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 >> 新药推荐 >> 来那度胺胶囊|REVLIMID(lenalidomide capsules)

来那度胺胶囊|REVLIMID(lenalidomide capsules)

2015-04-24 01:50:24  作者:新特药房  来源:互联网  浏览次数:192  文字大小:【】【】【
简介: 英文药名:REVLIMID(lenalidomide capsules) 中文药名: 瑞复美(来那度胺胶囊) 生产厂家:细胞基因公司药品介绍用途: 抗癌药物 适应症 来拉度胺主要用于具有5q缺失细胞遗传学异常的骨髓增生异常 ...

英文药名:REVLIMID(lenalidomide capsules)

中文药名: 瑞复美(来那度胺胶囊)

生产厂家:细胞基因公司
药品介绍
用途: 抗癌药物
适应症
来拉度胺主要用于具有5q缺失细胞遗传学异常的骨髓增生异常综合征所致的输血依赖性贫血患者的治疗。此外,到2006年1月为止,还有至少17项旨在评价来拉度胺其他适应症的临床试验在世界各地展开。
【药理及药代动力学】
来拉度胺的化学结构与沙利度胺相似。尽管来拉度胺的确切作用机制目前尚不十分清楚,但人们已经知道来拉度胺具有免疫调节及抗新血管生成作用。经口服给药后,来拉度胺迅速吸收入体内。体外实验显示,来拉度胺血浆蛋白结合率约为30%。约有2/3的来拉度胺以原形随尿液排泄,其消除半衰期约为3小时。
【注意事项】
在来拉度胺说明书中列出了一些比较重要的警告信息,包括①可能导致人体出生缺陷;②可能存在血液学方面的毒性反应(如中性粒细胞减少症和血小板减少症);③可能引起深部静脉血栓形成和肺动脉栓塞。由于来拉度胺的化学结构与沙利度胺相似,而后者又是人们所熟悉的一种致畸药物,因此妊娠女性服用来拉度胺后也有可能导致胎儿出生缺陷,甚至死亡。
用途
2005年9月,FDA肿瘤药物顾问委员会建议以来那度胺用于治疗输液依赖型贫血症。输液依赖型贫血症是由于5q染色体异常相关的骨髓增生异常综合征所引起的。10月,欧洲药品管理局接受了来那度胺的上市申请。同时,该品在欧洲获得了治疗骨髓增生异常综合征的罕用药物和治疗多发性骨髓瘤的罕用药物的地位。
celgene公司计划在2006年第一季度分别向美国FDA和欧洲药品管理局提交本品用于治疗复发型难治性多发性骨髓瘤的补充新药申请。
来那度胺对细胞内多种生物途径都有影响。来那度胺为口服制剂,且毒性较低
不良反应
来拉度胺最常见的不良反应为血小板减少症(61.5%)和中性粒细胞减少症(58.8%)。其他较常见的不良反应还包括腹泻、瘙痒、皮疹、疲劳、便秘、恶心、鼻咽炎、关节痛、发热、背痛、外周性水肿、咳嗽、头昏、头痛、肌肉痛性痉挛、呼吸困难和咽炎。
用药提示
医师每次向患者处方来拉度胺时都应为患者提供用药指导,医师或药师应借此机会向患者解释来拉度胺可能存在的风险。准备用药的患者最好同时采取两种不同的有效避孕措施来避孕。准备服用来拉度胺的女性患者应在医生的指导下签署知情同意书,以表明自己已经知道在服用来拉度胺时避孕的重要性。此外,所有有妊娠可能的女性患者在用药前还需进行两次妊娠测试(一次安排在给药前10~14天,另一次安排在给药前24小时之内),只有两次妊娠测试均为阴性结果者才能正式服用来拉度胺。


Revlimid 10mg(lenalidomide)
1. Name of the medicinal product
Revlimid 10 mg hard capsules
2. Qualitative and quantitative composition
Each capsule contains 10 mg of lenalidomide.
Excipient(s) with known effect:
Each capsule contains 294 mg of lactose, anhydrous.
For the full list of excipients, see section 6.1.
3. Pharmaceutical form
Hard capsule.
Blue-green/pale yellow capsules, size 0, 21.7 mm, marked “REV 10 mg”.
4. Clinical particulars
4.1 Therapeutic indications
Multiple myeloma
Revlimid is indicated for the treatment of adult patients with previously untreated multiple myeloma who are not eligible for transplant (see section 4.2).
Revlimid in combination with dexamethasone is indicated for the treatment of multiple myeloma in adult patients who have received at least one prior therapy.
Myelodysplastic syndromes
Revlimid is indicated for the treatment of patients with transfusion-dependent anaemia due to low- or intermediate-1-risk myelodysplastic syndromes associated with an isolated deletion 5q cytogenetic abnormality when other therapeutic options are insufficient or inadequate.
4.2 Posology and method of administration
Revlimid treatment should be supervised by a physician experienced in the use of anti-cancer therapies (see section 4.4, karyotype).
Posology
Newly diagnosed multiple myeloma
Lenalidomide in combination with dexamethasone until disease progression in patients who are not eligible for transplant
Lenalidomide treatment must not be started if the Absolute Neutrophil Counts (ANC) is < 1.0 x 109/L, and/or platelet counts are < 50 x 109/L.
Recommended dose
The recommended starting dose of lenalidomide is 25 mg orally once daily on days 1-21 of repeated 28-day cycles. The recommended dose of dexamethasone is 40 mg orally once daily on days 1, 8, 15 and 22 of repeated 28-day cycles. Patients may continue lenalidomide and dexamethasone therapy until disease progression or intolerance.
Dosing is continued or modified based upon clinical and laboratory findings (see section 4.4). For patients ≥75 years of age, the starting dose of dexamethasone is 20 mg/day on Days 1, 8, 15 and 22 of each 28-day treatment cycle. The recommended dose of lenalidomide for patients suffering from moderate renal impairment is 10 mg once daily.
Recommended dose adjustments during treatment and restart of treatment:
Dose adjustments, as summarised below, are recommended to manage grade 3 or 4 thrombocytopenia, neutropenia, or other grade 3 or 4 toxicity judged to be related to lenalidomide.
• Dose reduction steps

Lenalidomide

Dexamethasone

Starting dose

25 mg

40 mg

Dose level -1

20 mg

20 mg

Dose level -2

15 mg

12 mg

Dose level -3

10 mg

8 mg

Dose level- 4

5 mg

4 mg

Dose level -5

2.5 mg

NA

• Thrombocytopenia

When platelets

Recommended course

Fall to < 25 x 109/L

Stop lenalidomide dosing for remainder of cycleª

Return to ≥ 50 x 109/L

Decrease by one dose level when dosing resumed at next cycle

ª If Dose Limiting Toxicity occurs on > Day15 of a cycle, lenalidomide dosing will be interrupted for at least the remainder of the current 28-day cycle.
• Neutropenia

When neutrophils

Recommended course

First fall to < 0.5 x 109/L

Interrupt lenalidomide treatment

Return to ≥ 1 x 109/L when neutropenia is the only observed toxicity

Resume lenalidomide at Starting dose once daily

Return to ≥ 0.5 x 109/L when dose-dependent haematological toxicities other than neutropenia are observed

Resume lenalidomide at Dose level -1 once daily

For each subsequent drop below < 0.5 x 109/L

Interrupt lenalidomide treatment

Return to ≥ 0.5 x 109/L

Resume lenalidomide at next lower dose level once daily.

In case of neutropenia, the use of growth factors in patient management should be considered.
If the dose of lenalidomide was reduced for a hematologic DLT, the dose of lenalidomide may be re-introduced to the next higher dose level (up to the starting dose) at the discretion of the treating physician if continued lenalidomide / dexamethasone therapy resulted in improved bone marrow function (no DLT for at least 2 consecutive cycles and an ANC ≥1,500/µL with a platelet count ≥ 100,000/µL at the beginning of a new cycle at the current dose level).
Lenalidomide in combination with melphalan and prednisone followed by maintenance monotherapy in patients who are not eligible for transplant
Lenalidomide treatment must not be started if the ANC is < 1.5 x 109/L, and/or platelet counts are < 75 x 109/L.
Recommended dose
The recommended starting dose is lenalidomide 10 mg/day orally on days 1-21 of repeated 28-day cycles for up to 9 cycles, melphalan 0.18 mg/kg orally on days 1-4 of repeated 28 day cycles, prednisone 2 mg/kg orally on days 1-4 of repeated 28-day cycles. Patients who complete 9 cycles or who are unable to complete the combination therapy due to intolerance are treated with lenalidomide alone,10 mg/day orally on days 1-21 of repeated 28-day cycles given until disease progression. Dosing is continued or modified based upon clinical and laboratory findings (see section 4.4).
Recommended dose adjustments during treatment and restart of treatment:
Dose adjustments, as summarised below, are recommended to manage grade 3 or 4 thrombocytopenia, neutropenia, or other grade 3 or 4 toxicity judged to be related to lenalidomide.
• Dose reduction steps

Lenalidomide

Melphalan

Prednisone

Starting dose

10 mgª

0.18 mg/kg

2 mg/kg

Dose level -1

7.5 mg

0.14 mg/kg

1 mg/kg

Dose level -2

5 mg

0.10 mg/kg

0.5 mg/kg

Dose level -3

2.5 mg

NA

0.25 mg/kg

ª If neutropenia is the only toxicity at any dose level, add granulocyte colony stimulating factor (G-CSF) and maintain the dose level of lenalidomide
• Thrombocytopenia

When platelets

Recommended course

First fall to < 25 x 109/L

Interrupt lenalidomide treatment

Return to ≥ 25 x 109/L

Resume lenalidomide and melphalan at Dose level -1

For each subsequent drop below 30 x 109/L

Interrupt lenalidomide treatment

Return to ≥ 30 x 109/L

Resume lenalidomide at next lower dose level (Dose level -2 or -3) once daily.

• Neutropenia

When neutrophils

Recommended course

First fall to < 0.5 x 109/Lª

Interrupt lenalidomide treatment

Return to ≥ 0.5 x 109/L when neutropenia is the only observed toxicity

Resume lenalidomide at Starting dose once daily

Return to ≥ 0.5 x 109/L when dose-dependent haematological toxicities other than neutropenia are observed

Resume lenalidomide at Dose level -1 once daily

For each subsequent drop below < 0.5 x 109/L

Interrupt lenalidomide treatment

Return to ≥ 0.5 x 109/L

Resume lenalidomide at next lower dose level once daily.

ªIf the subject has not been receiving G-CSF therapy, initiate G-CSF therapy. On Day 1 of next cycle, continue G-CSF as needed and maintain dose of melphalan if neutropenia was the only DLT. Otherwise, decrease by one dose level at start of next cycle.
In case of neutropenia, the use of growth factors in patient management should be considered.
Multiple myeloma with at least one prior therapy
Recommended dose
The recommended starting dose of lenalidomide is 25 mg orally once daily on days 1-21 of repeated 28-day cycles. The recommended dose of dexamethasone is 40 mg orally once daily on days 1-4, 9-12, and 17-20 of each 28-day cycle for the first 4 cycles of therapy and then 40 mg once daily on days 1-4 every 28 days. Dosing is continued or modified based upon clinical and laboratory findings (see section 4.4). Prescribing physicians should carefully evaluate which dose of dexamethasone to use, taking into account the condition and disease status of the patient.
Lenalidomide treatment must not be started if the ANC < 1.0 x 109/L, and/or platelet counts < 75 x 109/L or, dependent on bone marrow infiltration by plasma cells, platelet counts < 30 x 109/L.
Recommended dose adjustments during treatment and restart of treatment
Dose adjustments, as summarised below, are recommended to manage grade 3 or 4 neutropenia or thrombocytopenia, or other grade 3 or 4 toxicity judged to be related to lenalidomide.
• Dose reduction steps

Starting dose

25 mg

Dose level -1

15 mg

Dose level -2

10 mg

Dose level -3

5 mg

• Thrombocytopenia

When platelets

Recommended course

First fall to < 30 x 109/L

Interrupt lenalidomide treatment

Return to ≥ 30 x 109/L

Resume lenalidomide at Dose level -1

For each subsequent drop below 30 x 109/L

Interrupt lenalidomide treatment

Return to ≥ 30 x 109/L

Resume lenalidomide at next lower dose level (Dose level -2 or -3) once daily. Do not dose below 5 mg once daily.

• Neutropenia

When neutrophils

Recommended course

First fall to < 0.5 x 109/L

Interrupt lenalidomide treatment

Return to ≥ 0.5 x 109/L when neutropenia is the only observed toxicity

Resume lenalidomide at Starting dose once daily

Return to ≥ 0.5 x 109/L when dose-dependent haematological toxicities other than neutropenia are observed

Resume lenalidomide at Dose level -1 once daily

For each subsequent drop below < 0.5 x 109/L

Interrupt lenalidomide treatment

Return to ≥ 0.5 x 109/L

Resume lenalidomide at next lower dose level (Dose level -1, -2 or -3) once daily. Do not dose below 5 mg once daily.

