Iraqi J Pharm Sci, Vol.31(1) 2022 Curcumin-tacrolimus interaction DOI: https://doi.org/10.31351/vol31iss1pp246-250 246 Effect of Curcumin at Various Doses on the Pharmacokinetic Profile of Tacrolimus in Healthy Rabbits Issam Mohammed Abushammala*,1, Belal Mohammed Mqat* and Abdallah Mohammed Hamdan* *Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University/Gaza, Palestine. Abstract The purpose of present study was to evaluate the effect of co-administration of curcumin (CUR) at various doses on the pharmacokinetic (PK) profile of tacrolimus (TAC), a CYP 3A4 substrate in healthy male rabbits. Healthy male rabbits (n=18) were employed in an in vivo, parallel-randomized study. Three groups of rabbits were selected and separated: The rabbits in the first group (control group) received 1 mg/kg TAC orally. Blood samples (1.5-2 mL) were drawn from rabbits' ear marginal veins at the following time frames: 15.0, 30.0, 45.0, 60.0, 90.0, 120.0, 150.0, 180.0 and 300 minutes after TAC administration and analyzed by using a TAC Chemiluminescent Enzyme Immunoassay (CLIA) detection kit. In the second and third groups (test groups), rabbits received TAC (1mg/kg) at identical conditions as in the control group with volumes equivalent to (30 and 90 mg/kg/day) of prepared CUR suspension in normal saline for seven continuous days. Blood samples from the control group were obtained on the eighth day. Non-compartmental analysis was used to derive different PK parameters of TAC for the three groups. When CUR was co-administered at both concentrations, statistically insignificant small changes between the control and testing groups were found. The current results revealed that the differences for the three groups in PK parameters as Cmax, Tmax, ke, AUC0-6 and AUC0-∞ were statistically insignificant (P>0.05). In conclusion, it has been found that CUR at the experimented doses does not affect the PK of TAC. Further confirmation of our findings is required before these results can be applied in patient care. Keywords: Tacrolimus, Curcumin, Pharmacokinetics لتاكروليموس في األرانب السليمة لتأثير الكركمين بجرعات مختلفة على ملف الحرائك الدوائية * عبدهللا محمد حمدان و *بالل محمد مقاط ،1*،عصام محمد ابو شمالة . غزة، فلسطين –قسم الصيدالنيات والصيدلة الصناعية، كلية الصيدلة، جامعة األزهر * الخالصة ( لعقار PK( بجرعات مختلفة على الخواص الحركية الدوائية )CURالكركمين ) أضافة مادةالغرض من هذه الدراسة هو تقييم تأثير في ذكور األرانب السليمة صحيا. في هذه الدراسة تم توظيف أرانب ذكور صحية CYP3A4بواسطة االنزيم الذي يستقلب( وTACالتاكروليموس ) ( في دراسة عشوائية متوازية. تم اختيار وفصل ثالث مجموعات من األرانب: تلقت األرانب في المجموعة األولى )المجموعة الضابطة( 18)ن = مل( من األوردة الهامشية ألذن األرانب في األطر الزمنية المحددة بعد 2-1.5م تم سحب عينات الدم )عن طريق الفم ومن ث TACمجم / كجم من 1 ( المضيئة اإلنزيمية المناعية المقايسة عن الكشف أدوات مجموعة باستخدام تحليلها و التاكروليموس دواء بدواء CLIAإعطاء الخاصة ) ختبار الثانية والثالثة )مجموعتي االختبار( ، أعطيت األرانب نفس الجرعة الخاصة بدواء التاكروليموس التاكروليموس. اما فيما يتعلق بمجموعتي اال المحضر في محلول ملحي عادي لسبعة CURمجم / كجم / يوم( من معلق 90و 30فى نفس الظروف كما في المجموعة الضابطة بتراكيز تعادل ) للمجموعات الثالث TACعينات الدم كالمجموعة الضابطة وتم تحديد معلمات الحركية الدوائية المختلفة لـ أيام متواصلة. في اليوم الثامن ، تم جمع عن المختبرة والمجموعات الضابطة المجموعة بين إحصائية داللة ذات غير اختالفات هناك كانت ، أنه لوحظ الجزئي. غير التحليل د باستخدام ke, max, TmaxC , ,. كشفت نتائجنا أن الفروق بين المجموعات الثالث في معامالت الحرائك الدوائية مثل في كال التركيزين CURاالضافة مع ∞-0and AUC 6-0AUC ( كانت غير ذات داللة إحصائيةp˃0.05 فى الختام ، لقد وجد ان الكوركمين عند الجرعات المجربة ال يؤثر على .) ن هناك حاجة إلى مزيد من التأكيد على هذه النتائج التي تم التوصل إليها قبل تطبيقها في رعاية المرضى.