Complementary medicines used in ulcerative colitis and unintended interactions with cytochrome P450-dependent drug-metabolizing enzymes

Yıl: 2022 Cilt: 52 Sayı: 5 Sayfa Aralığı: 1425 - 1447 Metin Dili: İngilizce DOI: 10.55730/1300-0144.5482 İndeks Tarihi: 27-12-2022

Complementary medicines used in ulcerative colitis and unintended interactions with cytochrome P450-dependent drug-metabolizing enzymes

Öz:
Ulcerative colitis (UC) is an idiopathic, chronic inflammatory disease with multiple genetic and a variety of environmental risk factors. Although current drugs significantly aid in controlling the disease, many people have led to the application of complementary therapies due to the common belief that they are natural and safe, as well as due to the consideration of the side effect of current drugs. Curcumin, cannabinoids, wheatgrass, Boswellia, wormwood and Aloe vera are among the most commonly used complementary medicines in UC. However, these treatments may have adverse and toxic effects due to unintended interactions with drugs or drug- metabolizing enzymes such as cytochrome P450s; thus, being ignorant of these interactions might cause deleterious effects with severe consequences. In addition, the lack of complete and controlled long-term studies with the use of these complementary medicines regarding drug metabolism pose additional risk and unsafety. Thus, this review aims to give an overview of the potential interactions of drug-metabolizing enzymes with the complementary botanical medicines used in UC, drawing attention to possible adverse effects.
Anahtar Kelime: Cytochrome P450 CYPs ulcerative colitis complementary medicines herbal products adverse effect

Belge Türü: Makale Makale Türü: Derleme Erişim Türü: Erişime Açık
  • 1. Szigethy E, McLafferty L, Goyal A. Inflammatory bowel disease. Child and Adolescent Psychiatric Clinics of North America 2010; 19(2): 301-318, doi: 10.1016/j.chc.2010.01.007 pmid: 20478501
  • 2. Kobayashi T, Siegmund B, Le Berre C, Wei SC, Ferrante M, Shen B, Bernstein CN, Danese S, Peyrin-Biroulet L, Hibi T. Ulcerative colitis. Nature Reviews Disease Primers 2020; 6(1): 74, doi: 10.1038/s41572-020-0205-x pmid: 32913180
  • 3. Hanauer SB. Inflammatory bowel disease: epidemiology, pathogenesis, and therapeutic opportunities. Inflammatory Bowel Diseases 2006; 12 Suppl 1: S3-9, doi: 10.1097/01. mib.0000195385.19268.68 pmid: 16378007
  • 4. Zhang YZ, Li YY. Inflammatory bowel disease: pathogenesis. World Journal of Gastroenterology 2014; 20(1): 91-99, doi: 10.3748/wjg.v20.i1.91 pmid: 24415861
  • 5. Chang JT. Pathophysiology of Inflammatory Bowel Diseases. The New England Journal of Medicine 2020; 383(27): 2652- 2664, doi: 10.1056/NEJMra2002697 pmid: 33382932
  • 6. Harbord M, Eliakim R, Bettenworth D, Karmiris K, Katsanos K, Kopylov U, Kucharzik T, Molnar T, Raine T, Sebastian S, de Sousa HT, Dignass A, Carbonnel F, European Cs, Colitis O. Third European Evidence-based Consensus on Diagnosis and Management of Ulcerative Colitis. Part 2: Current Management. Journal of Crohn’s and Colitis 2017; 11(7): 769- 784, doi: 10.1093/ecco-jcc/jjx009 pmid: 28513805
  • 7. Matsuoka K, Kobayashi T, Ueno F, Matsui T, Hirai F, Inoue N, Kato J, Kobayashi K, Kobayashi K, Koganei K, Kunisaki R, Motoya S, Nagahori M, Nakase H, Omata F, Saruta M, Watanabe T, Tanaka T, Kanai T, Noguchi Y, Takahashi KI, Watanabe K, Hibi T, Suzuki Y, Watanabe M, Sugano K, Shimosegawa T. Evidence-based clinical practice guidelines for inflammatory bowel disease. Journal of Gastroenterology 2018; 53(3): 305- 353, doi: 10.1007/s00535-018-1439-1 pmid: 29429045
  • 8. Cohen RD, Yu AP, Wu EQ, Xie J, Mulani PM, Chao J. Systematic review: the costs of ulcerative colitis in Western countries. Alimentary Pharmacology and Therapeutics 2010; 31(7): 693- 707, doi: 10.1111/j.1365-2036.2010.04234.x pmid: 20064142
  • 9. Null KD, Xu Y, Pasquale MK, Su C, Marren A, Harnett J, Mardekian J, Manuchehri A, Healey P. Ulcerative Colitis Treatment Patterns and Cost of Care. Value Health 2017; 20(6): 752-761, doi: 10.1016/j.jval.2017.02.005 pmid: 28577692
  • 10. Park KT, Ehrlich OG, Allen JI, Meadows P, Szigethy EM, Henrichsen K, Kim SC, Lawton RC, Murphy SM, Regueiro M, Rubin DT, Engel-Nitz NM, Heller CA. The Cost of Inflammatory Bowel Disease: An Initiative From the Crohn’s & Colitis Foundation. Inflammatory Bowel Diseases 2020; 26(1): 1-10, doi: 10.1093/ibd/izz104 pmid: 31112238
  • 11. Alulis S, Vadstrup K, Olsen J, Jorgensen TR, Qvist N, Munkholm P, Borsi A. The cost burden of Crohn’s disease and ulcerative colitis depending on biologic treatment status - a Danish register-based study. BMC Health Services Research 2021; 21(1): 836, doi: 10.1186/s12913-021-06816-3 pmid: 34407821
  • 12. Hilsden RJ, Verhoef MJ, Rasmussen H, Porcino A, DeBruyn JC. Use of complementary and alternative medicine by patients with inflammatory bowel disease. Inflammatory Bowel Diseases 2011; 17(2): 655-662, doi: 10.1002/ibd.21360 pmid: 20848543
  • 13. Ke F, Yadav PK, Ju LZ. Herbal medicine in the treatment of ulcerative colitis. Saudi Journal of Gastroenterology 2012; 18(1): 3-10, doi: 10.4103/1319-3767.91726 pmid: 22249085
  • 14. Ng SC, Lam YT, Tsoi KK, Chan FK, Sung JJ, Wu JC. Systematic review: the efficacy of herbal therapy in inflammatory bowel disease. Alimentary Pharmacology and Therapeutics 2013; 38(8): 854-863, doi: 10.1111/apt.12464 pmid: 23981095
  • 15. Langhorst J, Wulfert H, Lauche R, Klose P, Cramer H, Dobos GJ, Korzenik J. Systematic review of complementary and alternative medicine treatments in inflammatory bowel diseases. Journal of Crohn’s and Colitis 2015; 9(1): 86-106, doi: 10.1093/ecco-jcc/jju007 pmid: 25518050
  • 16. Akkol EK, Karpuz B, Sobarzo-Sanchez E, Khan H. A phytopharmacological overview of medicinal plants used for prophylactic and treatment of colitis. Food and Chemical Toxicology 2020; 144: 111628, doi: 10.1016/j.fct.2020.111628 pmid: 32738379
  • 17. Dubey V, Singh S. Alternative Herbal Medicines For Prevention And Treatment Of Ulcerative Colitis. European Journal of Molecular and Clinical Medicine 2020; 7(7): 4900-4907
  • 18. Picardo S, Altuwaijri M, Devlin SM, Seow CH. Complementary and alternative medications in the management of inflammatory bowel disease. Therapeutic Advances in Gastroenterology 2020; 13: 1756284820927550, doi: 10.1177/1756284820927550 pmid: 32523629
  • 19. Miller EC. Some current perspectives on chemical carcinogenesis in humans and experimental animals: Presidential Address. Cancer Research 1978; 38(6): 1479-1496, pmid: 348302
  • 20. Guengerich FP. Roles of cytochrome P-450 enzymes in chemical carcinogenesis and cancer chemotherapy. Cancer Research 1988; 48(11): 2946-2954, pmid: 3284639
  • 21. Guengerich FP, Kim DH. In vitro inhibition of dihydropyridine oxidationandaflatoxinB1activationinhumanlivermicrosomes by naringenin and other flavonoids. Carcinogenesis 1990; 11(12): 2275-2279, doi: 10.1093/carcin/11.12.2275 pmid: 2265479
  • 22. Arinc E, Sen A. Effects of in vivo benzene treatment on cytochrome-p450 and mixed-function oxidase activities of gilthead seabream (Sparus-aurata) liver-microsomes. Comparative Biochemistry and Physiology Part C: Toxicology and Pharmacology 1993; 104(1): 61-65, doi: 10.1016/0742- 8413(93)90112-x
  • 23. Arinc E, Sen A. In-vivo effects of the anesthetic, benzocaine, on liver microsomal cytochrome-P450 and mixed- function oxidase activities of gilthead seabream (Sparus- aurata). Comparative Biochemistry and Physiology Part C: Pharmacology, Toxicology and Endocrinology 1994; 107(3): 399-404, doi: 10.1016/1367-8280(94)90068-x
  • 24. Ozkarsli M, Sevim H, Sen A. In vivo effects of Urtica urens (dwarf nettle) on the expression of CYP1A in control and 3-methylcholanthrene-exposed rats. Xenobiotica 2008; 38(1): 48-61, doi: 10.1080/00498250701713968 pmid: 18098063
  • 25. Gibson GG, Skett P. Pharmacological and toxicological aspects of drug metabolism. In: Gibson GG, Skett P, editors. Introduction to Drug Metabolism. Boston, MA: Springer US, 1986: 175-198
  • 26. Sen A, Arinc E. Purification and Characterization of Cytochrome P450 Reductase from Liver Microsomes of Feral Leaping Mullet (Liza saliens). Journal of Biochemical and Molecular Toxicology 1998; 12(2): 103-113, doi: 10.1002/ (sici)1099-0461(1998)12:2<103::Aid-jbt5>3.0.Co;2-p pmid: 9443067
  • 27. Guengerich FP, Meanwell N, Georg GI, Wang S. Introduction: Drug Metabolism and Toxicology Special Issue. Journal of Medicinal Chemistry 2020; 63(12): 6249-6250, doi: 10.1021/ acs.jmedchem.0c00874 pmid: 32484351
  • 28. Nelson DR. The cytochrome p450 homepage. Human Genomics 2009; 4(1): 59-65, doi: 10.1186/1479-7364-4-1-59 pmid: 19951895
  • 29. Omura T. Structural diversity of cytochrome P450 enzyme system. Journal of Biochemistry 2010; 147(3): 297-306, doi: 10.1093/jb/mvq001 pmid: 20068028
  • 30. Gaedigk A, Ingelman-Sundberg M, Miller NA, Leeder JS, Whirl- Carrillo M, Klein TE, PharmVar Steering C. The Pharmacogene Variation (PharmVar) Consortium: Incorporation of the Human Cytochrome P450 (CYP) Allele Nomenclature Database. Clinical Pharmacology and Therapeutics 2018; 103(3): 399-401, doi: 10.1002/cpt.910 pmid: 29134625
