繁体中文
设为首页
加入收藏
当前位置:药品说明书与价格首页 >> 肿瘤 >> 肾癌 >> 药品推荐 >> 坦罗莫司注射剂Torisel(25MG/MLvial+1.8MLdiluent)/Kit

坦罗莫司注射剂Torisel(25MG/MLvial+1.8MLdiluent)/Kit

2012-11-18 14:26:41  作者:新特药房  来源:互联网  浏览次数:310  文字大小:【】【】【
简介: Temsirolimus(也叫CCI-779,Torisel)属于雷帕霉素(rapamycin)衍生物,一种具抗肿瘤活性的mTOR特异性抑制剂。目前已经受美国FDA批准以药品名-Torisel上市靶向治疗进行性肾细胞癌(RCC)。中文名称: ...

Temsirolimus(也叫CCI-779,Torisel)属于雷帕霉素(rapamycin)衍生物,一种具抗肿瘤活性的mTOR特异性抑制剂。目前已经受美国FDA批准以药品名-Torisel上市靶向治疗进行性肾细胞癌(RCC)。
中文名称: 西罗莫司脂化物
中文别名: 雷帕霉素42-[3-羟基-2-(羟甲基)-2-甲基丙酸酯
英文名称: Temsirolimus
英文别名: CCL-779;Rapamycin42-[3-Hydroxy-2-(hydroxymethyl)-2-methylpropanoate;(1R,2R,5S)-5-{(2S)-2-[(3S,6R,7E,9R,10R,12R,14S,15Z,17E,19E,21S,26R,27R,34aS)-9,27-dihydroxy-10,21-dimethoxy-6,8,12,14,20,26-hexamethyl-1,5,11,28,29-pentaoxo-1,4,5,6,9,10,11,12,13,14,21,22,23,24,25,26,27,28,29,31,32,33,34,34a-tetracosahydro-3H-23,27-epoxypyrido[2,1-c][1,4]oxazacyclohentriacontin-3-yl]propyl}-2-methoxycyclohexyl3-hydroxy-2-(hydroxymethyl)-2-methylpropanoate;4-{2-[(7E,15Z,17E,19E)-9,27-dihydroxy-10,21-dimethoxy-6,8,12,14,20,26-hexamethyl-1,5,11,28,29-pentaoxo-1,4,5,6,9,10,11,12,13,14,21,22,23,24,25,26,27,28,29,31,32,33,34,34a-tetracosahydro-3H-23,27-epoxypyrido[2,1-c][1,4]oxazacyclohentriacontin-3-yl]propyl}-2-methoxycyclohexyl3-hydroxy-2-(hydroxymethyl)-2-methylpropanoate
【作用机制与药理作用】
与肿瘤发生密切相关的多项细胞功能如:细胞生长增殖、细胞周期调控、生物合成、细胞迁移等均受控于mTOR;cyclinD、myc等多种癌基因的表达在翻译水平上亦受mTOR调节。现已发现,许多肿瘤伴有mTOR信号通路调节异常。越来越多的证据¨q1显示,细胞恶性转化依赖于mTOR信号通路。
细胞生长受到复杂、精细的网络调控。P13K—Akt—mTOR信号通路控制着众多在肿瘤发生发展中至关重要的细胞生物学过程,包括细胞凋亡、转录、翻译、代谢、血管新生以及细胞周期的调控。遗传学上的改变和生化条件引起的激活经常发生在恶变早期和肿瘤进展期,同时,信号通路激活程度也是肿瘤患者预后的重要指标。因此抑制该信号通路成为肿瘤预防和肿瘤靶向治疗的热点。对于该信号通路的各个激酶,均有多种抑制剂处于临床前和临床研究阶段。mTOR的抑制剂在靶向这一信号通路抑制剂中是开发最完善、研究最透彻的一类。
mTOR是一种保守的丝/苏氨酸激酶,在P13K—Akt—mToR信号通路中位于Akt的下游,控制着蛋白质的合成、血管新生和细胞周期的进程。mTOR的抑制剂有着广泛的体内抗瘤谱,在临床研究中表现出令人振奋的有效性和特异性。
本品即是一种mTOR抑制剂,它与细胞内FK—BP一12蛋白结合,形成蛋白-药物复合物,抑制细胞内的靶点mTOR激酶的活性。mTOR激酶是调节细胞增值、生长和细胞存活的重要蛋白质。当mTOR受到抑制时,将使P70s6K与s6核糖体蛋白进行磷酸化的能力受阻。体外研究表明,本品能抑制mTOR激酶的活性,同时导致诱导组织缺氧的因子HIF一1、HIF-2以及血管内皮因子的水平下降,进而阻止新生血管的发展。
【适应证】
晚期肾细胞癌(RCC)。
【用法与用量】
成人:推荐剂量25mg,静脉输注30~60min以上,每周1次。
【药物不良反应】
较常见的不良反应发生率为30%,包括皮疹、虚弱无力、黏膜炎、恶心、厌食、水肿;严重不良反应¨一1有:高敏反应、高血糖、高血脂、间质性肺炎、肠穿孔、急性肾衰等;实验室异常发生率为30%,有贫血、高血糖、高血脂、高三酰甘油血症、碱性磷酸(酯)酶升高、血清肌酸酐升高、淋巴细胞减少症、低磷酸盐血、血小板减少(症)、天冬氨酸转氨酶升高、白细胞减少症。
【注意事项】
①为使药物准确释放,最好使用输注泵,稀释好的药液应在6h内使用。
②应注意西罗莫司及其相关的过敏反应,肝功能损害,手术期间的伤口愈合,中枢神经系统肿瘤,每周监测血细胞计数以及每两周进行化学平板实验,血糖,血脂,肾功,呼吸恶化,胃肠症状(急腹症及血便)。

③老年人、孕妇、哺乳期妇女及儿童不推荐使用。
【药物相互作用】
①避免与强的cYP3A4抑制剂合用,如酮康唑、伊曲康唑、克拉霉素、茚地那韦、奈法唑酮、那非那韦、沙奎那韦、泰利霉素、优立康唑、葡萄柚汁。如果必须合用,应考虑减低本品剂量至每周12.5mg(调整剂量前停用1周CYP3A4抑制剂)。②避免与强的CYP3A4诱导剂合用,如,地塞米松、苯妥英、卡马西平、利福平、利福布丁、异福酰胺片、苯巴比妥、圣约翰麦芽汁。如果必须合用,应考虑增加本品剂量至每周50mg。③避免与活疫苗合用,避免密切接触已接种疫苗者。④与西尼替尼合用可产生附加的毒性反应(皮疹、痛风、蜂窝组织炎);与抗凝剂合用可引起颅内出血。
【临床评价】
一项评价本品与a.干扰素联(IFN)联合用药的安全性、耐受性及最大耐受量(MTD)的多中心I/II期临床试验,对71例受试者进行本品剂量递增研究。本品按5—25mg递增剂量,每周1次静脉给药;a—IFN每周3次,皮下给药6或9Mu,以出现口腔炎、疲劳、恶心、呕吐等确定限定毒性剂量。最常见的毒性反应为白细胞减少、低磷酸盐血症、无力、贫血及高三酰甘油血症。最终确定的推荐剂量为本品15mg/IFN6MU。接受推荐剂量的39例患者中,8%取得部分响应,36%至少在24周内病情稳定,所有患者的无进展生存期为9.1个月。结果表明:本品与d-IFN联合用药对于晚期RCC患者具有抗癌活性,毒性剂鼍范围可接受,因此,III期随机临床研究联合用药的推荐剂量为本品15mg/IFN6MU。
一项考察晚期RcC患者用药剂量的II期随机、双盲、对照试验中,11l例事先进行过治疗的晚期RcC患者,随机接受本品25,75或250mg治疗,每周1次。初级评价指标为客观应答率(ORR);次级评价指标是总生存期(OS)及非进展生存期(PFS)。结果3剂量组的ORR分别为5.0%,7.9%和8.1%;临床有效率分别为52.8%,55.3%和43.2%。所有患者中位总生存期为15个月,各剂量组生存期分布未见统计学意义上的差别。
对626例先前未进行治疗的晚期肾癌(RCC)患者进行了本品多中心、开放、随机III期临床疗效与安全性评价试验,旨在研究本品的总生存期(OS)、非进展生存期(PFs)及客观应答率(ORR)。患者平均年龄59岁(23—86岁);男性69%,女性31%;种族分布:白人91%,黑人4%,亚裔2%,其他3%。随机分成三组,即本品试验组209例(25mg,每周1次,静脉输注30一60min以上),本品(15mg)和d—IFN(最大剂量6Mu)联合用药组210例,及单独应用俚-IFN组(最大剂量18Mu)207例。中期分析结果表明:本品组与IFN组比较,中位生存期分别为10.9月和7.3月,有统计学意义的改善(P=0.00r78),显著延长患者生存期达49%;次级评价指标:本品能显著延长患者的无进展(病情不进一步恶化)生存期(5.5个月对3.1个月,P=0.0001);客观应答率分别为8.6%和4.8%(P=0.132)。本品和仅一IFN联合治疗组及单独应用o一IFN组比较,延长患者生存期15%,差别未见统计学意义,但与多种不良反应事件增加有关。
【规格与包装】注射液:25mg/mL/瓶+1.8mL稀释液/套。
【贮存条件】2—8℃下避光贮存。
 
