Potential Interactions between Increased Cytokines in COVID-19 and Drugs Used to Treat COVID-19

During COVID-19 infection, virus and host cell interactions lead to the acute production of very strong immune mediators. The clinical status caused by damage throughout the body is mostly due to excessive pro-inflammatory cytokine production from virus-induced macrophages and granulocytes. Under infectious and inflammatory conditions, clinical and experimental studies have demonstrated that hepatic and extrahepatic cytochrome P450 (CYP) enzymes and carrier proteins responsible for drug metabolism are specifically regulated by many cytokines. Downregulation of these enzymes by cytokines can cause an elevation in plasma drug levels and/or lead to adverse drug reactions and/or toxicity. Based on the knowledge of cytokine-drug interactions occurring in the infection and inflammation stage, the aim of this review was to ascertain the influence of uncontrolled cytokine release on the metabolism of drugs used alone or in combination to treat COVID-19 patients and predict drug-drug interactions causing adverse effects.

COVİD-19 da Sitokinler ve COVİD- 19 Tedavisinde Kullanılan İlaçlar Arasında Potansiyel İlaç Etkileşimleri

COVID-19 enfeksiyonu sırasında virus ve konakçı hücre etkileşimleri çok güçlü immun mediyatörlerin akut üretimine yol açmaktadır. Ağır klinik durum çoğunlukla virüsün indüklediği makrofaj ve granülositlerden aşırı inflammatuar sitokin üretimine bağlıdır. Enfeksiyöz ve inflammatuar koşullar altında, klinik ve deneysel çalışmalar göstermiştir ki, ilaç metabolizmasından sorumlu olan karaciğer ve karaciğer dışı sitokrom P450 (CYP) enzimleri ve taşıyıcı proteinler pek çok sitokinler tarafından spesifik olarak regüle edilmektedirler. Sitokinler tarafından bu enzimlerin downregülasyonu, plazma ilaç düzeylerinde yükselmeye neden olabilir ve/veya advers ilaç reaksiyonlarına ve/veya toksisiteye yol açabilir. İnfeksiyon ve inflammasyon koşullarında oluşan sitokin-ilaç etkileşimleri bilgilerimiz temelinde, bu derlemenin amacı, COVID-19 hastalarını tedavi etmek için tek başına veya kombinasyonla kullanılan ilaçların metabolizmaları üzerine kontrolsüz sitokin salınımının etkisini araştırmak ve advers ilaç etkilerine neden olabilecek ilaç- ilaç etkileşimlerini öngörmektir.

___

1. World Health Organization (2020). Naming the coronavirus disease (COVID-19) and the virus that causes it. https://www.who.int/emergencies/diseases/novel-coronavirus- 2019/technical-guidance/naming-the-coronavirus-disease-(covid- 2019)-and-the-virus-that-causes-it (accessed 21 January 2021).

2. Prajapat, M, Sarma, P, Shekhar, N, et al. Drug targets for corona virus: A systematic review, Indian Journal of Pharmacology 2020; 52: 56–65.

3. Kazak, A, Hintistan, S, Önal, B. COVID-19 Vaccine Development Studies in the World and Turkey, Celal Bayar University-Health Sciences Institute Journal, 2020, 7, 571-575.

4. Wang, F, Nie, J, Wang, H, et al. Characteristics of peripheral lymphocyte subset alteration in COVID-19 pneumonia, The Journal of Infectious Diseases, 2020, 221; 1762-1769.

5. Mehta, P, McAuley, D, F, Brown, M, Sanchez, E, Tattersall, R, S, Manson, J, J, H, L, H, Acros Speciality Collaboration, COVID-19: consider cytokine storm syndromes and İmmunosuppression, The Lancet, 2020, 395, 1034-1035.

6. Li, G, Fan, Y, Lai, Y, et al. Coronavirus infections and immune responses, Journal of Medical Viroogy, 2020, 92, 424-432.

7. Mc Gonagle, D, Sharif, K, O’Regan, A, Brigewood, C, Interleukin- 6 use in COVID-19 pneumonia related macrophage activation syndrome, Autoimmunity Reviews, 2020: 19, 102537.

8. Sen, E, S, Ramanan, A, V, Cytokine Storm Syndrome Associated with Hemorrhagic Fever and Other Viruses, in: R, O, Cron, E, M, Behrens, (Eds), Cytokine Storm Syndrome. Springer Nature Switzerland AG, 2019, 277-298.