In case of neutropenia, the physician should consider the use of growth factors in patient management.
Myelodysplastic syndromes
Lenalidomide treatment must not be started if the ANC < 0.5 x 109/L and/or platelet counts < 25 x 109/L.
Recommended dose
The recommended starting dose of lenalidomide is 10 mg orally once daily on days 1-21 of repeated 28-day cycles. Dosing is continued or modified based upon clinical and laboratory findings (see section 4.4).
Recommended dose adjustments during treatment and restart of treatment
Dose adjustments, as summarized below, are recommended to manage grade 3 or 4 neutropenia or thrombocytopenia, or other grade 3 or 4 toxicity judged to be related to lenalidomide.
• Dose reduction steps

Starting dose

10 mg once daily on days 1-21 every 28 days

Dose level -1

5.0 mg once daily on days 1-28 every 28 days

Dose level -2

2.5 mg once daily on days 1-28 every 28 days

Dose level -3

2.5 mg every other day 1-28 every 28 days

For patients who are dosed initially at 10 mg and who experience thrombocytopenia or neutropenia:
• Thrombocytopenia

When platelets

Recommended course

Fall to < 25 x 109/L

Interrupt lenalidomide treatment

Return to ≥ 25 x 109/L - < 50 x 109/L on at least 2 occasions for ≥ 7 days or when the platelet count recovers to ≥ 50 x 109/L at any time

Resume lenalidomide at next lower dose level (Dose level -1, -2 or -3)

• Neutropenia

When neutrophils

Recommended course

Fall to < 0.5 x 109/L

Interrupt lenalidomide treatment

Return to ≥ 0.5 x 109/L

Resume lenalidomide at next lower dose level (Dose level -1, -2 or -3)

Discontinuation of lenalidomide
Patients without at least a minor erythroid response within 4 months of therapy initiation, demonstrated by at least a 50% reduction in transfusion requirements or, if not transfused, a 1g/dl rise in haemoglobin, should discontinue lenalidomide treatment.
All patients
For other grade 3 or 4 toxicities judged to be related to lenalidomide, treatment should be stopped and only restarted at next lower dose level when toxicity has resolved to ≤ grade 2 depending on the physician's discretion.
Lenalidomide interruption or discontinuation should be considered for grade 2 or 3 skin rash. Lenalidomide must be discontinued for angioedema, grade 4 rash, exfoliative or bullous rash, or if Stevens-Johnson syndrome (SJS) or toxic epidermal necrolysis (TEN) is suspected, and should not be resumed following discontinuation from these reactions.
Special populations
Paediatric population
Revlimid should not be used in children and adolescents from birth to less than 18 years because of safety concerns (see section 4.4).
Older people
Currently available pharmacokinetic data are described in section 5.2. Lenalidomide has been used in clinical trials in multiple myeloma patients up to 91 years of age and in myelodysplastic syndromes patients up to 95 years of age (see section 5.1).
In patients with newly diagnosed multiple myeloma aged 75 years and older who received lenalidomide, there was a higher incidence of serious adverse reactions and adverse reactions that led to treatment discontinuation (see section 4.4). Patients with newly diagnosed multiple myeloma aged 75 years and older should be carefully assessed before treatment is considered (see section 4.4).
• Newly diagnosed multiple myeloma
For patients older than 75 years of age treated with lenalidomide in combination with dexamethasone, the starting dose of dexamethasone is 20 mg/day on Days 1, 8, 15 and 22 of each 28-day treatment cycle.
No dose adjustment is proposed for patients older than 75 years who are treated with lenalidomide in combination with melphalan and prednisone.
In clinical trials of newly diagnosed multiple myeloma in transplant non eligible patients, lenalidomide combined therapy was less tolerated in patients older than 75 years of age compared to the younger population. These patients discontinued at a higher rate due to intolerance (Grade 3 or 4 adverse events and serious adverse events), when compared to patients < 75years (see section 4.4).
• Multiple myeloma with at least one prior therapy
The percentage of multiple myeloma patients aged 65 or over was not significantly different between the lenalidomide/dexamethasone and placebo/dexamethasone groups. No overall difference in safety or efficacy was observed between these patients and younger patients, but greater pre-disposition of older individuals cannot be ruled out.
For myelodysplastic syndromes patients treated with lenalidomide, no overall difference in safety and efficacy was observed between patients aged over 65 and younger patients.
Because elderly patients are more likely to have decreased renal function, care should be taken in dose selection and it would be prudent to monitor renal function.
Patients with renal impairment
Lenalidomide is substantially excreted by the kidney; patients with greater degrees of renal impairment can have impaired treatment tolerance (see section 4.4). Care should be taken in dose selection and monitoring of renal function is advised.
No dose adjustments are required for patients with mild renal impairment and multiple myeloma or myelodysplastic syndromes. The following dose adjustments are recommended at the start of therapy and throughout treatment for patients with moderate or severe impaired renal function or end stage renal disease.
There are no Phase III trial experiences with End Stage Renal Disease (ESRD) (CLcr < 30 mL/min, requiring dialysis).
• Multiple myeloma

Renal function (CLcr)

Dose adjustment

(Days 1 to 21 of repeated 28- day cycles)

Moderate renal impairment

(30 ≤ CLcr < 50 mL/min)

10 mg once daily1

Severe renal impairment

(CLcr < 30 mL/min, not requiring dialysis)

7.5 mg once daily2

15 mg every other day

End Stage Renal Disease (ESRD)

(CLcr < 30 mL/min, requiring dialysis)

5 mg once daily. On dialysis days, the dose should be administered following dialysis.

1 The dose may be escalated to 15 mg once daily after 2 cycles if patient is not responding to treatment and is tolerating the treatment.
2 In countries where the 7.5 mg capsule is available.
After initiation of lenalidomide therapy, subsequent lenalidomide dose modification in renally impaired patients should be based on individual patient treatment tolerance, as described above.
• Myelodysplastic syndromes

Renal function (CLcr)

Dose adjustment

Moderate renal impairment

(30 ≤ CLcr < 50 mL/min)

Starting dose

5 mg once daily

(days 1-21 of repeated 28-day cycles)

Dose level -1

2.5 mg once daily

(days 1-28 of repeated 28-day cycles)

Dose level -2

2.5 mg once every other day

(days 1-28 of repeated 28-day cycles)

Severe renal impairment

(CLcr < 30 mL/min, not requiring dialysis)

Starting dose

2.5 mg once daily

(days 1-21 of repeated 28-day cycles)

Dose level -1

2.5 mg every other day

(days 1-28 of repeated 28-day cycles)

Dose level -2

2.5 mg twice a week

(days 1-28 of repeated 28-day cycles)

End Stage Renal Disease (ESRD)

(CLcr < 30 mL/min, requiring dialysis)

On dialysis days, the dose should be administered following dialysis.

Starting dose

2.5 mg once daily

(days 1-21 of repeated 28-day cycles)

Dose level -1

2.5 mg every other day

(days 1-28 of repeated 28-day cycles)

Dose level -2

2.5 mg twice a week

(days 1-28 of repeated 28-day cycles)

