Open menu Close menu Open Search Close search

AMERICAN DRUGS | ANATOMY | HEALTH TOPICS | HIV/AIDS GLOSSARY | DISEASES | HEALTH ARTICLES | GENOME | OCCUPATIONS

Glivec


Spanish Simplified Chinese French German Russian Hindi Arabic Portuguese
















Summary for the public


What is Glivec?

Glivec is a medicine that contains the active substance imatinib. It is available as capsules (orange-yellow: 50 mg; orange: 100 mg) and dark yellow/orange tablets (round: 100 mg; oval: 400 mg).


What is Glivec used for?

Glivec is an anticancer medicine. It is used to treat the following diseases:

  • chronic myeloid leukaemia (CML), a cancer of the white blood cells in which granulocytes (a type of white blood cell) start growing out of control. Glivec is used when the patients are ‘Philadelphia chromosome positive’ (Ph+). This means that some of their genes have re-arranged themselves to form a special chromosome called the Philadelphia chromosome. Glivec is used in adults and children who have been newly diagnosed with Ph+ CML, who are not eligible for a bone marrow transplant. It is also used in adults and children in the ‘chronic phase’ of the disease if it is not responding to interferon alpha (another anticancer medicine), and in more advanced phases of the disease (‘accelerated phase’ and ‘blast crisis’);
  • Ph+ acute lymphoblastic leukaemia (ALL), a type of cancer in which lymphocytes (another type of white blood cell) multiply too quickly. Glivec is used in combination with other anticancer medicines in adults who have been newly diagnosed with Ph+ ALL. It is also used alone to treat Ph+ ALL that has returned following previous treatment, or is not responding to other medicines. 
  • myelodysplastic or myeloproliferative diseases (MD/MPD), a group of diseases in which the body produces large numbers of abnormal blood cells. Glivec is used to treat adults with MD/MPD who have re-arrangements of the gene for platelet-derived growth factor receptor (PDGFR); 
  • advanced hypereosinophilic syndrome (HES) or chronic eosinophilic leukaemia (CEL), diseases in which eosinophils (another type of white blood cell) start growing out of control. Glivec is used to treat adults with HES or CEL who have a specific re-arrangement of two genes called FIP1L1 and PDGFRα;
  • gastrointestinal stromal tumours (GIST), a type of cancer (sarcoma) of the stomach and bowel, when there is uncontrolled growth of cells in the supporting tissues of these organs. Glivec is used to treat adults with GIST that cannot be removed with surgery or have spread to other parts of the body, and adults who are at risk of GIST coming back after surgical removal; 
  • dermatofibrosarcoma protuberans (DFSP), a type of cancer (sarcoma) in which cells in the tissue beneath the skin divide uncontrollably. Glivec is used to treat adults with DFSP that cannot be removed with surgery, and in adults who are not eligible for surgery when the cancer has returned after treatment or has spread to other parts of the body. 

Because the number of patients with these diseases is low, they are considered ‘rare’, and Glivec was designated an ‘orphan medicine’ (a medicine used in rare diseases) on various dates (see below).

The medicine can only be obtained with a prescription. 


How is Glivec used?

Glivec treatment should be started by a doctor who has experience in the treatment of patients with cancers of the blood or solid tumours. Glivec is given by mouth with a meal and a large glass of water to reduce the risk of irritation of the stomach and gut. The dose depends on the disease being treated, the age and condition of the patient, and the response to treatment, but it should not exceed 800 mg a day. For more information, see the Package Leaflet.


How does Glivec work?

The active substance in Glivec, imatinib, is a protein-tyrosine kinase inhibitor. This means that it blocks some specific enzymes known as tyrosine kinases. These enzymes can be found in some receptors on the surface of cancer cells, including the receptors that are involved in stimulating the cells to divide uncontrollably. By blocking these receptors, Glivec helps to control cell division.


How has Glivec been studied?

For CML, Glivec has been examined in four main studies involving 2,133 adults and one study of 54 children. These included a study involving 1,106 adults that compared Glivec with the combination of interferon alpha plus cytarabine (other anticancer medicines). This study measured how long the patients lived without their cancer getting worse.

For ALL, Glivec has been examined in three studies involving 456 adults, including one study comparing Glivec with standard chemotherapy (medicines used to kill cancer cells) in 55 newly-diagnosed patients.

For GIST, Glivec has been examined in two main studies. One involved 147 patients whose GIST could not be surgically removed or had spread to other parts of the body, and looked at whether the tumours shrank in size. This study did not compare Glivec with any other medicines. The other study compared Glivec with placebo (a dummy treatment) in 713 patients whose cancer had been removed with surgery. This study measured how long the patients lived without their cancer coming back. 

For MD/MPD (31 patients), HES and CEL (176 patients), and DFSP (18 patients), Glivec was not compared with any other medicines. These studies examined whether blood cell counts returned to normal levels, or whether the number of cancerous blood cells or the size of tumours fell. 


What benefit has Glivec shown during the studies?

Glivec was more effective than the comparator medicines. In patients with CML, the cancer had got worse in 16% of the patients taking Glivec after five years, compared with 28% of those taking interferon alpha plus cytarabine. Glivec was also better than standard chemotherapy in patients with ALL. In patients with GIST that had been removed with surgery, patients taking Glivec lived for longer than those taking placebo without their cancer coming back. In the non-comparative studies of CML, ALL and GIST, between 26 and 96% of patients showed a response to Glivec. Due to their rarity, limited data were available for the other diseases, but around two thirds of the patients showed at least a partial response to Glivec.


What is the risk associated with Glivec?

The most common side effects with Glivec (seen in more than 1 in 10 patients) are weight increase, neutropenia (low levels of the white blood cells that fight infection), thrombocytopenia (low blood platelet counts), anaemia (low red blood cell counts), headache, nausea (feeling sick), vomiting, diarrhoea, dyspepsia (indigestion), abdominal (tummy) pain, oedema (fluid retention), rash, muscle spasm and cramps, muscle and joint pain, and fatigue (tiredness). For the full list of all side effects reported with Glivec, see the Package Leaflet.

Glivec should not be used in people who may be hypersensitive (allergic) to imatinib or any of the other ingredients.


Why has Glivec been approved?

The Committee for Medicinal Products for Human Use (CHMP) decided that Glivec’s benefits are greater than its risks for the treatment of CML, ALL, MD/MPD, HES, CEL, GIST and DFSP. The Committee recommended that Glivec be given marketing authorisation.

Glivec was originally authorised under ‘Exceptional Circumstances’, because, as the diseases are rare, limited information was available at the time of the approval. As the company had supplied the additional information requested, the ‘Exceptional Circumstances’ were lifted on 13 April 2007.


Other information about Glivec

The European Commission granted a marketing authorisation valid throughout the European Union for Glivec to Novartis Europharm Limited on 7 November 2001. The marketing authorisation was renewed on 7 November 2006.

Authorisation details
Name: Glivec
EMEA Product number: EMEA/H/C/000406
Active substance: imatinib
INN or common name: imatinib
Therapeutic area: Leukemia, Myelogenous, Chronic, BCR-ABL PositiveGastrointestinal Stromal TumorsMyelodysplastic-Myeloproliferative DiseasesDermatofibrosarcomaPrecursor Cell Lymphoblastic Leukemia-LymphomaHypereosinophilic Syndrome
ATC Code: L01XE01
Treatment of rare diseases: This medicine has an "orphan designation" which means that it is used to treat life-threatening or chronically debilitating conditions that affect no more than five in 10,000 people in the European Union, or are medicines which, for economic reasons, would be unlikely to be developed without incentives.
Marketing Authorisation Holder: Novartis Europharm Ltd.
Revision: 13
Date of issue of Market Authorisation valid throughout the European Union: 07/11/2001
Contact address:
Novartis Europharm Limited
Wimblehurst Road
Horsham
West Sussex, RH12 5AB
United Kingdom