9. Huang, C, Wang, Y, Li, X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China, Lancet London England, 2020, 395, 497-506.

10. Li, H, Xiang, X, Ren, H, et al. Serum Amyloid A is a biomarker to distinguish the severity and prognosis of Coronavirus Disease 2019 (COVID-19), The Journal of Infection, 2020, 80, 646-655.

11. Stavropoulou, E, Pircalabioru, G, G, Bezirtzoglou, E. The Role of Cytochromes P450 in infection, Frontiers in Immunology, 2018, 31, 89.

12. Goralski, K, B, Ladda, A, M, O McNeil, J, Drug-Cytokine Interactions, in: Georgiev V, Pai MP, Kiser, J.J, Gubbins, P.O and Rodvold, K.A (Eds), Drug Interactions in Infectious Diseases: Mechanisms and Models of Drug Interactions, Switzerland, Springer International Publishing, 2018, 163-204.

13. Morgan, E, T, Impact of infectious and inflammatory disease on cytochrome P450-mediated drug metabolism and pharmacokinetics. Clinical Pharmacology and Therapeutics, 2009, 85, 434 - 438.

14. Aitken, A.E, Morgan, E.T, Gene-Specific Effects of Inflammatory Cytokines on Cytochrome P4502C, 2B6 and 3A4 mRNA Levels in Human Hepatocytes, Drug Metabolism & Disposition, 2007, 35, 1687-1693.

15. Aitken, A, Richardson, T.A, Morgan, E.T, Regulatıon of drug- metabolızıng enzymes and transporters in inflammation, Annual Review of Pharmacology and Toxicology, 2006, 46, 123–149.

16. Bleau, A, M, Maurel, P, Pichette, V, Interleukin-1beta, interleukin-6, tumour necrosis factor-alpha and interferon-gamma released by a viral infection and an aseptic inflammation reduce YP1A1, 1A2 and 3A6 expression in rabbit hepatocytes. European Journal of Pharmacology, 2003, 473,197-206.

17. Renton, K.W, Cytokines and Pharmacokinetic Drug Interactions. In House RV and Descotes J (Eds): Cytokines in Human Health: Immunotoxicology, Pathology, and Therapeutic Applications, Humana Press Inc., Totowa, NJ, USA, 2007, 275-296.

18. Haas, C, E. Drug–Cytokine Interactions, in: Piscitelli SC and Rodvold KA (Eds), Drug Interactions in Infectious Diseases. Humana Press Inc. Totowa, New Jersey, USA, 2001, pp 287-310.

19. Dickmann, L.J, Patel, S.K, Rock, D.A, Wienkers, L.C, Slatter, J.G, Effects of interleukin-6 (IL-6) and an anti-IL-6monoclonal antibody on drug-metabolizing enzymes in human hepatocyte culture, Drug Metabolism & Disposition, 2011, 39, 1415–1422.

20. Rendic, S, Guengerich, F.P, Update information on drug metabolism systems-2009, part II: summary of information on the effects of diseases and environmental factors on human cytochrome P450 (CYP) enzymes and trans-porters, Current Drug Metabolism, 2010, 11, 4–84.

21. Schmitt, C, Kuhn, B, Zhang, X, Kivitz, A.J, Grange, S, Disease– Drug–Drug Interaction Involving Tocilizumab and Simvastatin in Patients With Rheumatoid Arthritis, Clinical Pharmacology & Therapeutics, 2011, 89, 735-740.

22. Vial, T, Descotes, J. Clinical Adverse Effects of Cytokines on the Immune System, in: House RV, Descotes J. (Eds), Cytokines in Human Health: Immunotoxicology, Pathology, and Therapeutic Applications, Humana Press Inc., Totowa, NJ, USA, 2007, 319-349.

23.. Siddiqi, H.K, Mehra, M.R, COVID-19 illness in native and immunosuppressed states: A clinical−therapeutic staging proposal, Journal of Heart and Lung Transplantation, 39 (2020) 405-407.

24. Mahmudpour, M, Roozbeh, J, Keshavarz, M, Farrokhi, S, Nabipour, I. COVID-19 cytokine storm: The anger of inflammation, Cytokine, 2020, 133, 1551512.

