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 >> 上市新药 >> JAKAFI(RUXOLITINIB PHOSPHATE)Tablets

JAKAFI(RUXOLITINIB PHOSPHATE)Tablets

2015-01-18 06:59:03  作者:新特药房  来源:互联网  浏览次数:221  文字大小:【】【】【
简介: 美国批准第一个药物Jakafi(ruxolitinib)治疗一种罕见骨髓病2011年11月16日,美国食品药品监督管理局批准Jakafi (ruxolitinib)。是批准特异性治疗骨髓纤维化骨髓病患者的第一个药物。骨髓纤维化是一种 ...

美国批准第一个药物Jakafi(ruxolitinib)治疗一种罕见骨髓病
2011年11月16日,美国食品药品监督管理局批准Jakafi (ruxolitinib)。是批准特异性治疗骨髓纤维化骨髓病患者的第一个药物。
骨髓纤维化是一种疾病,导致制造血细胞器官如肝和脾骨髓被瘢痕组织替代。此病标志为脾脏肿大,贫血,白血细胞和血小板减低,和骨髓纤维化-相关症状。症状包括疲乏,腹部不适,肋下疼痛,饱感(早饱),肌肉和骨痛,瘙痒,和夜汗。
Jakafi,一种每天服药2次药丸,抑制酶被称为JAK 1和2 (Janus相关激酶)涉及调节血液和免疫学功能。JAK 1和2调节障碍伴随骨髓纤维化。
美国FDA药物评价和研究中心血液和肿瘤产品办公室主任Richard Pazdur, M.D.说:“Jakafi代表在肿瘤中详尽了解疾病特异性机制,允许药物靶向特异性分子通路的另一个实例”“临床试验导致这个批注集中在经常遇到的骨髓纤维化,包括脾脏肿大和疼痛患者的问题。”
在528例患者的两项临床试验中评价Jakafi的安全性和有效性。两项试验患者都是对可以得到骨髓纤维化治疗耐药或难治或对同种异体骨髓移植(即患者接受来自遗传上相似,但不一定相同供体的造血干细胞)不合格。所有患者有脾脏肿大和因疾病-相关症状的结果需要治疗。
在研究中被选择患者接受或Jakafi治疗,安慰剂(糖丸)或可得到的最佳治疗(羟基脲[hydroxyurea],一种化疗药,或糖皮质激素)。有更大百分率的接受Jakafi患者,当与接受安慰剂或可得到最佳治疗患比较时,经受脾脏大小减低大于35百分率。相似地,接受Jakafia比接受安慰剂患者,有更大患者比例见到骨髓纤维化-相关症状,包括腹部不适,夜汗,瘙痒和骨或肌肉痛减低50%以上.
用Jakafi治疗患者见到最严重副作用包括血小板计数减低,贫血,疲乏,腹泻,气短(呼吸困难),头痛,眩晕,和恶心。
Jakafi是在FDA的优先审评程序下被审评,即对可能提供显著进展超过可得到治疗的治疗或对当前没有适当治疗提供治疗加快6-个月审评药物的一种程序。
本治疗正在按处方药物用户费用法[Prescription Drug User Fee Act]下的目标日期2011年12月3日前被批准和已被指定为孤儿药物,确定疾病在美国影响人数少于200,000人。
批准日期:2011年11月16日; Jakafi由Wilmington, Del.Incyte Corp.公司制造。
JAKAFI - ruxolitinib tablet 
Incyte Corporation


HIGHLIGHTS OF PRESCRIBING INFORMATION
These highlights do not include all the information needed to use JAKAFI safely and effectively. See full prescribing information for JAKAFI.
JAKAFI™ (ruxolitinib) tablets, for oral use
Initial U.S. Approval: 2011
INDICATIONS AND USAGE
Jakafi is a kinase inhibitor indicated for treatment of patients with intermediate or high-risk myelofibrosis, including primary myelofibrosis, post-polycythemia vera myelofibrosis and post-essential thrombocythemia myelofibrosis. (1)
DOSAGE AND ADMINISTRATION
The starting dose of Jakafi is 20 mg given orally twice daily for patients with a platelet count greater than 200 X 109/L, and 15 mg twice daily for patients with a platelet count between 100 X 109/L and 200 X 109/L. (2.1)
Perform a complete blood count before initiating therapy with Jakafi. Monitor complete blood counts every 2 to 4 weeks until doses are stabilized, and then as clinically indicated. Modify dose for thrombocytopenia. (2.1) (2.2)
Increase dose based on response and as recommended to a maximum of 25 mg twice daily. Discontinue after 6 months if no spleen reduction or symptom improvement (2.3)
DOSAGE FORMS AND STRENGTHS
Tablets: 5 mg, 10 mg, 15 mg, 20 mg and 25 mg. (3)
CONTRAINDICATIONS
None. (4)
WARNINGS AND PRECAUTIONS
Thrombocytopenia, anemia and neutropenia can occur. Manage by dose reduction, or interruption or transfusion. (5.1)
Assess patients for signs and symptoms of infection and initiate appropriate treatment promptly. Serious infections should have resolved before starting therapy with Jakafi. (5.2)
ADVERSE REACTIONS
The most common hematologic adverse reactions (incidence > 20%) are thrombocytopenia and anemia. The most common non-hematologic adverse reactions (incidence >10%) are bruising, dizziness and headache. (6.1)
To report SUSPECTED ADVERSE REACTIONS, contact Incyte Corporation at 1-855-463-3463 or FDA at 1-800-FDA-1088 or www.fda.gov/medwatch.
DRUG INTERACTIONS
Strong CYP3A4 Inhibitors: Reduce Jakafi starting dose to 10 mg twice daily for patients with a platelet count greater than or equal to 100 X 109/L and concurrent use of strong CYP3A4 inhibitors. Avoid in patients with platelet counts less than 100 X 109/L. (2.4) (7.1)
USE IN SPECIFIC POPULATIONS
Renal Impairment: Reduce Jakafi starting dose to 10 mg twice daily for patients with moderate (CrCl 30-59 mL/min) or severe renal impairment (CrCl 15-29 mL/min) and a platelet count between 100 X 109/L and 150 X 109/L. Avoid in patients with end stage renal disease (CrCl less than 15 mL/min) not requiring dialysis and in patients with moderate or severe renal impairment and a platelet count less than 100 X 109/L. (2.5) (8.6)
Hepatic Impairment: Reduce Jakafi starting dose to 10 mg twice daily for patients with any degree of hepatic impairment and a platelet count between 100 X 109/L and 150 X 109/L. Avoid in patients with hepatic impairment with platelet counts less than 100 X 109/L. (2.5) (8.7)
Nursing Mothers: Discontinue nursing or discontinue the drug taking into account the importance of the drug to the mother. (8.3)
See 17 for PATIENT COUNSELING INFORMATION and FDA-approved patient labeling 
Revised: 11/2011
FULL PRESCRIBING INFORMATION: CONTENTS*