TORISEL—Powerful 1st-line evidence in poor-risk patients with advanced RCC
Median overall survival (OS)* of nearly 11 months was achieved with TORISEL1
Results from a phase 3, multicenter, 3-arm, randomized, open-label study conducted in 626 previously untreated patients with advanced RCC.1 Patients had ≥ 3 of 6 preselected prognostic risk factors.1 Patients received TORISEL (25 mg IV weekly) or IFN (maximum 18 MU subC 3 times weekly).2
49% significant increase in median OS with TORISEL vs IFN1
Median duration of treatment was 17 weeks (range 1-126 weeks) for TORISEL and 8 weeks (range 1-124 weeks) for IFN1
* Time from randomization to death.
A comparison is considered statistically significant if the P-value is <0.0159 (O'Brien-Fleming boundary at 446 deaths).
Based on log-rank test stratified by prior nephrectomy and region.
Based on Cox proportional hazard model stratified by prior nephrectomy and region.
Important Safety Information
TORISEL is contraindicated in patients with bilirubin >1.5 x ULN and should be used with caution when treating patients with mild hepatic impairment (bilirubin >1 - 1.5 x ULN or AST > ULN but bilirubin ≤ ULN). If TORISEL must be given to patients with mild hepatic impairment, reduce the dose of TORISEL to 15 mg/week. In a phase 1 study, the overall frequency of ≥ grade 3 adverse reactions and deaths, including deaths due to progressive disease, was greater in patients with baseline bilirubin > 1.5 x ULN.
Hypersensitivity/infusion reactions, including flushing, chest pain, dyspnea, hypotension, apnea, loss of consciousness, hypersensitivity and anaphylaxis, may occur very early in the first infusion or with subsequent infusions. Pretreat with an H1 antihistamine. TORISEL infusion should be interrupted in patients with infusion reactions and appropriate therapy given.
Serum glucose, serum cholesterol, and triglycerides should be tested before and during TORISEL treatment.
TORISEL is likely to result in hyperglycemia and hyperlipemia. This may result in the need for an increase in the dose of, or initiation of, insulin and/or oral hypoglycemic agent therapy and/or lipid-lowering agents, respectively.
TORISEL may result in immunosuppression. Patients should be carefully observed for the occurrence of infections, including opportunistic infections.
Cases of interstitial lung disease, some resulting in death, have occurred. Some patients were asymptomatic or had minimal symptoms. Patients should undergo baseline radiography prior to TORISEL therapy and periodically thereafter, even in the absence of clinical respiratory symptoms. Follow patients closely and, if clinically significant respiratory symptoms develop, consider withholding TORISEL until recovery of symptoms and radiographic improvement of pneumonitis findings. Some patients required TORISEL discontinuation and/or treatment with corticosteroids and/or antibiotics.
Cases of fatal bowel perforation occurred with TORISEL. These patients presented with fever, abdominal pain, metabolic acidosis, bloody stools, diarrhea, and/or acute abdomen.
Cases of rapidly progressive and sometimes fatal acute renal failure not clearly related to disease progression occurred in patients who received TORISEL.
Due to abnormal wound healing, use TORISEL with caution in the perioperative period.
Patients with central nervous system tumors (primary CNS tumor or metastases) and/or receiving anticoagulation therapy may be at an increased risk of developing intracerebral bleeding (including fatal outcomes) while receiving TORISEL.
Live vaccinations and close contact with those who received live vaccines should be avoided.
TORISEL may cause fetal harm. Patients and their partners should be advised to avoid pregnancy throughout treatment and for 3 months after TORISEL therapy has stopped.
Elderly patients may be more likely to experience certain adverse reactions including diarrhea, edema and pneumonia.
The most common (incidence ≥30%) adverse reactions observed with TORISEL are: rash (47%), asthenia (51%), mucositis (41%), nausea (37%), edema (35%), and anorexia (32%). The most common laboratory abnormalities (incidence ≥30%) are anemia (94%), hyperglycemia (89%), hyperlipemia (87%), hypertriglyceridemia (83%), elevated alkaline phosphatase (68%), elevated serum creatinine (57%), lymphopenia (53%), hypophosphatemia (49%), thrombocytopenia (40%), elevated AST (38%), and leukopenia (32%).
Most common grades 3/4 adverse events and laboratory abnormalities included asthenia (11%), dyspnea (9%), hemoglobin decreased (20%), lymphocytes decreased (16%), glucose increased (16%), phosphorus decreased (18%), and triglycerides increased (44%).
Pleural effusion, hemodynamically significant pericardial effusions requiring intervention, convulsions, rhabdomyolysis, Stevens-Johnson Syndrome, complex regional pain syndrome and extravasations have been reported during postmarketing use.
Strong inducers of CYP3A4/5 (eg, dexamethasone, rifampin) and strong inhibitors of CYP3A4 (eg, ketoconazole, atazanavir) may decrease and increase concentrations of the major metabolite of TORISEL, respectively. If alternatives cannot be used, dose modifications of TORISEL are recommended.
Avoid St. John's Wort which may decrease TORISEL plasma concentrations, and grapefruit juice which may increase plasma concentrations of the major metabolite of TORISEL.
The combination of TORISEL and sunitinib resulted in dose-limiting toxicity (Grade 3/4 erythematous maculopapular rash, and gout/cellulitis requiring hospitalization).
Please see the full Prescribing Information for TORISEL.
1 INDICATIONS AND USAGE

TORISEL is indicated for the treatment of advanced renal cell carcinoma.

2 DOSAGE AND ADMINISTRATION

2.1 Advanced Renal Cell Carcinoma

The recommended dose of TORISEL for advanced renal cell carcinoma is 25 mg infused over a 30–60 minute period once a week.

Treatment should continue until disease progression or unacceptable toxicity occurs.

2.2 Premedication

Patients should receive prophylactic intravenous diphenhydramine 25 to 50 mg (or similar antihistamine) approximately 30 minutes before the start of each dose of TORISEL [see Warnings and Precautions (5.1)].

2.3 Dosage Interruption/Adjustment

TORISEL should be held for absolute neutrophil count (ANC) < 1,000/mm3, platelet count < 75,000/mm3, or NCI CTCAE grade 3 or greater adverse reactions. Once toxicities have resolved to grade 2 or less, TORISEL may be restarted with the dose reduced by 5 mg/week to a dose no lower than 15 mg/week.

2.4 Dose Modification Guidelines

Hepatic Impairment: Use caution when treating patients with hepatic impairment. If TORISEL must be given in patients with mild hepatic impairment (bilirubin > 1 – 1.5 × ULN or AST >ULN but bilirubin ≤ULN), reduce the dose of TORISEL to 15 mg/week. TORISEL is contraindicated in patients with bilirubin >1.5 × ULN [see Contraindications (4), Warnings and Precautions (5.2) and Use in Specific Populations (8.7)].