Product Characteristics

ANNEX I

SUMMARY OF PRODUCT CHARACTERISTICS


1.
NAME OF THE MEDICINAL PRODUCT
Glivec 50 mg hard capsules
2.
QUALITATIVE AND QUANTITATIVE COMPOSITION
Each capsule contains 50 mg imatinib (as mesilate).
For a full list of excipients, see section 6.1.
3.
PHARMACEUTICAL FORM
Hard capsule
White to yellow powder in a light yellow to orange-yellow opaque capsule, marked “NVR SH”.
4.
CLINICAL PARTICULARS
4.1 Therapeutic indications
Glivec is indicated for the treatment of
adult and paediatric patients with newly diagnosed Philadelphia chromosome (bcr-abl) positive
(Ph+) chronic myeloid leukaemia (CML) for whom bone marrow transplantation is not
considered as the first line of treatment.
adult and paediatric patients with Ph+ CML in chronic phase after failure of interferon-alpha
therapy, or in accelerated phase or blast crisis.
adult patients with newly diagnosed Philadelphia chromosome positive acute lymphoblastic
leukaemia (Ph+ ALL) integrated with chemotherapy.
adult patients with relapsed or refractory Ph+ ALL as monotherapy.
adult patients with myelodysplastic/myeloproliferative diseases (MDS/MPD) associated with
platelet-derived growth factor receptor (PDGFR) gene re-arrangements.
adult patients with advanced hypereosinophilic syndrome (HES) and/or chronic eosinophilic
leukaemia (CEL) with FIP1L1-PDGFR rearrangement.
The effect of Glivec on the outcome of bone marrow transplantation has not been determined.
Glivec is indicated for
the treatment of adult patients with Kit (CD 117) positive unresectable and/or metastatic
malignant gastrointestinal stromal tumours (GIST).
the adjuvant treatment of adult patients who are at significant risk of relapse following resection
of Kit (CD117)-positive GIST. Patients who have a low or very low risk of recurrence should
not receive adjuvant treatment.
the treatment of adult patients with unresectable dermatofibrosarcoma protuberans (DFSP) and
adult patients with recurrent and/or metastatic DFSP who are not eligible for surgery.
In adult and paediatric patients, the effectiveness of Glivec is based on overall haematological and
cytogenetic response rates and progression-free survival in CML, on haematological and cytogenetic
response rates in Ph+ ALL, MDS/MPD, on haematological response rates in HES/CEL and on
objective response rates in adult patients with unresectable and/or metastatic GIST and DFSP and on
recurrence-free survival in adjuvant GIST. The experience with Glivec in patients with MDS/MPD
associated with PDGFR gene re-arrangements is very limited (see section 5.1). Except in newly
diagnosed chronic phase CML, there are no controlled trials demonstrating a clinical benefit or
increased survival for these diseases.
2
4.2 Posology and method of administration
Therapy should be initiated by a physician experienced in the treatment of patients with
haematological malignancies and malignant sarcomas, as appropriate.
The prescribed dose should be administered orally with a meal and a large glass of water to minimise
the risk of gastrointestinal irritations. Doses of 400 mg or 600 mg should be administered once daily,
whereas a daily dose of 800 mg should be administered as 400 mg twice a day, in the morning and in
the evening. For patients (children) unable to swallow the capsules, their content may be diluted in a
glass of either still water or apple juice. Since studies in animals have shown reproductive toxicity, and
the potential risk for the human foetus is unknown, women of child-bearing potential who open
capsules should be advised to handle the contents with caution and avoid skin-eye contact or
inhalation (see section 4.6). Hands should be washed immediately after handling open capsules.
Posology for CML in adult patients
The recommended dosage of Glivec is 400 mg/day for patients in chronic phase CML. Chronic phase
CML is defined when all of the following criteria are met: blasts < 15% in blood and bone marrow,
peripheral blood basophils < 20%, platelets > 100 x 10 9 /l.
The recommended dosage of Glivec is 600 mg/day for patients in accelerated phase. Accelerated
phase is defined by the presence of any of the following: blasts  15% but < 30% in blood or bone
marrow, blasts plus promyelocytes  30% in blood or bone marrow (providing < 30% blasts),
peripheral blood basophils  20%, platelets < 100 x 10 9 /l unrelated to therapy.
The recommended dose of Glivec is 600 mg/day for patients in blast crisis. Blast crisis is defined as
blasts  30% in blood or bone marrow or extramedullary disease other than hepatosplenomegaly.
Treatment duration: In clinical trials, treatment with Glivec was continued until disease progression.
The effect of stopping treatment after the achievement of a complete cytogenetic response has not
been investigated.
Dose increases from 400 mg to 600 mg or 800 mg in patients with chronic phase disease, or from
600 mg to a maximum of 800 mg (given as 400 mg twice daily) in patients with accelerated phase or
blast crisis may be considered in the absence of severe adverse drug reaction and severe non-
leukaemia-related neutropenia or thrombocytopenia in the following circumstances: disease
progression (at any time); failure to achieve a satisfactory haematological response after at least
3 months of treatment; failure to achieve a cytogenetic response after 12 months of treatment; or loss
of a previously achieved haematological and/or cytogenetic response. Patients should be monitored
closely following dose escalation given the potential for an increased incidence of adverse reactions at
higher dosages.
Posology for CML in children
Dosing for children should be on the basis of body surface area (mg/m 2 ). The dose of 340 mg/m 2 daily
is recommended for children with chronic phase CML and advanced phase CML (not to exceed the
total dose of 800 mg). Treatment can be given as a once daily dose or alternatively the daily dose may
be split into two administrations – one in the morning and one in the evening. The dose
recommendation is currently based on a small number of paediatric patients (see sections 5.1 and 5.2).
There is no experience with the treatment of children below 2 years of age.
Dose increases from 340 mg/m 2 daily to 570 mg/m 2 daily (not to exceed the total dose of 800 mg) may
be considered in children in the absence of severe adverse drug reaction and severe non-leukaemia-
related neutropenia or thrombocytopenia in the following circumstances: disease progression (at any
time); failure to achieve a satisfactory haematological response after at least 3 months of treatment;
failure to achieve a cytogenetic response after 12 months of treatment; or loss of a previously achieved
haematological and/or cytogenetic response. Patients should be monitored closely following dose
escalation given the potential for an increased incidence of adverse reactions at higher dosages.
3
Posology for Ph+ ALL
The recommended dose of Glivec is 600 mg/day for patients with Ph+ ALL. Haematological experts
in the management of this disease should supervise the therapy throughout all phases of care.
Treatment schedule: On the basis of the existing data, Glivec has been shown to be effective and safe
when administered at 600 mg/day in combination with chemotherapy in the induction phase, the
consolidation and maintenance phases of chemotherapy (see section 5.1) for adult patients with newly
diagnosed Ph+ ALL. The duration of Glivec therapy can vary with the treatment programme selected,
but generally longer exposures to Glivec have yielded better results.
For adult patients with relapsed or refractory Ph+ALL Glivec monotherapy at 600 mg/day is safe,
effective and can be given until disease progression occurs.
Posology for MDS/MPD
The recommended dose of Glivec is 400 mg/day for patients with MDS/MPD.
Treatment duration: In the only clinical trial performed up to now, treatment with Glivec was
continued until disease progression (see section 5.1). At the time of analysis, the treatment duration
was a median of 47 months (24 days - 60 months).
Posology for HES/CEL
The recommended dose of Glivec is 100 mg/day for patients with HES/CEL.
Dose increase from 100 mg to 400 mg may be considered in the absence of adverse drug reactions if
assessments demonstrate an insufficient response to therapy.
Treatment should be continued as long as the patient continues to benefit.
Posology for GIST
The recommended dose of Glivec is 400 mg/day for patients with unresectable and/or metastatic
malignant GIST.
Limited data exist on the effect of dose increases from 400 mg to 600 mg or 800 mg in patients
progressing at the lower dose (see section 5.1).
Treatment duration: In clinical trials in GIST patients, treatment with Glivec was continued until
disease progression. At the time of analysis, the treatment duration was a median of 7 months (7 days
to 13 months). The effect of stopping treatment after achieving a response has not been investigated.
The recommended dose of Glivec is 400 mg/day for the adjuvant treatment of adult patients following
resection of GIST. Optimal treatment duration is not yet established. Length of treatment in the
clinical trial supporting this indication was 12 months.
Posology for DFSP
The recommended dose of Glivec is 800 mg/day for patients with DFSP.
Dose adjustment for adverse reactions
Non-haematological adverse reactions
If a severe non-haematological adverse reaction develops with Glivec use, treatment must be withheld
until the event has resolved. Thereafter, treatment can be resumed as appropriate depending on the
initial severity of the event.
If elevations in bilirubin > 3 x institutional upper limit of normal (IULN) or in liver transaminases
> 5 x IULN occur, Glivec should be withheld until bilirubin levels have returned to < 1.5 x IULN and
transaminase levels to < 2.5 x IULN. Treatment with Glivec may then be continued at a reduced daily
dose. In adults the dose should be reduced from 400 to 300 mg or from 600 to 400 mg, or from
800 mg to 600 mg, and in children from 340 to 260 mg/m 2 /day.
4
Haematological adverse reactions
Dose reduction or treatment interruption for severe neutropenia and thrombocytopenia are
recommended as indicated in the table below.
Dose adjustments for neutropenia and thrombocytopenia:
HES/CEL (starting dose
100 mg)
ANC < 1.0 x 10 9 /l
and/or
platelets < 50 x 10 9 /l
1. Stop Glivec until ANC  1.5 x 10 9 /l and
platelets  75 x 10 9 /l.
2. Resume treatment with Glivec at previous
dose (i.e. before severe adverse reaction).
Chronic phase CML,
MDS/MPD and GIST
(starting dose 400 mg)
HES/CEL
(at dose 400 mg)
ANC < 1.0 x 10 9 /l
and/or
platelets < 50 x 10 9 /l
1. Stop Glivec until ANC  1.5 x 10 9 /l and
platelets  75 x 10 9 /l.
2. Resume treatment with Glivec at previous
dose (i.e. before severe adverse reaction).
3. In the event of recurrence of ANC
< 1.0 x 10 9 /l and/or platelets < 50 x 10 9 /l,
repeat step 1 and resume Glivec at
reduced dose of 300 mg.
Paediatric chronic phase
CML
(at dose 340 mg/m 2 )
ANC < 1.0 x 10 9 /l
and/or
platelets < 50 x 10 9 /l
1. Stop Glivec until ANC  1.5 x 10 9 /l and
platelets  75 x 10 9 /l.
2. Resume treatment with Glivec at previous
dose (i.e. before severe adverse reaction).
3. In the event of recurrence of ANC
< 1.0 x10 9 /l and/or platelets < 50 x10 9 /l,
repeat step 1 and resume Glivec at
reduced dose of 260 mg/m 2 .
Accelerated phase CML
and blast crisis and Ph+
ALL (starting dose
600 mg)
a ANC < 0.5 x 10 9 /l
and/or
platelets < 10 x 10 9 /l
1. Check whether cytopenia is related to
leukaemia (marrow aspirate or biopsy).
2. If cytopenia is unrelated to leukaemia,
reduce dose of Glivec to 400 mg.
3. If cytopenia persists for 2 weeks, reduce
further to 300 mg.
4. If cytopenia persists for 4 weeks and is
still unrelated to leukaemia, stop Glivec
until ANC  1 x 10 9 /l and platelets
 20 x 10 9 /l, then resume treatment at
300 mg.
Paediatric accelerated
phase CML and blast
crisis (starting dose
340 mg/m 2 )
a ANC < 0.5 x 10 9 /l
and/or
platelets < 10 x 10 9 /l
1. Check whether cytopenia is related to
leukaemia (marrow aspirate or biopsy).
2. If cytopenia is unrelated to leukaemia,
reduce dose of Glivec to 260 mg/m 2 .
3. If cytopenia persists for 2 weeks, reduce
further to 200 mg/m 2 .
4. If cytopenia persists for 4 weeks and is
still unrelated to leukaemia, stop Glivec
until ANC  1 x 10 9 /l and platelets
 20 x 10 9 /l, then resume treatment at
200 mg/m 2 .
5
DFSP
(at dose 800 mg)
ANC < 1.0 x 10 9 /l
and/or
platelets < 50 x 10 9 /l
1. Stop Glivec until ANC  1.5 x 10 9 /l and
platelets  75 x 10 9 /l.
2. Resume treatment with Glivec at 600 mg.
3. In the event of recurrence of ANC
< 1.0 x 10 9 /l and/or platelets < 50 x 10 9 /l,
repeat step 1 and resume Glivec at
reduced dose of 400 mg.
ANC = absolute neutrophil count
a occurring after at least 1 month of treatment
Paediatric use: There is no experience in children with CML below 2 years of age (see section 5.1).
There is limited experience in children with Ph+ ALL and very limited experience in children with
MDS/MPD and DFSP. There is no experience in children or adolescents with GIST and HES/CEL.
Hepatic insufficiency: Imatinib is mainly metabolised through the liver. Patients with mild, moderate
or severe liver dysfunction should be given the minimum recommended dose of 400 mg daily. The
dose can be reduced if not tolerated (see sections 4.4, 4.8 and 5.2).
Liver dysfunction classification:
Liver dysfunction Liver function tests
Mild Total bilirubin: = 1.5 ULN
AST: >ULN (can be normal or <ULN if total bilirubin is
>ULN)
Moderate Total bilirubin: >1.5–3.0 ULN
AST: any
Severe Total bilirubin: >3–10 ULN
AST: any
ULN = upper limit of normal for the institution
AST = aspartate aminotransferase
Renal insufficiency: Since the renal clearance of imatinib is negligible, a decrease in free imatinib
clearance is not expected in patients with renal insufficiency. Patients with mild or moderate renal
dysfunction (creatinine clearance = 20–59 ml/min) should be given the minimum recommended dose
of 400 mg daily as starting dose. Although very limited information is available, patients with severe
renal dysfunction (creatinine clearance = < 20 ml/min) or on dialysis could also start at the same dose
of 400 mg. However, in these patients caution is recommended. The dose can be reduced if not
tolerated, or increased for lack of efficacy (see sections 4.4 and 5.2).
Elderly patients: Imatinib pharmacokinetics have not been specifically studied in the elderly. No
significant age-related pharmacokinetic differences have been observed in adult patients in clinical
trials which included over 20% of patients age 65 and older. No specific dose recommendation is
necessary in the elderly.
4.3 Contraindications
Hypersensitivity to the active substance or to any of the excipients.
4.4 Special warnings and precautions for use
When Glivec is co-administered with other medicinal products, there is a potential for drug
interactions (see section 4.5).
Concomitant use of imatinib and medicinal products that induce CYP3A4 (e.g. dexamethasone,
phenytoin, carbamazepine, rifampicin, phenobarbital or Hypericum perforatum , also known as St.
6
John’s Wort) may significantly reduce exposure to Glivec, potentially increasing the risk of
therapeutic failure. Therefore, concomitant use of strong CYP3A4 inducers and imatinib should be
avoided (see section 4.5).
Clinical cases of hypothyroidism have been reported in thyroidectomy patients undergoing
levothyroxine replacement during treatment with Glivec (see section 4.5). TSH levels should be
closely monitored in such patients.
Metabolism of Glivec is mainly hepatic, and only 13% of excretion is through the kidneys. In patients
with hepatic dysfunction (mild, moderate or severe), peripheral blood counts and liver enzymes should
be carefully monitored (see sections 4.2, 4.8 and 5.2). It should be noted that GIST patients may have