1. INDICATIONS AND USAGE

Jakafi is indicated for treatment of patients with intermediate or high-risk myelofibrosis, including primary myelofibrosis, post-polycythemia vera myelofibrosis and post-essential thrombocythemia myelofibrosis.

2. DOSAGE AND ADMINISTRATION

2.1 Recommended Starting Dose

The recommended starting dose of Jakafi is based on platelet count (Table 1). A complete blood count (CBC) and platelet count must be performed before initiating therapy, every 2 to 4 weeks until doses are stabilized, and then as clinically indicated [see Warnings and Precautions (5.1)]. Doses may be titrated based on safety and efficacy.

Table 1: Proposed Jakafi Starting Doses
Platelet Count Starting Dose
Greater than 200 X 109/L 20 mg orally twice daily
100 X 109/L to 200 X 109/L 15 mg orally twice daily
2.2 Dose Modification Guidelines for Thrombocytopenia

Treatment Interruption

Interrupt treatment for platelet counts less than 50 X 109/L. After recovery of platelet counts above this level, dosing may be restarted or increased following recovery of platelet counts to acceptable levels. Table 2 illustrates the maximum allowable dose that may be used in restarting Jakafi after a previous interruption.

Table 2: Maximum Restarting Doses for Jakafi After Safety Interruption*
Current Platelet Count Maximum Dose When
Restarting Jakafi Treatment *
Greater than or equal to 125 X 109/L 20 mg twice daily
100 to less than 125 X 109/L 15 mg twice daily
75 to less than 100 X 109/L 10 mg twice daily for at least 2 weeks; if stable, may increase to 15 mg twice daily
50 to less than 75 X 109/L 5 mg twice daily for at least 2 weeks; if stable, may increase to 10 mg twice daily
Less than 50 X 109/L Continue hold
*Maximum doses are displayed. When restarting, begin with a dose at least 5 mg twice daily below the dose at interruption.

Dose Reductions

Dose reductions should be considered if the platelet counts decrease as outlined in Table 3 with the goal of avoiding dose interruptions for thrombocytopenia.

Table 3: Dosing Recommendations for Thrombocytopenia
  Dose at Time of Platelet Decline
Platelet Count 25 mg
twice daily
20 mg
twice daily
15 mg
twice daily
10 mg
twice daily
5 mg
twice daily
         
New Dose New Dose New Dose New Dose New Dose
         
100 to less than 125 X 109/L 20 mg
twice daily
15 mg
twice daily
No Change No Change No Change
75 to less than 100 X 109/L 10 mg
twice daily
10 mg
twice daily
10 mg
twice daily
No Change No Change
50 to less than 75 X 109/L 5 mg
twice daily
5 mg
twice daily
5 mg
twice daily
5 mg
twice daily
No Change
Less than 50 X 109/L Hold Hold Hold Hold Hold
2.3 Dose Modification Based on Response

If efficacy is considered insufficient and platelet and neutrophil counts are adequate, doses may be increased in 5 mg twice daily increments to a maximum of 25 mg twice daily. Doses should not be increased during the first 4 weeks of therapy and not more frequently than every 2 weeks. Discontinue treatment after 6 months if there is no spleen size reduction or symptom improvement since initiation of therapy with Jakafi.

Based on limited clinical data, long-term maintenance at a 5 mg twice daily dose has not shown responses and continued use at this dose should be limited to patients in whom the benefits outweigh the potential risks.

Consider dose increases in patients who meet all of the following conditions:

a. Failure to achieve a reduction from pretreatment baseline in either palpable spleen length of 50% or a 35% reduction in spleen volume as measured by CT or MRI;
b. Platelet count greater than 125 X 109/L at 4 weeks and platelet count never below 100 X 109/L;
c. ANC levels greater than 0.75 X 109/L.

2.4 Dose Adjustment with Concomitant Strong CYP3A4 Inhibitors

On the basis of pharmacokinetic studies in healthy volunteers, when administering Jakafi with strong CYP3A4 inhibitors (such as but not limited to boceprevir, clarithromycin, conivaptan, grapefruit juice, indinavir, itraconazole, ketoconazole, lopinavir/ritonavir, mibefradil, nefazodone, nelfinavir, posaconazole, ritonavir, saquinavir, telaprevir, telithromycin, voriconazole), the recommended starting dose is 10 mg twice daily for patients with a platelet count greater than or equal to 100 X 109/L. Additional dose modifications should be made with careful monitoring of safety and efficacy.

Concurrent administration of Jakafi with strong CYP3A4 inhibitors should be avoided in patients with platelet counts less than 100 X 109/L [see Drug Interactions (7.1)].

2.5 Organ Impairment

Renal Impairment

On the basis of pharmacokinetic studies in volunteers with renal impairment, the recommended starting dose is 10 mg twice daily for patients with a platelet count between 100 X 109/L and 150 X 109/L and moderate (CrCl 30-59 mL/min) or severe renal impairment (CrCl 15-29 mL/min). Additional dose modifications should be made with careful monitoring of safety and efficacy.

The recommended starting dose for patients with end stage renal disease on dialysis is 15 mg for patients with a platelet count between 100 X 109/L and 200 X 109/L or 20 mg for patients with a platelet count of greater than 200 X 109/L. Subsequent doses should be administered on dialysis days following each dialysis session. Additional dose modifications should be made with careful monitoring of safety and efficacy.