Concomitant Strong CYP3A4 Inhibitors: The concomitant use of strong CYP3A4 inhibitors should be avoided (e.g. ketoconazole, itraconazole, clarithromycin, atazanavir, indinavir, nefazodone, nelfinavir, ritonavir, saquinavir, telithromycin, and voriconazole). Grapefruit juice may also increase plasma concentrations of sirolimus (a major metabolite of temsirolimus) and should be avoided. If patients must be co-administered a strong CYP3A4 inhibitor, based on pharmacokinetic studies, a TORISEL dose reduction to 12.5 mg/week should be considered. This dose of TORISEL is predicted to adjust the AUC to the range observed without inhibitors. However, there are no clinical data with this dose adjustment in patients receiving strong CYP3A4 inhibitors. If the strong inhibitor is discontinued, a washout period of approximately 1 week should be allowed before the TORISEL dose is adjusted back to the dose used prior to initiation of the strong CYP3A4 inhibitor [see Warnings and Precautions (5.11) and Drug Interactions (7.2)].

Concomitant Strong CYP3A4 Inducers: The use of concomitant strong CYP3A4 inducers should be avoided (e.g. dexamethasone, phenytoin, carbamazepine, rifampin, rifabutin, rifampacin, phenobarbital). If patients must be co-administered a strong CYP3A4 inducer, based on pharmacokinetic studies, a TORISEL dose increase from 25 mg/week up to 50 mg/week should be considered. This dose of TORISEL is predicted to adjust the AUC to the range observed without inducers. However, there are no clinical data with this dose adjustment in patients receiving strong CYP3A4 inducers. If the strong inducer is discontinued the temsirolimus dose should be returned to the dose used prior to initiation of the strong CYP3A4 inducer [see Warnings and Precautions (5.11) and Drug Interactions (7.1)].

2.5 Instructions for Preparation

TORISEL must be stored under refrigeration at 2°–8°C (36°–46°F) and protected from light. During handling and preparation of admixtures, TORISEL should be protected from excessive room light and sunlight. Parenteral drug products should be inspected visually for particulate matter and discoloration prior to administration, whenever solution and container permit.

In order to minimize the patient exposure to the plasticizer DEHP (di-2-ethylhexyl phthalate), which may be leached from PVC infusion bags or sets, the final TORISEL dilution for infusion should be stored in bottles (glass, polypropylene) or plastic bags (polypropylene, polyolefin) and administered through polyethylene-lined administration sets.

TORISEL 25 mg/mL injection must be diluted with the supplied diluent before further dilution in 0.9% Sodium Chloride Injection, USP.

 Please note that both the TORISEL injection and diluent vials contain an overfill to ensure the recommended volume can be withdrawn.

Follow this two-step dilution process in an aseptic manner.

Step 1:

DILUTION OF TORISEL INJECTION 25 MG/ML WITH SUPPLIED DILUENT

  •  Each Vial of Torisel (temsirolimus) must first be mixed with 1.8 mL of the enclosed diluent. The resultant solution contains 30 mg/3 mL (10 mg/mL)
  •  Mix well by inversion of the vial. Allow sufficient time for the air bubbles to subside. The solution should be clear to slightly turbid, colorless to light-yellow solution, essentially free from visual particulates.

The concentrate-diluent mixture is stable below 25°C for up to 24 hours.

Step 2:

DILUTION OF CONCENTRATE-DILUENT MIXTURE WITH 0.9% SODIUM CHLORIDE INJECTION, USP

  •  Withdraw precisely the required amount of concentrate-diluent mixture containing temsirolimus 10 mg/mL as prepared in Step 1 from the vial (i.e., 2.5 mL for a temsirolimus dose of 25 mg) and further dilute into an infusion bag containing 250 mL of 0.9% Sodium Chloride Injection, USP
  •  Mix by inversion of the bag or bottle, avoiding excessive shaking, as this may cause foaming.

 The resulting solution should be inspected visually for particulate matter and discoloration prior to administration. The admixture of TORISEL in 0.9% Sodium Chloride Injection, USP should be protected from excessive room light and sunlight.

2.6 Administration

  •  Administration of the final diluted solution should be completed within six hours from the time that TORISEL is first added to 0.9% Solution Chloride Injection, USP.
  •  TORISEL is infused over a 30- to 60-minute period once weekly. The use of an infusion pump is the preferred method of administration to ensure accurate delivery of the product.
  •  Appropriate administration materials should be composed of glass, polyolefin, or polyethylene to avoid excessive loss of product and diethylhexylpthalate (DEHP) extraction. The administration materials should consist of non-DEHP, non-polyvinylchloride (PVC) tubing with appropriate filter. In the case when a PVC administration set has to be used, it should not contain DEHP. An in-line polyethersulfone filter with a pore size of not greater than 5 microns is recommended for administration to avoid the possibility of particles bigger than 5 microns being infused. If the administration set available does not have an in-line filter incorporated, a polyethersulfone filter should be added at the set (i.e., an end-filter) before the admixture reaches the vein of the patient. Different end-filters can be used, ranging in filter pore size from 0.2 microns up to 5 microns. The use of both an in-line and end-filter is not recommended.
  •  TORISEL, when diluted, contains polysorbate 80, which is known to increase the rate of DEHP extraction from PVC. This should be considered during the preparation and administration of TORISEL, including storage time elapsed when in direct contact with PVC following constitution.

 Compatibilities and Incompatibilities

 Undiluted TORISEL injection should not be added directly to aqueous infusion solutions. Direct addition of TORISEL injection to aqueous solutions will result in precipitation of drug. Always combine TORISEL injection with DILUENT for TORISEL before adding to infusion solutions. It is recommended that TORISEL be administered in 0.9% Sodium Chloride Injection after combining with diluent. The stability of TORISEL in other infusion solutions has not been evaluated. Addition of other drugs or nutritional agents to admixtures of TORISEL in 0.9% Sodium Chloride Injection has not been evaluated and should be avoided. Temsirolimus is degraded by both acids and bases, and thus combinations of temsirolimus with agents capable of modifying solution pH should be avoided.

3 DOSAGE FORMS AND STRENGTHS

TORISEL® (temsirolimus) is supplied as a kit consisting of the following:

TORISEL (temsirolimus) injection (25 mg/mL). The TORISEL vial contains temsirolimus at a concentration of 25 mg/mL. The vial contains an overfill of 0.2 mL to ensure the ability to withdraw the recommended dose.

DILUENT for TORISEL®. The DILUENT vial includes a deliverable volume of 1.8 mL. This vial contains an overfill in order to ensure that the appropriate volume can be withdrawn.

4 CONTRAINDICATIONS

TORISEL is contraindicated in patients with bilirubin > 1.5 × ULN [see Warnings and Precautions (5.2)].

5 WARNINGS AND PRECAUTIONS

5.1 Hypersensitivity/Infusion Reactions

 Hypersensitivity/infusion reactions, including but not limited to flushing, chest pain, dyspnea, hypotension, apnea, loss of consciousness, hypersensitivity and anaphylaxis, have been associated with the administration of temsirolimus. These reactions can occur very early in the first infusion, but may also occur with subsequent infusions. Patients should be monitored throughout the infusion and appropriate supportive care should be available. Temsirolimus infusion should be interrupted in all patients with severe infusion reactions and appropriate medical therapy administered.

 TORISEL should be used with caution in persons with known hypersensitivity to temsirolimus or its metabolites (including sirolimus), polysorbate 80, or to any other component (including the excipients) of TORISEL.

 An H1 antihistamine should be administered to patients before the start of the intravenous temsirolimus infusion. TORISEL should be used with caution in patients with known hypersensitivity to an antihistamine, or patients who cannot receive an antihistamine for other medical reasons.

 If a patient develops a hypersensitivity reaction during the TORISEL infusion, the infusion should be stopped and the patient should be observed for at least 30 to 60 minutes (depending on the severity of the reaction). At the discretion of the physician, treatment may be resumed with the administration of an H1-receptor antagonist (such as diphenhydramine), if not previously administered [see Dosage and Administration (2.2)], and/or an H2-receptor antagonist (such as intravenous famotidine 20 mg or intravenous ranitidine 50 mg) approximately 30 minutes before restarting the TORISEL infusion. The infusion may then be resumed at a slower rate (up to 60 minutes).

 A benefit-risk assessment should be done prior to the continuation of temsirolimus therapy in patients with severe or life-threatening reactions.

5.2 Hepatic Impairment

The safety and pharmacokinetics of TORISEL were evaluated in a dose escalation phase 1 study in 110 patients with normal or varying degrees of hepatic impairment. Patients with baseline bilirubin > 1.5 × ULN experienced greater toxicity than patients with baseline bilirubin ≤1.5 × ULN when treated with TORISEL. The overall frequency of ≥ grade 3 adverse reactions and deaths, including deaths due to progressive disease, were greater in patients with baseline bilirubin >1.5 × ULN due to increased risk of death [see Contraindications (4)].