Patients with hepatic impairment
Lenalidomide has not formally been studied in patients with impaired hepatic function and there are no specific dose recommendations.
Method of administration
Oral use.
Revlimid capsules should be taken at about the same time on the scheduled days. The capsules should not be opened, broken or chewed. The capsules should be swallowed whole, preferably with water, either with or without food. If less than 12 hours has elapsed since missing a dose, the patient can take the dose. If more than 12 hours has elapsed since missing a dose at the normal time, the patient should not take the dose, but take the next dose at the normal time on the following day.
4.3 Contraindications
• Hypersensitivity to the active substance or to any of the excipients, listed in section 6.1.
• Women who are pregnant.
• Women of childbearing potential unless all of the conditions of the Pregnancy Prevention Programme are met (see sections 4.4 and 4.6).
4.4 Special warnings and precautions for use
Pregnancy warning
Lenalidomide is structurally related to thalidomide. Thalidomide is a known human teratogenic active substance that causes severe life-threatening birth defects. Lenalidomide induced in monkeys malformations similar to those described with thalidomide (see sections 4.6 and 5.3). If lenalidomide is taken during pregnancy, a teratogenic effect of lenalidomide in humans is expected.
The conditions of the Pregnancy Prevention Programme must be fulfilled for all patients unless there is reliable evidence that the patient does not have childbearing potential.
Criteria for women of non-childbearing potential
A female patient or a female partner of a male patient is considered to have childbearing potential unless she meets at least one of the following criteria:
• Age ≥ 50 years and naturally amenorrhoeic for ≥ 1 year*
• Premature ovarian failure confirmed by a specialist gynaecologist
• Previous bilateral salpingo-oophorectomy, or hysterectomy
• XY genotype, Turner syndrome, uterine agenesis.
*Amenorrhoea following cancer therapy or during lactation does not rule out childbearing potential.
Counselling
For women of childbearing potential, lenalidomide is contraindicated unless all of the following are met:
• She understands the expected teratogenic risk to the unborn child
• She understands the need for effective contraception, without interruption, 4 weeks before starting treatment, throughout the entire duration of treatment, and 4 weeks after the end of treatment
• Even if a woman of childbearing potential has amenorrhea she must follow all the advice on effective contraception
• She should be capable of complying with effective contraceptive measures
• She is informed and understands the potential consequences of pregnancy and the need to rapidly consult if there is a risk of pregnancy
• She understands the need to commence contraceptive measures as soon as lenalidomide is dispensed following a negative pregnancy test
• She understands the need and accepts to undergo pregnancy testing every 4 weeks except in case of confirmed tubal sterilisation
• She acknowledges that she understands the hazards and necessary precautions associated with the use of lenalidomide.
For male patients taking lenalidomide, pharmacokinetic data has demonstrated that lenalidomide is present in human semen at extremely low levels during treatment and is undetectable in human semen 3 days after stopping the substance in the healthy subject (see section 5.2). As a precaution, all male patients taking lenalidomide must meet the following conditions:
• Understand the expected teratogenic risk if engaged in sexual activity with a pregnant woman or a woman of childbearing potential
• Understand the need for the use of a condom if engaged in sexual activity with a pregnant woman or a woman of childbearing potential not using effective contraception (even if the man has had a vasectomy), during treatment and for 1 week after dose interruptions and/or cessation of treatment.
• Understand that if his female partner becomes pregnant whilst he is taking Revlimid or shortly after he has stopped taking Revlimid, he should inform his treating physician immediately and that it is recommended to refer the female partner to a physician specialised or experienced in teratology for evaluation and advice.
The prescriber must ensure that for women of childbearing potential:
• The patient complies with the conditions of the Pregnancy Prevention Programme, including confirmation that she has an adequate level of understanding
• The patient has acknowledged the aforementioned conditions.
Contraception
Women of childbearing potential must use one effective method of contraception for 4 weeks before therapy, during therapy, and until 4 weeks after lenalidomide therapy and even in case of dose interruption unless the patient commits to absolute and continuous abstinence confirmed on a monthly basis. If not established on effective contraception, the patient must be referred to an appropriately trained health care professional for contraceptive advice in order that contraception can be initiated.
The following can be considered to be examples of suitable methods of contraception:
• Implant
• Levonorgestrel-releasing intrauterine system (IUS)
• Medroxyprogesterone acetate depot
• Tubal sterilisation
• Sexual intercourse with a vasectomised male partner only; vasectomy must be confirmed by two negative semen analyses
• Ovulation inhibitory progesterone-only pills (i.e. desogestrel)
Because of the increased risk of venous thromboembolism in patients with multiple myeloma taking lenalidomide in combination therapy, and to a lesser extent in patients with myelodysplastic syndromes taking lenalidomide monotherapy, combined oral contraceptive pills are not recommended (see also section 4.5). If a patient is currently using combined oral contraception the patient should switch to one of the effective methods listed above. The risk of venous thromboembolism continues for 4−6 weeks after discontinuing combined oral contraception. The efficacy of contraceptive steroids may be reduced during co-treatment with dexamethasone (see section 4.5).
Implants and levonorgestrel-releasing intrauterine systems are associated with an increased risk of infection at the time of insertion and irregular vaginal bleeding. Prophylactic antibiotics should be considered particularly in patients with neutropenia.
Copper-releasing intrauterine devices are generally not recommended due to the potential risks of infection at the time of insertion and menstrual blood loss which may compromise patients with neutropenia or thrombocytopenia.
Pregnancy testing
According to local practice, medically supervised pregnancy tests with a minimum sensitivity of 25 mIU/mL must be performed for women of childbearing potential as outlined below. This requirement includes women of childbearing potential who practice absolute and continuous abstinence. Ideally, pregnancy testing, issuing a prescription and dispensing should occur on the same day. Dispensing of lenalidomide to women of childbearing potential should occur within 7 days of the prescription.
Prior to starting treatment
A medically supervised pregnancy test should be performed during the consultation, when lenalidomide is prescribed, or in the 3 days prior to the visit to the prescriber once the patient had been using effective contraception for at least 4 weeks. The test should ensure the patient is not pregnant when she starts treatment with lenalidomide.
Follow-up and end of treatment
A medically supervised pregnancy test should be repeated every 4 weeks, including 4 weeks after the end of treatment, except in the case of confirmed tubal sterilisation. These pregnancy tests should be performed on the day of the prescribing visit or in the 3 days prior to the visit to the prescriber.
Men
Lenalidomide is present in human semen at extremely low levels during treatment and is undetectable in human semen 3 days after stopping the substance in the healthy subject (see section 5.2). As a precaution, and taking into account special populations with prolonged elimination time such as renal impairment, all male patients taking lenalidomide should use condoms throughout treatment duration, during dose interruption and for 1 week after cessation of treatment if their partner is pregnant or of childbearing potential and not using effective contraception (even if the man has had a vasectomy).
Additional precautions
Patients should be instructed never to give this medicinal product to another person and to return any unused capsules to their pharmacist at the end of treatment.
Patients should not donate blood during therapy or for 1 week following discontinuation of lenalidomide.
Educational materials, prescribing and dispensing restrictions
In order to assist patients in avoiding foetal exposure to lenalidomide, the Marketing Authorisation Holder will provide educational material to health care professionals to reinforce the warnings about the expected teratogenicity of lenalidomide, to provide advice on contraception before therapy is started, and to provide guidance on the need for pregnancy testing. The prescriber must inform male and female patients about the expected teratogenic risk and the strict pregnancy prevention measures as specified in the Pregnancy Prevention Programme and provide patients with appropriate patient educational brochure, patient card and/or equivalent tool in accordance to the national implemented patient card system. A national controlled distribution system has been implemented in collaboration with each National Competent Authority. The controlled distribution system includes the use of a patient card and/or equivalent tool for prescribing and/or dispensing controls, and the collecting of detailed data relating to the indication in order to monitor closely the off-label use within the national territory. Ideally, pregnancy testing, issuing a prescription and dispensing should occur on the same day. Dispensing of lenalidomide to women of childbearing potential should occur within 7 days of the prescription and following a medically supervised negative pregnancy test result.
Other special warnings and precautions for use
Cardiovascular disorders
Myocardial infarction
Myocardial infarction has been reported in patients receiving lenalidomide, particularly in those with known risk factors and within the first 12 months when used in combination with dexamethasone. Patients with known risk factors – including prior thrombosis – should be closely monitored, and action should be taken to try to minimize all modifiable risk factors (eg. smoking, hypertension, and hyperlipidaemia).
Venous and arterial thromboembolic events
In patients with multiple myeloma, the combination of lenalidomide with dexamethasone is associated with an increased risk of venous thromboembolism (predominantly deep vein thrombosis and pulmonary embolism) and arterial thromboembolism (predominantly myocardial infarction and cerebrovascular event). Venous thromboembolism was seen to a lesser extent with lenalidomide in combination with melphalan and prednisone in newly diagnosed multiple myeloma and with monotherapy in myelodysplastic syndromes. See sections 4.5 and 4.8.
In patients with myelodysplastic syndromes, treatment with lenalidomide monotherapy was also associated with a risk of venous thromboembolism (predominantly deep vein thrombosis and pulmonary embolism), but to a lesser extent than in patients with multiple myeloma – see sections 4.5 and 4.8.
Consequently, patients with known risk factors for thromboembolism – including prior thrombosis – should be closely monitored. Action should be taken to try to minimize all modifiable risk factors (e.g. smoking, hypertension, and hyperlipidaemia). Concomitant administration of erythropoietic agents or previous history of thromboembolic events may also increase thrombotic risk in these patients. Therefore, erythropoietic agents, or other agents that may increase the risk of thrombosis, such as hormone replacement therapy, should be used with caution in multiple myeloma patients receiving lenalidomide with dexamethasone. A haemoglobin concentration above 12 g/dl should lead to discontinuation of erythropoietic agents.
Patients and physicians are advised to be observant for the signs and symptoms of thromboembolism. Patients should be instructed to seek medical care if they develop symptoms such as shortness of breath, chest pain, arm or leg swelling. Prophylactic antithrombotic medicines should be recommended, especially in patients with additional thrombotic risk factors. The decision to take antithrombotic prophylactic measures should be made after careful assessment of an individual patient's underlying risk factors.
If the patient experiences any thromboembolic events, treatment must be discontinued and standard anticoagulation therapy started. Once the patient has been stabilised on the anticoagulation treatment and any complications of the thromboembolic event have been managed, the lenalidomide treatment may be restarted at the original dose dependent upon a benefit risk assessment. The patient should continue anticoagulation therapy during the course of lenalidomide treatment.
Neutropenia and thrombocytopenia
The major dose limiting toxicities of lenalidomide include neutropenia and thrombocytopenia. A complete blood cell count, including white blood cell count with differential count, platelet count, haemoglobin, and haematocrit should be performed at baseline, every week for the first 8 weeks of lenalidomide treatment and monthly thereafter to monitor for cytopenias. A dose reduction may be required (see section 4.2). In case of neutropenia, the physician should consider the use of growth factors in patient management. Patients should be advised to promptly report febrile episodes. Co-administration of lenalidomide with other myelosuppressive agents should be undertaken with caution.
• Newly diagnosed multiple myeloma in patients treated with lenalidomide in combination with low dose dexamethasone
Grade 4 neutropenia was observed in the lenalidomide arms in combination with low dose dexamethasone to a lesser extent than in the comparator arm (8.5% in the Rd [continuous treatment] and Rd18 [treatment for 18 four-week cycles] compared with 15% in the melphalan/prednisone/thalidomide arm, see section 4.8). Grade 4 febrile neutropenia episodes were consistent with the comparator arm (0.6 % in the Rd and Rd18 lenalidomide/dexamethasone-treated patients compared with 0.7% in the melphalan/prednisone/thalidomide arm, see section 4.8. Patients should be advised to promptly report febrile episodes and dose reductions may be required (see section 4.2).
Grade 3 or 4 thrombocytopenia was observed to a lesser extent in the Rd and Rd18 arms than in the comparator arm (8.1% vs 11.1%, respectively). Patients and physicians are advised to be observant for signs and symptoms of bleeding, including petechiae and epistaxes, especially in patients receiving concomitant medicinal products susceptible to induce bleeding (see section 4.8, Haemorrhagic disorders).
• Newly diagnosed multiple myeloma in patients treated with lenalidomide in combination with melphalan and prednisone
The combination of lenalidomide with melphalan and prednisone in in clinical trials of newly diagnosed multiple myeloma patients is associated with a higher incidence of grade 4 neutropenia (34.1% in melphalan, prednisone and lenalidomide arm followed by lenalidomide (MPR+R) and melphalan, prednisone and lenaldiomide followed by placebo (MPR+p) treated patients compared with 7.8% in MPp+p-treated patients; see section 4.8). Grade 4 febrile neutropenia episodes were observed infrequently (1.7% in MPR+R/MPR+p treated patients compared to 0.0 % in MPp+p treated patients; see section 4.8).
The combination of lenalidomide with melphalan and prednisone in clinical trials of multiple myeloma patients is associated with a higher incidence of grade 3 and grade 4 thrombocytopenia (40.4% in MPR+R/MPR+p treated patients, compared with 13.7% in MPp+p-treated patients; see section 4.8). Patients and physicians are advised to be observant for signs and symptoms of bleeding, including petechiae and epistaxes, especially in patients receiving concomitant medicinal products that increase susceptibility to bleeding (see section 4.8, Haemorrhagic disorders).
• Multiple myeloma with at least one prior therapy
The combination of lenalidomide with dexamethasone in multiple myeloma patients with at least one prior therapy is associated with a higher incidence of grade 4 neutropenia (5.1% in lenalidomide/dexamethasone-treated patients compared with 0.6% in placebo/dexamethasone-treated patients; see section 4.8). Grade 4 febrile neutropenia episodes were observed infrequently (0.6% in lenalidomide/dexamethasone-treated patients compared to 0.0% in placebo/dexamethasone treated patients; see section 4.8). Patients should be advised to promptly report febrile episodes. A dose reduction may be required (see section 4.2). In case of neutropenia, the physician should consider the use of growth factors in patient management.
The combination of lenalidomide with dexamethasone in multiple myeloma patients is associated with a higher incidence of grade 3 and grade 4 thrombocytopenia (9.9% and 1.4%, respectively, in lenalidomide/dexamethasone-treated patients compared to 2.3% and 0.0% in placebo/dexamethasone-treated patients; see section 4.8). Patients and physicians are advised to be observant for signs and symptoms of bleeding, including petechiae and epistaxes, especially in patients receiving concomitant medicinal products susceptible to induce bleeding (see section 4.8, Haemorrhagic disorders).
• Myelodysplastic syndromes
Lenalidomide treatment in myelodysplastic syndromes patients is associated with a higher incidence of grade 3 and 4 neutropenia and thrombocytopenia compared to patients on placebo (see section 4.8).
Infection with or without neutropenia
Patients with multiple myeloma are prone to develop infections including pneumonia. A higher rate of infections was observed with lenalidomide in combination with dexamethasone than with MPT. Grade ≥ 3 infections occurred within the context of neutropenia in less than one-third of the patients. Patients with known risk factors for infections should be closely monitored. All patients should be advised to seek medical attention promptly at the first sign of infection (eg, cough, fever, etc) thereby allowing for early management to reduce severity.
Renal impairment
Lenalidomide is substantially excreted by the kidney. Therefore care should be taken in dose selection and monitoring of renal function is advised in patients with renal impairment (see section 4.2).
Thyroid disorders
Cases of hypothyroidism and cases of hyperthyroidism have been reported. Optimal control of co-morbid conditions influencing thyroid function is recommended before start of treatment. Baseline and ongoing monitoring of thyroid function is recommended.
Peripheral neuropathy
Lenalidomide is structurally related to thalidomide, which is known to induce severe peripheral neuropathy. There was no increase in peripheral neuropathy observed with long term use of lenalidomide for the treatment of newly diagnosed multiple myeloma.
Tumour lysis syndrome
Because lenalidomide has anti-neoplastic activity the complications of tumour lysis syndrome may occur. The patients at risk of tumour lysis syndrome are those with high tumour burden prior to treatment. These patients should be monitored closely and appropriate precautions taken.
Allergic reactions
Cases of allergic reaction/hypersensitivity reactions have been reported in patients treated with lenalidomide (see section 4.8). Patients who had previous allergic reactions while treated with thalidomide should be monitored closely, as a possible cross-reaction between lenalidomide and thalidomide has been reported in the literature.
Severe skin reactions
SJS and TEN have been reported. Lenalidomide must be discontinued for exfoliative or bullous rash, or if SJS or TEN is suspected, and should not be resumed following discontinuation for these reactions. Interruption or discontinuation of lenalidomide should be considered for other forms of skin reaction depending on severity. Patients with a history of severe rash associated with thalidomide treatment should not receive lenalidomide.
Lactose intolerance
Revlimid capsules contain lactose. Patients with rare hereditary problems of galactose intolerance, the Lapp lactase deficiency or glucose-galactose malabsorption should not take this medicinal product.
Unused capsules
Patients should be advised never to give this medicinal product to another person and to return any unused capsules to their pharmacist at the end of the treatment.
Second primary malignancies
An increase of second primary malignancies (SPM) has been observed in clinical trials in previously treated myeloma patients receiving lenalidomide/dexamethasone (3.98 per 100 person-years) compared to controls (1.38 per 100 person-years). Non-invasive SPM comprise basal cell or squamous cell skin cancers. Most of the invasive SPMs were solid tumour malignancies.
In clinical trials of newly diagnosed multiple myeloma patients not eligible for transplant, a 4.9-fold increase in incidence rate of hematologic SPM (cases of AML, MDS) has been observed in patients receiving lenalidomide in combination with melphalan and prednisone until progression (1.75 per 100 person-years) compared with melphalan in combination with prednisone (0.36 per 100 person-years).
A 2.12-fold increase in incidence rate of solid tumor SPM has been observed in patients receiving lenalidomide (9 cycles) in combination with melphalan and prednisone (1.57 per 100 person-years) compared with melphalan in combination with prednisone (0.74 per 100 person-years).
In patients receiving lenalidomide in combination with dexamethasone until progression or for 18 months, the hematologic SPM incidence rate (0.16 per 100 person-years) was not increased as compared to thalidomide in combination with melphalan and prednisone (0.79 per 100 person-years).
A 1.3-fold increase in incidence rate of solid tumor SPM has been observed in patients receiving lenalidomide in combination with dexamethasone until progression or for 18 months (1.58 per 100 person-years) compared to thalidomide in combination with melphalan and prednisone (1.19 per 100 person-years).
In clinical trials of newly diagnosed multiple myeloma patients eligible for transplant, an increased incidence rate of hematologic SPM has been observed in patients receiving lenalidomide immediately following high-dose melphalan and Autologous Stem Cell Transplant (ASCT) compared with patients who received placebo (1.27 to 1.56 versus 0.46 to 0.53 per 100 person-years, respectively). Cases of B-cell malignancies (including Hodgkin's lymphoma) observed in the clinical trials were in patients who received lenalidomide in the post-ASCT setting.
The risk of occurrence of hematologic SPM must be taken into account before initiating treatment with Revlimid either in combination with melphalan or immediately following high-dose melphalan and ASCT. Physicians should carefully evaluate patients before and during treatment using standard cancer screening for occurrence of SPM and institute treatment as indicated.
Progression to acute myeloid leukaemia in low- and intermediate-1-risk MDS
• Karyotype
Baseline variables including complex cytogenetics are associated with progression to AML in subjects who are transfusion dependent and have a Del (5q) abnormality. In a combined analysis of two clinical trials of Revlimid in low- or intermediate-1-risk myelodysplastic syndromes, subjects who had a complex cytogenetics had the highest estimated 2-year cumulative risk of progression to AML (38.6%). The estimated 2-year rate of progression to AML in patients with an isolated Del (5q) abnormality was 13.8%, compared to 17.3% for patients with Del (5q) and one additional cytogenetic abnormality.
As a consequence, the benefit/risk ratio of Revlimid when MDS is associated with Del (5q) and complex cytogenetics is unknown.
• TP53 status
A TP53 mutation is present in 20 to 25% of lower-risk MDS Del 5q patients and is associated with a higher risk of progression to acute myeloid leukaemia (AML). In a post-hoc analysis of a clinical trial of Revlimid in low- or intermediate-1-risk myelodysplastic syndromes (MDS-004), the estimated 2-year rate of progression to AML was 27.5 % in patients with IHC-p53 positivity (1% cut-off level of strong nuclear staining, using immunohistochemical assessment of p53 protein as a surrogate for TP53 mutation status) and 3.6% in patients with IHC-p53 negativity (p=0.0038) (see section 4.8)
Hepatic disorders
Hepatic failure, including fatal cases, has been reported in patients treated with lenalidomide in combination therapy: acute hepatic failure, toxic hepatitis, cytolytic hepatitis, cholestatic hepatitis, and mixed cytolytic/cholestatic hepatitis have been reported. The mechanisms of severe drug-induced hepatotoxicity remain unknown although, in some cases, pre-existing viral liver disease, elevated baseline liver enzymes, and possibly treatment with antibiotics might be risk factors.
Abnormal liver function tests were commonly reported and were generally asymptomatic and reversible upon dosing interruption. Once parameters have returned to baseline, treatment at a lower dose may be considered.
Lenalidomide is excreted by the kidneys. It is important to dose adjust patients with renal impairment in order to avoid plasma levels which may increase the risk for higher haematological adverse reactions or hepatotoxicity. Monitoring of liver function is recommended, particularly when there is a history of or concurrent viral liver infection or when lenalidomide is combined with medicinal products known to be associated with liver dysfunction.
Newly diagnosed multiple myeloma patients
There was a higher rate of intolerance (grade 3 or 4 adverse events, serious adverse events, discontinuation) in patients with age > 75 years, ISS stage III, ECOG PS≤2 or CLcr<60 mL/min when lenalidomide is given in combination. Patients should be carefully assessed for their ability to tolerate lenalidomide in combination, with consideration to age, ISS stage III, ECOG PS≤2 or CLcr<60 mL/min (see section 4.2 and 4.8).
Cataract
Cataract has been reported with a higher frequency in patients receiving lenalidomide in combination with dexamethasone particularly when used for a prolonged time. Regular monitoring of visual ability is recommended.
4.5 Interaction with other medicinal products and other forms of interaction
Erythropoietic agents, or other agents that may increase the risk of thrombosis, such as hormone replacement therapy, should be used with caution in multiple myeloma patients receiving lenalidomide with dexamethasone (see sections 4.4 and 4.8).
Oral contraceptives
No interaction study has been performed with oral contraceptives. Lenalidomide is not an enzyme inducer. In an in vitro study with human hepatocytes, lenalidomide, at various concentrations tested did not induce CYP1A2, CYP2B6, CYP2C9, CYP2C19 and CYP3A4/5. Therefore, induction leading to reduced efficacy of medicinal products, including hormonal contraceptives, is not expected if lenalidomide is administered alone. However, dexamethasone is known to be a weak to moderate inducer of CYP3A4 and is likely to also affect other enzymes as well as transporters. It may not be excluded that the efficacy of oral contraceptives may be reduced during treatment. Effective measures to avoid pregnancy must be taken (see sections 4.4 and 4.6).
Warfarin
Co-administration of multiple doses of 10 mg of lenalidomide had no effect on the single dose pharmacokinetics of R- and S- warfarin. Co-administration of a single 25 mg dose of warfarin had no effect on the pharmacokinetics of lenalidomide. However, it is not known whether there is an interaction during clinical use (concomitant treatment with dexamethasone). Dexamethasone is a weak to moderate enzyme inducer and its effect on warfarin is unknown. Close monitoring of warfarin concentration is advised during the treatment.
Digoxin
Concomitant administration with lenalidomide 10 mg/day increased the plasma exposure of digoxin (0.5 mg, single dose) by 14% with a 90% CI (confidence interval) [0.52%-28.2%]. It is not known whether the effect will be different in the therapeutic situation (higher lenalidomide doses and concomitant treatment with dexamethasone). Therefore, monitoring of the digoxin concentration is advised during lenalidomide treatment.
Statins
There is an increased risk of rhabdomyolysis when statins are administered with lenalidomide, which may be simply additive. Enhanced clinical and laboratory monitoring is warranted notably during the first weeks of treatment.
Dexamethasone
Co-administration of single or multiple doses of dexamethasone (40 mg/ day) has no clinically relevant effect on the multiple dose pharmacokinetics of lenalidomide (25 mg/ day).
Interactions with P-glycoprotein (P-gp) inhibitors
In vitro, lenalidomide is a substrate of P-gp, but is not a P-gp inhibitor. Co-administration of multiple doses of the strong P-gp inhibitor quinidine (600 mg, twice daily) or the moderate P-gp inhibitor/substrate temsirolimus (25 mg) has no clinically relevant effect on the pharmacokinetics of lenalidomide (25 mg). Co-administration of lenalidomide does not alter the pharmacokinetics of temsirolimus.
4.6 Fertility, pregnancy and lactation
Women of childbearing potential / Contraception in males and females
Women of childbearing potential should use effective method of contraception. If pregnancy occurs in a woman treated with lenalidomide, treatment must be stopped and the patient should be referred to a physician specialised or experienced in teratology for evaluation and advice. If pregnancy occurs in a partner of a male patient taking lenalidomide, it is recommended to refer the female partner to a physician specialised or experienced in teratology for evaluation and advice.
Lenalidomide is present in human semen at extremely low levels during treatment and is undetectable in human semen 3 days after stopping the substance in the healthy subject (see section 5.2). As a precaution, and taking into account special populations with prolonged elimination time such as renal impairment, all male patients taking lenalidomide should use condoms throughout treatment duration, during dose interruption and for 1 week after cessation of treatment if their partner is pregnant or of childbearing potential and has no contraception.
Pregnancy
Lenalidomide is structurally related to thalidomide. Thalidomide is a known human teratogenic active substance that causes severe life-threatening birth defects.
Lenalidomide induced in monkeys malformations similar to those described with thalidomide (see section 5.3). Therefore, a teratogenic effect of lenalidomide is expected and lenalidomide is contraindicated during pregnancy (see section 4.3).
Breast-feeding
It is not known whether lenalidomide is excreted in human milk. Therefore breast-feeding should be discontinued during therapy with lenalidomide.
Fertility
A fertility study in rats with lenalidomide doses up to 500 mg/kg (approximately 200 to 500 times the human doses of 25 mg and 10 mg, respectively, based on body surface area) produced no adverse effects on fertility and no parental toxicity.
4.7 Effects on ability to drive and use machines
Lenalidomide has minor or moderate influence on the ability to drive and use machines. Fatigue, dizziness, somnolence and blurred vision have been reported with the use of lenalidomide. Therefore, caution is recommended when driving or operating machines.
4.8 Undesirable effects
Summary of the safety profile
Newly diagnosed multiple myeloma in patients treated with lenalidomide in combination with low dose dexamethasone
The serious adverse reactions observed more frequently (≥5%) with lenalidomide in combination with low dose dexamethasone (Rd and Rd18) than with melphalan, prednisone and thalidomide (MPT) were:
• Pneumonia (9.8%)
• Renal failure (including acute) (6.3%)
The adverse reactions observed more frequently with Rd or Rd18 than MPT were: diarrhoea (45.5%), fatigue (32.8%), back pain (32.0%), asthenia (28.2%), insomnia (27.6%), rash (24.3%), decreased appetite (23.1%), cough (22.7%), pyrexia (21.4%), and muscle spasms (20.5%).
Newly diagnosed multiple myeloma patients treated with lenalidomide in combination with melphalan and prednisone
The serious adverse reactions observed more frequently (≥5%) with melphalan prednisone, and lenalidomide followed by lenalidomide maintenance (MPR+R) or melphalan prednisone, and lenalidomide followed by placebo (MPR+p) than melphalan, prednisone and placebo followed by placebo (MPp+p) were:
• Febrile neutropenia (6.0%)
• Anaemia (5.3%)
The adverse reactions observed more frequently with MPR+R or MPR+ p than MPp+p were: neutropenia (83.3%), anaemia (70.7%), thrombocytopenia (70.0%), leukopenia (38.8%), constipation (34.0%), diarrhoea (33.3%), rash (28.9%), pyrexia (27.0%), peripheral oedema (25.0%), cough (24.0%), decreased appetite (23.7%), and asthenia (22.0%).
Multiple myeloma with at least one prior therapy
In two Phase III placebo-controlled studies, 353 patients with multiple myeloma were exposed to the lenalidomide/dexamethasone combination and 351 to the placebo/dexamethasone combination.
The most serious adverse reactions observed more frequently in lenalidomide/dexamethasone than placebo/dexamethasone combination were:
• Venous thromboembolism (deep vein thrombosis, pulmonary embolism) (see section 4.4)
• Grade 4 neutropenia (see section 4.4).
The observed adverse reactions which occurred more frequently with lenalidomide and dexamethasone than placebo and dexamethasone in pooled multiple myeloma clinical trials (MM-009 and MM-010) were fatigue (43.9%), neutropenia (42.2%), constipation (40.5%), diarrhoea (38.5%), muscle cramp (33.4%), anaemia (31.4%), thrombocytopenia (21.5%), and rash (21.2%).
Myelodysplastic syndromes
The overall safety profile of Revlimid in patients with myelodysplastic syndromes is based on data from a total of 286 patients from one Phase II study and one Phase III study (see section 5.1). In the Phase II, all 148 patients were on lenalidomide treatment. In the Phase III study, 69 patients were on lenalidomide 5 mg, 69 patients on lenalidomide 10 mg and 67 patients were on placebo during the double-blind phase of the study.
Most adverse reactions tended to occur during the first 16 weeks of therapy with lenalidomide.
Serious adverse reactions include:
• Venous thromboembolism (deep vein thrombosis, pulmonary embolism) (see section 4.4)
• Grade 3 or 4 neutropenia, febrile neutropenia and grade 3 or 4 thrombocytopenia (see section 4.4).
The most commonly observed adverse reactions which occurred more frequently in the lenalidomide groups compared to the control arm in the Phase III study were neutropenia (76.8%), thrombocytopenia (46.4%), diarrhoea (34.8%), constipation (19.6%), nausea (19.6%), pruritus (25.4%), rash (18.1%), fatigue (18.1%) and muscle spasms (16.7%).
Tabulated list of adverse reactions
Tabulated summary for combination therapy
The adverse reactions observed in patients treated for multiple myeloma are listed below by system organ class and frequency. Within each frequency grouping, adverse reactions are presented in order of decreasing seriousness. Frequencies are defined as: very common (≥ 1/10); common (≥ 1/100 to < 1/10); uncommon (≥ 1/1,000 to < 1/100); rare (≥ 1/10,000 to < 1/1,000); very rare (< 1/10,000), not known (cannot be estimated from the available data).
The following table is derived from data gathered during the multiple myeloma studies with combination therapy. . The data were not adjusted according to the greater duration of treatment in the lenalidomide/dexamethasone versus the placebo/dexamethasone arms in the pivotal multiple myeloma studies (see section 5.1).
Adverse reactions have been included under the appropriate category in the table below according to the highest frequency observed in any of the main clinical trials.
Table 1: ADRs reported in clinical studies in patients with multiple myeloma treated with lenalidomide in combination with dexamethasone, or with melphalan and prednisone