hepatic metastases which could lead to hepatic impairment.
Cases of liver injury, including hepatic failure and hepatic necrosis, have been observed with imatinib.
When imatinib is combined with high dose chemotherapy regimens, an increase in serious hepatic
reactions has been detected. Hepatic function should be carefully monitored in circumstances where
imatinib is combined with chemotherapy regimens also known to be associated with hepatic
dysfunction (see section 4.5 and 4.8).
Occurrences of severe fluid retention (pleural effusion, oedema, pulmonary oedema, ascites,
superficial oedema) have been reported in approximately 2.5% of newly diagnosed CML patients
taking Glivec. Therefore, it is highly recommended that patients be weighed regularly. An unexpected
rapid weight gain should be carefully investigated and if necessary appropriate supportive care and
therapeutic measures should be undertaken. In clinical trials, there was an increased incidence of these
events in elderly patients and those with a prior history of cardiac disease. Therefore, caution should
be exercised in patients with cardiac dysfunction.
Patients with cardiac disease or risk factors for cardiac failure should be monitored carefully, and any
patient with signs or symptoms consistent with cardiac failure should be evaluated and treated.
In patients with hypereosinophilic syndrome (HES) and cardiac involvement, isolated cases of
cardiogenic shock/left ventricular dysfunction have been associated with the initiation of imatinib
therapy. The condition was reported to be reversible with the administration of systemic steroids,
circulatory support measures and temporarily withholding imatinib. As cardiac adverse events have
been reported uncommonly with imatinib, a careful assessment of the benefit/risk of imatinib therapy
should be considered in the HES/CEL population before treatment initiation.
Myelodysplastic/myeloproliferative diseases with PDGFR gene re-arrangements could be associated
with high eosinophil levels. Evaluation by a cardiology specialist, performance of an echocardiogram
and determination of serum troponin should therefore be considered in patients with HES/CEL, and in
patients with MDS/MPD associated with high eosinophil levels before imatinib is administered. If
either is abnormal, follow-up with a cardiology specialist and the prophylactic use of systemic steroids
(1–2 mg/kg) for one to two weeks concomitantly with imatinib should be considered at the initiation
of therapy.
In the study in patients with unresectable and/or metastatic GIST, both gastrointestinal and intra-
tumoural haemorrhages were reported (see section 4.8). Based on the available data, no predisposing
factors (e.g. tumour size, tumour location, coagulation disorders) have been identified that place
patients with GIST at a higher risk of either type of haemorrhage. Since increased vascularity and
propensity for bleeding is a part of the nature and clinical course of GIST, standard practices and
procedures for the monitoring and management of haemorrhage in all patients should be applied.
Due to the possible occurrence of tumour lysis syndrome (TLS), correction of clinically significant
dehydration and treatment of high uric acid levels are recommended prior to initiation of Glivec (see
section 4.8).
7
Laboratory tests
Complete blood counts must be performed regularly during therapy with Glivec. Treatment of CML
patients with Glivec has been associated with neutropenia or thrombocytopenia. However, the
occurrence of these cytopenias is likely to be related to the stage of the disease being treated and they
were more frequent in patients with accelerated phase CML or blast crisis as compared to patients with
chronic phase CML. Treatment with Glivec may be interrupted or the dose may be reduced, as
recommended in section 4.2.
Liver function (transaminases, bilirubin, alkaline phosphatase) should be monitored regularly in
patients receiving Glivec.
In patients with impaired renal function, imatinib plasma exposure seems to be higher than that in
patients with normal renal function, probably due to an elevated plasma level of alpha-acid
glycoprotein (AGP), an imatinib-binding protein, in these patients. Patients with renal impairment
should be given the minimum starting dose. Patients with severe renal impairment should be treated
with caution. The dose can be reduced if not tolerated (see section 4.2 and 5.2).
Children and adolescents
There have been case reports of growth retardation occurring in children and pre-adolescents receiving
imatinib. The long-term effects of prolonged treatment with imatinib on growth in children are
unknown. Therefore, close monitoring of growth in children under imatinib treatment is recommended
(see section 4.8).
4.5 Interaction with other medicinal products and other forms of interaction
Active substances that may increase imatinib plasma concentrations:
Substances that inhibit the cytochrome P450 isoenzyme CYP3A4 activity (e.g. ketoconazole,
itraconazole, erythromycin, clarithromycin) could decrease metabolism and increase imatinib
concentrations. There was a significant increase in exposure to imatinib (the mean C max and AUC of
imatinib rose by 26% and 40%, respectively) in healthy subjects when it was co-administered with a
single dose of ketoconazole (a CYP3A4 inhibitor). Caution should be taken when administering
Glivec with inhibitors of the CYP3A4 family.
Active substances that may decrease imatinib plasma concentrations:
Substances that are inducers of CYP3A4 activity could increase metabolism and decrease imatinib
plasma concentrations. Co-medications which induce CYP3A4 (e.g. dexamethasone, phenytoin,
carbamazepine, rifampicin, phenobarbital, fosphenytoin, primidone or Hypericum perforatum , also
known as St. John’s Wort) may significantly reduce exposure to Glivec, potentially increasing the risk
of therapeutic failure. Pretreatment with multiple doses of rifampicin 600 mg followed by a single
400 mg dose of Glivec resulted in decrease in C max and AUC (0-∞) by at least 54% and 74%, of the
respective values without rifampicin treatment. Similar results were observed in patients with
malignant gliomas treated with Glivec while taking enzyme-inducing anti-epileptic drugs (EIAEDs)
such as carbamazepine, oxcarbazepine and phenytoin. The plasma AUC for imatinib decreased by
73% compared to patients not on EIAEDs. Concomitant use of rifampicin or other strong CYP3A4
inducers and imatinib should be avoided.
Active substances that may have their plasma concentration altered by Glivec
Imatinib increases the mean C max and AUC of simvastatin (CYP3A4 substrate) 2- and 3.5-fold,
respectively, indicating an inhibition of the CYP3A4 by imatinib. Therefore, caution is recommended
when administering Glivec with CYP3A4 substrates with a narrow therapeutic window (e.g.
cyclosporin or pimozide). Glivec may increase plasma concentration of other CYP3A4 metabolised
drugs (e.g. triazolo-benzodiazepines, dihydropyridine calcium channel blockers, certain HMG-CoA
reductase inhibitors, i.e. statins, etc.).
Because warfarin is metabolised by CYP2C9, patients who require anticoagulation should receive
low-molecular-weight or standard heparin.
8
In vitro Glivec inhibits the cytochrome P450 isoenzyme CYP2D6 activity at concentrations similar to
those that affect CYP3A4 activity. Imatinib at 400 mg twice daily had an inhibitory effect on
CYP2D6-mediated metoprolol metabolism, with metoprolol C max and AUC being increased by
approximately 23% (90%CI [1.16-1.30]). Dose adjustments do not seem to be necessary when
imatinib is co-administrated with CYP2D6 substrates, however caution is advised for CYP2D6
substrates with a narrow therapeutic window such as metoprolol. In patients treated with metoprolol
clinical monitoring should be considered.
In vitro , Glivec inhibits paracetamol O-glucuronidation (Ki value of 58.5 micromol/l at therapeutic
levels).
Caution should therefore be exercised when using Glivec and paracetamol concomitantly, especially
with high doses of paracetamol.
In thyroidectomy patients receiving levothyroxine, the plasma exposure to levothyroxine may be
decreased when Glivec is co-administered (see section 4.4). Caution is therefore recommended.
However, the mechanism of the observed interaction is presently unknown.
In Ph+ ALL patients, there is clinical experience of co-administering Glivec with chemotherapy (see
section 5.1), but drug-drug interactions between imatinib and chemotherapy regimens are not well
characterised. Imatinib adverse events, i.e. hepatotoxicity, myelosuppression or others, may increase
and it has been reported that concomitant use with L-asparaginase could be associated with increased
hepatotoxicity (see section 4.8). Therefore, the use of Glivec in combination requires special
precaution.
4.6 Fertility, pregnancy and lactation
Pregnancy
There are no adequate data on the use of imatinib in pregnant women. Studies in animals have
however shown reproductive toxicity (see section 5.3) and the potential risk for the foetus is unknown.
Glivec should not be used during pregnancy unless clearly necessary. If it is used during pregnancy,
the patient must be informed of the potential risk to the foetus. Women of childbearing potential must
be advised to use effective contraception during treatment.
Breast-feeding
There is limited information on imatinib distribution on human milk. Studies in two breast-feeding
women revealed that both imatinib and its active metabolite can be distributed into human milk. The
milk plasma ratio studied in a single patient was determined to be 0.5 for imatinib and 0.9 for the
metabolite, suggesting greater distribution of the metabolite into the milk. Considering the combined
concentration of imatinib and the metabolite and the maximum daily milk intake by infants, the total
exposure would be expected to be low (~10% of a therapeutic dose). However, since the effects of
low-dose exposure of the infant to imatinib are unknown, women taking imatinib should not breast-
feed.
4.7 Effects on ability to drive and use machines
No studies on the effects on the ability to drive and use machines have been performed. However,
patients should be advised that they may experience undesirable effects such as dizziness or blurred
vision during treatment with imatinib. Therefore, caution should be recommended when driving a car
or operating machinery.
9
4.8 Undesirable effects
Patients with advanced stages of malignancies may have numerous confounding medical conditions
that make causality of adverse reactions difficult to assess due to the variety of symptoms related to
the underlying disease, its progression, and the co-administration of numerous medicinal products.
In clinical trials in CML, drug discontinuation for drug-related adverse reactions was observed in 2.4%
of newly diagnosed patients, 4% of patients in late chronic phase after failure of interferon therapy,
4% of patients in accelerated phase after failure of interferon therapy and 5% of blast crisis patients
after failure of interferon therapy. In GIST the study drug was discontinued for drug-related adverse
reactions in 4% of patients.
The adverse reactions were similar in all indications, with two exceptions. There was more
myelosuppression seen in CML patients than in GIST, which is probably due to the underlying
disease. In the study in patients with unresectable and/or metastatic GIST, 7 (5%) patients experienced
CTC grade 3/4 GI bleeds (3 patients), intra-tumoural bleeds (3 patients) or both (1 patient). GI tumour
sites may have been the source of the GI bleeds (see section 4.4). GI and tumoural bleeding may be
serious and sometimes fatal. The most commonly reported ( 10%) drug-related adverse reactions in
both settings were mild nausea, vomiting, diarrhoea, abdominal pain, fatigue, myalgia, muscle cramps
and rash. Superficial oedemas were a common finding in all studies and were described primarily as
periorbital or lower limb oedemas. However, these oedemas were rarely severe and may be managed
with diuretics, other supportive measures, or by reducing the dose of Glivec.
When imatinib was combined with high dose chemotherapy in Ph+ ALL patients, transient liver
toxicity in the form of transaminase elevation and hyperbilirubinaemia were observed.
Miscellaneous adverse reactions such as pleural effusion, ascites, pulmonary oedema and rapid weight
gain with or without superficial oedema may be collectively described as “fluid retention”. These
reactions can usually be managed by withholding Glivec temporarily and with diuretics and other
appropriate supportive care measures. However, some of these reactions may be serious or life-
threatening and several patients with blast crisis died with a complex clinical history of pleural
effusion, congestive heart failure and renal failure. There were no special safety findings in paediatric
clinical trials.
Adverse reactions
Adverse reactions reported as more than an isolated case are listed below, by system organ class and
by frequency. Frequency categories are defined using the following convention: very common
(≥1/10), common (≥1/100 to <1/10), uncommon (≥1/1,000 to <1/100), rare (≥1/10,000 to <1/1,000),
very rare (<1/10,000), not known (cannot be estimated from the available data).
Within each frequency grouping, undesirable effects are presented in order of frequency, the most
frequent first.
10
Adverse reactions and their frequencies reported in Table 1 are based on the main registration studies.
Table 1
Adverse reactions in clinical studies
Infections and infestati ons
Uncommon: Herpes zoster, herpes simplex, nasopharyngitis, pneumonia 1 , sinusitis, cellulitis,
upper respiratory tract infection, influenza, urinary tract infection, gastroenteritis,
sepsis
Rare: Fungal infection
Neoplasm benign, mali gnant and unspecified (including cysts and polyps)
Rare: Tumour lysis syndrome
Blood and lymphatic sy stem disorders
Very common: Neutropenia, thrombocytopenia, anaemia
Common: Pancytopenia, febrile neutropenia
Uncommon: Thrombocythaemia, lymphopenia, bone marrow depression, eosinophilia,
lymphadenopathy
Rare: Haemolytic anaemia
Metabolism and nutrit ion disorders
Common:
Anorexia
Uncommon:
Hypokalaemia, increased appetite, hypophosphataemia, decreased appetite,
dehydration, gout, hyperuricaemia, hypercalcaemia, hyperglycaemia,
hyponatraemia
Rare:
Hyperkalaemia, hypomagnesaemia
Psychiatric disorders
Common: Insomnia
Uncommon: Depression, libido decreased, anxiety
Rare: Confusional state
Nervous system disord ers
Very common:
Headache 2
Common:
Dizziness, paraesthesia, taste disturbance, hypoaesthesia
Uncommon:
Migraine, somnolence, syncope, peripheral neuropathy, memory impairment,
sciatica, restless leg syndrome, tremor, cerebral haemorrhage
Rare:
Increased intracranial pressure, convulsions, optic neuritis
Eye disorders
Common: Eyelid oedema, lacrimation increased, conjunctival haemorrhage, conjunctivitis, dry
eye, blurred vision
Uncommon: Eye irritation, eye pain, orbital oedema, scleral haemorrhage, retinal haemorrhage,
blepharitis, macular oedema
Rare: Cataract, glaucoma, papilloedema
Ear and labyrinth diso rders
Uncommon:
Vertigo, tinnitus, hearing loss
Cardiac disorders
Uncommon:
Palpitations, tachycardia, cardiac failure congestive 3 , pulmonary oedema
Rare:
Arrhythmia, atrial fibrillation, cardiac arrest, myocardial infarction, angina pectoris,
pericardial effusion
Vascular disorders 4
Common: Flushing, haemorrhage
Uncommon: Hypertension, haematoma, peripheral coldness, hypotension, Raynaud’s
phenomenon
Respiratory, thoracic a nd mediastinal disorders
Common:
Dyspnoea, epistaxis, cough
Uncommon:
Pleural effusion 5 , pharyngolaryngeal pain, pharyngitis
Rare:
Pleuritic pain, pulmonary fibrosis, pulmonary hypertension, pulmonary
haemorrhage
11
 