  • 31. Buyukgoze O, Osmanoglu N, Arslan S, Sen A. Association of the CYP1A1*2A, GSTT1 null, GSTM1 null, mEPHX*3, and XRCC1-399 genetic polymorphisms with ulcerative colitis. International Journal of Colorectal Disease 2013; 28(4): 593- 595, doi: 10.1007/s00384-012-1507-6 pmid: 22664944
  • 32. Sen A, Stark H. Role of cytochrome P450 polymorphisms and functions in development of ulcerative colitis. World Journal of Gastroenterology 2019; 25(23): 2846-2862, doi: 10.3748/wjg. v25.i23.2846 pmid: 31249444
  • 33. Radha MH, Laxmipriya NP. Evaluation of biological properties and clinical effectiveness of Aloe vera: A systematic review. Journal of Traditional and Complementary Medicine 2015; 5(1): 21-26, doi: 10.1016/j.jtcme.2014.10.006 pmid: 26151005
  • 34. Akaberi M, Sobhani Z, Javadi B, Sahebkar A, Emami SA. Therapeutic effects of Aloe spp. in traditional and modern medicine: A review. Biomedicine and Pharmacotherapy 2016; 84: 759-772, doi: 10.1016/j.biopha.2016.09.096 pmid: 27716590
  • 35. Bendjedid S, Lekmine S, Tadjine A, Djelloul R, Bensouici C. Analysis of phytochemical constituents, antibacterial, antioxidant, photoprotective activities and cytotoxic effect of leaves extracts and fractions of Aloe vera. Biocatalysis and Agricultural Biotechnology 2021; 33, doi: 10.1016/j. bcab.2021.101991
  • 36. Lucini L, Pellizzoni M, Pellegrino R, Molinari GP, Colla G. Phytochemical constituents and in vitro radical scavenging activity of different Aloe species. Food Chemistry 2015; 170: 501-507, doi: 10.1016/j.foodchem.2014.08.034 pmid: 25306376
  • 37. Sun YN, Li W, Yang SY, Kang JS, Ma JY, Kim YH. Isolation and identification of chromone and pyrone constituents from Aloe and their anti-inflammatory activities. Journal of Functional Foods 2016; 21: 232-239, doi: 10.1016/j.jff.2015.12.026
  • 38. Mpiana PT, Ngbolua KT, Tshibangu DST, Kilembe JT, Gbolo BZ, Mwanangombo DT, Inkoto CL, Lengbiye EM, Mbadiko CM, Matondo A, Bongo GN, Tshilanda DD. Identification of potential inhibitors of SARS-CoV-2 main protease from Aloe vera compounds: A molecular docking study. Chemical Physics Letters 2020; 754: 137751, doi: 10.1016/j.cplett.2020.137751 pmid: 33518775
  • 39. Amoo SO, Aremu AO, Van Staden J. Unraveling the medicinal potential of South African Aloe species. Journal of Ethnopharmacology 2014; 153(1): 19-41, doi: 10.1016/j. jep.2014.01.036 pmid: 24509153
  • 40. Cardarelli M, Rouphael Y, Pellizzoni M, Colla G, Lucini L. Profile of bioactive secondary metabolites and antioxidant capacity of leaf exudates from eighteen Aloe species. Industrial Crops and Products 2017; 108: 44-51, doi: 10.1016/j. indcrop.2017.06.017
  • 41. Muller SO, Eckert I, Lutz WK, Stopper H. Genotoxicity of the laxative drug components emodin, aloe-emodin and danthron in mammalian cells: topoisomerase II mediated? Mutation Research 1996; 371(3-4): 165-173, doi: 10.1016/s0165- 1218(96)90105-6 pmid: 9008718
  • 42. Choi S, Chung M-H. A review on the relationship between aloe vera components and their biologic effects. Seminars in Integrative Medicine 2003; 1(1): 53-62, doi: 10.1016/s1543- 1150(03)00005-x
  • 43. Yang HN, Kim DJ, Kim YM, Kim BH, Sohn KM, Choi MJ, Choi YH. Aloe-induced toxic hepatitis. Journal of Korean Medical Science 2010; 25(3): 492-495, doi: 10.3346/jkms.2010.25.3.492 pmid: 20191055
  • 44. Kumar R, Singh AK, Gupta A, Bishayee A, Pandey AK. Therapeutic potential of Aloe vera-A miracle gift of nature. Phytomedicine 2019; 60: 152996, doi: 10.1016/j. phymed.2019.152996 pmid: 31272819
  • 45. Cui Y, Cheng Y, Guo Y, Xie Y, Yao W, Zhang W, Qian H. Evaluating the hepatoprotective efficacy of Aloe vera polysaccharides against subchronic exposure of aflatoxins B 1. Journal of the Taiwan Institute of Chemical Engineers 2017; 76: 10-17, doi: 10.1016/j.jtice.2017.03.040
  • 46. Djuv A, Nilsen OG. Aloe vera juice: IC(5)(0) and dual mechanistic inhibition of CYP3A4 and CYP2D6. Phytotherapy Research 2012; 26(3): 445-451, doi: 10.1002/ptr.3564 pmid: 21842479
  • 47. Yang MS, Yu CP, Huang CY, Chao PL, Lin SP, Hou YC. Aloe activated P-glycoprotein and CYP 3A: a study on the serum kinetics of aloe and its interaction with cyclosporine in rats. Food and Function 2017; 8(1): 315-322, doi: 10.1039/ c6fo00938g pmid: 28009901
  • 48. Ingelman-Sundberg M. Genetic polymorphisms of cytochrome P450 2D6 (CYP2D6): clinical consequences, evolutionary aspects and functional diversity. The Pharmacogenomics Journal 2005; 5(1): 6-13, doi: 10.1038/sj.tpj.6500285 pmid: 15492763
  • 49. Lynch T, Price A. The effect of cytochrome P450 metabolism on drug response, interactions, and adverse effects. American Family Physician 2007; 76(3): 391-396, pmid: 17708140
  • 50. Sultana B, Anwar F. Flavonols (kaempeferol, quercetin, myricetin) contents of selected fruits, vegetables and medicinal plants. Food Chemistry 2008; 108(3): 879-884, doi: 10.1016/j. foodchem.2007.11.053 pmid: 26065748
  • 51. He N, Edeki T. The inhibitory effects of herbal components on CYP2C9 and CYP3A4 catalytic activities in human liver microsomes. American Journal of Therapeutics 2004; 11(3): 206-212, doi: 10.1097/00045391-200405000-00009 pmid: 15133536
  • 52. Gougis P, Hilmi M, Geraud A, Mir O, Funck-Brentano C. Potential cytochrome P450-mediated pharmacokinetic interactions between herbs, food, and dietary supplements and cancer treatments. Critical Reviews in Oncology/Hematology 2021; 166: 103342, doi: 10.1016/j.critrevonc.2021.103342 pmid: 33930533
  • 53. Zhang YX, Li JS, Peng WW, Liu X, Yang GM, Chen LH, Cai BC. Comparative pharmacokinetics of aloe-emodin, rhein and emodin determined by liquid chromatography-mass spectrometry after oral administration of a rhubarb peony decoction and rhubarb extract to rats. Pharmazie 2013; 68(5): 333-339, pmid: 23802430
  • 54. Sridhar J, Liu J, Foroozesh M, Klein Stevens CL. Inhibition of cytochrome p450 enzymes by quinones and anthraquinones. Chemical Research in Toxicology 2012; 25(2): 357-365, doi: 10.1021/tx2004163 pmid: 22185593
  • 55. Liu Y, Mapa MST, Sprando RL. Anthraquinones inhibit cytochromes P450 enzyme activity in silico and in vitro. Journal of Applied Toxicology 2021; 41(9): 1438-1445, doi: 10.1002/jat.4134 pmid: 33438235
  • 56. Jiang W, Tian X, Wang Y, Sun Z, Dong P, Wang C, Huo X, Zhang B, Huang S, Deng S, Wang X, Ma X. The natural anthraquinones from Rheum palmatum induced the metabolic disorder of melatonin by inhibiting human CYP and SULT enzymes. Toxicology Letters 2016; 262: 27-38, doi: 10.1016/j. toxlet.2016.09.004 pmid: 27633141
  • 57. Wang X, Xin X, Sun Y, Zou L, Li H, Zhao Y, Li R, Peng Y, Zheng J. Chemical Reactivity of Aloe-Emodin and Its Hydroxylation Metabolites to Thiols. Chemical Research in Toxicology 2019; 32(2): 234-244, doi: 10.1021/acs.chemrestox.8b00248 pmid: 30672276
  • 58. Fei J, Peng Y, Tan H, Chen X, Yang J, Li J. Study on the electrochemical behavior and differential pulse voltammetric determination of rhein using a nanoparticle composite film- modified electrode. Bioelectrochemistry 2007; 70(2): 369-374, doi: 10.1016/j.bioelechem.2006.05.007 pmid: 16820331
  • 59. Tang JC, Yang H, Song XY, Song XH, Yan SL, Shao JQ, Zhang TL, Zhang JN. Inhibition of cytochrome P450 enzymes by rhein in rat liver microsomes. Phytotherapy Research 2009; 23(2): 159-164, doi: 10.1002/ptr.2572 pmid: 18814214
  • 60. Singh RP, Dhanalakshmi S, Rao AR. Chemomodulatory action of Aloe vera on the profiles of enzymes associated with carcinogen metabolism and antioxidant status regulation in mice. Phytomedicine 2000; 7(3): 209-219, doi: 10.1016/S0944- 7113(00)80006-9 pmid: 11185732
  • 61. Lin SY, Yang JH, Hsia TC, Lee JH, Chiu TH, Wei YH, Chung JG. Effect of inhibition of aloe-emodin on N-acetyltransferase activity and gene expression in human malignant melanoma cells (A375.S2). Melanoma Research 2005; 15(6): 489-494, doi: 10.1097/00008390-200512000-00002 pmid: 16314733
  • 62. Chung JG, Li YC, Lee YM, Lin JP, Cheng KC, Chang WC. Aloe-emodin inhibited N-acetylation and DNA adduct of 2-aminofluorene and arylamine N-acetyltransferase gene expression in mouse leukemia L 1210 cells. Leukemia Research 2003; 27(9): 831-840, doi: 10.1016/s0145-2126(03)00017-1 pmid: 12804642
  • 63. Hassan A, Othman Z, Siriphanich J. Pineapple (Ananas comosus L. Merr.). In: Yahia EM, editor Postharvest Biology and Technology of Tropical and Subtropical Fruits: Woodhead Publishing, 2011: 194-218e
  • 64. Nascimento RPD, Moya A, Machado A, Geraldi MV, Diez- Echave P, Vezza T, Galvez J, Cazarin CBB, Marostica Junior MR. Review on the potential application of non-phenolic compounds from native Latin American food byproducts in inflammatory bowel diseases. Food Research International 2021; 139: 109796, doi: 10.1016/j.foodres.2020.109796 pmid: 33509446
  • 65. Thomson AB, Keelan M, Thiesen A, Clandinin MT, Ropeleski M, Wild GE. Small bowel review: normal physiology part 1. Digestive Diseases and Sciences 2001; 46(12): 2567-2587, doi: 10.1023/a:1012794505897 pmid: 11768247