System Organ Class/ Preferred Term

All ADRs/Frequency

Grade 3−4 ADRs/Frequency

Infections and Infestations

Very Common

Pneumonia, Upper respiratory tract infection, Bacterial, viral and fungal infections (including opportunistic infections), Nasopharyngitis, Pharyngitis, Bronchitis

Common

Sepsis, Sinusitis

Common

Pneumonia, Bacterial, viral and fungal infections (including opportunistic infections), Sepsis, Bronchitis

Neoplasms Benign, Malignant and Unspecified (incl cysts and polyps)

Uncommon

Basal cell carcinoma

Squamous skin cancer^*

Common

Acute myeloid leukaemia, Myelodysplastic syndrome, Squamous cell carcinoma of skin**

Uncommon

T-cell type acute leukaemia, Basal cell carcinoma, Tumour lysis syndrome

Blood and Lymphatic System Disorders

Very Common

Neutropenia^, Thrombocytopenia ^, Anaemia, Haemorrhagic disorder ^, Leucopenias

Common

Febrile neutropenia, Pancytopenia

Uncommon

Haemolysis, Autoimmune haemolytic anaemia, Haemolytic anaemia

Very Common

Neutropenia^, Thrombocytopenia^, Anaemia, Leucopenias

Common

Febrile neutropenia^, Pancytopenia, Haemolytic anaemia

Uncommon

Hypercoagulation, Coagulopathy

Immune System Disorders

Uncommon

Hypersensitivity^

 

Endocrine Disorders

Common

Hypothyroidism

 

Metabolism and Nutrition Disorders

Very Common

Hypokalaemia, Hyperglycaemia, Hypocalcaemia, Decreased appetite, Weight decreased

Common

Hypomagnesaemia, Hyperuricaemia, Dehydration

Common

Hypokalaemia, Hyperglycaemia, Hypocalcaemia, Diabetes mellitus, Hypophosphataemia, Hyponatraemia, Hyperuricaemia, Gout, Decreased appetite, Weight decreased

Psychiatric Disorders

Very Common

Depression, Insomnia

Uncommon

Loss of libido

Common

Depression, Insomnia

Nervous System Disorders

Very Common

Peripheral neuropathies (excluding motor neuropathy), Dizziness, Tremor, Dysgeusia, Headache

Common

Ataxia, Balance impaired

Common

Cerebrovascular accident, Dizziness, Syncope

Uncommon

Intracranial haemorrhage ^, Transient ischaemic attack, Cerebral ischaemia

Eye Disorders

Very Common

Cataracts, Blurred vision

Common

Reduced visual acuity

Common

Cataract

Uncommon

Blindness

Ear and Labyrinth Disorders

Common

Deafness (Including Hypoacusis), Tinnitus

 

Cardiac Disorders

Common

Atrial fibrillation, Bradycardia

Uncommon

Arrhythmia, QT prolongation, Atrial flutter, Ventricular extrasystoles

Common

Myocardial infarction (including acute) ^, Atrial fibrillation, Congestive cardiac failure, Tachycardia, Cardiac failure, Myocardial ischaemia

Vascular Disorders

Very Common

Venous thromboembolic events, predominantly deep vein thrombosis and pulmonary embolism^

Common

Hypotension, Hypertension, Ecchymosis^

Very Common

Venous thromboembolic events, predominantly deep vein thrombosis and pulmonary embolism^

Common

Vasculitis

Uncommon

Ischemia, Peripheral ischemia, Intracranial venous sinus thrombosis

Respiratory, Thoracic and Mediastinal Disorders

Very Common

Dyspnoea, Epistaxis^

Common

Respiratory distress, Dyspnoea

Gastrointestinal Disorders

Very Common

Diarrhoea, Constipation, Abdominal pain, Nausea, Vomiting, Dyspepsia

Common

Gastrointestinal haemorrhage (including rectal haemorrhage, haemorrhoidal haemorrhage, peptic ulcer haemorrhage and gingival bleeding)^, Dry mouth, Stomatitis, Dysphagia

