Skip to main content
Elsevier - PMC COVID-19 Collection logoLink to Elsevier - PMC COVID-19 Collection
. 2020 May 12;151:102982. doi: 10.1016/j.critrevonc.2020.102982

Considerations for interactions of drugs used for the treatment of COVID-19 with anti-cancer treatments

Anya Jafari a,*, Sahar Dadkhahfar b, Sahra Perseh c
PMCID: PMC7217119  PMID: 32460133

Abstract

SARS-CoV2 infection is an emerging issue worldwide. Cancer patient are at increased risk of infection compared to general population. On the other hand, these patients are at major risk of drug interactions caused by renal and hepatic impairment background. Because of the long-term use of chemotherapy drugs, drug interactions are important in these patients especially with SARS-CoV2 treatments now. This paper is review of reported drug interactions of current treatments for COVID-19 and anticancer agents.

Keywords: Drug interaction, COVID-19, SARS-CoV2, Cancer, Chemotherapy

1. Introduction

The novel coronavirus (SARS-CoV2) has caused a growing pandemic and global issue now (Zhou et al., 2020). Cancer patients are at a greater risk of catastrophic outcomes of Coronavirus infectious disease 2019 (COVID-19) because of the older age, immunosuppressive condition and combined comorbidity. Cancer patients may become infected by SARS-CoV2 during chemotherapy or may need to receive chemotherapy after the resolution of their disease and receiving coronavirus treatment (Wang and Zhang, 2020).

Here we reviewed drug interactions of current covid-19 drugs with antineoplastic agents. Montamat SC et al., said that the risk of drug interactions is higher in cancer patient due to their underlying disease, older age and consumption of multiple medications (Montamat et al., 1989). The current paper is a review of the reported and expected drug interactions of current treatments for COVID-19 and anticancer agents.

2. Mechanisms of interaction

Drug-drug interactions (DDIs) commonly happen when two drugs with DDIs administered before 4–5 half-lives of one of them (Ito, 2011). DDIs can cause three results: decreased therapeutic effect/adverse effects, enhanced therapeutic effect/adverse effects or a new side effect that does not occur with each drug separately. In pharmacodynamic interaction, both drugs affect the same physiologic pathway (Blower et al., 2005). This effect can be inhibitory or inducible.

Pharmacokinetic interaction happens when one drug influences other drug’s absorption, distribution, metabolism or excretion (Blower et al., 2005). Drugs may affect the GI motility or PH, serum albumin concentration which can affect another drug absorption or distribution (Blower et al., 2005). Cytochrome P450 (CYP) with more than 50 isoenzymes is responsible for most of drug metabolism in liver. One drug can stimulate or inhibit its own CYP isoenzyme or other isoenzymes with further influencing the metabolism of other drugs that are metabolized with the same isoenzyme (Zhang et al., 2009). Another site of drug interactions occurs at P-glycoprotein that plays an important role in transporting drugs into cell on the cell membrane (Blower et al., 2005; Zhang et al., 2009).

3. Common therapeutic regiments for COVID-19

The most common proposed treatments for COVID-19 include chloroquine and hydroxychloroquine, azatinavir/ritonavir, lopinavir/ritonavir, remdesivir, tocilizumab and favipiravir (Baden and Rubin, 2020a, 2020b; Cascella et al., 2020; Dong et al., 2020; Guo et al., 2020).

3.1. Chloroquine

Chloroquine (CQ) and hydroxychloroquine (HCQ) are both 4-aminoquinoline agents that historically known as antimalaria drug from 1940s (Verbaanderd et al., 2017). These drugs have been used for treatment of rheumatoid arthritis, lupus erythematous, AIDS and recently COVID-19 (Ito, 2011; Solomon and Lee, 2009). Additionally, there are several in vivo and in vitro studies that confirm the anticancer effect of both CQ and HCQ. The most prominent evidences are three phase 3 clinical trials that used CQ during glioblastoma multiform with carmustin or temozolomide and showed positive results with added CQ. There are other phase 1–2 trials of addition the CQ to other chemotherapy regimens with hopeful results (Manic et al., 2014).Terminal elimination half-life of CQ is about 1–2 months and of 50 days for HCQ (Verbaanderd et al., 2017).

Short term use of CQ and HCQ is rarely associated with major side effects but serious side effects such as cardiomyopathy, irreversible retinopathy, myelosuppression and hypoglycemia have been reported after long-term use (Verbaanderd et al., 2017). Among the most serious adverse effects are cardiac side-effects such as atrioventricular block, bundle branch block, cardiac arrhythmia, cardiac failure, cardiomyopathy, electrocardiographic (ECG) changes including flattened T wave, T wave inversion, prolonged QT interval, widened QRS complex, hypotension, torsade’s de pointes, ventricular fibrillation and ventricular tachycardia (Page et al., 2016).

Tamoxifen, an antiestrogen agent, is administered for breast cancer patients who needs to take it for years (Regan et al., 2016). CQ decrease the level of tamoxifen by CYP2D6 inhibition effect (Blower et al., 2005; Marmor et al., 2016). Whether consuming concurrent CQ or HCQ could affect the efficacy of tamoxifen in breast cancer patients is not clear but should be considered.