Jakafi should be avoided in patients with end stage renal disease (CrCl less than 15 mL/min) not requiring dialysis and in patients with moderate or severe renal impairment with platelet counts less than 100 X 109/L [see Use in Specific Populations (8.6)].

Hepatic Impairment

On the basis of pharmacokinetic studies in volunteers with hepatic impairment, the recommended starting dose is 10 mg twice daily for patients with a platelet count between 100 X 109/L and 150 X 109/L. Additional dose modifications should be made with careful monitoring of safety and efficacy.

Jakafi should be avoided in patients with hepatic impairment with platelet counts less than 100 X 109/L [see Use in Specific Populations (8.7)].

2.6 Method of Administration

Jakafi is dosed orally and can be administered with or without food.

If a dose is missed, the patient should not take an additional dose, but should take the next usual prescribed dose.

When discontinuing Jakafi therapy for reasons other than thrombocytopenia, gradual tapering of the dose of Jakafi may be considered, for example by 5 mg twice daily each week.

For patients unable to ingest tablets, Jakafi can be administered through a nasogastric tube (8 French or greater) as follows:

  • Suspend one tablet in approximately 40 mL of water with stirring for approximately 10 minutes.
  • Within 6 hours after the tablet has dispersed, the suspension can be administered through a nasogastric tube using an appropriate syringe.

The tube should be rinsed with approximately 75 mL of water. The effect of tube feeding preparations on Jakafi exposure during administration through a nasogastric tube has not been evaluated.

3. DOSAGE FORMS AND STRENGTHS

5 mg tablets - round and white with "INCY" on one side and "5" on the other.

10 mg tablets - round and white with "INCY" on one side and "10" on the other.

15 mg tablets - oval and white with "INCY" on one side and "15" on the other.

20 mg tablets - capsule-shaped and white with "INCY" on one side and "20" on the other.

25 mg tablets - oval and white with "INCY" on one side and "25" on the other.

4. CONTRAINDICATIONS

None.

5. WARNINGS AND PRECAUTIONS

5.1 Thrombocytopenia, Anemia and Neutropenia

Treatment with Jakafi can cause hematologic adverse reactions, including thrombocytopenia, anemia and neutropenia. A complete blood count must be performed before initiating therapy with Jakafi [see Dosage and Administration (2.1)].

Patients with platelet counts of less than 200 X 109/L at the start of therapy are more likely to develop thrombocytopenia during treatment.

Thrombocytopenia was generally reversible and was usually managed by reducing the dose or temporarily withholding Jakafi. If clinically indicated, platelet transfusions may be administered [see Dosage and Administration (2.2), and Adverse Reactions (6.1)].

Patients developing anemia may require blood transfusions. Dose modifications of Jakafi for patients developing anemia may also be considered.

Neutropenia (ANC less than 0.5 X 109/L) was generally reversible and was managed by temporarily withholding Jakafi [see Adverse Reactions (6.1)].

Complete blood counts should be monitored as clinically indicated and dosing adjusted as required [see Dosage and Administration (2.2), and Adverse Reactions (6.1)].

5.2 Infections

Patients should be assessed for the risk of developing serious bacterial, mycobacterial, fungal and viral infections. Active serious infections should have resolved before starting therapy with Jakafi. Physicians should carefully observe patients receiving Jakafi for signs and symptoms of infection and initiate appropriate treatment promptly.

Herpes Zoster

Physicians should inform patients about early signs and symptoms of herpes zoster and advise patients to seek treatment as early as possible [see Adverse Reactions (6.1)].

6. ADVERSE REACTIONS

6.1 Clinical Trials Experience

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

The safety of Jakafi was assessed in 617 patients in six clinical studies with a median duration of follow-up of 10.9 months, including 301 patients with myelofibrosis in two Phase 3 studies.

In these two Phase 3 studies, patients had a median duration of exposure to Jakafi of 9.5 months (range 0.5 to 17 months), with 88.7% of patients treated for more than 6 months and 24.6% treated for more than 12 months. One hundred and eleven (111) patients started treatment at 15 mg twice daily and 190 patients started at 20 mg twice daily.

In a double-blind, randomized, placebo-controlled study of Jakafi, 155 patients were treated with Jakafi. The most frequent adverse drug reactions were thrombocytopenia and anemia [see Table 5]. Thrombocytopenia, anemia and neutropenia are dose related effects. The three most frequent non-hematologic adverse reactions were bruising, dizziness, and headache [see Table 4].

Discontinuation for adverse events, regardless of causality, was observed in 11.0% of patients treated with Jakafi and 10.6% of patients treated with placebo.

Following interruption or discontinuation of Jakafi, symptoms of myelofibrosis generally return to pretreatment levels over a period of approximately 1 week. There have been isolated cases of patients discontinuing Jakafi during acute intercurrent illnesses after which the patient's clinical course continued to worsen; however, it has not been established whether discontinuation of therapy contributed to the clinical course in these patients. When discontinuing therapy for reasons other than thrombocytopenia, gradual tapering of the dose of Jakafi may be considered [see Dosage and Administration (2.6)].

Table 4 presents the most common adverse reactions occurring in patients who received Jakafi in the double-blind, placebo-controlled study during randomized treatment.