  • 66. de Lencastre Novaes LC, Jozala AF, Lopes AM, de Carvalho Santos-Ebinuma V, Mazzola PG, Pessoa Junior A. Stability, purification, and applications of bromelain: A review. Biotechnology Progress 2016; 32(1): 5-13, doi: 10.1002/ btpr.2190 pmid: 26518672
  • 67. Blackwood AD, Salter J, Dettmar PW, Chaplin MF. Dietary fibre, physicochemical properties and their relationship to health. The Journal of the Royal Society for the Promotion of Health 2000; 120(4): 242-247, doi: 10.1177/146642400012000412 pmid: 11197452
  • 68. Hale LP, Greer PK, Trinh CT, Gottfried MR. Treatment with oral bromelain decreases colonic inflammation in the IL- 10-deficient murine model of inflammatory bowel disease. Clinical Immunology 2005; 116(2): 135-142, doi: 10.1016/j. clim.2005.04.011 pmid: 15936249
  • 69. Bhui K, Prasad S, George J, Shukla Y. Bromelain inhibits COX- 2 expression by blocking the activation of MAPK regulated NF- kappa B against skin tumor-initiation triggering mitochondrial death pathway. Cancer Letters 2009; 282(2): 167-176, doi: 10.1016/j.canlet.2009.03.003 pmid: 19339108
  • 70. Zhou Z, Wang L, Feng P, Yin L, Wang C, Zhi S, Dong J, Wang J, Lin Y, Chen D, Xiong Y, Peng J. Inhibition of Epithelial TNF- alpha Receptors by Purified Fruit Bromelain Ameliorates Intestinal Inflammation and Barrier Dysfunction in Colitis. Frontiers in Immunology 2017; 8: 1468, doi: 10.3389/ fimmu.2017.01468 pmid: 29176974
  • 71. Li KY, Wang JL, Wei JP, Gao SY, Zhang YY, Wang LT, Liu G. Fecal microbiota in pouchitis and ulcerative colitis. World Journal of Gastroenterology 2016; 22(40): 8929-8939, doi: 10.3748/wjg.v22.i40.8929 pmid: 27833384
  • 72. Hidaka M, Nagata M, Kawano Y, Sekiya H, Kai H, Yamasaki K, Okumura M, Arimori K. Inhibitory effects of fruit juices on cytochrome P450 2C9 activity in vitro. Bioscience, Biotechnology, and Biochemistry 2008; 72(2): 406-411, doi: 10.1271/bbb.70511 pmid: 18256496
  • 73. Petric Z, Zuntar I, Putnik P, Bursac Kovacevic D. Food-Drug Interactions with Fruit Juices. Foods 2020; 10(1), doi: 10.3390/ foods10010033 pmid: 33374399
  • 74. Helmy SA, El-Bedaiwy HM, El-Masry SM. Effect of pineapple juice on the pharmacokinetics of celecoxib and montelukast in humans. Therapeutic Delivery 2020; 11(5): 301-311, doi: 10.4155/tde-2020-0020 pmid: 32507029
  • 75. Paulson SK, Vaughn MB, Jessen SM, Lawal Y, Gresk CJ, Yan B, Maziasz TJ, Cook CS, Karim A. Pharmacokinetics of celecoxib after oral administration in dogs and humans: effect of food and site of absorption. Journal of Pharmacology and Experimental Therapeutics 2001; 297(2): 638-645, pmid: 11303053
  • 76. Werner U, Werner D, Rau T, Fromm MF, Hinz B, Brune K. Celecoxib inhibits metabolism of cytochrome P450 2D6 substrate metoprolol in humans. Clinical Pharmacology and Therapeutics 2003; 74(2): 130-137, doi: 10.1016/S0009- 9236(03)00120-6 pmid: 12891223
  • 77. Yanagawa Y, Sawada M, Deguchi T, Gonzalez FJ, Kamataki T. Stable expression of human CYP1A2 and N-acetyltransferases in Chinese hamster CHL cells: mutagenic activation of 2-amino-3-methylimidazo4,5-fquinoline and 2-amino-3,8- dimethylimidazo4,5-fquinoxaline. Cancer Research 1994; 54(13): 3422-3427, pmid: 8012961
  • 78. Platt KL, Edenharder R, Aderhold S, Muckel E, Glatt H. Fruits and vegetables protect against the genotoxicity of heterocyclic aromatic amines activated by human xenobiotic-metabolizing enzymes expressed in immortal mammalian cells. Mutation Research/Genetic Toxicology and Environmental Mutagenesis 2010; 703(2): 90-98, doi: 10.1016/j.mrgentox.2010.08.007 pmid: 20713174
  • 79. Tang T, Targan SR, Li ZS, Xu C, Byers VS, Sandborn WJ. Randomised clinical trial: herbal extract HMPL-004 in active ulcerative colitis - a double-blind comparison with sustained release mesalazine. Alimentary Pharmacology and Therapeutics 2011; 33(2): 194-202, doi: 10.1111/j.1365- 2036.2010.04515.x pmid: 21114791
  • 80. Sandborn WJ, Targan SR, Byers VS, Rutty DA, Mu H, Zhang X, Tang T. Andrographis paniculata extract (HMPL-004) for active ulcerative colitis. The American Journal of Gastroenterology 2013; 108(1): 90-98, doi: 10.1038/ajg.2012.340 pmid: 23044768
  • 81. Zhang L, Cao N, Wang Y, Wang Y, Wu C, Cheng X, Wang C. Improvement of Oxazolone-Induced Ulcerative Colitis in Rats Using Andrographolide. Molecules 2019; 25(1), doi: 10.3390/ molecules25010076 pmid: 31878303
  • 82. Vetvicka V, Vannucci L. Biological properties of andrographolide, an active ingredient of Andrographis Paniculata: a narrative review. Annals of Translational Medicine 2021; 9(14): 1186, doi: 10.21037/atm-20-7830 pmid: 34430627
  • 83. George J, Goodwin B, Liddle C, Tapner M, Farrell GC. Time- dependent expression of cytochrome P450 genes in primary cultures of well-differentiated human hepatocytes. Journal of Laboratory and Clinical Medicine 1997; 129(6): 638-648, doi: 10.1016/s0022-2143(97)90199-2 pmid: 9178731
  • 84. Liddle C, Goodwin BJ, George J, Tapner M, Farrell GC. Separate and interactive regulation of cytochrome P450 3A4 by triiodothyronine, dexamethasone, and growth hormone in cultured hepatocytes. The Journal of Clinical Endocrinology and Metabolism 1998; 83(7): 2411-2416, doi: 10.1210/ jcem.83.7.4877 pmid: 9661620
  • 85. LeCluyse E, Madan A, Hamilton G, Carroll K, DeHaan R, Parkinson A. Expression and regulation of cytochrome P450 enzymes in primary cultures of human hepatocytes. Journal of Biochemical and Molecular Toxicology 2000; 14(4): 177-188, doi: 10.1002/(sici)1099-0461(2000)14:4<177::aid- jbt1>3.0.co;2-4 pmid: 10789495
  • 86. Chatuphonprasert W, Jarukamjorn K, Kondo S, Nemoto N. Synergistic increases of metabolism and oxidation-reduction genes on their expression after combined treatment with a CYP1A inducer and andrographolide. Chemico-Biological Interactions 2009; 182(2-3): 233-238, doi: 10.1016/j. cbi.2009.09.001 pmid: 19737545
  • 87. Ooi JP, Kuroyanagi M, Sulaiman SF, Muhammad TS, Tan ML. Andrographolide and 14-deoxy-11, 12-didehydroandrographolide inhibit cytochrome P450s in HepG2 hepatoma cells. Life Sciences 2011; 88(9-10): 447-454, doi: 10.1016/j.lfs.2010.12.019 pmid: 21219911
  • 88. Tan ML, Lim LE. The effects of Andrographis paniculata (Burm.f.) Nees extract and diterpenoids on the CYP450 isoforms’ activities, a review of possible herb-drug interaction risks. Drug and Chemical Toxicology 2015; 38(3): 241-253, doi: 10.3109/01480545.2014.947504 pmid: 25156015
  • 89. Ashour ML, Youssef FS, Gad HA, Wink M. Inhibition of Cytochrome P450 (CYP3A4) Activity by Extracts from 57 Plants Used in Traditional Chinese Medicine (TCM). Pharmacognosy Magazine 2017; 13(50): 300-308, doi: 10.4103/0973-1296.204561 pmid: 28539725
  • 90. Yen CC, Liu YT, Lin YJ, Yang YC, Chen CC, Yao HT, Chen HW, Lii CK. Bioavailability of the diterpenoid 14-deoxy- 11,12-didehydroandrographolide in rats and up-regulation of hepatic drug-metabolizing enzyme and drug transporter expression. Phytomedicine 2019; 61: 15284,1 doi: 10.1016/j. phymed.2019.152841 pmid: 31035043
  • 91. Jarukamjorn K, Don-in K, Makejaruskul C, Laha T, Daodee S, Pearaksa P, Sripanidkulchai BO. Impact of Andrographis paniculata crude extract on mouse hepatic cytochrome P450 enzymes. Journal of Ethnopharmacology 2006; 105(3): 464- 467, doi: 10.1016/j.jep.2005.11.024 pmid: 16406417
  • 92. Jaruchotikamol A, Jarukamjorn K, Sirisangtrakul W, Sakuma T, Kawasaki Y, Nemoto N. Strong synergistic induction of CYP1A1 expression by andrographolide plus typical CYP1A inducers in mouse hepatocytes. Toxicology and Applied Pharmacology 2007; 224(2): 156-162, doi: 10.1016/j. taap.2007.07.008 pmid: 17825862
  • 93. Pekthong D, Martin H, Abadie C, Bonet A, Heyd B, Mantion G, Richert L. Differential inhibition of rat and human hepatic cytochrome P450 by Andrographis paniculata extract and andrographolide. Journal of Ethnopharmacology 2008; 115(3): 432-440, doi: 10.1016/j.jep.2007.10.013 pmid: 18053665
  • 94. Pekthong D, Blanchard N, Abadie C, Bonet A, Heyd B, Mantion G, Berthelot A, Richert L, Martin H. Effects of Andrographis paniculata extract and Andrographolide on hepatic cytochrome P450 mRNA expression and monooxygenase activities after in vivo administration to rats and in vitro in rat and human hepatocyte cultures. Chemico-Biological Interactions 2009; 179(2-3): 247-255, doi: 10.1016/j.cbi.2008.10.054 pmid: 19041297
  • 95. Chien CF, Wu YT, Lee WC, Lin LC, Tsai TH. Herb- drug interaction of Andrographis paniculata extract and andrographolide on the pharmacokinetics of theophylline in rats. Chemico-Biological Interactions 2010; 184(3): 458-4 65, doi: 10.1016/j.cbi.2010.01.017 pmid: 20096675
  • 96. Muthiah YD, Ong CE, Sulaiman SA, Ismail R. Inhibition of Human Cytochrome P450 2c8-catalyzed Amodiaquine N-desethylation: Effect of Five Traditionally and Commonly Used Herbs. Pharmacognosy Research 2016; 8(4): 292-297, doi: 10.4103/0974-8490.188886 pmid: 27695271
  • 97. Pan Y, Abd-Rashid BA, Ismail Z, Ismail R, Mak JW, Pook PC, Er HM, Ong CE. In vitro modulatory effects of Andrographis paniculata, Centella asiatica and Orthosiphon stamineus on cytochrome P450 2C19 (CYP2C19). Journal of Ethnopharmacology 2011; 133(2): 881-887, doi: 10.1016/j. jep.2010.11.026 pmid: 21093571