With regard to anti emetic drugs physicians should be noted that, granisetron is a safe antiemetic agent to be used concurrently with HCQ or CQ because unlike ondansetron, it does not appear to affect any CYP isoenzyme (Blower et al., 2005).

Ado-Trastuzumab Emtansine (TDM1) that is used to treat HER2-positive metastatic breast cancer, also metabolized largely by CYP3A4 but don’t have any effect on this enzyme. (Ballantyne and Dhillon, 2013) We summarized main interactions of this drug with anticancer medicines (Table1 ).

Table 1.

Chloroquine DDIs:

Covid-19 drug Type of interaction Result
Chloroquine Q-T interval prolongation Apalutamide, Leuprolide, Goserelin, Triptorelin,(Garnick, 2005) Increase Q-T prolongation probability
Eribulin (Perry, 2011), Ribociclib (Syed, 2017), Inotuzumab (Kebriaei et al., 2018), Gemtuzumab (Selby et al., 2019), Lenvatinib(Frampton, 2016), Dasatinib (Keam, 2008), Nilotinib (Kim et al., 2012), Cabozantinib and Ceritinib (Shah and Morganroth, 2015), Methadone (Barkin et al., 1998) Oxaliplatin(Chang et al., 2013)
Ondansetron(Charbit et al., 2005)
CYP3A4 induce Apalutamide(Pérez-Ruixo et al., 2020), Ivosidenib (Pérez-Ruixo et al., 2020), Fedratinib(Xu) Decrease the level of CQ
Dabrafenib(Ballantyne and Garnock-Jones, 2013), Encorafenib(Ballantyne and Garnock-Jones, 2013)
CYP3A4 inhibit Idelalisib(Ballantyne and Garnock-Jones, 2013), Crizotinib(Forde and Rudin, 2012), Fedratinib(Xu et al., 2014), Dasatinib(Haouala et al., 2011), Abiraterone(Benoist et al., 2016), Bicalutamide(Meulenbeld et al., 2013), Aprepitant(Majumdar et al., 2003), Imatinib(Majumdar et al., 2003) Increase the level of CQ
CYP2D6 inducers -- --
CYP2D6 inhibitors Dacomitinib(Bello et al., 2012), Abiraterone(Yang, 2011), Ondansetron(Blower et al., 2005), Methadone(Wu et al., 1993) Increase the level of QC
Pharmacodynamic synergism All chemotherapy agents Myelosuppression
Pharmacodynamic antagonism Sipuleucel-T(Cooper and Magwere, 2008; Plosker, 2011)
Effect on distribution MTX(Blower et al., 2005)

3.2. Protease inhibitors

Protease inhibitors (PIs) such as atazanavir, ritonavir, lopinavir have been for treatment of COVID-19 (Baden, Lindsey R. and Rubin, Eric J., 2020). Lopinavir is used in combination with ritonavir- Kaletra- to increase the lopinavir’s bioavailability (Eckhardt and Gulick, 2017). Reported half of lopinavir/ritonavir and atazanavir/ritonavir are 2–7/3−4 h, 8–9/5−6 h, respectively (Boffito et al., 2008; Chandwani and Shuter, 2008). About 85–95 % of these drugs bind to plasma proteins and metabolized in liver mainly by CYP3A4 isoenzyme (Makinson et al., 2010). Lopinavir is metabolized by CYP3A4 and on the other hand ritonavir is a potent inhibitor of CYP3A4, so the coadministration of them leads an increase in the effect of lopinavir; therefore, lopinavir is not used alone (Berretta et al., 2016; Makinson et al., 2010; Rudek et al., 2011).

Atazanavir, lopinavir/ritonavir also associated with Q-T interval prolongation so should be prescribed with caution with drugs with the same side effect such as tamoxifen, anthracyclines, dasatninb, lapatinib, nilotinib and sunitinib (Table 2 ) (Kebriaei et al., 2018) (DeRemer et al., 2008; Johnson et al., 2010; Lee et al., 2010; Pillai et al., 2014).

Table 2.

Protease inhibitors DDIs:

Covid-19 drug Type of interaction Result
Protease inhibitors* (Makinson et al., 2010; Pasin, 2015; Rudek et al., 2011) -- Platinum No effect
Inhibition of CYP3A4 Taxans Increase the level of docetaxel
Inhibition of CYP3A4 Vincaalkaloids Increase the level vincaalkaloids
-- Gemcitabine No effect
-- Topotecan No effect
Inhibition of CYP3A4 Irinotecan Increase the level of irinotecan
-- Pemetrexed No effect
-- Bevacizumab No effect
-- Cetuximab No effect
Inhibition of CYP3A4 Erlotinib Increase the level of erlotinib
Inhibition of CYP3A4 Gefitinib Increase the level of gefitinib
Inhibition of CYP3A4 Etoposide Expect to increase the etoposide toxicity
-- Anthracycline No effect
Inhibition of CYP3A4 Everolimus Increase the level of Everolimus
*

It should be mentioned that hyperbilirubinemia can be seen with atazanavir, but is not a guidance for chemotherapy drug adjustment dose (Rudek et al., 2011).

