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 >> 乐伐替尼硬胶囊|LENVIMA(lenvatinib hard capsules)

乐伐替尼硬胶囊|LENVIMA(lenvatinib hard capsules)

2015-06-22 01:59:01  作者:新特药房  来源:互联网  浏览次数:119  文字大小:【】【】【
简介:新型分子靶向治疗药物Lenvima(乐伐替尼 lenvatinib)胶囊获欧盟批准2015年6月3日,卫材(Eisai)自主研发的新型口服抗癌药Lenvima(乐伐替尼 lenvatinib)胶囊获欧盟批准,用于进展性、局部晚期或转移性放射 ...

—新型分子靶向治疗药物Lenvima(乐伐替尼 lenvatinib)胶囊获欧盟批准
2015年6月3日,卫材(Eisai)自主研发的新型口服抗癌药Lenvima(乐伐替尼 lenvatinib)胶囊获欧盟批准,用于进展性、局部晚期或转移性放射性碘难治(RAI)分化型(乳头状/滤泡状/嗜酸细胞)甲状腺癌(DTC)成人患者的治疗。这也是该药继今年2月和3月连获美国、日本批准之后,攻下的全球第3大主要市场。
在美国、日本、欧盟,lenvatinib均被授予孤儿药地位并通过优先审查通道获批
Lenvima是治疗分化型甲状腺癌的首个分子靶向治疗药物,该药作为一种具有重大公共卫生利益的创新药物,将帮助解决难治性分化型甲状腺癌(DTC)群体中存在的严重未获满足的巨大医疗需求。
Lenvatinib的获批,是基于一项III期SELECT研究的积极顶线数据。该研究是一项多中心、随机、双盲、安慰剂对照研究,调查了口服lenvatinib(24mg)治疗放射性碘131抵抗的分化型甲状腺癌(RR-DTC)的疗效。数据表明,与安慰剂相比,lenvatinib使无进展生存期(PFS)得到了统计学意义的显著延长(18.3个月 vs 3.6个月,p<0.0001)。此外,lenvatinib治疗组有64.8%的患者实现肿瘤缩小(客观缓解),安慰剂组数据仅为1.5%,达到了研究的主要终点。
目前,尽管大多数类型的甲状腺癌能够治疗,而一旦恶化鲜有治疗方案可供选择。分化型甲状腺癌(DTC)是最常见的甲状腺恶性肿瘤,近年来发病率稳步上升。据美国国家癌症研究所(NCI)数据,2014年美国新增甲状腺癌病例6.3万例;在欧洲,每年新增病例5.2万。
Lenvatinib是一种口服多受体酪氨酸激酶(RTK)抑制剂,具有新颖的结合模式,除抑制参与肿瘤增殖的其他促血管生成和致癌信号通路相关RTK外,还能够选择性抑制血管内皮生长因子(VEGF)受体的激酶活性。目前,卫材也正在评估lenvatinib用于其他类型肿瘤的治疗,包括肝癌、肾细胞癌、非小细胞肺癌等,该药在日本、欧盟及其他国家的监管审查正在进行中。
Lenvima 4mg and 10mg hard capsules
1. Name of the medicinal product
LENVIMA 4 mg hard capsules
LENVIMA 10 mg hard capsules
2. Qualitative and quantitative composition
Each 4mg hard capsule contains 4 mg of lenvatinib (as mesilate).
Each 10mg hard capsule contains 10 mg of lenvatinib (as mesilate).
For the full list of excipients, see section 6.1.
3. Pharmaceutical form
Hard capsule.
4mg capsule:
A yellowish-red body and yellowish-red cap, approximately 14.3 mm in length, marked in black ink with “Є” on the cap, and “LENV 4 mg” on the body.
10mg capsule:
A yellow body and yellowish-red cap, approximately 14.3 mm in length, marked in black ink with “Є” on the cap, and “LENV 10 mg” on the body.
4. Clinical particulars
4.1 Therapeutic indications
LENVIMA is indicated for the treatment of adult patients with progressive, locally advanced or metastatic, differentiated (papillary/follicular/Hürthle cell) thyroid carcinoma (DTC), refractory to radioactive iodine (RAI).
4.2 Posology and method of administration
LENVIMA treatment should be initiated and supervised by a health care professional experienced in the use of anticancer therapies.
Posology
The recommended daily dose of lenvatinib is 24 mg taken once daily. The daily dose is to be modified as needed according to the dose/toxicity management plan (see dose adjustment section below).
If a patient misses a dose, and it cannot be taken within 12 hours, then that dose should be skipped and the next dose should be taken at the usual time of administration. Treatment should continue as long as clinical benefit is observed or until unacceptable toxicity occurs.
Dose adjustment
Management of adverse reactions may require dose interruption, adjustment, or discontinuation of lenvatinib (see section 4.4). Mild to moderate adverse reactions (e.g., Grade 1 or 2) generally do not warrant interruption of lenvatinib, unless intolerable to the patient despite optimal management. Severe (e.g., Grade 3) or intolerable adverse reactions require interruption of lenvatinib until resolution or improvement of the reaction, after which treatment should be resumed at a reduced dose as suggested in Table 1. Treatment should be discontinued in case of life-threatening reactions (e.g., Grade 4) with the exception of laboratory abnormality judged to be non-life-threatening, in which case they should be managed as severe reaction (e.g., Grade 3).
Grades are based on the National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE).
Optimal medical management for nausea, vomiting, and diarrhoea should be initiated prior to any interruption or dose reduction of lenvatinib. Gastrointestinal toxicity should be actively managed in order to reduce the risk of development of renal impairment or failure (see section 4.4, Renal failure and impairment).
Table 1 Dose modifications from recommended daily dose

Dose level

Daily dose

Number of capsules

Recommended daily dose

24 mg orally once daily

Two 10 mg capsules plus one 4 mg capsule

First dose reduction

20 mg orally once daily

Two 10 mg capsules

Second dose reduction

14 mg orally once daily

One 10 mg capsule plus one 4 mg capsule

Third dose reduction

10 mg orally once dailya

One 10 mg capsule

a: Further dose reductions should be considered on an individual patient basis as limited data are available for doses below 10 mg.