25. Chen, Z, Hu, J, Zhang, Z, et al. Efficacy of hydroxychloroquine in patients with COVID-19: results of a randomized clinical trial, medRxiv. 2020, https://www.medrxiv.org/content/10.1101/2020.03.22.20040758v3

26. Gautret, P, Lagier, J.C, Parola, P, et al, Hydroxychloroquine and azithromycin as a treatment of COVID-19: results of an open-label non-randomized clinical trial, International Journal of Antimicrobial Agents, 2020, 56, 105949.

27. Molina, J.M, Delaugerre, C, Le, Goff J, Mela-Lima, B, Ponscarme, D, Goldwirt, L, de Castro N, No evidence of rapid antiviral clearance or clinical benefit with the combination of hydroxychloroquine and azithromycin in patients with severe COVID-19 infection, Médecine et Maladies Infectieuses, 2020, 50, 384.

28. Zhang, G, Hu, C, Luo, L, Fang, F, Chen, Y, Li, J, et al. Clinical features and outcomes of 221 patients with COVID-19 in Wuhan, China, medRxiv, https://www.medrxiv.org/content/10.1101/2020.03.02.20030452v1

29. Wang, Y, Zhang, D, Du, G, Du, R, Zhao, J, Jin, Y, et al, Remdesivir in adults with severe COVID-19: a randomised, double-blind, placebo-controlled, multicentre trial, The Lancet, 2020, 395, 1569- 1578.

30. Chen, C, Zhang, Y, Huang, J, Yin, P, Cheng, Z, Wu, J, et al, Favipiravir versus Arbidol for COVID-19: A Randomized Clinical Trial, medRxiv. https://www.medrxiv.org/content/10.1101/2020.03.17.20037432v4

31. Costanzo, M, de Giglio,, M, Roviello G.N, SARS-CoV-2: Recent Reports on Antiviral Therapies Based on Lopinavir/Ritonavir, Darunavir/Umifenovir, Hydroxychloroquine, Remdesivir, Favipiravir and Other Drugs for the Treatment of the New Coronavirus, Current Medicinal Chemistry, 2020, 27, 4536-4541.

32. Gattinoni, L, Chiumello, D, Caironi, P, Busana, M, Romitti, F, Brazzi, L, Camporota, L, COVID-19 pneumonia: different respiratory treatment for different phenotypes? Intensive Care Medicine, 2020, 46,1099-1102.

33. COVID-19 Treatment Guidelines Panel. Coronavirus Disease 2019 (COVID-19) Treatment Guidelines. National Institutes of Health, https://covid19treatmentguidelines.nih.gov/. (Last updated July 30, 2020).

34. COVID-19 Adult Treatment Guidelines. Coronavirus Disease 2019 (COVID-19). https://covid19bilgi.saglik.gov.tr/depo/algoritmalar/COVID19- PLKACILHASTAYONETIMI.pdf.

35. Russell, B, Moss, C, George, G, Santaolalla, A, Cope A, Papa S, Van Hemelrijck M, Associations between immune-suppressive and stimulating drugs and novel COVID-19—a systematic review of current evidence, ecancer. 2020, 14, 1022.

36. Yang, Z, Liu, J, Zhou, Y, Zhao, X, Zhao, Q, Liu J, The effect of corticosteroid treatment on patients with coronavirus infection: a systematic review and meta-analysis, Journal of Infection, 81 (2020) e13-e20.

37. Wang, Y, Jiang, W, He, Q, Wang, C, Liu, B, Zhou, P, Dong, N, Tong, Q, Early, low-dose and short-term application of corticosteroid treatment in patients with severe COVID-19 pneumonia: single- center experience from Wuhan, China. MedRxiv, 2020.

38. Fadel, R, Morrison, AR, Vahia, A, et al. COVID-19 Management Task Force, Early Short Course Corticosteroids in Hospitalized Patients with COVID-19, Clinical Infectious Diseases 2020, 71(16), 2114–2120.

39. Fu, B, Xu, X, Wei, H, Why tocilizumab could be an effective treatment for severe COVID‐19, Journal of Translational Medicine, 2020, 18, 164.

40. Ye, Q, Wang, B, Mao, J. The pathogenesis and treatment of the ‘Cytokine Storm’ in COVID-19, Journal of Infection, 2020, 80, 607- 613.