Use caution when treating patients with mild hepatic impairment. Concentrations of temsirolimus and its metabolite sirolimus were increased in patients with elevated AST or bilirubin levels. If TORISEL must be given in patients with mild hepatic impairment (bilirubin >1 – 1.5 × ULN or AST >ULN but bilirubin ≤ULN), reduce the dose of TORISEL to 15 mg/week [see Dosage and Administration (2.4)].

5.3 Hyperglycemia/Glucose Intolerance

The use of TORISEL is likely to result in increases in serum glucose. In the phase 3 trial, 89% of patients receiving TORISEL had at least one elevated serum glucose while on treatment, and 26% of patients reported hyperglycemia as an adverse event. This may result in the need for an increase in the dose of, or initiation of, insulin and/or oral hypoglycemic agent therapy. Serum glucose should be tested before and during treatment with TORISEL. Patients should be advised to report excessive thirst or any increase in the volume or frequency of urination.

5.4 Infections

The use of TORISEL may result in immunosuppression. Patients should be carefully observed for the occurrence of infections, including opportunistic infections [see Adverse Reactions (6.1)].

5.5 Interstitial Lung Disease

 Cases of interstitial lung disease, some resulting in death, occurred in patients who received TORISEL. Some patients were asymptomatic, or had minimal symptoms, with infiltrates detected on computed tomography scan or chest radiograph. Others presented with symptoms such as dyspnea, cough, hypoxia, and fever. Some patients required discontinuation of TORISEL and/or treatment with corticosteroids and/or antibiotics, while some patients continued treatment without additional intervention. Patients should be advised to report promptly any new or worsening respiratory symptoms.

 It is recommended that patients undergo baseline radiographic assessment by lung computed tomography scan or chest radiograph prior to the initiation of TORISEL therapy. Follow such assessments periodically, even in the absence of clinical respiratory symptoms.

 It is recommended that patients be followed closely for occurrence of clinical respiratory symptoms. If clinically significant respiratory symptoms develop, consider withholding TORISEL administration until after recovery of symptoms and improvement of radiographic findings related to pneumonitis. Empiric treatment with corticosteroids and/or antibiotics may be considered.

5.6 Hyperlipemia

The use of TORISEL is likely to result in increases in serum triglycerides and cholesterol. In the phase 3 trial, 87% of patients receiving TORISEL had at least one elevated serum cholesterol value and 83% had at least one elevated serum triglyceride value. This may require initiation, or increase in the dose, of lipid-lowering agents. Serum cholesterol and triglycerides should be tested before and during treatment with TORISEL.

5.7 Bowel Perforation

Cases of fatal bowel perforation occurred in patients who received TORISEL. These patients presented with fever, abdominal pain, metabolic acidosis, bloody stools, diarrhea, and/or acute abdomen. Patients should be advised to report promptly any new or worsening abdominal pain or blood in their stools.

5.8 Renal Failure

Cases of rapidly progressive and sometimes fatal acute renal failure not clearly related to disease progression occurred in patients who received TORISEL. Some of these cases were not responsive to dialysis.

5.9 Wound Healing Complications

Use of TORISEL has been associated with abnormal wound healing. Therefore, caution should be exercised with the use of TORISEL in the perioperative period.

5.10 Intracerebral Hemorrhage

Patients with central nervous system tumors (primary CNS tumor or metastases) and/or receiving anticoagulation therapy may be at an increased risk of developing intracerebral bleeding (including fatal outcomes) while receiving TORISEL.

5.11 Co-administration with Inducers or Inhibitors of CYP3A Metabolism

Agents Inducing CYP3A Metabolism:

Strong inducers of CYP3A4/5 such as dexamethasone, carbamazepine, phenytoin, phenobarbital, rifampin, rifabutin, and rifampacin may decrease exposure of the active metabolite, sirolimus. If alternative treatment cannot be administered, a dose adjustment should be considered. St. John's Wort may decrease TORISEL plasma concentrations unpredictably. Patients receiving TORISEL should not take St. John's Wort concomitantly. [see Dosage and Administration (2.4) and Drug Interactions (7.1)].

Agents Inhibiting CYP3A Metabolism:

Strong CYP3A4 inhibitors such as atazanavir, clarithromycin, indinavir, itraconazole, ketoconazole, nefazodone, nelfinavir, ritonavir, saquinavir, and telithromycin may increase blood concentrations of the active metabolite sirolimus. If alternative treatments cannot be administered, a dose adjustment should be considered. [see Dosage and Administration (2.4) and Drug Interactions (7.2)].

5.12 Concomitant use of TORISEL with sunitinib

The combination of TORISEL and sunitinib resulted in dose-limiting toxicity. Dose-limiting toxicities (Grade 3/4 erythematous maculopapular rash, and gout/cellulitis requiring hospitalization) were observed in two out of three patients treated in the first cohort of a phase 1 study at doses of TORISEL 15 mg IV per week and sunitinib 25 mg oral per day (Days 1–28 followed by a 2-week rest).

5.13 Vaccinations

The use of live vaccines and close contact with those who have received live vaccines should be avoided during treatment with TORISEL. Examples of live vaccines are: intranasal influenza, measles, mumps, rubella, oral polio, BCG, yellow fever, varicella, and TY21a typhoid vaccines.

5.14 Use in Pregnancy

There are no adequate and well-controlled studies of TORISEL in pregnant women. However, based on its mechanism of action, TORISEL may cause fetal harm when administered to a pregnant woman. Temsirolimus administered daily as an oral formulation caused embryo-fetal and intrauterine toxicities in rats and rabbits at human sub-therapeutic exposures. If this drug is used during pregnancy or if the patient becomes pregnant while taking the drug, the patient should be apprised of the potential hazard to the fetus. Women of childbearing potential should be advised to avoid becoming pregnant throughout treatment and for 3 months after TORISEL therapy has stopped. [see Use in Specific Populations (8.1)]

Men should be counseled regarding the effects of TORISEL on the fetus and sperm prior to starting treatment [see Nonclinical Toxicology (13.1)]. Men with partners of childbearing potential should use reliable contraception throughout treatment and are recommended to continue this for 3 months after the last dose of TORISEL.

5.15 Elderly Patients

 Based on the results of a phase 3 study, elderly patients may be more likely to experience certain adverse reactions including diarrhea, edema and pneumonia [see Use in Specific Populations (8.5)].

5.16 Monitoring Laboratory Tests

In the randomized, phase 3 trial, complete blood counts (CBCs) were checked weekly, and chemistry panels were checked every two weeks. Laboratory monitoring for patients receiving TORISEL may need to be performed more or less frequently at the physician's discretion.

6 ADVERSE REACTIONS

The following serious adverse reactions have been associated with TORISEL in clinical trials and are discussed in greater detail in other sections of the label [see Warnings and Precautions (5)].

Hypersensitivity/Infusion Reactions [see Warnings and Precautions (5.1)]
Hyperglycemia/Glucose Intolerance [see Warnings and Precautions (5.3)]
Interstitial Lung Disease [see Warnings and Precautions (5.5)]
Hyperlipemia [see Warnings and Precautions (5.6)]
Bowel Perforation [see Warnings and Precautions (5.7)]
Renal Failure [see Warnings and Precautions (5.8)]

The most common (≥ 30%) adverse reactions observed with TORISEL are rash, asthenia, mucositis, nausea, edema, and anorexia. The most common (≥ 30%) laboratory abnormalities observed with TORISEL are anemia, hyperglycemia, hyperlipemia, hypertriglyceridemia, lymphopenia, elevated alkaline phosphatase, elevated serum creatinine, hypophosphatemia, thrombocytopenia, elevated AST, and leukopenia.

6.1 Clinical Trials Experience

Because clinical trials are conducted under widely varying conditions, the adverse reaction rates observed cannot be directly compared to rates in other trials and may not reflect the rates observed in clinical practice.

In the Phase 3 randomized, open-label study of interferon alfa (IFN-α) alone, TORISEL alone, and TORISEL and IFN-α, a total of 616 patients were treated. Two hundred patients received IFN-α weekly, 208 received TORISEL 25 mg weekly, and 208 patients received a combination of TORISEL and IFN-α weekly [see Clinical Studies (14)].

