文档视界 最新最全的文档下载
当前位置:文档视界 › 复方卡力孜然酊联合他克莫司软膏治疗白癜风42例疗效观察

复方卡力孜然酊联合他克莫司软膏治疗白癜风42例疗效观察

复方卡力孜然酊联合他克莫司软膏治疗白癜风42例疗效观察
复方卡力孜然酊联合他克莫司软膏治疗白癜风42例疗效观察

他克莫司软膏说明书

快易捷医药网 【通用名】他克莫司软膏 【商品名】普特彼 【英文名】英文名:Tacrolimus Ointment 【汉语拼音】Ta Ke Mo Si Ruan Gao 本品主要成分及其化学名称为:他克莫司, 【性状】本品为白色至淡黄色软膏。 【处方组成】每克本品含他克莫司0.03%或0.1%(w/w),软膏基质为矿 物油、石蜡、碳酸丙烯酯、白凡士林和白蜡。 【适应症】本品适用于因潜在危险而不宜使用传统疗法、或对传统 疗法反应不充分、或无法耐受传统疗法的中到重度特应 性皮炎患者,作为短期或间歇性长期治疗。 0.03%和0.1%浓度的本品均可用于成人,但只有0.03%浓 度的本品可用于2岁及以上的儿童。 【用法与用量】成人 0.03%和0.1%他克莫司软膏 在患处皮肤涂上一薄层本品,轻轻擦匀,并完全覆盖, 一天两次,持续至特应性皮炎症状和体征消失后一周。 封包疗法可能会促进全身性吸收,其安全性未进行过评 价。本品不应采用封包敷料外用。 儿童 0.03%他克莫司软膏 在患处皮肤涂上一薄层本品,轻轻擦匀,并完全覆盖, 一天两次,持续至特应性皮炎症状和体征消失后一周。 封包疗法可能会促进全身性吸收,其安全性未进行过评 价。本品不应采用封包敷料外用。 【禁忌症】对他克莫司或制剂中任何其他成分有过敏史的患者禁用 本品。 【注意事项】外用本品可能会引起局部症状,如皮肤烧灼感(灼热感 、刺痛、疼痛)或瘙痒。局部症状最常见于使用本品的 最初几天,通常会随着特应性皮炎受累皮肤好转而消失 。应用0.1%浓度的本品治疗时,90%的皮肤烧灼感持续时 间介于2分钟至3小时(中位时间为15分钟)之间,90%的 瘙痒症状持续时间介于3分钟至10小时(中位时间为20分 钟)之间。 不推荐使用本品治疗Netherton综合征患者,因为可能会 增加他克莫司的全身性吸收。本品对弥漫性红皮病患者 治疗的安全性尚未建立。 【患者须知】使用本品的患者应接受下列信息和指导: 1.患者应在医生的指导下使用本品。本品仅供外用。 和任何外用药一样,患者或护工在用药后应洗手,如果 手部不是治疗区的话。

中国白癜风诊疗共识(2014版)

白癜风诊疗共识(2014版) 中国中西医结合学会皮肤性病专业委员会色素病学组 本指南以中国中西医结合学会皮肤性病专业委员会色素病学组制订的白癜风治疗共识(2009版)为基础,经色素病学组、中华医学会皮肤科分会白癜风研究中心部分专家及国内相关专家讨论制定。参加起草及讨 论的成员(按姓氏汉语拼音排序):傅雯雯、高天文、何黎、贾虹、李恒进、李铁男、李珊山、卢忠、鲁严、 李春英、李强、刘清、马东来、乔树芳、秦万章、宋智琦、孙越、宋秀祖、涂彩霞、温海、王玮蓁、许爱娥、项蕾红、张学军、张建中、郑志忠、赵广、朱光斗。 白癜风治疗目的是控制皮损发展,促进白斑复色。 一、选择治疗方法时主要考虑因素: 1.病期:分进展期和稳定期。进展期判定参考白癜风疾病活动度评分(VIDA)积分、同形反应、Wood灯。①VIDA积分:近6周内出现新皮损或原皮损扩大(+4分),近3个月出现新皮损或原皮损扩大(+3分),近6个月出现新皮损或原皮损扩大(+2分);近1年出现新皮损或原皮损扩大(+1分);至少稳定1年(0分);至少稳定1年且有自发色素再生(-1分)。总分>1分即为进展期,≥4分为快速进展期;②同形反应:皮肤损伤1年内局部出现白斑。损伤包括物理性(创伤、切割伤、抓伤)、机械性摩擦、化学性/热灼伤、过敏性(接触性皮炎)或刺激性反应(接种疫苗、纹身等)、慢性压力、炎症性皮肤病、治疗性(放射治疗、光疗)。白斑发生于持续的压力或摩擦部位,或者是衣物,饰品的慢性摩擦部位,形状特殊,明显由损伤诱发;③Wood灯:皮损颜色呈灰白色,边界欠清,wood灯下皮损面积大于目测面积,提示是进展期。皮损颜色是白色,边界清,wood灯下皮损面积≤目测面积,提示是稳定期。以上3条符合任何一条即可考虑病情进展;④可同时参考激光共聚焦扫描显微镜(简称皮肤CT)和皮肤镜的图像改变,辅以诊断。 2.白斑面积(手掌面积约为体表面积1%):1级为轻度,<l%;2级为中度,l%~5%; 3级为中重度,6%~50%;4级为重度,>50%。白斑面积也可按白癜风面积评分指数(vitiligo area scoring index,VASI)来判定。VASI=∑(身体各部占手掌单元数)×该区域色素脱失所占百分比,VASI值为0~100。 3.型别:根据2012年白癜风全球问题共识大会(VGICC)及专家讨论,分为节段型、非节段型、混合型及未定类型白癜风。①节段型白癜风:沿某一皮神经节段分布(完全或部分匹配皮肤节段),单侧的不对称的白癜风。少数可双侧多节段分布;②非节段型白癜风:包括散发型、泛发型、面肢端型和黏膜型。散发型指白斑≥2片,面积为1~3级;泛发型为白斑面积4级(>50%);面肢端型指白斑主要局限于头面、手足,尤其好发于指趾远端及面部腔口周围,可发展为散发型、泛发型;黏膜型指白斑分布于2个及以上黏膜部位,可发展为散发型、泛发型;③混合型白癜风:节段型和非节段型并存;④未定类型白癜风:指非节段型分布的单片皮损,面积为1级。 4.疗效:面部复色疗效好,口唇、手足部位复色疗效差。病程越短,疗效越好。儿童疗效优于成人。 二、治疗原则 (一)进展期白癜风: 1.未定类型(原称局限型):可外用糖皮质激素(简称激素)或钙调神经磷酸酶抑制剂(他克莫司软膏、吡美莫司乳膏)等,也可外用低浓度的光敏药,如浓度<0.1%的8-甲氧沙林(8-MOP);维生素D3衍生物;局部光疗可选窄谱中波紫外线(NB-UVB)、308nm准分子激光及准分子光。

他克莫司软膏英文说明书

xxxxxx Tacrolimus Capsules 0-1200-720-inf Due to intersubject variability in tacrolimus pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy. (See DOSAGE AND ADMINISTRATION ). Pharmacokinetic data indicate that whole blood concentrations rather than plasma concentrations serve as the more appropriate sampling compartment to describe tacrolimus pharmacokinetics. Absorption Absorption 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) and 18 ± 5% in healthy volunteers (N=16). A single dose study conducted in 32 healthy volunteers established the bioequivalence of the 1 mg and 5 mg capsules. Another single dose study in 32 healthy volunteers established the bioequivalence of the 0.5 mg and 1 mg capsules. Tacrolimus maximum blood concentrations (C max ) and area under the curve (AUC) appeared to increase in a dose-proportional fashion in 18 fasted healthy volunteers receiving a single oral dose of 3, 7, and 10 mg. In 18 kidney transplant patients, tacrolimus trough concentrations from 3 to 30 ng/mL measured at 10 to 12 hours post-dose (C min ) correlated well with the AUC (correlation coefficient 0.93). In 24 liver transplant patients over a concentration range of 10 to 60 ng/mL, the correlation coefficient was 0.94. 