
    1. Unis, G. et al. Mitochondrial mechanisms of nelfinavir toxicity in human brain microvascular endothelial cells. The FASEB Journal 30, 953.4‐.4 (2016).
    1. Lovegrove, F.E. & Kain, K.C. Chapter 6 ‐ malaria prevention. In: The Travel and Tropical Medicine Manual. 4th ed. (eds. Jong, E.C. & Sanford, C. ) 76–99 (W.B. Saunders, Edinburgh, 2008).
    1. de Kock, M. et al. Pharmacokinetics of sulfadoxine and pyrimethamine for intermittent preventive treatment of malaria during pregnancy and after delivery. CPT Pharmacometrics Syst. Pharmacol. 6, 430–438 (2017).
    1. Amici, C. , La Frazia, S. , Brunelli, C. , Balsamo, M. , Angelini, M. & Santoro, M.G. Inhibition of viral protein translation by indomethacin in vesicular stomatitis virus infection: role of eIF2alpha kinase PKR. Cell Microbiol. 17, 1391–1404 (2015).
    1. Nalamachu, S. & Wortmann, R. Role of indomethacin in acute pain and inflammation management: a review of the literature. Postgrad. Med. 126, 92–97 (2014).
    1. Pober, J.S. & Sessa, W.C. Evolving functions of endothelial cells in inflammation. Nat. Rev. Immunol. 7, 803–815 (2007).
    1. Zhang, R. et al. Tumor‐associated inflammatory microenvironment in non‐small cell lung cancer: correlation with FGFR1 and TLR4 expression via PI3K/Akt pathway. J. Cancer 10, 1004–1012 (2019).
    1. Geary, T.G. , Divo, A.D. , Jensen, J.B. , Zangwill, M. & Ginsburg, H. Kinetic modelling of the response of Plasmodium falciparum to chloroquine and its experimental testing in vitro. Implications for mechanism of action of and resistance to the drug. Biochem. Pharmacol. 40, 685–691 (1990).
    1. Pugin, J. , Dunn‐Siegrist, I. , Dufour, J. , Tissieres, P. , Charles, P.E. & Comte, R. Cyclic stretch of human lung cells induces an acidification and promotes bacterial growth. Am. J. Respir. Cell Mol. Biol. 38, 362–370 (2008).
    1. Drachman, N. , Kadlecek, S. , Pourfathi, M. , Xin, Y. , Profka, H. & Rizi, R. In vivo pH mapping of injured lungs using hyperpolarized [1‐(13) C]pyruvate. Magn. Reson. Med. 78, 1121–1130 (2017).
    1. Ginsburg, H. , Nissani, E. & Krugliak, M. Alkalinization of the food vacuole of malaria parasites by quinoline drugs and alkylamines is not correlated with their antimalarial activity. Biochem. Pharmacol. 38, 2645–2654 (1989).
    1. Vanderkooi, G. , Prapunwattana, P. & Yuthavong, Y. Evidence for electrogenic accumulation of mefloquine by malarial parasites. Biochem. Pharmacol. 37, 3623–3631 (1988).
    1. Fitch, C.D. , Chevli, R. & Gonzalez, Y. Chloroquine‐resistant Plasmodium falciparum: effect of substrate on chloroquine and amodiaquin accumulation. Antimicrob. Agents Chemother. 6, 757–762 (1974).
    1. Ritchie, E.C. , Block, J. & Nevin, R.L. Psychiatric side effects of mefloquine: applications to forensic psychiatry. J. Am. Acad. Psychiatry Law 41, 224–235 (2013).
    1. Gonzalez, D. , Schmidt, S. & Derendorf, H. Importance of relating efficacy measures to unbound drug concentrations for anti‐infective agents. Clin. Microbiol. Rev. 26, 274–288 (2013).
    1. Fox, L.M. & Saravolatz, L.D. Nitazoxanide: a new thiazolide antiparasitic agent. Clin. Infect. Dis. 40, 1173–1180 (2005).
    1. la Porte, C.J. , Sabo, J.P. , Beique, L. & Cameron, D.W. Lack of effect of efavirenz on the pharmacokinetics of tipranavir‐ritonavir in healthy volunteers. Antimicrob. Agents Chemother. 53, 4840–4844 (2009).
    1. Kruse, G. et al. The steady‐state pharmacokinetics of nelfinavir in combination with tenofovir in HIV‐infected patients. Antivir. Ther. 10, 349–355 (2005).
    1. Rainsford, K.D. et al. Effects of misoprostol on the pharmacokinetics of indomethacin in human volunteers. Clin. Pharmacol. Ther. 51, 415–421 (1992).
    1. Burger, D.M. , Agarwala, S. , Child, M. , Been‐Tiktak, A. , Wang, Y. & Bertz, R. Effect of rifampin on steady‐state pharmacokinetics of atazanavir with ritonavir in healthy volunteers. Antimicrobial Agents Chemother. 50, 3336–3342 (2006).