Special populations
Patients of age ≥75 years, of Asian race, with comorbidities (such as hypertension, and hepatic or renal impairment), or body weight below 60 kg appear to have reduced tolerability to lenvatinib (see section 4.8, Other special populations). All patients other than those with severe hepatic or renal impairment (see below) should initiate treatment at the recommended 24 mg dose, following which the dose should be further adjusted on the basis of individual tolerability.
Patients with hypertension
Blood pressure should be well controlled prior to treatment with lenvatinib, and should be regularly monitored during treatment (see section 4.4).
Patients with hepatic impairment
No adjustment of starting dose is required on the basis of hepatic function in patients with mild (Child-Pugh A) or moderate (Child-Pugh B) hepatic impairment. In patients with severe (Child-Pugh C) hepatic impairment, the recommended starting dose is 14 mg taken once daily. Further dose adjustments may be necessary on the basis of individual tolerability.
Patients with renal impairment
No adjustment of starting dose is required on the basis of renal function in patients with mild or moderate renal impairment. In patients with severe renal impairment, the recommended starting dose is 14 mg taken once daily. Further dose adjustments may be necessary based on individual tolerability. Patients with end‑stage renal disease were not studied, therefore the use of lenvatinib in these patients is not recommended.
Elderly population
No adjustment of starting dose is required on the basis of age. Limited data are available on use in patients aged ≥75 years.
Paediatric population
Lenvatinib must not be used in children younger than 2 years of age because of safety concerns identified in animal studies (see section 5.3). The safety and efficacy of lenvatinib in children aged 2 to <18 years have not yet been established (see section 5.1). No data are available.
Race
No adjustment of starting dose is required on the basis of race (see section 5.2). Limited data are available on use in patients from ethnic origins other than Caucasian or Asian.
Method of administration
Lenvatinib is for oral use. The capsules should be taken at about the same time each day, with or without food (see section 5.2). The capsules should be swallowed whole with water. Caregivers should not open the capsule, in order to avoid repeated exposure to the contents of the capsule.
4.3 Contraindications
Hypersensitivity to the active substance or to any of the excipients listed in section 6.1.
Breast-feeding (see section 4.6).
4.4 Special warnings and precautions for use
Hypertension
Hypertension has been reported in patients treated with lenvatinib, usually occurring early in the course of treatment (see section 4.8, Description of selected adverse reactions). Blood pressure should be well controlled prior to treatment with lenvatinib and, if patients are known to be hypertensive, they should be on a stable dose of antihypertensive therapy for at least 1 week prior to treatment with lenvatinib. The early detection and effective management of hypertension are important to minimise the need for lenvatinib dose interruptions and reductions. Antihypertensive agents should be started as soon as elevated BP is confirmed. Blood pressure should be monitored after 1 week of treatment with lenvatinib, then every 2 weeks for the first 2 months, and monthly thereafter. The choice of antihypertensive treatment should be individualized to the patient's clinical circumstances and follow standard medical practice. For previously normotensive subjects, monotherapy with one of the classes of antihypertensives should be started when elevated BP is observed. For those patients already on antihypertensive medication, the dose of the current agent may be increased, if appropriate, or one or more agents of a different class of antihypertensive should be added. For patients with hypertension and proteinuria, treatment with an angiotensin-converting enzyme inhibitor or angiotensin-II receptor antagonist is preferred. When necessary, manage hypertension as recommended in Table 2.
Table 2 Recommended management of hypertension

Blood Pressure (BP) level

Recommended action

Systolic BP ≥140 mmHg up to <160 mmHg or diastolic BP ≥90 mmHg up to <100 mmHg

Continue lenvatinib and initiate antihypertensive therapy, if not already receiving

OR

Continue lenvatinib and increase the dose of the current antihypertensive therapy or initiate additional antihypertensive therapy

 

Systolic BP ≥160 mmHg or

diastolic BP ≥100 mmHg

despite optimal antihypertensive therapy

1. Withhold lenvatinib

2. When systolic BP ≤150 mmHg, diastolic BP ≤95 mmHg, and patient has been on a stable dose of antihypertensive therapy for at least 48 hours, resume lenvatinib at a reduced dose (see section 4.2)

 

Life-threatening consequences

(malignant hypertension, neurological deficit, or hypertensive crisis)

 

Urgent intervention is indicated. Discontinue lenvatinib and institute appropriate medical management.