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 a high-fat meal (848 kcal, 46% fat): mean AUC and C max were decreased 37% and 77%, respectively; T max was lengthened 5-fold. A high-carbohydrate meal (668 kcal, 85% carbohydrate) decreased mean AUC and mean C 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 C max was reduced 71%, and mean AUC was reduced 39%, relative to the fasted condition. When administered 1.5 hours following the meal, mean C max was reduced 63%, and mean AUC was reduced 39%, relative to the fasted condition. In 11 liver transplant patients, tacrolimus capsules administered 15 minutes after a high fat (400 kcal, 34% fat) breakfast, resulted in decreased AUC (27 ± 18%) and C max (50 ± 19%), as compared to a fasted state. Distribution The plasma protein binding of tacrolimus is approximately 99% and is independent of concentration over a range of 5-50 ng/mL. Tacrolimus is bound mainly to albumin and alpha-1-acid glycoprotein, and has a 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 a U.S. study, the ratio of whole blood concentration to plasma concentration averaged 35 (range 12 to 67). Metabolism Tacrolimus is extensively metabolized by the mixed-function oxidase system, primarily the cytochrome P-450 system (CYP3A). A metabolic pathway leading to the formation of 8 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, a 31-demethyl metabolite has been reported to have the same activity as tacrolimus. Excretion The mean clearance following IV administration of tacrolimus is 0.040, 0.083, and 0.053, in healthy volunteers, adult kidney transplant patients, adult liver transplant patients, respectively. In man, less than 1% of the dose administered is excreted unchanged in urine. In a mass balance study of IV administered radiolabeled tacrolimus to 6 healthy volunteers, the mean recovery of radiolabel was 77.8±12.7%. Fecal elimination accounted for 92.4±1% 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. Special Populations Pediatric Pharmacokinetics of tacrolimus have been studied in liver transplantation patients, 0.7 to 13.2 years of age. Following oral administration to 9 patients, mean AUC and C max were 337 ± 167 ng±hr/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 ). Renal and Hepatic Insufficiency The mean pharmacokinetic parameters for tacrolimus following single administrations to patients with renal and hepatic impairment are given in the following table. Population (No. of Patients) Dose AUC 0 - t (ng·hr/mL) t ? (hr) V (L/kg) CI (L/hr/kg) Renal Impairment (n=12) 0.02 mg/kg/4hr IV 393 ± 123 (t = 60 hr) 26.3 ± 9.2 1.07 ±0.20 0.038 ± 0.014 Mild Hepatic Impairment (n=6) 0.02 mg/kg/4hr IV 367 ± 107 (t = 72 hr) 60.6 ± 43.8 Range: 27.8-141 3.1±1.6 0.042 ± 0.02 7.7 mg PO 488 ± 320 (t = 72 hr) 66.1 ± 44.8 Range: 29.5-138 3.7 ± 4.7* 0.034 ± 0.019* Severe Hepatic Impairment (n=6, IV) 0.02 mg/kg/4hr IV (n=2) 762 ± 204 (t = 120 hr) 198 ± 158 Range: 81-436 3.9 ± 1 0.017 ± 0.013 (n=5, PO)? 0.01 mg/kg/8hr IV (n=4) 8 mg PO (n=1) 289 ± 117 (t = 144 hr) 658 (t =120 hr) 119 ± 35 Range: 85-178 3.1 ± 3.4* 0.016 ± 0.011* 5 mg PO (n=4) 4 mg PO (n=1) 533 ± 156 (t = 144 hr) *corrected for bioavailability; ?1 patient did not receive the PO dose Renal Insufficiency Tacrolimus pharmacokinetics following a single IV administration were determined in 12 patients (7 not on dialysis and 5 on dialysis, serum creatinine of 3.9 ± 1.6 and 12 ± 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 (see previous table). Hepatic Insufficiency 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 6 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. Race A formal study to evaluate the pharmacokinetic disposition of tacrolimus in Black transplant patients has not been conducted. However, a retrospective comparison of Black and Caucasian kidney transplant patients indicated that Black patients required higher tacrolimus doses to attain similar trough concentrations. (See DOSAGE AND ADMINISTRATION ). Gender A formal study 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. A retrospective comparison of pharmacokinetics in healthy volunteers, and in kidney and liver transplant patients indicated no gender-based differences. CLINICAL STUDIES Liver Transplantation The safety and efficacy of tacrolimus-based immunosuppression following orthotopic liver transplantation were assessed in two prospective, randomized, non-blinded multicenter studies. The active control groups were treated with a cyclosporine-based immunosuppressive regimen. Both studies used concomitant adrenal corticosteroids as part of the immunosuppressive regimens. These studies were designed to evaluate whether the two regimens were therapeutically equivalent, with patient and graft survival at 12 months following transplantation as the primary endpoints. The tacrolimus-based immunosuppressive regimen was found to be equivalent to the cyclosporine-based immunosuppressive regimens. 