Uncommon

Colitis, Caecitis

Common

Diarrhoea, Constipation, Abdominal pain, Nausea, Vomiting

Hepatobiliary Disorders

Common

Abnormal liver function tests

Uncommon

Hepatic failure^

Common

Cholestasis, Abnormal liver function tests

Uncommon

Hepatic failure^

Skin and Subcutaneous Tissue Disorders

Very Common

Rashes, Pruritus

Common

Urticaria, Hyperhidrosis, Dry skin, Skin hyperpigmentation, Eczema, Erythema

Uncommon

Skin discolouration, Photosensitivity reaction

Common

Rashes

Musculoskeletal and Connective Tissue Disorders

Very Common

Muscle spasms, Bone pain, Musculoskeletal and connective tissue pain and discomfort, Arthralgia

Common

Muscular weakness, Joint swelling, Myalgia

Common

Muscular weakness, Bone pain

Uncommon

Joint swelling

Renal and Urinary Disorders

Very Common

Renal failure (including acute)

Common

Haematuria^, Urinary retention, Urinary incontinence

Uncommon

Acquired Fanconi syndrome

Uncommon

Renal tubular necrosis

Reproductive System and Breast Disorders

Common

Erectile dysfunction

 

General Disorders and Administration Site Conditions

Very Common

Fatigue, Oedema (including peripheral oedema), Pyrexia, Asthenia, Influenza like illness syndrome (including pyrexia, cough, myalgia, musculoskeletal pain, headache and rigors)

Common

Chest pain, Lethargy

Common

Fatigue, Pyrexia, Asthenia

Investigations

Common

C-reactive protein increased

 

Injury, Poisoning and Procedural Complications

Common

Fall, Contusion^

 
^See section 4.8 description of selected adverse reactions
* Squamous skin cancer was reported in clinical trials in previously treated myeloma patients with lenalidomide/dexamethasone compared to controls
** Squamous cell carcinoma of skin was reported in a clinical trial in newly diagnosed myeloma patients with lenalidomide/dexamethasone compared to controls
Tabulated summary from monotherapy
The adverse reactions observed in patients treated for myelodysplastic syndromes are listed below by system organ class and frequency. Within each frequency grouping, adverse reactions are presented in order of decreasing seriousness. Frequencies are defined as: very common (≥ 1/10); common (≥ 1/100 to < 1/10); uncommon (≥ 1/1,000 to < 1/100); rare (≥ 1/10,000 to < 1/1,000); very rare (< 1/10,000); not known (cannot be estimated from the available data).
The following table is derived from data gathered during the main studies in monotherapy for myelodysplastic syndromes.
Adverse reactions have been included under the appropriate category in the table below according to the highest frequency observed in any of the main clinical trials.
Table 2: ADRs reported in clinical trials in patients with myelodysplastic syndromes treated with lenalidomide#

System Organ Class/ Preferred Term

All ADRs/Frequency

Grade 3−4 ADRs/Frequency

Infections and Infestations

Very Common

Bacterial, viral and fungal infections (including opportunistic infections)

Very Common

Pneumonia

Common

Bacterial, viral and fungal infections (including opportunistic infections)

Blood and Lymphatic System Disorders

Very Common

Thrombocytopenia^, Neutropenia^, Leucopenias

Very Common

Thrombocytopenia^, Neutropenia^, Leucopenias

Common

Febrile Neutropenia^

Endocrine Disorders

Very Common

Hypothyroidism

 

Metabolism and Nutrition Disorders

Very Common

Decreased appetite

Common

Iron overload, Weight decreased

Common

Hyperglycaemia, Decreased appetite

Psychiatric Disorders

 

Common

Altered mood◊~

Nervous System Disorders

Very Common

Dizziness, Headache

Common

Paraesthesia

 

Cardiac Disorders

 

Common

Acute myocardial infarction^, Atrial fibrillation, Cardiac failure

Vascular Disorders

Common

Hypertension, Haematoma

Common

Venous thromboembolic events, predominantly deep vein thrombosis and pulmonary embolism^

Respiratory, Thoracic and Mediastinal Disorders

Very Common

Epistaxis^

Common

Bronchitis

Gastrointestinal Disorders

Very Common

Diarrhoea, Abdominal pain (including upper), Nausea, Vomiting, Constipation

Common

Dry mouth, Dyspepsia

Common

Diarrhoea, Nausea, Toothache

Hepatobiliary Disorders

Common

Abnormal liver function tests

Common

Abnormal liver function tests

Skin and Subcutaneous Tissue Disorders

Very Common

Rashes, Dry Skin, Pruritus

Common

Rashes, Pruritus

Musculoskeletal and Connective Tissue Disorders

Very Common

Muscle spasms, Musculoskeletal pain (including back pain and pain in extremity), Arthralgia, Myalgia

Common

Back pain

Renal and Urinary Disorders

 

Common

Renal failure

General Disorders and Administration Site Conditions

Very Common

Fatigue, Peripheral oedema, Influenza like illness syndrome (including pyrexia, cough, pharyngitis, myalgia, musculoskeletal pain, headache)

Common

Pyrexia

Injury, Poisoning and Procedural Complications

 

Common

Fall

^see section 4.8 description of selected adverse reactions
◊Adverse events reported as serious in myelodysplastic syndromes clinical trials
~Altered mood was reported as a common serious adverse event in the myelodysplastic syndromes Phase III study; it was not reported as a grade 3 or 4 adverse event
# Algorithm applied for myelodysplastic syndromes:
• Myelodysplastic syndromes Phase III study (double-blind safety population, difference between lenalidomide 5/10mg and placebo by initial dosing regimen occurring in at least 2 subjects)
o All treatment-emergent adverse events with ≥ 5% of subjects in lenalidomide and at least 2% difference in proportion between lenalidomide and placebo
o All treatment-emergent grade 3 or 4 adverse events in 1% of subjects in lenalidomide and at least 1% difference in proportion between lenalidomide and placebo
o All treatment-emergent serious adverse events in 1% of subjects in lenalidomide and at least 1% difference in proportion between lenalidomide and placebo
• Myelodysplastic syndromes Phase II study
o All treatment-emergent adverse events with ≥ 5% of lenalidomide treated subjects
o All treatment-emergent grade 3 or 4 adverse\events in 1% of lenalidomide treated subjects
o All treatment-emergent serious adverse events in 1% of lenalidomide treated subjects
• Algorithm applied for inclusion in the SmPC: All ADRs captured by the Phase III study algorithm are included in the EU SmPC. For these ADRs, an additional check of the frequency of the ADRs captured by the Phase II study algorithm was undertaken and, if the frequency of the ADRs in the Phase II study was higher than in the Phase III study, the event was included in the EU SmPC at the frequency it occurred in the Phase II study.
Tabulated summary of post-marketing adverse reactions
In addition to the above adverse reactions identified from the pivotal clinical trials, the following table is derived from data gathered from post-marketing data.
Table 3: ADRs reported in in post-marketing use in patients with multiple myeloma treated with lenalidomide

System Organ Class/ Preferred Term

All ADRs/Frequency

Grade 3−4 ADRs/Frequency

Neoplasms Benign, Malignant and Unspecified (incl cysts and polyps)

 

Rare

Tumour lysis syndrome

Endocrine Disorders

Common

Hyperthyroidism

 

Respiratory, Thoracic and Mediastinal Disorders

 

Not Known

Interstitial pneumonitis

Gastrointestinal Disorders

 

Not Known

Pancreatitis

Hepatobiliary Disorders

Not Known

Acute hepatic failure^, Hepatitis toxic^, Cytolytic hepatitis^, Cholestatic hepatitis^, Mixed cytolytic/cholestatic hepatitis^

Not Known

Acute hepatic failure^, Hepatitis toxic^

Skin and Subcutaneous Tissue Disorders

 

Uncommon

Angioedema

Rare

Stevens-Johnson Syndrome^, Toxic epidermal necrolysis^

Not Known

Leukocytoclastic vasculitis

^see section 4.8 description of selected adverse reactions
Description of selected adverse reactions
Teratogenicity
Lenalidomide is structurally related to thalidomide. Thalidomide is a known human teratogenic active substance that causes severe life-threatening birth defects. Lenalidomide induced in monkeys malformations similar to those described with thalidomide (see sections 4.6 and 5.3). If lenalidomide is taken during pregnancy, a teratogenic effect of lenalidomide in humans is expected.
Neutropenia and thrombocytopenia
• Newly diagnosed multiple myeloma patients treated with lenalidomide in combination with low dose dexamethasone
The combination of lenalidomide with low dose dexamethasone in newly diagnosed multiple myeloma patients is associated with a decreased incidence of grade 4 neutropenia (8.5% in Rd and Rd18, compared with 15% in MPT). Grade 4 febrile neutropenia was observed infrequently (0.6% compared with 0.7% in MPT).
The combination of lenalidomide with low dose dexamethasone in newly diagnosed multiple myeloma patients is associated with a decreased incidence of grade 3 and 4 thrombocytopenia (8.1 in Rd and Rd18 compared to 11% in MPT).
• Newly diagnosed multiple myeloma patients treated with lenalidomide in combination with melphalan and prednisone
The combination of lenalidomide with melphalan and prednisone in newly diagnosed multiple myeloma patients is associated with a higher incidence of grade 4 neutropenia (34.1% in MPR+R/MPR+p compared with 7.8% in MPp+p). There was a higher incidence of grade 4 febrile neutropenia observed (1.7% in MPR+R/MPR+p compared to 0.0% in MPp+p).
The combination of lenalidomide with melphalan and prednisone in newly diagnosed multiple myeloma patients is associated with a higher incidence of grade 3 and grade 4 thrombocytopenia (40.4% in MPR+R/MPR+p treated patients, compared with 13.7% in MPp+p-treated patients).
• Multiple myeloma with at least one prior therapy
The combination of lenalidomide with dexamethasone in multiple myeloma patients is associated with a higher incidence of grade 4 neutropenia (5.1% in lenalidomide/dexamethasone-treated patients compared with 0.6% in placebo/dexamethasone-treated patients). Grade 4 febrile neutropenia episodes were observed infrequently (0.6% in lenalidomide/dexamethasone-treated patients compared to 0.0% in placebo/dexamethasone treated patients).
The combination of lenalidomide with dexamethasone in multiple myeloma patients is associated with a higher incidence of grade 3 and grade 4 thrombocytopenia (9.9% and 1.4%, respectively, in lenalidomide/dexamethasone-treated patients compared to 2.3% and 0.0% in placebo/dexamethasone-treated patients).
• Myelodysplastic syndromes
In myelodysplastic syndromes patients, lenalidomide is associated with a higher incidence of grade 3 or 4 neutropenia (74.6% in lenalidomide-treated patients compared with 14.9% in patients on placebo in the Phase III study). Grade 3 or 4 febrile neutropenia episodes were observed in 2.2% of lenalidomide-treated patients compared with 0.0% in patients on placebo). Lenalidomide is associated with a higher incidence of grade 3 or 4 thrombocytopenia (37% in lenalidomide-treated patients compared with 1.5% in patients on placebo in the Phase III study).
Venous thromboembolism
An increased risk of DVT and PE is associated with the use of lenalidomide with dexamethasone in patients with multiple myeloma, and to a lesser extent in patients treated with melphalan and prednisone (see section 4.5). Concomitant administration of erythropoietic agents or previous history of DVT may also increase thrombotic risk in these patients.
Myocardial infarction
Myocardial infarction has been reported in patients receiving lenalidomide, particularly in those with known risk factors.
Haemorrhagic disorders
Haemorrhagic disorders are listed under several system organ classes: Blood and lymphatic system disorders; nervous system disorders (intracranial haemorrhage); respiratory, thoracic and mediastinal disorders (epistaxis); gastrointestinal disorders (gingival bleeding, haemorrhoidal haemorrhage, rectal haemorrhage); renal and urinary disorders (haematuria); injury, poisoning and procedural complications (contusion) and vascular disorders (ecchymosis).
Allergic reactions
Cases of allergic reaction/hypersensitivity reactions have been reported. A possible cross-reaction between lenalidomide and thalidomide has been reported in the literature.
Severe skin reactions
SJS and TEN have been reported. Patients with a history of severe rash associated with thalidomide treatment should not receive lenalidomide.
Second primary malignancies
*In clinical trials in previously treated myeloma patients with lenalidomide/dexamethasone compared to controls, mainly comprising of basal cell or squamous cell skin cancers.
Acute myeloid leukaemia
• Multiple myeloma
Cases of AML have been observed in clinical trials of newly diagnosed multiple myeloma in patients taking lenalidomide treatment in combination with melphalan or immediately following high dose melphalan and ASCT (see section 4.4). This increase was not observed in clinical trials of newly diagnosed multiple myeloma in patients taking lenalidomide in combination with low dose dexamethasone compared to thalidomide in combination with melphalan and prednisone.
• Myelodysplastic syndromes
Baseline variables including complex cytogenetics and TP53 mutation are associated with progression to AML in subjects who are transfusion dependent and have a Del (5q) abnormality (see section 4.4). The estimated 2-year cumulative risk of progression to AML were 13.8% in patients with an isolated Del (5q) abnormality compared to 17.3% for patients with Del (5q) and one additional cytogenetic abnormality and 38.6% in patients with a complex karyotype.
In a post-hoc analysis of a clinical trial of Revlimid in myelodysplastic syndromes, the estimated 2-year rate of progression to AML was 27.5 % in patients with IHC-p53 positivity and 3.6% in patients with IHC-p53 negativity (p=0.0038). In the patients with IHC-p53 positivity, a lower rate of progression to AML was observed amongst patients who achieved a transfusion independence (TI) response (11.1%) compared to a non-responder (34.8%).
Hepatic disorders
The following post-marketing adverse reactions have been reported (frequency unknown): acute hepatic failure and cholestasis (both potentially fatal), toxic hepatitis, cytolytic hepatitis, mixed cytolytic/cholestatic hepatitis.
Rhabdomyolysis
Rare cases of rhabdomyolysis have been observed, some of them when lenalidomide is administered with a statin.
Thyroid disorders
Cases of hypothyroidism and cases of hyperthyroidism have been reported (see section 4.4 Thyroid disorders).
Gastrointestinal disorders
Gastrointestinal perforations have been reported during treatment with lenalidomide. Gastrointestinal perforations may lead to septic complications and may be associated with fatal outcome.
Reporting of suspected adverse reactions
Reporting suspected adverse reactions after authorisation of the medicinal product is important. It allows continued monitoring of the benefit/risk balance of the medicinal product. Healthcare professionals are asked to report any suspected adverse reactions via the Yellow Card Scheme at www.mhra.gov.uk/yellowcard (Freephone 0808 100 3352).
4.9 Overdose
There is no specific experience in the management of lenalidomide overdose in patients, although in dose-ranging studies some patients were exposed to up to 150 mg, and in single-dose studies, some patients were exposed to up to 400 mg. The dose limiting toxicity in these studies was essentially haematological. In the event of overdose, supportive care is advised.
5. Pharmacological properties
5.1 Pharmacodynamic properties
Pharmacotherapeutic group: Other immunosuppressants. ATC code: L04AX04.
Mechanism of action
The lenalidomide mechanism of action includes anti-neoplastic, anti-angiogenic, pro-erythropoietic, and immunomodulatory properties. Specifically, lenalidomide inhibits proliferation of certain haematopoietic tumour cells (including MM plasma tumour cells and those with deletions of chromosome 5), enhances T cell- and Natural Killer (NK) cell-mediated immunity and increases the number of NK T cells, inhibits angiogenesis by blocking the migration and adhesion of endothelial cells and the formation of microvessels, augments foetal haemoglobin production by CD34+ haematopoietic stem cells, and inhibits production of pro-inflammatory cytokines (e.g., TNF-α and IL-6) by monocytes.
In MDS Del (5q), lenalidomide was shown to selectively inhibit the abnormal clone by increasing the apoptosis of Del (5q) cells.
Lenalidomide binds directly to cereblon, a component of a cullin ring E3 ubiquitin ligase enzyme complex that includes deoxyribonucleic acid (DNA) damage-binding protein 1(DDB1), cullin 4 (CUL4), and regulator of cullins 1 (Roc1). In the presence of lenalidomide, cereblon binds substrate proteins Aiolos and Ikaros which are lymphoid transcriptional factors, leading to their ubiquitination and subsequent degradation resulting in cytoxic and immunomodulatory effects.
Clinical efficacy and safety
Lenalidomide has been evaluated in two phase III studies in newly diagnosed multiple myeloma and two phase III studies in relapsed refractory multiple myeloma as described below.
Newly diagnosed multiple myeloma
Lenalidomide in combination with dexamethasone in patients who are not candidates for stem cell transplantation
The safety and efficacy of lenalidomide was assessed in a Phase III, multicenter, randomized, open-label, 3-arm study (MM-020) of patients who were at least 65 years of age or older or, if younger than 65 years of age, were not candidates for stem cell transplantation because they declined to undergo stem cell transplantation or stem cell transplantation is not available to the patient due to cost or other reason.The study (MM-020) compared lenalidomide and dexamethasone (Rd) given for 2 different durations of time (i.e., until progressive disease [Arm Rd] or for up to eighteen 28-day cycles [72 weeks, Arm Rd18]) to that of melphalan, prednisone and thalidomide (MPT) for a maximum of twelve 42-day cycles (72 weeks). Patients were randomized (1:1:1) to 1 of 3 treatment arms. Patients were stratified at randomization by age (≤75 versus >75 years), stage (ISS Stages I and II versus Stage III), and country.
Patients in the Rd and Rd18 arms took lenalidomide 25 mg once daily on Days 1 to 21 of 28-day cycles according to protocol arm. Dexamethasone 40 mg was dosed once daily on Days 1, 8, 15, and 22 of each 28-day cycle. Initial dose and regimen for Rd and Rd18 were adjusted according to age and renal function (see section 4.2). Patients >75 years received a dexamethasone dose of 20 mg once daily on Days 1, 8, 15, and 22 of each 28-day cycle. All patients received prophylactic anticoagulation (low molecular weight heparin, warfarin, heparin, low-dose aspirin) during the study.
The primary efficacy endpoint in the study was progression free survival (PFS). In total 1623 patients were enrolled into the study, with 535 patients randomized to Rd, 541 patients randomized to Rd18 and 547 patients randomized to MPT. The demographics and disease-related baseline characteristics of the patients were well balanced in all 3 arms. In general, study subjects had advanced-stage disease: of the total study population, 41% had ISS stage III, 9% had severe renal insufficiency (creatinine clearance [CLcr] < 30 mL/min). The median age was 73 in the 3 arms.
In an updated analysis of PFS, PFS2, OS and DR where the median follow up time for all surviving subjects was 45.5 months, the results of the study are presented in Table 4:
Table 4: Summary of overall efficacy data