Women of childbearing potential
Women of childbearing potential must use highly effective contraception while taking lenvatinib and for one month after stopping treatment (see section 4.6). It is currently unknown if lenvatinib increases the risk of thromboembolic events when combined with oral contraceptives.
Proteinuria
Proteinuria has been reported in patients treated with lenvatinib, usually occurring early in the course of treatment (see section 4.8, Description of selected adverse reactions). Urine protein should be monitored regularly. If urine dipstick proteinuria ≥2+ is detected, dose interruptions, adjustments, or discontinuation may be necessary (see section 4.2). LENVIMA should be discontinued in the event of nephrotic syndrome.
Renal failure and impairment
Renal impairment and renal failure have been reported in patients treated with lenvatinib (see section 4.8). The primary risk factor identified was dehydration and/or hypovolemia due to gastrointestinal toxicity. Gastrointestinal toxicity should be actively managed in order to reduce the risk of development of renal impairment or renal failure. Dose interruptions, adjustments, or discontinuation may be necessary (see section 4.2).
If patients have severe renal impairment, the initial dose of lenvatinib should be adjusted (see sections 4.2 and 5.2).
Cardiac failure
Cardiac failure (<1%) and decreased left ventricular ejection fraction have been reported in patients treated with lenvatinib (see section 4.8). Patients should be monitored for clinical symptoms or signs of cardiac decompensation, as dose interruptions, adjustments, or discontinuation may be necessary (see section 4.2).
Posterior reversible encephalopathy syndrome (PRES) / Reversible posterior leucoencephalopathy syndrome (RPLS)
Posterior reversible encephalopathy syndrome (PRES, also known as RPLS), has been reported in patients treated with lenvatinib (<1%; see section 4.8). PRES is a neurological disorder which can present with headache, seizure, lethargy, confusion, altered mental function, blindness, and other visual or neurological disturbances. Mild to severe hypertension may be present. Magnetic resonance imaging is necessary to confirm the diagnosis of PRES. Appropriate measures should be taken to control blood pressure (see section 4.4, Hypertension). In patients with signs or symptoms of PRES, dose interruptions, adjustments, or discontinuation may be necessary (see section 4.2).
Hepatotoxicity
Liver-related adverse reactions most commonly reported in patients treated with lenvatinib included increases in alanine aminotransferase, increases in aspartate aminotransferase, and increases in blood bilirubin. Hepatic failure and acute hepatitis (<1%; see section 4.8, Description of selected adverse reactions) have been reported in patients treated with lenvatinib. The hepatic failure cases were generally reported in patients with progressive liver metastases. Liver function tests should be monitored before initiation of treatment, then every 2 weeks for the first 2 months and monthly thereafter during treatment. In the case of hepatotoxicity, dose interruptions, adjustments, or discontinuation may be necessary (see section 4.2).
If patients have severe hepatic impairment, the initial dose of lenvatinib should be adjusted (see sections 4.2 and 5.2).
Haemorrhage
Serious cases of haemorrhage have been reported in patients treated with lenvatinib (see section 4.8 Description of selected adverse reactions). Cases of fatal intracranial haemorrhage have been reported in some patients with brain metastases. In the case of bleeding, dose interruptions, adjustments, or discontinuation may be necessary (see section 4.2).
Arterial thromboembolisms
Arterial thromboembolisms (cerebrovascular accident, transient ischaemic attack, and myocardial infarction) have been reported in patients treated with lenvatinib (see section 4.8). Lenvatinib has not been studied in patients who have had an arterial thromboembolism within the previous 6 months, and therefore should be used with caution in such patients. A treatment decision should be made based upon an assessment of the individual patient's benefit/risk. LENVIMA should be discontinued following an arterial thrombotic event.
Gastrointestinal perforation and fistula formation
Gastrointestinal perforation or fistulae have been reported in patients treated with lenvatinib (see section 4.8). In most cases, gastrointestinal perforation and fistulae occurred in patients with risk factors such as prior surgery or radiotherapy. In the case of a gastrointestinal perforation or fistula, dose interruptions, adjustments, or discontinuation may be necessary (see section 4.2).
QT interval prolongation
QT/QTc interval prolongation has been reported at a higher incidence in patients treated with lenvatinib than in patients treated with placebo (see section 4.8). Electrocardiograms should be monitored in all patients with a special attention for those with congenital long QT syndrome, congestive heart failure, bradyarrhythmias, and those taking medicinal products known to prolong the QT interval, including Class Ia and III antiarrhythmics. Electrolyte disturbances such as hypokalaemia, hypocalcaemia, or hypomagnesaemia increase the risk of QT prolongation, therefore electrolyte abnormalities should be monitored and corrected in all patients before starting treatment. Periodic monitoring of ECG and electrolytes (magnesium, potassium and calcium) should be considered during treatment.
Impairment of thyroid stimulating hormone suppression
Lenvatinib impairs exogenous thyroid suppression (see section 4.8, Description of selected adverse reactions). Thyroid stimulating hormone (TSH) levels should be monitored on a regular basis and thyroid hormone administration should be adjusted to reach appropriate TSH levels, according to the patient's therapeutic target.
Special populations
Limited data are available for patients of ethnic origin other than Caucasian or Asian, and in patients aged ≥75 years. Lenvatinib should be used with caution in such patients, given the reduced tolerability of lenvatinib in Asian and elderly patients (see section 4.8, Other special populations).
There are no data on the use of lenvatinib immediately following sorafenib or other anticancer treatments and there may be a potential risk for additive toxicities unless there is an adequate washout period between treatments. The minimal washout period in clinical trials was of 4 weeks.
4.5 Interaction with other medicinal products and other forms of interaction
Effect of other medicinal products on lenvatinib
Chemotherapeutic agents
Concomitant administration of lenvatinib, carboplatin, and paclitaxel has no significant impact on the pharmacokinetics of any of these 3 substances.
Effect of lenvatinib on other medicinal products
No data is available that can be used to exclude the risk that lenvatinib could be an inducer of CYP3A4 or Pgp in the gastrointestinal tract. This could potentially lead to decreased exposure to oral CYP3A4/Pgp substrates. This should be considered if co-administering oral CYP3A4/Pgp substrates for which retained efficacy is very important. CYP3A4 substrates known to have a narrow therapeutic index (e.g. astemizole, terfenadine, cisapride, pimozide, quinidine, bepridil or ergot alkaloids (ergotamine, dihydroergotamine)) should therefore be administered with caution in patients receiving lenvatinib.
Oral contraceptives
It is currently unknown whether lenvatinib may reduce the effectiveness of hormonal contraceptives, and therefore women using oral hormonal contraceptives should add a barrier method (see section 4.6).
4.6 Fertility, pregnancy and lactation
Women of childbearing potential
Women of childbearing potential should avoid becoming pregnant and use highly effective contraception while on treatment with lenvatinib and for at least one month after finishing treatment. It is currently unknown whether lenvatinib may reduce the effectiveness of hormonal contraceptives, and therefore women using oral hormonal contraceptives should add a barrier method.
Pregnancy
There are no data on the use of lenvatinib in pregnant women. Lenvatinib was embryotoxic and teratogenic when administered to rats and rabbits (see section 5.3).
Lenvatinib should not be used during pregnancy unless clearly necessary and after a careful consideration of the needs of the mother and the risk to the foetus.
Breast-feeding
It is not known whether lenvatinib is excreted in human milk. Lenvatinib and its metabolites are excreted in rat milk (see section 5.3). A risk to newborns or infants cannot be excluded and, therefore, lenvatinib is contraindicated during breast-feeding (see section 4.3).
Fertility
Effects in humans are unknown. However, testicular and ovarian toxicity has been observed in rats, dogs, and monkeys (see section 5.3).
4.7 Effects on ability to drive and use machines
Lenvatinib has a minor influence on the ability to drive and use machines, due to undesirable effects such as fatigue and dizziness. Patients who experience these symptoms should use caution when driving or operating machines.
4.8 Undesirable effects
Summary of the safety profile
The most frequently reported adverse reactions (occurring in ≥30% of patients) are hypertension (68.6%), diarrhoea (62.8%), decreased appetite (51.5%), weight decreased (49.1%), fatigue (45.8%), nausea (44.5%), proteinuria 36.9%), stomatitis (35.8%), vomiting (34.5%), dysphonia (34.1%), headache (34.1%), and palmar-plantar erythrodysaesthesia syndrome (PPE) (32.7%). Hypertension and proteinuria tend to occur early during lenvatinib treatment (see section 4.8, Description of selected adverse reactions). The majority of Grade 3 to 4 adverse reactions occurred during the first 6 months of treatment except for diarrhoea, which occurred throughout treatment, and weight loss, which tended to be cumulative over time.
The most important serious adverse reactions are renal failure and impairment (2.4%), cardiac failure (0.7%), intracranial tumor haemorrhage (0.7%), PRES / RPLS (0.2%), hepatic failure (0.2%), and arterial thromboembolisms (cerebrovascular accident (1.1%), transient ischaemic attack (0.7%), and myocardial infarction (0.9%).
In 452 patients with RAI-refractory DTC, dose reduction and discontinuation were the actions taken for an adverse reaction in 63.1% and 19.5% of patients, respectively. Adverse reactions that most commonly led to dose reductions (in ≥5% of patients) were hypertension, proteinuria, diarrhoea, fatigue, PPE, weight decreased, and decreased appetite. Adverse reactions that most commonly led to discontinuation of lenvatinib were proteinuria, asthenia, hypertension, cerebrovascular accident, diarrhoea, and pulmonary embolism.
Tabulated list of adverse reactions
Table 3 shows the incidence rates of adverse reactions observed in clinical trials.
Frequencies are defined as:
• Very common (≥1/10)
• Common (≥1/100 to <1/10)
• Uncommon (≥1/1,000 to <1/100)
Within each frequency category, undesirable effects are presented in order of decreasing seriousness.
Table 3 Adverse reactions reported in patients in clinical trials