معامالت الحركية لدواء التاكروليمس ولك تاكروليموس ، الكركمين ، حركية الدواء. المفتاحية: الكلمات Introduction Tacrolimus (TAC), a calcineurin inhibitor, is an important component of the traditional immunosuppressive regimen after renal transplantation. Due to its narrow therapeutic index and the fact that various factors intervene with its metabolism (1). TAC's PK pathways involve cytochrome P450 (CYP) 3A4 and P-glycoprotein (P-gp), and drugs that interact with these enzyme systems will influence TAC blood concentrations (2). Because TAC covers a restricted therapeutic window and lacks a strong correlation between dosage and serum concentration, it may cause toxicity, subtherapeutic failure, and organ rejection (3). TAC interacts with several other treatments used in transplantation therapy that are known CYP3A and/or P-gp inhibitors and/or inducers as a substrate of CYP3A enzymes and P-gp (4). 1Corresponding author E-mail: issam.abushammala@uv.es Received: 14/8/2021 Accepted: 20/10 /2021 Iraqi Journal of Pharmaceutical Science https://doi.org/10.31351/vol31iss1pp246-250 Iraqi J Pharm Sci, Vol.31(1) 2022 Curcumin-tacrolimus interaction 247 The CYP450 enzymes are membrane- bound hemoproteins that play a pivotal role in the detoxification of xenobiotics, cellular metabolism and homeostasis. Induction or inhibition of CYP450 enzymes is a major mechanism that underlies drug- drug interactions (5). Natural products have shown a promising source of components for the development of new drugs (6,7). One of the most common example is turmeric (Curcuma longa Linn.), which is a member of the Zingiberaceae family (8). The rising importance of natural remedies around the world has sparked concerns concerning herb-drug interactions. These interactions are especially important for drugs with narrow therapeutic indices and may either be pharmacodynamic or PK in nature when used with P-gp and CYP3A4 substrates (9,10). Herb-drug interactions are one of the most serious clinical concerns while using herbal remedies and prescribed drugs simultaneously (11). The likelihood of PK medication interactions involves two key challenges: pharmacotoxicity and treatment failure. The earlier can be produced by inhibiting drug metabolism and clearance enzymes, whereas the latter can be triggered by enzymatic stimulation resulting in increased drug metabolism (12). Numerous in vitro studies have reported that curcumin inhibited not only P-gp, but also CYP3A4. Based on these in vitro results, coadministration of curcumin should increase the oral bioavailability of substrates of P-gp or CYP 3A4 (13). The effect of co-administration of CUR at various doses on the PK profile of TAC, a CYP 3A4 substrate, was investigated in the present study. Materials and Methods Animals In this study, eighteen New Zealand strains of adult male rabbits weighing (3.1-3.4 kg) and aged (8-10) months were used. The Research and Ethics Committee granted permission to the Experimental Animal Care Facility, College of Pharmacy, Al- Azhar University of Gaza (AUG), Palestine. The rabbits were randomly split into three groups (six for each group). All rabbits were housed in controlled conditions with a 12-hour light/dark cycle at (25°C ± 2°C), pellet feeding, and free access to water (ad libitum). Prior to the trial, they also fasted over the night. Study design and blood sampling Eighteen healthy male rabbits were used in an in vivo, parallel-randomized controlled study. Three groups of rabbits were selected and separated: The rabbits in the first group (control group) received 1 mg/kg TAC orally from a hard capsule (5 mg, prograf®, astellas) kindly provided by the Palestinian Ministry of Health's medication stores. Blood samples (1.5-2 mL) were drawn from rabbits' ear marginal veins at the following time frames: 15.0, 30.0, 45.0, 60.0, 90.0, 120.0, 150.0, 180.0 and 300 minutes following TAC administration (14). In the second and third groups (test groups), rabbits were given TAC (1mg/kg) at the same conditions as in the control group with volumes equivalent to (30 and 90 mg/kg/day) from prepared CUR suspension in normal saline (Turmeric® Curcumin 550 mg, Jamieson) hard gelatin capsules purchased from a local pharmacy in Gaza, Palestine) for seven continuous days. On the 8th day, TAC was given one hour after the last dosage of CUR suspension, and blood was collected from the second and third test groups at the same time as the control group. Whole blood samples were stored in EDTA tubes at 2-8°C until they were tested (Whole blood sample is stable for up to 3 days). Blood samples analysis At the Medical Relief Society-Gaza