3.3. Favipiravir

Favipiravir is a pyrazine analog, originally made in Japan, oseltamivir resistant influenza (Furuta et al., 2013). The drug inhibits RNA-dependent RNA polymerase enzymes leading to prevention of virus replication (Furuta et al., 2009). The elimination half-life is about 2–5.5  with a protein binding of 54 % in plasma and metabolized by aldehyde oxidase (AO) and xanthine oxidase to its metabolite, T705M1, in liver and excreted to urine.(Du and Chen, 2020) Dose adjustment based on hepatic impairment is not needed however there is not enough humanized study about it (Du and Chen, 2020). The proposed dose for COVID-19 is the loading dose of 3200 mg on day 1 and maintenance dose of 1200 mg on day 2−14 (Du and Chen, 2020). Main side effects of favipiravir include mild to moderate diarrhea, elevated liver enzymes, testicular toxicity, increased blood uric acid and decrease in neutrophil count (2014). The rate of Q-T prolongation is low (8%) (2014). There is limited clinical data about DDIs and metabolism of favipiravir. In vivo study showed inhibitory effect of favipiravir on CYP2C8 isoenzyme (2014). As well as, other study revealed favipiravir inhibits AO (Du and Chen, 2020). So, more caution was necessary when used AO inhibitors such as tamoxifen or CYP2C8 substrates like paclitaxel with favipiravir (2014).

3.4. Ivermectin

Ivermectin is a broad spectrum antiparasitic agent that recently reported to have in vivo effect on SARS-CoV2 virus with 98 % elimination rate of virus RNA in or out of cells (Caly et al., 2020). It is used by oral or subcutaneous route and metabolized in liver by CYP3A4 isoenzyme (Canga et al., 2008; Zeng et al., 1998). Ivermectin binds to plasma protein by rate of 93 % and is mainly excreted in feces (Klotz et al., 1990). Its half-life based on administration route is around 12−20 hours (Canga et al., 2008). The absorption, distribution and elimination of ivermectin is dependent on P-glycoprotein, on the other hand, it is a potent inhibitor of P-glycoprotein (Ménez et al., 2012). The main interactions of this drug are caused by its effects on P-glycoprotein. Ivermectin may change the p-glycoprotein ABCB1 substrate (Ménez et al., 2012). The most reported side effects are reported in the context of its use an antiparasitic drug as the result of immunologic response to parasite, including skin rash, fever, headache, nausea and dizziness (Juarez et al., 2018). Drug interaction of Ivermectin and chemotherapy agents is an important issue both in treatment of cancer or COVID-19 (Canga et al., 2008).

There are not enough clinical data about ivermectin drug interactions. It is reasonable to cautiously administer this drug with drugs that are metabolized by CYP3A4 and induce or inhibit P-glycoproteins (Table 3 ) (Jiang et al., 2019; Mealey et al., 2003).

Table 3.

Ivermectin DDIs:

Covid-19 drug Type of interaction
Ivermectin P-glycoprotein substrates Doxorubicin (Kim, 2002)
Mitoxantrone (Kim, 2002)
Paclitaxel (Lin, 2003)
Vinblastine (Lin, 2003)
Vincristine (Kim, 2002)
Ivermectin (Lin, 2003)
Lapatinib (Cidon, 2017)
Lenvatinib (Cidon, 2017)
Sorafenib (Cidon, 2017)
Actinomycine D (Zhou, 2008)
Docetaxel (Zhou, 2008)
Imatinib (Zhou, 2008)
Irinotecan (Zhou, 2008)
Mitomycine C (Zhou, 2008)
Topotecan (Zhou, 2008)
P-glycoprotein inhibitors Etoposide (Kim, 2002)
Tamoxifen (Kim, 2002)
Ivermectin (Ménez et al., 2012)
Methadone (Kim, 2002)
Vinblastin (Zhou, 2008)
P-glycoprotein inducers Cisplatin (Zhou, 2008)
Daunorubicin (Zhou, 2008)
Doxorubicin (Zhou, 2008)
Vinblastine (Zhou, 2008)
Vincristine (Zhou, 2008)
Etoposide (Zhou, 2008)

3.5. Remdesivir

Remdisivir (RDV) is a new investigational antiviral agent, that have been used against coronavirus. RDV inhibits the RNA-dependent RNA polymerase (Al-Tawfiq et al., 2020; Gordon et al., 2020; Tchesnokov et al., 2019).

Its oral bioavailability is very low, so it is used intravenously (Mealey et al., 2003). Unfortunately, there are not any data about the drug pharmacokinetic and drug-drug interaction. In a phase II trial on remdesivir, 9 cases of side-effects were reported but 8 cases of them were not related to remdesivir (Tchesnokov et al., 2019).

While waiting for more definite evidence, physicians should prescribe this drug with caution in cases of multiple medications.

3.6. Tocilizumab

Tocilizumab (TCZ, Actemra) is an Ig G recombinant humanized monoclonal antibody that blocks the receptor of IL-6. TCZ has been used for treatment of rheumatoid arthritis (Zhang and Brennan, 2010). As the result of the cytokine storm the increased level of IL-6 is observed in the course of COVID-19, based on this finding, TCZ is currently used for treatment of this condition (Coomes and Haghbayan, 2020).