  • 98. Kar A, Pandit S, Mukherjee K, Bahadur S, Mukherjee PK. Safety assessment of selected medicinal food plants used in Ayurveda through CYP450 enzyme inhibition study. J Journal of the Science of Food and Agriculture 2017; 97(1): 333-340, doi: 10.1002/jsfa.7739 pmid: 27030208
  • 99. Nigam M, Atanassova M, Mishra AP, Pezzani R, Devkota HP, Plygun S, Salehi B, Setzer WN, Sharifi-Rad J. Bioactive Compounds and Health Benefits ofArtemisiaSpecies. Natural Product Communications 2019; 14(7): 1934578X19850354, doi: 10.1177/1934578x19850354
  • 100. Gilani AH, Janbaz KH. Preventive and curative effects of Artemisia absinthium on acetaminophen and CCl4-induced hepatotoxicity. General Pharmacology 1995; 26(2): 309-315, doi: 10.1016/0306-3623(94)00194-r pmid: 7590079
  • 101. Joshi RK. Volatile composition and antimicrobial activity of the essential oil of Artemisia absinthium growing in Western Ghats region of North West Karnataka, India. Pharmaceutical Biology 2013; 51(7): 888-892, doi: 10.3109/13880209.2013.768676 pmid: 23570523
  • 102. Chung KS, Choi HE, Shin JS, Cho EJ, Cho YW, Choi JH, Baek NI, Lee KT. Chemopreventive effects of standardized ethanol extract from the aerial parts of Artemisia princeps Pampanini cv. Sajabal via NF-kappaB inactivation on colitis- associated colon tumorigenesis in mice. Food and Chemical Toxicology 2015; 75: 14-23, doi: 10.1016/j.fct.2014.11.007 pmid: 25449198
  • 103. Ahamad J, Mir SR, Amin S. A Pharmacognostic Review on Artemisia Absinthium. International Research Journal of Pharmacy 2019; 10(1): 25-31, doi: 10.7897/2230- 8407.10015
  • 104. Batiha GE, Olatunde A, El-Mleeh A, Hetta HF, Al-Rejaie S, Alghamdi S, Zahoor M, Magdy Beshbishy A, Murata T, Zaragoza-Bastida A, Rivero-Perez N. Bioactive Compounds, Pharmacological Actions, and Pharmacokinetics of Wormwood (Artemisia absinthium). Antibiotics (Basel) 2020; 9(6), doi: 10.3390/antibiotics9060353 pmid: 32585887
  • 105. Yan SC, Wang YJ, Li YJ, Cai WY, Weng XG, Li Q, Chen Y, Yang Q, Zhu XX. Dihydroartemisinin Regulates the Th/Treg Balance by Inducing Activated CD4+ T cell Apoptosis via Heme Oxygenase-1 Induction in Mouse Models of Inflammatory Bowel Disease. Molecules 2019; 24(13), doi: 10.3390/ molecules24132475 pmid: 31284478
  • 106. Gunther S, Fagone P, Jalce G, Atanasov AG, Guignabert C, Nicoletti F. Role of MIF and D-DT in immune-inflammatory, autoimmune, and chronic respiratory diseases: from pathogenic factors to therapeutic targets. Drug Discovery Today 2019; 24(2): 428-439, doi: 10.1016/j.drudis.2018.11.003 pmid: 30439447
  • 107. Omer B, Krebs S, Omer H, Noor TO. Steroid-sparing effect of wormwood (Artemisia absinthium) in Crohn’s disease: a double- blind placebo-controlled study. Phytomedicine 2007; 14(2-3): 87-95, doi: 10.1016/j.phymed.2007.01.001 pmid: 17240130
  • 108. Lopes-Lutz D, Alviano DS, Alviano CS, Kolodziejczyk PP. Screening of chemical composition, antimicrobial and antioxidant activities of Artemisia essential oils. Phytochemistry 2008; 69(8): 1732-1738, doi: 10.1016/j.phytochem.2008.02.014 pmid: 18417176
  • 109. Nigam M, Atanassova M, Mishra AP, Pezzani R, Devkota HP, Plygun S, Salehi B, Setzer WN, Sharifi-Rad J. Bioactive Compounds and Health Benefits ofArtemisiaSpecies. Natural Product Communications 2019; 14(7), doi: 10.1177/1934578x19850354
  • 110. Govindarajan M, Benelli G. Artemisia absinthium-borne compounds as novel larvicides: effectiveness against six mosquito vectors and acute toxicity on non-target aquatic organisms. Parasitology Research 2016; 115(12): 4649-4661, doi: 10.1007/s00436-016-5257-1 pmid: 27630101
  • 111. Desrosiers MR, Mittelman A, Weathers PJ. Dried Leaf Artemisia Annua Improves Bioavailability of Artemisinin via Cytochrome P450 Inhibition and Enhances Artemisinin Efficacy Downstream. Biomolecules 2020; 10(2), doi: 10.3390/ biom10020254 pmid: 32046156
  • 112. Xing J, Kirby BJ, Whittington D, Wan Y, Goodlett DR. Evaluation of P450 inhibition and induction by artemisinin antimalarials in human liver microsomes and primary human hepatocytes. Drug Metabolism and Disposition 2012; 40(9): 1757-1764, doi: 10.1124/dmd.112.045765 pmid: 22679214
  • 113. Ericsson T, Masimirembwa C, Abelo A, Ashton M. The evaluation of CYP2B6 inhibition by artemisinin antimalarials in recombinant enzymes and human liver microsomes. Drug Metabolism Letters 2012; 6(4): 247-257, doi: 10.2174/1872312811206040004 pmid: 23506555
  • 114. Ibrahim SRM, Badr JM, Sayed KAE, Youssef DTA. A New Cytotoxic Sesquiterpene and Three Anti-inflammatory Flavonoids from Egyptian Tanacetum Santolinoides. Natural Product Communications 2019; 2(11): 1934578X0700201102, doi: 10.1177/1934578x0700201102
  • 115. Szopa A, Pajor J, Klin P, Rzepiela A, Elansary HO, Al-Mana FA, Mattar MA, Ekiert H. Artemisia absinthium L.-Importance in the History of Medicine, the Latest Advances in Phytochemistry and Therapeutical, Cosmetological and Culinary Uses. Plants (Basel) 2020; 9(9), doi: 10.3390/plants9091063 pmid: 32825178
  • 116. Ji HY, Kim SY, Kim DK, Jeong JH, Lee HS. Effects of eupatilin and jaceosidin on cytochrome p450 enzyme activities in human liver microsomes. Molecules 2010; 15(9): 6466-6475, doi: 10.3390/molecules15096466 pmid: 20877236
  • 117. Melillo de Magalhaes P, Dupont I, Hendrickx A, Joly A, Raas T, Dessy S, Sergent T, Schneider YJ. Anti-inflammatory effect and modulation of cytochrome P450 activities by Artemisia annua tea infusions in human intestinal Caco-2 cells. Food Chemistry 2012; 134(2): 864-871, doi: 10.1016/j.foodchem.2012.02.195 pmid: 23107701
  • 118. Attah AF, Fagbemi AA, Olubiyi O, Dada-Adegbola H, Oluwadotun A, Elujoba A, Babalola CP. Therapeutic Potentials of Antiviral Plants Used in Traditional African Medicine With COVID-19 in Focus: A Nigerian Perspective. Frontiers in Pharmacology 2021; 12: 596855, doi: 10.3389/ fphar.2021.596855 pmid: 33981214
  • 119. Radulovic NS, Gencic MS, Stojanovic NM, Randjelovic PJ, Stojanovic-Radic ZZ, Stojiljkovic NI. Toxic essential oils. Part V: Behaviour modulating and toxic properties of thujones and thujone-containing essential oils of Salvia officinalis L., Artemisia absinthium L., Thuja occidentalis L. and Tanacetum vulgare L. Food and Chemical Toxicology 2017; 105: 355-369, doi: 10.1016/j.fct.2017.04.044 pmid: 28472675
  • 120. Abdel-Tawab M, Werz O, Schubert-Zsilavecz M. Boswellia serrata: an overall assessment of in vitro, preclinical, pharmacokinetic and clinical data. Clinical Pharmacokinetics 2011; 50(6): 349-369, doi: 10.2165/11586800-000000000- 00000 pmid: 21553931
  • 121. Ammon HP. Boswellic Acids and Their Role in Chronic Inflammatory Diseases. Advances in Experimental Medicine and Biology 2016; 928: 291-327, doi: 10.1007/978-3-319- 41334-1_13 pmid: 27671822
  • 122. Efferth T, Oesch F. Anti-inflammatory and anti-cancer activities of frankincense: Targets, treatments and toxicities. Semin Cancer Biol 2020, doi: 10.1016/j.semcancer.2020.01.015 pmid: 32027979
  • 123. Gupta I, Parihar A, Malhotra P, Gupta S, Ludtke R, Safayhi H, Ammon HP. Effects of gum resin of Boswellia serrata in patients with chronic colitis. Planta Medica 2001; 67(5): 391- 395, doi: 10.1055/s-2001-15802 pmid: 11488449
  • 124. Gupta M, Singh N, Gulati M, Gupta R, Sudhakar K, Kapoor B. Herbal bioactives in treatment of inflammation: An overview. South African Journal of Botany 2021; 143: 205-225, doi: 10.1016/j.sajb.2021.07.027
  • 125. Hartmann RM, Fillmann HS, Martins MI, Meurer L, Marroni NP. Boswellia serrata has beneficial anti-inflammatory and antioxidant properties in a model of experimental colitis. Phytotherapy Research 2014; 28(9): 1392-1398, doi: 10.1002/ ptr.5142 pmid: 24619538
  • 126. Holleran G, Scaldaferri F, Gasbarrini A, Curro D. Herbal medicinal products for inflammatory bowel disease: A focus on those assessed in double-blind randomised controlled trials. Phytotherapy Research 2020; 34(1): 77-93, doi: 10.1002/ ptr.6517 pmid: 31701598
  • 127. Dey D, Chaskar S, Athavale N, Chitre D. Acute and chronic toxicity, cytochrome p450 enzyme inhibition, and HERG channel blockade studies with a polyherbal, ayurvedic formulation for inflammation. BioMed Research International 2015; 2015: 971982, doi: 10.1155/2015/971982 pmid: 25893199
  • 128. Frank A, Unger M. Analysis of frankincense from various Boswellia species with inhibitory activity on human drug metabolising cytochrome P450 enzymes using liquid chromatography mass spectrometry after automated on-line extraction. Journal of Chromatography A 2006; 1112(1-2): 255-262, doi: 10.1016/j.chroma.2005.11.116 pmid: 16364338
  • 129. Koeberle A, Henkel A, Verhoff M, Tausch L, Konig S, Fischer D, Kather N, Seitz S, Paul M, Jauch J, Werz O. Triterpene Acids from Frankincense and Semi-Synthetic Derivatives That Inhibit 5-Lipoxygenase and Cathepsin G. Molecules 2018; 23(2), doi: 10.3390/molecules23020506 pmid: 29495286
  • 130. Paoletti A, Gallo E, Benemei S, Vietri M, Lapi F, Volpi R, Menniti-Ippolito F, Gori L, Mugelli A, Firenzuoli F, Vannacci A. Interactions between Natural Health Products and Oral Anticoagulants: Spontaneous Reports in the Italian Surveillance System of Natural Health Products. Evidence- Based Complementary and Alternative Medicine 2011; 2011: 612150, doi: 10.1155/2011/612150 pmid: 21274401