System Organ Class

(MedDRA terminology*)

Very Common

Common

Uncommon

Infections and infestation

Urinary tract infection

 

 

Perineal abscess

Blood and lymphatic disorders

Thrombocytopeniaa

Lymphopeniaa

Splenic infarction

Endocrine disorders

 

 

Hypothyroidism

Blood thyroid stimulating hormone increased‡

 

 

 

Metabolism and nutrition disorders

Hypocalcaemia‡

Hypokalaemia

Weight decreased

Decreased appetite

Dehydration

Hypomagnesaemiab

Hypercholesterolaemiab

 

 

Psychiatric disorders

Insomnia

 

 

 

 

Nervous system disorders

Dizziness

Headache

Dysgeusia

Cerebrovascular accident

Posterior reversible encephalopathy syndrome Monoparesis

Transient ischaemic attack

 

Cardiac disorders

 

 

Myocardial infarctionc,†

Cardiac failure

Electrocardiogram QT prolonged

Ejection fraction decreased

 

 

Vascular disorders

Haemorrhaged, †,‡

Hypertensione,‡

Hypotension

 

 

 

 

 

Respiratory, thoracic and mediastinal disorders

Dysphonia

 

Pulmonary embolism†,

 

 

Gastrointestinal disorders

Diarrhoea

Gastrointestinal and abdominal painsf

Vomiting

Nausea

Oral inflammationg

Oral painh

Constipation

Dyspepsia

Dry mouth

Anal fistula

Flatulence

 

 

Hepatobiliary disorders

 

 

Aspartate aminotransferase increased

Hypoalbuminaemia

Alanine aminotransferase increased

Blood alkaline phosphatase increased

Hepatic function abnormal

Gamma-glutamyltransferase increasedk

Blood bilirubin increased

Hepatocellular damage/hepatitisi

Skin and subcutaneous tissue disorders

Palmar-plantar erythrodysaesthesia syndrome

Rash

Alopecia

Hyperkeratosis

 

 

Musculoskeletal and connective tissue disorders

Back pain

Arthralgia

Myalgia

Pain in extremity

Musculoskeletal pain

 

 

 

 

Renal and urinary disorders

Proteinuria

Renal failure cases j, †

Renal impairment

Blood creatinine increased

Blood urea increased

 

 