laboratory, whole blood samples were tested to detect TAC concentrations using the Maglumi 800 System and TAC detection kit. The TAC detection Kit is based on a chemiluminescent enzyme immunoassay (CLIA). It's utilized in hospitals to check TAC doses by doing fast TAC assays on whole blood. PK of TAC and statistical analysis The PK parameters, Cmax, Tmax, Ke, AUC0- 6, and AUC0-∞ were determined for the control and CUR-treated test groups. The Cmax and Tmax were accurately observed through plasma concentration versus time curves. The AUC0-6 was determined using the linear trapezoidal method. The AUC0- ∞value was calculated using the formula: AUC0-∞ = AUC0-6 + Ct / Ke, where Ct is the final measured blood concentration at time t and Ke is the elimination rate constant. The Ke was calculated using semilogarithmic dependency, which corresponds to first-order kinetics, by least- squares regression of blood concentration-time data points in the terminal region. The PK analysis was determined using a model- independent method (Non-Compartmental Approach), WinNonlin Professional Software (Version 6.3, Pharsight Corporation, Cary, NC), and (GraphPad Prism version 4.00; San Diego, CA, USA). The PK parameters of TAC alone (control group) and TAC co-administered with CUR in the first and second test groups were compared using statistical methods such as descriptive analysis and the Mann-Whitney test. The data was analyzed by using the SPSS program (version 22.0). A statistically significant difference was considered when P≤0.05. Iraqi J Pharm Sci, Vol.31(1) 2022 Curcumin-tacrolimus interaction 248 Results CUR's effect at various dosages on the PK parameters of TAC was explored in vivo. The PK parameters of TAC in the control group (first group) were compared to those of the test groups treated with CUR 30 and 90 mg/kg/day (second and third groups respectively) and the blood concentration- time profiles of TAC in the first group and in the co- administered with CUR (test groups) are shown in Figure 1 and Table 1. Figure 1. Plot of TAC concentration-time profile with and without CUR for the control, first and second test groups. Table 1. Calculated PK parameters of the control, the first and second test groups (6 for each). PK Parameters Groups Mean ± SD P-values aCmax (ng/mL) Control group 27.44 ± 5.83 0.668¥ 0.338§ First test group 25.78 ± 7.60 Second test group 31.20 ± 4.33 bTmax (hr -1) Control group 1.50 ± 0.50 0.405¥ 0.690§ First test group 1.90 ± 0.54 Second test group 1.65 ± 0.65 cke (hr-1) Control group 0.102 ± 0.04 0.736¥ 0.570§ First test group 0.124 ± 0.10 Second test group 0.091 ± 0.03 dAUC0-6 (ng*hr/mL) Control group 97.37 ± 18.06 0.524¥ 0.052§ First test group 112.42 ± 49.14 Second test group 130.02 ± 20.54 eAUC0-∞ (ng*hr/mL) Control group 204.49 ± 75.82 0.564¥ 0.679§ First test group 167.05 ± 71.42 Second test group 189.60 ± 25.80 (¥): P-value of the differences between the control and first test group; (§): P-value of the differences between the control and the second test group. *:P≤0.05 Statistical significance, SD: Standard deviation, amaximum blood concentration, btime to peak concentration, celimination rate constant, darea under the concentration-time profile curve from 0 to 6 hours and darea under the concentration-time profile curve from 0 to infinity. T im e ( h ) T a c ro li m u s ( T A C ) b lo o d c o n c e n tr a ti o n ( n g /m l) 0 .5 1 1 .5 2 2 .5 3 3 .5 4 4 .5 5 5 .5 6 0 1 0 2 0 3 0 4 0 C o n t r o l G r o u p : T A C a l o n e ( 1 m g / k g ) S e c o n d t e s t G r o u p ( T A C 1 m g / k g + C U R 9 0 m g / k g / d a y ) F i r s t t e s t G r o u p ( T A C 1 m g / k g + C U R 3 0 m g / k g / d a y ) Iraqi J Pharm Sci, Vol.31(1) 2022 Curcumin-tacrolimus interaction 249 Discussion TAC is an immunosuppressive agent that has emerged as a valuable therapeutic alternative to cyclosporine following solid organ transplantation (15). The most important human CYP isozyme is CYP3A4, which is involved in the metabolism of the majority of therapeutically prescribed drugs (16). TAC is a lipophilic compound that is metabolized by the CYP450 3A subfamily and is eliminated after extensive metabolism. Because of its low therapeutic