  • 131. Milic N, Milosevic N, Golocorbin Kon S, Bozic T, Abenavoli L, Borrelli F. Warfarin interactions with medicinal herbs. Natural Product Communications 2014; 9(8): 1211-1216, pmid: 25233607
  • 132. Weber CC, Reising K, Muller WE, Schubert-Zsilavecz M, Abdel-Tawab M. Modulation of Pgp function by boswellic acids. Planta Medica 2006; 72(6): 507-513, doi: 10.1055/s- 2006-931536 pmid: 16773534
  • 133. ElSohly MA, Radwan MM, Gul W, Chandra S, Galal A. Phytochemistry of Cannabis sativa L. In: Kinghorn AD, Falk H, Gibbons S, Kobayashi Ji, editors. Phytocannabinoids: Unraveling the Complex Chemistry and Pharmacology of Cannabis sativa. Cham: Springer International Publishing, 2017: 1-36
  • 134. Bonini SA, Premoli M, Tambaro S, Kumar A, Maccarinelli G, Memo M, Mastinu A. Cannabis sativa: A comprehensive ethnopharmacological review of a medicinal plant with a long history. Journal of Ethnopharmacology 2018; 227: 300-315, doi: 10.1016/j.jep.2018.09.004 pmid: 30205181
  • 135. Andre CM, Hausman JF, Guerriero G. Cannabis sativa: The Plant of the Thousand and One Molecules. Frontiers in Plant Sciences 2016; 7: 19, doi: 10.3389/fpls.2016.00019 pmid: 26870049
  • 136. Alves P, Amaral C, Teixeira N, Correia-da-Silva G. Cannabis sativa: Much more beyond Delta(9)-tetrahydrocannabinol. Pharmacological Research 2020; 157: 104822, doi: 10.1016/j. phrs.2020.104822 pmid: 32335286
  • 137. Tibirica E. The multiple functions of the endocannabinoid system: a focus on the regulation of food intake. Diabetology and Metabolic Syndrome 2010; 2: 5, doi: 10.1186/1758-5996-2- 5 pmid: 20180990
  • 138. Schicho R, Storr M. Cannabis finds its way into treatment of Crohn’s disease. Pharmacology 2014; 93(1-2): 1-3, doi: 10.1159/000356512 pmid: 24356243
  • 139. Ahmed W, Katz S. Therapeutic Use of Cannabis in Inflammatory Bowel Disease. Gastroenterology and Hepatology (N Y) 2016; 12(11): 668-679, pmid: 28035196
  • 140. Irving PM, Iqbal T, Nwokolo C, Subramanian S, Bloom S, Prasad N, Hart A, Murray C, Lindsay JO, Taylor A, Barron R, Wright S. A Randomized, Double-blind, Placebo-controlled, Parallel-group, Pilot Study of Cannabidiol-rich Botanical Extract in the Symptomatic Treatment of Ulcerative Colitis. Inflammatory Bowel Diseases 2018; 24(4): 714-724, doi: 10.1093/ibd/izy002 pmid: 29538683
  • 141. Kafil TS, Nguyen TM, MacDonald JK, Chande N. Cannabis for the treatment of ulcerative colitis. The Cochrane Database of Systematic Reviews 2018; 11: CD012954, doi: 10.1002/14651858.CD012954.pub2 pmid: 30406638
  • 142. Carvalho ACA, Souza GA, Marqui SV, Guiguer EL, Araujo AC, Rubira CJ, Goulart RA, Flato UAP, Bueno P, Buchaim RL, Barbalho SM. Cannabis and Canabidinoids on the Inflammatory Bowel Diseases: Going Beyond Misuse. International Journal of Molecular Sciences 2020; 21(8), doi: 10.3390/ijms21082940 pmid: 32331305
  • 143. Perisetti A, Rimu AH, Khan SA, Bansal P, Goyal H. Role of cannabis in inflammatory bowel diseases. Annals of Gastroenterology 2020; 33(2): 134-144, doi: 10.20524/ aog.2020.0452 pmid: 32127734
  • 144. Zendulka O, Dovrtelova G, Noskova K, Turjap M, Sulcova A, Hanus L, Jurica J. Cannabinoids and Cytochrome P450 Interactions. Current Drug Metabolism 2016; 17(3): 206-226, doi: 10.2174/1389200217666151210142051 pmid: 26651971
  • 145. Arnold WR, Weigle AT, Das A. Cross-talk of cannabinoid and endocannabinoid metabolism is mediated via human cardiac CYP2J2. Journal of Inorganic Biochemistry 2018; 184: 88-99, doi: 10.1016/j.jinorgbio.2018.03.016 pmid: 29689453
  • 146. Foster BC, Abramovici H, Harris CS. Cannabis and Cannabinoids: Kinetics and Interactions. The American Journal of Medicine 2019; 132(11): 1266-1270, doi: 10.1016/j. amjmed.2019.05.017 pmid: 31152723
  • 147. Greger J, Bates V, Mechtler L, Gengo F. A Review of Cannabis and Interactions With Anticoagulant and Antiplatelet Agents. The Journal of Clinical Pharmacology 2020; 60(4): 432-438, doi: 10.1002/jcph.1557 pmid: 31724188
  • 148. Amaral Silva D, Pate DW, Clark RD, Davies NM, El-Kadi AOS, Lobenberg R. Phytocannabinoid drug-drug interactions and their clinical implications. Pharmacology and Therapeutics 2020; 215: 107621, doi: 10.1016/j.pharmthera.2020.107621 pmid: 32615127
  • 149. Stout SM, Cimino NM. Exogenous cannabinoids as substrates, inhibitors, and inducers of human drug metabolizing enzymes: a systematic review. Drug Metabolism Reviews 2014; 46(1): 86- 95, doi: 10.3109/03602532.2013.849268 pmid: 24160757
  • 150. Lucas CJ, Galettis P, Schneider J. The pharmacokinetics and the pharmacodynamics of cannabinoids. Br The Journal of Clinical Pharmacology 2018; 84(11): 2477-2482, doi: 10.1111/ bcp.13710 pmid: 30001569
  • 151. Alsherbiny MA, Li CG. Medicinal Cannabis-Potential Drug Interactions. Medicines (Basel) 2018; 6(1), doi: 10.3390/ medicines6010003 pmid: 30583596
  • 152. Vazquez M, Guevara N, Maldonado C, Guido PC, Schaiquevich P. Potential Pharmacokinetic Drug-Drug Interactions between Cannabinoids and Drugs Used for Chronic Pain. BioMed Research International 2020; 2020: 3902740, doi: 10.1155/2020/3902740 pmid: 32855964
  • 153. Doohan PT, Oldfield LD, Arnold JC, Anderson LL. Cannabinoid Interactions with Cytochrome P450 Drug Metabolism: a Full-Spectrum Characterization. The American Association of Pharmaceutical Scientists Journal 2021; 23(4): 91, doi: 10.1208/s12248-021-00616-7 pmid: 34181150
  • 154. Yamaori S, Kushihara M, Yamamoto I, Watanabe K. Characterization of major phytocannabinoids, cannabidiol and cannabinol, as isoform-selective and potent inhibitors of human CYP1 enzymes. Biochemical Pharmacology 2010; 79(11): 1691- 1698, doi: 10.1016/j.bcp.2010.01.028 pmid: 20117100
  • 155. Rong C, Carmona NE, Lee YL, Ragguett RM, Pan Z, Rosenblat JD, Subramaniapillai M, Shekotikhina M, Almatham F, Alageel A, Mansur R, Ho RC, McIntyre RS. Drug-drug interactions as a result of co-administering Delta(9)-THC and CBD with other psychotropic agents. Expert Opinion on Drug Safety 2018; 17(1): 51-54, doi: 10.1080/14740338.2017.1397128 pmid: 29082802
  • 156. Yamaori S, Maeda C, Yamamoto I, Watanabe K. Differential inhibition of human cytochrome P450 2A6 and 2B6 by major phytocannabinoids. Forensic Toxicology 2011; 29(2): 117-124, doi: 10.1007/s11419-011-0112-7
  • 157. Yamaori S, Ebisawa J, Okushima Y, Yamamoto I, Watanabe K. Potent inhibition of human cytochrome P450 3A isoforms by cannabidiol: role of phenolic hydroxyl groups in the resorcinol moiety. Life Sciences 2011; 88(15-16): 730-736, doi: 10.1016/j. lfs.2011.02.017 pmid: 21356216
  • 158. Jiang R, Yamaori S, Okamoto Y, Yamamoto I, Watanabe K. Cannabidiol is a potent inhibitor of the catalytic activity of cytochrome P450 2C19. Drug Metabolism and Pharmacokinetics 2013; 28(4): 332-338, doi: 10.2133/dmpk. dmpk-12-rg-129 pmid: 23318708
  • 159. Yamaori S, Koeda K, Kushihara M, Hada Y, Yamamoto I, Watanabe K. Comparison in the in vitro inhibitory effects of major phytocannabinoids and polycyclic aromatic hydrocarbons contained in marijuana smoke on cytochrome P450 2C9 activity. Drug Metabolism and Pharmacokinetics 2012; 27(3): 294-300, doi: 10.2133/dmpk.dmpk-11-rg-107 pmid: 22166891
  • 160. Damkier P, Lassen D, Christensen MMH, Madsen KG, Hellfritzsch M, Pottegard A. Interaction between warfarin and cannabis. Basic and Clinical Pharmacology and Toxicology 2019; 124(1): 28-31, doi: 10.1111/bcpt.13152 pmid: 30326170
  • 161. Yamaori S, Okamoto Y, Yamamoto I, Watanabe K. Cannabidiol, a major phytocannabinoid, as a potent atypical inhibitor for CYP2D6. Drug Metabolism and Disposition 2011; 39(11): 2049-2056, doi: 10.1124/dmd.111.041384 pmid: 21821735
  • 162. Ammon HP, Wahl MA. Pharmacology of Curcuma longa. Planta Medica 1991; 57(1): 1-7 doi: 10.1055/s-2006-960004 pmid: 2062949
  • 163. Aggarwal BB, Sundaram C, Malani N, Ichikawa H. Curcumin: the Indian solid gold. Advances in Experimental Medicine and Biology 2007; 595: 1-75, doi: 10.1007/978-0-387-46401-5_1 pmid: 17569205
  • 164. Aggarwal BB, Harikumar KB. Potential therapeutic effects of curcumin, the anti-inflammatory agent, against neurodegenerative, cardiovascular, pulmonary, metabolic, autoimmune and neoplastic diseases. The International Journal of Biochemistry and Cell Biology 2009; 41(1): 40-59, doi: 10.1016/j.biocel.2008.06.010 pmid: 18662800
  • 165. Kunnumakkara AB, Bordoloi D, Padmavathi G, Monisha J, Roy NK, Prasad S, Aggarwal BB. Curcumin, the golden nutraceutical: multitargeting for multiple chronic diseases. British Journal of Pharmacology 2017; 174(11): 1325-1348, doi: 10.1111/bph.13621 pmid: 27638428
  • 166. Kocaadam B, Sanlier N. Curcumin, an active component of turmeric (Curcuma longa), and its effects on health. Critical Reviews in Food Science and Nutrition 2017; 57(13): 2889- 2895, doi: 10.1080/10408398.2015.1077195 pmid: 26528921
  • 167. Andrew R, Izzo AA. Principles of pharmacological research of nutraceuticals. British Journal of Pharmacology 2017; 174(11): 1177-1194, doi: 10.1111/bph.13779 pmid: 28500635
  • 168. Goel A, Kunnumakkara AB, Aggarwal BB. Curcumin as “Curecumin”: from kitchen to clinic. Biochemical Pharmacology 2008; 75(4): 787-809, doi: 10.1016/j. bcp.2007.08.016 pmid: 17900536