General disorders and administration site conditions

Fatigue

Asthenia

Oedema peripheral

Malaise

*: Medical Dictionary for Regulatory Activities (MedDRA) version 16.1. Preferred terms have been reassigned to the SOC most relevant to the target organ.
†: Includes cases with a fatal outcome.
‡: See section 4.8 Description of selected adverse reactions for further characterisation.
The following terms have been combined:
a: Thrombocytopenia includes thrombocytopenia and platelet count decreased. Lymphopenia includes lymphopenia and lymphocyte count decreased.
b: Hypomagnesaemia includes hypomagnesaemia and blood magnesium decreased. Hypercholesterolaemia includes hypercholesterolaemia and blood cholesterol increased.
c: Myocardial infarction includes myocardial infarction and acute myocardial infarction.
d: Haemorrhage includes: epistaxis, haemoptysis, haematuria, contusion, haematochezia, gingival bleeding, petechiae, pulmonary haemorrhage, rectal haemorrhage, blood urine present, haematoma, vaginal haemorrhage, conjunctival haemorrhage, haemorrhoidal haemorrhage, intracranial tumour haemorrhage, laryngeal haemorrhage, ecchymosis, increased tendency to bruise, post procedural haemorrhage, purpura, skin haemorrhage, aneurysm ruptured, arterial haemorrhage, eye haemorrhage, gastric haemorrhage, gastroduodenitis haemorrhagic, gastrointestinal haemorrhage, haematemesis, haemorrhage, haemorrhagic stroke, melaena, metrorrhagia, nail bed bleeding, pleural haemorrhage, postmenopausal haemorrhage, proctitis haemorrhagic, renal haematoma, splenic haemorrhage, splinter haemorrhages, subarachnoid haemorrhage, tracheal haemorrhage, tumour haemorrhage.
e: Hypertension includes: hypertension, hypertensive crisis, blood pressure diastolic increased, and blood pressure increased.
f: Gastrointestinal and abdominal pain includes: abdominal discomfort, abdominal pain, abdominal pain lower, abdominal pain upper, abdominal tenderness, epigastric discomfort, and gastrointestinal pain.
g: Oral inflammation includes: aphthous stomatitis, stomatitis, glossitis, mouth ulceration, and mucosal inflammation.
h: Oral pain includes: oral pain, glossodynia, and oropharyngeal pain.
i: Hepatocellular damage and hepatitis includes: drug-induced liver injury, hepatic steatosis, and cholestatic liver injury.
j: Renal failure cases includes: acute prerenal failure, renal failure, renal failure acute, and renal tubular necrosis.
Description of selected adverse reactions
Hypertension (see section 4.4)
In the pivotal Phase 3 SELECT trial (see section 5.1), hypertension (including hypertension, hypertensive crisis, blood pressure diastolic increased, and blood pressure increased) was reported in 72.8% of lenvatinib-treated patients and 16.0% of patients in the placebo-treated group. The median time to onset in lenvatinib-treated patients was 16 days. Reactions of Grade 3 or higher (including 1 reaction of Grade 4) occurred in 44.4% of lenvatinib-treated patients compared with 3.8% of placebo-treated patients. The majority of cases recovered or resolved following dose interruption or reduction, which occurred in 13.0% and 13.4% of patients, respectively. In 1.1% of patients, hypertension led to permanent treatment discontinuation.
Proteinuria (see section 4.4)
In the pivotal Phase 3 SELECT trial (see section 5.1), proteinuria was reported in 33.7% of lenvatinib-treated patients and 3.1% of patients in the placebo-treated group. The median time to onset was 6.7 weeks. Grade 3 reactions occurred in 10.7% of lenvatinib-treated patients and none in placebo-treated patients. The majority of cases had an outcome of recovered or resolved following dose interruption or reduction, which occurred in 16.9% and 10.7% of patients, respectively. Proteinuria led to permanent treatment discontinuation in 0.8% of patients.
Hepatotoxicity (see section 4.4)
In the pivotal Phase 3 SELECT trial (see section 5.1), the most commonly reported liver-related adverse reactions were hypoalbuminaemia (9.6% lenvatinib vs. 1.5% placebo) and elevations of liver enzyme levels, including increases in alanine aminotransferase (7.7% lenvatinib vs. 0 placebo), aspartate aminotransferase (6.9% lenvatinib vs. 1.5% placebo), and blood bilirubin (1.9% lenvatinib vs. 0 placebo). The median time to onset of liver reactions in lenvatinib-treated patients was 12.1 weeks. Liver-related reactions of Grade 3 or higher (including 1 Grade 5 case of hepatic failure) occurred in 5.4% of lenvatinib-treated patients compared with 0.8% in placebo-treated patients. Liver-related reactions led to dose interruptions and reductions in 4.6% and 2.7% of patients, respectively, and to permanent discontinuation in 0.4%.
Amongst 1108 patients treated with lenvatinib, there were 3 cases (0.3%) of hepatic failure, all with a fatal outcome. One occurred in a patient with no liver metastases. There was also a case of acute hepatitis in a patient without liver metastases.
Haemorrhage (see section 4.4)
In the pivotal Phase 3 SELECT trial (see section 5.1), haemorrhage was reported in 34.9% of lenvatinib-treated patients versus 18.3% of placebo-treated patients. Reactions that occurred at an incidence of ≥0.75% above placebo were: epistaxis (11.9%), haematuria (6.5%), contusion (4.6%), gingival bleeding (2.3%), haematochezia (2.3%), rectal haemorrhage (1.5%), haematoma (1.1%), haemorrhoidal haemorrhage (1.1%), laryngeal haemorrhage (1.1%), petechiae (1.1%), and intracranial tumour haemorrhage (0.8%). When adjusted to account for the 4-fold greater duration of exposure in the lenvatinib versus the placebo arm, the following reactions occurred less frequently on lenvatinib than placebo: haemoptysis (0.05 episodes/subject-year on lenvatinib vs. 0.21 episodes/subject-year on placebo) and pulmonary haemorrhage (0.02 episodes/subject-year on lenvatinib vs. 0.09 episodes/subject-year on placebo).
The median time to first onset in lenvatinib-treated patients was 10.1 weeks. No differences between lenvatinib- and placebo-treated patients were observed in the incidences of serious reactions (3.4% vs. 3.8%), reactions leading to premature discontinuation (1.1% vs. 1.5%), or reactions leading to dose interruption (3.4% vs. 3.8%) or reduction (0.4% vs. 0).
Amongst 1108 patients treated with lenvatinib, 3 patients (0.3%) had a Grade 4 haemorrhage and 5 patients (0.5%) had a Grade 5 reaction including arterial haemorrhage, haemorrhagic stroke, intracranial tumour haemorrhage, haemoptysis and tumour haemorrhage.
Hypocalcaemia (see section 4.4, QT interval prolongation)
In the pivotal Phase 3 SELECT trial (see section 5.1), hypocalcaemia was reported in 12.6% of lenvatinib-treated patients vs. no cases in the placebo arm. The median time to first onset in lenvatinib-treated patients was 11.1 weeks. Reactions of Grade 3 or 4 severity occurred in 5.0% of lenvatinib-treated vs 0 placebo-treated patients. Most reactions resolved following supportive treatment, without dose interruption or reduction, which occurred in 1.5% and 1.1% of patients, respectively; 1 patient with Grade 4 hypocalcaemia discontinued treatment permanently.
Blood thyroid stimulating hormone increased (see section 4.4 Impairment of thyroid stimulating hormone suppression)
In the pivotal Phase 3 SELECT trial (see section 5.1), 88% of all patients had a baseline TSH level less than or equal to 0.5 mU/L. In those patients with a normal TSH at baseline, elevation of TSH level above 0.5 mU/L was observed post baseline in 57% of lenvatinib-treated patients as compared with 14% of placebo-treated patients.
Paediatric population
Clinical data are not yet available in this population (see section 4.2).
Other special populations
Elderly
Patients of age ≥75 years were more likely to experience Grade 3 to 4 hypertension, proteinuria, decreased appetite, and dehydration.
Gender
Females had a higher incidence of hypertension (including Grade 3 or 4 hypertension), proteinuria, and PPE, while males had a higher incidence of decreased ejection fraction and gastrointestinal perforation and fistula formation.
Ethnic origin
Asian patients had a higher incidence than Caucasian patients of peripheral oedema, hypertension, fatigue, PPE, proteinuria, thrombocytopenia, and blood thyroid stimulating hormone increased.
Baseline hypertension
Patients with baseline hypertension had a higher incidence of Grade 3 to 4 hypertension, proteinuria, diarrhoea, and dehydration, and experienced more serious cases of dehydration, hypotension, pulmonary embolism, malignant pleural effusion, atrial fibrillation, and GI symptoms (abdominal pain, diarrhoea, vomiting).
Hepatic impairment
Patients with baseline hepatic impairment had a higher incidence of hypertension and PPE, and a higher incidence of Grade 3 or 4 hypertension, asthenia, fatigue, and hypocalcaemia compared with patients with normal hepatic function.
Renal impairment
Patients with baseline renal impairment had a higher incidence of Grade 3 to 4 hypertension, proteinuria, fatigue, stomatitis, oedema peripheral, thrombocytopenia, dehydration, prolonged electrocardiogram QT, hypothyroidism, hyponatraemia, blood thyroid stimulating hormone increased, pneumonia compared with subjects with normal renal function. These patients also had a higher incidence of renal reactions and a trend towards a higher incidence of liver reactions.
Patients with body weight <60 kg
Patients with low body weight (<60 kg) had a higher incidence of PPE, proteinuria, of grade 3-4 hypocalcaemia and hyponatraemia, and a trend towards a higher incidence of grade 3-4 decreased appetite.
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 Yellow Card Scheme Website: www.mhra.gov.uk/yellowcard.
4.9 Overdose
The highest doses of lenvatinib studied clinically were 32 mg and 40 mg per day. Accidental medication errors resulting in single doses of 40 to 48 mg have also occurred in clinical trials. The most frequently observed adverse drug reactions at these doses were hypertension, nausea, diarrhea, fatigue, stomatitis, proteinuria, headache, and aggravation of PPE. There have also been reports of overdose with lenvatinib involving single administrations of 6 to 10 times the recommended daily dose. These cases were associated with adverse reactions consistent with the known safety profile of lenvatinib (i.e., renal and cardiac failure), or were without adverse reactions.
Symptoms and Management
There is no specific antidote for overdose with lenvatinib. In case of suspected overdose, lenvatinib should be withheld and appropriate supportive care given as required.
5. Pharmacological properties
5.1 Pharmacodynamic properties
Pharmacotherapeutic group: antineoplastic agents, protein kinase inhibitors, ATC code: L01XE29
Lenvatinib is a multikinase inhibitor which has shown mainly antiangiogenic properties in vitro and in vivo, and direct inhibition of tumour growth was also observed in in vitro models.
Mechanism of action
Lenvatinib is a receptor tyrosine kinase (RTK) inhibitor that selectively inhibits the kinase activities of vascular endothelial growth factor (VEGF) receptors VEGFR1 (FLT1), VEGFR2 (KDR), and VEGFR3 (FLT4), in addition to other proangiogenic and oncogenic pathway-related RTKs including fibroblast growth factor (FGF) receptors FGFR1, 2, 3, and 4, the platelet derived growth factor (PDGF) receptor PDGFRα, KIT, and RET.
Although not studied directly with lenvatinib, the mechanism of action (MOA) for hypertension is postulated to be mediated by the inhibition of VEGFR2 in vascular endothelial cells. Similarly, although not studied directly, the MOA for proteinuria is postulated to be mediated by downregulation of VEGFR1 and VEGFR2 in the podocytes of the glomerulus.
The mechanism of action for hypothyroidism is not fully elucidated.
Clinical efficacy
Radioiodine-refractory differentiated thyroid cancer
The SELECT study was a multicentre, randomised, double-blind, placebo-controlled trial that was conducted in 392 patients with radioiodine-refractory differentiated thyroid cancer with independent, centrally reviewed, radiographic evidence of disease progression within 12 months (+1 month window) prior to enrollment. Radioiodine-refractory was defined as one or more measurable lesions either with a lack of iodine uptake or with progression in spite of radioactive-iodine (RAI) therapy, or having a cumulative activity of RAI of >600 mCi or 22 GBq with the last dose at least 6 months prior to study entry. Randomisation was stratified by geographic region (Europe, North America, and Other), prior VEGF/VEGFR-targeted therapy (patients may have received 0 or 1 prior VEGF/VEGFR-targeted therapy), and age (≤65 years or >65 years). The main efficacy outcome measure was progression-free survival (PFS) as determined by blinded independent radiologic review using Response Evaluation Criteria in Solid Tumours (RECIST) 1.1. Secondary efficacy outcome measures included overall response rate and overall survival. Patients in the placebo arm could opt to receive lenvatinib treatment at the time of confirmed disease progression.
Eligible patients with measurable disease according to RECIST 1.1 were randomised 2:1 to receive lenvatinib 24 mg once daily (n=261) or placebo (n=131). Baseline demographics and disease characteristics were well balanced for both treatment groups. Of the 392 patients randomised, 76.3% were naïve to prior VEGF/VEGFR-targeted therapies, 49.0% were female, 49.7% were European, and the median age was 63 years. Histologically, 66.1% had a confirmed diagnosis of papillary thyroid cancer and 33.9% had follicular thyroid cancer which included Hürthle cell 14.8% and clear cell 3.8%. Metastases were present in 99% of the patients: lungs in 89.3%, lymph nodes in 51.5%, bone in 38.8%, liver in 18.1%, pleura in 16.3%, and brain in 4.1%. The majority of patients had an ECOG performance status of 0; 42.1% had a status of 1; 3.9% had a status above 1. The median cumulative RAI activity administered prior to study entry was 350 mCi (12.95 GBq).
A statistically significant prolongation in PFS was demonstrated in lenvatinib-treated patients compared with those receiving placebo (p<0.0001) (see figure 1). The positive effect on PFS was seen across the subgroups of age (above or below 65 years), sex, race, histological subtype, geographic region, and those who received 0 or 1 prior VEGF/VEGFR-targeted therapy. Following independent review confirmation of disease progression, 109 (83.2%) patients randomised to placebo had crossed over to open-label lenvatinib at the time of the primary efficacy analysis.
The objective response rate (complete response [CR] plus partial response [PR]) per independent radiological review was significantly (p<0.0001) higher in the lenvatinib-treated group (64.8%) than in the placebo-treated group (1.5%). Four (1.5%) subjects treated with lenvatinib attained a CR and 165 subjects (63.2%) had a PR, while no subjects treated with placebo had a CR and 2 (1.5%) subjects had a PR.
The median time to first dose reduction was 2.8 months. The median time to objective responsive was 2.0 (95% CI: 1.9, 3.5) months; however, of the patients who experienced a complete or partial response to lenvatinib, 70.4% were observed to develop the response on or within 30 days of being on the 24-mg dose.
An overall survival analysis was confounded by the fact that placebo-treated subjects with confirmed disease progression had the option to cross over to open-label lenvatinib. There was no statistically significant difference in overall survival between the treatment groups at the time of the primary efficacy analysis (HR=0.73; 95%CI: 0.50, 1.07, p=0.1032). The median OS had not been reached for either the lenvatinib group or the placebo crossover group.
Table 4 Efficacy results