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 a cyclosporine-based immunosuppressive regimen (CBIR). In 10 of the 12 sites, the same CBIR protocol was used, while 2 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 8 clinical sites in Europe; prior to surgery, 270 were randomized to the tacrolimus-based immunosuppressive regimen and 275 to CBIR. In this study, each center used its local standard CBIR 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 equivalent to those in the CBIR treatment groups in both studies. The overall 1-year patient survival (CBIR and tacrolimus-based treatment groups combined) was 88% in the U.S. study and 78% in the European study. The overall 1-year graft survival (CBIR and tacrolimus-based treatment groups combined) was 81% in the U.S. study and 73% in the European study. In both studies, the median time to convert from IV to oral tacrolimus capsules dosing was 2 days. Because of the nature of the study design, comparisons of differences in secondary endpoints, such as incidence of acute rejection, refractory rejection or use of OKT3 for steroid-resistant rejection, could not be reliably made. Kidney Transplantation Tacrolimus/azathioprine Tacrolimus-based immunosuppression in conjunction with azathioprine and corticosteroids following kidney transplantation was assessed in a Phase 3 randomized, multicenter, non-blinded, prospective study. 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 a serum creatinine ≤ 4 mg/dL (median of 4 days after transplantation, range 1 to 14 days). Patients less than 6 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 1 year patient and graft survival was 96.1% and 89.6%, respectively and was equivalent between treatment arms. Because of the nature of the study design, comparisons of differences in secondary endpoints, such as incidence of acute rejection, refractory rejection or use of OKT3 for steroid-resistant rejection, could not be reliably made. Tacrolimus/mycophenolate mofetil(MMF) Tacrolimus-based immunosuppression in conjunction with MMF, corticosteroids, and induction has been studied. In a 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 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 study was conducted outside the United States; the study population was 93% Caucasian. In this study, mortality at 12 months in patients receiving tacrolimus/MMF was similar (2.7%) compared to patients receiving cyclosporine/MMF (3.3% and 1.8%) or sirolimus/MMF (3 %). Patients in the tacrolimus group exhibited higher estimated creatinine clearance rates (eCL cr ) using the Cockcroft-Gault formula (Table 1) and experienced fewer efficacy failures, defined as biopsy proven acute rejection (BPAR), graft loss, death, and/or lost to follow-up (Table 2) 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 ). Table 1: Estimated Creatinine Clearance at 12 Months in Study 1 Group eCLcr [mL/min] at Month 12a N MEAN SD MEDIAN Treatment Difference with Group C (99.2%CI b ) (A) CsA/MMF/CS 390 56.5 25.8 56.9 -8.6 (-13.7,-3.7) (B) CsA/MMF/CS/Daclizumab 399 58.9 25.6 60.9 -6.2 (-11.2,-1.2) (C) Tac/MMF/CS/Daclizumab 401 65.1 27.4 66.2 -(D) Siro/MMF/CS/Daclizumab 399 56.2 27.4 57.3 -8.9 (-14.1, -3.9) Total 1589 59.2 26.8 60.5 Key: CsA=Cyclosporine, CS=Corticosteroids, Tac=Tacrolimus, Siro=Sirolimus a) All death/graft loss (n=41,27, 23 and 42 in Groups A, B, C and D) and patients whose last recorded creatinine values were prior to month 3 visit (n=10, 9, 7 and 9 in Groups A, B, C and D) were inputed with GFR of 10 mL/min; a subject’s last observed creatinine value from month 3 on was used for the remainder of subjects with missing creatinine at month 12 (n=11, 12, 15 and 19 for Groups A, B, C and D). Weight was also imputed in the calculation of estimated GFR, if missing b) Adjusted for multiple (6) pairwise comparisons using Bonferroni corrections. Table 2: Incidence of BPAR, Graft Loss, Death or Loss to Follow-up at 12 Months in Study 1 A B C D N=390 N=399 N=401 N=399 Overall Failure 141 (36.2%) 126(31.6%) 82 (20.4%) 185 (46.4%) Components of efficacy failure BPAR 113(29%) 106 (26.6%) 60 (15%) 152(38.1%) Graft loss excluding death 28 (7.2%) 20 (5%) 12 (3%) 30 (7.5%) Mortality 13 (3.3%) 7(1.8%) 11(2.7%) 12 (3%) Lost to follow-up 5(1.3%) 7(1.8%) 5 (1.3%) 6(1.5%) Treatment Difference of efficacy failure compared to 15.8% (7.1%, 11.2% (2.7%, -26% (17.2%, Group C (99.2% CI a ) 24.3%) 19.5%) 34.7%) Group A =CsA/MMF/CS, B =CsA/MMF/CS/Daclizumab, C=Tac/MMF/CS/Daclizumab, and D=Siro/MMF/CS/Daclizumab a) Adjusted for multiple (6) pairwise comparisons using Bonferroni corrections. The protocol-specified target tacrolimus trough concentrations (C ,Tac) were 3-7 ng/mL; however, the trough observed median C ,Tac approximated 7 ng/mL throughout the 12 month study (Table 3).troughs Table 3: Tacrolimus Whole Blood Trough Concentrations (Study 1) Time Median (P10-P90 a ) tacrolimus whole blood trough concentrations (ng/mL) Day 30 (N=366) 6.9 (4.4-11.3) Day90 (N=351) 6.8 (4.1-10.7) Day 180 (N=355) 6.5 (4 - 9.6) Day 365 (N=346) 6.5 (3.8 -10) a) Range of C trough , Tac that excludes lowest 10% and highest 10% of C trough , Tac The protocol-specified target cyclosporine trough concentrations (C trough ,CsA) for Group B were 50-100 ng/mL; however, the observed median C trougs , CsA approximated 100 ng/mL throughout the 12 month study. The protocol-specified target C trougs ,CsA for Group A were 150-300 ng/mL for the first 3 months and 100-200 ng/mL from month 4 to month 12; the observed median C trougs , CsA approximated 225 ng/mL for the first 3 months and 140 ng/mL from month 4 to month 12. While patients in all groups started MMF at 1g BID, the MMF dose was reduced to <2 g/day in 63% of patients in the tacrolimus treatment arm by month 12 (Table 4); approximately 50% of these MMF dose reductions were due to adverse events. By comparison, the MMF dose was reduced to <2 g/day in 49% and 45% of patients in the two cyclosporine arms (Group A and Group B, respectively), by month 12 and approximately 40% of MMF dose reductions were due to adverse events. Table 4: MMF Dose Over Time in tacrolimus/MMF (Group C) (Study 1) Time period (Days) Time-averaged MMF dose (g/day)a <2 2 >2 0-30 (N=364) 37% 60% 2% 0-90 (N=373) 47% 51% 2% 0-180 (N=377) 56% 42% 2% 0-365 (N=380) 63% 36% 1% Time-averaged MMF dose = (total MMF dose)/(duration of treatment) a) Percentage of patients for each time-averaged MMF dose range during various treatment periods. Two g/day of time-averaged MMF dose means that MMF dose was not reduced in those patients during the treatment periods. In a 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 1 gram BID, basiliximab induction, and corticosteroids. In this study, the rate for the combined endpoint of biopsy proven acute rejection, 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 (4.2%) compared to those receiving cyclosporine/MMF (2.4%), including cases attributed to overimmunosuppression (Table 5). Table 5: Incidence of BPAR, Graft Loss, Death or Loss to Follow-up at 12 Months in Study 2 Tacrolimus/MMF (n=212) Cyclosporine/MMF (n=212) Overall Failure 32(15.1%) 36 (17%) Components of efficacy failure BPAR 16 (7.5%) 29 (13.7%) Graft loss excluding death 6 (2.8%) 4(1.9%) Mortality 9 (4.2%) 5 (2.4%) Lost to follow-up 4(1.9%) 1 (0.5%) Treatment Difference of efficacy failure compared to tacrolimus/MMF group (95% CI a ) - 1.9% (-5.2%, 9%) a) 95% confidence interval calculated using Fisher’s Exact Test The protocol-specified target tacrolimus whole blood trough concentrations (C trough ,Tac) in Study 2 were 7-16 ng/mL for the first three months and 5-15 ng/mL thereafter. The observed median C troughs ,Tac approximated 10 ng/mL during the first three months and 8 ng/mL from month 4 to month 12 (Table 6). Table 6: Tacrolimus Whole Blood Trough Concentrations (Study 2) Time Median (P10-P90a ) tacrolimus whole blood trough concentrations ng/mL Day 30 (N=174) 10.5 (6.3 -16.8) Day 60 (N=179) 9.2(5.9-15.3) Day 120 (N=176) 8.3 (4.6 -13.3) Day 180(N=171) 7.8 (5.5 -13.2) Day 365 (N=178) 7.1(4.2-12.4) a) Range of C troughs , Tac that excludes lowest 10% and highest 10% of C troughs Tac The protocol-specified target cyclosporine whole blood concentrations (Ctr o ugh,CsA) were 125 to 400 ng/ mL for the first three months, and 100 to 300 ng/mL thereafter. The observed median Ctroughs, CsA approximated 280 ng/mL during the first three months and 190 ng/mL from month 4 to month 12. Patients in both groups started MMF at 1g BID. The MMF dose was reduced to <2 g/day by month 12 in 62% of patients in the tacrolimus/MMF group (Table 7) and in 47% of patients in the cyclosporine/MMF group. Approximately 63% and 55% of these MMF dose reductions were because of adverse events in the tacrolimus/MMF group and the cyclosporine/MMF group, respectively. Table 