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 >> Gilotrif(Afatinib filmcoated tablets)

Gilotrif(Afatinib filmcoated tablets)

2015-04-14 13:14:17  作者:新特药房  来源:互联网  浏览次数:457  文字大小:【】【】【
简介:英文药名:Gilotrif(Afatinib filmcoated tablets) 中文药名:阿法替尼片 生产厂家:德国勃林格殷格翰公司药品介绍【中文名】阿法替尼片afatinib【商品名】Gilotrif【英文名】BIBW2992【适应证】适 ...

英文药名:Gilotrif(Afatinib filmcoated tablets)

中文药名:阿法替尼片

生产厂家:德国勃林格殷格翰
药品介绍
【中文名】阿法替尼片afatinib
【商品名】Gilotrif
【英文名】BIBW2992
【适应证】适用于晚期非小细胞肺癌(NSCLC)及HER2阳性的晚期乳腺癌患者。
【用法用量】BIBW2992有效剂量是每天20mg,标准剂量为40mg。应空腹服用阿法替尼所有剂量至少进食前1小时或后2小时。
阿法替尼由勃林格殷格翰公司研制开发:2013年7月在美国以Gilotrif为商品名获得批准,2013年9月在欧盟获得批准上市。
Gilotrif产品优势
阿法替尼是一种新型、针对ErbB家族的不可逆性的阻滞剂,其可以选择且有效的阻滞ErbB家族受体(EGFR,HER2[ErbB2]和ErbB4)的信号传导以及ErbB的磷酸转移。与可逆性的EGFR酪氨酸激酶抑制剂(如厄洛替尼和吉非替尼)不同,阿法替尼能与ErbB受体网络形成共价结合,不可逆的和完全的中断信号传导,这会带来持续且广谱的抗有丝分裂活性。
与可逆EGFR酪氨酸激酶抑制剂相比,阿法替尼的抗肿瘤活性更强,并且对EGFR酪氨酸激酶抑制剂的敏感性,对抑制剂耐药的细胞株的作用,以及对非小细胞肺癌异种移植物模型的作用也较强。数个随机对照研究的结果支持——在EGFR突变的患者中,应采用EGFR酪氨酸激酶抑制剂作为标准的一线治疗方案,结果显示与普通化疗相比,采用上述方案时,肿瘤对治疗的反应率高,并且无进展生存期也延长。
阿法替尼是一种口服的不可逆的ErbB家族阻滞剂,具有选择性,可有效的阻滞ErbB家族受体(EGFR,HER2[ErbB2]和ErbB4)的信号传导以及ErbB的磷酸转移;
阿法替尼能与ErbB受体网络形成共价结合,完全的中断该网络中信号传导,且作用不可逆,这会带来持续且广谱的抗有丝分裂活性;
在亚洲EGFR突变阳性的非小细胞肺癌患者中,采用阿法替尼作为一线治疗方案能显著改善患者的无进展生存期,并且该方案安全,可被患者很好的耐受,在上述患者人群中,应将阿法替尼作为一线治疗方案。
阿法替尼是一种口服的不可逆的ErbB家族阻滞剂,在EGFR突变阳性的晚期非小细胞肺癌患者的一线治疗中,与培美曲塞和顺铂相比,其能延长上述患者群体的无进展生存期。

Giotrif 20 mg/30 mg/40 mg/50 mg film-coated tablets
1. Name of the medicinal product
GIOTRIF 20 mg film-coated tablets
GIOTRIF 30 mg film-coated tablets
GIOTRIF 40 mg film-coated tablets
GIOTRIF 50 mg film-coated tablets
2. Qualitative and quantitative composition
GIOTRIF 20 mg film-coated tablets
One film-coated tablet contains 20 mg afatinib (as dimaleate).
Excipient with known effect: One film-coated tablet contains 118 mg lactose (as monohydrate).
GIOTRIF 30 mg film-coated tablets
One film-coated tablet contains 30 mg afatinib (as dimaleate).
Excipient with known effect: One film-coated tablet contains 176 mg lactose (as monohydrate).
GIOTRIF 40 mg film-coated tablets
One film-coated tablet contains 40 mg afatinib (as dimaleate).
Excipient with known effect: One film-coated tablet contains 235 mg lactose (as monohydrate).
GIOTRIF 50 mg film-coated tablets
One film-coated tablet contains 50 mg afatinib (as dimaleate).
Excipient with known effect: One film-coated tablet contains 294 mg lactose (as monohydrate).
For the full list of excipients, see section 6.1.
3. Pharmaceutical form
Film-coated tablet (tablet).
GIOTRIF 20 mg film-coated tablets
White to yellowish, round, biconvex and bevel-edged film-coated tablet debossed with the code “T20” on one side and the Boehringer Ingelheim company logo on the other.
GIOTRIF 30 mg film-coated tablets
Dark blue, round, biconvex and bevel-edged film-coated tablet debossed with the code “T30” on one side and the Boehringer Ingelheim company logo on the other.
GIOTRIF 40 mg film-coated tablets
Light blue, round, biconvex and bevel-edged film-coated tablet debossed with the code “T40” on one side and the Boehringer Ingelheim company logo on the other.
GIOTRIF 50 mg film-coated tablets
Dark blue, oval, biconvex film-coated tablet debossed with the code “T50” on one side and the Boehringer Ingelheim company logo on the other.
4. Clinical particulars
4.1 Therapeutic indications
GIOTRIF as monotherapy is indicated for the treatment of
• Epidermal Growth Factor Receptor (EGFR) TKI-naïve adult patients with locally advanced or metastatic non-small cell lung cancer (NSCLC) with activating EGFR mutation(s);
• locally advanced or metastatic NSCLC of squamous histology progressing on or after platinum-based chemotherapy (see section 5.1).
4.2 Posology and method of administration
Treatment with GIOTRIF should be initiated and supervised by a physician experienced in the use of anticancer therapies.
EGFR mutation status should be established prior to initiation of GIOTRIF therapy (see section 4.4).
Posology
The recommended dose is 40 mg once daily.
This medicinal product should be taken without food. Food should not be consumed for at least 3 hours before and at least 1 hour after taking this medicinal product (see sections 4.5 and 5.2).
GIOTRIF treatment should be continued until disease progression or until no longer tolerated by the patient (see Table 1 below).
Dose escalation
A dose escalation to a maximum of 50 mg/day may be considered in patients who tolerate a 40 mg/day starting dose (i.e. absence of diarrhoea, skin rash, stomatitis, and other adverse reactions with CTCAE Grade > 1) in the first cycle of treatment (21 days for EGFR mutation positive NSCLC and 28 days for squamous NSCLC). The dose should not be escalated in any patients with a prior dose reduction. The maximum daily dose is 50 mg.
Dose adjustment for adverse reactions
Symptomatic adverse reactions (e.g. severe/persistent diarrhoea or skin related adverse reactions) may be successfully managed by treatment interruption and dose reductions or treatment discontinuation of GIOTRIF as outlined in Table 1 (see sections 4.4 and 4.8).
Table 1: Dose adjustment information for adverse reactions

