BOXED WARNING SECTION.


BOXED WARNING MALIGNANCIES AND SERIOUS INFECTIONS. oIncreased risk of development of lymphoma and other malignancies, particularly of the skin, due to immunosuppression [see Warnings and Precautions (5.2)]. oIncreased susceptibility to bacterial, viral, fungal, and protozoal infections, including opportunistic infections [see Warnings and Precautions (5.3, 5.4, 5.5)]. oOnly physicians experienced in immunosuppressive therapy and management of organ transplant patients should prescribe tacrolimus. Patients receiving the drug should be managed in facilities equipped and staffed with adequate laboratory and supportive medical resources. The physician responsible for maintenance therapy should have complete information requisite for the follow-up of the patient [see Warnings and Precautions (5.1)]. oIncreased risk of development of lymphoma and other malignancies, particularly of the skin, due to immunosuppression [see Warnings and Precautions (5.2)]. oIncreased susceptibility to bacterial, viral, fungal, and protozoal infections, including opportunistic infections [see Warnings and Precautions (5.3, 5.4, 5.5)]. oOnly physicians experienced in immunosuppressive therapy and management of organ transplant patients should prescribe tacrolimus. Patients receiving the drug should be managed in facilities equipped and staffed with adequate laboratory and supportive medical resources. The physician responsible for maintenance therapy should have complete information requisite for the follow-up of the patient [see Warnings and Precautions (5.1)]. WARNING: MALIGNANCIES AND SERIOUS INFECTIONSSee full prescribing information for complete boxed warningoIncreased risk of development of lymphoma and other malignancies, particularly of the skin, due to immunosuppression 5.2) oIncreased susceptibility to bacterial, viral, fungal, and protozoal infections, including opportunistic infections 5.3, 5.4, 5.5) oOnly physicians experienced in immunosuppressive therapy and management of organ transplant patients should prescribe tacrolimus (5.1) oIncreased risk of development of lymphoma and other malignancies, particularly of the skin, due to immunosuppression 5.2) oIncreased susceptibility to bacterial, viral, fungal, and protozoal infections, including opportunistic infections 5.3, 5.4, 5.5) oOnly physicians experienced in immunosuppressive therapy and management of organ transplant patients should prescribe tacrolimus (5.1).

MECHANISM OF ACTION SECTION.


12.1 Mechanism of Action. Tacrolimus inhibits T-lymphocyte activation, although the exact mechanism of action is not known. Experimental evidence suggests that tacrolimus binds to an intracellular protein, FKBP-12. complex of tacrolimus-FKBP-12, calcium, calmodulin, and calcineurin is then formed and the phosphatase activity of calcineurin inhibited. This effect may prevent the dephosphorylation and translocation of nuclear factor of activated T-cells (NF-AT), nuclear component thought to initiate gene transcription for the formation of lymphokines (such as interleukin-2, gamma interferon). The net result is the inhibition of T-lymphocyte activation (i.e., immunosuppression).Tacrolimus prolongs the survival of the host and transplanted graft in animal transplant models of liver, kidney, heart, bone marrow, small bowel and pancreas, lung and trachea, skin, cornea, and limb.In animals, tacrolimus has been demonstrated to suppress some humoral immunity and, to greater extent, cell-mediated reactions such as allograft rejection, delayed type hypersensitivity, collagen-induced arthritis, experimental allergic encephalomyelitis, and graft versus host disease.

ADVERSE REACTIONS SECTION.


6 ADVERSE REACTIONS. The following serious and otherwise important adverse drug reactions are discussed in greater detail in other sections of labeling:oLymphoma and Other Malignancies [see Boxed Warning, Warnings and Precautions 5.2)] oSerious Infections [see Boxed Warning, Warnings and Precautions (5.3)] oPolyoma Virus Infections [see Boxed Warning, Warnings and Precautions 5.4)] oCMV Infections [see Boxed Warning, Warnings and Precautions 5.5)] oNew Onset Diabetes After Transplant [see Warnings and Precautions (5.6)] oNephrotoxicity [see Warnings and Precautions 5.7)] oNeurotoxicity [see Warnings and Precautions (5.8)] oHyperkalemia [see Warnings and Precautions 5.9)] oHypertension [see Warnings and Precautions 5.10)] oAnaphylaxis with Tacrolimus Injection [see Warnings and Precautions 5.11)] oMyocardial Hypertrophy [see Warnings and Precautions (5.15)] oPure Red Cell Aplasia [see Warnings and Precautions (5.17)] oGastrointestinal Perforation [see Warnings and Precautions (5.18)] oLymphoma and Other Malignancies [see Boxed Warning, Warnings and Precautions 5.2)] oSerious Infections [see Boxed Warning, Warnings and Precautions (5.3)] oPolyoma Virus Infections [see Boxed Warning, Warnings and Precautions 5.4)] oCMV Infections [see Boxed Warning, Warnings and Precautions 5.5)] oNew Onset Diabetes After Transplant [see Warnings and Precautions (5.6)] oNephrotoxicity [see Warnings and Precautions 5.7)] oNeurotoxicity [see Warnings and Precautions (5.8)] oHyperkalemia [see Warnings and Precautions 5.9)] oHypertension [see Warnings and Precautions 5.10)] oAnaphylaxis with Tacrolimus Injection [see Warnings and Precautions 5.11)] oMyocardial Hypertrophy [see Warnings and Precautions (5.15)] oPure Red Cell Aplasia [see Warnings and Precautions (5.17)] oGastrointestinal Perforation [see Warnings and Precautions (5.18)] oKidney Transplant: The most common adverse reactions >= 30%) were infection, tremor, hypertension, abnormal renal function, constipation, diarrhea, headache, abdominal pain, insomnia, nausea, hypomagnesemia, urinary tract infection, hypophosphatemia, peripheral edema, asthenia, pain, hyperlipidemia, hyperkalemia, anemia 6.1) oLiver Transplant: The most common adverse reactions (>= 40%) were tremor, headache, diarrhea, hypertension, nausea, abnormal renal function, abdominal pain, insomnia, paresthesia, anemia, pain, fever, asthenia, hyperkalemia, hypomagnesemia, and hyperglycemia 6.1) oHeart Transplant: The most common adverse reactions (>= 15%) were abnormal renal function, hypertension, diabetes mellitus, CMV infection, tremor, hyperglycemia, leukopenia, infection, anemia, bronchitis, pericardial effusion, urinary tract infection and hyperlipemia 6.1) To report SUSPECTED ADVERSE REACTIONS, contact Accord Healthcare Inc. at 1-866-941-7875 or www.accordhealthcare.us or FDA at 1-800-FDA-1088 or www.fda.gov/medwatch. oKidney Transplant: The most common adverse reactions >= 30%) were infection, tremor, hypertension, abnormal renal function, constipation, diarrhea, headache, abdominal pain, insomnia, nausea, hypomagnesemia, urinary tract infection, hypophosphatemia, peripheral edema, asthenia, pain, hyperlipidemia, hyperkalemia, anemia 6.1) oLiver Transplant: The most common adverse reactions (>= 40%) were tremor, headache, diarrhea, hypertension, nausea, abnormal renal function, abdominal pain, insomnia, paresthesia, anemia, pain, fever, asthenia, hyperkalemia, hypomagnesemia, and hyperglycemia 6.1) oHeart Transplant: The most common adverse reactions (>= 15%) were abnormal renal function, hypertension, diabetes mellitus, CMV infection, tremor, hyperglycemia, leukopenia, infection, anemia, bronchitis, pericardial effusion, urinary tract infection and hyperlipemia 6.1) 6.1 Clinical Studies Experience. 6.2 Postmarketing Adverse Reactions. The following adverse reactions have been reported from worldwide marketing experience with tacrolimus. Because these reactions are reported voluntarily from population of uncertain size it is not always possible to reliably estimate their frequency or establish causal relationship to drug exposure. Decisions to include these reactions in labeling are typically based on one or more of the following factors: (1) seriousness of the reaction, (2) frequency of the reporting, or (3) strength of causal connection to the drug. Other reactions include: CardiovascularAtrial fibrillation, atrial flutter, cardiac arrhythmia, cardiac arrest, electrocardiogram wave abnormal, flushing, myocardial infarction, myocardial ischaemia, pericardial effusion, QT prolongation, Torsade de Pointes, venous thrombosis deep limb, ventricular extrasystoles, ventricular fibrillation, myocardial hypertrophy [see Warnings and Precautions (5.15)] Gastrointestinal Bile duct stenosis, colitis, enterocolitis, gastroenteritis, gastrooesophageal reflux disease, hepatic cytolysis, hepatic necrosis, hepatotoxicity, impaired gastric emptying, liver fatty, mouth ulceration, pancreatitis haemorrhagic, pancreatitis necrotizing, stomach ulcer, venoocclusive liver disease Hemic/Lymphatic Agranulocytosis, disseminated intravascular coagulation, hemolytic anemia, neutropenia, pancytopenia, thrombocytopenic purpura, thrombotic thrombocytopenic purpura, pure red cell aplasia [see Warnings and Precautions (5.17)] Infections Cases of progressive multifocal leukoencephalopathy (PML), sometimes fatal; -polyoma virus-associated nephropathy, (PVAN) including graft loss [see Warnings and Precautions (5.4)] Metabolic/NutritionalGlycosuria, increased amylase including pancreatitis, weight decreasedMiscellaneousFeeling hot and cold, feeling jittery, hot flushes, multi-organ failure, primary graft dysfunction Nervous SystemCarpal tunnel syndrome, cerebral infarction, hemiparesis, leukoencephalopathy, mental disorder, mutism, posterior reversible encephalopathy syndrome (PRES) [see Warnings and Precautions (5.8) progressive multifocal leukoencephalopathy (PML) [see Warnings and Precautions 5.4)] quadriplegia, speech disorder, syncope Respiratory Acute respiratory distress syndrome, interstitial lung disease, lung infiltration, respiratory distress, respiratory failure Skin Stevens-Johnson syndrome, toxic epidermal necrolysis Special Senses Blindness, blindness cortical, hearing loss including deafness, photophobiaUrogenital Acute renal failure, cystitis haemorrhagic, hemolytic-uremic syndrome, micturition disorder.

CARCINOGENESIS & MUTAGENESIS & IMPAIRMENT OF FERTILITY SECTION.


13.1 Carcinogenesis, Mutagenesis, Impairment of Fertility. Carcinogenicity studies were conducted in male and female rats and mice. In the 80-week mouse oral study and in the 104-week rat oral study, no relationship of tumor incidence to tacrolimus dosage was found. The highest dose used in the mouse was 3.0 mg/kg/day (0.9 to 2.2 times the AUC at clinical doses of 0.075 to 0.2 mg/kg/day) and in the rat was 5.0 mg/kg/day (0.265 to 0.65 times the AUC at clinical doses of 0.075 to 0.2 mg/kg/day) [see Boxed Warning and Warnings and Precautions 5.2)]. 104-week dermal carcinogenicity study was performed in mice with tacrolimus ointment (0.03% to 3%), equivalent to tacrolimus doses of 1.1 to 118 mg/kg/day or 3.3 to 354 mg/m 2/day. In the study, the incidence of skin tumors was minimal and the topical application of tacrolimus was not associated with skin tumor formation under ambient room lighting. However, statistically significant elevation in the incidence of pleomorphic lymphoma in high dose male (25/50) and female animals (27/50) and in the incidence of undifferentiated lymphoma in high dose female animals (13/50) was noted in the mouse dermal carcinogenicity study. Lymphomas were noted in the mouse dermal carcinogenicity study at daily dose of 3.5 mg/kg (0.1% tacrolimus ointment). No drug-related tumors were noted in the mouse dermal carcinogenicity study at daily dose of 1.1 mg/kg (0.03% tacrolimus ointment). The relevance of topical administration of tacrolimus in the setting of systemic tacrolimus use is unknown. The implications of these carcinogenicity studies to the human condition are limited; doses of tacrolimus were administered that likely induced immunosuppression in these animals impairing their immune systems ability to inhibit unrelated carcinogenesis. No evidence of genotoxicity was seen in bacterial (Salmonella and E. coli) or mammalian (Chinese hamster lung-derived cells) in vitro assays of mutagenicity, the in vitro CHO/HGPRT assay of mutagenicity, or in vivo clastogenicity assays performed in mice; tacrolimus did not cause unscheduled DNA synthesis in rodent hepatocytes. Tacrolimus given orally at 1.0 mg/kg (0.8 to 2.2 times the clinical dose range of 0.075 to 0.2 mg/kg/day based on body surface area) to male and female rats, prior to and during mating, as well as to dams during gestation and lactation, was associated with embryolethality and adverse effects on female reproduction. Effects on female reproductive function (parturition) and embryolethal effects were indicated by higher rate of pre-implantation loss and increased numbers of undelivered and nonviable pups. When given at 3.2 mg/kg (2.6 to 6.9 times the clinical dose range based on body surface area), tacrolimus was associated with maternal and paternal toxicity as well as reproductive toxicity including marked adverse effects on estrus cycles, parturition, pup viability, and pup malformations.

CLINICAL PHARMACOLOGY SECTION.