Lenvatinib

N=261

Placebo

N=131

Progression-Free Survival (PFS)a

Number of progressions or deaths (%)

107 (41.0)

113 (86.3)

Median PFS in months (95% CI)

18.3 (15.1, NE)

3.6 (2.2, 3.7)

Hazard ratio (99% CI)b,c

0.21 (0.14, 0.31)

P-valueb

<0.0001

Patients who had received 0 prior

VEGF/VEGFR-targeted therapy (%)

195 (74.7)

104 (79.4)

Number of progressions or deaths

76

88

Median PFS in months (95% CI)

18.7 (16.4, NE)

>

3.6 (2.1, 5.3)

Hazard ratio (95% CI)b,c

0.20 (0.14, 0.27)

Patients who had received 1 prior

VEGF/VEGFR-targeted therapy (%)

66 (25.3)

27 (20.6)

Number of progressions or deaths

31

25

Median PFS in months (95% CI)

15.1 (8.8, NE)

3.6 (1.9, 3.7)

Hazard ratio (95% CI)b,c

0.22 (0.12, 0.41)

Objective Response Ratea

Number of objective responders (%)

169 (64.8)

2 (1.5)

(95% CI)

(59.0, 70.5)

(0.0, 3.6)

P-valueb

<0.0001

Number of complete responses

4

0

Number of partial responses

165

2

Median time to objective response,d months (95% CI)

2.0 (1.9, 3.5)

5.6 (1.8, 9.4)

Duration of response,d months, median (95% CI)

NE (16.8, NE)

NE (NE, NE)

Overall Survival

Number of deaths (%)

71 (27.2)

47 (35.9)

Median OS in months (95% CI)

NE (22.0, NE)

NE (20.3, NE)

Hazard ratio (95% CI)b, e

0.73 (0.50, 1.07)