  • 169. Dosoky NS, Setzer WN. Chemical Composition and Biological Activities of Essential Oils of Curcuma Species. Nutrients 2018; 10(9), doi: 10.3390/nu10091196 pmid: 30200410
  • 170. Soleimani V, Sahebkar A, Hosseinzadeh H. Turmeric (Curcuma longa) and its major constituent (curcumin) as nontoxic and safe substances: Review. Phytotherapy Research 2018; 32(6): 985-995, doi: 10.1002/ptr.6054 pmid: 29480523 ŞEN / Turk J Med Sci1443
  • 171. Ayati Z, Ramezani M, Amiri MS, Moghadam AT, Rahimi H, Abdollahzade A, Sahebkar A, Emami SA. Ethnobotany, Phytochemistry and Traditional Uses of Curcuma spp. and Pharmacological Profile of Two Important Species (C. longa and C. zedoaria): A Review. Current Pharmaceutical Design 2019; 25(8): 871-935, doi: 10.2174/13816128256661904021639 40 pmid: 30947655
  • 172. Sharifi-Rad J, Rayess YE, Rizk AA, Sadaka C, Zgheib R, Zam W, Sestito S, Rapposelli S, Neffe-Skocinska K, Zielinska D, Salehi B, Setzer WN, Dosoky NS, Taheri Y, El Beyrouthy M, Martorell M, Ostrander EA, Suleria HAR, Cho WC, Maroyi A, Martins N. Turmeric and Its Major Compound Curcumin on Health: Bioactive Effects and Safety Profiles for Food, Pharmaceutical, Biotechnological and Medicinal Applications. Frontiers in Pharmacology 2020; 11: 01021, doi: 10.3389/fphar.2020.01021 pmid: 33041781
  • 173. Ahmad RS, Hussain MB, Sultan MT, Arshad MS, Waheed M, Shariati MA, Plygun S, Hashempur MH. Biochemistry, Safety, Pharmacological Activities, and Clinical Applications of Turmeric: A Mechanistic Review. Evidence-Based Complementary and Alternative Medicine 2020; 2020: 7656919, doi: 10.1155/2020/7656919 pmid: 32454872
  • 174. Abd El-Hack ME, El-Saadony MT, Swelum AA, Arif M, Abo Ghanima MM, Shukry M, Noreldin A, Taha AE, El-Tarabily KA. Curcumin, the active substance of turmeric: its effects on health and ways to improve its bioavailability. Journal of the Science of Food and Agriculture 2021; 101(14): 5747-5762, doi: 10.1002/jsfa.11372 pmid: 34143894
  • 175. Quispe C, Cruz-Martins N, Manca ML, Manconi M, Sytar O, Hudz N, Shanaida M, Kumar M, Taheri Y, Martorell M, Sharifi-Rad J, Pintus G, Cho WC. Nano- Derived Therapeutic Formulations with Curcumin in Inflammation-Related Diseases. Oxidative Medicine and Cellular Longevity 2021; 2021: 3149223, doi: 10.1155/2021/3149223 pmid: 34584616
  • 176. Holt PR, Katz S, Kirshoff R. Curcumin therapy in inflammatory bowel disease: a pilot study. Digestive Diseases and Sciences 2005; 50(11): 2191-2193, doi: 10.1007/s10620-005-3032-8 pmid: 16240238
  • 177. Hanai H, Iida T, Takeuchi K, Watanabe F, Maruyama Y, Andoh A, Tsujikawa T, Fujiyama Y, Mitsuyama K, Sata M, Yamada M, Iwaoka Y, Kanke K, Hiraishi H, Hirayama K, Arai H, Yoshii S, Uchijima M, Nagata T, Koide Y. Curcumin maintenance therapy for ulcerative colitis: randomized, multicenter, double- blind, placebo-controlled trial. Clinical and Gastroenterology & Hepatology 2006; 4(12): 1502-1506, doi: 10.1016/j. cgh.2006.08.008 pmid: 17101300
  • 178. Singla V, Pratap Mouli V, Garg SK, Rai T, Choudhury BN, Verma P, Deb R, Tiwari V, Rohatgi S, Dhingra R, Kedia S, Sharma PK, Makharia G, Ahuja V. Induction with NCB-02 (curcumin) enema for mild-to-moderate distal ulcerative colitis - a randomized, placebo-controlled, pilot study. Journal of Crohn’s and Colitis 2014; 8(3): 208-214, doi: 10.1016/j. crohns.2013.08.006 pmid: 24011514
  • 179. Lang A, Salomon N, Wu JC, Kopylov U, Lahat A, Har-Noy O, Ching JY, Cheong PK, Avidan B, Gamus D, Kaimakliotis I, Eliakim R, Ng SC, Ben-Horin S. Curcumin in Combination With Mesalamine Induces Remission in Patients With Mild- to-Moderate Ulcerative Colitis in a Randomized Controlled Trial. Clinical Gastroenterology and Hepatology 2015; 13(8): 1444-1449, doi: 10.1016/j.cgh.2015.02.019 pmid: 25724700
  • 180. Kedia S, Bhatia V, Thareja S, Garg S, Mouli VP, Bopanna S, Tiwari V, Makharia G, Ahuja V. Low dose oral curcumin is not effective in induction of remission in mild to moderate ulcerative colitis: Results from a randomized double blind placebo controlled trial. World J Gastrointest Pharmacology and Therapeutics 2017; 8(2): 147-154, doi: 10.4292/wjgpt. v8.i2.147 pmid: 28533925
  • 181. Price RJ, Scott MP, Giddings AM, Walters DG, Stierum RH, Meredith C, Lake BG. Effect of butylated hydroxytoluene, curcumin, propyl gallate and thiabendazole on cytochrome P450 forms in cultured human hepatocytes. Xenobiotica 2008; 38(6): 574-586, doi: 10.1080/00498250802008615 pmid: 18570159
  • 182. Volak LP, Ghirmai S, Cashman JR, Court MH. Curcuminoids inhibit multiple human cytochromes P450, UDP- glucuronosyltransferase, and sulfotransferase enzymes, whereas piperine is a relatively selective CYP3A4 inhibitor. Drug Metabolism and Disposition 2008; 36(8): 1594-1605, doi: 10.1124/dmd.108.020552 pmid: 18480186
  • 183. Liu AC, Zhao LX, Xing J, Liu T, Du FY, Lou HX. Pre-treatment with curcumin enhances plasma concentrations of losartan and its metabolite EXP3174 in rats. Biological and Pharmaceutical Bulletin 2012; 35(2): 145-150, doi: 10.1248/bpb.35.145 pmid: 22293343
  • 184. Koe XF, Tengku Muhammad TS, Chong AS, Wahab HA, Tan ML. Cytochrome P450 induction properties of food and herbal-derived compounds using a novel multiplex RT-qPCR in vitro assay, a drug-food interaction prediction tool. Food Science & Nutrition 2014; 2(5): 500-520, doi: 10.1002/fsn3.122 pmid: 25473508
  • 185. Liju VB, Jeena K, Kuttan R. Chemopreventive activity of turmeric essential oil and possible mechanisms of action. Asian Pacific Journal of Cancer Prevention 2014; 15(16): 6575-6580, doi: 10.7314/apjcp.2014.15.16.6575 pmid: 25169490
  • 186. Bahramsoltani R, Rahimi R, Farzaei MH. Pharmacokinetic interactions of curcuminoids with conventional drugs: A review. Journal of Ethnopharmacology 2017; 209: 1-12, doi: 10.1016/j.jep.2017.07.022 pmid: 28734960
  • 187. Sun DX, Fang ZZ, Zhang YY, Cao YF, Yang L, Yin J. Inhibitory effects of curcumenol on human liver cytochrome P450 enzymes. Phytotherapy Research 2010; 24(8): 1213-1216, doi: 10.1002/ptr.3102 pmid: 20148399
  • 188. Jinno H, Tanaka-Kagawa T, Ohno A, Makino Y, Matsushima E, Hanioka N, Ando M. Functional characterization of cytochrome P450 2B6 allelic variants. Drug Metabolism and Disposition 2003; 31(4): 398-403, doi: 10.1124/dmd.31.4.398 pmid: 12642465
  • 189. Appiah-Opong R, Commandeur JN, van Vugt-Lussenburg B, Vermeulen NP. Inhibition of human recombinant cytochrome P450s by curcumin and curcumin decomposition products. Toxicology 2007; 235(1-2): 83-91, doi: 10.1016/j. tox.2007.03.007 pmid: 17433521
  • 190. Kim SB, Cho SS, Cho HJ, Yoon IS. Modulation of Hepatic Cytochrome P450 Enzymes by Curcumin and its Pharmacokinetic Consequences in Sprague-dawley Rats. Pharmacognosy Magazine 2015; 11(Suppl 4): S580-584, doi: 10.4103/0973-1296.172965 pmid: 27013798
  • 191. Rodriguez Castano P, Parween S, Pandey AV. Bioactivity of Curcumin on the Cytochrome P450 Enzymes of the Steroidogenic Pathway. International Journal of Molecular Sciences 2019; 20(18), doi: 10.3390/ijms20184606 pmid: 31533365
  • 192. Bamba Y, Yun YS, Kunugi A, Inoue H. Compounds isolated from Curcuma aromatica Salisb. inhibit human P450 enzymes. Journal of Natural Medicines 2011; 65(3-4): 583-587, doi: 10.1007/s11418-011-0507-0 pmid: 21287405
  • 193. Liu Q, Dang DS, Chen YF, Yan M, Shi GB, Zhao QC. The influence of omeprazole on platelet inhibition of clopidogrel in various CYP2C19 mutant alleles. Genetic Testing and Molecular Biomarkers 2012; 16(11): 1293-1297, doi: 10.1089/ gtmb.2012.0119 pmid: 23046071
  • 194. Holmes MV, Perel P, Shah T, Hingorani AD, Casas JP. CYP2C19 genotype, clopidogrel metabolism, platelet function, and cardiovascular events: a systematic review and meta- analysis. The Journal of the American Medical Association 2011; 306(24): 2704-2714, doi: 10.1001/jama.2011.1880 pmid: 22203539
  • 195. Jarrar YB, Lee SJ. Molecular functionality of CYP2C9 polymorphisms and their influence on drug therapy. Drug Metabolism and Drug Interactions 2014; 29(4): 211-220, doi: 10.1515/dmdi-2014-0001 pmid: 24825094
  • 196. Hou XL, Takahashi K, Kinoshita N, Qiu F, Tanaka K, Komatsu K, Takahashi K, Azuma J. Possible inhibitory mechanism of Curcuma drugs on CYP3A4 in 1alpha,25 dihydroxyvitamin D3 treated Caco-2 cells. International Journal of Pharmaceutics 2007; 337(1-2): 169-177, doi: 10.1016/j.ijpharm.2006.12.035 pmid: 17270371
  • 197. Zhang W, Lim LY. Effects of spice constituents on P-glycoprotein-mediated transport and CYP3A4-mediated metabolism in vitro. Drug Metabolism and Disposition 2008; 36(7): 1283-1290, doi: 10.1124/dmd.107.019737 pmid: 18385293
  • 198. Hsieh YW, Huang CY, Yang SY, Peng YH, Yu CP, Chao PD, Hou YC. Oral intake of curcumin markedly activated CYP 3A4: in vivo and ex-vivo studies. Scientific Reports 2014; 4: 6587, doi: 10.1038/srep06587 pmid: 25300360
  • 199. Mohajeri M, Behnam B, Cicero AFG, Sahebkar A. Protective effects of curcumin against aflatoxicosis: A comprehensive review. Journal of Cellular Physiology 2018; 233(4): 3552-3577, doi: 10.1002/jcp.26212 pmid: 29034472