12 CLINICAL PHARMACOLOGY. 12.1 Mechanism of Action. Tacrolimus inhibits T-lymphocyte activation, although the exact mechanism of action is not known. Experimental evidence suggests that tacrolimus binds to an intracellular protein, FKBP-12. complex of tacrolimus-FKBP-12, calcium, calmodulin, and calcineurin is then formed and the phosphatase activity of calcineurin inhibited. This effect may prevent the dephosphorylation and translocation of nuclear factor of activated T-cells (NF-AT), nuclear component thought to initiate gene transcription for the formation of lymphokines (such as interleukin-2, gamma interferon). The net result is the inhibition of T-lymphocyte activation (i.e., immunosuppression).Tacrolimus prolongs the survival of the host and transplanted graft in animal transplant models of liver, kidney, heart, bone marrow, small bowel and pancreas, lung and trachea, skin, cornea, and limb.In animals, tacrolimus has been demonstrated to suppress some humoral immunity and, to greater extent, cell-mediated reactions such as allograft rejection, delayed type hypersensitivity, collagen-induced arthritis, experimental allergic encephalomyelitis, and graft versus host disease.. 12.3 Pharmacokinetics. Tacrolimus activity is primarily due to the parent drug. The pharmacokinetic parameters (mean+-S.D.) of tacrolimus have been determined following intravenous (IV) and/or oral (PO) administration in healthy volunteers, and in kidney transplant, liver transplant, and heart transplant patients Table 14). Table 14. Pharmacokinetics Parameters (mean+-S.D.) of Tacrolimus in Healthy Volunteers and PatientsPopulationNRoute(Dose)ParametersC max (ng/mL)T max (hr)AUC(ngohr/mL)t 1/2 (hr)CI(L/hr/kg)V(L/kg)HealthyVolunteers8IV(0.025 mg/kg/4hr)not applicable598 AUC 0-120 +- 12534.2+- 7.70.040+- 0.0091.91+- 0.3116PO(5 mg)29.7+- 7.21.6+- 0.7243 AUC 0-72 +- 7334.8+- 11.4 0.041 Corrected for individual bioavailability +- 0.008 1.94 +- 0.53 KidneyTransplantPatients26IV(0.02 mg/kg/12 hr) 294 AUC 0-inf +- 26218.8+- 16.7 0.083+- 0.050 1.41+- 0.66 PO(0.2 mg/kg/day) 19.2+- 10.3 3.0203 +- 42not availablePO(0.3 mg/kg/day) 24.2+- 15.81.5288 +- 93 LiverTransplantPatients 17IV(0.05 mg/kg/12 hr) 3300 +- 213011.7+- 3.9 0.053+- 0.0170.85+- 0.30 PO(0.3 mg/kg/day) 68.5+- 30.0 2.3+- 1.5 519 +- 179 HeartTransplant Patients11IV (0.01 mg/kg/day as continuous infusion) 954 AUC 0-t +- 334 23.6 +- 9.22 0.051 +- 0.015 11PO (0.075 mg/kg/day) Determined after the first dose 14.7 7.79 2.1 [0.5-6.0] Median [range] 82.7 AUC 0-12 +- 63.2 14PO (0.15 mg/kg/day) 24.5 +- 13.7 1.5 [0.4-4.0] 142 +- 116 Due to intersubject variability in tacrolimus pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy [see Dosage and Administration (2.6)] Pharmacokinetic data indicate that whole blood concentrations rather than plasma concentrations serve as the more appropriate sampling compartment to describe tacrolimus pharmacokinetics. AbsorptionAbsorption of tacrolimus from the gastrointestinal tract after oral administration is incomplete and variable. The absolute bioavailability of tacrolimus was 17+-10% in adult kidney transplant patients (N=26), 22+-6% in adult liver transplant patients (N=17), 23+-9% in adult heart transplant patients (N= 11) and 18+-5% in healthy volunteers (N=16).A single dose trial conducted in 32 healthy volunteers established the bioequivalence of the mg and mg capsules. Another single dose trial in 32 healthy volunteers established the bioequivalence of the 0.5 mg and mg capsules. Tacrolimus maximum blood concentrations (C max) and area under the curve (AUC) appeared to increase in dose-proportional fashion in 18 fasted healthy volunteers receiving single oral dose of 3, 7, and 10 mg. In 18 kidney transplant patients, tacrolimus trough concentrations from to 30 ng/mL measured at 10 to 12 hours post-dose (Cmin) correlated well with the AUC (correlation coefficient 0.93). In 24 liver transplant patients over concentration range of 10 to 60 ng/mL, the correlation coefficient was 0.94. In 25 heart transplant patients over concentration range of to 24 ng/mL, the correlation coefficient was 0.89 after an oral dose of 0.075 or 0.15 mg/kg/day at steady-state. Food Effects The rate and extent of tacrolimus absorption were greatest under fasted conditions. The presence and composition of food decreased both the rate and extent of tacrolimus absorption when administered to 15 healthy volunteers.The effect was most pronounced with high-fat meal (848 kcal, 46% fat): mean AUC and max were decreased 37% and 77%, respectively; max was lengthened 5-fold. high-carbohydrate meal (668 kcal, 85% carbohydrate) decreased mean AUC and mean max by 28% and 65%, respectively. In healthy volunteers (N=16), the time of the meal also affected tacrolimus bioavailability. When given immediately following the meal, mean max was reduced 71%, and mean AUC was reduced 39%, relative to the fasted condition. When administered 1.5 hours following the meal, mean max was reduced 63%, and mean AUC was reduced 39%, relative to the fasted condition. In 11 liver transplant patients, tacrolimus administered 15 minutes after high fat (400 kcal, 34% fat) breakfast, resulted in decreased AUC (27+-18%) and max (50+-19%), as compared to fasted state. Tacrolimus capsules should be taken consistently every day either with or without food because the presence and composition of food decreases the bioavailability of tacrolimus [see Dosage and Administration (2.5)] DistributionThe plasma protein binding of tacrolimus is approximately 99% and is independent of concentration over range of to 50 ng/mL. Tacrolimus is bound mainly to albumin and alpha-1-acid glycoprotein, and has high level of association with erythrocytes. The distribution of tacrolimus between whole blood and plasma depends on several factors, such as hematocrit, temperature at the time of plasma separation, drug concentration, and plasma protein concentration. In U.S. trial, the ratio of whole blood concentration to plasma concentration averaged 35 (range 12 to 67).MetabolismTacrolimus is extensively metabolized by the mixed-function oxidase system, primarily the cytochrome P-450 system (CYP3A). metabolic pathway leading to the formation of possible metabolites has been proposed. Demethylation and hydroxylation were identified as the primary mechanisms of biotransformation in vitro. The major metabolite identified in incubations with human liver microsomes is 13-demethyl tacrolimus. In in vitro studies, 31-demethyl metabolite has been reported to have the same activity as tacrolimus. ExcretionThe mean clearance following IV administration of tacrolimus is 0.040, 0.083, and 0.053 and 0.051 L/hr/kg in healthy volunteers, adult kidney transplant patients, adult liver transplant patients, and adult heart transplant patients, respectively. In man, less than 1% of the dose administered is excreted unchanged in urine. In mass balance study of IV administered radiolabeled tacrolimus to healthy volunteers, the mean recovery of radiolabel was 77.8+-12.7%. Fecal elimination accounted for 92.4+-1.0% and the elimination half-life based on radioactivity was 48.1+-15.9 hours whereas it was 43.5+-11.6 hours based on tacrolimus concentrations. The mean clearance of radiolabel was 0.029+-0.015 L/hr/kg and clearance of tacrolimus was 0.029+-0.009 L/hr/kg. When administered PO, the mean recovery of the radiolabel was 94.9+-30.7%. Fecal elimination accounted for 92.6+-30.7%, urinary elimination accounted for 2.3+-1.1% and the elimination half-life based on radioactivity was 31.9+-10.5 hours whereas it was 48.4+-12.3 hours based on tacrolimus concentrations. The mean clearance of radiolabel was 0.226+-0.116 L/hr/kg and clearance of tacrolimus 0.172+-0.088 L/hr/kg.Specific PopulationsPediatricPharmacokinetics of tacrolimus have been studied in liver transplantation patients, 0.7 to 13.2 years of age. Following IV administration of 0.037 mg/kg/day dose to 12 pediatric patients, mean terminal half-life, volume of distribution and clearance were 11.5+-3.8 hours, 2.6+-2.1 L/kg and 0.138+-0.071 L/hr/kg, respectively. Following oral administration to patients, mean AUC and max were 337+-167 ngohr/mL and 48.4+-27.9 ng/mL, respectively. The absolute bioavailability was 31+-24%. Whole blood trough concentrations from 31 patients less than 12 years old showed that pediatric patients needed higher doses than adults to achieve similar tacrolimus trough concentrations [see Dosage and Administration 2.2)] Pharmacokinetics of tacrolimus have also been studied in kidney transplantation patients, 8.2+-2.4 years of age. Following IV infusion of 0.06 (range 0.06 to 0.09) mg/kg/day to 12 pediatric patients (8 male and female), mean terminal half-life and clearance were 10.2+-5.0 (range 3.4 to 25) hours and 0.12+-0.04 (range 0.06 to 0.17) L/hr/kg, respectively. Following oral administration to the same patients, mean AUC and max were 181+-65 (range 81 to 300) ngohr/mL and 30+-11 (range 14 to 49) ng/mL, respectively. The absolute bioavailability was 19+-14 (range 5.2 to 56) %. Renal and Hepatic ImpairmentThe mean pharmacokinetic parameters for tacrolimus following single administrations to patients with renal and hepatic impairment are given in Table 15. Table 15. Pharmacokinetic In Renal and Hepatic Impaired PatientsPopulation(No. of Patients)DoseAUC 0-t (ng.hr/mL)t 1/2 (hr)V(L/kg)CI(L/hr/kg)RenalImpairment(n=12) 0.02mg/kg/4hrIV 393+-123(t=60 hr) 26.3+-9.21.07+-0.20 0.038+-0.014 Mild Hepatic Impairment(n=6) 0.02mg/kg/4hrIV367+-107(t=72 hr)60.6+-43.8Range: 27.8 to 1413.1+-1.60.042+-0.027.7 mgPO 488+-320(t=72 hr) 66.1+-44.8Range: 29.5 to 138 3.7+-4.7 corrected for bioavailability 0.034+-0.019 SevereHepaticImpairment(n=6, IV)0.02 mg/kg/4hrIV (n=2)0.01 mg/kg/8hrIV (n=4)762+-204(t=120 hr)289+-117(t=144 hr)198+-158Range: 81 to 4363.9+-1.00.017+-0.013(n=5, PO) patient did not receive the PO dose mg PO(n=1)5 mg PO(n=4)4 mg PO(n=1) 658(t=120 hr)533+-156 (t=144 hr) 119+-35Range: 85 to 178 3.1+-3.4 0.016+-0.011 Renal Impairment: Tacrolimus pharmacokinetics following single IV administration were determined in 12 patients (7 not on dialysis and on dialysis, serum creatinine of 3.9+-1.6 and 12.0+-2.4 mg/dL, respectively) prior to their kidney transplant. The pharmacokinetic parameters obtained were similar for both groups. The mean clearance of tacrolimus in patients with renal dysfunction was similar to that in normal volunteers Table 15) [see Dosage and Administration (2.3) and Use in Specific Populations 8.6)] Hepatic Impairment: Tacrolimus pharmacokinetics have been determined in six patients with mild hepatic dysfunction (mean Pugh score: 6.2) following single IV and oral administrations. The mean clearance of tacrolimus in patients with mild hepatic dysfunction was not substantially different from that in normal volunteers (see previous table). Tacrolimus pharmacokinetics were studied in patients with severe hepatic dysfunction (mean Pugh score: >10). The mean clearance was substantially lower in patients with severe hepatic dysfunction, irrespective of the route of administration [see Dosage and Administration 2.4) and Use in Specific Populations 8.7)]. RaceThe pharmacokinetics of tacrolimus have been studied following single IV and oral administration of tacrolimus to 10 African-American, 12 Latino-American, and 12 Caucasian healthy volunteers. There were no significant pharmacokinetic differences among the three ethnic groups following 4-hour IV infusion of 0.015 mg/kg. However, after single oral administration of mg, mean (+-SD) tacrolimus max in African-Americans (23.6+-12.1 ng/mL) was significantly lower than in Caucasians (40.2+-12.6 ng/mL) and the Latino-Americans (36.2+-15.8 ng/mL) (p <0.01). Mean AUC0-inf tended to be lower in African-Americans (203+-115 ngohr/mL) than Caucasians (344+-186 ngohr/mL) and Latino-Americans (274+-150 ngohr/mL). The mean (+-SD) absolute oral bioavailability (F) in African-Americans (12+-4.5%) and Latino-Americans (14+-7.4%) was significantly lower than in Caucasians (19+-5.8%, p=0.011). There was no significant difference in mean terminal 1/2 among the three ethnic groups (range from approximately 25 to 30 hours). retrospective comparison of African-American and Caucasian kidney transplant patients indicated that African-American patients required higher tacrolimus doses to attain similar trough concentrations [see Dosage and Administration (2.1)]. GenderA formal trial to evaluate the effect of gender on tacrolimus pharmacokinetics has not been conducted, however, there was no difference in dosing by gender in the kidney transplant trial. retrospective comparison of pharmacokinetics in healthy volunteers, and in kidney, liver and heart transplant patients indicated no gender-based differences. Drug InteractionsFrequent monitoring of whole blood concentrations and appropriate dosage adjustments of tacrolimus are recommended when concomitant use of the following drugs with tacrolimus is initiated or discontinued [see Drug Interactions (7)] Telaprevir: In single dose study in healthy volunteers, coadministration of tacrolimus (0.5 mg single dose) with telaprevir (750 mg three times daily for 13 days) increased the tacrolimus dose-normalized max by 9.3-fold and AUC by 70-fold compared to tacrolimus alone [see Drug Interactions 7.3)]. Boceprevir: In single dose study in 12 subjects, coadministration of tacrolimus (0.5 mg single dose) with boceprevir (800 mg three times daily for 11 days) increased tacrolimus maxby 9.9-fold and AUC by 17-fold compared to tacrolimus alone [see Drug Interactions (7.3)]. Nelfinavir: Based on clinical study of liver transplant recipients, co-administration of tacrolimus with nelfinavir increased blood concentrations of tacrolimus significantly and, as result, reduction in the tacrolimus dose by an average of 16-fold was needed to maintain mean trough tacrolimus blood concentrations of 9.7 ng/mL. It is recommended to avoid concomitant use of tacrolimus and nelfinavir unless the benefits outweigh the risks [see Drug Interactions 7.3)]. Rifampin: In study of normal volunteers, significant decrease in tacrolimus oral bioavailability (14+-6% vs. 7+-3%) was observed with concomitant rifampin administration (600 mg). In addition, there was significant increase in tacrolimus clearance (0.036+-0.008 L/hr/kg vs. 0.053+-0.010 L/hr/kg) with concomitant rifampin administration [see Drug Interactions 7.7)]. Magnesium-aluminum-hydroxide: In single-dose crossover study in healthy volunteers, co-administration of tacrolimus and magnesium-aluminum-hydroxide resulted in 21% increase in the mean tacrolimus AUC and 10% decrease in the mean tacrolimus Cmax relative to tacrolimus administration alone [see Drug Interactions 7.10)]. Ketoconazole: In study of normal volunteers, significant increase in tacrolimus oral bioavailability (14+-5% vs. 30+-8%) was observed with concomitant ketoconazole administration (200 mg). The apparent oral clearance of tacrolimus during ketoconazole administration was significantly decreased compared to tacrolimus alone (0.430+-0.129 L/hr/kg vs. 0.148+-0.043 L/hr/kg). Overall, IV clearance of tacrolimus was not significantly changed by ketoconazole co-administration, although it was highly variable between patients [see Drug Interactions (7.4)]. Voriconazole (see complete prescribing information for VFEND(R)): Repeat oral dose administration of voriconazole (400 mg every 12 hours for one day, then 200 mg every 12 hours for days) increased tacrolimus (0.1 mg/kg single dose) Cmax and AUC in healthy subjects by an average of 2-fold (90% CI: 1.9, 2.5) and 3-fold (90% CI: 2.7, 3.8), respectively [see Drug Interactions (7.4)] Posaconazole (see complete prescribing information for Noxafil(R)): Repeat oral administration of posaconazole (400 mg twice daily for days) increased tacrolimus (0.05 mg/kg single dose) Cmax and AUC in healthy subjects by an average of 2-fold (90% CI: 2.01, 2.42) and 4.5-fold (90% CI 4.03, 5.19), respectively [see Drug Interactions (7.4)]. Caspofungin (see complete prescribing information for CANCIDAS(R)): Caspofungin reduced the blood AUC0-12 of tacrolimus by approximately 20%, peak blood concentration (C max) by 16%, and 12-hour blood concentration (C12hr) by 26% in healthy adult subjects when tacrolimus (2 doses of 0.1 mg/kg 12 hours apart) was administered on the 10th day of CANCIDAS (R) 70 mg daily, as compared to results from control period in which tacrolimus was administered alone [see Drug Interactions 7.4)]. (0.01 mg/kg/day as continuous infusion) 954 AUC 0-t +- 334 +- 9.22 +- 0.015 14.7 7.79 2.1 [0.5-6.0] Median [range] 82.7 AUC 0-12 +- 63.2 1.5 [0.4-4.0] 142 +- 116 In 18 kidney transplant patients, tacrolimus trough concentrations from to 30 ng/mL measured at 10 to 12 hours post-dose (Cmin) correlated well with the AUC (correlation coefficient 0.93). In 24 liver transplant patients over concentration range of 10 to 60 ng/mL, the correlation coefficient was 0.94. In 25 heart transplant patients over concentration range of to 24 ng/mL, the correlation coefficient was 0.89 after an oral dose of 0.075 or 0.15 mg/kg/day at steady-state.

CLINICAL STUDIES SECTION.