Gastrointestinal disord ers
Very common: Nausea, diarrhoea, vomiting, dyspepsia, abdominal pain 6
Common: Flatulence, abdominal distension, gastro-oesophageal reflux, constipation, dry
mouth, gastritis
Uncommon: Stomatitis, mouth ulceration, gastrointestinal haemorrhage 7 , eructation, melaena,
oesophagitis, ascites, gastric ulcer, haematemesis, cheilitis, dysphagia, pancreatitis
Rare: Colitis, ileus, inflammatory bowel disease
Hepatobiliary disorder s
Common: Increased hepatic enzymes
Uncommon: Hyperbilirubinaemia, hepatitis, jaundice
Rare: Hepatic failure 8 , hepatic necrosis
Skin and subcutaneous tissue disorders
Very common: Periorbital oedema, dermatitis/eczema/rash
Common: Pruritus, face oedema, dry skin, erythema, alopecia, night sweats, photosensitivity
reaction
Uncommon: Rash pustular, contusion, sweating increased, urticaria, ecchymosis, increased
tendency to bruise, hypotrichosis, skin hypopigmentation, dermatitis exfoliative,
onychoclasis, folliculitis, petechiae, psoriasis, purpura, skin hyperpigmentation,
bullous eruptions
Rare: Acute febrile neutrophilic dermatosis (Sweet’s syndrome), nail discolouration,
angioneurotic oedema, rash vesicular, erythema multiforme, leucocytoclastic
vasculitis, Stevens-Johnson syndrome, acute generalised exanthematous pustulosis
(AGEP)
Musculoskeletal and co nnective tissue disorders
Very common: Muscle spasm and cramps, musculoskeletal pain including myalgia, arthralgia, bone
pain 9
Common: Joint swelling
Uncommon: Joint and muscle stiffness
Rare: Muscular weakness, arthritis, rhabdomyolysis/myopathy
Renal and urinary diso rders
Uncommon: Renal pain, haematuria, renal failure acute, urinary frequency increased
Reproductive system a nd breast disorders
Uncommon: Gynaecomastia, erectile dysfunction, menorrhagia, menstruation irregular, sexual
dysfunction, nipple pain, breast enlargement, scrotal oedema
Rare: Haemorrhagic corpus luteum/haemorrhagic ovarian cyst
General disorders and administration site conditions
Very common:
Fluid retention and oedema, fatigue
Common:
Weakness, pyrexia, anasarca, chills, rigors
Uncommon:
Chest pain, malaise
12
 
Investigations
Very common:
Weight increased
Common :
Weight decreased
Uncommon :
Blood creatinine increased, blood creatine phosphokinase increased, blood lactate
dehydrogenase increased, blood alkaline phosphatase increased
Rare:
Blood amylase increased
1 Pneumonia was reported most commonly in patients with transformed CML and in patients with
GIST.
2 Headache was the most common in GIST patients.
3 On a patient-year basis, cardiac events including congestive heart failure were more commonly
observed in patients with transformed CML than in patients with chronic CML.
4 Flushing was most common in GIST patients and bleeding (haematoma, haemorrhage) was
most common in patients with GIST and with transformed CML (CML-AP and CML-BC).
5 Pleural effusion was reported more commonly in patients with GIST and in patients with
transformed CML (CML-AP and CML-BC) than in patients with chronic CML.
6+7 Abdominal pain and gastrointestinal haemorrhage were most commonly observed in GIST
patients.
8
Musculoskeletal pain and related events were more commonly observed in patients with CML
than in GIST patients.
13
9
Some fatal cases of hepatic failure and of hepatic necrosis have been reported.
 
The following types of reactions have been reported mainly from post-marketing experience with
Glivec. This includes spontaneous case reports as well as serious adverse events from ongoing studies,
the expanded access programmes, clinical pharmacology studies and exploratory studies in
unapproved indications. Because these reactions are reported from a population of uncertain size, it is
not always possible to reliably estimate their frequency or establish a causal relationship to imatinib
exposure.
Table 2
Adverse reactions from post-marketing reports
Neoplasm benign, mali gnant and unspecified (including cysts and polyps)
Not known: Tumour haemorrhage/tumour necrosis
Immune system disord ers
Not known: Anaphylactic shock
Nervous system disord ers
Not known:
Cerebral oedema
Eye disorders
Not known:
Vitreous haemorrhage
Cardiac disorders
Not known:
Pericarditis, cardiac tamponade
Vascular disorders
Not known: Thrombosis/embolism
Respiratory, thoracic a nd mediastinal disorders
Not known: Acute respiratory failure 1 , interstitial lung disease
Gastrointestinal disord ers
Not known: Ileus/intestinal obstruction, gastrointestinal perforation, diverticulitis
Skin and subcutaneous tissue disorders
Not known: Palmoplantar erythrodysesthesia syndrome
Not known: Lichenoid keratosis, lichen planus
Not known: Toxic epidermal necrolysis
Musculoskeletal and co nnective tissue disorders
Not known:
Avascular necrosis/hip necrosis
Not known:
Growth retardation in children
1
Fatal cases have been reported in patients with advanced disease, severe infections, severe
neutropenia and other serious concomitant conditions.
Laboratory test abnormalities
Haematology
In CML, cytopenias, particularly neutropenia and thrombocytopenia, have been a consistent finding in
all studies, with the suggestion of a higher frequency at high doses  750 mg (phase I study).
However, the occurrence of cytopenias was also clearly dependent on the stage of the disease, the
frequency of grade 3 or 4 neutropenias (ANC < 1.0 x 10 9 /l) and thrombocytopenias (platelet count
< 50 x 10 9 /l) being between 4 and 6 times higher in blast crisis and accelerated phase (59–64% and
44–63% for neutropenia and thrombocytopenia, respectively) as compared to newly diagnosed
patients in chronic phase CML (16.7% neutropenia and 8.9% thrombocytopenia). In newly diagnosed
chronic phase CML grade 4 neutropenia (ANC < 0.5 x 10 9 /l) and thrombocytopenia (platelet count
< 10 x 10 9 /l) were observed in 3.6% and < 1% of patients, respectively. The median duration of the
neutropenic and thrombocytopenic episodes usually ranged from 2 to 3 weeks, and from 3 to 4 weeks,
respectively. These events can usually be managed with either a reduction of the dose or an
interruption of treatment with Glivec, but can in rare cases lead to permanent discontinuation of
treatment. In paediatric CML patients the most frequent toxicities observed were grade 3 or 4
cytopenias involving neutropenia, thrombocytopenia and anaemia. These generally occur within the
first several months of therapy.
14
 