CTCAEa Adverse reactions

Recommended dosing

Grade 1 or Grade 2

No interruption b

No dose adjustment

Grade 2 (prolonged c or intolerable) or Grade ≥ 3

Interrupt until Grade 0/1 b

Resume with dose reduction by 10 mg decrements d

a NCI Common Terminology Criteria for Adverse Events
b In case of diarrhoea, anti-diarrhoeal medicinal products (e.g. loperamide) should be taken immediately and continued for persistent diarrhoea until loose bowel movements cease.
c > 48 hours of diarrhoea and/or > 7 days of rash
d If patient cannot tolerate 20 mg/day, permanent discontinuation of GIOTRIF should be considered
Interstitial Lung Disease (ILD) should be considered if a patient develops acute or worsening of respiratory symptoms in which case treatment should be interrupted pending evaluation. If ILD is diagnosed, GIOTRIF should be discontinued and appropriate treatment initiated as necessary (see section 4.4).
Missed dose
If a dose is missed, it should be taken within the same day as soon as the patient remembers. However, if the next scheduled dose is due within 8 hours then the missed dose must be skipped.
Use of P-glycoprotein (P-gp) inhibitors
If P-gp inhibitors need to be taken, they should be administered using staggered dosing, i.e. the P-gp inhibitor dose should be taken as far apart in time as possible from the GIOTRIF dose. This means preferably 6 hours (for P-gp inhibitors dosed twice daily) or 12 hours (for P-gp inhibitors dosed once daily) apart from GIOTRIF (see section 4.5).
Patients with renal impairment
The safety, pharmacokinetics and efficacy of this medicinal product have not been studied in a dedicated trial in patients with renal impairment. Adjustments to the starting dose are not necessary in patients with mild or moderate renal impairment. Treatment in patients with severely impaired renal function (< 30 mL/min creatinine clearance) is not recommended (see section 5.2).
Patients with hepatic impairment
Exposure to afatinib is not significantly changed in patients with mild (Child Pugh A) or moderate (Child Pugh B) hepatic impairment (see section 5.2). Adjustments to the starting dose are not necessary in patients with mild or moderate hepatic impairment. This medicinal product has not been studied in patients with severe (Child Pugh C) hepatic impairment. Treatment in this population is not recommended (see section 4.4).
Paediatric population
There is no relevant use of GIOTRIF in the paediatric population in the indication of NSCLC. Therefore, treatment of children or adolescents with this medicinal product is not recommended.
Method of administration
This medicinal product is for oral use. The tablets should be swallowed whole with water. If swallowing of whole tablets is not possible, these can be dispersed in approximately 100 ml of noncarbonated drinking water. No other liquids should be used. The tablet should be dropped into the water without crushing it, and stirred occasionally for up to 15 min until it is broken up into very small particles. The dispersion should be consumed immediately. The glass should be rinsed with approximately 100 ml of water which should also be consumed. The dispersion can also be administered through a gastric tube.
4.3 Contraindications
Hypersensitivity to afatinib or to any of the excipients listed in section 6.1.
4.4 Special warnings and precautions for use
Assessment of EGFR mutation status
When assessing the EGFR mutation status of a patient, it is important that a well-validated and robust methodology is chosen to avoid false negative or false positive determinations.
Diarrhoea
Diarrhoea, including severe diarrhoea, has been reported during treatment with GIOTRIF (see section 4.8). Diarrhoea may result in dehydration with or without renal impairment, which in rare cases has resulted in fatal outcomes. Diarrhoea usually occurred within the first 2 weeks of treatment. Grade 3 diarrhoea most frequently occurred within the first 6 weeks of treatment.
Proactive management of diarrhoea including adequate hydration combined with anti-diarrhoeal medicinal products especially within the first 6 weeks of the treatment is important and should start at first signs of diarrhoea. Antidiarrhoeal medicinal products (e.g. loperamide) should be used and if necessary their dose should be escalated to the highest recommended approved dose. Anti-diarrhoeal medicinal products should be readily available to the patients so that treatment can be initiated at first signs of diarrhoea and continued until loose bowel movements cease for 12 hours. Patients with severe diarrhoea may require interruption and dose reduction or discontinuation of therapy with GIOTRIF (see section 4.2). Patients who become dehydrated may require administration of intravenous electrolytes and fluids.
Skin related adverse events
Rash/acne has been reported in patients treated with this medicinal product (see section 4.8). In general, rash manifests as a mild or moderate erythematous and acneiform rash, which may occur or worsen in areas exposed to sun. For patients who are exposed to sun, protective clothing, and use of sun screen is advisable. Early intervention (such as emollients, antibiotics) of dermatologic reactions can facilitate continuous GIOTRIF treatment. Patients with severe skin reactions may also require temporary interruption of therapy, dose reduction (see section 4.2), additional therapeutic intervention, and referral to a specialist with expertise in managing these dermatologic effects.
Bullous, blistering and exfoliative skin conditions have been reported including rare cases suggestive of Stevens-Johnson syndrome. Treatment with this medicinal product should be interrupted or discontinued if the patient develops severe bullous, blistering or exfoliating conditions (see section 4.8).
Female gender, lower body weight, and underlying renal impairment
Higher exposure to afatinib has been observed in female patients, patients with lower body weight and those with underlying renal impairment (see section 5.2). This could result in a higher risk of developing adverse reactions in particular diarrhoea, rash/acne and stomatitis. Closer monitoring is recommended in patients with these risk factors.
Interstitial Lung Disease (ILD)
There have been reports of ILD or ILD-like adverse reactions (such as lung infiltration, pneumonitis, acute respiratory distress syndrome, allergic alveolitis), including fatalities, in patients receiving GIOTRIF for treatment of NSCLC. ILD-like adverse reactions were reported in 0.7% of patients treated with GIOTRIF across all clinical trials (including 0.5% of patients with CTCAE Grade ≥ 3 ILD-like adverse reactions). Patients with a history of ILD have not been studied.
Careful assessment of all patients with an acute onset and/or unexplained worsening of pulmonary symptoms (dyspnoea, cough, fever) should be performed to exclude ILD. Treatment with this medicinal product should be interrupted pending investigation of these symptoms. If ILD is diagnosed, GIOTRIF should be permanently discontinued and appropriate treatment initiated as necessary (see section 4.2).
Severe hepatic impairment
Hepatic failure, including fatalities, has been reported during treatment with this medicinal product in less than 1% of patients. In these patients, confounding factors have included pre-existing liver disease and/or comorbidities associated with progression of underlying malignancy. Periodic liver function testing is recommended in patients with pre-existing liver disease. In the pivotal trials Grade 3 alanine aminotransferase (ALT) and aspartate aminotransferase (AST) elevations were observed in 2.4% (LUX-Lung-3) and 1.6% (LUX-Lung 8) of patients with normal baseline liver tests treated with 40 mg/day. In LUX-Lung-3 Grade 3 ALT/AST elevations were about 3.5 fold higher in patients with abnormal baseline liver tests. There were no Grade 3 ALT/AST elevations in patients with abnormal baseline liver tests in LUX-Lung 8 (see section 4.8). Dose interruption may become necessary in patients who experience worsening of liver function (see section 4.2). In patients who develop severe hepatic impairment while taking GIOTRIF, treatment should be discontinued.
Keratitis
Symptoms such as acute or worsening eye inflammation, lacrimation, light sensitivity, blurred vision, eye pain and/or red eye should be referred promptly to an ophthalmology specialist. If a diagnosis of ulcerative keratitis is confirmed, treatment should be interrupted or discontinued. If keratitis is diagnosed, the benefits and risks of continuing treatment should be carefully considered. This medicinal product should be used with caution in patients with a history of keratitis, ulcerative keratitis or severe dry eye. Contact lens use is also a risk factor for keratitis and ulceration (see section 4.8).
Left ventricular function
Left ventricular dysfunction has been associated with HER2 inhibition. Based on the available clinical trial data, there is no suggestion that this medicinal product causes an adverse reaction on cardiac contractility. However, this medicinal product has not been studied in patients with abnormal left ventricular ejection fraction (LVEF) or those with significant cardiac history. In patients with cardiac risk factors and those with conditions that can affect LVEF, cardiac monitoring, including an assessment of LVEF at baseline and during treatment, should be considered. In patients who develop relevant cardiac signs/symptoms during treatment, cardiac monitoring including LVEF assessment should be considered.
In patients with an ejection fraction below the institution's lower limit of normal, cardiac consultation as well as treatment interruption or discontinuation should be considered.
P-glycoprotein (P-gp) interactions
Concomitant treatment with strong inducers of P-gp may decrease exposure to afatinib (see section 4.5).
Lactose
This medicinal product contains lactose. Patients with rare hereditary conditions of galactose intolerance, the Lapp lactase deficiency or glucose-galactose malabsorption should not take this medicinal product.
4.5 Interaction with other medicinal products and other forms of interaction
Interactions with drug transport systems
Effects of P-gp and breast cancer resistance protein (BCRP) inhibitors on afatinib
In vitro studies have demonstrated that afatinib is a substrate of P-gp and BCRP. When the strong P-gp and BCRP inhibitor ritonavir (200 mg twice a day for 3 days) was administered 1 hour before a single dose of 20 mg GIOTRIF, exposure to afatinib increased by 48% (area under the curve (AUC0-∞)) and 39% (maximum plasma concentration (Cmax)). In contrast, when ritonavir was administered simultaneously or 6 hours after 40 mg GIOTRIF, the relative bioavailability of afatinib was 119% (AUC0-∞) and 104% (Cmax) and 111% (AUC0-∞) and 105% (Cmax), respectively. Therefore, it is recommended to administer strong P-gp inhibitors (including but not limited to ritonavir, cyclosporine A, ketoconazole, itraconazole, erythromycin, verapamil, quinidine, tacrolimus, nelfinavir, saquinavir, and amiodarone) using staggered dosing, preferably 6 hours or 12 hours apart from GIOTRIF (see section 4.2).
Effects of P-gp inducers on afatinib
Pre-treatment with rifampicin (600 mg once daily for 7 days), a potent inducer of P-gp, decreased the plasma exposure to afatinib by 34% (AUC0-∞) and 22% (Cmax) after administration of a single dose of 40 mg GIOTRIF. Strong P-gp inducers (including but not limited to rifampicin, carbamazepine, phenytoin, phenobarbital or St. John's wort (Hypericum perforatum)) may decrease exposure to afatinib (see section 4.4).
Effects of afatinib on P-gp substrates
Based on in vitro data, afatinib is a moderate inhibitor of P-gp. However, based on clinical data it is considered unlikely that GIOTRIF treatment will result in changes of the plasma concentrations of other P-gp substrates.
Interactions with BCRP
In vitro studies indicated that afatinib is a substrate and an inhibitor of the transporter BCRP. Afatinib may increase the bioavailability of orally administered BCRP substrates (including but not limited to rosuvastatin and sulfasalazine).
Food effect on afatinib
Co-administration of a high-fat meal with GIOTRIF resulted in a significant decrease of exposure to afatinib by about 50% in regard to Cmax and 39% in regard to AUC0-∞. This medicinal product should be administered without food (see sections 4.2 and 5.2).
4.6 Fertility, pregnancy and lactation
Women of childbearing potential
As a precautionary measure, women of childbearing potential should be advised to avoid becoming pregnant while receiving treatment with GIOTRIF. Adequate contraceptive methods should be used during therapy and for at least 1 month after the last dose.
Pregnancy
Mechanistically, all EGFR targeting medicinal products have the potential to cause foetal harm.
Animal studies with afatinib did not indicate direct or indirect harmful effects with respect to reproductive toxicity (see section 5.3). Studies in animals have shown no signs of teratogenicity up to and including maternally lethal dose levels. Adverse changes were restricted to toxic dose levels. However, systemic exposures achieved in animals were either in a similar range or below the levels observed in patients (see section 5.3).
There are no or limited amount of data from the use of this medicinal product in pregnant women. The risk for humans is thus unknown. If used during pregnancy or if the patient becomes pregnant while or after receiving GIOTRIF, she should be informed of the potential hazard to the foetus.
Breast-feeding
Available pharmacokinetic data in animals have shown excretion of afatinib in milk (see section 5.3). Based on this, it is likely that afatinib is excreted in human milk. A risk to the breast-feeding child cannot be excluded. Mothers should be advised against breast-feeding while receiving this medicinal product.
Fertility
Fertility studies in humans have not been performed with afatinib. Available non-clinical toxicology data have shown effects on reproductive organs at higher doses. Therefore, an adverse effect of this medicinal product on human fertility cannot be excluded.
4.7 Effects on ability to drive and use machines
GIOTRIF has minor influence on the ability to drive and use machines. During treatment, ocular adverse reactions (conjunctivitis, dry eye, keratitis) have been reported in some patients (see section 4.8) which may affect patients ability to drive or use machines.
4.8 Undesirable effects
Summary of the safety profile
The types of adverse reactions (ADRs) were generally associated with the EGFR inhibitory mode of action of afatinib. The summary of all ADRs is shown in Table 2. The most frequent ADRs were diarrhoea and skin related adverse events (see section 4.4) as well as stomatitis and paronychia (see also Table 3 and 4). Overall, dose reduction (see section 4.2) led to a lower frequency of common adverse reactions.
In patients treated with once daily GIOTRIF 40 mg, dose reductions due to ADRs occurred in 57% of the patients in the LUX-Lung 3 trial and in 25% of the patients in the LUX-Lung 8 trial. Discontinuation due to ADRs diarrhoea and rash/acne was 1.3% and 0% in LUX-Lung 3 and 3.8% and 2.0% in LUX-Lung 8, respectively.
ILD-like adverse reactions were reported in 0.7% of afatinib treated patients. Bullous, blistering and exfoliative skin conditions have been reported including rare cases suggestive of Stevens-Johnson syndrome although in these cases there were potential alternative aetiologies (see section 4.4).
Tabulated list of adverse reactions
Table 2 summarises the frequencies of ADRs from all NSCLC trials with daily GIOTRIF doses of 40 mg or 50 mg as monotherapy. The following terms are used to rank the ADRs by frequency: 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). Within each frequency grouping, adverse reactions are presented in order of decreasing seriousness.
Table 2: Summary of ADRs per frequency category