Rd (N = 541)

Rd18 (N = 540)

MPT (N = 542)

Investigator-assessed PFS ☐ (months)

     

Mediana PFS time, months (95% CI)b

26.0 (20.7, 29.7)

21.0 (19.7, 22.4)

21.9 (19.8, 23.9)

HR [95% CI]c; p-valued

     

Rd vs MPT

0.69 (0.59, 0.80); <0.001

Rd vs Rd18

0.71 (0.61, 0.83); <0.001

Rd18 vs MPT

0.99 (0.86, 1.14); <0.001

PFS2e ☐ (months)

     

Mediana PFS time, months (95% CI)b

42.9 (38.1, 47.4)

40.0 (36.2, 44.2)

35.0 (30.4, 37.8)

HR [95% CI]c; p-valued

     

Rd vs MPT

0.74 (0.63, 0.86); <0.001

Rd vs Rd18

0.92 (0.78, 1.08); 0.316

Rd18 vs MPT

0.80 (0.69, 0.93); 0.004

Overall survival (months)

     

Mediana OS time, months (95% CI)b

58.9 (56.0, NE)

56.7 (50.1, NE)

48.5 (44.2, 52.0)

HR [95% CI]c; p-valued

     

Rd vs MPT

0.75 (0.62, 0.90); 0.002

Rd vs Rd18

0.91 (0.75, 1.09); 0.305

Rd18 vs MPT

0.83 (0.69, 0.99); 0.034

Follow-up (months)

     

Medianf (min, max): all patients

40.8 (0.0, 65.9)

40.1 (0.4, 65.7)

38.7 (0.0, 64.2)

Myeloma responseg n (%)

     

CR

81 (15.1)

77 (14.2)

51 (9.3)

VGPR

152 (28.4)

154 (28.5)

103 (18.8)

PR

169 (31.6)

166 (30.7)

187 (34.2)

Overall response: CR, VGPR, or PR

402 (75.1)

397 (73.4)

341 (62.3)

Duration of response ☐ (months)h

     

Mediana (95% CI)b

35.0 (27.9, 43.4)

22.1 (20.3, 24.0)

22.3 (20.2, 24.9)

AMT = antimyeloma therapy; CI = confidence interval; CR = complete response; d = low-dose dexamethasone; HR = hazard ratio; IMWG = International Myeloma Working Group; IRAC = Independent Response Adjudication Committee; M = melphalan; max = maximum; min = minimum; NE = not estimable; OS = overall survival; P = prednisone; PFS = progression-free survival; PR = partial response; R = lenalidomide; Rd = Rd given until documentation of progressive disease; Rd18 = Rd given for ≥ 18 cycles; SE = standard error; T = thalidomide; VGPR = very good partial response; vs = versus.
a The median is based on the Kaplan-Meier estimate.
b The 95% CI about the median.
c Based on Cox proportional hazards model comparing the hazard functions associated with the indicated treatment arms.
d The p-value is based on the unstratified log-rank test of Kaplan-Meier curve differences between the indicated treatment arms.
e Exploratory endpoint (PFS2)
f The median is the univariate statistic without adjusting for censoring.
g Best assessment of adjudicated response during the treatment phase of the study (for definitions of each response category, Data cutoff date = 24 May 2013).
h data cut 24 May 2014
Lenalidomide in combination with melphalan and prednisone followed by maintenance monotherapy in patients who are not eligible for transplantation
The safety and efficacy of lenalidomide was assessed in a Phase III multicenter, randomized double blind 3 arm study (MM-015) of patients who were 65 years or older and had a serum creatinine < 2.5 mg/dL. The study compared lenalidomide in combination with melphalan and prednisone (MPR) with or without lenalidomide maintenance monotherapy until disease progression, to that of melphalan and prednisone for a maximum of 9 cycles. Patients were randomized in a 1:1:1 ratio to one of 3 treatment arms. Patients were stratified at randomisation by age (≤ 75 vs. > 75 years) and stage (ISS; Stages I and II vs. stage III).
This study investigated the use of combination therapy of MPR (melphalan 0.18 mg/kg orally on days 1-4 of repeated 28-day cycles; prednisone 2 mg/kg orally on days 1-4 of repeated 28-day cycles; and lenalidomide 10 mg/day orally on days 1-21 of repeated 28-day cycles) for induction therapy, up to 9 cycles. Patients who completed 9 cycles or who were unable to complete 9 cycles due to intolerance proceeded to maintenance monotherapy starting with lenalidomide 10 mg orally on days 1-21 of repeated 28-day cycles until disease progression.
The primary efficacy endpoint in the study was progression free survival (PFS). In total 459 patients were enrolled into the study, with 152 patients randomized to MPR+R, 153 patients randomized to MPR+p and 154 patients randomized to MPp+p. The demographics and disease-related baseline characteristics of the patients were well balanced in all 3 arms; notably, approximately 50% of the patients enrolled in each arm had the following characteristics; ISS Stage III, and creatinine clearance < 60 mL/min. The median age was 71 in the MPR+R and MPR+p arms and 72 in the MPp+p arm.
In an analysis of PFS, PFS2, OS using a cut off of April 2013 where the median follow up time for all surviving subjects was 62.4 months, the results of the study are presented in Table 5
Table 5: Summary of overall efficacy data

MPR+R

(N = 152)

MPR+p

(N = 153)

MPp +p

(N = 154)

Investigator-assessed PFS ☐ (months)

 

Mediana PFS time, months (95% CI)

27.4 (21.3, 35.0)

14.3 (13.2, 15.7)

13.1 (12.0, 14.8)

HR [95% CI]; p-value

 

MPR+R vs MPp+p

0.37 (0.27, 0.50); <0.001

MPR+R vs MPR+p

0.47 (0.35, 0.65); <0.001

MPR+p vs MPp +p

0.78 (0.60, 1.01); 0.059

PFS2 ☐ (months) ¤

 

Mediana PFS time, months (95% CI)

39.7 (29.2, 48.4)

27.8 (23.1, 33.1)

28.8 (24.3, 33.8)

HR [95% CI]; p-value

 

MPR+R vs MPp+p

0.70 (0.54, 0.92); 0.009

MPR+R vs MPR+p

0.77 (0.59, 1.02); 0.065

MPR+p vs MPp +p

0.92 (0.71, 1.19); 0.051

Overall survival (months)

 

Mediana OS time, months (95% CI)

55.9 (49.1, 67.5)

51.9 (43.1, 60.6)

53.9 (47.3, 64.2)

HR [95% CI]; p-value

 

MPR+R vs MPp+p

0.95 (0.70, 1.29); 0.736

MPR+R vs MPR+p

0.88 (0.65, 1.20); 0.43

MPR+p vs MPp +p

1.07 (0.79, 1.45); 0.67

Follow-up (months)

 

Median (min, max): all patients

48.4 (0.8, 73.8)

46.3 (0.5, 71.9)

50.4 (0.5, 73.3)

Investigator-assessed Myeloma response n (%)

 

CR

30 (19.7)

17 (11.1)

9 (5.8)

PR

90 (59.2)

99 ( 64.7)

75 (48.7)

Stable Disease (SD)

24 (15.8)

31 (20.3)

63 (40.9)

Response Not Evaluable (NE)

8 (5.3)

4 (2.6)

7 (4.5)

Investigator-assessed Duration of response (CR+PR) ☐ (months)

     

Mediana (95% CI)

26.5 (19.4, 35.8)

12.4 (11.2, 13.9)

12.0 (9.4, 14.5)