7: MMF Dose Over Time in the tacrolimus/MMF group (Study 2) Time period (Days) Time-averaged MMF dose (g/day)a <2 2 >2 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% Time-averaged MMF dose=(total MMF dose)/(duration of treatment) a) Percentage of patients for each time-averaged MMF dose range during various treatment periods. Two g/day of time-averaged MMF dose means that MMF dose was not reduced in those patients during the treatment periods. INDICATIONS AND USAGE Tacrolimus capsules are indicated for the prophylaxis of organ rejection in patients receiving allogeneic liver, or kidney transplants. It is recommended that tacrolimus be used concomitantly with adrenal corticosteroids. In kidney transplant recipients, it is recommended that tacrolimus be used in conjunction with azathioprine or mycophenolate mofetil (MMF). The safety and efficacy of the use of tacrolimus with sirolimus has not been established (see CLINICAL STUDIES ). CONTRAINDICATIONS Tacrolimus capsules are contraindicated in patients with a hypersensitivity to tacrolimus. WARNINGS (See boxed WARNING .) Post-Transplant Diabetes Mellitus Insulin-dependent post-transplant diabetes mellitus (PTDM) was reported in 20% of tacrolimus-treated kidney transplant patients without pretransplant history of diabetes mellitus in the Phase III study (See Tables Below). The median time to onset of PTDM was 68 days. Insulin dependence was reversible in 15% of these PTDM patients at one year and in 50% at 2 years post transplant. Black and Hispanic kidney transplant patients were at an increased risk of development of PTDM. Incidence of Post Transplant Diabetes Mellitus and Insulin Use at 2 Years in Kidney Transplant Recipients in the Phase III study Status of PTDM* Tacrolimus CBIR Patients without pretransplant history of diabetes mellitus. 151 151 New onset PTDM*, 1st Year 30/151 (20%) 6/151 (4%) Still insulin dependent at one year in those without prior history of diabetes. 25/151 (17%) 5/151 (3%) New onset PTDM* post 1 year 1 Patients with PTDM* at 2 years 16/151 (11%) 5/151 (3%) * use of insulin for 30 or more consecutive days, with <5 day gap, without a prior history of insulin dependent diabetes mellitus or non insulin dependent diabetes mellitus. Development of Post Transplant Diabetes Mellitus by Race and by Treatment Group during First Year Post Kidney Transplantation in the Phase III study Tacrolimus CBIR Patient Race No. of Patients at Risk Patients Who Developed PTDM* No. of Patients At Risk Patients Who Developed PTDM* Black 41 15 (37%) 36 3 (8%) Hispanic 17 5 (29%) 18 1 (6%) Caucasian 82 10 (12%) 87 1 (1%) Other 11 0 (0%) 10 1 (10%) Total 151 30 (20%) 151 6 (4%) *use of insulin for 30 or more consecutive days, with <5 day gap, without a prior history of insulin dependent diabetes mellitus or non insulin dependent diabetes mellitus. Insulin-dependent post-transplant diabetes mellitus was reported in 18% and 11% of tacrolimus-treated liver transplant patients and was reversible in 45% and 31% of these patients at 1 year post transplant, in the U.S. and European randomized studies, respectively (See Table below). Hyperglycemia was associated with the use of tacrolimus in 47% and 33% of liver transplant recipients in the U.S. and European randomized studies, respectively, and may require treatment (see ADVERSE REACTIONS ) Incidence of Post Transplant Diabetes Mellitus and Insulin Use at 1 Year in Liver Transplant Recipients Status of PTDM* U.S. Study European Study Tacrolimus CBIR Tacrolimus CBIR Patients at risk** 239 236 239 249 New Onset PTDM* 42(18%) 30(13%) 26(11%) 12(5%) Patients still on insulin at 1 year 23(10%) 19(8%) 18(8%) 6 (2%) * use of insulin for 30 or more consecutive days, with < 5 day gap, without a prior history of insulin dependent diabetes mellitus or non insulin dependent diabetes mellitus ** Patients without pretransplant history of diabetes mellitus Nephrotoxicity Tacrolimus can cause nephrotoxicity, particularly when used in high doses. 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 (see ADVERSE REACTIONS ). More overt nephrotoxicity is seen early after transplantation, characterized by increasing serum creatinine and a decrease in urine output. 