41. Feldmann, M, Maini, R.N, Woody, J, N, et al, Trials of anti-tumour necrosis factor therapy for COVID-19 are urgently needed, The Lancet, 2020, 395, 1407-1409.

42. Jamilloux, Y, Henry, T, Belot, A, et al. Should we stimulate or suppress immune responses in COVID-19? Cytokine and anti- cytokine interventions, Autoimmunity Reviews, 2020, 19, 102567.

43. Chorin, E, Dai, M, Shulman, E, et al, The QT interval in patients with COVID-19 treated with hydroxychloroquine and azithromycin, medRxiv 2020.04.02.20047050.

44. Borba, M, G, S, Val, F, F, A, Sampaio, V, S, et al, Effect of high vs low doses of chloroquine diphosphate as adjunctive therapy for patients hospitalized with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection: a randomized clinical trial, JAMA, 2020, 3, e208857.

45. Magagnoli, J, Narendran, S, Pereira, F, et al., Outcomes of hydroxychloroquine usage in United States veterans hospitalized with Covid-19, medRxiv, 2020.

46. Kany, S, Vollrath, J.T, Relja, B. Cytokines in Inflammatory Disease, The International Journal of Molecular Sciences, 2019, 20, 6008.

47. Cressman, A.M, Petrovic, V, Piquette-Miller, M. Inflammation- mediated changes in drug transporter expression/activity: implications for therapeutic drug response, Expert Review of Clinical Pharmacology, 2012, 5, 69- 89.

48. Petrovic, V, Teng, S, Piquette-Miller, M. Regulation of drug transporters during infection and inflammation, Molecular Intervention 2007, 7, 99- 111.

49. Heemskerk, S, Peters, J.G.P, Louisse, J, Sagar, S, Russel, F.G.M, Masereeuw, R. Regulation of P-glycoprotein in renal proximal tubule epithelial cells by LPS and TNF-alpha, Journal of Biomedical and Biotechnology, 2010, 525180.

50. Bezirtzoglou, E, Intestinal cytochromes P450 regulating the intestinal micro-biota and its probiotic profile, Microbial Ecology in Health Disease, 2012, 23, 1–10.

51. Clark, E.S, Jones, B.C, Human cytochromes P450 and their role in metabolism based drug-drug interaction, in: Rodrigues A, D (Ed), Drug–Drug Interactions: Drugs and the Pharmaceutical Sciences, Marcel Dekker, New York, USA, 2002, pp. 55–88.

52. Chang, K.C, Bell, T.D, Lauer, B.A, Chai, H. Altered Theophylline Pharmacokinetics During Acute Respiratory Viral Illness, The Lancet 1978, 27, 1132-1133.

53. Renton, KW, Knickle, LC. Regulation of cytochrome P450 during infectious disease. Can Journal of Physiology and Pharmacology, 1990, 68, 777-781.

54. Overton, C, L, Hudder, A, Novak, R, F. The CYP2E Subfamily, in: Ioannides C (Ed), Cytochromes P450. Role in the Metabolism and Toxicity of Drugs and other Xenobiotics. RSC Publishing, Cambridge, UK, 2008, pp. 282.

55. Zhou, J, Li, F. Potential pharmacokinetic interactions of therapeutic cytokines or cytokine modulators on small-molecule drugs: mechanistic understanding via studies using in vitro systems. Drug Metabolism and Drug Interactions, 2014, 29, 17–28.

56. Tufan, A, Avanoğlu, Güler, A, Matuccı-Cerınıc, M. COVID-19, immune system response, hyper inflammation and repurposing antirheumatic drugs. Turkish Journal of Medical Science, 2020, 50, 620-632.

57. Sanaee, F, Clements, J, D, Waugh, A, W, G,. Fedorak, R, N, Lewanczuk, R, Jamali, F. Drug-disease interaction: Crohn’s disease elevates verapamil plasma concentrations but reduces response to the drug proportional to disease activity, British Journal of Clinical Pharmacology, 2011, 72, 787-797.

58. Tapner, M, Liddle, C, Goodwin, B, George, J, Farrell, G,C. Interferon gamma down-regulates cytochrome P450 3A genes in primary cultures of well-differentiated rat hepatocytes, Hepatology, 1996, 24, 367-373.