14 CLINICAL STUDIES. 14.1 Kidney Transplantation. Tacrolimus/azathioprine (AZA)Tacrolimus-based immunosuppression in conjunction with azathioprine and corticosteroids following kidney transplantation was assessed in randomized, multicenter, non-blinded, prospective trial. There were 412 kidney transplant patients enrolled at 19 clinical sites in the United States. Study therapy was initiated when renal function was stable as indicated by serum creatinine <= mg/dL (median of days after transplantation, range to 14 days). Patients less than years of age were excluded. There were 205 patients randomized to tacrolimus-based immunosuppression and 207 patients were randomized to cyclosporine-based immunosuppression. All patients received prophylactic induction therapy consisting of an antilymphocyte antibody preparation, corticosteroids and azathioprine. Overall year patient and graft survival was 96.1% and 89.6%, respectively.Data from this trial of tacrolimus in conjunction with azathioprine indicate that during the first three months of that trial, 80% of the patients maintained trough concentrations between to 20 ng/mL, and then between to 15 ng/mL, through year.Tacrolimus/mycophenolate mofetil (MMF)Tacrolimus-based immunosuppression in conjunction with MMF, corticosteroids, and induction has been studied. In randomized, open-label, multi-center trial (Study 1), 1589 kidney transplant patients received tacrolimus (Group C, n=401), sirolimus (Group D, n=399), or one of two cyclosporine (CsA) regimens (Group A, n=390 and Group B, n=399) in combination with MMF and corticosteroids; all patients, except those in one of the two cyclosporine groups, also received induction with daclizumab. The trial was conducted outside the United States; the trial population was 93% Caucasian. In this trial, mortality at 12 months in patients receiving tacrolimus/MMF was similar (3%) compared to patients receiving cyclosporine/MMF (3% and 2%) or sirolimus/MMF (3%). Patients in the tacrolimus group exhibited higher estimated creatinine clearance rates (eCLcr) using the Cockcroft-Gault formula (Table 16) and experienced fewer efficacy failures, defined as biopsy proven acute rejection (BPAR), graft loss, death, and/or lost to follow-up (Table 17) in comparison to each of the other three groups. Patients randomized to tacrolimus /MMF were more likely to develop diarrhea and diabetes after the transplantation and experienced similar rates of infections compared to patients randomized to either cyclosporine/MMF regimen [see Adverse Reactions 6.1)]. Table 16. Estimated Creatinine Clearance at 12 Months (Study 1) GroupeCL cr [mL/min] at Month 12 All death/graft loss (n=41, 27, 23 and 42 in Groups A, B, and D) and patients whose last recorded creatinine values were prior to month visit (n=10, 9, and in Groups A, B, and D, respectively) were inputed with Glomerular Filtration Rate (GFR) of 10 mL/min; subjects last observed creatinine value from month on was used for the remainder of subjects with missing creatinine at month 12 (n=11, 12, 15 and 19 for Groups A, B, and D, respectively). Weight was also imputed in the calculation of estimated GFR, if missing. NMEANSDMEDIANTreatment Difference with Group (99.2% CI Adjusted for multiple (6) pairwise comparisons using Bonferroni corrections.) (A) CsA/MMF/CS 39056.525.856.9-8.6 (-13.7, -3.7)(B) CsA/MMF/CS/Daclizumab39958.925.660.9-6.2 (-11.2, -1.2)(C) Tac/MMF/CS/Daclizumab 40165.127.466.2-(D) Siro/MMF/CS/Daclizumab 39956.227.457.3-8.9 (-14.1, -3.9)Total158959.226.860.5Key: CsA=Cyclosporine, CS=Corticosteroids, Tac=Tacrolimus, Siro=SirolimusTable 17. Incidence of BPAR, Graft Loss, Death or Loss to Follow-up at 12 Months (Study 1) Key: Group A=CsA/MMF/CS, B=CsA/MMF/CS/Daclizumab, C=Tac/MMF/CS/Daclizumab, and D=Siro/MMF/CS/DaclizumabGroup AN=390Group BN=399Group CN=401Group DN=399Overall Failure141 (36.2%)126 (31.6%)82 (20.4%)185 (46.4%)Components of efficacy failureBPAR113 (29.0%)106 (26.6%)60 (15.0%)152 (38.1%)Graft loss excluding death28 (7.2%)20 (5.0%)12 (3.0%)30 (7.5%)Mortality13 (3.3%)7 (1.8%)11 (2.7%)12 (3.0%)Lost to follow-up5 (1.3%)7 (1.8%)5 (1.3%)6 (1.5%)Treatment Difference of efficacy failure compared to Group (99.2% CI 15.8% (7.1%, 24.3%)11.2% (2.7%, 19.5%)-26.0% (17.2%, 34.7%)The protocol-specified target tacrolimus trough concentrations (C trough, Tac) were to ng/mL; however, the observed median troughs, Tac approximated ng/mL throughout the 12 month trial Table 18). Approximately 80% of patients maintained tacrolimus whole blood concentrations between to 11 ng/mL through year post-transplant. Table 18. Tacrolimus Whole Blood Trough Concentrations (Study 1)Time Median (P10 to P90 tacrolimus whole blood trough concentrations (ng/mL)Day 30 (N=366)6.9 (4.4 to 11.3)Day 90 (N=351)6.8 (4.1 to 10.7)Day 180 (N=355)6.5 (4.0 to 9.6)Day 365 (N=346)6.5 (3.8 to 10.0)The protocol-specified target cyclosporine trough concentrations (Ctrough, CsA) for Group were 50 to 100 ng/mL; however, the observed median troughs, CsA approximated 100 ng/mL throughout the 12 month trial. The protocol-specified target Ctroughs, CsA for Group were 150 to 300 ng/mL for the first months and 100 to 200 ng/mL from month to month 12; the observed median troughs, CsA approximated 225 ng/mL for the first months and 140 ng/mL from month to month 12. While patients in all groups started MMF at 1gram twice daily, the MMF dose was reduced to less than g per day in 63% of patients in the tacrolimus treatment arm by month 12 (Table 19); approximately 50% of these MMF dose reductions were due to adverse reactions. By comparison, the MMF dose was reduced to less than g per day in 49% and 45% of patients in the two cyclosporine arms (Group and Group B, respectively), by month 12 and approximately 40% of MMF dose reductions were due to adverse reactions. Table 19. MMF Dose Over Time in Tacrolimus/MMF (Group C) (Study 1)Key: Time-averaged MMF dose (total MMF dose)/(duration of treatment)Time period (Days)Time-averaged MMF dose (grams per day) Less than 2.02.0Greater than 2.00 to 30 (N=364)37%60%2%0 to 90 (N=373)47%51%2%0 to 180 (N=377)56%42%2%0 to 365 (N=380)63%36%1%In second randomized, open-label, multi-center trial (Study 2), 424 kidney transplant patients received tacrolimus (N=212) or cyclosporine (N=212) in combination with MMF gram twice daily, basiliximab induction, and corticosteroids. In this trial, the rate for the combined endpoint of BPAR, graft failure, death, and/or lost to follow-up at 12 months in the tacrolimus/MMF group was similar to the rate in the cyclosporine/MMF group. There was, however, an imbalance in mortality at 12 months in those patients receiving tacrolimus/MMF (4%) compared to those receiving cyclosporine/MMF (2%), including cases attributed to overimmunosuppression (Table 20). Table 20. Incidence of BPAR, Graft Loss, Death or Loss to Follow-up at 12 Months (Study 2)Tacrolimus/MMFCyclosporine/MMF(N=212)(N=212)Overall Failure32 (15.1%)36 (17.0%)Components of efficacy failureBPAR16 (7.5%)29 (13.7%)Graft loss excluding death6 (2.8%)4 (1.9%)Mortality9 (4.2%)5 (2.4%)Lost to follow-up4 (1.9%)1 (0.5%)Treatment Difference of efficacy failure compared to Tacrolimus/MMF group (95% CI 1.9% (-5.2%, 9.0%)The protocol-specified target tacrolimus whole blood trough concentrations (C trough,Tac) in Study were to 16 ng/mL for the first three months and to 15 ng/mL thereafter. The observed median troughs, Tac approximated 10 ng/mL during the first three months and ng/mL from month to month 12 Table 21). Approximately 80% of patients maintained tacrolimus whole trough blood concentrations between to 16 ng/mL during months through and, then, between to 12 ng/mL from month through year. Table 21. Tacrolimus Whole Blood Trough Concentrations (Study 2)Time Median (P10 to P90 tacrolimus whole blood trough concentrations (ng/mL)Day 30 (N=174)10.5 (6.3 to 16.8)Day 60 (N=179)9.2 (5.9 to 15.3)Day 120 (N=176)8.3 (4.6 to 13.3)Day 180 (N=171)7.8 (5.5 to 13.2)Day 365 (N=178)7.1 (4.2 to 12.4)The protocol-specified target cyclosporine whole blood concentrations (C trough, CsA) were 125 to 400 ng/mL for the first three months, and 100 to 300 ng/mL thereafter. The observed median troughs, CsA approximated 280 ng/mL during the first three months and 190 ng/mL from month to month 12. Patients in both groups started MMF at 1gram twice daily. The MMF dose was reduced to less than grams per day by month 12 in 62% of patients in the tacrolimus/MMF group Table 22) and in 47% of patients in the cyclosporine/MMF group. Approximately 63% and 55% of these MMF dose reductions were because of adverse reactions in the tacrolimus/MMF group and the cyclosporine/MMF group, respectively [see Adverse Reactions (6.1)]. Table 22. MMF Dose Over Time in the Tacrolimus/MMF Group (Study 2)Key: Time-averaged MMF dose=(total MMF dose)/(duration of treatment)Time period (Days)Time-averaged MMF dose (g/day) Less than 2.02.0Greater than 2.0 0-30 (N=212)25%69%6%0-90 (N=212)41%53%6%0-180 (N=212)52%41%7%0-365 (N=212)62%34%4%. 14.2 Liver Transplantation. The safety and efficacy of tacrolimus-based immunosuppression following orthotopic liver transplantation were assessed in two prospective, randomized, non-blinded multicenter trials. The active control groups were treated with cyclosporine-based immunosuppressive regimen (CsA/AZA). Both trials used concomitant adrenal corticosteroids as part of the immunosuppressive regimens. These trials compared patient and graft survival rates at 12 months following transplantation.In one trial, 529 patients were enrolled at 12 clinical sites in the United States; prior to surgery, 263 were randomized to the tacrolimus-based immunosuppressive regimen and 266 to the CsA/AZA. In 10 of the 12 sites, the same CsA/AZA protocol was used, while sites used different control protocols. This trial excluded patients with renal dysfunction, fulminant hepatic failure with Stage IV encephalopathy, and cancers; pediatric patients (<= 12 years old) were allowed.In the second trial, 545 patients were enrolled at clinical sites in Europe; prior to surgery, 270 were randomized to the tacrolimus-based immunosuppressive regimen and 275 to CsA/AZA. In this trial, each center used its local standard CsA/AZA protocol in the active-control arm. This trial excluded pediatric patients, but did allow enrollment of subjects with renal dysfunction, fulminant hepatic failure in Stage IV encephalopathy, and cancers other than primary hepatic with metastases.One-year patient survival and graft survival in the tacrolimus-based treatment groups were similar to those in the CsA/AZA treatment groups in both trials. The overall 1-year patient survival (CsA/AZA and tacrolimus-based treatment groups combined) was 88% in the U.S. trial and 78% in the European trial. The overall 1-year graft survival (CsA/AZA and tacrolimus-based treatment groups combined) was 81% in the U.S. trial and 73% in the European trial. In both trials, the median time to convert from IV to oral tacrolimus dosing was days.Although there is lack of direct correlation between tacrolimus concentrations and drug efficacy, data from clinical trials of liver transplant patients have shown an increasing incidence of adverse reactions with increasing trough blood concentrations. Most patients are stable when trough whole blood concentrations are maintained between to 20 ng/mL. Long-term post-transplant patients often are maintained at the low end of this target range. Data from the U.S. clinical trial show that the median trough blood concentrations, measured at intervals from the second week to one year post-transplantation ranged from 9.8 ng/mL to 19.4 ng/mL. 14.3 Heart Transplantation. Two open-label, randomized, comparative trials evaluated the safety and efficacy of tacrolimus-based and cyclosporine-based immunosuppression in primary orthotopic heart transplantation. In trial conducted in Europe, 314 patients received regimen of antibody induction, corticosteroids and azathioprine in combination with tacrolimus or cyclosporine modified for 18 months. In 3-arm trial conducted in the US, 331 patients received corticosteroids and tacrolimus plus sirolimus, tacrolimus plus mycophenolate mofetil (MMF) or cyclosporine modified plus MMF for year.In the European trial, patient/graft survival at 18 months post-transplant was similar between treatment arms, 92% in the tacrolimus group and 90% in the cyclosporine group. In the U.S. trial, patient and graft survival at 12 months was similar with 93% survival in the tacrolimus plus MMF group and 86% survival in the cyclosporine modified plus MMF group. In the European trial, the cyclosporine trough concentrations were above the pre-defined target range (i.e., 100 to 200 ng/mL) at Day 122 and beyond in 32 to 68% of the patients in the cyclosporine treatment arm, whereas the tacrolimus trough concentrations were within the pre-defined target range (i.e., to 15 ng/mL) in 74 to 86% of the patients in the tacrolimus treatment arm. Data from this European trial indicate that from week to months post-transplant, approximately 80% of patients maintained trough concentrations between to 20 ng/mL and, from months through 18 months post-transplant, approximately 80% of patients maintained trough concentrations between to 18 ng/mL. The U.S. trial contained third arm of combination regimen of sirolimus, mg per day, and full-dose tacrolimus; however, this regimen was associated with increased risk of wound healing complications, renal function impairment, and insulin-dependent post-transplant diabetes mellitus, and is not recommended [see Warnings and Precautions (5.12)] The U.S. trial contained third arm of combination regimen of sirolimus, mg per day, and full-dose tacrolimus; however, this regimen was associated with increased risk of wound healing complications, renal function impairment, and insulin-dependent post-transplant diabetes mellitus, and is not recommended [see Warnings and Precautions (5.12)].

CONTRAINDICATIONS SECTION.


4 CONTRAINDICATIONS. Tacrolimus capsules are contraindicated in patients with hypersensitivity to tacrolimus. Tacrolimus injection is contraindicated in patients with hypersensitivity to HCO-60 (polyoxyl 60 hydrogenated castor oil). Hypersensitivity symptoms reported include dyspnea, rash, pruritus, and acute respiratory distress syndrome [see Adverse Reactions (6)] . oHypersensitivity to tacrolimus or HCO-60 (polyoxyl 60 hydrogenated castor oil) (4) oHypersensitivity to tacrolimus or HCO-60 (polyoxyl 60 hydrogenated castor oil) (4).

DESCRIPTION SECTION.


11 DESCRIPTION. Tacrolimus capsules, USP are available for oral administration containing 0.5 mg, mg or mg of tacrolimus. Inactive ingredients include lactose monohydrate, hypromellose E5, croscarmellose sodium, and magnesium stearate. The 0.5 mg capsule shell contains gelatin, titanium dioxide, iron oxide yellow and sodium lauryl sulfate, the mg capsule shell contains gelatin, titanium dioxide and sodium lauryl sulfate, and the mg capsule shell contains gelatin, titanium dioxide, iron oxide red and sodium lauryl sulfate.Tacrolimus, previously known as FK506, is the active ingredient in tacrolimus. Tacrolimus is macrolide immunosuppressant produced by Streptomyces tsukubaensis. Chemically, tacrolimus is designated as [3 S-[3 R[ E(1S,3S,4 S)], S,5R,8S,9E,12R,14 R,15S,16 R,18S,19 S,26aR]] -5,6,8,11,12,13,14,15,16,17,18,19,24,25,26,26a-hexadecahydro-5,19-dihydroxy-3-[2-(4-hydroxy-3-methoxycyclohexyl)-1-methylethenyl]-14,16-dimethoxy-4,10,12,18-tetramethyl-8-(2-propenyl)-15,19-epoxy-3H-pyrido[2,1- c][1,4] oxaazacyclotricosine-1,7,20,21(4H,23H)-tetrone, monohydrate. The chemical structure of tacrolimus is:Tacrolimus has an empirical formula of C44H 69NO12oH 2O and formula weight of 822.03. Tacrolimus appears as white crystals or crystalline powder. It is practically insoluble in water, freely soluble in ethanol, and very soluble in methanol and chloroform. Structure.

DOSAGE & ADMINISTRATION SECTION.