Body System

Very common

(≥1/10)

Common

(≥1/100 to <1/10)

Uncommon

(≥1/1,000 to < 1/100)

Infections and infestations

Paronychia1

Cystitis

 

Metabolism and nutrition disorders

Decreased appetite

Dehydration

Hypokalaemia

 

Nervous system disorders

 

Dysgeusia

 

Eye disorders

 

Conjunctivitis

Dry eye

Keratitis

Respiratory, thoracic and mediastinal disorders

Epistaxis

Rhinorrhoea

Interstitial lung disease

Gastrointestinal disorders

Diarrhoea

Stomatitis2

Nausea

Vomiting

Dyspepsia

Cheilitis

Pancreatitis

Hepatobiliary disorders

 

Alanine aminotransferase increased

Aspartate aminotransferase increased

 

Skin and subcutaneous tissue disorders

Rash3

Dermatitis acneiform4

Pruritus5

Dry skin6

Palmar-plantar erythrodysaesthesia syndrome

 

Musculoskeletal and connective tissue disorders

 

Muscle spasms

 

Renal and urinary disorders

 

Renal impairment/ Renal failure

 

General disorders and administration site conditions

 

Pyrexia

 

Investigations

 

Weight decreased

1 Includes Paronychia, Nail infection, Nail bed infection
2 Includes Stomatitis, Aphthous stomatitis, Mucosal inflammation, Mouth ulceration, Oral mucosa erosion, Mucosal erosion, Mucosal ulceration
3 Includes group of rash preferred terms
4 Includes Acne, Acne pustular, Dermatitis acneiform
5 Includes Pruritus, Pruritus generalised
6 Includes Dry skin, Skin chapped
Description of selected adverse reactions
Very common ADRs in GIOTRIF-treated patients occurring in at least 10% of patients in trial LUX-Lung 3 are summarised by National Cancer Institute-Common Toxicity Criteria (NCI-CTC) Grade in Table 3.
Table 3: Very common ADRs in trial LUX-Lung 3

GIOTRIF

(40 mg/day)