In the study in patients with unresectable and/or metastatic GIST, grade 3 and 4 anaemia was reported
in 5.4% and 0.7% of patients, respectively, and may have been related to gastrointestinal or intra-
tumoural bleeding in at least some of these patients. Grade 3 and 4 neutropenia was seen in 7.5% and
2.7% of patients, respectively, and grade 3 thrombocytopenia in 0.7% of patients. No patient
developed grade 4 thrombocytopenia. The decreases in white blood cell (WBC) and neutrophil counts
occurred mainly during the first six weeks of therapy, with values remaining relatively stable
thereafter.
Biochemistry
Severe elevation of transaminases (<5%) or bilirubin (<1%) was seen in CML patients and was
usually managed with dose reduction or interruption (the median duration of these episodes was
approximately one week). Treatment was discontinued permanently because of liver laboratory
abnormalities in less than 1% of CML patients. In GIST patients (study B2222), 6.8% of grade 3 or 4
ALT (alanine aminotransferase) elevations and 4.8% of grade 3 or 4 AST (aspartate aminotransferase)
elevations were observed. Bilirubin elevation was below 3%.
There have been cases of cytolytic and cholestatic hepatitis and hepatic failure; in some of them
outcome was fatal, including one patient on high dose paracetamol.
4.9 Overdose
Experience with doses higher than the recommended therapeutic dose is limited. Isolated cases of
Glivec overdose have been reported spontaneously and in the literature. In the event of overdose the
patient should be observed and appropriate symptomatic treatment given. Generally the reported
outcome in these cases was “improved” or “recovered”. Events that have been reported at different
dose ranges are as follows:
Adult population
1200 to 1600 mg (duration varying between 1 to 10 days): Nausea, vomiting, diarrhoea, rash,
erythema, oedema, swelling, fatigue, muscle spasms, thrombocytopenia, pancytopenia, abdominal
pain, headache, decreased appetite.
1800 to 3200 mg (as high as 3200 mg daily for 6 days): Weakness, myalgia, increased creatine
phosphokinase, increased bilirubin, gastrointestinal pain.
6400 mg (single dose): One case reported in the literature of one patient who experienced nausea,
vomiting, abdominal pain, pyrexia, facial swelling, decreased neutrophil count, increased
transaminases.
8 to 10 g (single dose): Vomiting and gastrointestinal pain have been reported.
Paediatric population
One 3-year-old male exposed to a single dose of 400 mg experienced vomiting, diarrhoea and anorexia
and another 3-year-old male exposed to a single dose of 980 mg dose experienced decreased white
blood cell count and diarrhoea.
In the event of overdose, the patient should be observed and appropriate supportive treatment given.
5.
PHARMACOLOGICAL PROPERTIES
5.1 Pharmacodynamic properties
Pharmacotherapeutic group: protein-tyrosine kinase inhibitor, ATC code: L01XE01
Imatinib is a protein-tyrosine kinase inhibitor which potently inhibits the Bcr-Abl tyrosine kinase at
the in vitro , cellular and in vivo levels. The compound selectively inhibits proliferation and induces
apoptosis in Bcr-Abl positive cell lines as well as fresh leukaemic cells from Philadelphia chromosome
positive CML and acute lymphoblastic leukaemia (ALL) patients.
15
In vivo the compound shows anti-tumour activity as a single agent in animal models using Bcr-Abl
positive tumour cells.
Imatinib is also an inhibitor of the receptor tyrosine kinases for platelet-derived growth factor (PDGF),
PDGF-R, and stem cell factor (SCF), c-Kit, and inhibits PDGF- and SCF-mediated cellular events. In
vitro , imatinib inhibits proliferation and induces apoptosis in gastrointestinal stromal tumour (GIST)
cells, which express an activating kit mutation. Constitutive activation of the PDGF receptor or the
Abl protein tyrosine kinases as a consequence of fusion to diverse partner proteins or constitutive
production of PDGF have been implicated in the pathogenesis of MDS/MPD, HES/CEL and DFSP.
Imatinib inhibits signalling and proliferation of cells driven by dysregulated PDGFR and Abl kinase
activity.
Clinical studies in chronic myeloid leukaemia
The effectiveness of Glivec is based on overall haematological and cytogenetic response rates and
progression-free survival. Except in newly diagnosed chronic phase CML, there are no controlled
trials demonstrating a clinical benefit, such as improvement in disease-related symptoms or increased
survival.
Three large, international, open-label, non-controlled phase II studies were conducted in patients with
Philadelphia chromosome positive (Ph+) CML in advanced, blast or accelerated phase disease, other
Ph+ leukaemias or with CML in the chronic phase but failing prior interferon-alpha (IFN) therapy.
One large, open-label, multicentre, international randomised phase III study has been conducted in
patients with newly diagnosed Ph+ CML. In addition, children have been treated in two phase I
studies and one phase II study.
In all clinical studies 38–40% of patients were  60 years of age and 10–12% of patients were
 70 years of age.
Chronic phase, newly diagnosed: This phase III study in adult patients compared treatment with either
single-agent Glivec or a combination of interferon-alpha (IFN) plus cytarabine (Ara-C). Patients
showing lack of response (lack of complete haematological response (CHR) at 6 months, increasing
WBC, no major cytogenetic response (MCyR) at 24 months), loss of response (loss of CHR or MCyR)
or severe intolerance to treatment were allowed to cross over to the alternative treatment arm. In the
Glivec arm, patients were treated with 400 mg daily. In the IFN arm, patients were treated with a
target dose of IFN of 5 MIU/m 2 /day subcutaneously in combination with subcutaneous Ara-C
20 mg/m 2 /day for 10 days/month.
A total of 1,106 patients were randomised, 553 to each arm. Baseline characteristics were well
balanced between the two arms. Median age was 51 years (range 18–70 years), with 21.9% of patients
≥ 60 years of age. There were 59% males and 41% females; 89.9% caucasian and 4.7% black patients.
Seven years after the last patient had been recruited, the median duration of first-line treatment was 82
and 8 months in the Glivec and IFN arms, respectively. The median duration of second-line treatment
with Glivec was 64 months. Overall, in patients receiving first-line Glivec, the average daily dose
delivered was 406 ± 76 mg. The primary efficacy endpoint of the study is progression-free survival.
Progression was defined as any of the following events: progression to accelerated phase or blast
crisis, death, loss of CHR or MCyR, or in patients not achieving a CHR an increasing WBC despite
appropriate therapeutic management. Major cytogenetic response, haematological response, molecular
response (evaluation of minimal residual disease), time to accelerated phase or blast crisis and survival
are main secondary endpoints. Response data are shown in Table 3.
16
Table 3
Response in newly diagnosed CML Study (84-month data)
Glivec
IFN+Ara-C
(Best response rates)
n=553
n=553
Haematological response
CHR rate n (%)
534 (96.6%)*
313 (56.6%)*
[95% CI]
[94.7%, 97.9%]
[52.4%, 60.8%]
Cytogenetic response
Major response n (%)
490 (88.6%)*
129 (23.3%)*
[95% CI]
[85.7%, 91.1%]
[19.9%, 27.1%]
Complete CyR n (%)
456 (82.5%)*
64 (11.6%)*
Partial CyR n (%)
34 (6.1%)
65 (11.8%)
Molecular response **
Major response at 12 months (%)
153/305=50.2%
8/83=9.6%
Major response at 24 months (%)
73/104=70.2%
3/12=25%
Major response at 84 months (%)
102/116=87.9%
3/4=75%
* p<0.001, Fischer’s exact test
** molecular response percentages are based on available samples
Haematological response criteria (all responses to be confirmed after 4 weeks):
WBC < 10 x 10 9 /l, platelet < 450 x 10 9 /l, myelocyte+metamyelocyte < 5% in blood, no blasts and
promyelocytes in blood, basophils < 20%, no extramedullary involvement
Cytogenetic response criteria: complete (0% Ph+ metaphases), partial (1–35%), minor (36–65%) or
minimal (66–95%). A major response (0–35%) combines both complete and partial responses.
Major molecular response criteria : in the peripheral blood reduction of ≥ 3 logarithms in the
amount of Bcr-Abl transcripts (measured by real-time quantitative reverse transcriptase PCR assay)
over a standardised baseline.
Rates of complete haematological response, major cytogenetic response and complete cytogenetic
response on first-line treatment were estimated using the Kaplan-Meier approach, for which non-
responses were censored at the date of last examination. Using this approach, the estimated cumulative
response rates for first-line treatment with Glivec improved from 12 months of therapy to 84 months
of therapy as follows: CHR from 96.4% to 98.4% and CCyR from 69.5% to 87.2%, respectively.
With 7 years follow-up, there were 93 (16.8%) progression events in the Glivec arm: 37 (6.7%)
involving progression to accelerated phase/blast crisis, 31 (5.6%) loss of MCyR, 15 (2.7%) loss of
CHR or increase in WBC, and 10 (1.8%) CML unrelated deaths. In contrast, there were 165 (29.8%)
events in the IFN+Ara-C arm, of which 130 occurred during first-line treatment with IFN+Ara-C.
The estimated rate of patients free of progression to accelerated phase or blast crisis at 84 months was
significantly higher in the Glivec arm compared to the IFN arm (92.5% versus 85.1%, p<0.001). The
annual rate of progression to accelerated phase or blast crisis decreased with time on therapy and was
less than 1% annually in the fourth and fifth years. The estimated rate of progression-free survival at
84 months was 81.2% in the Glivec arm and 60.6% in the control arm (p<0.001). The yearly rates of
progression of any type for Glivec also decreased over time.
A total of 71 (12.8%) and 85 (15.4%) patients died in the Glivec and IFN+Ara-C groups, respectively.
At 84 months the estimated overall survival is 86.4% (83, 90) vs. 83.3% (80, 87) in the randomised
Glivec and the IFN+Ara-C groups, respectively (p=0.073, log-rank test). This time-to-event endpoint
is strongly affected by the high crossover rate from IFN+Ara-C to Glivec. The effect of Glivec
treatment on survival in chronic phase, newly diagnosed CML has been further examined in a
retrospective analysis of the above reported Glivec data with the primary data from another Phase III
study using IFN+Ara-C (n=325) in an identical regimen. In this retrospective analysis, the superiority
17
 