Treatment with the combination of TORISEL 15 mg and IFN-α was associated with an increased incidence of multiple adverse reactions and did not result in a significant increase in overall survival when compared with IFN-α alone.

Table 1 shows the percentage of patients experiencing treatment emergent adverse reactions. Reactions reported in at least 10% of patients who received TORISEL 25 mg alone or IFN-α alone are listed. Table 2 shows the percentage of patients experiencing selected laboratory abnormalities. Data for the same adverse reactions and laboratory abnormalities in the IFN-α alone arm are shown for comparison.

Table 1 – Adverse Reactions Reported in at Least 10% of Patients Who Received 25 mg IV TORISEL or IFN-α in the Randomized Trial
TORISEL
25 mg
IFN-α
n=208 n=200
Adverse Reaction All
Grades
n (%)
Grades
3&4
n (%)
All
Grades
n (%)
Grades
3&4
n (%)
Common Toxicity Criteria for Adverse Events (CTCAE), Version 3.0.
Includes edema, facial edema, and peripheral edema
Includes aphthous stomatitis, glossitis, mouth ulceration, mucositis, and stomatitis
Includes infections not otherwise specified (NOS) and the following infections that occurred infrequently as distinct entities: abscess, bronchitis, cellulitis, herpes simplex, and herpes zoster
Includes cystitis, dysuria, hematuria, urinary frequency, and urinary tract infection
Includes eczema, exfoliative dermatitis, maculopapular rash, pruritic rash, pustular rash, rash (NOS), and vesiculobullous rash
Includes taste loss and taste perversion
General disorders
106 (51) 23 (11) 127 (64) 52 (26)
Asthenia
  Edema† 73 (35) 7 (3) 21 (11) 1 (1)
  Pain 59 (28) 10 (5) 31 (16) 4 (2)
  Pyrexia 50 (24) 1 (1) 99 (50) 7 (4)
  Weight Loss 39 (19) 3 (1) 50 (25) 4 (2)
  Headache 31 (15) 1 (1) 30 (15) 0 (0)
  Chest Pain 34 (16) 2 (1) 18 (9) 2 (1)
  Chills 17 (8) 1 (1) 59 (30) 3 (2)
Gastrointestinal disorders
  Mucositis‡ 86 (41) 6 (3) 19 (10) 0 (0)
  Anorexia 66 (32) 6 (3) 87 (44) 8 (4)
  Nausea 77 (37) 5 (2) 82 (41) 9 (5)
  Diarrhea 56 (27) 3 (1) 40 (20) 4 (2)
  Abdominal Pain 44 (21) 9 (4) 34 (17) 3 (2)
  Constipation 42 (20) 0 (0) 36 (18) 1 (1)
  Vomiting 40 (19) 4 (2) 57 (29) 5 (3)
Infections
  Infections§ 42 (20) 6 (3) 19 (10) 4 (2)
  Urinary tract infection 31 (15) 3 (1) 24 (12) 3 (2)
  Pharyngitis 25 (12) 0 (0) 3 (2) 0 (0)
  Rhinitis 20 (10) 0 (0) 4 (2) 0 (0)
Musculoskeletal and connective tissue disorders
  Back Pain 41 (20) 6 (3) 28 (14) 7 (4)
  Arthralgia 37 (18) 2 (1) 29 (15) 2 (1)
  Myalgia 16 (8) 1 (1) 29 (15) 2 (1)
Respiratory, thoracic and mediastinal disorders
  Dyspnea 58 (28) 18 (9) 48 (24) 11 (6)
  Cough 53 (26) 2 (1) 29 (15) 0 (0)
  Epistaxis 25 (12) 0 (0) 7 (4) 0 (0)
Skin and subcutaneous tissue disorders
  Rash# 97 (47) 10 (5) 14 (7) 0 (0)
  Pruritus 40 (19) 1 (1) 16 (8) 0 (0)
  Nail Disorder 28 (14) 0 (0) 1 (1) 0 (0)
  Dry Skin 22 (11) 1 (1) 14 (7) 0 (0)
  Acne 21 (10) 0 (0) 2 (1) 0 (0)
Nervous system disorders
  DysgeusiaÞ 41 (20) 0 (0) 17 (9) 0 (0)
  Insomnia 24 (12) 1 (1) 30 (15) 0 (0)
  Depression 9 (4) 0 (0) 27 (14) 4 (2)

The following selected adverse reactions were reported less frequently (<10%).

Gastrointestinal Disorders – Fatal bowel perforation occurred in 1 patient (1%).

Eye Disorders - Conjunctivitis (including lacrimation disorder) occurred in 15 patients (7%).

Immune System - Allergic/Hypersensitivity reactions occurred in 18 patients (9%).

Angioneurotic edema-type reactions (including delayed reactions occurring two months following initiation of therapy) have been observed in some patients who received TORISEL and ACE inhibitors concomitantly.

Infections - Pneumonia occurred in 17 patients (8%); upper respiratory tract infection occurred in 14 patients (7%).

General Disorders and Administration Site Conditions - Impaired wound healing occurred in 3 patients (1%).

Respiratory, Thoracic and Mediastinal Disorders – Interstitial lung disease occurred in 5 patients (2%), including rare fatalities.

Vascular - Hypertension occurred in 14 patients (7%); venous thromboembolism (including deep vein thrombosis and pulmonary embolus [including fatal outcomes]) occurred in 5 patients (2%); thrombophlebitis occurred in 2 patients (1%).

Table 2 – Incidence of Selected Laboratory Abnormalities in Patients Who Received 25 mg IV TORISEL or IFN-α in the Randomized Trial
TORISEL
25 mg
IFN-α
n=208 n=200
Laboratory Abnormality All
Grades*
n (%)
Grades
3&4*
n (%)
All
Grades*
n (%)
Grades
3&4*
n (%)
NCI CTC version 3.0
Grade 1 toxicity may be under-reported for lymphocytes and neutrophils
Any 208 (100) 162 (78) 195 (98) 144 (72)
Hematology
  Hemoglobin Decreased 195 (94) 41 (20) 180 (90) 43 (22)
  Lymphocytes Decreased† 110 (53) 33 (16) 106 (53) 48 (24)
  Neutrophils Decreased† 39 (19) 10 (5) 58 (29) 19 (10)
  Platelets Decreased 84 (40) 3 (1) 51 (26) 0 (0)
  Leukocytes Decreased 67 (32) 1 (1) 93 (47) 11 (6)
Chemistry
  Alkaline Phosphatase Increased 141 (68) 7 (3) 111 (56) 13 (7)
  AST Increased 79 (38) 5 (2) 103 (52) 14 (7)
  Creatinine Increased 119 (57) 7 (3) 97 (49) 2 (1)
  Glucose Increased 186 (89) 33 (16) 128 (64) 6 (3)
  Phosphorus Decreased 102 (49) 38 (18) 61 (31) 17 (9)
  Total Bilirubin Increased 16 (8) 2 (1) 25 (13) 4 (2)
  Total Cholesterol Increased 181 (87) 5 (2) 95 (48) 2 (1)
  Triglycerides Increased 173 (83) 92 (44) 144 (72) 69 (35)
  Potassium Decreased 43 (21) 11 (5) 15 (8) 0 (0)
6.2 Post-marketing and Other Clinical Experience

The following adverse reactions have been identified during post approval use of TORISEL. Because these reactions are reported voluntarily from a population of uncertain size, it is not always possible to readily estimate their frequency or establish a causal relationship to drug exposure.

The following adverse reactions have been observed in patients receiving temsirolimus: pleural effusion, hemodynamically significant pericardial effusions requiring intervention, convulsions, rhabdomyolysis, Stevens-Johnson Syndrome, and complex regional pain syndrome (reflex sympathetic dystrophy).

There are also postmarketing reports of temsirolimus extravasations resulting in swelling, pain, warmth, and erythema.

7 DRUG INTERACTIONS

7.1 Agents Inducing CYP3A Metabolism

Co-administration of TORISEL with rifampin, a potent CYP3A4/5 inducer, had no significant effect on temsirolimus Cmax (maximum concentration) and AUC (area under the concentration versus the time curve) after intravenous administration, but decreased sirolimus Cmax by 65% and AUC by 56% compared to TORISEL treatment alone. If alternative treatment cannot be administered, a dose adjustment should be considered [see Dosage and Administration (2.4)].