CI = confidence interval; CR = complete response; HR = Hazard Rate; M = melphalan; NE = not estimable; OS = overall survival; p = placebo; P = prednisone;
PD = progressive disease; PR = partial response; R = lenalidomide; SD = stable disease; VGPR = very good partial response.
ª The median is based on the Kaplan-Meier estimate
¤PFS2 (an exploratory endpoint) was defined for all patients (ITT) as time from randomization to start of 3rd line antimyeloma therapy (AMT) or death for all randomized patients
Supportive newly diagnosed multiple myeloma studies
An open-label, randomized, multicenter, Phase III study (ECOG E4A03) was conducted in 445 patients with newly diagnosed multiple myeloma; 222 patients were randomized to the lenalidomide/low dose dexamethasone arm, and 223 were randomized to the lenalidomide/standard dose dexamethasone arm. Patients randomized to the lenalidomide/standard dose dexamethasone arm received lenalidomide 25 mg/day, Days 1 to 21 every 28 days plus dexamethasone 40 mg/day on Days 1 to 4, 9 to 12, and 17 to 20 every 28 days for the first four cycles. Patients randomized to the lenalidomide/low dose dexamethasone arm received lenalidomide 25 mg/day, Days 1 to 21 every 28 days plus low dose dexamethasone – 40 mg/day on Days 1, 8, 15, and 22 every 28 days. In the lenalidomide/low dose dexamethasone group, 20 patients (9.1%) underwent at least one dose interruption compared to 65 patients (29.3%) in the lenalidomide/standard dose dexamethasone arm.
In a post-hoc analysis, lower mortality was observed in the lenalidomide/low dose dexamethasone arm 6.8% (15/220) compared to the lenalidomide/standard dose dexamethasone arm 19.3% (43/223), in the newly diagnosed multiple myeloma patient population, with a median follow up of 72.3 weeks.
However with a longer follow-up, the difference in overall survival in favour of lenalidomide/ low dose dexamethasone tends to decrease.
Multiple myeloma with at least one prior therapy
The efficacy and safety of lenalidomide were evaluated in two Phase III multi-centre, randomised, double-blind, placebo-controlled, parallel-group controlled studies (MM-009 and MM-010) of lenalidomide plus dexamethasone therapy versus dexamethasone alone in previously treated patients with multiple myeloma. Out of 353 patients in the MM-009 and MM-010 studies who received lenalidomide/dexamethasone, 45.6% were aged 65 or over. Of the 704 patients evaluated in the MM-009 and MM-010 studies, 44.6% were aged 65 or over.
In both studies, patients in the lenalidomide/dexamethasone (len/dex) group took 25 mg of lenalidomide orally once daily on Days 1 to 21 and a matching placebo capsule once daily on Days 22 to 28 of each 28-day cycle. Patients in the placebo/dexamethasone (placebo/dex) group took 1 placebo capsule on Days 1 to 28 of each 28-day cycle. Patients in both treatment groups took 40 mg of dexamethasone orally once daily on Days 1 to 4, 9 to 12, and 17 to 20 of each 28-day cycle for the first 4 cycles of therapy. The dose of dexamethasone was reduced to 40 mg orally once daily on Days 1 to 4 of each 28-day cycle after the first 4 cycles of therapy. In both studies, treatment was to continue until disease progression. In both studies, dose adjustments were allowed based on clinical and laboratory finding.
The primary efficacy endpoint in both studies was time to progression (TTP). In total, 353 patients were evaluated in the MM-009 study; 177 in the lenalidomide/dexamethasone group and 176 in the placebo/dexamethasone group and, in total, 351 patients were evaluated in the MM-010 study; 176 in the lenalidomide/dexamethasone group and 175 in the placebo/dexamethasone group.
In both studies, the baseline demographic and disease-related characteristics were comparable between the lenalidomide/dexamethasone and placebo/dexamethasone groups. Both patient populations presented a median age of 63 years, with a comparable male to female ratio. The ECOG performance status was comparable between both groups, as was the number and type of prior therapies.
Pre-planned interim analyses of both studies showed that lenalidomide/dexamethasone was statistically significantly superior (p < 0.00001) to dexamethasone alone for the primary efficacy endpoint, TTP (median follow-up duration of 98.0 weeks). Complete response and overall response rates in the lenalidomide/dexamethasone arm were also significantly higher than the dexamethasone/placebo arm in both studies. Results of these analyses subsequently led to an unblinding in both studies, in order to allow patients in the placebo/dexamethasone group to receive treatment with the lenalidomide/dexamethasone combination.
An extended follow-up efficacy analysis was conducted with a median follow-up of 130.7 weeks. Table 6 summarises the results of the follow-up efficacy analyses – pooled studies MM-009 and MM-010.
In this pooled extended follow-up analysis, the median TTP was 60.1 weeks (95% CI: 44.3, 73.1) in patients treated with lenalidomide/dexamethasone (N = 353) versus 20.1 weeks (95% CI: 17.7, 20.3) in patients treated with placebo/dexamethasone (N = 351). The median progression free survival was 48.1 weeks (95% CI: 36.4, 62.1) in patients treated with lenalidomide/dexamethasone versus 20.0 weeks (95% CI: 16.1, 20.1) in patients treated with placebo/dexamethasone. The median duration of treatment was 44.0 weeks (min: 0.1, max: 254.9) for lenalidomide/dexamethasone and 23.1 weeks (min: 0.3, max: 238.1) for placebo/dexamethasone. Complete response (CR), partial response (PR) and overall response (CR+PR) rates in the lenalidomide/dexamethasone arm remain significantly higher than in the dexamethasone/placebo arm in both studies. The median overall survival in the extended follow-up analysis of the pooled studies is 164.3 weeks (95% CI: 145.1, 192.6) in patients treated with lenalidomide/dexamethasone versus 136.4 weeks (95% CI: 113.1, 161.7) in patients treated with placebo/dexamethasone. Despite the fact that 170 out of the 351 patients randomised to placebo/dexamethasone received lenalidomide after disease progression or after the studies were unblinded, the pooled analysis of overall survival demonstrated a statistically significant survival advantage for lenalidomide/dexamethasone relative to placebo/dexamethasone (hazard ratio = 0.833, 95% CI = [0.687, 1.009], p=0.045).
Table 6: Summary of results of efficacy analyses as of cut-off date for extended follow-up — pooled studies MM-009 and MM-010 (cut-offs 23 July 2008 and 2 March 2008, respectively)

Endpoint

len/dex

(N=353)

placebo/dex

(N=351)

 

Time to event

   

Hazard ratio [95% CI], p-value a

Time to progression

Median [95% CI], weeks

60.1 [44.3, 73.1]

20.1 [17.7, 20.3]

0.350 [0.287, 0.426], p < 0.001

Progression free survival

Median [95% CI], weeks

48.1

[36.4, 62.1]

20.0 [16.1, 20.1]

0.393 [0.326, 0.473]

p < 0.001

Overall survival

Median [95% CI], weeks

1-year Overall survival rate

164.3 [145.1, 192.6]

82%

136.4 [113.1, 161.7]

75%

0.833 [0.687, 1.009]

p = 0.045

Response rate

   

Odds ratio [95% CI], p-value b

Overall response [n, %]

Complete response [n, %]

212 (60.1)

58 (16.4)

75 (21.4)

11 (3.1)

5.53 [3.97, 7.71], p < 0.001

6.08 [3.13, 11.80], p < 0.001

a: Two-tailed log rank test comparing survival curves between treatment groups.
b: Two-tailed continuity-corrected chi-square test.
Myelodysplastic syndromes
The efficacy and safety of lenalidomide were evaluated in patients with transfusion-dependent anaemia due to low- or intermediate-1-risk myelodysplastic syndromes associated with a deletion 5q cytogenetic abnormality, with or without additional cytogenetic abnormalities, in two main studies: a Phase III, multicentre, randomised, double-blind, placebo-controlled, 3-arm study of two doses of oral lenalidomide (10 mg and 5 mg) versus placebo (MDS-004); and a Phase II, a multicentre, single-arm, open-label study of lenalidomide (10 mg) (MDS-003).
The results presented below represent the intent-to-treat population studied in MDS-003 and MDS-004; with the results in the isolated Del (5q) sub-population also shown separately (see section 4.1 for the approved indication).
In study MDS-004, in which 205 patients were equally randomised to receive lenalidomide 10 mg, 5 mg or placebo, the primary efficacy analysis consisted of a comparison of the transfusion-independence response rates of the 10 mg and 5 mg lenalidomide arms versus the placebo arm (double-blind phase 16 to 52 weeks and open-label up to a total of 156 weeks). Patients who did not have evidence of at least a minor erythroid response after 16 weeks were to be discontinued from treatment. Patients who had evidence of at least a minor erythroid response could continue therapy until erythroid relapse, disease progression or unacceptable toxicity. Patients, who initially received placebo or 5 mg lenalidomide and did not achieve at least a minor erythroid response after 16 weeks of treatment were permitted to switch from placebo to 5 mg lenalidomide or continue lenalidomide treatment at higher dose (5 mg to 10 mg).
In, study MDS-003, in which 148 patients received lenalidomide at a dose of 10 mg, the primary efficacy analysis consisted of an evaluation of the efficacy of lenalidomide treatments to achieve haematopoietic improvement in subjects with low- or intermediate-1 risk myelodysplastic syndromes.
Table 7: Summary of efficacy results – studies MDS-004 (double-blind phase) and MDS-003, intent-to-treat population

Endpoint

MDS-004

N = 205

MDS-003

N = 148

10 mg

N = 69

5 mg††

N = 69

Placebo*

N = 67

10 mg

N = 148

Transfusion Independence

(≥ 182 days) #

38 (55.1%)

24 (34.8%)

4 (6.0%)

86 (58.1%)

Transfusion Independence

(≥ 56 days) #

42 (60.9%)

33 (47.8%)

5 (7.5%)

97 (65.5%)

Median Time to Transfusion Independence (weeks)

4.6

4.1

0.3

4.1

Median Duration of Transfusion Independence (weeks)