of Glivec over IFN+Ara-C in overall survival was demonstrated (p<0.001); within 42 months, 47
(8.5%) Glivec patients and 63 (19.4%) IFN+Ara-C patients had died.
The degree of cytogenetic response and molecular response had a clear effect on long-term outcomes
in patients on Glivec. Whereas an estimated 96% (93%) of patients with CCyR (PCyR) at 12 months
were free of progression to accelerated phase/blast crisis at 84 months, only 81% of patients without
MCyR at 12 months were free of progression to advanced CML at 84 months (p<0.001 overall,
p=0.25 between CCyR and PCyR). For patients with reduction in Bcr-Abl transcripts of at least
3 logarithms at 12 months, the probability of remaining free from progression to accelerated
phase/blast crisis was 99% at 84 months. Similar findings were found based on a 18-months landmark
analysis.
In this study, dose escalations were allowed from 400 mg daily to 600 mg daily, then from 600 mg
daily to 800 mg daily. After 42 months of follow-up, 11 patients experienced a confirmed loss (within
4 weeks) of their cytogenetic response. Of these 11 patients, 4 patients escalated up to 800 mg daily, 2
of whom regained a cytogenetic response (1 partial and 1 complete, the latter also achieving a
molecular response), while of the 7 patients who did not escalate the dose, only one regained a
complete cytogenetic response. The percentage of some adverse reactions was higher in the 40 patients
in whom the dose was increased to 800 mg daily compared to the population of patients before dose
increase (n=551). The more frequent adverse reactions included gastrointestinal haemorrhages,
conjunctivitis and elevation of transaminases or bilirubin. Other adverse reactions were reported with
lower or equal frequency.
Chronic phase, Interferon failure: 532 adult patients were treated at a starting dose of 400 mg. The
patients were distributed in three main categories: haematological failure (29%), cytogenetic failure
(35%), or intolerance to interferon (36%). Patients had received a median of 14 months of prior IFN
therapy at doses  25 x 10 6 IU/week and were all in late chronic phase, with a median time from
diagnosis of 32 months. The primary efficacy variable of the study was the rate of major cytogenetic
response (complete plus partial response, 0 to 35% Ph+ metaphases in the bone marrow).
In this study 65% of the patients achieved a major cytogenetic response that was complete in 53%
(confirmed 43%) of patients (Table 4). A complete haematological response was achieved in 95% of
patients.
Accelerated phase : 235 adult patients with accelerated phase disease were enrolled. The first
77 patients were started at 400 mg, the protocol was subsequently amended to allow higher dosing and
the remaining 158 patients were started at 600 mg.
The primary efficacy variable was the rate of haematological response, reported as either complete
haematological response, no evidence of leukaemia (i.e. clearance of blasts from the marrow and the
blood, but without a full peripheral blood recovery as for complete responses), or return to chronic
phase CML. A confirmed haematological response was achieved in 71.5% of patients (Table 4).
Importantly, 27.7% of patients also achieved a major cytogenetic response, which was complete in
20.4% (confirmed 16%) of patients. For the patients treated at 600 mg, the current estimates for
median progression-free-survival and overall survival were 22.9 and 42.5 months, respectively.
Myeloid blast crisis: 260 patients with myeloid blast crisis were enrolled. 95 (37%) had received prior
chemotherapy for treatment of either accelerated phase or blast crisis (“pretreated patients”) whereas
165 (63%) had not (“untreated patients”). The first 37 patients were started at 400 mg, the protocol
was subsequently amended to allow higher dosing and the remaining 223 patients were started at
600 mg.
The primary efficacy variable was the rate of haematological response, reported as either complete
haematological response, no evidence of leukaemia, or return to chronic phase CML using the same
criteria as for the study in accelerated phase. In this study, 31% of patients achieved a haematological
response (36% in previously untreated patients and 22% in previously treated patients). The rate of
response was also higher in the patients treated at 600 mg (33%) as compared to the patients treated at
18
400 mg (16%, p=0.0220). The current estimate of the median survival of the previously untreated and
treated patients was 7.7 and 4.7 months, respectively.
Lymphoid blast crisis : a limited number of patients were enrolled in phase I studies (n=10). The rate of
haematological response was 70% with a duration of 2 3 months.
Table 4
Response in adult CML studies
Study 0110
37-month data
Chronic phase,
IFN failure
(n=532)
Study 0109
40.5-month data
Accelerated phase
(n=235)
Study 0102
38-month data
Myeloid blast
crisis
(n=260)
% of patients (CI 95% )
Haematological response 1
95% (92.3–96.3)
71% (65.3–77.2)
31% (25.2–36.8)
Complete haematological
response (CHR)
95%
42%
8%
No evidence of leukaemia
(NEL)
Not applicable
12%
5%
Return to chronic phase (RTC)
Not applicable
17%
18%
Major cytogenetic response 2
65% (61.2–69.5)
28% (22.0–33.9)
15% (11.2–20.4)
Complete
53%
20%
7%
(Confirmed 3 ) [95% CI]
(43%) [38.6–47.2] (16%) [11.3–21.0]
(2%) [0.6–4.4]
Partial
12%
7%
8%
1 Haematological response criteria (all responses to be confirmed after 4 weeks):
CHR: Study 0110 [WBC < 10 x 10 9 /l, platelets < 450 x 10 9 /l, myelocyte+metamyelocyte < 5% in
blood, no blasts and promyelocytes in blood, basophils < 20%, no extramedullary
involvement] and in studies 0102 and 0109 [ANC  1.5 x 10 9 /l, platelets  100 x 10 9 /l, no
blood blasts, BM blasts < 5% and no extramedullary disease]
NEL Same criteria as for CHR but ANC  1 x 10 9 /l and platelets  20 x 10 9 /l (0102 and 0109
only)
RTC < 15% blasts BM and PB, < 30% blasts+promyelocytes in BM and PB, < 20% basophils in
PB, no extramedullary disease other than spleen and liver (only for 0102 and 0109).
BM = bone marrow, PB = peripheral blood
2 Cytogenetic response criteria:
A major response combines both complete and partial responses: complete (0% Ph+ metaphases),
partial (1–35%)
3 Complete cytogenetic response confirmed by a second bone marrow cytogenetic evaluation
performed at least one month after the initial bone marrow study.
Paediatric patients : A total of 26 paediatric patients of age < 18 years with either chronic phase CML
(n=11) or CML in blast crisis or Ph+ acute leukaemias (n=15) were enrolled in a dose-escalation phase
I trial. This was a population of heavily pretreated patients, as 46% had received prior BMT and 73% a
prior multi-agent chemotherapy. Patients were treated at doses of Glivec of 260 mg/m 2 /day (n=5),
340 mg/m 2 /day (n=9), 440 mg/m 2 /day (n=7) and 570 mg/m 2 /day (n=5). Out of 9 patients with chronic
phase CML and cytogenetic data available, 4 (44%) and 3 (33%) achieved a complete and partial
cytogenetic response, respectively, for a rate of MCyR of 77%.
A total of 51 paediatric patients with newly diagnosed and untreated CML in chronic phase have been
enrolled in an open-label, multicentre, single-arm phase II trial. Patients were treated with Glivec
340 mg/m 2 /day, with no interruptions in the absence of dose limiting toxicity. Glivec treatment
induces a rapid response in newly diagnosed paediatric CML patients with a CHR of 78% after
8 weeks of therapy. The high rate of CHR is accompanied by the development of a complete
cytogenetic response (CCyR) of 65% which is comparable to the results observed in adults.
Additionally, partial cytogenetic response (PCyR) was observed in 16% for a MCyR of 81%. The
19
 