7.2 Agents Inhibiting CYP3A Metabolism

Co-administration of TORISEL with ketoconazole, a potent CYP3A4 inhibitor, had no significant effect on temsirolimus Cmax or AUC; however, sirolimus AUC increased 3.1-fold, and Cmax increased 2.2-fold compared to TORISEL alone. If alternative treatment cannot be administered, a dose adjustment should be considered. [see Dosage and Administration (2.4)].

7.3 Interactions with Drugs Metabolized by CYP2D6

The concentration of desipramine, a CYP2D6 substrate, was unaffected when 25 mg of TORISEL was co-administered. No clinically significant effect is anticipated when temsirolimus is co-administered with agents that are metabolized by CYP2D6 or CYP3A4.

8 USE IN SPECIFIC POPULATIONS

8.1 Pregnancy

Teratogenic Effects

Pregnancy Category D [see Warnings and Precautions (5.14)].

Women of childbearing potential should be advised to avoid becoming pregnant throughout treatment and for 3 months after TORISEL therapy has stopped. Temsirolimus can cause fetal harm when administered to a pregnant woman. If this drug is used during pregnancy, or if the patient becomes pregnant while taking this drug, the patient should be apprised of the potential hazard to the fetus.

Temsirolimus administered daily as an oral formulation caused embryo-fetal and intrauterine toxicities in rats and rabbits at human sub-therapeutic exposures. Embryo-fetal adverse effects in rats consisted of reduced fetal weight and reduced ossifications, and in rabbits included reduced fetal weight, omphalocele, bifurcated sternabrae, notched ribs, and incomplete ossifications.

In rats, the intrauterine and embryo-fetal adverse effects were observed at the oral dose of 2.7 mg/m2/day (approximately 0.04-fold the AUC in cancer patients at the human recommended dose). In rabbits, the intrauterine and embryo-fetal adverse effects were observed at the oral dose of ≥7.2 mg/m2/day (approximately 0.12-fold the AUC in cancer patients at the recommended human dose).

8.3 Nursing Mothers

It is not known whether TORISEL is excreted into human milk, and due to the potential for tumorigenicity shown for sirolimus (active metabolite of TORISEL) in animal studies, a decision should be made whether to discontinue nursing or discontinue TORISEL, taking into account the importance of the drug to the mother.

8.4 Pediatric Use

Limited data are available on the use of temsirolimus in pediatric patients. The effectiveness of temsirolimus in pediatric patients with advanced recurrent/refractory solid tumors has not been established.

TORISEL was studied in 71 patients (59 patients ages 1 to 17 years and 12 patients ages 18 to 21 years) with relapsed/refractory solid tumors in a phase 1–2 safety and exploratory pharmacodynamic study.

In phase 1, 19 pediatric patients with advanced recurrent/refractory solid tumors received TORISEL at doses ranging from 10 mg/m2 to 150 mg/m2 as a 60-minute intravenous infusion once weekly in three-week cycles.

In phase 2, 52 pediatric patients with recurrent/relapsed neuroblastoma, rhabdomyosarcoma or high grade glioma received TORISEL at a weekly dose of 75 mg/m2. One of 19 patients with neuroblastoma achieved a partial response. There were no objective responses in pediatric patients with recurrent/relapsed rhabdomyosarcoma or high grade glioma.

Adverse reactions associated with TORISEL were similar to those observed in adults. The most common adverse reactions (≥ 20%) in pediatric patients receiving the 75 mg/m2 dose included thrombocytopenia, infections, asthenia/fatigue, fever, pain, leukopenia, rash, anemia, hyperlipidemia, increased cough, stomatitis, anorexia, increased plasma levels of alanine aminotransferase and aspartate aminotransferase, hypercholesterolemia, hyperglycemia, abdominal pain, headache, arthralgia, upper respiratory infection, nausea and vomiting, neutropenia, hypokalemia and hypophosphatemia.

Pharmacokinetics

In phase 1 of the above mentioned pediatric trial, the single dose and multiple dose total systemic exposure (AUC) of temsirolimus and sirolimus were less than dose-proportional over the dose range of 10 to 150 mg/m2.

In the phase 2 portion, the multiple dose (Day 1, Cycle 2) pharmacokinetics of TORISEL 75 mg/m2 were characterized in an additional 35 patients ages 28 days to 21 years (median age of 8 years). The geometric mean body surface adjusted clearance of temsirolimus and sirolimus was 9.45 L/h/m2 and 9.26 L/h/m2, respectively. The mean elimination half-life of temsirolimus and sirolimus was 31 hours and 44 hours, respectively.

The exposure (AUCss) to temsirolimus and sirolimus was approximately 6-fold and 2-fold higher, respectively than the exposure in adult patients receiving a 25 mg intravenous infusion.

8.5 Geriatric Use

Clinical studies of TORISEL did not include sufficient numbers of subjects aged 65 and older to determine whether they respond differently from younger subjects. Based on the results of a phase 3 study, elderly patients may be more likely to experience certain adverse reactions including diarrhea, edema and pneumonia [see Warnings and Precautions (5.15)].

8.6 Renal Impairment

No clinical studies were conducted with TORISEL in patients with decreased renal function. Less than 5% of total radioactivity was excreted in the urine following a 25 mg intravenous dose of [14C]-labeled temsirolimus in healthy subjects. Renal impairment is not expected to markedly influence drug exposure, and no dosage adjustment of TORISEL is recommended in patients with renal impairment.

TORISEL has not been studied in patients undergoing hemodialysis.

8.7 Hepatic Impairment

TORISEL was evaluated in a dose escalation phase 1 study in 110 patients with normal or varying degrees of hepatic impairment as defined by AST and bilirubin levels and patients with liver transplant (Table 3). Patients with moderate and severe hepatic impairment had increased rates of adverse reactions and deaths, including deaths due to progressive disease, during the study (Table 3).

Table 3 – Adverse Reactions in Patients with Advanced Malignancies Plus Normal or Impaired Hepatic Function
Hepatic Function* TORISEL Dose Range Adverse Reactions
Grade ≥ 3†
n (%)
Death‡
n (%)
*
Hepatic Function Groups: normal = bilirubin and AST ≤ULN; mild = bilirubin >1 – 1.5 × ULN or AST >ULN but bilirubin ≤ULN; moderate = bilirubin >1.5 – 3 × ULN; severe = bilirubin >3 × ULN; liver transplant = any bilirubin and AST.
Common Terminology Criteria for Adverse Events, version 3.0, including all causality.
Includes deaths due to progressive disease and adverse reactions.
Normal (n=25) 25 – 175 20 (80.0) 2 (8.0)
Mild (n=39) 10 – 25 32 (82.1) 5 (12.8)
Moderate (n=20) 10 – 25 19 (95.0) 8 (40.0)
Severe (n=24) 7.5 – 15 23 (95.8) 13 (54.2)
Liver Transplant (n=2) 10 1 (50.0) 0 (0)

TORISEL is contraindicated in patients with bilirubin >1.5 × ULN [see Contraindications (4), and Warnings and Precautions (5.2)]. Use caution when treating patients with mild hepatic impairment. If TORISEL must be given in patients with mild hepatic impairment (bilirubin >1 – 1.5 × ULN or AST >ULN but bilirubin ≤ULN), reduce the dose of TORISEL to 15 mg/week [see Dosage and Administration (2.4)]. Because there is a need for dosage adjustment based upon hepatic function, assessment of AST and bilirubin levels is recommended before initiation of TORISEL and periodically thereafter.

10 OVERDOSAGE

There is no specific treatment for TORISEL intravenous overdose. TORISEL has been administered to patients with cancer in phase 1 and 2 trials with repeated intravenous doses as high as 220 mg/m2. The risk of several serious adverse events, including thrombosis, bowel perforation, interstitial lung disease (ILD), seizure, and psychosis, is increased with doses of TORISEL greater than 25 mg.

11 DESCRIPTION

Temsirolimus, an inhibitor of mTOR, is an antineoplastic agent.