2 DOSAGE AND ADMINISTRATION. Summary of Initial Oral Dosage Recommendation and Observed Whole Blood Trough Concentrations 2.1, 2.2). Patient PopulationRecommended Initial Oral Dosage (two divided doses every 12 hours)Observed Whole Blood Trough ConcentrationsAdult Kidney transplant In combination with azathioprine0.2 mg/kg/daymonth to 3: to 20 ng/mLmonth to 12: to 15 ng/mL In combination with MMF/IL-2 receptor antagonist 0.1 mg/kg/daymonth to 12: to 11 ng/mLAdult Liver transplant Pediatric Liver transplant 0.10 to 0.15 mg/kg/day0.15 to 0.20 mg/kg/day month to 12: to 20 ng/mLmonth to 12: to 20 ng/mL Adult Heart transplant 0.075 mg/kg/day month to 3: 10 to 20 ng/mL month >=4: to 15 ng/mL oCareful and frequent monitoring of tacrolimus trough concentrations is recommended; Black patients may require higher doses in order to achieve comparable trough concentrations (2.1) oHepatic/Renal impaired patients should receive doses at the lowest value of the recommended initial oral dosing range 2.3, 2.4) oAdminister capsules consistently with or without food; do not drink grapefruit juice (2.5, 7.2) Adult Heart transplant month to 3: 10 to 20 ng/mL. month >=4: to 15 ng/mL oCareful and frequent monitoring of tacrolimus trough concentrations is recommended; Black patients may require higher doses in order to achieve comparable trough concentrations (2.1) oHepatic/Renal impaired patients should receive doses at the lowest value of the recommended initial oral dosing range 2.3, 2.4) oAdminister capsules consistently with or without food; do not drink grapefruit juice (2.5, 7.2) 2.1 Dosage in Adult Kidney, Liver, or Heart Transplant Patients. The initial oral dosage recommendations for adult patients with kidney, liver, or heart transplants along with recommendations for whole blood trough concentrations are shown in Table 1. The initial dose of tacrolimus capsules should be administered no sooner than hours after transplantation in the liver and heart transplant patients. In kidney transplant patients, the initial dose of tacrolimus capsules may be administered within 24 hours of transplantation, but should be delayed until renal function has recovered. For blood concentration monitoring details see Dosage and Administration (2.6). Table 1. Summary of Initial Oral Dosage Recommendations and Observed Whole Blood Trough Concentrations in AdultsPatient PopulationRecommended Tacrolimus Capsules Initial Oral DosageNote: daily doses should be administered as two divided doses, every 12 hoursObserved Tacrolimus Whole Blood Trough ConcentrationsAdult kidney transplant patientsIn combination with azathioprine0.2 mg/kg/daymonth to 3: to 20 ng/mLmonth to 12: to 15 ng/mLIn combination with MMF/IL-2 receptor antagonist In second smaller trial, the initial dose of tacrolimus was 0.15 to 0.2 mg/kg/day and observed tacrolimus concentrations were to 16 ng/mL during month to and to 12 ng/mL during month to 12 [see Clinical Studies (14.1)]. 0.1 mg/kg/daymonth to 12: to 11 ng/mLAdult liver transplant patients0.10 to 0.15 mg/kg/daymonth to 12: to 20 ng/mLAdult heart transplant patients0.075 mg/kg/daymonth to : 10 to 20 ng/mLmonth >=4: to 15 ng/mLDosing should be titrated based on clinical assessments of rejection and tolerability. Lower tacrolimus capsules dosages than the recommended initial dosage may be sufficient as maintenance therapy. Adjunct therapy with adrenal corticosteroids is recommended early post-transplant. The data in kidney transplant patients indicate that the Black patients required higher dose to attain comparable trough concentrations compared to Caucasian patients (Table 2). Table 2. Comparative Dose and Trough Concentrations Based on RaceTime After TransplantCaucasian n=114 Blackn=56 Dose(mg/kg) Trough Concentrations(ng/mL) Dose(mg/kg) Trough Concentrations (ng/mL)Day 70.1812.00.2310.9Month 10.1712.80.2612.9Month 60.1411.80.2411.5Month 120.1310.10.1911.0Initial Dose InjectionTacrolimus injection should be used only as continuous IV infusion and when the patient cannot tolerate oral administration of tacrolimus capsules. Tacrolimus injection should be discontinued as soon as the patient can tolerate oral administration of tacrolimus capsules, usually within to days. In patient receiving an IV infusion, the first dose of oral therapy should be given to 12 hours after discontinuing the IV infusion. The observed trough concentrations described above pertain to oral administration of tacrolimus only; while monitoring tacrolimus concentrations in patients receiving tacrolimus injection as continuous IV infusion may have some utility, the observed concentrations will not represent comparable exposures to those estimated by the trough concentrations observed in patients on oral therapy.The recommended starting dose of tacrolimus injection is 0.03 to 0.05 mg/kg/day in kidney and liver transplant and 0.01 mg/kg/day in heart transplant given as continuous IV infusion. Adult patients should receive doses at the lower end of the dosing range. Concomitant adrenal corticosteroid therapy is recommended early post-transplantation.Anaphylactic reactions have occurred with injectables containing castor oil derivatives, such as tacrolimus injection [see Warnings and Precautions 5.11)] 2.2 Dosage in Pediatric Liver Transplant Patients. The initial oral dosage recommendations for pediatric patients with liver transplants along with recommendations for whole blood trough concentrations are shown in Table 3. For blood concentration monitoring details see Dosage and Administration (2.6). If necessary, pediatric patients may start on an IV dose of 0.03 to 0.05 mg/kg/day. Table 3. Summary of Initial Oral Dosage Recommendations and Observed Whole Blood Trough Concentrations in ChildrenPatient PopulationRecommended Tacrolimus Capsules Initial Oral DosageNote: daily doses should be administered as two divided doses, every 12 hours Observed Tacrolimus Whole Blood Trough ConcentrationsPediatric liver transplant patients0.15 to 0.20 mg/kg/dayMonth to 12: to 20 ng/mLPediatric liver transplantation patients without pre-existing renal or hepatic dysfunction have required and tolerated higher doses than adults to achieve similar blood concentrations.Experience in pediatric kidney and heart transplantation patients is limited.. 2.3 Dosage Adjustment in Patients with Renal Impairment. Due to its potential for nephrotoxicity, consideration should be given to dosing tacrolimus capsules at the lower end of the therapeutic dosing range in patients who have received liver or heart transplant and have pre-existing renal impairment. Further reductions in dose below the targeted range may be required. In kidney transplant patients with post-operative oliguria, the initial dose of tacrolimus capsules should be administered no sooner than hours and within 24 hours of transplantation, but may be delayed until renal function shows evidence of recovery.. 2.4 Dosage Adjustments in Patients with Hepatic Impairment. Due to the reduced clearance and prolonged half-life, patients with severe hepatic impairment (Child Pugh >= 10) may require lower doses of tacrolimus capsules. Close monitoring of blood concentrations is warranted. The use of tacrolimus capsules in liver transplant recipients experiencing post-transplant hepatic impairment may be associated with increased risk of developing renal insufficiency related to high whole-blood concentrations of tacrolimus. These patients should be monitored closely and dosage adjustments should be considered. Some evidence suggests that lower doses should be used in these patients [see Dosage and Administration 2.1), Use in Specific Populations (8.7) and Clinical Pharmacology 12.3)]. 2.5 Administration Instructions. It is recommended that patients initiate oral therapy with tacrolimus capsules if possible. Initial dosage and observed tacrolimus whole blood trough concentrations for adults are shown in Table and for pediatrics in Table [see Dosage and Administration 2.1, 2.2)]; for blood concentration monitoring details in kidney transplant patients [see Dosage and Administration 2.1)]. It is important to take tacrolimus capsules consistently every day either with or without food because the presence and composition of food decreases the bioavailability of tacrolimus capsules [see Clinical Pharmacology 12.3)]. Patients should not eat grapefruit or drink grapefruit juice in combination with tacrolimus capsules [see Drug Interactions 7.2)]. Tacrolimus capsules should not be used simultaneously with cyclosporine. Tacrolimus capsules or cyclosporine should be discontinued at least 24 hours before initiating the other. In the presence of elevated tacrolimus capsules or cyclosporine concentrations, dosing with the other drug usually should be further delayed.In patients unable to take oral tacrolimus capsules, therapy may be initiated with tacrolimus injection as continuous IV infusion. If IV therapy is necessary, conversion from IV to oral tacrolimus is recommended as soon as oral therapy can be tolerated. This usually occurs within to days. In patients receiving an IV infusion, the first dose of oral therapy should be given to 12 hours after discontinuing the IV infusion.. 2.6 Therapeutic Drug Monitoring Monitoring of tacrolimus blood concentrations in conjunction with other laboratory and clinical parameters is considered an essential aid to patient management for the evaluation of rejection, toxicity, dose adjustments and compliance. Observed whole blood trough concentrations can be found in Table 1. Factors influencing frequency of monitoring include but are not limited to hepatic or renal dysfunction, the addition or discontinuation of potentially interacting drugs and the post-transplant time. Blood concentration monitoring is not replacement for renal and liver function monitoring and tissue biopsies. Data from clinical trials show that tacrolimus whole blood concentrations were most variable during the first week post-transplantation. The relative risks of toxicity and efficacy failure are related to tacrolimus whole blood trough concentrations. Therefore, monitoring of whole blood trough concentrations is recommended to assist in the clinical evaluation of toxicity and efficacy failure.Methods commonly used for the assay of tacrolimus include high performance liquid chromatography with tandem mass spectrometric detection (HPLC/MS/MS) and immunoassays. Immunoassays may react with metabolites as well as parent compound. Therefore assay results obtained with immunoassays may have positive bias relative to results of HPLC/MS. The bias may depend upon the specific assay and laboratory. Comparison of the concentrations in published literature to patient concentrations using the current assays must be made with detailed knowledge of the assay methods and biological matrices employed. Whole blood is the matrix of choice and specimens should be collected into tubes containing ethylene diamine tetraacetic acid (EDTA) anti-coagulant. Heparin anti-coagulation is not recommended because of the tendency to form clots on storage. Samples which are not analyzed immediately should be stored at room temperature or in refrigerator and assayed within days; see assay instructions for specifics. If samples are to be kept longer they should be deep frozen at -20 C. One study showed drug recovery 90% for samples stored at -20 for months, with reduced recovery observed after months.

DOSAGE FORMS & STRENGTHS SECTION.


3 DOSAGE FORMS AND STRENGTHS. oTacrolimus capsules, USP are available as Oblong, hard capsule for oral administration contains tacrolimus as follows: o0.5 mg, light-yellow color, imprinted with TCR on the capsule cap and 0.5 on capsule body.o1 mg, white color, imprinted with TCR on the capsule cap and 1 on capsule body. o5 mg, pink color, imprinted with TCR on the capsule cap and 5 on capsule body.. oTacrolimus capsules, USP are available as Oblong, hard capsule for oral administration contains tacrolimus as follows: o0.5 mg, light-yellow color, imprinted with TCR on the capsule cap and 0.5 on capsule body.o1 mg, white color, imprinted with TCR on the capsule cap and 1 on capsule body. o5 mg, pink color, imprinted with TCR on the capsule cap and 5 on capsule body.. o0.5 mg, light-yellow color, imprinted with TCR on the capsule cap and 0.5 on capsule body.. o1 mg, white color, imprinted with TCR on the capsule cap and 1 on capsule body. o5 mg, pink color, imprinted with TCR on the capsule cap and 5 on capsule body.. oCapsules: 0.5 mg, mg and mg (3) oCapsules: 0.5 mg, mg and mg (3).

DRUG INTERACTIONS SECTION.


7 DRUG INTERACTIONS. Since tacrolimus is metabolized mainly by CYP3A enzymes, drugs or substances known to inhibit these enzymes may increase tacrolimus whole blood concentrations. Drugs known to induce CYP3A enzymes may decrease tacrolimus whole blood concentrations [see Warnings and Precautions 5.13) and Clinical Pharmacology 12.3)]. Dose adjustments may be needed along with frequent monitoring of tacrolimus whole blood trough concentrations when tacrolimus is administered with CYP3A inhibitors or inducers. In addition, patients should be monitored for adverse reactions including changes in renal function and QT prolongation [see Warnings and Precautions (5.7) and (5.14)] . oMycophenolic Acid Products: Can increase MPA exposure after crossover from cyclosporine to tacrolimus; monitor for MPA-related adverse reactions and adjust MMF or MPA-dose as needed (7.1) oNelfinavir and Grapefruit Juice: Increased tacrolimus concentrations via CYP3A inhibition; avoid concomitant use 7.2, 7.3) oCYP3A Inhibitors: Increased tacrolimus concentrations; monitor concentrations and adjust tacrolimus dose as needed with concomitant use (5.13, 7.3, 7.4, 7.5, 7.6) oCYP3A4 Inducers: Decreased tacrolimus concentrations; monitor concentrations and adjust tacrolimus dose as needed with concomitant use (5.13, 7.7, 7.8, 7.9) oMycophenolic Acid Products: Can increase MPA exposure after crossover from cyclosporine to tacrolimus; monitor for MPA-related adverse reactions and adjust MMF or MPA-dose as needed (7.1) oNelfinavir and Grapefruit Juice: Increased tacrolimus concentrations via CYP3A inhibition; avoid concomitant use 7.2, 7.3) oCYP3A Inhibitors: Increased tacrolimus concentrations; monitor concentrations and adjust tacrolimus dose as needed with concomitant use (5.13, 7.3, 7.4, 7.5, 7.6) oCYP3A4 Inducers: Decreased tacrolimus concentrations; monitor concentrations and adjust tacrolimus dose as needed with concomitant use (5.13, 7.7, 7.8, 7.9) 7.1 Mycophenolic Acid Products. With given dose of mycophenolic acid (MPA) products, exposure to MPA is higher with tacrolimus co-administration than with cyclosporine co-administration because cyclosporine interrupts the enterohepatic recirculation of MPA while tacrolimus does not. Clinicians should be aware that there is also potential for increased MPA exposure after crossover from cyclosporine to tacrolimus in patients concomitantly receiving MPA-containing products.. 7.2 Grapefruit Juice. Grapefruit juice inhibits CYP3A-enzymes resulting in increased tacrolimus whole blood trough concentrations, and patients should avoid eating grapefruit or drinking grapefruit juice with tacrolimus [see Dosage and Administration 2.5)] . 7.3 Protease Inhibitors. Most protease inhibitors inhibit CYP3A enzymes and may increase tacrolimus whole blood concentrations. It is recommended to avoid concomitant use of tacrolimus with nelfinavir unless the benefits outweigh the risks [see Clinical Pharmacology (12.3)] Whole blood concentrations of tacrolimus are markedly increased when co-administered with telaprevir or with boceprevir [see Clinical Pharmacology 12.3)]. Monitoring of tacrolimus whole blood concentrations and tacrolimus-associated adverse reactions, and appropriate adjustments in the dosing regimen of tacrolimus are recommended when tacrolimus and protease inhibitors (e.g., ritonavir, telaprevir, boceprevir) are used concomitantly. 7.4 Antifungal Agents. Frequent monitoring of whole blood concentrations and appropriate dosage adjustments of tacrolimus are recommended when concomitant use of the following antifungal drugs with tacrolimus is initiated or discontinued [see Clinical Pharmacology (12.3)] Azoles: Voriconazole, posaconazole, itraconazole, ketoconazole, fluconazole and clotrimazole inhibit CYP3A metabolism of tacrolimus and increase tacrolimus whole blood concentrations. When initiating therapy with voriconazole or posaconazole in patients already receiving tacrolimus, it is recommended that the tacrolimus dose be initially reduced to one-third of the original dose and the subsequent tacrolimus doses be adjusted based on the tacrolimus whole blood concentrations.Caspofungin is an inducer of CYP3A and decreases whole blood concentrations of tacrolimus. 7.5 Calcium Channel Blockers. Verapamil, diltiazem, nifedipine, and nicardipine inhibit CYP3A metabolism of tacrolimus and may increase tacrolimus whole blood concentrations. Monitoring of whole blood concentrations and appropriate dosage adjustments of tacrolimus are recommended when these calcium channel blocking drugs and tacrolimus are used concomitantly.. 7.6 Antibacterials. Erythromycin, clarithromycin, troleandomycin and chloramphenicol inhibit CYP3A metabolism of tacrolimus and may increase tacrolimus whole blood concentrations. Monitoring of blood concentrations and appropriate dosage adjustments of tacrolimus are recommended when these drugs and tacrolimus are used concomitantly.. 7.7 Antimycobacterials. Rifampin [see Clinical Pharmacology 12.3)] and rifabutin are inducers of CYP3A enzymes and may decrease tacrolimus whole blood concentrations. Monitoring of whole blood concentrations and appropriate dosage adjustments of tacrolimus are recommended when these antimycobacterial drugs and tacrolimus are used concomitantly. 7.8 Anticonvulsants. Phenytoin, carbamazepine and phenobarbital induce CYP3A enzymes and may decrease tacrolimus whole blood concentrations. Monitoring of whole blood concentrations and appropriate dosage adjustments of tacrolimus are recommended when these drugs and tacrolimus are used concomitantly.Concomitant administration of phenytoin with tacrolimus may also increase phenytoin plasma concentrations. Thus, frequent monitoring phenytoin plasma concentrations and adjusting the phenytoin dose as needed are recommended when tacrolimus and phenytoin are administered concomitantly.. 7.9 St. Johns Wort Hypericum perforatum) St. Johns Wort induces CYP3A enzymes and may decrease tacrolimus whole blood concentrations. Monitoring of whole blood concentrations and appropriate dosage adjustments of tacrolimus are recommended when St. Johns Wort and tacrolimus are co-administered. 7.10 Gastric Acid Suppressors/Neutralizers. Lansoprazole and omeprazole, as CYP2C19 and CYP3A4 substrates, may potentially inhibit the CYP3A4 metabolism of tacrolimus and thereby substantially increase tacrolimus whole blood concentrations, especially in transplant patients who are intermediate or poor CYP2C19 metabolizers, as compared to those patients who are efficient CYP2C19 metabolizers.Cimetidine may also inhibit the CYP3A4 metabolism of tacrolimus and thereby substantially increase tacrolimus whole blood concentrations.Coadministration with magnesium and aluminum hydroxide antacids increase tacrolimus whole blood concentrations [see Clinical Pharmacology 12.3)]. Monitoring of whole blood concentrations and appropriate dosage adjustments of tacrolimus are recommended when these drugs and tacrolimus are used concomitantly. 7.11 Others. Bromocriptine, nefazodone, metoclopramide, danazol, ethinyl estradiol, amiodarone, methylprednisolone and herbal products containing schisandra sphenanthera extracts may inhibit CYP3A metabolism of tacrolimus and increase tacrolimus whole blood concentrations. Monitoring of blood concentrations and appropriate dosage adjustments of tacrolimus are recommended when these drugs and tacrolimus are co-administered.

GERIATRIC USE SECTION.


8.5 Geriatric Use. Clinical trials of tacrolimus did not include sufficient numbers of subjects aged 65 and over to determine whether they respond differently from younger subjects. Other reported clinical experience has not identified differences in responses between the elderly and younger patients. In general, dose selection for an elderly patient should be cautious, usually starting at the low end of the dosing range, reflecting the greater frequency of decreased hepatic, renal, or cardiac function, and of concomitant disease or other drug therapy.

HOW SUPPLIED SECTION.