Table 4: Adverse Reactions Occurring in Patients on Jakafi in the Double-blind, Placebo-controlled Study During Randomized Treatment
a National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE), version 3.0
b includes contusion, ecchymosis, hematoma, injection site hematoma, periorbital hematoma, vessel puncture site hematoma, increased tendency to bruise, petechiae, purpura
c includes dizziness, postural dizziness, vertigo, balance disorder, Meniere's Disease, labyrinthitis
d includes urinary tract infection, cystitis, urosepsis, urinary tract infection bacterial, kidney infection, pyuria, bacteria urine, bacteria urine identified, nitrite urine present
e includes weight increased, abnormal weight gain
f includes herpes zoster and post-herpetic neuralgia
  Jakafi
(N=155)
Placebo
(N=151)
Adverse Reactions All Gradesa
(%)
Grade 3
(%)
Grade 4
(%)
All Grades
(%)
Grade 3
(%)
Grade 4
(%)
Bruisingb 23.2 0.6 0 14.6 0 0
Dizzinessc 18.1 0.6 0 7.3 0 0
Headache 14.8 0 0 5.3 0 0
Urinary Tract Infectionsd 9.0 0 0 5.3 0.7 0.7
Weight Gaine 7.1 0.6 0 1.3 0.7 0
Flatulence 5.2 0 0 0.7 0 0
Herpes Zosterf 1.9 0 0 0.7 0 0

Description of Selected Adverse Drug Reactions

Anemia

In the two Phase 3 clinical studies, median time to onset of first CTCAE Grade 2 or higher anemia was approximately 6 weeks. One patient (0.3%) discontinued treatment because of anemia. In patients receiving Jakafi, mean decreases in hemoglobin reached a nadir of approximately 1.5 to 2.0 g/dL below baseline after 8 to 12 weeks of therapy and then gradually recovered to reach a new steady state that was approximately 1.0 g/dL below baseline. This pattern was observed in patients regardless of whether they had received transfusions during therapy.

In the randomized, placebo-controlled study, 60% of patients treated with Jakafi and 38% of patients receiving placebo received red blood cell transfusions during randomized treatment. Among transfused patients, the median number of units transfused per month was 1.2 in patients treated with Jakafi and 1.7 in placebo treated patients.

Thrombocytopenia

In the two Phase 3 clinical studies, in patients who developed Grade 3 or 4 thrombocytopenia, the median time to onset was approximately 8 weeks. Thrombocytopenia was generally reversible with dose reduction or dose interruption. The median time to recovery of platelet counts above 50 X 109/L was 14 days. Platelet transfusions were administered to 4.7% of patients receiving Jakafi and to 4.0% of patients receiving control regimens. Discontinuation of treatment because of thrombocytopenia occurred in 0.7% of patients receiving Jakafi and 0.9% of patients receiving control regimens. Patients with a platelet count of 100 X 109/L to 200 X 109/L before starting Jakafi had a higher frequency of Grade 3 or 4 thrombocytopenia compared to patients with a platelet count greater than 200 X 109/L (16.5% versus 7.2%).

Neutropenia

In the two Phase 3 clinical studies, 1.0% of patients reduced or stopped Jakafi because of neutropenia.

Table 5 provides the frequency and severity of clinical hematology abnormalities reported for patients receiving treatment with Jakafi or placebo in the placebo-controlled study.

Table 5: Worst Hematology Laboratory Abnormalities in the Placebo-controlled Studya
a Presented values are worst Grade values regardless of baseline
b National Cancer Institute Common Terminology Criteria for Adverse Events, version 3.0
  Jakafi
(N=155)
Placebo
(N=151)
Laboratory Parameter All Gradesb
(%)
Grade 3
(%)
Grade 4
(%)
All Grades
(%)
Grade 3
(%)
Grade 4
(%)
Thrombocytopenia 69.7 9.0 3.9 30.5 1.3 0
Anemia 96.1 34.2 11.0 86.8 15.9 3.3
Neutropenia 18.7 5.2 1.9 4.0 0.7 1.3

Additional Data from the Placebo-controlled Study

25.2% of patients treated with Jakafi and 7.3% of patients treated with placebo developed newly occurring or worsening Grade 1 abnormalities in alanine transaminase (ALT). The incidence of greater than or equal to Grade 2 elevations was 1.9% for Jakafi with 1.3% Grade 3 and no Grade 4 ALT elevations.

17.4% of patients treated with Jakafi and 6.0% of patients treated with placebo developed newly occurring or worsening Grade 1 abnormalities in aspartate transaminase (AST). The incidence of Grade 2 AST elevations was 0.6% for Jakafi with no Grade 3 or 4 AST elevations.

16.8% of patients treated with Jakafi and 0.7% of patients treated with placebo developed newly occurring or worsening Grade 1 elevations in cholesterol. The incidence of Grade 2 cholesterol elevations was 0.6% for Jakafi with no Grade 3 or 4 cholesterol elevations.

7. DRUG INTERACTIONS

7.1 Drugs That Inhibit or Induce Cytochrome P450 Enzymes

Ruxolitinib is predominantly metabolized by CYP3A4.

Strong CYP3A4 inhibitors: The Cmax and AUC of ruxolitinib increased 33% and 91%, respectively, with Jakafi administration (10 mg single dose) following ketoconazole 200 mg twice daily for four days, compared to receiving Jakafi alone in healthy subjects. The half-life was also prolonged from 3.7 to 6.0 hours with concurrent use of ketoconazole. The change in the pharmacodynamic marker, pSTAT3 inhibition, was consistent with the corresponding ruxolitinib AUC following concurrent administration with ketoconazole.

When administering Jakafi with strong CYP3A4 inhibitors a dose reduction is recommended [see Dosage and Administration (2.4)]. Patients should be closely monitored and the dose titrated based on safety and efficacy.

Mild or moderate CYP3A4 inhibitors: There was an 8% and 27% increase in the Cmax and AUC of ruxolitinib, respectively, with Jakafi administration (10 mg single dose) following erythromycin, a moderate CYP3A4 inhibitor, at 500 mg twice daily for 4 days, compared to receiving Jakafi alone in healthy subjects. The change in the pharmacodynamic marker, pSTAT3 inhibition was consistent with the corresponding exposure information.

No dose adjustment is recommended when Jakafi is coadministered with mild or moderate CYP3A4 inhibitors (eg, erythromycin).

CYP3A4 inducers: The Cmax and AUC of ruxolitinib decreased 32% and 61%, respectively, with Jakafi administration (50 mg single dose) following rifampin 600 mg once daily for 10 days, compared to receiving Jakafi alone in healthy subjects. In addition, the relative exposure to ruxolitinib's active metabolites increased approximately 100%. This increase may partially explain the reported disproportionate 10% reduction in the pharmacodynamic marker pSTAT3 inhibition.