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. Care should be taken in using tacrolimus with other nephrotoxic drugs. In particular, to avoid excess nephrotoxicity, tacrolimus should not be used simultaneously with cyclosporine. tacrolimus or cyclosporine should be discontinued at least 24 hours prior to initiating the other. In the presence of elevated tacrolimus or cyclosporine concentrations, dosing with the other drug usually should be further delayed. Hyperkalemia Mild to severe hyperkalemia was reported in 31% of kidney transplant recipients and in 45% and 13% of liver transplant recipients treated with tacrolimus capsules in the U.S. and European randomized trials, respectively, and may require treatment (see ADVERSE REACTIONS ). Serum potassium levels should be monitored and potassium-sparing diuretics should not be used during tacrolimus capsules therapy (see PRECAUTIONS ). Neurotoxicity Tacrolimus capsules can cause neurotoxicity, particularly when used in high doses. Neurotoxicity, including tremor, headache, and other changes in motor function, mental status, and sensory function were reported in approximately 55% of liver transplant recipients in the two randomized studies. Tremor occurred more often in tacrolimus capsules-treated kidney transplant patients (54%) compared to cyclosporine-treated patients. The incidence of other neurological events in kidney transplant patients was similar in the two treatment groups (see ADVERSE REACTI ONS ). Tremor and headache have been associated with high whole-blood concentrations of tacrolimus and may respond to dosage adjustment. Seizures have occurred in adult and pediatric patients receiving tacrolimus capsules (see ADVERSE REACTIONS ). Coma and delirium also have been associated with high plasma concentrations of tacrolimus. Patients treated with tacrolimus have been reported to develop posterior reversible encephalopathy syndrome (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. Malignacy and Lymphoproliferative Disorders As in patients receiving other immunosuppressants, patients receiving tacrolimus capsules are at increased risk of developing lymphomas and other malignancies, particularly of the skin. The risk appears to be related to the intensity and duration of immunosuppression rather than to the use of any specific agent. A lymphoproliferative disorder (LPD) related to Epstein-Barr Virus (EBV) infection has been reported in immunosuppressed organ transplant recipients. The risk of LPD appears greatest in young children who are at risk for primary EBV infection while immunosuppressed or who are switched to tacrolimus capsules following long-term immunosuppression therapy. Because of the danger of oversuppression of the immune system which can increase susceptibility to infection, combination immunosuppressant therapy should be used with caution. Latent Viral Infections Immunosuppressed patients are at increased risk for opportunistic infections, including latent viral infections. These include BK virus associated nephropathy and JC virus associated progressive multifocal leukoencephalopathy (PML) which have been observed in patients receiving tacrolimus. These infections may lead to serious, including fatal, outcomes. PRECAUTIONS General Hypertension is a common adverse effect of tacrolimus therapy (see ADVERSE REACTIONS ). Mild or moderate hypertension is more frequently reported than severe hypertension. Antihypertensive therapy may be required; the control of blood pressure can be accomplished with any of the common antihypertensive agents. Since tacrolimus may cause hyperkalemia, potassium-sparing diuretics should be avoided. While calcium-channel blocking agents can be effective in treating tacrolimus-associated hypertension, care should be taken since interference with tacrolimus metabolism may require a dosage reduction (see Drug Interactions ). Renally and Hepatically Impaired Patients For patients with renal insufficiency some evidence suggests that lower doses should be used (see CLINICAL PHARMACOLOGY and DOSAGE AND ADMINISTRATION ). 