N=229

Pemetrexed/ Cisplatin

N=111

NCI-CTC Grade

Any Grade

3

4

Any Grade

3

4

MedDRA Preferred Term

%

%

%

%

%

%

Infections and infestations

Paronychia1

57.6

11.4

0

0

0

0

Metabolism and nutrition disorders

Decreased appetite

20.5

3.1

0

53.2

2.7

0

Respiratory, thoracic and mediastinal disorders

Epistaxis

13.1

0

0

0.9

0.9

0

Gastrointestinal disorders

Diarrhoea

Stomatitis2

Cheilitis

95.2

69.9

12.2

14.4

8.3

0

0

0.4

0

15.3

13.5

0.9

0

0.9

0

0

0

0

Skin and subcutaneous tissue disorders

Rash3

Dermatitis acneiform4

Dry skin5

Pruritus6

70.3

34.9

29.7

19.2

14

2.6

0.4

0.4

0

0

0

0

6.3

0

1.8

0.9

0

0

0

0

0

0

0

0

Investigations

Weight decreased

10.5

0

0

9.0

0

0

1 Includes Paronychia, Nail infection, Nail bed infection
2 Includes Stomatitis, Aphthous stomatitis, Mucosal inflammation, Mouth ulceration, Oral mucosa erosion, Mucosal erosion, Mucosal ulceration
3 Includes group of rash preferred terms
4 Includes Acne, Acne pustular, Dermatitis acneiform
5 Includes Dry skin, Skin chapped
6 Includes Pruritus, Pruritus generalised
Liver function test abnormalities
Liver function test abnormalities (including elevated ALT and AST) were observed in patients receiving GIOTRIF 40 mg. These elevations were mainly transient and did not lead to discontinuation. Grade 2 (> 2.5 to 5.0 times upper limit of normal (ULN)) ALT elevations occurred in < 8% of patients treated with this medicinal product. Grade 3 (> 5.0 to 20.0 times ULN) elevations occurred in <4% of patients treated with GIOTRIF (see section 4.4).
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:
United Kingdom
Yellow Card Scheme
Website: www.mhra.gov.uk/yellowcard
Ireland
HPRA Pharmacovigilance
Earlsfort Terrace
IRL - Dublin 2
Tel: +353 1 6764971
Fax: +353 1 6762517
Website: www.hpra.ie
e-mail: medsafety@hpra.ie
Malta
ADR Reporting
The Medicines Authority
Post-Licensing Directorate
203 Level 3, Rue D'Argens
GŻR-1368 Gżira
Website: www.medicinesauthority.gov.mt
e-mail: postlicensing.medicinesauthority@gov.mt
Description of selected adverse reactions
Very common ADRs in GIOTRIF-treated patients occurring in at least 10% of patients in trial LUX-Lung 8 are summarised by National Cancer Institute-Common Toxicity Criteria (NCI-CTC) Grade in Table 4.
Table 4: Very common ADRs in trial LUX-Lung 8*

GIOTRIF

(40 mg/day)

N=392

erlotinib

N=395

NCI-CTC Grade

Any Grade

3

4

Any Grade

3

4

MedDRA Preferred Term

%

%

%

%

%

%

Infections and infestations

Paronychia1

11.0

0.5

0

5.1

0.3

0

Metabolism and nutrition disorders

Decreased appetite

24.7

3.1

0

26.1

2.0

0

Gastrointestinal disorders

Diarrhoea

Stomatitis2

Nausea

74.7

30.1

20.7

9.9

4.1

1.5

0.8

0

0

41.3

10.6

16.2

3.0

0.5

1.0

0.3

0

0.3

Skin and subcutaneous tissue disorders

Rash3

Dermatitis acneiform4

60.7

14.0

5.4

1.3

0

0

56.7

18.0

8.1

2.5

0

0

* Reporting the frequency of patients with all causality AEs
1 Includes Paronychia, Nail infection, Nail bed infection
2 Includes Stomatitis, Aphthous stomatitis, Mucosal inflammation, Mouth ulceration, Oral mucosa erosion, Mucosal erosion, Mucosal ulceration
3 Includes group of rash preferred terms
4 Includes Acne, Acne pustular, Dermatitis acneiform
Liver function test abnormalities
Liver function test abnormalities (including elevated ALT and AST) were observed in patients receiving GIOTRIF 40 mg. These elevations were mainly transient and did not lead to discontinuation. Grade 2 ALT elevations occurred in 1% and Grade 3 elevations occurred in 0.8% of patients treated with GIOTRIF (see section 4.4).
4.9 Overdose
Symptoms
The highest dose of afatinib studied in a limited number of patients in Phase I clinical trials was 160 mg once daily for 3 days and 100 mg once daily for 2 weeks. The adverse reactions observed at these doses were primarily dermatological (rash/acne) and gastrointestinal events (especially diarrhoea). Overdose in 2 healthy adolescents involving the ingestion of 360 mg each of afatinib (as part of a mixed drug ingestion) was associated with adverse events of nausea, vomiting, asthenia, dizziness, headache, abdominal pain and elevated amylase (< 1.5 times ULN). Both individuals recovered from these adverse events.
Treatment
There is no specific antidote for overdose with this medicinal product. In cases of suspected overdose, GIOTRIF should be withheld and supportive care initiated.
If indicated, elimination of unabsorbed afatinib may be achieved by emesis or gastric lavage.
5. Pharmacological properties
5.1 Pharmacodynamic properties
Pharmacotherapeutic group: antineoplastic agents, protein kinase inhibitors, ATC code: L01XE13.
Mechanism of action
Afatinib is a potent and selective, irreversible ErbB Family Blocker. Afatinib covalently binds to and irreversibly blocks signalling from all homo- and heterodimers formed by the ErbB family members EGFR (ErbB1), HER2 (ErbB2), ErbB3 and ErbB4.
Pharmacodynamic effects
Aberrant ErbB signalling triggered by receptor mutations, and/or amplification, and/or receptor ligand overexpression contributes to the malignant phenotype. Mutation in EGFR defines a distinct molecular subtype of lung cancer.
In non-clinical disease models with ErbB pathway deregulation, afatinib as a single agent effectively blocks ErbB receptor signalling resulting in tumour growth inhibition or tumour regression. NSCLC tumours with common activating EGFR mutations (Del 19, L858R) and several less common EGFR mutations in exon 18 (G719X) and exon 21 (L861Q) are particularly sensitive to afatinib treatment in non-clinical and clinical settings.
Afatinib retains significant anti-tumour activity in NSCLC cell lines in vitro and/or tumour models in vivo (xenografts or transgenic models) driven by mutant EGFR isoforms known to be resistant to the reversible EGFR inhibitors erlotinib and gefitinib such as T790M or T854A. Clinically, activity in tumours harbouring the T790M mutation in exon 20 has also been shown. Limited non-clinical and/or clinical activity was observed in NSCLC tumours with insertion mutations in exon 20.
Clinical efficacy and safety
GIOTRIF in patients with Non-Small Cell Lung Cancer (NSCLC) with EGFR mutations
LUX-Lung 3
In the first-line setting, the efficacy and safety of GIOTRIF in patients with EGFR mutation-positive locally advanced or metastatic NSCLC (stage IIIB or IV) were assessed in a global, randomised, multicentre, open-label trial. Patients were screened for the presence of 29 different EGFR mutations using a polymerase chain reaction (PCR)-based method (TheraScreen®: EGFR29 Mutation Kit, Qiagen Manchester Ltd). Patients were randomised (2:1) to receive GIOTRIF 40 mg once daily or up to 6 cycles of pemetrexed/cisplatin. Among the patients randomised, 65% were female, the median age was 61 years, the baseline ECOG performance status was 0 (39%) or 1 (61%), 26% were Caucasian and 72% were Asian. 89% of patients had common EGFR mutations (Del 19 or L858R).
The primary endpoint was progression free survival (PFS) by independent review; the secondary endpoints included overall survival and objective response rate. At the time of the analysis, 14 Nov 2013, 176 patients (76.5%) in the afatinib arm and 70 patients (60.9%) in the chemotherapy arm experienced an event contributing to the PFS analysis, i.e. disease progression as determined by central independent review or death. The efficacy results are provided in Figure 1, Tables 5 and 6.
LUX-Lung 6
The efficacy and safety of GIOTRIF in Asian patients with Stage IIIB/IV EGFR mutation-positive locally advanced or metastatic adenocarcinoma of the lung was evaluated in a randomised, multicentre, open-label trial. Similar to LUX-Lung 3, patients with previously untreated NSCLC were screened for EGFR mutations using TheraScreen®: EGFR29 Mutation Kit (Qiagen Manchester Ltd). Among randomized patients, 65% were female, the median age was 58 years and all patients were of Asian ethnicity. Patients with common EGFR mutations accounted for 89% of the study population.
The primary endpoint was PFS as assessed by central independent review; secondary endpoints included OS and ORR.
Both trials demonstrated significant improvement in PFS of EGFR mutation positive patients treated with GIOTRIF compared to chemotherapy. The efficacy results are summarized in Figure 1 (LUX-Lung 3) and Tables 5 and 6 (LUX-Lung 3 and 6). Table 6 shows outcomes in the subgroups of patients with two common EGFR mutations – Del 19 and L858R.
Figure 1: Kaplan-Meier curve for PFS by independent review by treatment group in trial LUX-Lung 3 (Overall Population)


Table 5: Efficacy results of GIOTRIF vs. pemetrexed/cisplatin (LUX-Lung 3) gemcitabine/cisplatin (LUX-Lung 6) (Independent review)

LUX-Lung 3

LUX-Lung 6

GIOTRIF
 

(N=230)

Pemetrexed/ Cisplatin

(N=115)

GIOTRIF
 

(N=242)

Gemcitabine/ Cisplatin

(N=122)

Progression-free survival

Months (median)

11.2

6.9

11.0

5.6

Hazard Ratio (HR)