NR

NR

NR

114.4

Median Increase in Hgb, g/dL

6.4

5.3

2.6

5.6

Subjects treated with lenalidomide 10 mg on 21 days of 28-day cycles
Subjects treated with lenalidomide 5 mg on 28 days of 28-day cycles
* The majority of patients on placebo discontinued the double-blind treatment for lack of efficacy after 16 weeks of treatment before entering the open-label phase
#Associated with an increase in Hgb of ≥ 1g/dL
∞ Not reached (i.e. the median was not reached)
In MDS-004, a significant larger proportion of patients with myelodysplastic syndromes achieved the primary endpoint of transfusion independence (>182 days) on lenalidomide 10 mg compared with placebo (55.1% vs. 6.0%). Amongst the 47 patients with an isolated Del (5q) cytogenetic abnormality and treated with lenalidomide 10 mg, 27 patients (57.4%) achieved red blood cell transfusion independence.
The median time to transfusion independence in the lenalidomide 10 mg arm was 4.6 weeks. The median duration of transfusion independence was not reached in any of the treatment arms, but should exceed 2 years for the lenalidomide-treated subjects. The median increase in haemoglobin (Hgb) from baseline in the 10 mg arm was 6.4 g/dL.
Additional endpoints of the study included cytogenetic response (in the 10 mg arm major and minor cytogenetic responses were observed in 30.0% and 24.0% of subjects, respectively), assessment of Health Related Quality of Life (HRQoL) and progression to acute myeloid leukaemia. Results of the cytogenetic response and HRQoL were consistent with the findings of the primary endpoint and in favour of lenalidomide treatment compared to placebo.
In MDS-003, a large proportion of patients with myelodysplastic syndromes achieved transfusion independence (>182 days) on lenalidomide 10 mg (58.1%). The median time to transfusion independence was 4.1 weeks. The median duration of transfusion independence was 114.4 weeks. The median increase in haemoglobin (Hgb) was 5.6 g/dL. Major and minor cytogenetic responses were observed in 40.9% and 30.7% of subjects, respectively.
A large proportion of subjects enrolled in MDS-003 (72.9%) and MDS-004 (52.7%) had received prior erythropoiesis-stimulating agents.
Paediatric population
The European Medicines Agency has waived the obligation to submit the results of studies with Revlimid in all subsets of the paediatric population in multiple myeloma and myelodysplastic syndromes (see section 4.2 for information on paediatric use).
5.2 Pharmacokinetic properties
Lenalidomide has an asymmetric carbon atom and can therefore exist as the optically active forms S(-) and R(+). Lenalidomide is produced as a racemic mixture. Lenalidomide is generally more soluble in organic solvents but exhibits the greatest solubility in 0.1N HCl buffer.
Absorption
Lenalidomide is rapidly absorbed following oral administration in healthy volunteers, under fasting conditions, with maximum plasma concentrations occurring between 0.5 and 2 hours post-dose. In patients, as well as in healthy volunteers, the maximum concentration (Cmax) and area-under-the-concentration time curve (AUC) increase proportionally with increases in dose. Multiple dosing does not cause marked medicinal product accumulation. In plasma, the relative exposures of the S- and R- enantiomers of lenalidomide are approximately 56% and 44%, respectively.
Co-administration with a high-fat and high-calorie meal in healthy volunteers reduces the extent of absorption, resulting in an approximately 20% decrease in area under the concentration versus time curve (AUC) and 50% decrease in Cmax in plasma. However, in the main multiple myeloma and myelodysplastic syndromes registration trials where the efficacy and safety were established for lenalidomide, the medicinal product was administered without regard to food intake. Thus, lenalidomide can be administered with or without food.
Population pharmacokinetic analyses indicate that the oral absorption rate of lenalidomide is similar between MM and MDS patients.
Distribution
In vitro (14C)-lenalidomide binding to plasma proteins was low with mean plasma protein binding at 23% and 29% in multiple myeloma patients and healthy volunteers, respectively.
Lenalidomide is present in human semen (< 0.01% of the dose) after administration of 25 mg/day and the medicinal product is undetectable in semen of a healthy subject 3 days after stopping the substance (see section 4.4).
Biotransformation and elimination
Results from human in vitro metabolism studies indicate that lenalidomide is not metabolised by cytochrome P450 enzymes suggesting that administration of lenalidomide with medicinal products that inhibit cytochrome P450 enzymes is not likely to result in metabolic medicinal product interactions in humans. In vitro studies indicate that lenalidomide has no inhibitory effect on CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A, or UGT1A1. Therefore, lenalidomide is unlikely to cause any clinically relevant medicinal product interactions when co-administered with substrates of these enzymes.
In vitro studies indicate that lenalidomide is not a substrate of human breast cancer resistance protein (BCRP), multidrug resistance protein (MRP) transporters MRP1, MRP2, or MRP3, organic anion transporters (OAT) OAT1 and OAT3, organic anion transporting polypeptide 1B1 (OATP1B1), organic cation transporters (OCT) OCT1 and OCT2, multidrug and toxin extrusion protein (MATE) MATE1, and organic cation transporters novel (OCTN) OCTN1 and OCTN2.
It is unknown whether lenalidomide is a human bile salt export pump (BSEP), BCRP, MRP2, OAT1, OAT3, OATP1B1, OATP1B3, or OCT2 inhibitor in vivo, though it has no inhibitory effect at in vitro concentrations up to 20 µM.
A majority of lenalidomide is eliminated through urinary excretion. The contribution of renal excretion to total clearance in subjects with normal renal function was 90%, with 4% of lenalidomide eliminated in faeces.
Lenalidomide is poorly metabolized as 82% of the dose is excreted unchanged in urine. Hydroxy-lenalidomide and N-acetyl-lenalidomide represent 4.59% and 1.83% of the excreted dose, respectively. The renal clearance of lenalidomide exceeds the glomerular filtration rate and therefore is at least actively secreted to some extent.
At doses of 5 to 25 mg/day, half-life in plasma is approximately 3 hours in healthy volunteers and ranges from 3 to 5 hours in patients with multiple myeloma or myelodysplastic syndromes.
Older people
No dedicated clinical studies have been conducted to evaluate pharmacokinetics of lenalidomide in the elderly. Population pharmacokinetic analyses included patients with ages ranging from 39 to 85 years old and indicate that age does not influence lenalidomide clearance (exposure in plasma). Because elderly patients are more likely to have decreased renal function, care should be taken in dose selection and it would be prudent to monitor renal function.
Renal impairment
The pharmacokinetics of lenalidomide was studied in subjects with renal impairment due to nonmalignant conditions. In this study, two methods were used to classify renal function: the urinary creatinine clearance measured over 24 hours and the creatinine clearance estimated by Cockcroft-Gault formula. The results indicate that as renal function decreases (< 50 mL/min), the total drug clearance decreases proportionally resulting in an increase in AUC. The AUC was increased by approximately 2.5, 4 and 5-fold in subjects with moderate renal impairment, severe renal impairment, and end-stage renal disease, respectively, compared to the group combining subjects with normal renal function and subjects with mild renal impairment. The half-life of lenalidomide increased from approximately 3.5 hours in subjects with creatinine clearance > 50 mL/min to more than 9 hours in subjects with reduced renal function < 50 mL/min. However, renal impairment did not alter the oral absorption of lenalidomide. The Cmax was similar between healthy subjects and patients with renal impairment. Approximately 30% of the medicinal product in the body was removed during a single 4-hour dialysis session. Recommended dose adjustments in patients with impaired renal function are described in section 4.2.
Hepatic impairment
Population pharmacokinetic analyses included patients with mild hepatic impairment (N=16, total bilirubin >1 to ≤1.5 x ULN or AST > ULN) and indicate that mild hepatic impairment does not influence lenalidomide clearance (exposure in plasma). There are no data available for patients with moderate to severe hepatic impairment.
Other intrinsic factors
Population pharmacokinetic analyses indicate that body weight (33- 135 kg), gender, race and type of haematological malignancy (MM or MDS) do not have a clinically relevant effect on lenalidomide clearance in adult patients.
5.3 Preclinical safety data
An embryofoetal development study has been conducted in monkeys administered lenalidomide at doses from 0.5 and up to 4 mg/kg/day. Findings from this study indicate that lenalidomide produced external malformations including non-patent anus and malformations of upper and lower extremities (bent, shortened, malformed, malrotated and/or absent part of the extremities, oligo and/or polydactyly) in the offspring of female monkeys who received the drug during pregnancy.
Various visceral effects (discoloration, red foci at different organs, small colourless mass above atrio-ventricular valve, small gall bladder, malformed diaphragm) were also observed in single foetuses.
Lenalidomide has a potential for acute toxicity; minimum lethal doses after oral administration were > 2000 mg/kg/day in rodents. Repeated oral administration of 75, 150 and 300 mg/kg/day to rats for up to 26 weeks produced a reversible treatment-related increase in kidney pelvis mineralisation in all 3 doses, most notably in females. The no observed adverse effect level (NOAEL) was considered to be less than 75 mg/kg/day, and is approximately 25-fold greater than the human daily exposure based on AUC exposure. Repeated oral administration of 4 and 6 mg/kg/day to monkeys for up to 20 weeks produced mortality and significant toxicity (marked weight loss, reduced red and white blood cell and platelet counts, multiple organ haemorrhage, gastrointestinal tract inflammation, lymphoid, and bone marrow atrophy). Repeated oral administration of 1 and 2 mg/kg/day to monkeys for up to 1 year produced reversible changes in bone marrow cellularity, a slight decrease in myeloid/erythroid cell ratio and thymic atrophy. Mild suppression of white blood cell count was observed at 1 mg/kg/day corresponding to approximately the same human dose based on AUC comparisons.
In vitro (bacterial mutation, human lymphocytes, mouse lymphoma, Syrian Hamster Embryo cell transformation) and in vivo (rat micronucleus) mutagenicity studies revealed no drug related effects at either the gene or chromosomal level. Carcinogenicity studies with lenalidomide have not been conducted.
Developmental toxicity studies were previously conducted in rabbits. In these studies, rabbits were administered 3, 10 and 20 mg/kg/day orally. An absence of the intermediate lobe of the lung was observed at 10 and 20 mg/kg/day with dose dependence and displaced kidneys were observed at 20 mg/kg/day. Although it was observed at maternotoxic levels they may be attributable to a direct effect. Soft tissue and skeletal variations in the foetuses were also observed at 10 and 20 mg/kg/day.
6. Pharmaceutical particulars
6.1 List of excipients
Capsule contents
Lactose, anhydrous
Cellulose, microcrystalline
Croscarmellose sodium
Magnesium stearate
Capsule shell
Gelatin
Titanium dioxide (E171)
Indigo carmine (E132)
Yellow iron oxide (E172)
Printing ink
Shellac
Propylene glycol
Black iron oxide (E172)
Potassium hydroxide
6.2 Incompatibilities
Not applicable.
6.3 Shelf life
3 years.
6.4 Special precautions for storage
This medicinal product does not require any special storage conditions.
6.5 Nature and contents of container
Polyvinylchloride (PVC) / Polychlorotrifluoroethylene (PCTFE) / Aluminium foil blisters containing 7 hard capsules.
Pack size of 21 capsules.
6.6 Special precautions for disposal and other handling
Any unused product or waste material should be disposed of in accordance with local requirements.
7. Marketing authorisation holder
Celgene Europe Limited
1 Longwalk Road
Stockley Park
Uxbridge
UB11 1DB
United Kingdom
8. Marketing authorisation number(s)
EU/1/07/391/002
9. Date of first authorisation/renewal of the authorisation
Date of first authorisation: 14 June 2007
Date of latest renewal: 14 June 2012
10. Date of revision of the text
17/03/2015
Detailed information on this medicinal product is available on the website of the European Medicines Agency: http://www.ema.europa.eu/.
来那度胺扩大用于复发性套细胞淋巴瘤
Celgene公司于 6月5日披露,美国FDA已批准该公司Revlimid(lenalidomide capsules,来那度胺胶囊)的补充药申请。新获准的适应症为先前已经使用过两种药物,包括bortezomib(硼替佐米)治疗后复发或病情恶化的套细胞淋巴瘤(MCL)。
套细胞淋巴瘤患者治疗仍然存在着巨大的未能满足的需求。Revlimid的获准为患者提供了一种新的选择,Revlimid是治疗淋巴瘤首个口服药物。”
Revlimid的获准建立在一项名为MCL-001的II期-多中心-单臂-开放标签研究基础上,这项研究目的是评估Revlimid对于之前接受利妥昔单抗、环磷酰胺、一种蒽环类(或米托蒽醌)和硼替佐米单独或联合使用治疗134名MCL患者的结果。患者必须是有病史记载难治性的(定义为用硼替佐米或含硼替佐米治疗方案,在治疗过程中没有任何应答——部分缓解或好转),或复发性疾病(定义为用硼替佐米或含有硼替佐米方案治疗后一年内恶化)。肌酐清除率≥60mL/min的患者每28天分别给予Revlimid每日一次25毫克,共21天。肌酸酐清除率≥30ml/min和<60mL/min的患者每28天Revlimid的剂量为10mg,每天一次共21天。
在这项研究中,主要终点为根据国际研讨会淋巴瘤疗效标准修改版本由一个独立的评审委员会放射扫描检查总应答率——26%(34/133)(95%CI为18.4,33.9),完全应答率(CR / CRU)为7%(9/133)(95%CI为3.1,12.5)。应答持续时间中位数为16.6个月(95%CI,7.7%,26.7)。
Revlimid是沙利度胺(thalidomide)的类似物,在怀孕期间禁止使用,否则可能导致出生缺陷或胎儿死亡。它的处方配发受到严格限制。Revlimid可能会导致明显的白细胞和血小板减少。Revlimid治疗的患者中有深静脉血栓和肺栓塞发生。过敏反应,包括致命性的过敏、血管神经性水肿、史蒂文斯-约翰逊综合征、中毒性表皮坏死松解症在以与Revlimid治疗的患者已有报道。有报道在Revlimid治疗期间出现肿瘤溶解综合征,包括死亡。在研究使用Revlimid治疗慢性淋巴细胞白血病和淋巴瘤过程中发生严重的肿瘤耀斑反应病例。以Revlimid与地塞米松(dexamethasone)联合治疗的患者中发现有肝衰竭,包括死亡病例。在接受Revlimid的多发性骨髓瘤患者观察对照试验中第二原发恶性肿瘤的发生率较高。The most common grade 3/4 adverse reactions reported in ≥5% of patients were neutropenia (43%), thrombocytopenia (28%), anemia (11%), pneumonia (9%), fatigue (7%), leukopenia (7%), febrile neutropenia (6%), diarrhea (6%), and dyspnea (6%).最常见的3/4级不良反应发生率≥5%,有中性粒细胞减少(43%)、血小板减少(28%)、贫血(11%)、肺炎(9%)、疲劳(7%)、白细胞减少(7 %)、发热性中性粒细胞减少(6%)、腹泻(6%)、呼吸困难(6%)。
此前Revlimid已获准用于治疗多发性骨髓瘤、骨髓增生异常综合征.
Revlimid 2.5 mg 5mg 7.5mg 10mg 15mg 20mg 25mg hard capsules
Revlimid 2.5mg(http://www.medicines.org.uk/emc/medicine/19841
Revlimid 5mg(http://www.medicines.org.uk/emc/medicine/29492
Revlimid 7.5mg(http://www.medicines.org.uk/emc/medicine/29478
Revlimid 10mg(http://www.medicines.org.uk/emc/medicine/29491)
Revlimid 15mg(http://www.medicines.org.uk/emc/medicine/29479)
Revlimid 20 mg hard capsules(http://www.medicines.org.uk/emc/medicine/30048
Revlimid 25mg(http://www.medicines.org.uk/emc/medicine/29490
http://www.oneyao.net

------------------------------------------------------
产地国家:德国
原产地英文商品名:
Revlimid 10mg/capsules  21capsules/box
原产地英文药品名:
lenalidomide 
中文参考商品译名:
瑞复美胶囊 10毫克/胶囊 21胶囊/盒
中文参考药品译名:
来那度胺
生产厂家中文参考译名:
细胞基因公司
生产厂家英文名:
Celgene
------------------------------------------------------
产地国家:德国
原产地英文商品名:
Revlimid 15mg/capsules 21capsules/box
原产地英文药品名:
lenalidomide 
中文参考商品译名:
瑞复美胶囊  15毫克/胶囊 21胶囊/盒
中文参考药品译名:
来那度胺
生产厂家中文参考译名:
细胞基因公司
生产厂家英文名:
Celgene
------------------------------------------------------
产地国家:德国
原产地英文商品名:
Revlimid 25mg/capsules  21capsules/box
原产地英文药品名:
lenalidomide 
中文参考商品译名:
瑞复美胶囊 25毫克/胶囊 21胶囊/盒
中文参考药品译名:
来那度胺
生产厂家中文参考译名:
细胞基因公司
生产厂家英文名:
Celgene

责任编辑:admin


相关文章
Revlimid Capsules(Lenalidomide Hydrate)来那度胺胶囊
Revlimid(Lenalidomide)capsules
雷利米得胶囊REVLIMID(来那度胺 LENALIDOMIDE)
来那度胺胶囊Revlimid(Lenalidomide)
Revlimid(来那度胺胶囊)为骨髓增生异常综合症的首选药
来那度胺胶囊|Revlimid(Lenalidomide)
雷纳利多米得REVLIMID(LENALIDOMIDE,雷利米得,来那度胺)
多发性骨髓瘤治疗药Revlimid在欧洲批准上市
Revlimid(Lenalidomide) 用于治疗多发性骨髓瘤能降低复发
来那度胺(Lenalidomide/Revlimid)FDA批准用于治疗骨髓增生异常综合征
 

最新文章

更多

· COTELLIC(COBIMETINIB)...
· Darzalex(daratumumab)...
· TAGRISSO(osimertinib t...
· ZYKADIA(CERITINIB, LDK...
· YONDELIS(trabectedin f...
· Nasea OD Tablets(Ramo...
· DOXIL(doxorubicin hydr...
· KYTRIL Injection(盐酸...
· NEULASTA(非格司亭注射...
· KEYTRUDA(派姆单抗 pemb...

推荐文章

更多

· COTELLIC(COBIMETINIB)...
· Darzalex(daratumumab)...
· TAGRISSO(osimertinib t...
· ZYKADIA(CERITINIB, LDK...
· YONDELIS(trabectedin f...
· Nasea OD Tablets(Ramo...
· DOXIL(doxorubicin hydr...
· KYTRIL Injection(盐酸...
· NEULASTA(非格司亭注射...
· KEYTRUDA(派姆单抗 pemb...

热点文章

更多

· DOXIL(doxorubicin hydr...
· TAGRISSO(osimertinib t...
· Nasea OD Tablets(Ramo...
· KYTRIL Injection(盐酸...
· YONDELIS(trabectedin f...
· COTELLIC(COBIMETINIB)...
· ZYKADIA(CERITINIB, LDK...
· Darzalex(daratumumab)...