P-value b, e

0.1032

CI, confidence interval; NE, not estimable; OS, overall survival; PFS, progression-free survival; RPSFT, rank preserving structural failure time model; VEGF/VEGFR, vascular endothelial growth factor / vascular endothelial growth factor receptor.
a: Independent radiologic review.
b: Stratified by region (Europe vs. North America vs. Other), age group (≤65 year vs >65 years), and previous VEGF/VEGFR-targeted therapy (0 vs. 1).
c: Estimated with Cox proportional hazard model.
d: Estimated using the Kaplan-Meier method; the 95% CI was constructed with a generalised Brookmeyer and Crowley method in patients with a best overall response of complete response or partial response.
e: Not adjusted for crossover effect.
Figure 1 Kaplan-Meier Plot of Progression-Free Survival
CI, confidence interval; NE, not estimable.
QT interval prolongation
A single 32-mg dose of lenvatinib did not prolong the QT/QTc interval based on results from a thorough QT study in healthy volunteers; however, QT/QTc interval prolongation has been reported at a higher incidence in patients treated with lenvatinib than in patients treated with placebo (see sections 4.4 and 4.8).
Paediatric population
The European Medicines Agency (EMA) has deferred the obligation to submit the results of a study with lenvatinib in one or more subsets of the paediatric population in the treatment of radioiodine-refractory differentiated thyroid cancer.
5.2 Pharmacokinetic properties
Pharmacokinetic parameters of lenvatinib have been studied in healthy adult subjects, adult subjects with hepatic impairment, renal impairment, and solid tumors.
Absorption
Lenvatinib is rapidly absorbed after oral administration with tmax typically observed from 1 to 4 hours postdose. Food does not affect the extent of absorption, but slows the rate of absorption. When administered with food to healthy subjects, peak plasma concentrations are delayed by 2 hours. Absolute bioavailability has not been determined in humans; however, data from a mass-balance study suggests that it is in the order of 85%. Lenvatinib exhibited good oral bioavailability in dogs (70.4%) and monkeys (78.4%).
Distribution
In vitro binding of lenvatinib to human plasma proteins is high and ranged from 98% to 99% (0.3 ‑ 30 μg/mL, mesilate). This binding was mainly to albumin with minor binding to α1-acid glycoprotein and γ‑globulin.
In vitro, the lenvatinib blood-to-plasma concentration ratio ranged from 0.589 to 0.608 (0.1 – 10 μg/mL, mesyilate).
Lenvatinib is a substrate for P-gp and BCRP. Lenvatinib is not a substrate for OAT1, OAT3, OATP1B1, OATP1B3, OCT1, OCT2, or the BSEP.
In patients, the median apparent volume of distribution (Vz/F) of the first dose ranged from 50.5 L to 92 L and was generally consistent across the dose groups from 3.2 mg to 32 mg. The analogous median apparent volume of distribution at steady-state (Vz/Fss) was also generally consistent and ranged from 43.2 L to 121 L.
Biotransformation
In vitro, cytochrome P450 3A4 was demonstrated as the predominant (>80%) isoform involved in the P450‑mediated metabolism of lenvatinib. However, in vivo data indicated that non-P450-mediated pathways contributed to a significant portion of the overall metabolism of lenvatinib. Consequently, in vivo, inducers and inhibitors of CYP 3A4 had a minimal effect on lenvatinib exposure (see section 4.5).
In human liver microsomes, the demethylated form of lenvatinib (M2) was identified as the main metabolite. M2' and M3', the major metabolites in human faeces, were formed from M2 and lenvatinib, respectively, by aldehyde oxidase.
In plasma samples collected up to 24 hours after administration, lenvatinib constituted 97% of the radioactivity in plasma radiochromatograms while the M2 metabolite accounted for an additional 2.5%. Based on AUC(0 – inf), lenvatinib accounted for 60% and 64% of the total radioactivity in plasma and blood, respectively.
Data from a human mass balance/excretion study indicate lenvatinib is extensively metabolised in humans. The main metabolic pathways in humans were identified as oxidation by aldehyde oxidase, demethylation via CYP3A4, glutathione conjugation with elimination of the O-aryl group (chlorbenzyl moiety), and combinations of these pathways followed by further biotransformations (e.g., glucuronidation, hydrolysis of the glutathione moiety, degradation of the cysteine moiety, and intramolecular rearrangement of the cysteinylglycine and cysteine conjugates with subsequent dimerisation). These in vivo metabolic routes align with the data provided in the in vitro studies using human biomaterials.
In vitro transporter studies
For the following transporters, clinically relevant inhibition was excluded based on a cutoff of IC50> 50 × Cmax,unbound.
Lenvatinib showed minimal or no inhibitory activities toward P-gp-mediated and BCRP-mediated transport activities. Similarly, no induction of P-gp mRNA expression was observed .
Lenvatinib showed minimal or no inhibitory effect on OATP1B3. In human liver cytosol, lenvatinib did not inhibit aldehyde oxidase activity.
Elimination
Plasma concentrations decline bi-exponentially following Cmax. The mean terminal exponential half-life of lenvatinib is approximately 28 hours.
Following administration of radiolabelled lenvatinib to 6 patients with solid tumours, approximately two-thirds and one-fourth of the radiolabel were eliminated in the faeces and urine, respectively. The M3 metabolite was the predominant analyte in excreta (~17% of the dose), followed by M2' (~11% of the dose) and M2 (~4.4 of the dose).
Linearity/non-linearity
Dose proportionality and accumulation
In patients with solid tumours administered single and multiple doses of lenvatinib once daily, exposure to lenvatinib (Cmax and AUC) increased in direct proportion to the administered dose over the range of 3.2 to 32 mg once-daily.
Lenvatinib displays minimimal accumulation at steady state. Over this range, the median accumulation index (Rac) ranged from 0.96 (20 mg) to 1.54 (6.4 mg).
Special populations
Hepatic impairment
The pharmacokinetics of lenvatinib following a single 10-mg dose were evaluated in 6 subjects each with mild and moderate hepatic impairment (Child-Pugh A and Child-Pugh B, respectively). A 5-mg dose was evaluated in 6 subjects with severe hepatic impairment (Child-Pugh C). Eight healthy, demographically matched subjects served as controls and received a 10-mg dose. The median half-life was comparable in subjects with mild, moderate, and severe hepatic impairment as well as those with normal hepatic function and ranged from 26 hours to 31 hours. The percentage of the dose of lenvatinib excreted in urine was low in all cohorts (<2.16% across treatment cohorts).
Lenvatinib exposure, based on dose-adjusted AUC0-t and AUC0-inf data, was 119%, 107%, and 180% of normal for subjects with mild, moderate, and severe hepatic impairment, respectively. It is unknown whether there is a change in the plasma protein binding in hepatically impaired subjects. See section 4.2 for dosing recommendation.
Renal impairment
The pharmacokinetics of lenvatinib following a single 24-mg dose were evaluated in 6 subjects each with mild, moderate, and severe renal impairment, and compared with 8 healthy, demographically matched subjects. Subjects with end-stage renal disease were not studied.
Lenvatinib exposure, based on AUC0-inf data, was 101%, 90%, and 122% of normal for subjects with mild, moderate, and severe hepatic impairment, respectively. It is unknown whether there is a change in the plasma protein binding in renally impaired subjects. See section 4.2 for dosing recommendation.
Age, sex, weight, race
Based on a population pharmacokinetic analysis of patients receiving up to 24 mg lenvatinib once daily, age, sex, weight, and race (Japanese vs. other, Caucasian vs. other) had no significant effects on clearance (see section 4.2).
Paediatric Population
Paediatric patients have not been studied.
5.3 Preclinical safety data
In the repeated-dose toxicity studies (up to 39 weeks), lenvatinib caused toxicologic changes in various organs and tissues related to the expected pharmacologic effects of lenvatinib including glomerulopathy, testicular hypocellularity, ovarian follicular atresia, gastrointestinal changes, bone changes, changes to the adrenals (rats and dogs), and arterial (arterial fibrinoid necrosis, medial degeneration, or haemorrhage) lesions in rats, dogs, and cynomolgus monkeys. Elevated transaminase levels asociated with signs of hepatotoxicity, were also observed in rats, dogs and monkeys. Reversibility of the toxicologic changes was observed at the end of a 4-week recovery period in all animal species investigated.
Genotoxicity
Lenvatinib was not genotoxic.
Carcinogenicity studies have not been conducted with lenvatinib.
Reproductive and developmental toxicity
No specific studies with lenvatinib have been conducted in animals to evaluate the effect on fertility. However, testicular (hypocellularity of the seminiferous epithelium)and ovarian changes (follicular atresia) were observed in repeated-dose toxicity studies in animals at exposures 11 to 15 times (rat) or 0.6 to 7 times (monkey) the anticipated clinical exposure (based on AUC) at the maximum tolerated human dose. These findings were reversible at the end of a 4-week recovery period.
Administration of lenvatinib during organogenesis resulted in embryolethality and teratogenicity in rats (foetal external and skeletal anomalies) at exposures below the clinical exposure (based on AUC) at the maximum tolerated human dose, and rabbits (foetal external, visceral or skeletal anomalies) based on body surface area; mg/m2 at the maximum tolerated human dose. These findings indicate that lenvatinib has a teratogenic potential, likely related to the pharmacologic activity of lenvatinib as an antiangiogenic agent.
Lenvatinib and its metabolites are excreted in rat milk.
Juvenile animal toxicity studies
Mortality was the dose-limiting toxicity in juvenile rats in which dosing was initiated on postnatal day (PND) 7 or PND21 and was observed at exposures that were respectively 125- or 12-fold lower compared with the exposure at which mortality was observed in adult rats, suggesting an increasing sensitivity to toxicity with decreasing age. Therefore mortality may be attributed to complications related to primary duodenal lesions with possible contribution from additional toxicities in immature target organs.
The toxicity of lenvatinib was more prominent in younger rats (dosing initiated on PND7) compared with those with dosing initiated on PND21 and mortality and some toxicities were observed earlier in the juvenile rats at 10 mg/kg compared with adult rats administered the same dose level. Growth retardation, secondary delay of physical development, and lesions attributable to pharmacologic effects (incisors, femur [epiphyseal growth plate], kidneys, adrenals, and duodenum) were also observed in juvenile rats.
6. Pharmaceutical particulars
6.1 List of excipients
Capsule contents
Calcium carbonate
Mannitol
Microcrystalline cellulose
Hydroxypropylcellulose
Low-substituted hydroxypropylcellulose
Talc
Capsule shell
Hypromellose
Titanium dioxide (E171)
Yellow iron oxide (E172)
Red iron oxide (E172)
Printing ink
Shellac
Black iron oxide (E172)
Potassium hydroxide
Propylene glycol
6.2 Incompatibilities
Not applicable.
6.3 Shelf life
3 years.
6.4 Special precautions for storage
Do not store above 25°C. Store in the original blister in order to protect from moisture.
6.5 Nature and contents of container
Polyamide/Aluminium/PVC/Aluminium blisters containing 10 capsules. Each carton contains 30 capsules
6.6 Special precautions for disposal and other handling
Any unused medicinal product or waste material should be disposed of in accordance with local requirements.
Caregivers should not open the capsule, in order to avoid repeated exposure to the contents of the capsule.
7. Marketing authorisation holder
Eisai Europe Ltd.
European Knowledge Centre
Mosquito Way
Hatfield
Herts AL10 9SN
United Kingdom
8. Marketing authorisation number(s)
4mg capsules: EU/1/15/1002/001
10mg capsules: EU/1/15/1002/002
9. Date of first authorisation/renewal of the authorisation
28th May 2015
10. Date of revision of the text
28th May 2015
Detailed information on this medicinal product is available on the website of the European Medicines Agency http://www.ema.europa.eu.
LENVIMA(Lenvatinib Mesilate)包装规格:
美国上市包装
10毫克*30胶囊
14毫克*30胶囊
20毫克*30胶囊
24毫克*30胶囊