(95%CI)

0.58

(0.43-0.78)

0.28

(0.20-0.39)

p-value1

0.0002

<0.0001

1-year PFS Rate

48.1%

22.0%

46.7%

2.1%

Objective Response Rate (CR+PR)2

56.5%

22.6%

67.8%

23.0%

Odds Ratio (OR)

(95%CI)

4.80

(2.89-8.08)

7.57

(4.52-12.68)

p-value1

<0.0001

<0.0001

Overall Survival (OS)

Months (median)

28.2

28.2

23.1

23.5

Hazard Ratio (HR)

(95%CI)

0.88

(0.66-1.17)

0.93

(0.72-1.22)

p-value1

0.3850

0.6137

1 p-value for PFS/OS based on stratified log-rank test; p-value for Objective Response Rate based on logistic regression
2 CR=complete response; PR=partial response
Table 6: PFS and OS efficacy results of GIOTRIF vs pemetrexed/cisplatin (LUX-Lung 3) gemcitabine/cisplatin (LUX-Lung 6) in the pre-defined EGFR mutation subgroups Del 19 and L858R (Independent review)

LUX-Lung 3

LUX-Lung 6

Del19

GIOTRIF
 

(N=112)

Pemetrexed/ Cisplatin

(N=57)

GIOTRIF
 

(N=124)

Gemcitabine/ Cisplatin

(N=62)

Progression-free survival

Months (median)

13.8

5.6

13.1

5.6

Hazard Ratio (HR)

(95%CI)

0.26

(0.17-0.42)

0.20

(0.13-0.33)

p-value1

<0.0001

<0.0001

Overall Survival (OS)

Months (median)

33.3

21.1

31.4

18.4

Hazard Ratio (HR)

(95%CI)

0.54

(0.36-0.79)

0.64

(0.44-0.94)

p-value1

0.0015

0.0229

L858R

GIOTRIF
 

(N=91)

Pemetrexed/ Cisplatin

(N=47)

GIOTRIF
 

(N=92)

Gemcitabine/ Cisplatin

(N=46)

Progression-free survival

Months (median)

10.8

8.1

9.6

5.6

Hazard Ratio (HR)

(95%CI)

0.75

(0.48-1.19)

0.31

(0.19-0.52)

p-value1

0.2191

<0.0001

Overall Survival (OS)

Months (median)

27.6

40.3

19.6

24.3

Hazard Ratio (HR)

(95%CI)

1.30

(0.80-2.11)

1.22

(0.81-1.83)

p-value1

0.2919

0.3432

1 p-value for PFS/OS based on stratified log-rank test
In the pre-defined subgroup of common mutations (combined Del 19 and L858R) for GIOTRIF and chemotherapy, the median PFS was 13.6 months vs. 6.9 months (HR 0.48; 95% CI 0.35-0.66; p<0.0001; N=307) in LUX-Lung 3, and 11.0 months vs. 5.6 months (HR 0.24; 95% CI 0.17-0.35; p<0.0001; N=324) in LUX-Lung 6, respectively.
PFS benefit was accompanied by improvement in disease-related symptoms and delayed time to deterioration (see Table 7). Mean scores over time for overall quality of life, global health status and physical, role, cognitive, social and emotional functioning were significantly better for GIOTRIF.
Table 7: Symptom outcomes for GIOTRIF vs. chemotherapy in trials LUX-Lung 3 and LUX-Lung 6 (EORTC QLQ-C30 & QLQ-LC13)

LUX-Lung 3

Cough

Dyspnoea

Pain

% of patients improved a

67% vs. 60%;

p=0.2133

65% vs. 50%;

p=0.0078

60% vs. 48%;

p=0.0427

Delay of median time to deterioration (months) a,b

27.0 vs. 8.0

HR 0.60; p=0.0062

10.4 vs. 2.9

HR 0.68; p=0.0129

4.2 vs. 3.1

HR 0.83; p=0.1882

 

LUX-Lung 6

Cough

Dyspnoea

Pain

% of patients improved a

76% vs. 55%;

p=0.0003

71% vs. 48%;

p<0.0001

65% vs. 47%;

p=0.0017

Delay of median time to deterioration (months) a,b

31.1 vs. 10.3

HR 0.46; p=0.0001

7.7 vs. 1.7

HR 0.53; p<0.0001

6.9 vs. 3.4

HR 0.70; p=0.0220

a values presented for GIOTRIF vs. chemotherapy, p-value based on logistic regression
b p-value for time to deterioration based on stratified log-rank test
LUX-Lung 2
LUX-Lung 2 was a single arm Phase II trial in 129 EGFR TKI-naïve patients with stage IIIB or IV lung adenocarcinoma with EGFR mutations. Patients were enrolled in the first-line (N=61) or second-line setting (N=68) (i.e. after failure of 1 prior chemotherapy regimen). In 61 patients treated in the first-line setting, confirmed ORR was 65.6% and DCR was 86.9% according to independent review. The median PFS was 12.0 months by independent review. Efficacy was similarly high in the group of patients who had received prior chemotherapy (N=68; ORR 57.4%; median PFS by independent review 8 months). The updated median OS for first- and second-line was 31.7 months and 23.6 months, respectively.
GIOTRIF in patients with NSCLC of squamous histology
The efficacy and safety of GIOTRIF as second-line treatment for patients with advanced NSCLC of squamous histology was investigated in a randomized open-label global Phase III trial LUX-Lung 8. Patients who received at least 4 cycles of platinum-based therapy in the first line setting were subsequently randomized 1:1 to daily GIOTRIF 40 mg or erlotinib 150 mg until progression. Randomization was stratified by race (Eastern Asian vs non Eastern Asian). The primary endpoint was PFS; OS was the key secondary endpoint. Other secondary endpoints included ORR, DCR, change in tumour size and HRQOL.
Among 795 patients randomized, the majority were males (84%), white (73%), current or former smokers (95%) with baseline performance status ECOG 1 (67%) and ECOG 0 (33%).
Second-line GIOTRIF significantly improved PFS and OS of patients with squamous NSCLC compared to erlotinib. The efficacy results at the time of the primary analysis of OS including all randomized patients are summarized in Figure 2 and Table 8.
Table 8: Efficacy results for GIOTRIF vs erlotinib in LUX-Lung 8, based on primary analysis of OS, including all randomized patients

GIOTRIF
 

(N=398)

Erlotinib
 

(n=397)

Hazard Ratio/ Odds Ratio

(95%CI)

p-value2

PFS

Months (median)

2.63

1.94

HR 0.81

(0.69, 0.96)

0.0103

OS

Months (median)

Alive at 12 months

Alive at 18 months

7.92

36.4%

22.0%

6.77

28.2%

14.4%

HR 0.81

(0.69, 0.95)

0.0077

Objective Response Rate (CR+PR)1

5.5%

2.8%

OR 2.06

(0.98, 4.32)

0.0551

Duration of response

Months (median)

7.29

3.71

 

 

1CR=complete response; PR=partial response
2p-value for PFS/OS based on stratified log-rank test; p-value for Objective Response Rate and Disease Control Rate based on logistic regression
The overall survival hazard ratio in patients < 65 years of age was 0.68 (95% CI 0.55, 0.85) and in patients 65 years of age and older it was 0.95 (95% CI 0.76, 1.19).
Figure 2: Kaplan-Meier Curve for OS by treatment group in LUX Lung 8


PFS benefit was accompanied by improvement in disease-related symptoms and delayed time to deterioration (see Table 9).
Table 9: Symptom outcomes for GIOTRIF vs. erlotinib in trial LUX-Lung 8 (EORTC QLQ-C30 & QLQ-LC13)