No dose adjustment is recommended when Jakafi is coadministered with a CYP3A4 inducer. Patients should be closely monitored and the dose titrated based on safety and efficacy.

8. USE IN SPECIFIC POPULATIONS

8.1 Pregnancy

Pregnancy Category C

There are no adequate and well-controlled studies of Jakafi in pregnant women. In embryofetal toxicity studies, treatment with ruxolitinib resulted in an increase in late resorptions and reduced fetal weights at maternally toxic doses. Jakafi should be used during pregnancy only if the potential benefit justifies the potential risk to the fetus.

Ruxolitinib was administered orally to pregnant rats or rabbits during the period of organogenesis, at doses of 15, 30 or 60 mg/kg/day in rats and 10, 30 or 60 mg/kg/day in rabbits. There was no evidence of teratogenicity. However, decreases of approximately 9% in fetal weights were noted in rats at the highest and maternally toxic dose of 60 mg/kg/day. This dose results in an exposure (AUC) that is approximately 2 times the clinical exposure at the maximum recommended dose of 25 mg twice daily. In rabbits, lower fetal weights of approximately 8% and increased late resorptions were noted at the highest and maternally toxic dose of 60 mg/kg/day. This dose is approximately 7% the clinical exposure at the maximum recommended dose.

In a pre- and post-natal development study in rats, pregnant animals were dosed with ruxolitinib from implantation through lactation at doses up to 30 mg/kg/day. There were no drug-related adverse findings in pups for fertility indices or for maternal or embryofetal survival, growth and development parameters at the highest dose evaluated (34% the clinical exposure at the maximum recommended dose of 25 mg twice daily).

8.3 Nursing Mothers

It is not known whether ruxolitinib is excreted in human milk. Ruxolitinib and/or its metabolites were excreted in the milk of lactating rats with a concentration that was 13-fold the maternal plasma. Because many drugs are excreted in human milk and because of the potential for serious adverse reactions in nursing infants from Jakafi, a decision should be made to discontinue nursing or to discontinue the drug, taking into account the importance of the drug to the mother.

8.4 Pediatric Use

The safety and effectiveness of Jakafi in pediatric patients have not been established.

8.5 Geriatric Use

Of the total number of myelofibrosis patients in clinical studies with Jakafi, 51.9% were 65 years of age and older. No overall differences in safety or effectiveness of Jakafi were observed between these patients and younger patients.

8.6 Renal Impairment

The safety and pharmacokinetics of single dose Jakafi (25 mg) were evaluated in a study in healthy subjects [CrCl 72-164 mL/min (N=8)] and in subjects with mild [CrCl 53-83 mL/min (N=8)], moderate [CrCl 38-57 mL/min (N=8)], or severe renal impairment [CrCl 15-51 mL/min (N=8)]. Eight (8) additional subjects with end stage renal disease requiring hemodialysis were also enrolled.

The pharmacokinetics of ruxolitinib was similar in subjects with various degrees of renal impairment and in those with normal renal function. However, plasma AUC values of ruxolitinib metabolites increased with increasing severity of renal impairment. This was most marked in the subjects with end stage renal disease requiring hemodialysis. The change in the pharmacodynamic marker, pSTAT3 inhibition, was consistent with the corresponding increase in metabolite exposure. Ruxolitinib is not removed by dialysis; however, the removal of some active metabolites by dialysis cannot be ruled out.

When administering Jakafi to patients with moderate (CrCl 30-59 mL/min) or severe renal impairment (CrCl 15-29 mL/min) with a platelet count between 100 X 109/L and 150 X 109/L and patients with end stage renal disease on dialysis a dose reduction is recommended [see Dosage and Administration (2.5)].

8.7 Hepatic Impairment

The safety and pharmacokinetics of single dose Jakafi (25 mg) were evaluated in a study in healthy subjects (N=8) and in subjects with mild [Child-Pugh A (N=8)], moderate [Child-Pugh B (N=8)], or severe hepatic impairment [Child-Pugh C (N=8)]. The mean AUC for ruxolitinib was increased by 87%, 28% and 65%, respectively, in patients with mild, moderate and severe hepatic impairment compared to patients with normal hepatic function. The terminal elimination half-life was prolonged in patients with hepatic impairment compared to healthy controls (4.1-5.0 hours versus 2.8 hours). The change in the pharmacodynamic marker, pSTAT3 inhibition, was consistent with the corresponding increase in ruxolitinib exposure except in the severe (Child-Pugh C) hepatic impairment cohort where the pharmacodynamic activity was more prolonged in some subjects than expected based on plasma concentrations of ruxolitinib.

When administering Jakafi to patients with any degree of hepatic impairment and with a platelet count between 100 X 109/L and 150 X 109/L, a dose reduction is recommended [see Dosage and Administration (2.5)].

10. OVERDOSAGE

There is no known antidote for overdoses with Jakafi. Single doses up to 200 mg have been given with acceptable acute tolerability. Higher than recommended repeat doses are associated with increased myelosuppression including leukopenia, anemia and thrombocytopenia. Appropriate supportive treatment should be given.

Hemodialysis is not expected to enhance the elimination of ruxolitinib.

11. DESCRIPTION

Ruxolitinib phosphate is a kinase inhibitor with the chemical name (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphate and a molecular weight of 404.36. Ruxolitinib phosphate has the following structural formula:

Ruxolitinib phosphate is a white to off-white to light pink powder and is soluble in aqueous buffers across a pH range of 1 to 8.

Jakafi (ruxolitinib) Tablets are for oral administration. Each tablet contains ruxolitinib phosphate equivalent to 5 mg, 10 mg, 15 mg, 20 mg and 25 mg of ruxolitinib free base together with microcrystalline cellulose, lactose monohydrate, magnesium stearate, colloidal silicon dioxide, sodium starch glycolate, povidone and hydroxypropyl cellulose.

12. CLINICAL PHARMACOLOGY

12.1 Mechanism of Action

Ruxolitinib, a kinase inhibitor, inhibits Janus Associated Kinases (JAKs) JAK1 and JAK2 which mediate the signaling of a number of cytokines and growth factors that are important for hematopoiesis and immune function. JAK signaling involves recruitment of STATs (signal transducers and activators of transcription) to cytokine receptors, activation and subsequent localization of STATs to the nucleus leading to modulation of gene expression.