There is no information regarding the metabolism and plasma protein binding of tocilizumab. Its half- life depending on administration dose and routes is 11–13 days (Grange et al., 2011).

Reported adverse effects are upper respiratory tract infection, neutropenia, headache, hypertension, ALT elevation and lipid profile changes. Mild reaction in infusion site is common (Oldfield et al., 2009; Sheppard et al., 2017). One study showed that TCZ has no effect on QT interval (Grange et al., 2011).

Among the drugs that has concurrently used with TCZ, It seems that co-administration of TCZ has no effect on MTX pharmacokinetic (Schmitt et al., 2012). Increasing the level of IL-6 is the consequence of blocking IL-6 receptors by TCZ. Elevated IL-6 reduces CYP450 activity, TCZ may reverse this reduced activity of CYPA50. This issue must be kept in mind when we want to administer TCZ with other agents that metabolized by CYP450 (Schmitt et al., 2011). TCZ is an immunosuppressive agent; therefore, there is a concern toward its administration in cancer patients who are already immunosuppressed as the result of chemotherapy. It should also be noted that due to the long half-life of TCZ monitoring of this interaction may be necessary for 1–2 months after the discontinuation of TCZ (Roche Pharma, 2013).

4. Conclusion

In conclusion, there are limited data about the metabolism or drug interactions some of the current COVID-19 treatments. But some of them have been studied more. Knowing of the metabolism, half-life and drug interactions of the current covid-19 treatments help physicians to make a better and quick decisions and manage correctly.

Declaration of Competing Interest

There are no conflict of interest.

CRediT authorship contribution statement

Anya Jafari: Data curation, Writing - original draft. Sahar Dadkhahfar: Writing - review & editing. Sahra Perseh: Data curation.

Acknowledgements

None declared.