Temsirolimus is a white to off-white powder with a molecular formula of C56H87NO16 and a molecular weight of 1030.30. It is non-hygroscopic. Temsirolimus is practically insoluble in water and soluble in alcohol. It has no ionizable functional groups, and its solubility is independent of pH.

The chemical name of temsirolimus is (3S,6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34aS)-9,10,12,13,14,21,22,23,24,25,26,27,32,33,34,34a-Hexadecahydro-9,27-dihydroxy-3-[(1R)-2-[(1S,3R,4R)-4-hydroxy-3-methoxycyclohexyl]-1-methylethyl]-10,21-dimethoxy-6,8,12,14,20,26-hexamethyl-23, 27-epoxy-3H-pyrido[2,1-c][1,4]oxaazacyclohentriacontine-1,5,11,28,29(4H,6H,31H)-pentone 4'-[2,2-bis(hydroxymethyl)propionate]; or Rapamycin, 42-[3-hydroxy-2-(hydroxymethyl)-2-methylpropanoate].

TORISEL (temsirolimus) injection, 25 mg/mL, is a clear, colorless to light yellow, non-aqueous, ethanolic, sterile solution. TORISEL (temsirolimus) injection requires two dilutions prior to intravenous infusion. TORISEL (temsirolimus) injection should be diluted only with the supplied DILUENT for TORISEL®.

DILUENT for TORISEL® is a sterile, non-aqueous solution that is supplied with TORISEL injection, as a kit.

TORISEL (temsirolimus) injection, 25 mg/mL:

Active ingredient: temsirolimus (25 mg/mL)

Inactive ingredients: dehydrated alcohol (39.5% w/v), dl-alpha-tocopherol (0.075% w/v), propylene glycol (50.3% w/v), and anhydrous citric acid (0.0025% w/v).

DILUENT for TORISEL®

Inactive ingredients: polysorbate 80 (40.0% w/v), polyethylene glycol 400 (42.8% w/v) and dehydrated alcohol (19.9% w/v).

After the TORISEL (temsirolimus) injection vial has been diluted with DILUENT for TORISEL®, in accordance with the instructions in section 2.5, the solution contains 35.2% alcohol.

TORISEL (temsirolimus) injection and DILUENT for TORISEL® are filled in clear glass vials with butyl rubber stoppers.

12 CLINICAL PHARMACOLOGY

12.1 Mechanism of Action

Temsirolimus is an inhibitor of mTOR (mammalian target of rapamycin). Temsirolimus binds to an intracellular protein (FKBP-12), and the protein-drug complex inhibits the activity of mTOR that controls cell division. Inhibition of mTOR activity resulted in a G1 growth arrest in treated tumor cells. When mTOR was inhibited, its ability to phosphorylate p70S6k and S6 ribosomal protein, which are downstream of mTOR in the PI3 kinase/AKT pathway was blocked. In in vitro studies using renal cell carcinoma cell lines, temsirolimus inhibited the activity of mTOR and resulted in reduced levels of the hypoxia-inducible factors HIF-1 and HIF-2 alpha, and the vascular endothelial growth factor.

12.2 Pharmacodynamics

Effects on Electrocardiogram There were no clinically relevant QT changes observed at the recommended dose for TORISEL. In a randomized, single-blinded, crossover study, 58 healthy subjects received TORISEL 25 mg, placebo, and a single oral dose of moxifloxacin 400 mg. A supratherapeutic TORISEL dose was not studied in this randomized QT trial. The largest difference between the upper bound 2-sided 90% CI for the mean difference between TORISEL and placebo-corrected QT interval was less than 10 ms. In a different trial in 69 patients with a hematologic malignancy, TORISEL doses up to 175mg were studied. No patient with a normal QTcF at baseline had an increase in QTcF >60 ms. Additionally, there were no patients with a QTcF interval greater than 500 ms.

12.3 Pharmacokinetics

Absorption

Following administration of a single 25 mg dose of TORISEL in patients with cancer, mean temsirolimus Cmax in whole blood was 585 ng/mL (coefficient of variation, CV =14%), and mean AUC in blood was 1627 ng•h/mL (CV=26%). Typically Cmax occurred at the end of infusion. Over the dose range of 1 mg to 25 mg, temsirolimus exposure increased in a less than dose proportional manner while sirolimus exposure increased proportionally with dose. Following a single 25 mg intravenous dose in patients with cancer, sirolimus AUC was 2.7-fold that of temsirolimus AUC, due principally to the longer half-life of sirolimus.

Distribution

Following a single 25 mg intravenous dose, mean steady-state volume of distribution of temsirolimus in whole blood of patients with cancer was 172 liters. Both temsirolimus and sirolimus are extensively partitioned into formed blood elements.

Metabolism

Cytochrome P450 3A4 is the major isozyme responsible for the formation of five temsirolimus metabolites. Sirolimus, an active metabolite of temsirolimus, is the principal metabolite in humans following intravenous treatment. The remainder of the metabolites account for less than 10% of radioactivity in the plasma. In human liver microsomes temsirolimus was an inhibitor of CYP2D6 and 3A4. However, there was no effect observed in vivo when temsirolimus was administered with desipramine (a CYP2D6 substrate), and no effect is anticipated with substrates of CYP3A4 metabolism.

Elimination

Elimination is primarily via the feces. After a single IV dose of [14C]-temsirolimus approximately 82% of total radioactivity was eliminated within 14 days, with 4.6% and 78% of the administered radioactivity recovered in the urine and feces, respectively. Following a single 25 mg dose of TORISEL in patients with cancer, temsirolimus mean (CV) systemic clearance was 16.2 (22%) L/h. Temsirolimus exhibits a bi-exponential decline in whole blood concentrations and the mean half-lives of temsirolimus and sirolimus were 17.3 hr and 54.6 hr, respectively.

Drug-Transport Systems - P-glycoprotein

Temsirolimus is a substrate of the efflux transporter P-glycoprotein (Pgp) in vitro. If TORISEL is administered with drugs that inhibit Pgp, increased concentrations of temsirolimus are likely and caution should be exercised.

In vitro, temsirolimus inhibited human Pgp (IC50 value of 2µM). If TORISEL is administered with drugs that are substrates of Pgp, increased concentrations of the substrate drug are likely and caution should be exercised.

Effects of Age and Gender

In population pharmacokinetic-based data analyses, no relationship was apparent between drug exposure and patient age or gender.

13 NONCLINICAL TOXICOLOGY

13.1 Carcinogenesis, Mutagenesis, Impairment of Fertility

Carcinogenicity studies have not been conducted with temsirolimus. However, sirolimus, the major metabolite of temsirolimus in humans, was carcinogenic in mice and rats. The following effects were reported in mice and/or rats in the carcinogenicity studies conducted with sirolimus: lymphoma, hepatocellular adenoma and carcinoma, and testicular adenoma.

Temsirolimus was not genotoxic in a battery of in vitro (bacterial reverse mutation in Salmonella typhimurium and Escherichia coli, forward mutation in mouse lymphoma cells, and chromosome aberrations in Chinese hamster ovary cells) and in vivo (mouse micronucleus) assays.

In male rats, the following fertility effects were observed: decreased number of pregnancies, decreased sperm concentration and motility, decreased reproductive organ weights, and testicular tubular degeneration. These effects were observed at oral temsirolimus doses ≥ 3 mg/m2/day (approximately 0.2-fold the human recommended intravenous dose). Fertility was absent at 30 mg/m2/day.

In female rats, an increased incidence of pre- and post-implantation losses occurred at oral doses ≥ 4.2 mg/m2/day (approximately 0.3-fold the human recommended intravenous dose), resulting in decreased numbers of live fetuses.

14 CLINICAL STUDIES

A phase 3, multi-center, three-arm, randomized, open-label study was conducted in previously untreated patients with advanced renal cell carcinoma (clear cell and non-clear cell histologies). The objectives were to compare Overall Survival (OS), Progression-Free Survival (PFS), Objective Response Rate (ORR), and safety in patients receiving IFN-α to those receiving TORISEL or TORISEL plus IFN-α. Patients in this study had 3 or more of 6 pre-selected prognostic risk factors (less than one year from time of initial RCC diagnosis to randomization, Karnofsky performance status of 60 or 70, hemoglobin less than the lower limit of normal, corrected calcium of greater than 10 mg/dL, lactate dehydrogenase > 1.5 times the upper limit of normal, more than one metastatic organ site). Patients were stratified for prior nephrectomy status within three geographic regions and were randomly assigned (1:1:1) to receive IFN-α alone (n=207), TORISEL alone (25 mg weekly; n=209), or the combination arm (n=210).