Myelofibrosis (MF) is a myeloproliferative neoplasm (MPN) known to be associated with dysregulated JAK1 and JAK2 signaling. In a mouse model of JAK2V617F-positive MPN, oral administration of ruxolitinib prevented splenomegaly, preferentially decreased JAK2V617F mutant cells in the spleen and decreased circulating inflammatory cytokines (eg, TNF-α, IL-6).

12.2 Pharmacodynamics

Ruxolitinib inhibits cytokine induced STAT3 phosphorylation in whole blood from healthy subjects and MF patients. Jakafi administration resulted in maximal inhibition of STAT3 phosphorylation 2 hours after dosing which returned to near baseline by 10 hours in both healthy subjects and myelofibrosis patients.

12.3 Pharmacokinetics

Absorption

In clinical studies, ruxolitinib is rapidly absorbed after oral Jakafi administration with maximal plasma concentration (Cmax) achieved within 1 to 2 hours post-dose. Based on a mass balance study in humans, oral absorption of ruxolitinib was estimated to be at least 95%. Mean ruxolitinib Cmax and total exposure (AUC) increased proportionally over a single dose range of 5 to 200 mg. There were no clinically relevant changes in the pharmacokinetics of ruxolitinib upon administration of Jakafi with a high-fat meal, with the mean Cmax moderately decreased (24%) and the mean AUC nearly unchanged (4% increase).

Distribution

The apparent volume of distribution of ruxolitinib at steady-state is 53 to 65 L in myelofibrosis patients. Binding to plasma proteins in vitro is approximately 97%, mostly to albumin.

Metabolism

In vitro studies suggest that CYP3A4 is the major enzyme responsible for metabolism of ruxolitinib. Ruxolitinib is the predominant entity in humans representing approximately 60% of the drug-related material in circulation. Two major and active metabolites were identified in plasma of healthy subjects representing 25% and 11% of parent AUC. These two metabolites have one-fifth and one-half of ruxolitinib's pharmacological activity, respectively. The sum total of all active metabolites contributes 18% of the overall pharmacodynamics of ruxolitinib.

Elimination

Following a single oral dose of [14C]-labeled ruxolitinib in healthy adult subjects, elimination was predominately through metabolism with 74% of radioactivity excreted in urine and 22% excretion via feces. Unchanged drug accounted for less than 1% of the excreted total radioactivity. The mean elimination half-life of ruxolitinib is approximately 3 hours and the mean half-life of ruxolitinib + metabolites is approximately 5.8 hours.

Effects of Age, Gender, or Race

In healthy subjects, no significant differences in ruxolitinib pharmacokinetics were observed with regard to gender and race. In a population pharmacokinetic evaluation in myelofibrosis patients, no relationship was apparent between oral clearance and patient age or race, and in women, clearance was 17.7 L/h and in men, 22.1 L/h with 39% inter-subject variability.

Drug Interactions

In vitro, ruxolitinib and its M18 metabolite are not inhibitors of CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6 or CYP3A4. Ruxolitinib is not an inducer of CYP1A2, CYP2B6 or CYP3A4 at clinically relevant concentrations.

In vitro, ruxolitinib and its M18 metabolite are not inhibitors of the P-gp, BCRP, OATP1B1, OATP1B3, OCT1, OCT2, OAT1 or OAT3 transport systems at clinically relevant concentrations. Ruxolitinib is not a substrate for the P-gp transporter.

12.4 Thorough QT Study

The effect of single dose ruxolitinib 25 mg and 200 mg on QTc interval was evaluated in a randomized, placebo-, and active-controlled (moxifloxacin 400 mg) four-period crossover thorough QT study in 47 healthy subjects. In a study with demonstrated ability to detect small effects, the upper bound of the one-sided 95% confidence interval for the largest placebo adjusted, baseline-corrected QTc based on Fridericia correction method (QTcF) was below 10 ms, the threshold for regulatory concern. The dose of 200 mg is adequate to represent the high exposure clinical scenario.

13. NONCLINICAL TOXICOLOGY

13.1. Carcinogenesis, Mutagenesis, Impairment of Fertility

Ruxolitinib was not carcinogenic in the 6 month Tg.rasH2 transgenic mouse model. A 2-year carcinogenicity study in the rat is ongoing.

Ruxolitinib was not mutagenic in a bacterial mutagenicity assay (Ames test) or clastogenic in in vitro chromosomal aberration assay (cultured human peripheral blood lymphocytes) or in vivo in a rat bone marrow micronucleus assay.

In a fertility study, ruxolitinib was administered to male rats prior to and throughout mating and to female rats prior to mating and up to the implantation day (gestation day 7). Ruxolitinib had no effect on fertility or reproductive function in male or female rats at doses of 10, 30 or 60 mg/kg/day. However, in female rats doses of greater than or equal to 30 mg/kg/day resulted in increased post-implantation loss. The exposure (AUC) at the dose of 30 mg/kg/day is approximately 34% the clinical exposure at the maximum recommended dose of 25 mg twice daily.

14. CLINICAL STUDIES

Two randomized Phase 3 studies (Studies 1 and 2) were conducted in patients with myelofibrosis (either primary myelofibrosis, post-polycythemia vera myelofibrosis or post-essential thrombocythemia-myelofibrosis). In both studies, patients had palpable splenomegaly at least 5 cm below the costal margin and risk category of intermediate 2 (2 prognostic factors) or high risk (3 or more prognostic factors) based on the International Working Group Consensus Criteria (IWG).

The starting dose of Jakafi was based on platelet count. Patients with a platelet count between 100 and 200 X 109/L were started on Jakafi 15 mg twice daily and patients with a platelet count greater than 200 X 109/L were started on Jakafi 20 mg twice daily. Doses were then individualized based upon tolerability and efficacy with maximum doses of 20 mg twice daily for patients with platelet counts between 100 to less than or equal to 125 X 109/L, of 10 mg twice daily for patients with platelet counts between 75 to less than or equal to 100 X 109/L, and of 5 mg twice daily for patients with platelet counts between 50 to less than or equal to 75 X 109/L.