    1. Tett, S.E. , Cutler, D.J. , Beck, C. & Day, R.O. Concentration‐effect relationship of hydroxychloroquine in patients with rheumatoid arthritis–a prospective, dose ranging study. J. Rheumatol. 27, 1656–1660 (2000).
    1. Shida, Y. , Takahashi, N. , Nohda, S. & Hirama, T. Pharmacokinetics and pharmacodynamics of eltrombopag in healthy Japanese males. Jpn. J. Clin. Pharmacol. Ther. 42, 11–20 (2011).
    1. US Food and Drug Administration (FDA) . Abbott L. Clinical Pharmacology and Biopharmaceutics review of Kaletra oral solution (NDA#021251). <> (2020). Accessed April 13, 2020.
    1. Na‐Bangchang, K. , Limpaibul, L. , Thanavibul, A. , Tan‐Ariya, P. & Karbwang, J. The pharmacokinetics of chloroquine in healthy Thai subjects and patients with Plasmodium vivax malaria. Br. J. Clin. Pharmacol. 38, 278–281 (1994).
    1. Krudsood, S. et al. New fixed‐dose artesunate‐mefloquine formulation against multidrug‐resistant Plasmodium falciparum in adults: a comparative phase iib safety and pharmacokinetic study with standard‐dose nonfixed artesunate plus mefloquine. Antimicrob. Agents Chemother. 54, 3730–3737 (2010).
    1. Liu, P. , Ruhnke, M. , Meersseman, W. , Paiva, J.A. , Kantecki, M. & Damle, B. Pharmacokinetics of anidulafungin in critically ill patients with candidemia/invasive candidiasis. Antimicrob. Agents Chemother. 57, 1672–1676 (2013).
    1. Raja, A. , Lebbos, J. & Kirkpatrick, P. Atazanavir sulphate. Nat. Rev. Drug Discov. 2, 857–858 (2003).
    1. Harrison, T.S. & Scott, L.J. Atazanavir. Drugs 65, 2309–2336 (2005).
    1. Murdoch, D. & Plosker, G.L. Anidulafungin. Drugs 64, 2249–2258 (2004).
    1. Goel, P. & Gerriets, V. Chloroquine. In: StatPearls. (StatPearls Publishing, Treasure Island (FL). 2020.
    1. Garnock‐Jones, K.P. Eltrombopag. Drugs 71, 1333–1353 (2011).
    1. Pharmaceuticals and Medical Devices Agency (PMDA) . Report on the Deliberation Results. <> (2014).
    1. Ben‐Zvi, I. , Kivity, S. , Langevitz, P. & Shoenfeld, Y. Hydroxychloroquine: from malaria to autoimmunity. Clin. Rev. Allergy Immunol. 42, 145–153 (2012).
    1. Yeh, K.C. Pharmacokinetic overview of indomethacin and sustained‐release indomethacin. Am. J. Med. 79, 3–12 (1985).
    1. Corbett, A.H. , Lim, M.L. & Kashuba, A.D. Kaletra (lopinavir/ritonavir). Ann. Pharmacother. 36, 1193–1203 (2002).
    1. Price, R.N. et al. Artesunate/mefloquine treatment of multi‐drug resistant falciparum malaria. Trans. R Soc. Trop. Med. Hyg. 91, 574–577 (1997).
    1. McHutchison, J.G. et al. A randomized, double‐blind, placebo‐controlled dose‐escalation trial of merimepodib (VX‐497) and interferon‐alpha in previously untreated patients with chronic hepatitis C. Antivir. Ther. 10, 635–643 (2005).
    1. James, J.S. Nelfinavir (Viracept) approved: fourth protease inhibitor available. AIDS Treat. News 267, 1–2 (1997).
    1. Chen, W. , Mook, R.A. , Premont, R.T. & Wang, J. Niclosamide: beyond an antihelminthic drug. Cell. Signal. 41, 89–96 (2018).
    1. Anderson, V.R. & Curran, M.P. Nitazoxanide. Drugs 67, 1947–1967 (2007).
    1. Porche, D.J. Ritonavir (Norvir). J. Assoc. Nurses AIDS Care 8, 81–83 (1997).
    1. Miller, K.D. , Lobel, H.O. , Satriale, R.F. , Kuritsky, J.N. , Stern, R. & Campbell, C.C. Severe cutaneous reactions among American travelers using pyrimethamine‐sulfadoxine (Fansidar) for malaria prophylaxis. Am. J. Trop. Med. Hyg. 35, 451–458 (1986).
    1. Orman, J.S. & Perry, C.M. Tipranavir: a review of its use in the management of HIV infection. Drugs 68, 1435–1463 (2008).

Source: PubMed

3
Abonner