majority of patients who achieved a CCyR developed the CCyR between months 3 and 10 with a
median time to response based on the Kaplan-Meier estimate of 5.6 months.
The European Medicines Agency has waived the obligation to submit the results of studies with
Glivec in all subsets of the paediatric population in Philadelphia chromosome (bcr-abl translocation)-
positive chronic myeloid leukaemia (see section 4.2 for information on paediatric use).
Clinical studies in Ph+ ALL
Newly diagnosed Ph+ ALL : In a controlled study (ADE10) of imatinib versus chemotherapy induction
in 55 newly diagnosed patients aged 55 years and over, imatinib used as single agent induced a
significantly higher rate of complete haematological response than chemotherapy (96.3% vs. 50%;
p=0.0001). When salvage therapy with imatinib was administered in patients who did not respond or
who responded poorly to chemotherapy, it resulted in 9 patients (81.8%) out of 11 achieving a
complete haematological response. This clinical effect was associated with a higher reduction in bcr-
abl transcripts in the imatinib-treated patients than in the chemotherapy arm after 2 weeks of therapy
(p=0.02). All patients received imatinib and consolidation chemotherapy (see Table 5) after induction
and the levels of bcr-abl transcripts were identical in the two arms at 8 weeks. As expected on the
basis of the study design, no difference was observed in remission duration, disease-free survival or
overall survival, although patients with complete molecular response and remaining in minimal
residual disease had a better outcome in terms of both remission duration (p=0.01) and disease-free
survival (p=0.02).
The results observed in a population of 211 newly diagnosed Ph+ ALL patients in four uncontrolled
clinical studies (AAU02, ADE04, AJP01 and AUS01) are consistent with the results described above.
Imatinib in combination with chemotherapy induction (see Table 5) resulted in a complete
haematological response rate of 93% (147 out of 158 evaluable patients) and in a major cytogenetic
response rate of 90% (19 out of 21 evaluable patients). The complete molecular response rate was 48%
(49 out of 102 evaluable patients). Disease-free survival (DFS) and overall survival (OS) constantly
exceeded 1 year and were superior to historical control (DFS p<0.001; OS p<0.0001) in two studies
(AJP01 and AUS01).
20
Table 5
Chemotherapy regimen used in combination with imatinib
Study ADE10
Prephase DEX 10 mg/m 2 oral, days 1-5; CP 200 mg/m 2 i.v., days 3, 4, 5; MTX
12 mg intrathecal, day 1
Remission induction DEX 10 mg/m 2 oral, days 6-7, 13-16; VCR 1 mg i.v., days 7, 14; IDA
8 mg/m 2 i.v. (0.5 h), days 7, 8, 14, 15; CP 500 mg/m 2 i.v.(1 h) day 1; Ara-
C 60 mg/m 2 i.v., days 22-25, 29-32
Consolidation
therapy I, III, V
MTX 500 mg/m 2 i.v. (24 h), days 1, 15; 6-MP 25 mg/m 2 oral, days 1-20
Consolidation
therapy II, IV
Ara-C 75 mg/m 2 i.v. (1 h), days 1-5; VM26 60 mg/m 2 i.v. (1 h), days 1-5
Study AAU02
Induction therapy ( de
novo Ph+ ALL)
Daunorubicin 30 mg/m 2 i.v., days 1-3, 15-16; VCR 2 mg total dose i.v.,
days 1, 8, 15, 22; CP 750 mg/m 2 i.v., days 1, 8; prednisone 60 mg/m 2
oral, days 1-7, 15-21; IDA 9 mg/m 2 oral, days 1-28; MTX 15 mg
intrathecal, days 1, 8, 15, 22; Ara-C 40 mg intrathecal, days 1, 8, 15, 22;
methylprednisolone 40 mg intrathecal, days 1, 8, 15, 22
Consolidation ( de
novo Ph+ ALL)
Ara-C 1,000 mg/m 2 /12 h i.v.(3 h), days 1-4; mitoxantrone 10 mg/m 2 i.v.
days 3-5; MTX 15 mg intrathecal, day 1; methylprednisolone 40 mg
intrathecal, day 1
Study ADE04
Prephase
DEX 10 mg/m 2 oral, days 1-5; CP 200 mg/m 2 i.v., days 3-5; MTX 15 mg
intrathecal, day 1
Induction therapy I
DEX 10 mg/m 2 oral, days 1-5; VCR 2 mg i.v., days 6, 13, 20;
daunorubicin 45 mg/m 2 i.v., days 6-7, 13-14
Induction therapy II
CP 1 g/m 2 i.v. (1 h), days 26, 46; Ara-C 75 mg/m 2 i.v. (1 h), days 28-31,
35-38, 42-45; 6-MP 60 mg/m 2 oral, days 26-46
Consolidation
therapy
DEX 10 mg/m 2 oral, days 1-5; vindesine 3 mg/m 2 i.v., day 1; MTX
1.5 g/m 2 i.v. (24 h), day 1; etoposide 250 mg/m 2 i.v. (1 h) days 4-5; Ara-
C 2x 2 g/m 2 i.v. (3 h, q 12 h), day 5
Study AJP01
Induction therapy
CP 1.2 g/m 2 i.v. (3 h), day 1; daunorubicin 60 mg/m 2 i.v. (1 h), days 1-3;
vincristine 1.3 mg/m 2 i.v., days 1, 8, 15, 21; prednisolone 60 mg/m 2 /day
oral
Consolidation
therapy
Alternating chemotherapy course: high dose chemotherapy with MTX
1 g/m 2 i.v. (24 h), day 1, and Ara-C 2 g/m 2 i.v. (q 12 h), days 2-3, for
4 cycles
Maintenance
VCR 1.3 g/m 2 i.v., day 1; prednisolone 60 mg/m 2 oral, days 1-5
Study AUS01
Hyper-CVAD regimen: CP 300 mg/m 2 i.v. (3 h, q 12 h), days 1-3;
vincristine 2 mg i.v., days 4, 11; doxorubicine 50 mg/m 2 i.v. (24 h),
day 4; DEX 40 mg/day on days 1-4 and 11-14, alternated with MTX
1 g/m 2 i.v. (24 h), day 1, Ara-C 1 g/m 2 i.v. (2 h, q 12 h), days 2-3 (total of
8 courses)
Maintenance VCR 2 mg i.v. monthly for 13 months; prednisolone 200 mg oral, 5 days
per month for 13 months
All treatment regimens include administration of steroids for CNS prophylaxis.
Ara-C: cytosine arabinoside; CP: cyclophosphamide; DEX: dexamethasone; MTX: methotrexate;
6-MP: 6-mercaptopurine VM26: Teniposide; VCR: vincristine; IDA: idarubicine; i.v.:
intravenous
21
Induction-
consolidation therapy
Relapsed/refractory Ph+ ALL: When imatinib was used as single agent in patients with
relapsed/refractory Ph+ ALL, it resulted, in the 53 out of 411 patients evaluable for response, in a
haematological response rate of 30% (9% complete) and a major cytogenetic response rate of 23%.
(Of note, out of the 411 patients, 353 were treated in an expanded access program without primary
response data collected.) The median time to progression in the overall population of 411 patients with
relapsed/refractory Ph+ ALL ranged from 2.6 to 3.1 months, and median overall survival in the
401 evaluable patients ranged from 4.9 to 9 months. The data was similar when re-analysed to include
only those patients age 55 or older.
Clinical studies in MDS/MPD
Experience with Glivec in this indication is very limited and is based on haematological and
cytogenetic response rates. There are no controlled trials demonstrating a clinical benefit or increased
survival. One open label, multicentre, phase II clinical trial (study B2225) was conducted testing
Glivec in diverse populations of patients suffering from life-threatening diseases associated with Abl,
Kit or PDGFR protein tyrosine kinases. This study included 7 patients with MDS/MPD who were
treated with Glivec 400 mg daily. Three patients presented a complete haematological response (CHR)
and one patient experienced a partial haematological response (PHR). At the time of the original
analysis, three of the four patients with detected PDGFR gene rearrangements developed
haematological response (2 CHR and 1 PHR). The age of these patients ranged from 20 to 72 years. In
addition a further 24 patients with MDS/MPD were reported in 13 publications. 21 patients were
treated with Glivec 400 mg daily, while the other 3 patients received lower doses. In eleven patients
PDGFR gene rearrangements was detected, 9 of them achieved a CHR and 1 PHR. The age of these
patients ranged from 2 to 79 years. In a recent publication updated information from 6 of these
11 patients revealed that all these patients remained in cytogenetic remission (range 32-38 months).
The same publication reported long term follow-up data from 12 MDS/MPD patients with PDGFR
gene rearrangements (5 patients from study B2225). These patients received Glivec for a median of
47 months (range 24 days – 60 months). In 6 of these patients follow-up now exceeds 4 years. Eleven
patients achieved rapid CHR; ten had complete resolution of cytogenetic abnormalities and a decrease
or disappearance of fusion transcripts as measured by RT-PCR. Haematological and cytogenetic
responses have been sustained for a median of 49 months (range 19-60) and 47 months (range 16-59),
respectively. The overall survival is 65 months since diagnosis (range 25-234). Glivec administration
to patients without the genetic translocation generally results in no improvement.
Clinical studies in HES/CEL
One open-label, multicentre, phase II clinical trial (study B2225) was conducted testing Glivec in
diverse populations of patients suffering from life-threatening diseases associated with Abl, Kit or
PDGFR protein tyrosine kinases. In this study, 14 patients with HES/CEL were treated with 100 mg to
1,000 mg of Glivec daily. A further 162 patients with HES/CEL, reported in 35 published case reports
and case series received Glivec at doses from 75 mg to 800 mg daily. Cytogenetic abnormalities were
evaluated in 117 of the total population of 176 patients. In 61 of these 117 patients FIP1L1-PDGFRα
fusion kinase was identified. An additional four HES patients were found to be FIP1L1-PDGFRα-
positive in other 3 published reports. All 65 FIP1L1-PDGFRα fusion kinase positive patients achieved
a CHR sustained for months (range from 1+ to 44+ months censored at the time of the reporting). As
reported in a recent publication 21 of these 65 patients also achieved complete molecular remission
with a median follow-up of 28 months (range 13-67 months). The age of these patients ranged from 25
to 72 years. Additionally, improvements in symptomatology and other organ dysfunction
abnormalities were reported by the investigators in the case reports. Improvements were reported in
cardiac, nervous, skin/subcutaneous tissue, respiratory/thoracic/mediastinal,
musculoskeletal/connective tissue/vascular, and gastrointestinal organ systems.
Clinical studies in unresectable and/or metastatic GIST
One phase II, open-label, randomised, uncontrolled multinational study was conducted in patients with
unresectable or metastatic malignant gastrointestinal stromal tumours (GIST). In this study
147 patients were enrolled and randomised to receive either 400 mg or 600 mg orally once daily for up
to 36 months. These patients ranged in age from 18 to 83 years old and had a pathologic diagnosis of
Kit-positive malignant GIST that was unresectable and/or metastatic. Immunohistochemistry was
22
routinely performed with Kit antibody (A-4502, rabbit polyclonal antiserum, 1:100; DAKO
Corporation, Carpinteria, CA) according to analysis by an avidin-biotin-peroxidase complex method
after antigen retrieval.
The primary evidence of efficacy was based on objective response rates. Tumours were required to be
measurable in at least one site of disease, and response characterisation based on Southwestern
Oncology Group (SWOG) criteria. Results are provided in Table 6.
Table 6
Best tumour response in trial STIB2222 (GIST)
Best response
All doses (n=147)
400 mg (n=73)
600 mg (n=74)
n (%)
Complete response
1 (0.7)
Partial response
98 (66.7)
Progressive disease
23 (15.6)
Not evaluable
5 (3.4)
Unknown
2 (1.4)
There were no differences in response rates between the two dose groups. A significant number of
patients who had stable disease at the time of the interim analysis achieved a partial response with
longer treatment (median follow-up 31 months). Median time to response was 13 weeks (95% C.I. 12–
23). Median time to treatment failure in responders was 122 weeks (95% C.I 106–147), while in the
overall study population it was 84 weeks (95% C.I 71–109). The median overall survival has not been
reached. The Kaplan-Meier estimate for survival after 36-month follow-up is 68%.
In two clinical studies (study B2222 and an intergroup study S0033) the daily dose of Glivec was
escalated to 800 mg in patients progressing at the lower daily doses of 400 mg or 600 mg. The daily
dose was escalated to 800 mg in a total of 103 patients; 6 patients achieved a partial response and 21
stabilisation of their disease after dose escalation for an overall clinical benefit of 26%. From the
safety data available, escalating the dose to 800 mg daily in patients progressing at lower doses of
400 mg or 600 mg daily does not seem to affect the safety profile of Glivec.
Clinical study in adjuvant GIST
In the adjuvant setting, Glivec was investigated in a multicentre, double-blind, long-term, placebo-
controlled phase III study (Z9001) involving 773 patients. The ages of these patients ranged from 18
to 91 years. Patients were included who had a histological diagnosis of primary GIST expressing Kit
protein by immunochemistry and a tumour size ≥ 3 cm in maximum dimension, with complete gross
resection of primary GIST within 14-70 days prior to registration. After resection of primary GIST,
patients were randomised to one of the two arms: Glivec at 400 mg/day or matching placebo for one
year.
The primary endpoint of the study was recurrence-free survival (RFS), defined as the time from date
of randomisation to the date of recurrence or death from any cause.
Glivec significantly prolonged RFS, with 75% of patients being recurrence-free at 38 months in the
Glivec group vs. 20 months in the placebo group (95% CIs, [30 - non-estimable]; [14 - non-estimable],
respectively); (hazard ratio = 0.398 [0.259-0.610], p<0.0001). At one year the overall RFS was
significantly better for Glivec (97.7%) vs. placebo (82.3%), (p<0.0001). The risk of recurrence was
thus reduced by approximately 89% as compared with placebo (hazard ratio = 0.113 [0.049-0.264]).
The risk of recurrence in patients after surgery of their primary GIST was retrospectively assessed
based on the following prognostic factors: tumour size, mitotic index, tumour location. Mitotic index
data were available for 556 of the 773 intention-to-treat (ITT) population. The results of subgroup
analyses according to the United States National Institutes of Health (NIH) and the Armed Forces
23
Stable disease
18 (12.2)
 