Study 1

Study 1 was a double-blind, randomized, placebo-controlled study in 309 patients who were refractory to or were not candidates for available therapy. The median age was 68 years (range 40 to 91 years) with 61% of patients older than 65 years and 54% were male. Fifty percent (50%) of patients had primary myelofibrosis, 31% had post-polycythemia vera myelofibrosis and 18% had post-essential thrombocythemia myelofibrosis. Twenty-one percent (21%) of patients had red blood cell transfusions within 8 weeks of enrollment in the study. The median hemoglobin count was 10.5 g/dL and the median platelet count was 251 X 109/L. Patients had a median palpable spleen length of 16 cm below the costal margin, with 81% having a spleen length 10 cm or greater below the costal margin. Patients had a median spleen volume as measured by magnetic resonance imaging (MRI) or computed tomography (CT) of 2595 cm3 (range 478 cm3 to 8881 cm3). (The upper limit of normal is approximately 300 cm3).

Patients were dosed with Jakafi or matching placebo. The primary efficacy endpoint was the proportion of patients achieving greater than or equal to a 35% reduction from baseline in spleen volume at Week 24 as measured by MRI or CT.

Secondary endpoints included duration of a 35% or greater reduction in spleen volume and proportion of patients with a 50% or greater reduction in Total Symptom Score from baseline to Week 24 as measured by the modified Myelofibrosis Symptom Assessment Form (MFSAF) v2.0 diary.

Study 2

Study 2 was an open-label, randomized study in 219 patients. Patients were randomized 2:1 to Jakafi versus best available therapy. Best available therapy was selected by the investigator on a patient-by-patient basis. In the best available therapy arm, the medications received by more than 10% of patients were hydroxyurea (47%) and glucocorticoids (16%). The median age was 66 years (range 35 to 85 years) with 52% of patients older than 65 years and 57% were male. Fifty-three percent (53%) of patients had primary myelofibrosis, 31% had post-polycythemia vera myelofibrosis and 16% had post-essential thrombocythemia myelofibrosis. Twenty-one percent (21%) of patients had red blood cell transfusions within 8 weeks of enrollment in the study. The median hemoglobin count was 10.4 g/dL and the median platelet count was 236 X 109/L. Patients had a median palpable spleen length of 15 cm below the costal margin, with 70% having a spleen length 10 cm or greater below the costal margin. Patients had a median spleen volume as measured by MRI or CT of 2381 cm3 (range 451 cm3 to 7765 cm3).

The primary efficacy endpoint was the proportion of patients achieving 35% or greater reduction from baseline in spleen volume at Week 48 as measured by MRI or CT.

A secondary endpoint in Study 2 was the proportion of patients achieving a 35% or greater reduction of spleen volume as measured by MRI or CT from baseline to Week 24.

Study 1 and 2 Efficacy Results

Efficacy analyses of the primary endpoint in Studies 1 and 2 are presented in Table 6 below. A significantly larger proportion of patients in the Jakafi group achieved a 35% or greater reduction in spleen volume from baseline in both studies compared to placebo in Study 1 and best available therapy in Study 2. A similar proportion of patients in the Jakafi group achieved a 50% or greater reduction in palpable spleen length.

Table 6: Percent of Patients with 35% or Greater Reduction from Baseline in Spleen Volume at Week 24 in Study 1 and at Week 48 in Study 2 (Intent to Treat)
  Study 1 Study 2
  Jakafi
(N=155)
Placebo
(N=154)
Jakafi
(N=146)
Best Available
Therapy
(N=73)
Time Points Week 24 Week 48
Number (%) of Patients with Spleen Volume Reduction by 35% or More 65 (41.9) 1 (0.7) 41 (28.5) 0
P-value < 0.0001 < 0.0001

Figure 1 shows the percent change from baseline in spleen volume for each patient at Week 24 (Jakafi N=139, placebo N=106) or the last evaluation prior to Week 24 for patients who did not complete 24 weeks of randomized treatment (Jakafi N=16, placebo N=47). One (1) patient (placebo) with a missing baseline spleen volume is not included.

In Study 1, myelofibrosis symptoms were a secondary endpoint and were measured using the modified Myelofibrosis Symptom Assessment Form (MFSAF) v2.0 diary. The modified MFSAF is a daily diary capturing the core symptoms of myelofibrosis (abdominal discomfort, pain under left ribs, night sweats, itching, bone/muscle pain and early satiety). Symptom scores ranged from 0 to 10 with 0 representing symptoms "absent" and 10 representing "worst imaginable" symptoms. These scores were added to create the daily total score, which has a maximum of 60.

Table 7 presents assessments of Total Symptom Score from baseline to Week 24 in Study 1 including the proportion of patients with at least a 50% reduction (ie, improvement in symptoms). At baseline, the mean Total Symptom Score was 18.0 in the Jakafi group and 16.5 in the placebo group. A higher proportion of patients in the Jakafi group had a 50% or greater reduction in Total Symptom Score than in the placebo group, with a median time to response of less than 4 weeks.

Table 7: Improvement in Total Symptom Score
  Jakafi
(N=148)
Placebo
(N=152)
Number (%) of Patients with 50% or Greater Reduction in Total Symptom Score by Week 24 68 (45.9) 8 (5.3)
P-value < 0.0001

Figure 2 shows the percent change from baseline in Total Symptom Score for each patient at Week 24 (Jakafi N=129, placebo N=103) or the last evaluation on randomized therapy prior to Week 24 for patients who did not complete 24 weeks of randomized treatment (Jakafi N=16, placebo N=42). Results are excluded for 5 patients with a baseline Total Symptom Score of zero, 8 patients with missing baseline and 6 patients with insufficient post-baseline data.

Figure 3 displays the proportion of patients with at least a 50% improvement in each of the individual symptoms that comprise the Total Symptom Score indicating that all 6 of the symptoms contributed to the higher Total Symptom Score response rate in the group treated with Jakafi.