The ITT population for this interim analysis included 626 patients. Demographics were comparable between the three treatment arms with regard to age, gender, and race. The mean age of all groups was 59 years (range 23–86). Sixty-nine percent were male and 31% were female. The racial distribution for all groups was 91% White, 4% Black, 2% Asian, and 3% other. Sixty-seven percent of patients had a history of prior nephrectomy.

The median duration of treatment in the TORISEL arm was 17 weeks (range 1–126 weeks). The median duration of treatment on the IFN arm was 8 weeks (range 1–124 weeks).

There was a statistically significant improvement in OS (time from randomization to death) in the TORISEL 25 mg arm compared to IFN-α. The combination of TORISEL 15 mg and IFN-α did not result in a significant increase in overall survival when compared with IFN-α alone. Figure 1 is a Kaplan-Meier plot of OS in this study. The evaluations of PFS (time from randomization to disease progression or death) and ORR, were based on blinded independent radiologic assessment of tumor response. Efficacy results are summarized in Table 4.

Table 4- Summary of Efficacy Results of TORISEL vs. IFN-α
TORISEL
n = 209
IFN-α
n = 207
P-value Hazard Ratio
(95% CI)
CI = confidence interval; NA = not applicable
Based on log-rank test stratified by prior nephrectomy and region.
Based on Cox proportional hazard model stratified by prior nephrectomy and region.
A comparison is considered statistically significant if the p-value is <0.0159 (O'Brien-Fleming boundary at 446 deaths).
Not adjusted for multiple comparisons.
Based on Cochran-Mantel-Haenszel test stratified by prior nephrectomy and region.
Median Overall Survival
Months (95% CI)
10.9 (8.6, 12.7) 7.3 (6.1, 8.8) 0.0078‡ 0.73 (0.58, 0.92)
Median Progression-Free Survival
Months (95% CI)
5.5 (3.9, 7.0) 3.1 (2.2, 3.8) 0.0001§ 0.66 (0.53, 0.81)
Overall Response Rate
% (95% CI)
8.6 (4.8, 12.4) 4.8 (1.9, 7.8) 0.1232§¶ NA

Figure 1: Kaplan-Meier Curves for Overall Survival – TORISEL vs. IFN

15 REFERENCES

  1. NIOSH Alert: Preventing occupational exposures to antineoplastic and other hazardous drugs in healthcare settings. 2004. U.S. Department of Health and Human Services, Public Health Service, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, DHHS (NIOSH) Publication No. 2004-165.
  2. OSHA Technical Manual, TED 1-0.15A, Section VI: Chapter 2. Controlling Occupational Exposure to Hazardous Drugs. OSHA, 1999. http://www.osha.gov/dts/osta/otm/otm_vi/otm_vi_2.html
  3. American Society of Health-System Pharmacists. ASHP guidelines on handling hazardous drugs. Am J Health-Syst Pharm. (2006) 63:1172–1193.
  4. Polovich, M., White, J. M., & Kelleher, L.O. (eds.) 2005. Chemotherapy and biotherapy guidelines and recommendations for practice (2nd. ed.) Pittsburgh, PA: Oncology Nursing Society.

16 HOW SUPPLIED/STORAGE AND HANDLING

NDC 0008-1179-01 TORISEL® (temsirolimus) injection, 25 mg/mL.

STORAGE AND HANDLING

Each kit is supplied in a single carton containing one single-use vial of 25 mg/mL of temsirolimus and one DILUENT vial which includes a deliverable volume of 1.8 mL, and must be stored at 2°–8° C (36°–46° F). Protect from light.

一般名
テムシロリムス(Temsirolimus) 
トーリセル点滴静注液25mg:1バイアル(希釈用液 1バイアル付き)

製造販売 
ファイザー株式会社
完整资料附件1:http://www.info.pmda.go.jp/go/pack/4291418A1025_1_03/
完整资料附件2:http://www.drugsdb.eu/

靶向治疗肾癌药物Torisel上市
坦罗莫司(Temsirolimus,CCI-779)是一种特异性mTOR抑制剂。在Ⅲ期临床研究中对晚期肾细胞癌具有良好的疗效,延长总生存期至10.9个月。其主要不良反应包括代谢失调(高血脂、高血糖)、黏膜炎、皮疹和肺炎等。2007年5月美国FDA批准坦罗莫司用于难治性晚期肾细胞癌的一线治疗,具有广泛的应用前景。
美国FDA已经批准Torisel(Temsirolimus)上市用于治疗进行肾细胞癌(RCC)。Torisel是第一个治疗肾癌的靶向治疗药物,将在今年7月正式上市。
肾细胞癌占全部肾肿瘤的85%。美国癌症协会预计今年将有新诊断肾癌51190例,其中40%在诊断时已经是晚期或发展阶段。Torisel是唯一上市的特异抑制mTOR激酶的药物,mTOR激酶是调节细胞增值、生长和细胞存活重要的蛋白质。在体外研究中发现,Torisel抑制mTOR激酶后导致一定的血管生长因子如血管内皮生长因子的水平下降,进而阻止新生血管的发展。作为上市后安全监测措施,惠氏将提供2套完整的数据,其一已经完成,另一个关于肝细胞毒的实验即将开始。Torisel也是目前唯一能够显著延长患者生存期的药物。惠氏为Torisel进行了包括626名患者的III期临床研究,共分为三组。Torisel单独一组,Torisel和alpha干扰素混合组以及alpha干扰素单独一组。结果显示,Torisel比alpha干扰素显著延长患者生存率达49%(10.9个月对7.3个月);在次级评价指标上,Torisel比alpha干扰素显著延长患者的无进展(病情不进一步恶化)生存期(5.5个月对3.1个月)。Torisel和alpha干扰素组合使用没有显示更好的作用。
-----------------------------------------------------------
产地国家: 美国
原产地英文商品名:
Torisel (25MG/MLvial+1.8MLdiluent)/Kit
原产地英文药品名:
Temsirolimus
原产地英文化合物名称:
Antineoplastic (mTOR kinase inhibitor)
中文参考商品译名:
Torisel (25毫克/毫升瓶+1.8毫升稀释液)/套盒
中文参考药品译名:
Temsirolimus
中文参考化合物名称:
Antineoplastic (mTOR kinase inhibitor)
生产厂家中文参考译名:
美国惠氏公司
生产厂家英文名:
Wyeth Pharmaceuticals Inc

责任编辑:admin


相关文章
坦罗莫司注射剂|TORISEL(TEMSIROLIMUS)
Sutent(Sunitinib Malate Capsules)
索拉非尼司特片|NEXAVAR(Sorafenib Tosilate tablets)
依维莫司片|AFINITOR(everolimus Tablets)
AFINITOR(everolimus Tablets)依维莫司片
依维莫司片|Afinitor(everolimus Tablets)
阿西替尼片INLYTA(axitinib Tablels)
盐酸帕唑帕尼片|VOTRIENT(PAZOPANIB HYDROCHLORIDE)
TORISEL(Temsirolimus Injection)
坦罗莫司作为晚期肾细胞癌二线用药临床研究
 

最新文章

更多

· 坦罗莫司注射剂|TORISEL...
· 依维莫司片|AFINITOR(ev...
· 舒尼替尼胶囊|Sutent(su...
· AFINITOR(everolimus Ta...
· AFINITOR(依维莫司片,...
· AFINITOR dispersible t...
· 干扰素注射液Sumiferon(...
· 舒尼替尼胶囊SUTENT(索...
· 索坦胶囊Sutent(Sunitin...
· 阿西替尼片|Inlyta(Axi...

推荐文章

更多

· 坦罗莫司注射剂|TORISEL...
· 依维莫司片|AFINITOR(ev...
· 舒尼替尼胶囊|Sutent(su...
· AFINITOR(everolimus Ta...
· AFINITOR(依维莫司片,...
· AFINITOR dispersible t...
· 干扰素注射液Sumiferon(...
· 舒尼替尼胶囊SUTENT(索...
· 索坦胶囊Sutent(Sunitin...
· 阿西替尼片|Inlyta(Axi...

热点文章

更多