References

  1. Al-Tawfiq J.A., Al-Homoud A.H., Memish Z.A. 2020. Remdesivir as a Possible Therapeutic Option for the COVID-19. Travel Medicine and Infectious Disease. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Baden L.R., Rubin E.J. Covid-19—the search for effective therapy. Mass Med. Soc. 2020 doi: 10.1056/NEJMe2005477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Baden L.R., Rubin E.J. Covid-19 — the search for effective therapy. N. Engl. J. Med. 2020 doi: 10.1056/NEJMe2005477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Ballantyne A., Dhillon S. Trastuzumab emtansine: first global approval. Drugs. 2013;73(7):755–765. doi: 10.1007/s40265-013-0050-2. [DOI] [PubMed] [Google Scholar]
  5. Ballantyne A.D., Garnock-Jones K.P. Dabrafenib: first global approval. Drugs. 2013;73(12):1367–1376. doi: 10.1007/s40265-013-0095-2. [DOI] [PubMed] [Google Scholar]
  6. Barkin R.L., Barkin D.S., Barkin S.J., Barkin S.A. Opiate, opioids, and centrally acting analgesics and drug interactions: the emerging role of the psychiatrist. Med. Update Psychiatr. 1998;3(6):171–175. [Google Scholar]
  7. Bello C.L., LaBadie R.R., Ni G., Boutros T., McCormick C., Ndongo M.N. The effect of dacomitinib (PF-00299804) on CYP2D6 activity in healthy volunteers who are extensive or intermediate metabolizers. Cancer Chemother. Pharmacol. 2012;69(4):991–997. doi: 10.1007/s00280-011-1793-7. [DOI] [PubMed] [Google Scholar]
  8. Benoist G.E., Hendriks R.J., Mulders P.F., Gerritsen W.R., Somford D.M., Schalken J.A., van Oort I.M., Burger D.M., van Erp N.P. Pharmacokinetic aspects of the two novel oral drugs used for metastatic castration-resistant prostate cancer: abiraterone acetate and enzalutamide. Clin. Pharmacokinet. 2016;55(11):1369–1380. doi: 10.1007/s40262-016-0403-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Berretta M., Caraglia M., Martellotta F., Zappavigna S., Lombardi A., Fierro C., Atripaldi L., Muto T., Valente D., De Paoli P. Drug–drug interactions based on pharmacogenetic profile between highly active antiretroviral therapy and antiblastic chemotherapy in cancer patients with HIV infection. Front. Pharmacol. 2016;7:71. doi: 10.3389/fphar.2016.00071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Blower P., De Wit R., Goodin S., Aapro M. Drug–drug interactions in oncology: why are they important and can they be minimized? Crit. Rev. Oncol. Hematol. 2005;55(2):117–142. doi: 10.1016/j.critrevonc.2005.03.007. [DOI] [PubMed] [Google Scholar]
  11. Boffito M., Else L., Back D., Taylor J., Khoo S., Sousa M., Pozniak A., Gazzard B., Moyle G. Pharmacokinetics of atazanavir/ritonavir once daily and lopinavir/ritonavir twice and once daily over 72 h following drug cessation. Antivir. Ther. 2008;13(7):901–907. [PubMed] [Google Scholar]
  12. 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. 2020 doi: 10.1016/j.antiviral.2020.104787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Canga A.G., Prieto A.M.S., Liébana M.J.D., Martínez N.F., Vega M.S., Vieitez J.J.G. The pharmacokinetics and interactions of ivermectin in humans—a mini-review. AAPS J. 2008;10(1):42–46. doi: 10.1208/s12248-007-9000-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Cascella M., Rajnik M., Cuomo A., Dulebohn S.C., Di Napoli R. StatPearls Publishing; 2020. Features, Evaluation and Treatment Coronavirus (COVID-19), StatPearls [Internet] [PubMed] [Google Scholar]
  15. Chandwani A., Shuter J. Lopinavir/ritonavir in the treatment of HIV-1 infection: a review. Ther. Clin. Risk Manag. 2008;4(5):1023. doi: 10.2147/tcrm.s3285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Chang R.-Y., Lee M.-Y., Kan C.-B., Hsu W.-P., Hsiao P.-C. Oxaliplatin-induced acquired long QT syndrome with torsades de pointes and myocardial injury in a patient with dilated cardiomyopathy and rectal cancer. J. Chin. Med. Assoc. 2013;76(8):466–469. doi: 10.1016/j.jcma.2013.05.001. [DOI] [PubMed] [Google Scholar]
  17. Charbit B., Albaladejo P., Funck-Brentano C., Legrand M., Samain E., Marty J. Prolongation of QTc interval after postoperative nausea and vomiting treatment by droperidol or ondansetron. Anesthesiology. 2005;102(6):1094–1100. doi: 10.1097/00000542-200506000-00006. [DOI] [PubMed] [Google Scholar]
  18. Cidon E.U. 2017. Tyrosine Kinases Inhibitors: Interactions and Safe Use. [Google Scholar]
  19. Coomes E.A., Haghbayan H. Interleukin-6 in COVID-19: a systematic review and meta-analysis. medRxiv. 2020;2020:2003. doi: 10.1002/rmv.2141. 2030.20048058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Cooper R., Magwere T. Chloroquine: novel uses & manifestations. Indian J. Med. Res. 2008;127(4) [PubMed] [Google Scholar]
  21. DeRemer D.L., Ustun C., Natarajan K. Nilotinib: a second-generation tyrosine kinase inhibitor for the treatment of chronic myelogenous leukemia. Clin. Ther. 2008;30(11):1956–1975. doi: 10.1016/j.clinthera.2008.11.014. [DOI] [PubMed] [Google Scholar]