index, TAC requires blood level monitoring. Therefore, metabolic studies, such as drug-drug interaction and metabolite identification studies, are vital and urgent for the development of clinically optimal medication use (17). Herb-drug interactions are among the most frequent medical concerns when taking herbs and pharmaceutical prescriptions concurrently (11). In addition, TAC bioavailability decreased when concurrently administered with St John’s wort (SJW), cranberry, rooibos tea, and boldo in human models by induction of CYP450 system isoenzyme and/or P-gp efflux pump (18-21). Meanwhile, TAC bioavailability was enhanced in human and/or animal models when grapefruit juice, schisandra, pomelo, and ginger were given concurrently, presumably due to an inhibitory effect on the CYP450 system or the P-gp efflux pump (22-24). The PK investigations evaluating the interaction between herbal supplements and the bioavailability of various therapeutically monitored drugs, such as digoxin, cyclosporine and carbamazepine revealed that herbal supplements have a clinically insignificant effect on the PK profiles of digoxin and cyclosporine (25-27). Our findings, statistical examination of TAC PK parameters revealed statistically insignificant differences between the three groups (Table 1). The control group's mean Cmax decreased slightly from 27.44±5.83 ng/mL to 25.78±7.60 ng/mL in the first test group and a slight increase to 31.20±4.33 ng/mL in the second test group were observed. Similarly, Liu and his colleagues found comparable results when they investigated the effect of various CUR administrations (25 and 50 mg/kg) on the PK parameters of warfarin alone (control group). In this investigation, the increasing in Cmax in the herbal treated groups from 1.14±0.33 to 1.49±0.38 and 1.15±0.29 µg/mL was insignificant when compared to the control group (P>0.05). However, in the large dose CUR groups (100 mg/kg) were statistically significant (28). Also, the current results revealed that the decrease in AUC0-∞ from 204.49±75.82 to 167.05±7.10 in the first test group and 189.60±25.80 ng*hr/mL in the second test group respectively, was also similar and statistically insignificant (P>0.05). Furthermore, the remaining PK parameters showed insignificant variations, including AUC0-6, Tmax and Ke between the control and tested groups. Additional investigations need to be conducted with higher doses of curcumin and over longer periods of time, or recruit a larger number of animals, to support the interpretation of previous research findings. Conclusion Drug interactions with herbs are critically important for patient safety, especially as herbal remedies become more popular and are used more frequently. The CUR had systemically insignificant effect on the PK profile of TAC in this investigation at the dosages examined. Ethical Statement The study was approved and performed under ethical principles laid down by the Faculty of Pharmacy, Al-Azhar University-Gaza, Palestine. Conflict of Interest No conflicts of interest relevant to this article. Acknowledgments The authors wish to express deepest gratitude and thanks to all staff in the Faculty of Pharmacy at Al-Azhar University-Gaza for their kind support. References 1. Schutte-Nutgen K, Tholking G, and Suwelack B, et al. Tacrolimus- Pharmacokinetic Considerations for Clinicians. Current Drug Metabolism. 2018;19:342-350. 2. Van Gelder T. Drug interactions with tacrolimus. Drug Safty. 2002;25:707-712. 3. Zhai X, Chen C, and Xu X, et al. Marked change in blood tacrolimus concentration levels due to grapefruit in a renal transplant patient. Journal of Clinical Pharmacy and Therapeutics. 2019; 44:819-822. 4. Christians U, Jacobsen W, and Benet LZ, et al. Mechanisms of clinically relevant drug interactions associated with tacrolimus. Clinical Pharmacokinetics. 2002;41:813-851. 5. Manikandan P, Nagini S. Cytochrome P450 Structure, Function and Clinical Significance: A Review. Current Drug Targets. 2018;19:38- 54. 6. Newman DJ, Cragg GM, Snader KM. Natural products as sources of new drugs over the period. Journal of Natural Products. 2003;66:1022e1037. 