http://www.oneyao.net/article/2015/0520/article_35421.html
欧洲上市包装
4毫克*30胶囊
10毫克*30胶囊


http://www.medicines.org.uk/emc/medicine/30412
日本包装上市包装
4mg
20胶囊(PTP10C×2)


10毫克
20胶囊(PTP10C×2


http://www.kegg.jp/medicus-bin/japic_med?japic_code=00065427

责任编辑:admin


相关文章
DARZALEX(daratumumab solution for infusion)
强生抗癌药Darzalex获欧盟批准用于治疗难治性和复发性多发性骨髓瘤
FDA批准Vaxchora为预防成年人霍乱的疫苗
Netspot(gallium Ga 68 dotatate)注射剂
阿特朱单抗注射液|TECENTRIQ(atezolizumab injection)
FDA批准新诊断影像剂Netspot检测罕见神经内分泌肿瘤
TECENTRIQ(atezolizumab injection)
Tecentriq(atezolizumab)注射液
Tecentriq(atezolizumab)注射液为首个治疗膀胱癌
FDA批准Tecentriq(atezolizumab)为治疗膀胱癌新靶向药
Lenvima(lenvatinib/E7080)capsules
 

最新文章

更多

· Empliciti powder solut...
· DARZALEX(daratumumab s...
· 左旋亚叶酸钙注射剂|FUS...
· CABOMETYX(cabozantinib...
· Portrazza(necitumumab...
· Axumin(Fluciclovine F ...
· 阿特朱单抗注射液|TECEN...
· Imbruvica(ibrutinib c...
· G-LASTA Subcutaneous I...
· Venetoclax(Venclexta)T...

推荐文章

更多

· Empliciti powder solut...
· DARZALEX(daratumumab s...
· 左旋亚叶酸钙注射剂|FUS...
· CABOMETYX(cabozantinib...
· Portrazza(necitumumab...
· Axumin(Fluciclovine F ...
· 阿特朱单抗注射液|TECEN...
· Imbruvica(ibrutinib c...
· G-LASTA Subcutaneous I...
· Venetoclax(Venclexta)T...

热点文章

更多

· CABOMETYX(cabozantinib...
· Empliciti powder solut...
· Portrazza(necitumumab...
· 左旋亚叶酸钙注射剂|FUS...
· DARZALEX(daratumumab s...