  22. Dong L., Hu S., Gao J. Discovering drugs to treat coronavirus disease 2019 (COVID-19) Drug Discov. Ther. 2020;14(1):58–60. doi: 10.5582/ddt.2020.01012. [DOI] [PubMed] [Google Scholar]
  23. Du Y.X., Chen X.P. Favipiravir: pharmacokinetics and concerns about clinical trials for 2019‐nCoV infection. Clin. Pharmacol. Ther. 2020 doi: 10.1002/cpt.1844. [DOI] [PubMed] [Google Scholar]
  24. Eckhardt B.J., Gulick R.M. Elsevier; 2017. Drugs for HIV infection, Infectious Diseases; pp. 1293–1308. e1292. [Google Scholar]
  25. Forde P.M., Rudin C.M. Crizotinib in the treatment of non-small-cell lung cancer. Expert Opin. Pharmacother. 2012;13(8):1195–1201. doi: 10.1517/14656566.2012.688029. [DOI] [PubMed] [Google Scholar]
  26. Frampton J.E. Lenvatinib: a review in refractory thyroid cancer. Target. Oncol. 2016;11(1):115–122. doi: 10.1007/s11523-015-0416-3. [DOI] [PubMed] [Google Scholar]
  27. Furuta Y., Takahashi K., Shiraki K., Sakamoto K., Smee D.F., Barnard D.L., Gowen B.B., Julander J.G., Morrey J.D. T-705 (favipiravir) and related compounds: novel broad-spectrum inhibitors of RNA viral infections. Antiviral Res. 2009;82(3):95–102. doi: 10.1016/j.antiviral.2009.02.198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Furuta Y., Gowen B.B., Takahashi K., Shiraki K., Smee D.F., Barnard D.L. Favipiravir (T-705), a novel viral RNA polymerase inhibitor. Antiviral Res. 2013;100(2):446–454. doi: 10.1016/j.antiviral.2013.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Garnick, M., 2005. Methods for treating long QT syndrome. Google Patents.
  30. Gordon C.J., Tchesnokov E.P., Feng J.Y., Porter D.P., Götte M. The antiviral compound remdesivir potently inhibits RNA-dependent RNA polymerase from Middle East respiratory syndrome coronavirus. J. Biol. Chem. 2020;295(15):4773–4779. doi: 10.1074/jbc.AC120.013056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Grange S., Schmitt C., Banken L., Kuhn B., Zhang X. Thorough QT/QTc study of tocilizumab after single-dose administration at therapeutic and supratherapeutic doses in healthy subjects. Int. J. Clin. Pharmacol. Ther. 2011;49(11):648–655. doi: 10.5414/cp201549. [DOI] [PubMed] [Google Scholar]
  32. Guo Y.-R., Cao Q.-D., Hong Z.-S., Tan Y.-Y., Chen S.-D., Jin H.-J., Tan K.-S., Wang D.-Y., Yan Y. The origin, transmission and clinical therapies on coronavirus disease 2019 (COVID-19) outbreak–an update on the status. Mil. Med. Res. 2020;7(1):1–10. doi: 10.1186/s40779-020-00240-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Haouala A., Widmer N., Duchosal M.A., Montemurro M., Buclin T., Decosterd L.A. Drug interactions with the tyrosine kinase inhibitors imatinib, dasatinib, and nilotinib. Blood. 2011;117(8):e75–e87. doi: 10.1182/blood-2010-07-294330. [DOI] [PubMed] [Google Scholar]
  34. Ito S. Pharmacokinetics 101. Paediatr. Child Health. 2011;16(9):535–536. doi: 10.1093/pch/16.9.535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Jiang L., Wang P., Sun Y.-J., Wu Y.-J. Ivermectin reverses the drug resistance in cancer cells through EGFR/ERK/Akt/NF-κB pathway. J. Exp. Clin. Cancer Res. 2019;38(1):1–18. doi: 10.1186/s13046-019-1251-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Johnson F.M., Agrawal S., Burris H., Rosen L., Dhillon N., Hong D., Blackwood‐Chirchir A., Luo F.R., Sy O., Kaul S. Phase 1 pharmacokinetic and drug‐interaction study of dasatinib in patients with advanced solid tumors. Cancer. 2010;116(6):1582–1591. doi: 10.1002/cncr.24927. [DOI] [PubMed] [Google Scholar]
  37. Juarez M., Schcolnik-Cabrera A., Dueñas-Gonzalez A. The multitargeted drug ivermectin: from an antiparasitic agent to a repositioned cancer drug. Am. J. Cancer Res. 2018;8(2):317. [PMC free article] [PubMed] [Google Scholar]
  38. Keam S.J. Dasatinib. BioDrugs. 2008;22(1):59–69. doi: 10.2165/00063030-200822010-00007. [DOI] [PubMed] [Google Scholar]
  39. Kebriaei P., Cutler C., De Lima M., Giralt S., Lee S.J., Marks D., Merchant A., Stock W., Van Besien K., Stelljes M. Management of important adverse events associated with inotuzumab ozogamicin: expert panel review. Bone Marrow Transplant. 2018;53(4):449–456. doi: 10.1038/s41409-017-0019-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Kim R.B. Drugs as P-glycoprotein substrates, inhibitors, and inducers. Drug Metab. Rev. 2002;34(1-2):47–54. doi: 10.1081/dmr-120001389. [DOI] [PubMed] [Google Scholar]
  41. Kim T.D., Le Coutre P., Schwarz M., Grille P., Levitin M., Fateh-Moghadam S., Giles F.J., Dörken B., Haverkamp W., Köhncke C. Clinical cardiac safety profile of nilotinib. Haematologica. 2012;97(6):883–889. doi: 10.3324/haematol.2011.058776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Klotz U., Ogbuokiri J., Okonkwo P. Ivermectin binds avidly to plasma proteins. Eur. J. Clin. Pharmacol. 1990;39(6):607–608. doi: 10.1007/BF00316107. [DOI] [PubMed] [Google Scholar]