7. Pandit S, Kim H, and Kim J, et al. Separation of an effective fraction from turmeric against Streptococcus mutans biofilms by the comparison of curcuminoid content and anti- acidogenic activity. Food Chemistry. 2011;126:1565-1570. Iraqi J Pharm Sci, Vol.31(1) 2022 Curcumin-tacrolimus interaction 250 8. Paramasivam M, Poi R, and Banerjee H, et al. High-performance thin layer chromatographic method for quantitative determination of curcuminoids in Curcuma longa germplasm. Food Chemistry. 2009;113:640-644. 9. Tarirai C, Viljoen AM, Hamman JH. Herb-drug pharmacokinetic interactions reviewed. Expert Opinion on Drug Metabolism and Toxicology. 2010;6:1515-1538. 10. Bahramsoltani R, Rahimi R, Farzaei MH. Pharmacokinetic interactions of curcuminoids with conventional drugs: A review. Journal of Ethnopharmacology. 2017;209:1-12. 11. Quintieri L, Palatini P, Nassi A, Ruzza P, et al. Flavonoids diosmetin and luteolin inhibit midazolam metabolism by human liver microsomes and recombinant CYP 3A4 and CYP3A5 enzymes. Biochemical Pharmacology. 2008; 75:1426-1437. 12. Fasinu PS, Bouic PJ, Rosenkranz B. An overview of the evidence and mechanisms of herb-drug interactions. Frontiers in Pharmacology. 2012;3:69. 13. Hsieh YW, Huang CY, and Yang SY, et al. Oral intake of curcumin markedly activated CYP 3A4: in vivo and ex-vivo studies. Scientific Reports. 2014;4:6587. 14. Parasuraman S, Raveendran R, Kesavan R. Blood sample collection in small laboratory animals, Journal of Pharmacology and Pharmacotherapeutics. 2010; 1:87–93. 15. Staatz CE, Tett SE. Clinical pharmacokinetics and pharmacodynamics of tacrolimus in solid organ transplantation. Clinical Pharmacokinetics. 2004;43:623-653. 16. Wanwimolruk S, Prachayasittikul V. Cytochrome P450 enzyme mediated herbal drug interactions (Part 1). EXCLI Journal. 2014;13:347-391. 17. Iwasaki K. Metabolism of tacrolimus (FK506) and recent topics in clinical pharmacokinetics. Drug Metabolism and Pharmacokinetics. 2007;22:328-335. 18. Hebert MF, Park JM, and Chen YL, et al. Effects of St John’s wort (Hypericum perforatum) on tacrolimus pharmacokinetics in healthy volunteers. Journal of Clinical Pharmacology. 2004;44: 89–94. 19. Dave AA, Samuel J. Suspected Interaction of Cranberry Juice Extracts and Tacrolimus Serum Levels: A Case Report. Cureus. 2016;8:610. 20. Beppu N, Mori T, and Kato J, et al. Drug-herbal interaction between tacrolimus and rooibos tea in a recipient of allogeneic hematopoietic stem cell transplantation. Journal of Hematopoietic Cell Transplantation. 2013;2:109-111. 21. Carbajal R, Yisfalem A, and Pradhan N, et al. Case report: boldo (Peumus boldus) and tacrolimus interaction in a renal transplant patient. Transplantion Proceedings. 2014;46:2400-2402. 22. Peynaud D, Charpiat B, and Vial T, et al. Tacrolimus severe overdosage after intake of masked grapefruit in orange marmalade. European Journal of Clinical Pharmacology. 2007;63:721-2. 23. Xiao-ling Q, Jia-li L, and Si-han W, et al. Co- administration of Wuzhi tablet (Schisandra sphenanthera extract) alters tacrolimus pharmacokinetics in a dose- and time- dependent manner in rats. Journal of Ethnopharmacology. 2020;263:113233. 24. Egashira K, Sasaki H, and Higuchi S, et al. Food-drug interaction of tacrolimus with pomelo, ginger, and turmeric juice in rats. Drug Metabolism and Pharmacokinetics. 2012;27:242-247. 25. Mauro VF, Mauro LS, and Kleshinski JF, et al. Impact of Ginkgo biloba on the pharmacokinetics of digoxin. American journal of therapeutics. 2003;10:247-251. 26. Xin HW, Wu XC, and Li Q, et al. The effects of berberine on the pharmacokinetics of cyclosporin A in healthy volunteers. Methods and Findings in Experimantal and Clinical Pharmacology. 2006;28:25-29. 27. Abushammala IM, El-Shaikh Ali FK, Abu Shammaleh KF, Taha MM, Miqdad MY. Effect of Panax ginseng on Carbamazepine Pharmacokinetics in Rabbits. Turk J Pharm Sci. 2021;25:18(1):17-20. 28. Liu AC, Zhao LX, Lou HX. Curcumin alters the pharmacokinetics of warfarin and clopidogrel in Wistar rats but has no effect on anticoagulation or antiplatelet aggregation. Planta Medica. 2013;79:971-977. This work is licensed under a Creative Commons Attribution 4.0 International License. http://creativecommons.org/licenses/by/4.0/ http://creativecommons.org/licenses/by/4.0/