16 HOW SUPPLIED/STORAGE AND HANDLING. 16.1 Tacrolimus Capsules, USP. strength1 mg(containing mg tacrolimus)shape/colorcapsule/whitebranding on capsule cap/body TCR on cap and 1 on body 100 count carton (10 10 blisters)NDC 0904-6425-61Store and DispenseStore at 25C (77F); excursions permitted to 15C to 30C (59F to 86F).

INDICATIONS & USAGE SECTION.


1 INDICATIONS AND USAGE. Tacrolimus is calcineurin-inhibitor immunosuppressant indicated foroProphylaxis of organ rejection in patients receiving allogeneic liver, kidney or heart transplants (1.1, 1.2, 1.3) oUse concomitantly with adrenal corticosteroids; in kidney and heart transplant, use in conjunction with azathioprine or mycophenolate mofetil (MMF) (1.1, 1.2, 1.3) oLimitations of Use 1.4): oDo not use simultaneously with cyclosporineoIntravenous use reserved for patients who can not tolerate capsules orallyoUse with sirolimus is not recommended in liver and heart transplant; use with sirolimus in kidney transplant has not been established. oProphylaxis of organ rejection in patients receiving allogeneic liver, kidney or heart transplants (1.1, 1.2, 1.3) oUse concomitantly with adrenal corticosteroids; in kidney and heart transplant, use in conjunction with azathioprine or mycophenolate mofetil (MMF) (1.1, 1.2, 1.3) oLimitations of Use 1.4): oDo not use simultaneously with cyclosporineoIntravenous use reserved for patients who can not tolerate capsules orallyoUse with sirolimus is not recommended in liver and heart transplant; use with sirolimus in kidney transplant has not been established. oDo not use simultaneously with cyclosporine. oIntravenous use reserved for patients who can not tolerate capsules orally. oUse with sirolimus is not recommended in liver and heart transplant; use with sirolimus in kidney transplant has not been established. 1.1 Prophylaxis of Organ Rejection in Kidney Transplant. Tacrolimus capsules are indicated for the prophylaxis of organ rejection in patients receiving allogeneic kidney transplants. It is recommended that tacrolimus capsules be used concomitantly with azathioprine or mycophenolate mofetil (MMF) and adrenal corticosteroids [see Clinical Studies (14.1)] Therapeutic drug monitoring is recommended for all patients receiving tacrolimus capsules [see Dosage and Administration (2.6)]. 1.2 Prophylaxis of Organ Rejection in Liver Transplant. Tacrolimus capsules are indicated for the prophylaxis of organ rejection in patients receiving allogeneic liver transplants. It is recommended that tacrolimus capsules be used concomitantly with adrenal corticosteroids [see Clinical Studies (14.2) ]. Therapeutic drug monitoring is recommended for all patients receiving tacrolimus capsules [see Dosage and Administration (2.6)] . 1.3 Prophylaxis of Organ Rejection in Heart Transplant. Tacrolimus capsules are indicated for the prophylaxis of organ rejection in patients receiving allogeneic heart transplants. It is recommended that tacrolimus capsules be used concomitantly with azathioprine or mycophenolate mofetil (MMF) and adrenal corticosteroids [see Clinical Studies 14.3)] Therapeutic drug monitoring is recommended for all patients receiving tacrolimus capsules [see Dosage and Administration (2.6)]. Tacrolimus capsules are indicated for the prophylaxis of organ rejection in patients receiving allogeneic heart transplants. It is recommended that tacrolimus capsules be used concomitantly with azathioprine or mycophenolate mofetil (MMF) and adrenal corticosteroids [see Clinical Studies 14.3)] Therapeutic drug monitoring is recommended for all patients receiving tacrolimus capsules [see Dosage and Administration (2.6)]. 1.4 Limitations of Use. Tacrolimus capsules should not be used simultaneously with cyclosporine [see Dosage and Administration (2.5)]. Tacrolimus injection should be reserved for patients unable to take tacrolimus capsules orally [see Dosage and Administration (2.1) and Warnings and Precautions (5.11)] Use with sirolimus is not recommended in liver and heart transplant. The safety and efficacy of tacrolimus capsules with sirolimus has not been established in kidney transplant [see Warnings and Precautions 5.12)].

INFORMATION FOR PATIENTS SECTION.


17 PATIENT COUNSELING INFORMATION. 17.1 Administration. Advise patients to: oTake tacrolimus capsules at the same 12-hour intervals everyday to achieve consistent blood concentrations.oTake tacrolimus capsules consistently either with or without food because the presence and composition of food decreases the bioavailability of tacrolimus capsules.oNot to eat grapefruit or drink grapefruit juice in combination with tacrolimus capsules [see Drug Interactions 7.2)]. oTake tacrolimus capsules at the same 12-hour intervals everyday to achieve consistent blood concentrations.. oTake tacrolimus capsules consistently either with or without food because the presence and composition of food decreases the bioavailability of tacrolimus capsules.. oNot to eat grapefruit or drink grapefruit juice in combination with tacrolimus capsules [see Drug Interactions 7.2)]. 17.2 Development of Lymphoma and Other Malignancies. Inform patients they are at increased risk of developing lymphomas and other malignancies, particularly of the skin, due to immunosuppression. Advise patients to limit exposure to sunlight and ultraviolet (UV) light by wearing protective clothing and use sunscreen with high protection factor [see Warnings and Precautions 5.2)] . 17.3 Increased Risk of Infection. Inform patients they are at increased risk of developing variety of infections, including opportunistic infections, due to immunosuppression and to contact their physician if they develop any symptoms of infection [see Warnings and Precautions 5.3, 5.4, 5.5)] . 17.4 New Onset Diabetes After Transplant. Inform patients that tacrolimus capsules can cause diabetes mellitus and should be advised to contact their physician if they develop frequent urination, increased thirst or hunger [see Warnings and Precautions (5.6)] . 17.5 Nephrotoxicity. Inform patients that tacrolimus capsules can have toxic effects on the kidney that should be monitored. Advise patients to attend all visits and complete all blood tests ordered by their medical team [see Warnings and Precautions 5.7)]. 17.6 Neurotoxicity. Inform patients that they are at risk of developing adverse neurologic effects including seizure, altered mental status, and tremor. Advise patients to contact their physician should they develop vision changes, deliriums, or tremors [see Warnings and Precautions (5.8)] . 17.7 Hyperkalemia. Inform patients that tacrolimus capsules can cause hyperkalemia. Monitoring of potassium levels may be necessary, especially with concomitant use of other drugs known to cause hyperkalemia [see Warnings and Precautions 5.9)] . 17.8 Hypertension. Inform patients that tacrolimus capsules can cause high blood pressure which may require treatment with anti-hypertensive therapy [see Warnings and Precautions (5.10)] . 17.9 Drug Interactions. Instruct patients to tell their health care providers when they start or stop taking all the medicines, including prescription medicines and non-prescription medicines, natural or herbal remedies, nutritional supplements and vitamins [see Drug Interactions 7)]. 17.10 Pregnant Women and Nursing Mothers. Instruct patients to tell their healthcare provider if they plan to become pregnant or breast-feed their infant [see Use in Specific Populations 8.1, 8.3)] 17.11 Immunizations. Inform patients that tacrolimus capsules can interfere with the usual response to immunizations and that they should avoid live vaccines [see Warnings and Precautions 5.16)]. Rx onlyManufactured For:Accord Healthcare, Inc.,1009 Slater Road,Suite 210-B,Durham, NC 27703, USA. Manufactured By:Intas Pharmaceuticals Limited,Plot No 457, 458,Village Matoda,Bavla Road, Ta.- Sanand,Dist.- Ahmedabad 382210.India.10 0108 679378Distributed By:MAJOR(R) PHARMACEUTICALS17177 Laurel Park Dr., Suite 233Livonia, MI 48152Refer to package label for Distributors NDC Number Issued November 2017.

NONCLINICAL TOXICOLOGY SECTION.


13 NONCLINICAL TOXICOLOGY. 13.1 Carcinogenesis, Mutagenesis, Impairment of Fertility. Carcinogenicity studies were conducted in male and female rats and mice. In the 80-week mouse oral study and in the 104-week rat oral study, no relationship of tumor incidence to tacrolimus dosage was found. The highest dose used in the mouse was 3.0 mg/kg/day (0.9 to 2.2 times the AUC at clinical doses of 0.075 to 0.2 mg/kg/day) and in the rat was 5.0 mg/kg/day (0.265 to 0.65 times the AUC at clinical doses of 0.075 to 0.2 mg/kg/day) [see Boxed Warning and Warnings and Precautions 5.2)]. 104-week dermal carcinogenicity study was performed in mice with tacrolimus ointment (0.03% to 3%), equivalent to tacrolimus doses of 1.1 to 118 mg/kg/day or 3.3 to 354 mg/m 2/day. In the study, the incidence of skin tumors was minimal and the topical application of tacrolimus was not associated with skin tumor formation under ambient room lighting. However, statistically significant elevation in the incidence of pleomorphic lymphoma in high dose male (25/50) and female animals (27/50) and in the incidence of undifferentiated lymphoma in high dose female animals (13/50) was noted in the mouse dermal carcinogenicity study. Lymphomas were noted in the mouse dermal carcinogenicity study at daily dose of 3.5 mg/kg (0.1% tacrolimus ointment). No drug-related tumors were noted in the mouse dermal carcinogenicity study at daily dose of 1.1 mg/kg (0.03% tacrolimus ointment). The relevance of topical administration of tacrolimus in the setting of systemic tacrolimus use is unknown. The implications of these carcinogenicity studies to the human condition are limited; doses of tacrolimus were administered that likely induced immunosuppression in these animals impairing their immune systems ability to inhibit unrelated carcinogenesis. No evidence of genotoxicity was seen in bacterial (Salmonella and E. coli) or mammalian (Chinese hamster lung-derived cells) in vitro assays of mutagenicity, the in vitro CHO/HGPRT assay of mutagenicity, or in vivo clastogenicity assays performed in mice; tacrolimus did not cause unscheduled DNA synthesis in rodent hepatocytes. Tacrolimus given orally at 1.0 mg/kg (0.8 to 2.2 times the clinical dose range of 0.075 to 0.2 mg/kg/day based on body surface area) to male and female rats, prior to and during mating, as well as to dams during gestation and lactation, was associated with embryolethality and adverse effects on female reproduction. Effects on female reproductive function (parturition) and embryolethal effects were indicated by higher rate of pre-implantation loss and increased numbers of undelivered and nonviable pups. When given at 3.2 mg/kg (2.6 to 6.9 times the clinical dose range based on body surface area), tacrolimus was associated with maternal and paternal toxicity as well as reproductive toxicity including marked adverse effects on estrus cycles, parturition, pup viability, and pup malformations.

NURSING MOTHERS SECTION.


8.3 Nursing Mothers. Tacrolimus is excreted in human milk. As the effect of chronic exposure to tacrolimus in healthy infants is not established, patients maintained on tacrolimus should discontinue nursing taking into consideration importance of drug to the mother.

OVERDOSAGE SECTION.


10 OVERDOSAGE. Limited overdosage experience is available. Acute overdosages of up to 30 times the intended dose have been reported. Almost all cases have been asymptomatic and all patients recovered with no sequelae. Acute overdosage was sometimes followed by adverse reactions consistent with those listed in Adverse Reactions (6) (including tremors, abnormal renal function, hypertension, and peripheral edema); in one case of acute overdosage, transient urticaria and lethargy were observed. Based on the poor aqueous solubility and extensive erythrocyte and plasma protein binding, it is anticipated that tacrolimus is not dialyzable to any significant extent; there is no experience with charcoal hemoperfusion. The oral use of activated charcoal has been reported in treating acute overdoses, but experience has not been sufficient to warrant recommending its use. General supportive measures and treatment of specific symptoms should be followed in all cases of overdosage. In acute oral and IV toxicity studies, mortalities were seen at or above the following doses: in adult rats, 52 times the recommended human oral dose; in immature rats, 16 times the recommended oral dose; and in adult rats, 16 times the recommended human IV dose (all based on body surface area corrections).

PACKAGE LABEL.PRINCIPAL DISPLAY PANEL.


Package/Label Display Panel Tacrolimus Capsules USP1 mg100 Capsules. carton label.

PEDIATRIC USE SECTION.


8.4 Pediatric Use. The safety and efficacy of tacrolimus in pediatric kidney and heart transplant patients have not been established. Successful liver transplants have been performed in pediatric patients (ages up to 16 years) using tacrolimus. Two randomized active-controlled trials of tacrolimus in primary liver transplantation included 56 pediatric patients. Thirty-one patients were randomized to tacrolimus-based and 25 to cyclosporine-based therapies. Additionally, minimum of 122 pediatric patients were studied in an uncontrolled trial of tacrolimus in living related donor liver transplantation. Pediatric patients generally required higher doses of tacrolimus to maintain blood trough concentrations of tacrolimus similar to adult patients [see Dosage and Administration 2.2)].

PHARMACOKINETICS SECTION.