16. HOW SUPPLIED/STORAGE AND HANDLING

Jakafi (ruxolitinib) Tablets are available as follows:

Jakafi Trade Presentations NDC Number Strength Description Tablets per Bottle
50881-005-60 5 mg Round tablet with "INCY" on one side and "5" on the other  60
50881-010-60 10 mg Round tablet with "INCY" on one side and "10" on the other 60
50881-015-60 15 mg Oval tablet with "INCY" on one side and "15" on the other 60
50881-020-60 20 mg Capsule shaped tablet with "INCY" on one side and "20" on the other 60
50881-025-60 25 mg Oval tablet with "INCY" on one side and "25" on the other 60
Store at room temperature 20°C to 25°C (68°F to 77°F); excursions permitted between 15°C and 30°C (59°F and 86°F) [see USP Controlled Room Temperature].
Jakafi(ruxolitinib)“磷酸鲁索替尼”在美获批首款治疗真性红细胞增多症
2014年12月4日,美国FDA批准鲁索替尼;商品名Jakafi)用于治疗真性红细胞增多症患者,这是一种慢性骨髓疾病。在美国鲁索替尼是获批用于这一疾病的首款药物。
当骨髓制造太多红细胞时会发生真性红细胞增多症,患者可能经历白细胞及血小板增多。血细胞过多可导致脾脏肿大、出血问题及皮肤附近静脉发生血栓(静脉炎)。此外,这种疾病使患者处于中风或心脏病发作风险增加的境地。
鲁索替尼的新适应症旨在治疗对羟基脲没有充分响应或不能耐受的真性红细胞增多症患者,羟基脲是另一款通常用来降低血液中红细胞及血小板数量的药物。鲁索替尼通过抑制参与调节血液及免疫功能的Janus相关激酶(JAK)1和2发挥作用。这款药物获批治疗真性红细胞增多症将帮助降低脾肿大的发生及静脉切开术的需求,静脉切开术是将体内过多血液移出体外的一种手术。
“鲁索替尼获批用于真性红细胞增多症凸显了开发药物以与我们日益增长的疾病机理知识相匹配的重要性,,”FDA药物评价与研究中心血液及肿瘤产品办公室主任、医学博士Pazdur称。“用来评价鲁索替尼的试验证实减少脾脏体积及静脉切开术来控制这一疾病的临床意义。”
鲁索替尼治疗真性红细胞增多症的安全性及有效性在一项由222名受试者参与的试验中得到评价,这些受试者患有该疾病至少有24周时间,他们对羟基脲已没有充分响应或不能耐受,实施过静脉切开术及显示脾肿大。受试者被随机配给鲁索替尼或最好的现有可供使用治疗药物。
这项研究旨在检测在治疗第8周及继续治疗完成32周时对静脉切开术的需求,及在第32周时脾脏体积减小35%的情况。结果显示,21%的鲁索替尼治疗受试者经历了静脉切开术需求减少及脾脏体积减小,相比之下,接受现有最好治疗的受试者只有1%的患者达到这一指标。
在真性红细胞增多症受试者中,最常见的与使用鲁索替尼相关的副作用有低红细胞计数 (贫血)及低血小板计数(血小板减少症)。最常见的非血液相关副作用有头晕、便秘和带状疱疹。
FDA对鲁索替尼用于真性红细胞增多症的审评是通过优先审评程序完成的,因为在其申请资料被提交时,这款药物证实与现有治疗药物相比,对一种严重疾病治疗的安全性及有效性有明显的改善。优先审评可为一款药物的审评提供一个加快的审评过程。鲁索替尼因旨在治疗一种罕见疾病,其还获得孤儿药资格。
2011年,FDA批准鲁索替尼用于患有另一种骨髓疾病-中或高风险骨髓纤维化患者的治疗,包括原发性骨髓纤维化、真性红细胞增多性骨髓纤维化及原发性血小板增多性骨髓纤维化。鲁索替尼由特拉华州威尔明顿的Incyte公司上市销售。

责任编辑:admin


相关文章
威罗菲尼片ZELBORAF(VEMURAFENIB Tablets)
Zydelig(idelalisib filmcoated tablets)
Sutent(Sunitinib Malate Capsules)
索拉非尼司特片|NEXAVAR(Sorafenib Tosilate tablets)
磷酸鲁索替尼片|Jakavi(ruxilitinib Tablets)
卡博替尼混合胶囊|Cometriq(cabozantinib capsules)
卡博替尼[混合]胶囊|COMETRIQ(CABOZANTINIB S-MALATE)
尼达尼布胶囊|Ofev(nintedanib)capsules
阿法替尼片|Gilotrif(Afatinib filmcoated tablets)
舒尼替尼胶囊|Sutent(sunitinib malate cap)
 

最新文章

更多

· LYMPHOSEEK KIT(TECHNE...
· ADEMPAS(RIOCIGUAT)TABL...
· RIXUBIS(Coagulation Fa...
· GLIADEL Wafer(Polifepr...
· 尼达尼布软胶囊|Ofev(ni...
· 碘克沙醇注射剂VISIPAQU...
· SAVAYSA(EDOXABAN TOSYL...
· VIEKIRA PAK(DASABUVIR...
· TECHNIVIE(ombitasvir, ...
· ODOMZO(sonidegib 原名L...

推荐文章

更多

· LYMPHOSEEK KIT(TECHNE...
· ADEMPAS(RIOCIGUAT)TABL...
· RIXUBIS(Coagulation Fa...
· GLIADEL Wafer(Polifepr...
· 尼达尼布软胶囊|Ofev(ni...
· 碘克沙醇注射剂VISIPAQU...
· SAVAYSA(EDOXABAN TOSYL...
· VIEKIRA PAK(DASABUVIR...
· TECHNIVIE(ombitasvir, ...
· ODOMZO(sonidegib 原名L...

热点文章

更多

· GLIADEL Wafer(Polifepr...
· LYMPHOSEEK KIT(TECHNE...
· RIXUBIS(Coagulation Fa...
· ADEMPAS(RIOCIGUAT)TABL...