59. Ashino, T, Arima, Y, Shioda, S, Iwakura, Y, Numazawa, S, Yoshida, T. Effect of interleukin-6 neutralization on cyp3a11 and metallothionein-1/2 expressions in arthritic mouse liver, European Journal of Pharmacology, 2007, 558, 199-207.

60. Frye, R, F, Zgheib, N, K, Matzke, G, R, et al, Liver disease selectively modulates cytochrome P450--mediated metabolism, Clinical Pharmacology and Therapeutics, 2006, 80, 235-245.

61. Muntanı, J, Longo, V, Mitjavila, M, T, Gervası, P.G, Ingelman- Sundberg M. Effect of carrageenan-induced granuloma on hepatic cytochrome P-450 isozymes in rats, Inflammation, 1995, 19, 143- 156.

62. Getachew, Y, James, L, Lee, W.M, Thiele, D.L, Miller B.C, Susceptibility to acetamino-phen (APAP) toxicity unexpectedly is decreased during acute viral hepatitis in mice, Biochemical Pharmacology, 2010, 79, 1363-1371.

63. Kraemer, M.J, Furukawa C.T, Koup J.R, Shapiro, G.G, Pierson, W, E, Bierman, C.W, Altered Theophylline Clearance During an Influenza B Outbreak, Pediatrics, 1982, 69, 476-80.

64. Liu, J, Cao, R, Xu, M, et al, Hydroxychloroquine, a less toxic derivative of chloroquine, is effective in inhibiting SARS-CoV-2 infection in vitro, Cell Discovery, 2020, 6, 16.

65. Keyaerts, E, Vijgen, L, Maes, P, Neyts, J, van Ranst, M, In vitro inhibition of severe acute respiratory syndrome coronavirus by chloroquine, Biochemical and Biophysical Research Communications, 2004, 323, 264-268.

66. Yavuz, S.Ş, Ünal, S, Antiviral treatment of COVID-19, Turkish Journal of Medical Science, 2020, 50, 611-619.

67. Albertson, T.E, Chloroquine and Other Aminoquinolines, in: Olson KR (Ed), Poisoning & Drug Overdose. McGraw-Hill Education, 2018, 194-196.

68. Lee, J.Y, Vinayagamoorthy, N, Han, K, et al, Association of polymorphisms of cytochrome P450 2D6 with blood hydroxychloroquine levels in patients with systemic lupus erythematosus. Arthritis and Rhematology, 2016, 68, 184-190.

69. Wisniowska, B, Tylutki, Z, Wyszogrodzka, G, Polak, S. Drug-drug interactions and QT prolongation as a commonly assessed cardiac effect- comprehensive overview of clinical trials, BMC Pharmacology and Toxicology, 2016, 17, 12.

70. Fohnera, A.E, Sparreboomb, A, Altmana, R.B, Klein, T.E, Pharm GKB summary: macrolide antibiotic pathway, pharmacokinetics/pharmacodynamics, Pharmacogenet Genomics. 2017, 27, 164-167.

71. Faragon, J.J, Jehan, Z, Budak, J.Z, Drug Interactions with Antiretroviral Medications. © National HIV in Curriculum Section 3: Antiretroviral Therapy, https://www.hiv.uw.edu/go/antiretroviral- therapy/drug-drug-interactions/core-concept/all.

72. Chinello, P, Petrosillo, N, Pittalis, S, Biava, G, Ippolito, G, Nicastri, E, on behalf of the INMI Ebola Team, QTc interval prolongation during favipiravir therapy in an Ebolavirus-infected patient, PLOS Neglected Tropical Diseases, 2017, 11(12), e0006034.

73. https://www.micromedexsolutions.com/micromedex2/librarian Accessed July 2, 2020

74. Brown, K.C, Paul, S, Kashuba, A.D, Drug interactions with new and investigational antiretrovirals, Clinical Pharmacokinetics, 2009, 48, 211–241.

75. Yang, B, B, Baughman, S, Sullivan, J.T, Pharmacokinetics of anakinra in subjects with different levels of renal function, International Journal of Clinical Pharmacology and Therapeutics, 2003, 74, 85-94.

76. Pelkonen, O, Turpeinen, M, Hakkola, J, Honkakoski, P, Hukkanen, J, Raunio, H. Inhibition and induction of human cytochrome P450 enzymes: current status, Archives of Toxicology, 2008, 82, 667–715.