  • 200. Raucy JL. Regulation of CYP3A4 expression in human hepatocytes by pharmaceuticals and natural products. Drug Metabolism and Disposition 2003; 31(5): 533-539, doi: 10.1124/dmd.31.5.533 pmid: 12695340
  • 201. Graber-Maier A, Buter KB, Aeschlimann J, Bittel C, Kreuter M, Drewe J, Gutmann H. Effects of Curcuma extracts and curcuminoids on expression of P-glycoprotein and cytochrome P450 3A4 in the intestinal cell culture model LS180. Planta Medica 2010; 76(16): 1866-1870, doi: 10.1055/s-0030-1249980 pmid: 20509107
  • 202. Seah TC, Tay YL, Tan HK, Muhammad TS, Wahab HA, Tan ML. Determination of CYP3A4 Inducing Properties of Compounds Using a Laboratory-Developed Cell-Based Assay. International Journal of Toxicology 2015; 34(5): 454-468, doi: 10.1177/1091581815599335 pmid: 26268769
  • 203. Lee SJ, Hedstrom OR, Fischer K, Wang-Buhler JL, Sen A, Cok I, Buhler DR. Immunohistochemical localization and differential expression of cytochrome P450 3A27 in the gastrointestinal tract of rainbow trout. Toxicology and Applied Pharmacology 2001; 177(2): 94-102, doi: 10.1006/taap.2001.9289 pmid: 11740908
  • 204. Agus HH, Tekin P, Bayav M, Semiz A, Sen A. Drug Interaction Potential of the Seed Extract of Urtica urens L. (Dwarf Nettle). Phytotherapy Research 2009; 23(12): 1763-1770, doi: 10.1002/ ptr.2848 pmid: 19441062
  • 205. Wang D, Sadee W. CYP3A4 intronic SNP rs35599367 (CYP3A4*22) alters RNA splicing. Pharmacogenetics and Genomics 2016; 26(1): 40-43, doi: 10.1097/ FPC.0000000000000183 pmid: 26488616
  • 206. Kato M, Chiba K, Hisaka A, Ishigami M, Kayama M, Mizuno N, Nagata Y, Takakuwa S, Tsukamoto Y, Ueda K, Kusuhara H, Ito K, Sugiyama Y. The intestinal first-pass metabolism of substrates of CYP3A4 and P-glycoprotein-quantitative analysis based on information from the literature. Drug Metabolism and Pharmacokinetics 2003; 18(6): 365-372, doi: 10.2133/ dmpk.18.365 pmid: 15618757
  • 207. Chen J, Huang Z, Chen Z, Yuan M, Liu Y, Zhu Y. The effect of carbon on surface quality of solid-state-sintered silicon carbide as optical materials. Materials Characterization 2014; 89: 7-12, doi: 10.1016/j.matchar.2013.12.005
  • 208. Celik G, Akca H, Sen A. Investigation of aromotase inhibition by several dietary vegetables in human non-small cell lung cancer cell lines. Turkish Journal of Biochemistry 2013; 38(2): 207-217, doi: 10.5505/tjb.2013.62634 pmid: 17067750
  • 209. Srivilai J, Rabgay K, Khorana N, Waranuch N, Nuengchamnong N, Ingkaninan K. A new label-free screen for steroid 5alpha- reductase inhibitors using LC-MS. Steroids 2016; 116: 67-75, doi: 10.1016/j.steroids.2016.10.007 pmid: 27789379
  • 210. Pimkaew P, Kublbeck J, Petsalo A, Jukka J, Suksamrarn A, Juvonen R, Auriola S, Piyachaturawat P, Honkakoski P. Interactions of sesquiterpenes zederone and germacrone with the human cytochrome P450 system. Toxicology in Vitro 2013; 27(6): 2005-2012, doi: 10.1016/j.tiv.2013.07.004 pmid: 23850985
  • 211. Araki Y, Andoh A, Fujiyama Y, Kanauchi O, Takenaka K, Higuchi A, Bamba T. Germinated barley foodstuff exhibits different adsorption properties for hydrophilic versus hydrophobic bile acids. Digestion 2001; 64(4): 248-254, doi: 10.1159/000048869 pmid: 11842282
  • 212. Bamba T, Kanauchi O, Andoh A, Fujiyama Y. A new prebiotic from germinated barley for nutraceutical treatment of ulcerative colitis. Journal of Gastroenterology and Hepatology 2002; 17(8): 818-824, doi: 10.1046/j.1440-1746.2002.02709.x pmid: 12164955
  • 213. Kanauchi O, Suga T, Tochihara M, Hibi T, Naganuma M, Homma T, Asakura H, Nakano H, Takahama K, Fujiyama Y, Andoh A, Shimoyama T, Hida N, Haruma K, Koga H, Mitsuyama K, Sata M, Fukuda M, Kojima A, Bamba T. Treatment of ulcerative colitis by feeding with germinated barley foodstuff: first report of a multicenter open control trial. Journal of Gastroenterology 2002; 37 Suppl 14(14): 67-72, doi: 10.1007/BF03326417 pmid: 12572869
  • 214. Hanai H, Kanauchi O, Mitsuyama K, Andoh A, Takeuchi K, Takayuki I, Araki Y, Fujiyama Y, Toyonaga A, Sata M, Kojima A, Fukuda M, Bamba T. Germinated barley foodstuff prolongs remission in patients with ulcerative colitis. International Journal of Molecular Medicine 2004; 13(5): 643-647, pmid: 15067363
  • 215 Mitsuyama K, Saiki T, Kanauchi O, Iwanaga T, Tomiyasu N, Nishiyama T, Tateishi H, Shirachi A, Ide M, Suzuki A, Noguchi K, Ikeda H, Toyonaga A, Sata M. Treatment of ulcerative colitis with germinated barley foodstuff feeding: a pilot study. Alimentary Pharmacology and Therapeutics 1998; 12(12): 1225-1230, doi: 10.1046/j.1365-2036.1998.00432.x pmid: 9882030
  • 216 Kanauchi O, Oshima T, Andoh A, Shioya M, Mitsuyama K. Germinated barley foodstuff ameliorates inflammation in mice with colitis through modulation of mucosal immune system. Scandian Journal of Gastroenterology 2008; 43(11): 1346-1352, doi: 10.1080/00365520802245411 pmid: 18618333
  • 217 Triantafyllidi A, Xanthos T, Papalois A, Triantafillidis JK. Herbal and plant therapy in patients with inflammatory bowel disease. Annals of Gastroenterology 2015; 28(2): 210-220, pmid: 25830661
  • 218 Ben-Arye E, Goldin E, Wengrower D, Stamper A, Kohn R, Berry E. Wheat grass juice in the treatment of active distal ulcerative colitis: a randomized double-blind placebo- controlled trial. Scandian Journal of Gastroenterology 2002; 37(4): 444-449, doi: 10.1080/003655202317316088 pmid: 11989836
  • 219 Suzuki H, Kaneko T, Mizokami Y, Narasaka T, Endo S, Matsui H, Yanaka A, Hirayama A, Hyodo I. Therapeutic efficacy of the Qing Dai in patients with intractable ulcerative colitis. World Journal of Gastroenterology 2013; 19(17): 2718-2722, doi: 10.3748/wjg.v19.i17.2718 pmid: 23674882
  • 220 Zhang F, Li Y, Xu F, Chu Y, Zhao W. Comparison of Xilei- san, a Chinese herbal medicine, and dexamethasone in mild/ moderate ulcerative proctitis: a double-blind randomized clinical trial. Journal of Alternative and Complementary Medicine 2013; 19(10): 838-842, doi: 10.1089/acm.2012.0296 pmid: 23383973
  • 221 Sugimoto S, Naganuma M, Kiyohara H, Arai M, Ono K, Mori K, Saigusa K, Nanki K, Takeshita K, Takeshita T, Mutaguchi M, Mizuno S, Bessho R, Nakazato Y, Hisamatsu T, Inoue N, Ogata H, Iwao Y, Kanai T. Clinical Efficacy and Safety of Oral Qing-Dai in Patients with Ulcerative Colitis: A Single-Center Open-Label Prospective Study. Digestion 2016; 93(3): 193-201, doi: 10.1159/000444217 pmid: 26959688
  • 222 Naganuma M, Sugimoto S, Mitsuyama K, Kobayashi T, Yoshimura N, Ohi H, Tanaka S, Andoh A, Ohmiya N, Saigusa K, Yamamoto T, Morohoshi Y, Ichikawa H, Matsuoka K, Hisamatsu T, Watanabe K, Mizuno S, Suda W, Hattori M, Fukuda S, Hirayama A, Abe T, Watanabe M, Hibi T, Suzuki Y, Kanai T, Group IS. Efficacy of Indigo Naturalis in a Multicenter Randomized Controlled Trial of Patients With Ulcerative Colitis. Gastroenterology 2018; 154(4): 935-947, doi: 10.1053/j. gastro.2017.11.024 pmid: 29174928
  • 223 Naganuma M. Treatment with indigo naturalis for inflammatory bowel disease and other immune diseases. Immunological Medicine 2019; 42(1): 16-21, doi: 10.1080/25785826.2019.1599158 pmid: 31034341
  • 224 Saiki JP, Andreasson JOL, Grimes KV, Frumkin LR, Sanjines E, Davidson MG, Park KT, Limketkai B. Treatment- refractory ulcerative colitis responsive to indigo naturalis. BMJ Open Gastroenterology 2021; 8(1), doi: 10.1136/ bmjgast-2021-000813 pmid: 34969665
  • 225 Gu S, Xue Y, Gao Y, Shen S, Zhang Y, Chen K, Xue S, Pan J, Tang Y, Zhu H, Wu H, Dou D. Mechanisms of indigo naturalis on treating ulcerative colitis explored by GEO gene chips combined with network pharmacology and molecular docking. Scientific Reports 2020; 10(1): 15204, doi: 10.1038/ s41598-020-71030-w pmid: 32938944
  • 226 Kondo S, Araki T, Okita Y, Yamamoto A, Hamada Y, Katsurahara M, Horiki N, Nakamura M, Shimoyama T, Yamamoto T, Takei Y, Kusunoki M. Colitis with wall thickening and edematous changes during oral administration of the powdered form of Qing-dai in patients with ulcerative colitis: a report of two cases. Clinical Journal of Gastroenterology 2018; 11(4): 268- 272, doi: 10.1007/s12328-018-0851-7 pmid: 29549501
  • 227 Mimura J, Ema M, Sogawa K, Fujii-Kuriyama Y. Identification of a novel mechanism of regulation of Ah (dioxin) receptor function. Genes and Development 1999; 13(1): 20-25, doi: 10.1101/gad.13.1.20 pmid: 9887096
  • 228 Sen A, Arinc E. Further immunochemical and biocatalytic characterization of CYP1A1 from feral leaping mullet liver (Liza saliens) microsomes. Comparative Biochemistry and Physiology Part C: Toxicology and Pharmacology 2000; 126(3): 235-244, doi: 1104867310.1016/s0742-8413(00)00117-1 pmid: 11048673
  • 229 Spink BC, Hussain MM, Katz BH, Eisele L, Spink DC. Transient induction of cytochromes P450 1A1 and 1B1 in MCF-7 human breast cancer cells by indirubin. Biochemical Pharmacology 2003; 66(12): 2313-2321, doi: 10.1016/j.bcp.2003.08.019 pmid: 14637189
  • 230 Adachi J, Mori Y, Matsui S, Matsuda T. Comparison of gene expression patterns between 2,3,7,8-tetrachlorodibenzo- p-dioxin and a natural arylhydrocarbon receptor ligand, indirubin. Toxicological Sciences 2004; 80(1): 161-169, doi: 10.1093/toxsci/kfh129 pmid: 15056799