Cough

Dyspnoea

Pain

% of patients improveda, c

43% vs. 35%;

p=0.0294

51% vs. 44%;

p=0.0605

40% vs. 39%;

p=0.7752

Delay of time to deterioration (months)b, c

4.5 vs. 3.7

HR 0.89; p=0.2562

2.6 vs. 1.9

HR 0.79; p=0.0078

2.5 vs. 2.4

HR 0.99; p=0.8690

a values presented for GIOTRIF vs. erlotinib, p-value based on logistic regression
b p-value for time to deterioration based on stratified log-rank test
c p-values were not adjusted for multiplicity
Efficacy in EGFR-negative tumours has not been established.
Paediatric population
The European Medicines Agency has waived the obligation to submit the results of trials with this medicinal product in all subsets of the paediatric population in NSCLC indications (see section 4.2 for information on paediatric use).
5.2 Pharmacokinetic properties
Absorption
Following oral administration of GIOTRIF, Cmax of afatinib were observed approximately 2 to 5 hours post dose. Cmax and AUC0-∞ values increased slightly more than proportionally in the dose range from 20 mg to 50 mg GIOTRIF. Systemic exposure to afatinib is decreased by 50% (Cmax) and 39% (AUC0-∞), when administered with a high-fat meal compared to administration in the fasted state. Based on population pharmacokinetic data derived from clinical trials in various tumour types, an average decrease of 26% in AUC,ss was observed when food was consumed within 3 hours before or 1 hour after taking GIOTRIF. Therefore, food should not be consumed for at least 3 hours before and at least 1 hour after taking GIOTRIF (see sections 4.2 and 4.5).
Distribution
In vitro binding of afatinib to human plasma proteins is approximately 95%. Afatinib binds to proteins both non-covalently (traditional protein binding) and covalently.
Biotransformation
Enzyme-catalyzed metabolic reactions play a negligible role for afatinib in vivo. Covalent adducts to proteins were the major circulating metabolites of afatinib.
Elimination
In humans, excretion of afatinib is primarily via the faeces. Following administration of an oral solution of 15 mg afatinib, 85.4% of the dose was recovered in the faeces and 4.3% in urine. The parent compound afatinib accounted for 88% of the recovered dose. Afatinib is eliminated with an effective half-life of approximately 37 hours. Thus, steady state plasma concentrations of afatinib were achieved within 8 days of multiple dosing of afatinib resulting in an accumulation of 2.77-fold (AUC0-∞) and 2.11-fold (Cmax). In patients treated with afatinib for more than 6 months a terminal half-life of 344 h was estimated.
Special populations
Renal impairment
Less than 5% of a single dose of afatinib is excreted via the kidneys. The safety, pharmacokinetics and efficacy of GIOTRIF have not been studied specifically in patients with renal impairment. Based on population pharmacokinetic data derived from clinical trials in various tumour types, no dose adjustments appear necessary in patients with mild or moderate renal impairment (see “Population pharmacokinetic analysis in special populations” below and section 4.2).
Hepatic impairment
Afatinib is eliminated mainly by biliary/faecal excretion. Subjects with mild (Child Pugh A) or moderate (Child Pugh B) hepatic impairment had similar exposure in comparison to healthy volunteers following a single dose of 50 mg GIOTRIF. This is consistent with population pharmacokinetic data derived from clinical trials in various tumour types (see “Population pharmacokinetic analysis in special populations” below). No starting dose adjustments appear necessary in patients with mild or moderate hepatic impairment (see section 4.2). The pharmacokinetics of afatinib have not been studied in subjects with severe (Child Pugh C) hepatic dysfunction (see section 4.4).
Population pharmacokinetic analysis in special populations
A population pharmacokinetic analysis was performed in 927 cancer patients (764 with NSCLC) receiving GIOTRIF monotherapy. No starting dose adjustment was considered necessary for any of the following covariates tested.
Age
No significant impact of age (range: 28 years - 87 years) on the pharmacokinetics of afatinib could be observed.
Body weight
Plasma exposure (AUC,ss) was increased by 26% for a 42 kg patient (2.5th percentile) and decreased by 22% for a 95 kg patient (97.5th percentile) relative to a patient weighing 62 kg (median body weight of patients in the overall patient population).
Gender
Female patients had a 15% higher plasma exposure (AUC,ss, body weight corrected) than male patients.
Race
Race had no effect on the pharmacokinetics of afatinib based on a population pharmacokinetic analysis, including patients of Asian, White, and Black racial groups. Data on Black racial groups was limited.
Renal impairment
Exposure to afatinib moderately increased with lowering of the creatinine clearance (CrCL, calculated according to Cockcroft Gault), i.e. for a patient with a CrCL of 60 mL/min or 30 mL/min exposure (AUC,ss) to afatinib increased by 13% and 42%, respectively, and decreased by 6% and 20% for a patient with CrCL of 90 mL/min or 120 mL/min, respectively, compared to a patient with the CrCL of 79 mL/min (median CrCL of patients in the overall patient population analysed).
Hepatic impairment
Patients with mild and moderate hepatic impairment as identified by abnormal liver tests did not correlate with any significant change in afatinib exposure. There was limited data available for moderate and severe hepatic impairment.
Other patient characteristics/intrinsic factors
Other patient characteristics/intrinsic factors found with a significant impact on afatinib exposure were: ECOG performance score, lactate dehydrogenase levels, alkaline phosphatase levels and total protein. The individual effect sizes of these covariates were considered not clinically relevant. Smoking history, alcohol consumption (limited data), or presence of liver metastases had no significant impact on the pharmacokinetics of afatinib.
Other information on drug-drug interactions
Interactions with drug uptake transport systems
In vitro data indicated that drug-drug interactions with afatinib due to inhibition of OATB1B1, OATP1B3, OATP2B1, OAT1, OAT3, OCT1, OCT2, and OCT3 transporters are considered unlikely.
Interactions with Cytochrome P450 (CYP) enzymes
In humans it was found that enzyme-catalyzed metabolic reactions play a negligible role for the metabolism of afatinib. Approximately 2% of the afatinib dose was metabolized by FMO3 and the CYP3A4-dependent N-demethylation was too low to be quantitatively detected. Afatinib is not an inhibitor or an inducer of CYP enzymes. Therefore, this medicinal product is unlikely to interact with other medicines that modulate or are metabolised by CYP enzymes.
Effect of UDP-glucuronosyltransferase 1A1 (UGT1A1) inhibition on afatinib
In vitro data indicated that drug-drug interactions with afatinib due to inhibition of UGT1A1 are considered unlikely.
5.3 Preclinical safety data
Oral administration of single doses to mice and rats indicated a low acute toxic potential of afatinib. In oral repeated-dose studies for up to 26 weeks in rats or 52 weeks in minipigs the main effects were identified in the skin (dermal changes, epithelial atrophy and folliculitis in rats), the gastrointestinal tract (diarrhoea, erosions in the stomach, epithelial atrophy in rats and minipigs) and the kidneys (papillary necrosis in rats). Depending on the finding, these changes occurred at exposures below, in the range of or above clinically relevant levels. Additionally, in various organs pharmacodynamically mediated atrophy of epithelia was observed in both species.
Reproduction toxicity
Based on the mechanism of action, all EGFR targeting medicinal products including GIOTRIF have the potential to cause foetal harm. The embryo-foetal development studies performed on afatinib revealed no indication of teratogenicity. The respective total systemic exposure (AUC) was either slightly above (2.2 times in rats) or below (0.3 times in rabbits) compared with levels in patients.
Radiolabelled afatinib administered orally to rats on Day 11 of lactation was excreted in the breast milk of the dams.
A fertility study in male and female rats up to the maximum tolerated dose revealed no significant impact on fertility. The total systemic exposure (AUC0-24) in male and female rats was in the range or less than that observed in patients (1.3 times and 0.51 times, respectively).
A study in rats up to the maximum tolerated doses revealed no significant impact on pre-/postnatal development. The highest total systemic exposure (AUC0-24) in female rats was less than that observed in patients (0.23 times).
Phototoxicity
An in vitro 3T3 test showed that afatinib may have phototoxicity potential.
Carcinogenicity
Carcinogenicity studies have not been conducted with GIOTRIF.
6. Pharmaceutical particulars
6.1 List of excipients
Tablet core
Lactose monohydrate
Cellulose, microcrystalline (E460)
Silica, colloidal anhydrous (E551)
Crospovidone type A
Magnesium stearate (E470b)
Film-coating
GIOTRIF 20 mg film-coated tablets
Hypromellose (E464)
Macrogol 400
Titanium dioxide (E171)
Talc (E553b)
Polysorbate 80 (E433)
GIOTRIF 30, 40 and 50 mg film-coated tablets
Hypromellose (E464)
Macrogol 400
Titanium dioxide (E171)
Talc (E553b)
Polysorbate 80 (E433)
Indigo carmine (E132) aluminium hydroxide
6.2 Incompatibilities
Not applicable.
6.3 Shelf life
3 years.
6.4 Special precautions for storage
Store in the original package in order to protect from moisture and light.
6.5 Nature and contents of container
PVC/PVDC perforated unit dose blister. Each blister is packed together with a desiccant sachet in a laminated aluminium pouch and contains 7 x 1 film-coated tablets. Pack sizes of 7 x 1, 14 x 1 or 28 x 1 film-coated tablets.
Not all pack sizes may be marketed.
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.
7. Marketing authorisation holder
Boehringer Ingelheim International GmbH
Binger Strasse 173
D-55216 Ingelheim am Rhein
Germany
8. Marketing authorisation number(s)
GIOTRIF 20 mg film-coated tablets
EU/1/13/879/001
EU/1/13/879/002
EU/1/13/879/003
GIOTRIF 30 mg film-coated tablets
EU/1/13/879/004
EU/1/13/879/005
EU/1/13/879/006
GIOTRIF 40 mg film-coated tablets
EU/1/13/879/007
EU/1/13/879/008
EU/1/13/879/009
GIOTRIF 50 mg film-coated tablets
EU/1/13/879/010
EU/1/13/879/011
EU/1/13/879/012
9. Date of first authorisation/renewal of the authorisation
Date of first authorisation: 25 September 2013
10. Date of revision of the text
03/2016
Detailed information on this medicinal product is available on the website of the European Medicines Agency http://www.ema.europa.eu