77. Zhang, X, Peck, R. Clinical pharmacology of tocilizumab for the treatment of patients with rheumatoid arthritis. Expert Review of Clinical Pharmacolology, 2011, 4, 539-558.

78. Clarivet, B, Robin, P, Pers, Y.M, et al, Tocilizumab and mesenteric arterial thrombosis: drug-drug interaction with anticoagulants metabolized by CYP 450 and/or by P-glycoprotein, European Journal of Clinical Pharmacology, 2016, 72, 1413-1414.

79. Kim, S, Östör, A.J.K, Nisar, M.K, Interleukin-6 and cytochrome- P450, reason for concern? Rheumatolology International, 2012, 32, 2601-2604.

80. Zhang, X, Schmitt, C, Grange, S, et al. Disease-drug interaction studies of tocilizumab with cytochrome P450 substrates in vitro and in vivo. Clinical Pharmacolology and Therapeutics, 2009, 85, S59.

81. Papamichael, K, van Stappen, T, Jairath V, et al, Review article: Pharmacological aspects of anti-TNF biosimilars in inflammatory bowel diseases, Alimentary Pharmacology and Therapeutics, 2015, 42, 1158-1169.

82. Danese, S, Sans, M, Scaldaferri, F, et al, TNF-alpha blockade down- regulates the CD40/CD40L pathway in the mucosal microcirculation: a novel anti-inflammatory mechanism of infliximab in Crohn's disease, The Journal of Immunology, 2006, 176, 2617-2624.

83. Lee, J.I, Zhang, L, Men, A.Y, Kenna, L.A, Huang, S.M, CYP- Mediated Therapeutic Protein-Drug Interactions Clinical Findings, Proposed Mechanisms and Regulatory Implications, Clinical Pharmacokinetics, 2010, 49, 295-310.

84. Dinarello, C.A, Novick, D, Kim, S. Interleukin-18 and IL-18 binding protein, The Frontiers in Immunology, 2013, 4, 289.

85. Gabay, C, Fautrel, B, Rech, J, et al. Open-label, multicentre, dose- escalating phase II clinical trial on the safety and efficacy of tadekinig alfa (IL-18BP) in adult-onset Still’s disease, Annals of Rheumatologic Disease, 2018, 77, 840-847.

86. Sanders, J.M, Monogue, M.L, Jodlowski, T.Z, Cutrell J.B, Pharmacologic Treatments for Coronavirus Disease, 2019, (COVID- 19), A Review, JAMA 2020, 323, 1824-1836.

87. Zhou, Q, Chen, V, Shannon, C.P, et al, Interferon-a2b Treatment for COVID-19, The Frontiers in Immunology, 11, 1061.

88. Caocci, G, La Nasa, G, Could ruxolitinib be effective in patients with COVID-19 infection at risk of acute respiratory distress syndrome (ARDS)? Annals of Hematology, 2020, 14, 1-2.

89. Stebbing, J, Phelan, A, Griffin, I, Tucker, C, Oechsle, O, Smith, D, Richardson, P. COVID-19: combining antiviral and anti- inflammatory treatments, Lancet Infectious Diseases, 2020, 20, 400- 402.

90. Russell, C.D, Millar, J.E, Baillie, J.K, Clinical evidence does not support corticosteroid treatment for 2019-nCoV lung injury, The Lancet, 2020, 395, 473-475.

91. Low-cost dexamethasone reduces death by up to one third in hospitalised patients with severe respiratory complications of COVID-19. Published 16 June, 2020. https://www.ox.ac.uk/news/2020-06-16-low-cost-dexamethasone- reduces-death-one-third-hospitalised-patients-severe

92. Daveluy, A, Raignoux, C, Miremont-Salamé G, et al, Drug interactions between inhaled corticosteroids and enzymatic inhibitors, European Journal of Clinical Pharmacology, 2009, 65, 743-745.

93. Features of 20 133 UK patients in hospital with covid-19 using the ISARIC WHO Clinical Characterisation Protocol: prospective observational cohort study, British Medical Journal, 2020, 369, m1985
Celal Bayar Üniversitesi Sağlık Bilimleri Enstitüsü Dergisi-Cover
  • ISSN: 2147-9607
  • Yayın Aralığı: Yılda 4 Sayı
  • Başlangıç: 2014
  • Yayıncı: Manisa Celal Bayar Üniversitesi Sağlık Bilimleri Enstitüsü