  43. Lee H.A., Hyun S.A., Park S.G., Kim K.S. Electrophysiological effects of the anti-cancer drug lapatinib on cardiac ion channels. J. Pharmacol. Toxicol. Methods. 2010;2(62):e35. doi: 10.1111/j.1742-7843.2010.00556.x. [DOI] [PubMed] [Google Scholar]
  44. Lin J.H. Drug–drug interaction mediated by inhibition and induction of P-glycoprotein. Adv. Drug Deliv. Rev. 2003;55(1):53–81. doi: 10.1016/s0169-409x(02)00171-0. [DOI] [PubMed] [Google Scholar]
  45. Majumdar A.K., McCrea J.B., Panebianco D.L., Hesney M., Dru J., Constanzer M., Goldberg M.R., Murphy G., Gottesdiener K.M., Lines C.R. Effects of aprepitant on cytochrome P450 3A4 activity using midazolam as a probe. Clin. Pharmacol. Ther. 2003;74(2):150–156. doi: 10.1016/S0009-9236(03)00123-1. [DOI] [PubMed] [Google Scholar]
  46. Makinson A., Pujol J.-L., Le Moing V., Peyriere H., Reynes J. Interactions between cytotoxic chemotherapy and antiretroviral treatment in human immunodeficiency virus-infected patients with lung cancer. J. Thorac. Oncol. 2010;5(4):562–571. doi: 10.1097/JTO.0b013e3181d3ccf2. [DOI] [PubMed] [Google Scholar]
  47. Manic G., Obrist F., Kroemer G., Vitale I., Galluzzi L. Chloroquine and hydroxychloroquine for cancer therapy. Mol. Cell. Oncol. 2014;1(1) doi: 10.4161/mco.29911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Marmor M.F., Kellner U., Lai T.Y., Melles R.B., Mieler W.F. Recommendations on screening for chloroquine and hydroxychloroquine retinopathy (2016 revision) Ophthalmology. 2016;123(6):1386–1394. doi: 10.1016/j.ophtha.2016.01.058. [DOI] [PubMed] [Google Scholar]
  49. Mealey K.L., Northrup N.C., Bentjen S.A. Increased toxicity of P-glycoprotein-substrate chemotherapeutic agents in a dog with the MDR1 deletion mutation associated with ivermectin sensitivity. J. Am. Vet. Med. Assoc. 2003;223(10):1453–1455. doi: 10.2460/javma.2003.223.1453. [DOI] [PubMed] [Google Scholar]
  50. Ménez C., Mselli-Lakhal L., Foucaud-Vignault M., Balaguer P., Alvinerie M., Lespine A. Ivermectin induces P-glycoprotein expression and function through mRNA stabilization in murine hepatocyte cell line. Biochem. Pharmacol. 2012;83(2):269–278. doi: 10.1016/j.bcp.2011.10.010. [DOI] [PubMed] [Google Scholar]
  51. Meulenbeld H.J., de Bono J.S., Tagawa S.T., Whang Y.E., Li X., Heath K.H., Zandvliet A.S., Ebbinghaus S.W., Hudes G.R., de Wit R. Tolerability, safety and pharmacokinetics of ridaforolimus in combination with bicalutamide in patients with asymptomatic, metastatic castration-resistant prostate cancer (CRPC) Cancer Chemother. Pharmacol. 2013;72(4):909–916. doi: 10.1007/s00280-013-2250-6. [DOI] [PubMed] [Google Scholar]
  52. Montamat S.C., Cusack B.J., Vestal R.E. Management of drug therapy in the elderly. N. Engl. J. Med. 1989;321(5):303–309. doi: 10.1056/NEJM198908033210507. [DOI] [PubMed] [Google Scholar]
  53. Oldfield V., Dhillon S., Plosker G.L. Tocilizumab. Drugs. 2009;69(5):609–632. doi: 10.2165/00003495-200969050-00007. [DOI] [PubMed] [Google Scholar]
  54. Page R.L., O’Bryant C.L., Cheng D., Dow T.J., Ky B., Stein C.M., Spencer A.P., Trupp R.J., Lindenfeld J. Drugs that may cause or exacerbate heart failure: a scientific statement from the American Heart Association. Circulation. 2016;134(6):e32–e69. doi: 10.1161/CIR.0000000000000426. [DOI] [PubMed] [Google Scholar]
  55. Pasin V. Atazanavir/everolimus/ritonavir interaction. Reactions. 2015;1569 38-19. [Google Scholar]
  56. Pérez-Ruixo C., Pérez-Blanco J.S., Chien C., Yu M., Ouellet D., Pérez-Ruixo J.-J., Ackaert O. Population pharmacokinetics of apalutamide and its active metabolite N-desmethyl-apalutamide in healthy and castration-resistant prostate cancer subjects. Clin. Pharmacokinet. 2020;59(2):229–244. doi: 10.1007/s40262-019-00808-7. [DOI] [PubMed] [Google Scholar]
  57. Perry C.M. Eribulin. Drugs. 2011;71(10):1321–1331. doi: 10.2165/11207520-000000000-00000. [DOI] [PubMed] [Google Scholar]
  58. Pharmaceuticals and Medical Devices Agency: Avigan (favipiravir) Review Report 2014 https://www.pmda.go.jp/files/000210319.pdf. (Accessed April 15 2020).
  59. Pillai V.C., Parise R.A., Christner S.M., Rudek M.A., Beumer J.H., Venkataramanan R. Potential interactions between HIV drugs, ritonavir and efavirenz and anticancer drug, nilotinib—a study in primary cultures of human hepatocytes that is applicable to HIV patients with cancer. J. Clin. Pharmacol. 2014;54(11):1272–1279. doi: 10.1002/jcph.333. [DOI] [PubMed] [Google Scholar]
  60. Plosker G.L. Sipuleucel-T. Drugs. 2011;71(1):101–108. doi: 10.2165/11206840-000000000-00000. [DOI] [PubMed] [Google Scholar]
  61. Regan M.M., Francis P.A., Pagani O., Fleming G.F., Walley B.A., Viale G., Colleoni M., Láng I., Gómez H.L., Tondini C. Absolute benefit of adjuvant endocrine therapies for premenopausal women with hormone receptor–positive, human epidermal growth factor receptor 2–negative early breast cancer: TEXT and SOFT trials. J. Clin. Oncol. 2016;34(19):2221. doi: 10.1200/JCO.2015.64.3171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Roche Pharma A. Eu Summary of Product Characteristics 2013. 2013. RoActemra 20 mg/mL concentrate for solution for infusion. [Google Scholar]