阿法替尼(GIOTRIF)疗效和安全性再添证据
•新的疗效和安全性分析结果进一步充实了之前已经公布的、日益丰富的关于阿法替尼相较于标准化疗(培美曲塞/顺铂和吉西他滨/顺铂)的临床数据库
•最新数据证实,不可逆性ErbB家族阻滞剂阿法替尼具有可控的安全性,而且适用于EGFR突变阳性的亚洲和非亚洲肺癌患者的长期治疗。
•阿法替尼已经在美国、欧盟、墨西哥、智利获准应用于EGFR突变阳性的非小细胞肺癌(NSCLC)患者。在亚洲地区,阿法替尼在台湾获得了上述批准。该药在其他亚洲国家的注册申请正处于审批过程中。
•全球肺癌联盟与勃林格殷格翰公司合作启动“倾听肺癌患者的心声”旨在收集肺癌患者在真实世界中的感受
德国殷格翰2013年11月11日,来自两项关键性的、大规模、III期、注册临床试验(LUX-Lung 3研究和LUX-Lung 6研究)的数据显示,阿法替尼相较于化疗应用于亚洲和非亚洲患者具有优越的疗效和可控的安全性。针对上述试验所开展的最新分析证实,阿法替尼作为一线治疗应用于EGFR突变阳性的非小细胞肺癌(NSCLC)亚洲和非亚洲患者具有一致的安全性。
在NSCLC患者亚群中获得的最新数据也进一步证实了阿法替尼的疗效。基于LUX-Lung研究的其他分析也显示了阿法替尼应用于伴有不常见的EGFR突变类型以及伴有转移性脑病的NSCLC患者的安全性。
在今年的世界肺癌大会(WCLC)上公布的研究结果进一步充实了日益丰富的强大的临床证据库,这些证据支持阿法替尼作为一线治疗药物应用于亚洲和非亚洲患有特定类型的晚期肺癌患者(即表皮生长因子受体(EGFR)突变阳性的NSCLC)。在中国、韩国和泰国开展的全球性临床试验 LUX-Lung 3研究及其并行试验LUX-Lung 6研究是迄今为止在EGFR突变阳性的NSCLC患者中开展过的规模最大的注册临床试验。上述试验早已证实,接受阿法替尼治疗的患者在肿瘤重新开始生长之前的生存(无进展生存,PFS)时间达到将近一年,而接受化疗的患者则略超过半年。
具体而言,上述试验显示:
•LUX-Lung 3研究:阿法替尼治疗组和培美曲塞/顺铂治疗组的PFS分别为11.1个月和6.9个月
•LUX-Lung 6研究:阿法替尼治疗组和培美曲塞/顺铂治疗组的PFS分别为11.0个月和5.6个月
(这些数据来自基于独立性回顾的主要分析)
在LUX-Lung 6研究中,接受阿法替尼治疗组有47%患者在治疗1年后仍然存活,而且无疾病进展,化疗组则仅为2%。肿瘤进展的延迟还伴有患者肺癌相关性症状(例如气促、咳嗽和胸痛)的改善以及通过肺癌标准问卷评估的生活质量的改善。
在亚洲,肺癌占所有癌症的比例超过了14%,肺癌死亡占所有癌症死亡的比例超过了18%,尽管肺癌发生率在不同地区会有所不同。东亚地区的发病率最高,在中国大陆,每年新诊断的肺癌病例数量超过五十万,在日本和台湾地区则分别超过八万六千例和九千例。由于肺癌的预后较差,从最初开始就选择最佳的治疗方案就变得非常重要。
基于LUX-Lung 3研究和LUX-Lung 6研究最新开展的安全性汇总分析证实了之前已经报告的阿法替尼应用于EGFR突变阳性的亚洲和非亚洲NSCLC患者的不良事件(AE)和耐受性特征。在上述两大患者人群中,最常见的3级副反应的发生率是相似的,包括腹泻、皮疹/痤疮和口腔炎(口腔黏膜的炎症)。此外,阿法替尼的药代动力学暴露结果在上述两大患者人群中也无差异。在此之前,来自LUX-Lung 3研究和LUX-Lung6研究的数据已经证实,阿法替尼的副反应是可预测、可控和可逆的,而且停药率较低。
“最新的亚组分析证实,阿法替尼在亚洲患者中和非亚洲患者中的安全性特征是一致的,”来自位于台湾地区台北市的台湾大学医学院肿瘤研究中心主任、LUX-Lung 3研究的主要研究者 James Chih-Hsin Yang教授如此评价道。“这些数据为阿法替尼作为EGFR突变阳性的肺癌患者的有价值的治疗选择提供了进一步的支持,EGFR突变阳性的肺癌在亚洲的发生率是西方国家三倍。”
另一场独立的口头演讲则基于迄今为止规模最大的、针对伴有不常见EGFR突变类型的患者的疗效数据集,这些汇总数据来自3项前瞻性阿法替尼临床试验。这些数据显示,阿法替尼应用于伴有罕见的特定类型EGFR突变的肺癌患者的活性与该药应用于伴有常见类型的EGFR突变(缺失19, L858R)的患者的活性处于同样的范围之内。
一项基于LUX-Lung 3研究的亚组分析显示,阿法替尼可作为伴有常见类型的EGFR突变和转移性脑病的肺癌患者的有效的一线治疗选择。
“阿法替尼所能提供的临床受益已经通过大规模的、设计严谨的LUX-Lung临床试验计划获得了明确的体现,这些结论现在又获得了基于重要的亚组患者人群的、强有力的疗效和安全性数据的进一步支持,”勃林格殷格翰公司全球医学高级副总裁Klaus Dugi如此说道。“这些分析结果非常振奋人心,而且有望使阿法替尼成为伴有EGFR突变的亚洲和非亚洲肺癌患者的有价值的治疗选择的重要补充。”
此外,全球肺癌联盟(GLCC)还在WCLC上宣布了一项由勃林格殷格翰公司(BI)支持的创意活动,此项活动旨在进一步了解并理解肺癌患者及其家庭在现实世界中所面临的挑战。“倾听肺癌患者的心声”项目将收集来自于那些被肺癌疾病所累及的人群的反馈,包括患者及其护理者、家人、朋友、医疗专业人士和同事等。
“倾听肺癌患者的心声”项目采用独特的方法收集患者的反馈,尽可能减少基于书面文本的反馈格式,主要呈现基于视觉的调查材料。通过采用简明浏览格式的新型在线工具,受调研者只需要点击预先定义的图像、标志和基于网络的视觉材料中的元素,就能针对与之相关的议题提供反馈。这些视觉元素代表了肺癌患者及其护理人员以及家人在生活中所面临的挑战以及问题的重要性。受调研者所选取的图像或标志将透露他们最为迫切的问题和顾虑,从而反映肺癌患者随着时间推移在现实世界中的日常事项和经历。
这是有史以来首次采用上述设计方法来“倾听”肺癌患者在现实世界中的感受,并在医疗服务提供方与患者的真实处境之间搭建起沟通桥梁,从而消除间隔。调研结果将会与肺癌社区分享,从中获得的心得也将与为患者提供服务的相关人士进行分享,进而推动旨在更好地满足患者需求的新型项目、服务和宣教材料的开发。
“我们需要理解患者的感受,从而更好地为他们提供帮助,”勃林格殷格翰公司的全球医学高级副总裁Klaus Dugi教授如此说道,“患者始终处于我们在肿瘤领域所作出的承诺的核心位置,通过与GLCC的共同努力,我们就能利用‘倾听肺癌患者的心声’项目为开展全球性活动提供信息,同时有助于更加有效地支持肺癌社区,最终改善患者的生活,而且这一改善作用将不局限于治疗本身。”
“倾听肺癌患者的心声”项目的网站将于11月中旬上线,届时也将与“全球肺癌关注月”的活动相呼应。
关于阿法替尼数据在WCLC上的公布情况
安全性亚组分析
基于LUX-Lung 3研究和LUX-Lung 6研究的最新汇总性亚组分析总共纳入了468名EGFR突变阳性的患者(404名亚洲患者和64名非亚洲患者),上述患者接受阿法替尼40 mg每日一次治疗,直到发生疾病进展或发生不可耐受的不良事件为止。基于预先定义的研究标准,阿法替尼的给药剂量可增至50 mg每日一次、或降至30 mg或20 mg每日一次。所有患者报告了至少1例不良事件,最常见的不良事件包括腹泻、皮疹/痤疮和口腔炎(口腔黏膜的炎症)。导致停药的药物相关性不良事件的发生率在亚洲患者和非亚洲患者中分别为7.2%和4.7%,低于化疗组的28%。
针对不常见的突变类型的亚组分析
此项分析基于被纳入LUX-Lung 2研究(II期临床试验)、LUX-Lung 3研究和LUX-Lung 6研究(两者均为III期临床试验)的EGFR突变阳性的患者。患者所伴有的突变被分为常见(Del 19 或 L858R)和不常见(所有其他单个或多个突变)两大类。通过独立性回顾对客观反应率、疾病控制、反应时间和无进展生存(PFS)进行评估。
针对转移性脑病的亚组分析
在LUX-Lung 3研究中,EGFR突变阳性的患者以2:1的比例随机接受阿法替尼40 mg 每日一次治疗或最多达6个疗程的培美曲塞/顺铂标准剂量方案。伴有稳定性脑转移的患者被允许纳入。由研究者在筛选期记录脑转移。每6周进行一次肿瘤评估,直到48周为止,在此之后每12周进行一次肿瘤评估,直到出现疾病进展为止。
关于LUX-Lung 3试验
LUX-Lung 3试验是规模最大的、随机、开放标记、III期注册临床试验,此项试验针对阿法替尼与由两种化疗药物培美曲塞和顺铂组成的化疗方案作为一线治疗选择应用于伴有EGFR突变的IIIb期或IV期NSCLC患者的效果进行了比较。此项试验在全球范围内纳入了345名EGFR突变阳性的NSCLC患者。LUX-Lung 3试验也是第一项在EGFR突变阳性的NSCLC患者中开展的、将培美曲塞/顺铂作为对照药物的临床试验。
在LUX-Lung 3试验中,与阿法替尼有关的最常见的3级不良事件是腹泻(14%)、皮疹(16%)和甲沟炎(11%)。与化疗(培美曲塞/顺铂)相关的最常见的3级不良事件是中性粒细胞减少症(15%)、虚弱(13%)和白细胞减少(8%)。在此项试验中,与治疗有关的不良事件有关的停药率较低(阿法替尼治疗组织和化疗组的停药率分别为8%和12%)。在阿法替尼治疗组中,有1%患者由于腹泻而停药。
关于 LUX-Lung 6试验
LUX-Lung 6试验是迄今为止在伴有EGFR突变阳性的亚洲晚期肺癌患者中开展的规模最大(n = 364)、前瞻性、注册临床试验。LUX-Lung 6试验是一项多中心、随机、开放标记的III期临床试验,对于阿法替尼相较化疗(顺铂/吉西他滨)作为一线治疗方案应用于伴有EGFR突变的晚期和转移性NSCLC的效果进行了考察。
此项试验共纳入了364名来自中国、韩国和泰国的患者(IIIB/IV期,功能状态评分0–1,之前未接受过化疗),这些患者以2:1的比例随机接受每日口服阿法替尼*40mg (n=242) 或静脉接受顺铂/吉西他滨(第1天75 mg/m2/第1天和第8天1000mg/m2,每21天一个疗程,共6个疗程,n=122)。主要终点是由中心独立审核而确定的无进展生存期。
顺铂/吉西他滨是目前在中国、韩国和泰国被经常使用的NSCLC一线治疗选择。
关于阿法替尼
在欧盟、台湾地区、智利和墨西哥,阿法替尼以GIOTRIF®为商品名获准应用于肿瘤细胞具有表皮生长因子受体(EGFR)突变的转移性NSCLC的治疗。
阿法替尼在美国以GILOTRIFTM为商品名获得批准作为一线治疗药物应用于通过经FDA批准的检测方法检出存在表皮生长因子受体(EGFR)外显子19缺失或外显子21(L858R)替代突变的转移性非小细胞肺癌(NSCLC)患者。
阿法替尼是一种不可逆性ErbB家族抑制剂,该药能够不可逆性地阻断EGFR (ErbB1)以及ErbB家族的其他相关成员,上述受体在某些最为常见的肿瘤和某些死亡率较高的肿瘤的生长和播散过程中发挥了关键性的作用。阿法替尼与受体的共价、不可逆性结合与其他化合物与受体的可逆性结合的不同之处在于能够提供持久的、选择性的、完全性的ErbB家族的阻断,从而可能带来独特的治疗受益。
阿法替尼目前正处于针对NSCLC和头颈癌的III期临床试验过程中。
关于肺癌
肺癌是在全球范围内最为常见的一种癌症,每年的新发病例数达到一百六十万,其中超过半数(54%)发生在亚洲。总体而言,肺癌死亡人数占到所有亚洲癌症死亡人数18.5%。肺癌约占所有新发癌症的13%,吸烟是肺癌的主要病因。
有10%至15%的白种人NSCLC患者和 40%的亚洲NSCLC患者的肿瘤存在EGFR突变,其中约有90%的病例伴有两种EGFR突变中的一种(Del19或 L858R)。