12.3 Pharmacokinetics. Tacrolimus activity is primarily due to the parent drug. The pharmacokinetic parameters (mean+-S.D.) of tacrolimus have been determined following intravenous (IV) and/or oral (PO) administration in healthy volunteers, and in kidney transplant, liver transplant, and heart transplant patients Table 14). Table 14. Pharmacokinetics Parameters (mean+-S.D.) of Tacrolimus in Healthy Volunteers and PatientsPopulationNRoute(Dose)ParametersC max (ng/mL)T max (hr)AUC(ngohr/mL)t 1/2 (hr)CI(L/hr/kg)V(L/kg)HealthyVolunteers8IV(0.025 mg/kg/4hr)not applicable598 AUC 0-120 +- 12534.2+- 7.70.040+- 0.0091.91+- 0.3116PO(5 mg)29.7+- 7.21.6+- 0.7243 AUC 0-72 +- 7334.8+- 11.4 0.041 Corrected for individual bioavailability +- 0.008 1.94 +- 0.53 KidneyTransplantPatients26IV(0.02 mg/kg/12 hr) 294 AUC 0-inf +- 26218.8+- 16.7 0.083+- 0.050 1.41+- 0.66 PO(0.2 mg/kg/day) 19.2+- 10.3 3.0203 +- 42not availablePO(0.3 mg/kg/day) 24.2+- 15.81.5288 +- 93 LiverTransplantPatients 17IV(0.05 mg/kg/12 hr) 3300 +- 213011.7+- 3.9 0.053+- 0.0170.85+- 0.30 PO(0.3 mg/kg/day) 68.5+- 30.0 2.3+- 1.5 519 +- 179 HeartTransplant Patients11IV (0.01 mg/kg/day as continuous infusion) 954 AUC 0-t +- 334 23.6 +- 9.22 0.051 +- 0.015 11PO (0.075 mg/kg/day) Determined after the first dose 14.7 7.79 2.1 [0.5-6.0] Median [range] 82.7 AUC 0-12 +- 63.2 14PO (0.15 mg/kg/day) 24.5 +- 13.7 1.5 [0.4-4.0] 142 +- 116 Due to intersubject variability in tacrolimus pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy [see Dosage and Administration (2.6)] Pharmacokinetic data indicate that whole blood concentrations rather than plasma concentrations serve as the more appropriate sampling compartment to describe tacrolimus pharmacokinetics. AbsorptionAbsorption of tacrolimus from the gastrointestinal tract after oral administration is incomplete and variable. The absolute bioavailability of tacrolimus was 17+-10% in adult kidney transplant patients (N=26), 22+-6% in adult liver transplant patients (N=17), 23+-9% in adult heart transplant patients (N= 11) and 18+-5% in healthy volunteers (N=16).A single dose trial conducted in 32 healthy volunteers established the bioequivalence of the mg and mg capsules. Another single dose trial in 32 healthy volunteers established the bioequivalence of the 0.5 mg and mg capsules. Tacrolimus maximum blood concentrations (C max) and area under the curve (AUC) appeared to increase in dose-proportional fashion in 18 fasted healthy volunteers receiving single oral dose of 3, 7, and 10 mg. In 18 kidney transplant patients, tacrolimus trough concentrations from to 30 ng/mL measured at 10 to 12 hours post-dose (Cmin) correlated well with the AUC (correlation coefficient 0.93). In 24 liver transplant patients over concentration range of 10 to 60 ng/mL, the correlation coefficient was 0.94. In 25 heart transplant patients over concentration range of to 24 ng/mL, the correlation coefficient was 0.89 after an oral dose of 0.075 or 0.15 mg/kg/day at steady-state. Food Effects The rate and extent of tacrolimus absorption were greatest under fasted conditions. The presence and composition of food decreased both the rate and extent of tacrolimus absorption when administered to 15 healthy volunteers.The effect was most pronounced with high-fat meal (848 kcal, 46% fat): mean AUC and max were decreased 37% and 77%, respectively; max was lengthened 5-fold. high-carbohydrate meal (668 kcal, 85% carbohydrate) decreased mean AUC and mean max by 28% and 65%, respectively. In healthy volunteers (N=16), the time of the meal also affected tacrolimus bioavailability. When given immediately following the meal, mean max was reduced 71%, and mean AUC was reduced 39%, relative to the fasted condition. When administered 1.5 hours following the meal, mean max was reduced 63%, and mean AUC was reduced 39%, relative to the fasted condition. In 11 liver transplant patients, tacrolimus administered 15 minutes after high fat (400 kcal, 34% fat) breakfast, resulted in decreased AUC (27+-18%) and max (50+-19%), as compared to fasted state. Tacrolimus capsules should be taken consistently every day either with or without food because the presence and composition of food decreases the bioavailability of tacrolimus [see Dosage and Administration (2.5)] DistributionThe plasma protein binding of tacrolimus is approximately 99% and is independent of concentration over range of to 50 ng/mL. Tacrolimus is bound mainly to albumin and alpha-1-acid glycoprotein, and has high level of association with erythrocytes. The distribution of tacrolimus between whole blood and plasma depends on several factors, such as hematocrit, temperature at the time of plasma separation, drug concentration, and plasma protein concentration. In U.S. trial, the ratio of whole blood concentration to plasma concentration averaged 35 (range 12 to 67).MetabolismTacrolimus is extensively metabolized by the mixed-function oxidase system, primarily the cytochrome P-450 system (CYP3A). metabolic pathway leading to the formation of possible metabolites has been proposed. Demethylation and hydroxylation were identified as the primary mechanisms of biotransformation in vitro. The major metabolite identified in incubations with human liver microsomes is 13-demethyl tacrolimus. In in vitro studies, 31-demethyl metabolite has been reported to have the same activity as tacrolimus. ExcretionThe mean clearance following IV administration of tacrolimus is 0.040, 0.083, and 0.053 and 0.051 L/hr/kg in healthy volunteers, adult kidney transplant patients, adult liver transplant patients, and adult heart transplant patients, respectively. In man, less than 1% of the dose administered is excreted unchanged in urine. In mass balance study of IV administered radiolabeled tacrolimus to healthy volunteers, the mean recovery of radiolabel was 77.8+-12.7%. Fecal elimination accounted for 92.4+-1.0% and the elimination half-life based on radioactivity was 48.1+-15.9 hours whereas it was 43.5+-11.6 hours based on tacrolimus concentrations. The mean clearance of radiolabel was 0.029+-0.015 L/hr/kg and clearance of tacrolimus was 0.029+-0.009 L/hr/kg. When administered PO, the mean recovery of the radiolabel was 94.9+-30.7%. Fecal elimination accounted for 92.6+-30.7%, urinary elimination accounted for 2.3+-1.1% and the elimination half-life based on radioactivity was 31.9+-10.5 hours whereas it was 48.4+-12.3 hours based on tacrolimus concentrations. The mean clearance of radiolabel was 0.226+-0.116 L/hr/kg and clearance of tacrolimus 0.172+-0.088 L/hr/kg.Specific PopulationsPediatricPharmacokinetics of tacrolimus have been studied in liver transplantation patients, 0.7 to 13.2 years of age. Following IV administration of 0.037 mg/kg/day dose to 12 pediatric patients, mean terminal half-life, volume of distribution and clearance were 11.5+-3.8 hours, 2.6+-2.1 L/kg and 0.138+-0.071 L/hr/kg, respectively. Following oral administration to patients, mean AUC and max were 337+-167 ngohr/mL and 48.4+-27.9 ng/mL, respectively. The absolute bioavailability was 31+-24%. Whole blood trough concentrations from 31 patients less than 12 years old showed that pediatric patients needed higher doses than adults to achieve similar tacrolimus trough concentrations [see Dosage and Administration 2.2)] Pharmacokinetics of tacrolimus have also been studied in kidney transplantation patients, 8.2+-2.4 years of age. Following IV infusion of 0.06 (range 0.06 to 0.09) mg/kg/day to 12 pediatric patients (8 male and female), mean terminal half-life and clearance were 10.2+-5.0 (range 3.4 to 25) hours and 0.12+-0.04 (range 0.06 to 0.17) L/hr/kg, respectively. Following oral administration to the same patients, mean AUC and max were 181+-65 (range 81 to 300) ngohr/mL and 30+-11 (range 14 to 49) ng/mL, respectively. The absolute bioavailability was 19+-14 (range 5.2 to 56) %. Renal and Hepatic ImpairmentThe mean pharmacokinetic parameters for tacrolimus following single administrations to patients with renal and hepatic impairment are given in Table 15. Table 15. Pharmacokinetic In Renal and Hepatic Impaired PatientsPopulation(No. of Patients)DoseAUC 0-t (ng.hr/mL)t 1/2 (hr)V(L/kg)CI(L/hr/kg)RenalImpairment(n=12) 0.02mg/kg/4hrIV 393+-123(t=60 hr) 26.3+-9.21.07+-0.20 0.038+-0.014 Mild Hepatic Impairment(n=6) 0.02mg/kg/4hrIV367+-107(t=72 hr)60.6+-43.8Range: 27.8 to 1413.1+-1.60.042+-0.027.7 mgPO 488+-320(t=72 hr) 66.1+-44.8Range: 29.5 to 138 3.7+-4.7 corrected for bioavailability 0.034+-0.019 SevereHepaticImpairment(n=6, IV)0.02 mg/kg/4hrIV (n=2)0.01 mg/kg/8hrIV (n=4)762+-204(t=120 hr)289+-117(t=144 hr)198+-158Range: 81 to 4363.9+-1.00.017+-0.013(n=5, PO) patient did not receive the PO dose mg PO(n=1)5 mg PO(n=4)4 mg PO(n=1) 658(t=120 hr)533+-156 (t=144 hr) 119+-35Range: 85 to 178 3.1+-3.4 0.016+-0.011 Renal Impairment: Tacrolimus pharmacokinetics following single IV administration were determined in 12 patients (7 not on dialysis and on dialysis, serum creatinine of 3.9+-1.6 and 12.0+-2.4 mg/dL, respectively) prior to their kidney transplant. The pharmacokinetic parameters obtained were similar for both groups. The mean clearance of tacrolimus in patients with renal dysfunction was similar to that in normal volunteers Table 15) [see Dosage and Administration (2.3) and Use in Specific Populations 8.6)] Hepatic Impairment: Tacrolimus pharmacokinetics have been determined in six patients with mild hepatic dysfunction (mean Pugh score: 6.2) following single IV and oral administrations. The mean clearance of tacrolimus in patients with mild hepatic dysfunction was not substantially different from that in normal volunteers (see previous table). Tacrolimus pharmacokinetics were studied in patients with severe hepatic dysfunction (mean Pugh score: >10). The mean clearance was substantially lower in patients with severe hepatic dysfunction, irrespective of the route of administration [see Dosage and Administration 2.4) and Use in Specific Populations 8.7)]. RaceThe pharmacokinetics of tacrolimus have been studied following single IV and oral administration of tacrolimus to 10 African-American, 12 Latino-American, and 12 Caucasian healthy volunteers. There were no significant pharmacokinetic differences among the three ethnic groups following 4-hour IV infusion of 0.015 mg/kg. However, after single oral administration of mg, mean (+-SD) tacrolimus max in African-Americans (23.6+-12.1 ng/mL) was significantly lower than in Caucasians (40.2+-12.6 ng/mL) and the Latino-Americans (36.2+-15.8 ng/mL) (p <0.01). Mean AUC0-inf tended to be lower in African-Americans (203+-115 ngohr/mL) than Caucasians (344+-186 ngohr/mL) and Latino-Americans (274+-150 ngohr/mL). The mean (+-SD) absolute oral bioavailability (F) in African-Americans (12+-4.5%) and Latino-Americans (14+-7.4%) was significantly lower than in Caucasians (19+-5.8%, p=0.011). There was no significant difference in mean terminal 1/2 among the three ethnic groups (range from approximately 25 to 30 hours). retrospective comparison of African-American and Caucasian kidney transplant patients indicated that African-American patients required higher tacrolimus doses to attain similar trough concentrations [see Dosage and Administration (2.1)]. GenderA formal trial to evaluate the effect of gender on tacrolimus pharmacokinetics has not been conducted, however, there was no difference in dosing by gender in the kidney transplant trial. retrospective comparison of pharmacokinetics in healthy volunteers, and in kidney, liver and heart transplant patients indicated no gender-based differences. Drug InteractionsFrequent monitoring of whole blood concentrations and appropriate dosage adjustments of tacrolimus are recommended when concomitant use of the following drugs with tacrolimus is initiated or discontinued [see Drug Interactions (7)] Telaprevir: In single dose study in healthy volunteers, coadministration of tacrolimus (0.5 mg single dose) with telaprevir (750 mg three times daily for 13 days) increased the tacrolimus dose-normalized max by 9.3-fold and AUC by 70-fold compared to tacrolimus alone [see Drug Interactions 7.3)]. Boceprevir: In single dose study in 12 subjects, coadministration of tacrolimus (0.5 mg single dose) with boceprevir (800 mg three times daily for 11 days) increased tacrolimus maxby 9.9-fold and AUC by 17-fold compared to tacrolimus alone [see Drug Interactions (7.3)]. Nelfinavir: Based on clinical study of liver transplant recipients, co-administration of tacrolimus with nelfinavir increased blood concentrations of tacrolimus significantly and, as result, reduction in the tacrolimus dose by an average of 16-fold was needed to maintain mean trough tacrolimus blood concentrations of 9.7 ng/mL. It is recommended to avoid concomitant use of tacrolimus and nelfinavir unless the benefits outweigh the risks [see Drug Interactions 7.3)]. Rifampin: In study of normal volunteers, significant decrease in tacrolimus oral bioavailability (14+-6% vs. 7+-3%) was observed with concomitant rifampin administration (600 mg). In addition, there was significant increase in tacrolimus clearance (0.036+-0.008 L/hr/kg vs. 0.053+-0.010 L/hr/kg) with concomitant rifampin administration [see Drug Interactions 7.7)]. Magnesium-aluminum-hydroxide: In single-dose crossover study in healthy volunteers, co-administration of tacrolimus and magnesium-aluminum-hydroxide resulted in 21% increase in the mean tacrolimus AUC and 10% decrease in the mean tacrolimus Cmax relative to tacrolimus administration alone [see Drug Interactions 7.10)]. Ketoconazole: In study of normal volunteers, significant increase in tacrolimus oral bioavailability (14+-5% vs. 30+-8%) was observed with concomitant ketoconazole administration (200 mg). The apparent oral clearance of tacrolimus during ketoconazole administration was significantly decreased compared to tacrolimus alone (0.430+-0.129 L/hr/kg vs. 0.148+-0.043 L/hr/kg). Overall, IV clearance of tacrolimus was not significantly changed by ketoconazole co-administration, although it was highly variable between patients [see Drug Interactions (7.4)]. Voriconazole (see complete prescribing information for VFEND(R)): Repeat oral dose administration of voriconazole (400 mg every 12 hours for one day, then 200 mg every 12 hours for days) increased tacrolimus (0.1 mg/kg single dose) Cmax and AUC in healthy subjects by an average of 2-fold (90% CI: 1.9, 2.5) and 3-fold (90% CI: 2.7, 3.8), respectively [see Drug Interactions (7.4)] Posaconazole (see complete prescribing information for Noxafil(R)): Repeat oral administration of posaconazole (400 mg twice daily for days) increased tacrolimus (0.05 mg/kg single dose) Cmax and AUC in healthy subjects by an average of 2-fold (90% CI: 2.01, 2.42) and 4.5-fold (90% CI 4.03, 5.19), respectively [see Drug Interactions (7.4)]. Caspofungin (see complete prescribing information for CANCIDAS(R)): Caspofungin reduced the blood AUC0-12 of tacrolimus by approximately 20%, peak blood concentration (C max) by 16%, and 12-hour blood concentration (C12hr) by 26% in healthy adult subjects when tacrolimus (2 doses of 0.1 mg/kg 12 hours apart) was administered on the 10th day of CANCIDAS (R) 70 mg daily, as compared to results from control period in which tacrolimus was administered alone [see Drug Interactions 7.4)]. (0.01 mg/kg/day as continuous infusion) 954 AUC 0-t +- 334 +- 9.22 +- 0.015 14.7 7.79 2.1 [0.5-6.0] Median [range] 82.7 AUC 0-12 +- 63.2 1.5 [0.4-4.0] 142 +- 116 In 18 kidney transplant patients, tacrolimus trough concentrations from to 30 ng/mL measured at 10 to 12 hours post-dose (Cmin) correlated well with the AUC (correlation coefficient 0.93). In 24 liver transplant patients over concentration range of 10 to 60 ng/mL, the correlation coefficient was 0.94. In 25 heart transplant patients over concentration range of to 24 ng/mL, the correlation coefficient was 0.89 after an oral dose of 0.075 or 0.15 mg/kg/day at steady-state.

PREGNANCY SECTION.


8.1 Pregnancy. Pregnancy Category - There are no adequate and well-controlled studies in pregnant women. Tacrolimus is transferred across the placenta. The use of tacrolimus during pregnancy in humans has been associated with neonatal hyperkalemia and renal dysfunction. Tacrolimus given orally to pregnant rabbits at 0.5 to 4.3 times the clinical dose and pregnant rats at 0.8 to 6.9 times the clinical dose was associated with an increased incidence of fetal death in utero, fetal malformations (cardiovascular, skeletal, omphalocele, and gallbladder agenesis) and maternal toxicity. Tacrolimus should be used during pregnancy only if the potential benefit to the mother justifies the potential risk to the fetus. In pregnant rabbits, tacrolimus at oral doses of 0.32 and 1.0 mg/kg, 0.5 to 4.3 times the clinical dose range (0.075 to 0.2 mg/kg) based on body surface area, was associated with maternal toxicity as well as an increased incidence of abortions. At the mg/kg dose, fetal rabbits showed an increased incidence of malformations (ventricular hypoplasia, interventricular septal defect, bulbous aortic arch, stenosis of ductus arteriosus, interrupted ossification of vertebral arch, vertebral and rib malformations, omphalocele, and gallbladder agenesis) and developmental variations. In pregnant rats, tacrolimus at oral doses of 3.2 mg/kg, 2.6 to 6.9 times the clinical dose range was associated with maternal toxicity, an increase in late resorptions, decreased numbers of live births, and decreased pup weight and viability. Tacrolimus, given orally to pregnant rats after organogenesis and during lactation at 1.0 and 3.2 mg/kg, 0.8 to 6.9 times the recommended clinical dose range was associated with reduced pup weights and pup viability (3.2 mg/kg only); among the high dose pups that died early, an increased incidence of kidney hydronephrosis was observed.

SPL PATIENT PACKAGE INSERT SECTION.