  63. Rudek M.A., Flexner C., Ambinder R.F. Use of antineoplastic agents in patients with cancer who have HIV/AIDS. Lancet Oncol. 2011;12(9):905–912. doi: 10.1016/S1470-2045(11)70056-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Schmitt C., Kuhn B., Zhang X., Kivitz A., Grange S. Disease–drug–drug interaction involving tocilizumab and simvastatin in patients with rheumatoid arthritis. Clin. Pharmacol. Ther. 2011;89(5):735–740. doi: 10.1038/clpt.2011.35. [DOI] [PubMed] [Google Scholar]
  65. Schmitt C., Kuhn B., Zhang X., Kivitz A., Grange S. Tocilizumab has no clinically relevant effects on methotrexate pharmacokinetics in patients with rheumatoid arthritis. Int. J. Clin. Pharmacol. Ther. 2012;50(3):218–223. doi: 10.5414/cp201613. [DOI] [PubMed] [Google Scholar]
  66. Selby C., Yacko L.R., Glode A.E. Gemtuzumab ozogamicin: back again. J. Adv. Pract. Oncol. 2019;10(1):68. [PMC free article] [PubMed] [Google Scholar]
  67. Shah R.R., Morganroth J. Update on cardiovascular safety of tyrosine kinase inhibitors: with a special focus on QT interval, left ventricular dysfunction and overall risk/benefit. Drug Saf. 2015;38(8):693–710. doi: 10.1007/s40264-015-0300-1. [DOI] [PubMed] [Google Scholar]
  68. Sheppard M., Laskou F., Stapleton P.P., Hadavi S., Dasgupta B. Taylor & Francis; 2017. Tocilizumab (Actemra) [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Solomon V.R., Lee H. Chloroquine and its analogs: a new promise of an old drug for effective and safe cancer therapies. Eur. J. Pharmacol. 2009;625(1-3):220–233. doi: 10.1016/j.ejphar.2009.06.063. [DOI] [PubMed] [Google Scholar]
  70. Syed Y.Y. Ribociclib: first global approval. Drugs. 2017;77(7):799–807. doi: 10.1007/s40265-017-0742-0. [DOI] [PubMed] [Google Scholar]
  71. Tchesnokov E.P., Feng J.Y., Porter D.P., Götte M. Mechanism of inhibition of Ebola virus RNA-dependent RNA polymerase by remdesivir. Viruses. 2019;11(4):326. doi: 10.3390/v11040326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Verbaanderd C., Maes H., Schaaf M.B., Sukhatme V.P., Pantziarka P., Sukhatme V., Agostinis P., Bouche G. Repurposing Drugs in Oncology (ReDO)—chloroquine and hydroxychloroquine as anti-cancer agents. ecancermedicalscience. 2017:11. doi: 10.3332/ecancer.2017.781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Wang H., Zhang L. Risk of COVID-19 for patients with cancer. Lancet Oncol. 2020;21(4):e181. doi: 10.1016/S1470-2045(20)30149-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Wu D., Otton S., Sproule B., Busto U., Inaba T., Kalow W., Sellers E. Inhibition of human cytochrome P450 2D6 (CYP2D6) by methadone. Br. J. Clin. Pharmacol. 1993;35(1):30–34. doi: 10.1111/j.1365-2125.1993.tb05666.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Xu C., Djebli N., Kanamaluru V., Bridgewater D.S. Physiologically based pharmacokinetic (PBPK) approach to discern potential population differences in patients with refractory solid tumors and healthy subjects: the effects of fedratinib on CYP3A4 substrate midazolam. J. Pharmacokinet. Pharmacodyn. 2014 SPRINGER/PLENUM PUBLISHERS 233 SPRING ST, NEW YORK, NY 10013 USA, pp. S10-S10. [Google Scholar]
  76. Yang L.P. Abiraterone acetate. Drugs. 2011;71(15):2067–2077. doi: 10.2165/11208080-000000000-00000. [DOI] [PubMed] [Google Scholar]
  77. Zeng Z., Andrew N., Arison B., Luffer-Atlas D., Wang R. Identification of cytochrome P4503A4 as the major enzyme responsible for the metabolism of ivermectin by human liver microsomes. Xenobiotica. 1998;28(3):313–321. doi: 10.1080/004982598239597. [DOI] [PubMed] [Google Scholar]
  78. Zhang X., Brennan B.J. Pharmaceutical Sciences Encyclopedia: Drug Discovery, Development, and Manufacturing; 2010. Disease–Drug–Drug Interaction Assessments for Tocilizumab—A Monoclonal Antibody Against Interleukin‐6 Receptor to Treat Patients with Rheumatoid Arthritis; pp. 1–16. [Google Scholar]
  79. Zhang L., Zhang Y.D., Zhao P., Huang S.-M. Predicting drug–drug interactions: an FDA perspective. AAPS J. 2009;11(2):300–306. doi: 10.1208/s12248-009-9106-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Zhou S.-F. Structure, function and regulation of P-glycoprotein and its clinical relevance in drug disposition. Xenobiotica. 2008;38(7-8):802–832. doi: 10.1080/00498250701867889. [DOI] [PubMed] [Google Scholar]
  81. Zhou F., Yu T., Du R., Fan G., Liu Y., Liu Z., Xiang J., Wang Y., Song B., Gu X. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020 doi: 10.1016/S0140-6736(20)30566-3. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Critical Reviews in Oncology/Hematology are provided here courtesy of Elsevier

RESOURCES