责任编辑:admin


相关文章
卡赞替尼片|Cabometyx(Cabozantinib Tablets)
Cabometyx(cabozantinib)临床治疗晚期肾细胞癌
曲美替尼片|MEKINIST(trametinib tablets)
Tafinlar(Dabrafenib Mesilate Capsules)
Alecensa Capsule(盐酸阿雷替尼胶囊)
XELJANZ Filmtabl(tofacitinib Tablets)枸橼酸托法替尼片
阿西替尼片|INLYTA(axitinib tablets)
INLYTA Filmtabl tab(axitinib 阿西替尼片)
TAGRISSO(osimertinib)临床治疗小细胞肺癌
Caprelsa(vandetanib filmcoated tablets)
抗血癌新药IMBRUVICA(ibrutinib)获欧盟批准扩展治疗巨球蛋白血症
 

最新文章

更多

· Imbruvica(ibrutinib c...
· G-LASTA Subcutaneous I...
· Venetoclax(Venclexta)T...
· 曲美替尼片|MEKINIST(tr...
· CYRAMZA(ramucirumab in...
· 西罗莫司片|Rapalimus(...
· Lonsurf tablet(triflu...
· 色瑞替尼胶囊|Zykadia(...
· Tagrisso(osimertinib ...
· CYRAMZA Inf Konz(Ramu...

推荐文章

更多

· Imbruvica(ibrutinib c...
· G-LASTA Subcutaneous I...
· Venetoclax(Venclexta)T...
· 曲美替尼片|MEKINIST(tr...
· CYRAMZA(ramucirumab in...
· 西罗莫司片|Rapalimus(...
· Lonsurf tablet(triflu...
· 色瑞替尼胶囊|Zykadia(...
· Tagrisso(osimertinib ...
· CYRAMZA Inf Konz(Ramu...

热点文章

更多

· Venetoclax(Venclexta)T...
· 曲美替尼片|MEKINIST(tr...
· Imbruvica(ibrutinib c...
· G-LASTA Subcutaneous I...