PATIENT INFORMATION. Tacrolimus Capsules (ta-KROE-li-mus) Read this Patient Information before you start taking tacrolimus capsules and each time you get refill. There may be new information. This information does not take the place of talking with your doctor about your medical condition or your treatment. What is the most important information should know about tacrolimus capsules Tacrolimus capsules can cause serious side effects, including: 1.Increased risk of cancer. People who take tacrolimus capsules have an increased risk of getting some kinds of cancer, including skin and lymph gland cancer (lymphoma). 2.Increased risk of infection. Tacrolimus capsule is medicine that affects your immune system. Tacrolimus capsules can lower the ability of your immune system to fight infections. Serious infections can happen in people receiving tacrolimus capsules that can cause death. 3.Call your doctor right away if you have symptoms of an infection such as:ofeverosweats or chillsocough or flu-like symptomsomuscle achesowarm, red, or painful areas on your skinWhat are tacrolimus capsulesTacrolimus capsules are prescription medicine used with other medicines to help prevent organ rejection in people who have had kidney, liver, or heart transplant and tacrolimus capsules are not for use with medicines called cyclosporines (Gengraf (R), Neoral (R), and Sandimune(R)). Tacrolimus capsules are not for use with medicine called sirolimus (Rapamune (R)) in people who have had liver or heart transplants. It is not known if tacrolimus capsules are safe and effective when used with sirolimus in people who have had kidney transplants.It is not known if tacrolimus capsules are safe and effective in children who have had kidney or heart transplants.Who Should Not Take Tacrolimus CapsulesDo not take tacrolimus capsules if you are allergic to tacrolimus or any of the ingredients in tacrolimus capsules. See the end of this leaflet for complete list of ingredients in tacrolimus capsules. What should tell my doctor before taking tacrolimus capsulesBefore you take tacrolimus capsules, tell your doctor if you:oplan to receive any live vaccinesohave or have had liver, kidney or heart problemsoare pregnant or plan to become pregnant. Tacrolimus capsules may harm your unborn baby. Talk to your doctor if you are pregnant or plan to become pregnant.oAre breastfeeding or plan to breastfeed. Tacrolimus can pass into your breast milk. You and your doctor should decide if you will take tacrolimus capsules or breastfeed. You should not do both. Tell your doctor about all the medicines you take, including prescription and non-prescription medicines, vitamins, and herbal supplements. Especially tell your doctor if you take:ocyclosporine (Gengraf (R), Neoral (R), and Sandimune(R)) osirolimus (Rapamune (R)) onelfinavir (Viracept(R)) otelaprevir (IncivekTM) oboceprevir (VictrelisTM) oamiodarone (Cordarone TM, NexteroneTM, PaceroneTM) Ask your doctor or pharmacist if you are not sure if you take any of the medicines listed above. Tacrolimus capsules may affect the way other medicines work, and other medicines may affect how tacrolimus capsules works. Know the medicines you take. Keep list of your medicines and show it to your doctor and pharmacist when you get new medicine.How Should Take Tacrolimus CapsulesoTake tacrolimus capsules exactly as your doctor tells you to take it. oYour doctor will tell you how many tacrolimus capsules to take and when to take them.oYour doctor may change your tacrolimus capsules dose if needed. oDo not stop taking or change your dose of tacrolimus capsules without talking to your doctor. oTake tacrolimus capsules with or without food.oTake tacrolimus capsules the same way everyday. For example, if you choose to take tacrolimus capsules with food, you should always take tacrolimus capsules with food.oTake tacrolimus capsules at the same time each day, 12 hours apart. For example, if you take your first dose at 7:00 a.m. you should take your second dose at 7:00 p.m. oTaking tacrolimus capsules at the same time each day helps to keep enough medicine in your body to give your transplanted organ the around-the-clock medicine it needs.oDo not eat grapefruit or drink grapefruit juice while taking tacrolimus capsules. oIf you take too much tacrolimus capsules, call your doctor or go to the nearest hospital emergency room right away.What should avoid while taking tacrolimus capsulesoWhile you take tacrolimus capsules you should not receive any live vaccines such as: oflu vaccine through your noseomeaslesomumpsorubellaopolio by mouthoBCG (TB vaccine)oyellow feverochicken pox (varicella)otyphoid oAvoid exposure to sunlight and UV light such as tanning machines. Wear protective clothing and use sunscreen.What are the possible side effects of tacrolimus capsules Tacrolimus capsules may cause serious side effects, including:oSee What the most important information should know about tacrolimus capsulesohigh blood sugar (diabetes). Your doctor may do certain tests to check for diabetes while you take tacrolimus capsules. Call your doctor right away if you have: ofrequent urinationoincreased thirst or hungeroblurred visionoconfusionodrowsinessoloss of appetiteofruity smell on your breathonausea, vomiting, or stomach painokidney problems. Your doctor may do certain tests to check your kidney function while you take tacrolimus capsules. onervous system problems. Call your doctor right away if you get any of these symptoms while taking tacrolimus capsules. These could be signs of serious nervous system problem: oconfusionocomaomuscle tremorsonumbness and tinglingoheadacheoseizuresovision changesohigh levels of potassium in your blood. Your doctor may do certain tests to check your potassium level while you take tacrolimus capsules. ohigh blood pressure. Your doctor will monitor your blood pressure while you take tacrolimus capsules. oheart problems (myocardial hypertrophy). Tell your doctor right away if you get any of these symptoms of heart problems while taking tacrolimus capsules: oshortness of breathochest painofeel lightheadedofeel faintThe most common side effects of tacrolimus capsules in people receiving kidney transplant are:oinfection otremors (shaking of the body)ohigh blood pressureokidney problemsoconstipationodiarrheaoheadacheostomach pain otrouble sleepingonauseaolow levels of phosphate in your blood oswelling of the hands, ankles, or legs oweakness opainohigh levels of fat in your bloodohigh levels of potassium in your bloodolow red blood cell count (anemia)The most common side effects of tacrolimus capsules in people receiving liver transplants are:oshaking of the body tremors oheadacheodiarrheaohigh blood pressureonauseaokidney problemsostomach pain otrouble sleeping onumbness or tingling in your hands or feetoanemiaopain ofeveroweaknessohigh levels of potassium in the bloodolow levels of magnesium in the blood The most common side effects of tacrolimus capsules for heart transplant patients are:okidney problemsohigh blood pressureTell your doctor if you have any side effect that bothers you or that does not go away.These are not all the possible side effects of tacrolimus capsules. For more information, ask your doctor or pharmacist. Call your doctor for medical advice about side effects. You may report side effects to FDA at 1-800-FDA-1088. How should store tacrolimus capsulesoStore tacrolimus capsules at 59o to 86oF (15C to 30o C). oSafely throw away medicine that is out of date or no longer needed.Keep tacrolimus capsules and all medicines out of reach of children.General information about the safe and effective use of tacrolimus capsulesMedicines are sometimes prescribed for purposes other than those listed in Patient Information leaflet. Do not use tacrolimus capsules for condition for which it was not prescribed. Do not give tacrolimus capsules to other people, even if they have the same symptoms that you have. It may harm them.How Does Tacrolimus Capsules Protect My New OrganThe bodys immune system protects the body against anything that it does not recognize as part of the body. For example, when the immune system detects virus or bacteria it tries to get rid of it to prevent infection. When person has liver, kidney or heart transplant, the immune system does not recognize the new organ as part of the body and tries to get rid of it, too. This is called rejection. Tacrolimus capsules protect your new organ by slowing down the bodys immune system.This Patient Information leaflet summarizes the most important information about tacrolimus capsules. If you would like more information, talk with your doctor. You can ask your pharmacist or doctor for information about tacrolimus capsules that is written for health professionals. For more information, go to www.accordhealthcare.us or call Accord Healthcare at 1-866-941-7875. What are the ingredients in tacrolimus capsulesActive ingredient: tacrolimusInactive ingredients: lactose monohydrate, hypromellose E5, croscarmellose sodium, and magnesium stearate. The 0.5 mg capsule shell contains gelatin, titanium dioxide, iron oxide yellow and sodium lauryl sulfate, the mg capsule shell contains gelatin, titanium dioxide and sodium lauryl sulfate, and the mg capsule shell contains gelatin, titanium dioxide, iron oxide red and sodium lauryl sulfate. This Patient Information has been approved by the U.S. Food and Drug Administration.Manufactured For:Accord Healthcare, Inc.,1009 Slater Road,Suite 210-B,Durham, NC 27703, USA. Manufactured By:Intas Pharmaceuticals Limited,Plot No 457, 458,Village Matoda,Bavla Road, Ta.- Sanand,Dist.- Ahmedabad 382210.India.10 0108 679378Distributed By:MAJOR(R) PHARMACEUTICALS17177 Laurel Park Dr., Suite 233Livonia, MI 48152Refer to package label for Distributors NDC Number Issued November 2017. 1.Increased risk of cancer. People who take tacrolimus capsules have an increased risk of getting some kinds of cancer, including skin and lymph gland cancer (lymphoma). 2.Increased risk of infection. Tacrolimus capsule is medicine that affects your immune system. Tacrolimus capsules can lower the ability of your immune system to fight infections. Serious infections can happen in people receiving tacrolimus capsules that can cause death. 3.Call your doctor right away if you have symptoms of an infection such as:ofeverosweats or chillsocough or flu-like symptomsomuscle achesowarm, red, or painful areas on your skin. ofever. osweats or chills. ocough or flu-like symptoms. omuscle aches. owarm, red, or painful areas on your skin. Tacrolimus capsules are not for use with medicine called sirolimus (Rapamune (R)) in people who have had liver or heart transplants. oplan to receive any live vaccines. ohave or have had liver, kidney or heart problems. oare pregnant or plan to become pregnant. Tacrolimus capsules may harm your unborn baby. Talk to your doctor if you are pregnant or plan to become pregnant.. oAre breastfeeding or plan to breastfeed. Tacrolimus can pass into your breast milk. You and your doctor should decide if you will take tacrolimus capsules or breastfeed. You should not do both. ocyclosporine (Gengraf (R), Neoral (R), and Sandimune(R)) osirolimus (Rapamune (R)) onelfinavir (Viracept(R)) otelaprevir (IncivekTM) oboceprevir (VictrelisTM) oamiodarone (Cordarone TM, NexteroneTM, PaceroneTM) oTake tacrolimus capsules exactly as your doctor tells you to take it. oYour doctor will tell you how many tacrolimus capsules to take and when to take them.. oYour doctor may change your tacrolimus capsules dose if needed. oDo not stop taking or change your dose of tacrolimus capsules without talking to your doctor. oTake tacrolimus capsules with or without food.. oTake tacrolimus capsules the same way everyday. For example, if you choose to take tacrolimus capsules with food, you should always take tacrolimus capsules with food.. oTake tacrolimus capsules at the same time each day, 12 hours apart. For example, if you take your first dose at 7:00 a.m. you should take your second dose at 7:00 p.m. oTaking tacrolimus capsules at the same time each day helps to keep enough medicine in your body to give your transplanted organ the around-the-clock medicine it needs.. oTaking tacrolimus capsules at the same time each day helps to keep enough medicine in your body to give your transplanted organ the around-the-clock medicine it needs.. oDo not eat grapefruit or drink grapefruit juice while taking tacrolimus capsules. oIf you take too much tacrolimus capsules, call your doctor or go to the nearest hospital emergency room right away.. oWhile you take tacrolimus capsules you should not receive any live vaccines such as: oflu vaccine through your noseomeaslesomumpsorubellaopolio by mouthoBCG (TB vaccine)oyellow feverochicken pox (varicella)otyphoid oflu vaccine through your nose. omeasles. omumps. orubella. opolio by mouth. oBCG (TB vaccine). oyellow fever. ochicken pox (varicella). otyphoid. oAvoid exposure to sunlight and UV light such as tanning machines. Wear protective clothing and use sunscreen.. oSee What the most important information should know about tacrolimus capsules. ohigh blood sugar (diabetes). Your doctor may do certain tests to check for diabetes while you take tacrolimus capsules. Call your doctor right away if you have: ofrequent urinationoincreased thirst or hungeroblurred visionoconfusionodrowsinessoloss of appetiteofruity smell on your breathonausea, vomiting, or stomach pain. ofrequent urination. oincreased thirst or hunger. oblurred vision. oconfusion. odrowsiness. oloss of appetite. ofruity smell on your breath. onausea, vomiting, or stomach pain. okidney problems. Your doctor may do certain tests to check your kidney function while you take tacrolimus capsules. onervous system problems. Call your doctor right away if you get any of these symptoms while taking tacrolimus capsules. These could be signs of serious nervous system problem: oconfusionocomaomuscle tremorsonumbness and tinglingoheadacheoseizuresovision changes. oconfusion. ocoma. omuscle tremors. onumbness and tingling. oheadache. oseizures. ovision changes. ohigh levels of potassium in your blood. Your doctor may do certain tests to check your potassium level while you take tacrolimus capsules. ohigh blood pressure. Your doctor will monitor your blood pressure while you take tacrolimus capsules. oheart problems (myocardial hypertrophy). Tell your doctor right away if you get any of these symptoms of heart problems while taking tacrolimus capsules: oshortness of breathochest painofeel lightheadedofeel faint. oshortness of breath. ochest pain. ofeel lightheaded. ofeel faint. oinfection otremors (shaking of the body). ohigh blood pressure. okidney problems. oconstipation. odiarrhea. oheadache. ostomach pain otrouble sleeping. onausea. olow levels of phosphate in your blood oswelling of the hands, ankles, or legs oweakness opain. ohigh levels of fat in your blood. ohigh levels of potassium in your blood. olow red blood cell count (anemia). oshaking of the body tremors oheadache. odiarrhea. ohigh blood pressure. onausea. okidney problems. ostomach pain otrouble sleeping onumbness or tingling in your hands or feet. oanemia. opain ofever. oweakness. ohigh levels of potassium in the blood. olow levels of magnesium in the blood okidney problems. ohigh blood pressure. oStore tacrolimus capsules at 59o to 86oF (15C to 30o C). oSafely throw away medicine that is out of date or no longer needed.

SPL UNCLASSIFIED SECTION.


5.2 Lymphoma and Other Malignancies. Patients receiving immunosuppressants, including tacrolimus, are at increased risk of developing lymphomas and other malignancies, particularly of the skin [see Boxed Warning]. The risk appears to be related to the intensity and duration of immunosuppression rather than to the use of any specific agent. As usual for patients with increased risk for skin cancer, exposure to sunlight and UV light should be limited by wearing protective clothing and using sunscreen with high protection factor.Post transplant lymphoproliferative disorder (PTLD) has been reported in immunosuppressed organ transplant recipients. The majority of PTLD events appear related to Epstein Barr Virus (EBV) infection. The risk of PTLD appears greatest in those individuals who are EBV seronegative, population which includes many young children.

USE IN SPECIFIC POPULATIONS SECTION.


8 USE IN SPECIFIC POPULATIONS. oPregnancy: Based on animal data may cause fetal harm. Use only if the potential benefit justifies the risk 8.1) oNursing Mothers: Discontinue nursing taking into consideration importance of drug to mother (8.3) oHepatic/Renal impaired patients: Administer at the lower end of the recommended starting dose. Monitor function in patients with impaired renal function 2.3, 2.4, 8.6, 8.7) oPregnancy: Based on animal data may cause fetal harm. Use only if the potential benefit justifies the risk 8.1) oNursing Mothers: Discontinue nursing taking into consideration importance of drug to mother (8.3) oHepatic/Renal impaired patients: Administer at the lower end of the recommended starting dose. Monitor function in patients with impaired renal function 2.3, 2.4, 8.6, 8.7) 8.1 Pregnancy. Pregnancy Category - There are no adequate and well-controlled studies in pregnant women. Tacrolimus is transferred across the placenta. The use of tacrolimus during pregnancy in humans has been associated with neonatal hyperkalemia and renal dysfunction. Tacrolimus given orally to pregnant rabbits at 0.5 to 4.3 times the clinical dose and pregnant rats at 0.8 to 6.9 times the clinical dose was associated with an increased incidence of fetal death in utero, fetal malformations (cardiovascular, skeletal, omphalocele, and gallbladder agenesis) and maternal toxicity. Tacrolimus should be used during pregnancy only if the potential benefit to the mother justifies the potential risk to the fetus. In pregnant rabbits, tacrolimus at oral doses of 0.32 and 1.0 mg/kg, 0.5 to 4.3 times the clinical dose range (0.075 to 0.2 mg/kg) based on body surface area, was associated with maternal toxicity as well as an increased incidence of abortions. At the mg/kg dose, fetal rabbits showed an increased incidence of malformations (ventricular hypoplasia, interventricular septal defect, bulbous aortic arch, stenosis of ductus arteriosus, interrupted ossification of vertebral arch, vertebral and rib malformations, omphalocele, and gallbladder agenesis) and developmental variations. In pregnant rats, tacrolimus at oral doses of 3.2 mg/kg, 2.6 to 6.9 times the clinical dose range was associated with maternal toxicity, an increase in late resorptions, decreased numbers of live births, and decreased pup weight and viability. Tacrolimus, given orally to pregnant rats after organogenesis and during lactation at 1.0 and 3.2 mg/kg, 0.8 to 6.9 times the recommended clinical dose range was associated with reduced pup weights and pup viability (3.2 mg/kg only); among the high dose pups that died early, an increased incidence of kidney hydronephrosis was observed.. 8.3 Nursing Mothers. Tacrolimus is excreted in human milk. As the effect of chronic exposure to tacrolimus in healthy infants is not established, patients maintained on tacrolimus should discontinue nursing taking into consideration importance of drug to the mother.. 8.4 Pediatric Use. The safety and efficacy of tacrolimus in pediatric kidney and heart transplant patients have not been established. Successful liver transplants have been performed in pediatric patients (ages up to 16 years) using tacrolimus. Two randomized active-controlled trials of tacrolimus in primary liver transplantation included 56 pediatric patients. Thirty-one patients were randomized to tacrolimus-based and 25 to cyclosporine-based therapies. Additionally, minimum of 122 pediatric patients were studied in an uncontrolled trial of tacrolimus in living related donor liver transplantation. Pediatric patients generally required higher doses of tacrolimus to maintain blood trough concentrations of tacrolimus similar to adult patients [see Dosage and Administration 2.2)] . 8.5 Geriatric Use. Clinical trials of tacrolimus did not include sufficient numbers of subjects aged 65 and over to determine whether they respond differently from younger subjects. Other reported clinical experience has not identified differences in responses between the elderly and younger patients. In general, dose selection for an elderly patient should be cautious, usually starting at the low end of the dosing range, reflecting the greater frequency of decreased hepatic, renal, or cardiac function, and of concomitant disease or other drug therapy.. 8.6 Use in Renal Impairment. The pharmacokinetics of tacrolimus in patients with renal impairment was similar to that in healthy volunteers with normal renal function. However, consideration should be given to dosing tacrolimus at the lower end of the therapeutic dosing range in patients who have received liver or heart transplant and have pre-existing renal impairment. Further reductions in dose below the targeted range may be required [see Dosage and Administration (2.3) and Clinical Pharmacology 12.3)] . 8.7 Use in Hepatic Impairment. The mean clearance of tacrolimus was substantially lower in patients with severe hepatic impairment (mean Child-Pugh score: >10) compared to healthy volunteers with normal hepatic function. Close monitoring of tacrolimus trough concentrations is warranted in patients with hepatic impairment [see Clinical Pharmacology 12.3)]. The use of tacrolimus in liver transplant recipients experiencing post-transplant hepatic impairment may be associated with increased risk of developing renal insufficiency related to high whole-blood trough concentrations of tacrolimus. These patients should be monitored closely and dosage adjustments should be considered. Some evidence suggests that lower doses should be used in these patients [see Dosage and Administration (2.3) and Clinical Pharmacology (12.3)].