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 levels 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 ). Myocardial Hypertrophy Myocardial hypertrophy has been reported in association with the administration of tacrolimus capsules, and is generally manifested by echocardiographically demonstrated concentric increases in left ventricular posterior wall and interventricular septum thickness. Hypertrophy has been observed in infants, children and adults. This condition appears reversible in most cases following dose reduction or discontinuance of therapy. In a group of 20 patients with pre- and post-treatment echocardiograms who showed evidence of myocardial hypertrophy, mean tacrolimus whole blood concentrations during the period prior to diagnosis of myocardial hypertrophy ranged from 11 to 53 ng/mL in infants (N=10, age 0.4 to 2 years), 4 to 46 ng/ mL in children (N=7, age 2 to 15 years) and 11 to 24 ng/mL in adults (N=3, age 37 to 53 years). 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. Information for Patients Patients should be informed of the need for repeated appropriate laboratory tests while they are receiving tacrolimus capsules. They should be given complete dosage instructions, advised of the potential risks during pregnancy, and informed of the increased risk of neoplasia. Patients should be informed that changes in dosage should not be undertaken without first consulting their physician. Patients should be informed that tacrolimus capsules can cause diabetes mellitus and should be advised of the need to see their physician if they develop frequent urination, increased thirst or hunger. As with other immunosuppressive agents, owing to the potential risk of malignant skin changes, exposure to sunlight and ultraviolet (UV) light should be limited by wearing protective clothing and using a sunscreen with a high protection factor. Laboratory Tests Serum creatinine, potassium, and fasting glucose should be assessed regularly. Routine monitoring of metabolic and hematologic systems should be performed as clinically warranted. Drug Interactions Due to the potential for additive or synergistic impairment of renal function, care should be taken when administering tacrolimus capsules with drugs that may be associated with renal dysfunction. These include, but are not limited to, aminoglycosides, amphotericin B, and cisplatin. Initial clinical experience with the co-administration of tacrolimus and cyclosporine resulted in additive/synergistic nephrotoxicity. Patients switched from cyclosporine to tacrolimus should receive the first tacrolimus capsules dose no sooner than 24 hours after the last cyclosporine dose. Dosing may be further delayed in the presence of elevated cyclosporine levels. Drugs that May Alter Tacrolimus Concentrations Since tacrolimus is metabolized mainly by the CYP3A enzyme systems, substances known to inhibit these enzymes may decrease the metabolism or increase bioavailability of tacrolimus as indicated by increased whole blood or plasma concentrations. Drugs known to induce these enzyme systems may result in an increased metabolism of tacrolimus or decreased bioavailability as indicated by decreased whole blood or plasma concentrations. Monitoring of blood concentrations and appropriate dosage adjustments are essential when such drugs are used concomitantly. *Drugs That May Increase Tacrolimus Blood Concentrations Calcium Channel Blockers Antifungal Agents Macrolide Antibiotics diltiazem clotrimazole clarithromycin nicardipine fluconazole erythromycin nifedipine itraconazole troleandomycin verapamil ketoconazole** voriconazole Gastrointestinal Prokinetic Agents Other Drugs cisapride bromocriptine metoclopramide chloramphenicol cimetidine cyclosporine danazol ethinyl estradiol methylprednisolone lansoprazole*** omeprazole protease inhibitors nefazodone magnesium-aluminum-hydroxide * This table is not all inclusive **In a study of 6 normal volunteers, a 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. *** Lansoprazole (CYP2C19, CYP3A4 substrate) may potentially inhibit CYP3A4-mediated 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. *Drugs That May Decrease Tacrolimus Blood Concentrations Anticonvulsants Antimicrobials carbamazepine rifabutin phenobarbital caspofungin phenytoin rifampin Herbal Preparations Other Drugs St. John’s Wort sirolimus *This table is not all inclusive. St. John’s Wort (Hypericum perforatum) induces CYP3A4 and P-glycoprotein. Since tacrolimus is a substrate for CYP3A4, there is the potential that the use of St. John’s Wort in patients receiving tacrolimus capsules could result in reduced tacrolimus levels. In a single-dose crossover study in healthy volunteers, co-administration of tacrolimus and magnesium-aluminum-hydroxide resulted in a 21% increase in the mean tacrolimus AUC and a 