  • 231 Sinz M, Kim S, Ferguson S, LeCluyse E. Evaluating and Predicting Human Cytochrome P450 Enzyme Induction. Pharmaceutical Sciences Encyclopedia, 2010: 1-31
  • 232 Kumagai T, Aratsu Y, Sugawara R, Sasaki T, Miyairi S, Nagata K. Indirubin, a component of Ban-Lan-Gen, activates CYP3A4 gene transcription through the human pregnane X receptor. Drug Metabolism and Pharmacokinetics 2016; 31(2): 139-145, doi: 10.1016/j.dmpk.2016.01.002 pmid: 26987505
  • 233 Xu H-H, Ma Z-C, Shi Q-L, Yang S-H, Jiang L, Chen X-M, Gao Y. Synergistic effect and different toxicities of adjuvant components of Realgar–Indigo Naturalis formula. Chinese Herbal Medicines 2018; 10(2): 137-144, doi: 10.1016/j. chmed.2018.03.001
  • 234 Greenfield SM, Green AT, Teare JP, Jenkins AP, Punchard NA, Ainley CC, Thompson RP. A randomized controlled study of evening primrose oil and fish oil in ulcerative colitis. Alimentary Pharmacology and Therapeutics 1993; 7(2 ): 159- 166, doi: 10.1111/j.1365-2036.1993.tb00085.x pmid: 8485269
  • 235. Brinkhaus B, Hentschel C, Von Keudell C, Schindler G, Lindner M, Stutzer H, Kohnen R, Willich SN, Lehmacher W, Hahn EG. Herbal medicine with curcuma and fumitory in the treatment of irritable bowel syndrome: a randomized, placebo-controlled, double-blind clinical trial. Scandian Journal of Gastroenterology 2005; 40(8): 936-943, doi: 10.1080/00365520510023134 pmid: 16173134
  • 236. Emendorfer F, Emendorfer F, Bellato F, Noldin VF, Cechinel-Filho V, Yunes RA, Delle Monache F, Cardozo AM. Antispasmodic activity of fractions and cynaropicrin from Cynara scolymus on guinea-pig ileum. Biological and Pharmaceutical Bulletin 2005; 28(5): 902-904, doi: 10.1248/ bpb.28.902 pmid: 15863902
  • 237. Huber R, Ditfurth AV, Amann F, Guthlin C, Rostock M, Trittler R, Kummerer K, Merfort I. Tormentil for active ulcerative colitis: an open-label, dose-escalating study. Journal of Clinical Gastroenterology 2007; 41(9): 834-838, doi: 10.1097/ MCG.0b013e31804b2173 pmid: 17881930
  • 238. Kim JM, Kang HW, Cha MY, Yoo D, Kim N, Kim IK, Ku J, Kim S, Ma SH, Jung HC, Song IS, Kim JS. Novel guggulsterone derivative GG-52 inhibits NF-kappaB signaling in intestinal epithelial cells and attenuates acute murine colitis. Laboratory Investigation 2010; 90(7): 1004-1015, doi: 10.1038/ labinvest.2010.54 pmid: 20195240
  • 239. Wan H, Chen Y. Effects of antidepressive treatment of Saint John’s wort extract related to autonomic nervous function in women with irritable bowel syndrome. Int The International Journal of Psychiatry in Medicine 2010; 40(1): 45-56, doi: 10.2190/PM.40.1.d pmid: 20565044
  • 240. Kudo T, Okamura S, Zhang Y, Masuo T, Mori M. Topical application of glycyrrhizin preparation ameliorates experimentally induced colitis in rats. World Journal of Gastroenterology 2011; 17(17): 2223-2228, doi: 10.3748/wjg. v17.i17.2223 pmid: 21633533
  • 241. Gong Y, Zha Q, Li L, Liu Y, Yang B, Liu L, Lu A, Lin Y, Jiang M. Efficacy and safety of Fufangkushen colon-coated capsule in the treatment of ulcerative colitis compared with mesalazine: a double-blinded and randomized study. Journal of Ethnopharmacology 2012; 141(2): 592-598, doi: 10.1016/j. jep.2011.08.057 pmid: 21911045
  • 242. Fukunaga K, Ohda Y, Hida N, Iimuro M, Yokoyama Y, Kamikozuru K, Nagase K, Nakamura S, Miwa H, Matsumoto T. Placebo controlled evaluation of Xilei San, a herbal preparation in patients with intractable ulcerative proctitis. Journal of Gastroenterology and Hepatology 2012; 27(12): 1808-1815, doi: 10.1111/j.1440-1746.2012.07215.x pmid: 22775479
  • 243. Khanna R, MacDonald JK, Levesque BG. Peppermint oil for the treatment of irritable bowel syndrome: a systematic review and meta-analysis. Journal of Clinical Gastroenterology 2014; 48(6): 505-512 doi: 10.1097/MCG.0b013e3182a88357 pmid: 24100754
  • 244. Karakurt S, Semiz A, Celik G, Gencler-Ozkan AM, Sen A, Adali O. Contribution of ellagic acid on the antioxidant potential of medicinal plant Epilobium hirsutum. Nutrition and Cancer 2016; 68(1): 173-183, doi: 10.1080/01635581.2016.1115092 pmid: 26700224
  • 245. Ozgun-Acar O, Celik-Turgut G, Gazioglu I, Kolak U, Ozbal S, Ergur BU, Arslan S, Sen A, Topcu G. Capparis ovata treatment suppresses inflammatory cytokine expression and ameliorates experimental allergic encephalomyelitis model of multiple sclerosis in C57BL/6 mice. Journal of Neuroimmunology 2016; 298: 106-116, doi: 10.1016/j.jneuroim.2016.07.010 pmid: 27609283
  • 246. Sebepos-Rogers GM, Rampton DS. Herbs and Inflammatory Bowel Disease. Gastroenterology Clinics of North America 2017; 46(4): 809-824, doi: 10.1016/j.gtc.2017.08.009 pmid: 29173523
  • 247. Gazioglu I, Semen S, Acar OO, Kolak U, Sen A, Topcu G. Triterpenoids and steroids isolated from Anatolian Capparis ovata and their activity on the expression of inflammatory cytokines. Pharmaceutical Biology 2020; 58(1): 925-931, doi: 10.1080/13880209.2020.1814356 pmid: 32915696
  • 248. Semiz A, Ozgun Acar O, Cetin H, Semiz G, Sen A. Suppression of Inflammatory Cytokines Expression with Bitter Melon (Momordica Charantia) in TNBS-instigated Ulcerative Colitis. Journal of Translational Internal Medicine 2020; 8(3): 177-187, doi: 10.2478/jtim-2020-0027 pmid: 33062594
  • 249. Suntar I, Cevik CK, Ceribasi AO, Gokbulut A. Healing effects of Cornus mas L. in experimentally induced ulcerative colitis in rats: From ethnobotany to pharmacology. Journal of Ethnopharmacology 2020; 248: 112322, doi: 10.1016/j. jep.2019.112322 pmid: 31644942
  • 250. Yu H, Li TN, Ran Q, Huang QW, Wang J. Strobilanthes cusia (Nees) Kuntze, a multifunctional traditional Chinese medicinal plant, and its herbal medicines: A comprehensive review. Journal of Ethnopharmacology 2021; 265: 113325, doi: 10.1016/j.jep.2020.113325 pmid: 32889034
  • 251. Zhu X, Yang Y, Gao W, Jiang B, Shi L. Capparis spinosa Alleviates DSS-Induced Ulcerative Colitis via Regulation of the Gut Microbiota and Oxidative Stress. Evidence-Based Complementary and Alternative Medicine 2021; 2021: 1227876, doi: 10.1155/2021/1227876 pmid: 34956375
APA SEN A (2022). Complementary medicines used in ulcerative colitis and unintended interactions with cytochrome P450-dependent drug-metabolizing enzymes. , 1425 - 1447. 10.55730/1300-0144.5482
Chicago SEN Alaattin Complementary medicines used in ulcerative colitis and unintended interactions with cytochrome P450-dependent drug-metabolizing enzymes. (2022): 1425 - 1447. 10.55730/1300-0144.5482
MLA SEN Alaattin Complementary medicines used in ulcerative colitis and unintended interactions with cytochrome P450-dependent drug-metabolizing enzymes. , 2022, ss.1425 - 1447. 10.55730/1300-0144.5482
AMA SEN A Complementary medicines used in ulcerative colitis and unintended interactions with cytochrome P450-dependent drug-metabolizing enzymes. . 2022; 1425 - 1447. 10.55730/1300-0144.5482
Vancouver SEN A Complementary medicines used in ulcerative colitis and unintended interactions with cytochrome P450-dependent drug-metabolizing enzymes. . 2022; 1425 - 1447. 10.55730/1300-0144.5482
IEEE SEN A "Complementary medicines used in ulcerative colitis and unintended interactions with cytochrome P450-dependent drug-metabolizing enzymes." , ss.1425 - 1447, 2022. 10.55730/1300-0144.5482
ISNAD SEN, Alaattin. "Complementary medicines used in ulcerative colitis and unintended interactions with cytochrome P450-dependent drug-metabolizing enzymes". (2022), 1425-1447. https://doi.org/10.55730/1300-0144.5482
APA SEN A (2022). Complementary medicines used in ulcerative colitis and unintended interactions with cytochrome P450-dependent drug-metabolizing enzymes. Turkish Journal of Medical Sciences, 52(5), 1425 - 1447. 10.55730/1300-0144.5482
Chicago SEN Alaattin Complementary medicines used in ulcerative colitis and unintended interactions with cytochrome P450-dependent drug-metabolizing enzymes. Turkish Journal of Medical Sciences 52, no.5 (2022): 1425 - 1447. 10.55730/1300-0144.5482
MLA SEN Alaattin Complementary medicines used in ulcerative colitis and unintended interactions with cytochrome P450-dependent drug-metabolizing enzymes. Turkish Journal of Medical Sciences, vol.52, no.5, 2022, ss.1425 - 1447. 10.55730/1300-0144.5482
AMA SEN A Complementary medicines used in ulcerative colitis and unintended interactions with cytochrome P450-dependent drug-metabolizing enzymes. Turkish Journal of Medical Sciences. 2022; 52(5): 1425 - 1447. 10.55730/1300-0144.5482
Vancouver SEN A Complementary medicines used in ulcerative colitis and unintended interactions with cytochrome P450-dependent drug-metabolizing enzymes. Turkish Journal of Medical Sciences. 2022; 52(5): 1425 - 1447. 10.55730/1300-0144.5482
IEEE SEN A "Complementary medicines used in ulcerative colitis and unintended interactions with cytochrome P450-dependent drug-metabolizing enzymes." Turkish Journal of Medical Sciences, 52, ss.1425 - 1447, 2022. 10.55730/1300-0144.5482
ISNAD SEN, Alaattin. "Complementary medicines used in ulcerative colitis and unintended interactions with cytochrome P450-dependent drug-metabolizing enzymes". Turkish Journal of Medical Sciences 52/5 (2022), 1425-1447. https://doi.org/10.55730/1300-0144.5482