WARNINGS AND PRECAUTIONS SECTION.


5 WARNINGS AND PRECAUTIONS. oLymphoma and Other Malignancies: Risk of lymphomas, including post transplant lymphoproliferative disorder (PTLD); appears related to intensity and duration of use. Avoid prolonged exposure to UV light and sunlight (5.2) Serious infections: Increased risk of bacterial, viral, fungal and protozoal infections, including opportunistic infections: combination immunosuppression should be used with caution (5.3) oPolyoma Virus Infections: Serious, sometimes fatal outcomes, including polyoma virus-associated nephropathy (PVAN), mostly due to BK virus, and JC virus-associated progressive multifocal leukoencephalopathy (PML); consider reducing immunosuppression (5.4) oCytomegalovirus (CMV) Infections: Increased risk of CMV viremia and disease; consider reducing immunosuppression 5.5) oNew Onset Diabetes After Transplant: Monitor blood glucose (5.6) oNephrotoxicity: Acute and/or chronic; reduce the dose; use caution with other nephrotoxic drugs 5.7) oNeurotoxicity: Risk of Posterior Reversible Encephalopathy Syndrome, monitor for neurologic abnormalities; reduce or discontinue tacrolimus and other immunosuppressants (5.8) oHyperkalemia: Monitor serum potassium levels. Careful consideration should be given prior to use of other agents also associated with hyperkalemia 5.9) oHypertension: May require antihypertensive therapy. Monitor relevant drug-drug interactions 5.10) oAnaphylactic Reactions with IV formulation: Observe patients receiving tacrolimus injection for signs and symptoms of anaphylaxis (5.11) oUse with Sirolimus: Not recommended in liver and heart transplant due to increased risk of serious adverse reactions (5.12) oMyocardial Hypertrophy: Consider dosage reduction or discontinuation (5.15) oImmunizations: Use of live vaccines should be avoided (5.16) oPure Red Cell Aplasia: Discontinuation should be considered (5.17) oLymphoma and Other Malignancies: Risk of lymphomas, including post transplant lymphoproliferative disorder (PTLD); appears related to intensity and duration of use. Avoid prolonged exposure to UV light and sunlight (5.2) Serious infections: Increased risk of bacterial, viral, fungal and protozoal infections, including opportunistic infections: combination immunosuppression should be used with caution (5.3) oPolyoma Virus Infections: Serious, sometimes fatal outcomes, including polyoma virus-associated nephropathy (PVAN), mostly due to BK virus, and JC virus-associated progressive multifocal leukoencephalopathy (PML); consider reducing immunosuppression (5.4) oCytomegalovirus (CMV) Infections: Increased risk of CMV viremia and disease; consider reducing immunosuppression 5.5) oNew Onset Diabetes After Transplant: Monitor blood glucose (5.6) oNephrotoxicity: Acute and/or chronic; reduce the dose; use caution with other nephrotoxic drugs 5.7) oNeurotoxicity: Risk of Posterior Reversible Encephalopathy Syndrome, monitor for neurologic abnormalities; reduce or discontinue tacrolimus and other immunosuppressants (5.8) oHyperkalemia: Monitor serum potassium levels. Careful consideration should be given prior to use of other agents also associated with hyperkalemia 5.9) oHypertension: May require antihypertensive therapy. Monitor relevant drug-drug interactions 5.10) oAnaphylactic Reactions with IV formulation: Observe patients receiving tacrolimus injection for signs and symptoms of anaphylaxis (5.11) oUse with Sirolimus: Not recommended in liver and heart transplant due to increased risk of serious adverse reactions (5.12) oMyocardial Hypertrophy: Consider dosage reduction or discontinuation (5.15) oImmunizations: Use of live vaccines should be avoided (5.16) oPure Red Cell Aplasia: Discontinuation should be considered (5.17) 5.1 Management of Immunosuppression. Only physicians experienced in immunosuppressive therapy and management of organ transplant patients should use tacrolimus. Patients receiving the drug should be managed in facilities equipped and staffed with adequate laboratory and supportive medical resources. The physicians responsible for maintenance therapy should have complete information requisite for the follow up of the patient [see Boxed Warning] . 5.2 Lymphoma and Other Malignancies. Patients receiving immunosuppressants, including tacrolimus, are at increased risk of developing lymphomas and other malignancies, particularly of the skin [see Boxed Warning]. The risk appears to be related to the intensity and duration of immunosuppression rather than to the use of any specific agent. As usual for patients with increased risk for skin cancer, exposure to sunlight and UV light should be limited by wearing protective clothing and using sunscreen with high protection factor.Post transplant lymphoproliferative disorder (PTLD) has been reported in immunosuppressed organ transplant recipients. The majority of PTLD events appear related to Epstein Barr Virus (EBV) infection. The risk of PTLD appears greatest in those individuals who are EBV seronegative, population which includes many young children.. 5.3 Serious Infections. Patients receiving immunosuppressants, including tacrolimus, are at increased risk of developing bacterial, viral, fungal, and protozoal infections, including opportunistic infections [see Boxed Warning and Warnings and Precautions 5.4, 5.5)]. These infections may lead to serious, including fatal, outcomes. Because of the danger of oversuppression of the immune system which can increase susceptibility to infection, combination immunosuppressant therapy should be used with caution. 5.4 Polyoma Virus Infections. Patients receiving immunosuppressants, including tacrolimus, are at increased risk for opportunistic infections, including polyoma virus infections. Polyoma virus infections in transplant patients may have serious, and sometimes fatal, outcomes. These include polyoma virus-associated nephropathy (PVAN), mostly due to BK virus infection, and JC virus-associated progressive multifocal leukoencephalopathy (PML) which have been observed in patients receiving tacrolimus [see Adverse Reactions 6.2)] PVAN is associated with serious outcomes, including deteriorating renal function and kidney graft loss [see Adverse Reactions 6.2)] Patient monitoring may help detect patients at risk for PVAN. Cases of PML have been reported in patients treated with tacrolimus. PML, which is sometimes fatal, commonly presents with hemiparesis, apathy, confusion, cognitive deficiencies and ataxia. Risk factors for PML include treatment with immunosuppressant therapies and impairment of immune function. In immunosuppressed patients, physicians should consider PML in the differential diagnosis in patients reporting neurological symptoms and consultation with neurologist should be considered as clinically indicated. Reductions in immunosuppression should be considered for patients who develop evidence of PVAN or PML. Physicians should also consider the risk that reduced immunosuppression represents to the functioning allograft. 5.5 Cytomegalovirus (CMV) Infections. Patients receiving immunosuppressants, including tacrolimus, are at increased risk of developing CMV viremia and CMV disease. The risk of CMV disease is highest among transplant recipients seronegative for CMV at time of transplant who receive graft from CMV seropositive donor. Therapeutic approaches to limiting CMV disease exist and should be routinely provided. Patient monitoring may help detect patients at risk for CMV disease. Consideration should be given to reducing the amount of immunosuppression in patients who develop CMV viremia and/or CMV disease. 5.6 New Onset Diabetes After Transplant. Tacrolimus was shown to cause new onset diabetes mellitus in clinical trials of kidney, liver, and heart transplantation. New onset diabetes after transplantation may be reversible in some patients. Black and Hispanic kidney transplant patients are at an increased risk. Blood glucose concentrations should be monitored closely in patients using tacrolimus [see Adverse Reactions 6.1)] . 5.7 Nephrotoxicity. Tacrolimus, like other calcineurin-inhibitors, can cause acute or chronic nephrotoxicity, particularly when used in high doses. Acute nephrotoxicity is most often related to vasoconstriction of the afferent renal arteriole, is characterized by increasing serum creatinine, hyperkalemia, and/or decrease in urine output, and is typically reversible. Chronic calcineurin-inhibitor nephrotoxicity is associated with increased serum creatinine, decreased kidney graft life, and characteristic histologic changes observed on renal biopsy; the changes associated with chronic calcineurin-inhibitor nephrotoxicity are typically progressive. Patients with impaired renal function should be monitored closely as the dosage of tacrolimus may need to be reduced. In patients with persistent elevations of serum creatinine who are unresponsive to dosage adjustments, consideration should be given to changing to another immunosuppressive therapy. Based on reported adverse reactions terms related to decreased renal function, nephrotoxicity was reported in approximately 52% of kidney transplantation patients and in 40% and 36% of liver transplantation patients receiving tacrolimus in the U.S. and European randomized trials, respectively, and in 59% of heart transplantation patients in European randomized trial [see Adverse Reactions (6.1)]. Due to the potential for additive or synergistic impairment of renal function, care should be taken when administering tacrolimus with drugs that may be associated with renal dysfunction. These include, but are not limited to, aminoglycosides, ganciclovir, amphotericin B, cisplatin, nucleotide reverse transcriptase inhibitors (e.g., tenofovir) and protease inhibitors (e.g., ritonavir, indinavir). Similarly, care should be exercised when administering with CYP3A4 inhibitors such as antifungal drugs (e.g., ketoconazole), calcium channel blockers (e.g., diltiazem, verapamil), and macrolide antibiotics (e.g., clarithromycin, erythromycin, troleandomycin) which will result in increased tacrolimus whole blood concentrations due to inhibition of tacrolimus metabolism [see Drug Interactions 7.3, 7.4, 7.5, 7.6)] . 5.8 Neurotoxicity. Tacrolimus may cause spectrum of neurotoxicities, particularly when used in high doses. The most severe neurotoxicities include posterior reversible encephalopathy syndrome (PRES), delirium, and coma. Patients treated with tacrolimus have been reported to develop PRES. Symptoms indicating PRES include headache, altered mental status, seizures, visual disturbances and hypertension. Diagnosis may be confirmed by radiological procedure. If PRES is suspected or diagnosed, blood pressure control should be maintained and immediate reduction of immunosuppression is advised. This syndrome is characterized by reversal of symptoms upon reduction or discontinuation of immunosuppression. Coma and delirium, in the absence of PRES, have also been associated with high plasma concentrations of tacrolimus. Seizures have occurred in adult and pediatric patients receiving tacrolimus [see Adverse Reactions 6.1)] Less severe neurotoxicities, include tremors, paresthesias, headache, and other changes in motor function, mental status, and sensory function [see Adverse Reactions (6.1)] Tremor and headache have been associated with high whole-blood concentrations of tacrolimus and may respond to dosage adjustment. 5.9 Hyperkalemia. Hyperkalemia has been reported with tacrolimus use. Serum potassium levels should be monitored. Careful consideration should be given prior to use of other agents also associated with hyperkalemia (e.g., potassium-sparing diuretics, ACE inhibitors, angiotensin receptor blockers) during tacrolimus therapy [see Adverse Reactions( 6.1)]. 5.10 Hypertension. Hypertension is common adverse effect of tacrolimus therapy and may require antihypertensive therapy [see Adverse Reactions (6.1)]. The control of blood pressure can be accomplished with any of the common antihypertensive agents, though careful consideration should be given prior to use of antihypertensive agents associated with hyperkalemia (e.g., potassium-sparing diuretics, ACE inhibitors, angiotensin receptor blockers) [see Warnings and Precautions 5.9)] Calcium-channel blocking agents may increase tacrolimus blood concentrations and therefore require dosage reduction of tacrolimus [see Drug Interactions 7.5)]. 5.11 Anaphylactic Reactions with Tacrolimus Injection. Anaphylactic reactions have occurred with injectables containing castor oil derivatives, including tacrolimus, in small percentage of patients (0.6%). The exact cause of these reactions is not known. Tacrolimus injection should be reserved for patients who are unable to take tacrolimus capsules [see Indications and Usage (1.4)] Patients receiving tacrolimus injection should be under continuous observation for at least the first 30 minutes following the start of the infusion and at frequent intervals thereafter. If signs or symptoms of anaphylaxis occur, the infusion should be stopped. An aqueous solution of epinephrine should be available at the bedside as well as source of oxygen.. 5.12 Use with Sirolimus. The safety and efficacy of tacrolimus with sirolimus has not been established in kidney transplant patients. Use of sirolimus with tacrolimus in studies of de novo liver transplant patients was associated with an excess mortality, graft loss, and hepatic artery thrombosis (HAT) and is not recommended [see Indications and Usage (1.4)]. Use of sirolimus (2 mg per day) with tacrolimus in heart transplant patients in U.S. trial was associated with increased risk of renal function impairment, wound healing complications, and insulin-dependent post-transplant diabetes mellitus, and is not recommended [see Clinical Studies (14.3)]. Use of sirolimus with tacrolimus in studies of de novo liver transplant patients was associated with an excess mortality, graft loss, and hepatic artery thrombosis (HAT) and is not recommended [see Indications and Usage (1.4)]. Use of sirolimus (2 mg per day) with tacrolimus in heart transplant patients in U.S. trial was associated with increased risk of renal function impairment, wound healing complications, and insulin-dependent post-transplant diabetes mellitus, and is not recommended [see Clinical Studies (14.3)]. 5.13 Use with CYP3A4 Inhibitors and Inducers. When coadministering tacrolimus with strong CYP3A4-inhibitors (e.g., telaprevir, boceprevir, ritonavir, ketoconazole, itraconazole, voriconazole, clarithromycin) and strong inducers (e.g., rifampin, rifabutin) adjustments in the dosing regimen of tacrolimus and subsequent frequent monitoring of tacrolimus whole blood trough concentrations and tacrolimus associated adverse reactions are recommended [see Drug Interactions (7)] . 5.14 QT Prolongation. Tacrolimus may prolong the QT/QTc interval and may cause Torsade de Pointes. Avoid tacrolimus in patients with congenital long QT syndrome. In patients with congestive heart failure, bradyarrhythmias, those taking certain antiarrhythmic medications or other medicinal products that lead to QT prolongation, and those with electrolyte disturbances such as hypokalemia, hypocalcemia, or hypomagnesemia, consider obtaining electrocardiograms and monitoring electrolytes (magnesium, potassium, calcium) periodically during treatment.When coadministering tacrolimus with other substrates and/or inhibitors of CYP3A4 that also have the potential to prolong the QT interval, reduction in tacrolimus dose, frequent monitoring of tacrolimus whole blood concentrations, and monitoring for QT prolongation is recommended. Use of tacrolimus with amiodarone has been reported to result in increased tacrolimus whole blood concentrations with or without concurrent QT prolongation [see Drug Interactions (7)] . 5.15 Myocardial Hypertrophy. Myocardial hypertrophy has been reported in infants, children, and adults, particularly those with high tacrolimus trough concentrations, and is generally manifested by echocardiographically demonstrated concentric increases in left ventricular posterior wall and interventricular septum thickness. This condition appears reversible in most cases following dose reduction or discontinuance of therapy. In patients who develop renal failure or clinical manifestations of ventricular dysfunction while receiving tacrolimus therapy, echocardiographic evaluation should be considered. If myocardial hypertrophy is diagnosed, dosage reduction or discontinuation of tacrolimus should be considered [see Adverse Reactions (6.2)]. 5.16 Immunizations. The use of live vaccines should be avoided during treatment with tacrolimus; examples include (not limited to) the following: intranasal influenza, measles, mumps, rubella, oral polio, BCG, yellow fever, varicella, and TY21a typhoid vaccines.. 5.17 Pure Red Cell Aplasia. Cases of pure red cell aplasia (PRCA) have been reported in patients treated with tacrolimus. mechanism for tacrolimus-induced PRCA has not been elucidated. All patients reported risk factors for PRCA such as parvovirus B19 infection, underlying disease, or concomitant medications associated with PRCA. If PRCA is diagnosed, discontinuation of tacrolimus should be considered [see Adverse Reactions (6.2)]. 5.18 Gastrointestinal Perforation. Gastrointestinal perforation has been reported in patients treated with tacrolimus; all reported cases were considered to be complication of transplant surgery or accompanied by infection, diverticulum, or malignant neoplasm. As gastrointestinal perforation may be serious or life-threatening, appropriate medical/surgical management should be instituted promptly [see Adverse Reactions (6.1)].