10% decrease in the mean tacrolimus C max relative to tacrolimus administration alone. In a study of 6 normal volunteers, a significant decrease in tacrolimus oral bioavailability (14 ± 6% vs. 7 ± 3%) was observed with concomitant rifampin administration (600 mg). In addition, there was a significant increase in tacrolimus clearance (0.036 ± 0.008 L/hr/kg vs. 0.053 ± 0.01 L/hr/kg) with concomitant rifampin administration. Interaction studies with drugs used in HIV therapy have not been conducted. However, care should be exercised when drugs that are nephrotoxic (e.g., ganciclovir) or that are metabolized by CYP3A (e.g., nelfinavir, ritonavir) are administered concomitantly with tacrolimus. Based on a clinical study of 5 liver transplant recipients, co-administration of tacrolimus with nelfinavir increased blood concentrations of tacrolimus significantly and, as a result, a 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. Thus, frequent monitoring of tacrolimus blood concentrations and appropriate dosage adjustments are essential when nelfinavir is used concomitantly. Tacrolimus may affect the pharmacokinetics of other drugs (e.g., phenytoin) and increase their concentration. Grapefruit juice affects CYP3A-mediated metabolism and should be avoided (see DOSAGE AND ADMINISTRATION ). Following co-administration of tacrolimus and sirolimus (2 or 5 mg/day) in stable renal transplant patients, mean tacrolimus AUC 0-12 and C min decreased approximately by 30% relative to tacrolimus alone. Mean tacrolimus AUC 0-12 and C min following co-administration of 1 mg/day of sirolimus decreased approximately 3% and 11%, respectively. The safety and efficacy of tacrolimus used in combination with sirolimus for the prevention of graft rejection has not been established and is not recommended. Other Drug Interactions Immunosuppressants may affect vaccination. Therefore, during treatment with tacrolimus capsules, vaccination may be less effective. The use of live vaccines should be avoided; live vaccines may include, but are not limited to measles, mumps, rubella, oral polio, BCG, yellow fever, and TY 21a typhoid.1 At a given MMF dose, mycophenolic acid (MPA) exposure is higher with tacrolimus co-administration than with cyclosporine co-administration due to the differences in the interruption of the enterohepatic recirculation of MPA. Clinicians should be aware that there is also a potential for increased MPA exposure after crossover from cyclosporine to tacrolimus in patients concomitantly receiving MMF or MPA. Carcinogenesis, Mutagenesis, Impairment of Fertility An increased incidence of malignancy is a recognized complication of immunosuppression in recipients of organ transplants. The most common forms of neoplasms are non-Hodgkin’s lymphomas and carcinomas of the skin. As with other immunosuppressive therapies, the risk of malignancies in tacrolimus recipients may be higher than in the normal, healthy population. Lymphoproliferative disorders associated with Epstein-Barr Virus infection have been seen. It has been reported that reduction or discontinuation of immunosuppression may cause the lesions to regress. 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. Carcinogenicity studies were carried out in male and female rats and mice. In the 80-week mouse study and in the 104-week rat study no relationship of tumor incidence to tacrolimus dosage was found. The highest doses used in the mouse and rat studies were 0.8 - 2.5 times (mice) and 3.5 - 7.1 times (rats) the recommended clinical dose range of 0.1 - 0.2 mg/kg/day when corrected for body surface area. No impairment of fertility was demonstrated in studies of male and female rats. Tacrolimus, given orally at 1 mg/kg (0.7 - 1.4X the recommended clinical dose range of 0.1 - 0.2 mg/kg/day based on body surface area corrections) to male and female rats, prior to and during mating, as well as to dams during gestation and lactation, was associated with embryolethality and with adverse effects on female reproduction. Effects on female reproductive function (parturition) and embryolethal effects were indicated by a higher rate of pre-implantation loss and increased numbers of undelivered and nonviable pups. When given at 3.2 mg/kg (2.3 - 4.6X the recommended clinical dose range based on body surface area correction), 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.

相关文档