Institute of Pathology (AFIP) risk classifications are shown in Table 7. No benefit was observed in the
low and very low risk groups. No overall survival benefit has been observed.
Table 7
Summary of Z9001 trial RFS analyses by NIH and AFIP risk classifications
Risk
criteria
Risk Level
% of
patients
No. of events /
No. of patients
Overall hazard
ratio (95%CI)*
RFS rates (%)
12 month
24 month
Glivec vs placebo
Glivec vs
placebo
Glivec vs
placebo
Low
29.5
0/86 vs. 2/90
N.E.
100 vs. 98.7 100 vs. 95.5
NIH
Intermediate
25.7
4/75 vs. 6/78
0.59 (0.17; 2.10) 100 vs. 94.8 97.8 vs. 89.5
High
44.8
21/140 vs. 51/127
0.29 (0.18; 0.49) 94.8 vs. 64.0 80.7 vs. 46.6
Very Low
20.7
0/52 vs. 2/63
N.E.
100 vs. 98.1 100 vs. 93.0
AFIP
Low
25.0
2/70 vs. 0/69
N.E.
100 vs. 100
97.8 vs. 100
Moderate
24.6
2/70 vs. 11/67
0.16 (0.03; 0.70) 97.9 vs. 90.8 97.9 vs. 73.3
High
29.7
16/84 vs. 39/81
0.27 (0.15; 0.48) 98.7 vs. 56.1 79.9 vs. 41.5
* Full follow-up period; NE – Not estimable
Clinical studies in DFSP
One phase II, open label, multicentre clinical trial (study B2225) was conducted including 12 patients
with DFSP treated with Glivec 800 mg daily. The age of the DFSP patients ranged from 23 to
75 years; DFSP was metastatic, locally recurrent following initial resective surgery and not considered
amenable to further resective surgery at the time of study entry. The primary evidence of efficacy was
based on objective response rates. Out of the 12 patients enrolled, 9 responded, one completely and 8
partially. Three of the partial responders were subsequently rendered disease free by surgery. The
median duration of therapy in study B2225 was 6.2 months, with a maximum duration of 24.3 months.
A further 6 DFSP patients treated with Glivec were reported in 5 published case reports, their ages
ranging from 18 months to 49 years. The adult patients reported in the published literature were
treated with either 400 mg (4 cases) or 800 mg (1 case) Glivec daily. The paediatric patient received
400 mg/m 2 /daily, subsequently increased to 520 mg/m 2 /daily. 5 patients responded, 3 completely and
2 partially. The median duration of therapy in the published literature ranged between 4 weeks and
more than 20 months. The translocation t(17:22)[(q22:q13)], or its gene product, was present in nearly
all responders to Glivec treatment.
5.2 Pharmacokinetic properties
Pharmacokinetics of Glivec
The pharmacokinetics of Glivec have been evaluated over a dosage range of 25 to 1,000 mg. Plasma
pharmacokinetic profiles were analysed on day 1 and on either day 7 or day 28, by which time plasma
concentrations had reached steady state.
Absorption
Mean absolute bioavailability for the capsule formulation is 98%. There was high between-patient
variability in plasma imatinib AUC levels after an oral dose. When given with a high-fat meal, the rate
of absorption of imatinib was minimally reduced (11% decrease in C max and prolongation of t max by
1.5 h), with a small reduction in AUC (7.4%) compared to fasting conditions. The effect of prior
gastrointestinal surgery on drug absorption has not been investigated.
Distribution
At clinically relevant concentrations of imatinib, binding to plasma proteins was approximately 95%
on the basis of in vitro experiments, mostly to albumin and alpha-acid-glycoprotein, with little binding
to lipoprotein.
24
 
Metabolism
The main circulating metabolite in humans is the N-demethylated piperazine derivative, which shows
similar in vitro potency to the parent. The plasma AUC for this metabolite was found to be only 16%
of the AUC for imatinib. The plasma protein binding of the N-demethylated metabolite is similar to
that of the parent compound.
Imatinib and the N-demethyl metabolite together accounted for about 65% of the circulating
radioactivity (AUC (0-48h) ). The remaining circulating radioactivity consisted of a number of minor
metabolites.
The in vitro results showed that CYP3A4 was the major human P450 enzyme catalysing the
biotransformation of imatinib. Of a panel of potential comedications (acetaminophen, aciclovir,
allopurinol, amphotericin, cytarabine, erythromycin, fluconazole, hydroxyurea, norfloxacin, penicillin
V) only erythromycin (IC 50 50 µM) and fluconazole (IC 50 118 µM) showed inhibition of imatinib
metabolism which could have clinical relevance.
Imatinib was shown in vitro to be a competitive inhibitor of marker substrates for CYP2C9, CYP2D6
and CYP3A4/5. K i values in human liver microsomes were 27, 7.5 and 7.9 mol/l, respectively.
Maximal plasma concentrations of imatinib in patients are 2–4 mol/l, consequently an inhibition of
CYP2D6 and/or CYP3A4/5-mediated metabolism of co-administered drugs is possible. Imatinib did
not interfere with the biotransformation of 5-fluorouracil, but it inhibited paclitaxel metabolism as a
result of competitive inhibition of CYP2C8 (K i = 34.7 µM). This K i value is far higher than the
expected plasma levels of imatinib in patients, consequently no interaction is expected upon co-
administration of either 5-fluorouracil or paclitaxel and imatinib.
Elimination
Based on the recovery of compound(s) after an oral 14 C-labelled dose of imatinib, approximately 81%
of the dose was recovered within 7 days in faeces (68% of dose) and urine (13% of dose). Unchanged
imatinib accounted for 25% of the dose (5% urine, 20% faeces), the remainder being metabolites.
Plasma pharmacokinetics
Following oral administration in healthy volunteers, the t ½ was approximately 18 h, suggesting that
once-daily dosing is appropriate. The increase in mean AUC with increasing dose was linear and dose
proportional in the range of 25–1,000 mg imatinib after oral administration. There was no change in
the kinetics of imatinib on repeated dosing, and accumulation was 1.5–2.5-fold at steady state when
dosed once daily.
Pharmacokinetics in GIST patients
In patients with GIST steady-state exposure was 1.5-fold higher than that observed for CML patients
for the same dosage (400 mg daily). Based on preliminary population pharmacokinetic analysis in
GIST patients, there were three variables (albumin, WBC and bilirubin) found to have a statistically
significant relationship with imatinib pharmacokinetics. Decreased values of albumin caused a
reduced clearance (CL/f); and higher levels of WBC led to a reduction of CL/f. However, these
associations are not sufficiently pronounced to warrant dose adjustment. In this patient population, the
presence of hepatic metastases could potentially lead to hepatic insufficiency and reduced metabolism.
Population pharmacokinetics
Based on population pharmacokinetic analysis in CML patients, there was a small effect of age on the
volume of distribution (12% increase in patients > 65 years old). This change is not thought to be
clinically significant. The effect of bodyweight on the clearance of imatinib is such that for a patient
weighing 50 kg the mean clearance is expected to be 8.5 l/h, while for a patient weighing 100 kg the
clearance will rise to 11.8 l/h. These changes are not considered sufficient to warrant dose adjustment
based on kg bodyweight. There is no effect of gender on the kinetics of imatinib.
25
Pharmacokinetics in children
As in adult patients, imatinib was rapidly absorbed after oral administration in paediatric patients in
both phase I and phase II studies. Dosing in children at 260 and 340 mg/m 2 /day achieved the same
exposure, respectively, as doses of 400 mg and 600 mg in adult patients. The comparison of AUC (0-24)
on day 8 and day 1 at the 340 mg/m 2 /day dose level revealed a 1.7-fold drug accumulation after
repeated once-daily dosing.
Organ function impairment
Imatinib and its metabolites are not excreted via the kidney to a significant extent. Patients with mild
and moderate impairment of renal function appear to have a higher plasma exposure than patients with
normal renal function. The increase is approximately 1.5- to 2-fold, corresponding to a 1.5-fold
elevation of plasma AGP, to which imatinib binds strongly. The free drug clearance of imatinib is
probably similar between patients with renal impairment and those with normal renal function, since
renal excretion represents only a minor elimination pathway for imatinib (see sections 4.2 and 4.4).
Although the results of pharmacokinetic analysis showed that there is considerable inter-subject
variation, the mean exposure to imatinib did not increase in patients with varying degrees of liver
dysfunction as compared to patients with normal liver function (see sections 4.2, 4.4 and 4.8).
5.3 Preclinical safety data
The preclinical safety profile of imatinib was assessed in rats, dogs, monkeys and rabbits.
Multiple dose toxicity studies revealed mild to moderate haematological changes in rats, dogs and
monkeys, accompanied by bone marrow changes in rats and dogs.
The liver was a target organ in rats and dogs. Mild to moderate increases in transaminases and slight
decreases in cholesterol, triglycerides, total protein and albumin levels were observed in both species.
No histopathological changes were seen in rat liver. Severe liver toxicity was observed in dogs treated
for 2 weeks, with elevated liver enzymes, hepatocellular necrosis, bile duct necrosis, and bile duct
hyperplasia.
Renal toxicity was observed in monkeys treated for 2 weeks, with focal mineralisation and dilation of
the renal tubules and tubular nephrosis. Increased blood urea nitrogen (BUN) and creatinine were
observed in several of these animals. In rats, hyperplasia of the transitional epithelium in the renal
papilla and in the urinary bladder was observed at doses  6 mg/kg in the 13-week study, without
changes in serum or urinary parameters. An increased rate of opportunistic infections was observed
with chronic imatinib treatment.
In a 39-week monkey study, no NOAEL (no observed adverse effect level) was established at the
lowest dose of 15 mg/kg, approximately one-third the maximum human dose of 800 mg based on
body surface. Treatment resulted in worsening of normally suppressed malarial infections in these
animals.
Imatinib was not considered genotoxic when tested in an in vitro bacterial cell assay (Ames test), an in
vitro mammalian cell assay (mouse lymphoma) and an in vivo rat micronucleus test. Positive
genotoxic effects were obtained for imatinib in an in vitro mammalian cell assay (Chinese hamster
ovary) for clastogenicity (chromosome aberration) in the presence of metabolic activation. Two
intermediates of the manufacturing process, which are also present in the final product, are positive for
mutagenesis in the Ames assay. One of these intermediates was also positive in the mouse lymphoma
assay.
In a study of fertility, in male rats dosed for 70 days prior to mating, testicular and epididymal weights
and percent motile sperm were decreased at 60 mg/kg, approximately equal to the maximum clinical
dose of 800 mg/day, based on body surface area. This was not seen at doses ≤ 20 mg/kg. A slight to
moderate reduction in spermatogenesis was also observed in the dog at oral doses  30 mg/kg. When
26
female rats were dosed 14 days prior to mating and through to gestational day 6, there was no effect on
mating or on number of pregnant females. At a dose of 60 mg/kg, female rats had significant post-
implantation foetal loss and a reduced number of live foetuses. This was not seen at doses ≤ 20 mg/kg.
In an oral pre- and postnatal development study in rats, red vaginal discharge was noted in the
45 mg/kg/day group on either day 14 or day 15 of gestation. At the same dose, the number of stillborn
pups as well as those dying between postpartum days 0 and 4 was increased. In the F 1 offspring, at the
same dose level, mean body weights were reduced from birth until terminal sacrifice and the number
of litters achieving criterion for preputial separation was slightly decreased. F 1 fertility was not
affected, while an increased number of resorptions and a decreased number of viable foetuses was
noted at 45 mg/kg/day. The no observed effect level (NOEL) for both the maternal animals and the F 1
generation was 15 mg/kg/day (one quarter of the maximum human dose of 800 mg).
Imatinib was teratogenic in rats when administered during organogenesis at doses ≥ 100 mg/kg,
approximately equal to the maximum clinical dose of 800 mg/day, based on body surface area.
Teratogenic effects included exencephaly or encephalocele, absent/reduced frontal and absent parietal
bones. These effects were not seen at doses ≤ 30 mg/kg.
In the 2-year rat carcinogenicity study administration of imatinib at 15, 30 and 60 mg/kg/day resulted
in a statistically significant reduction in the longevity of males at 60 mg/kg/day and females at
≥30 mg/kg/day. Histopathological examination of decedents revealed cardiomyopathy (both sexes),
chronic progressive nephropathy (females) and preputial gland papilloma as principal causes of death
or reasons for sacrifice. Target organs for neoplastic changes were the kidneys, urinary bladder,
urethra, preputial and clitoral gland, small intestine, parathyroid glands, adrenal glands and non-
glandular stomach.
Papilloma/carcinoma of the preputial/clitoral gland were noted from 30 mg/kg/day onwards,
representing approximately 0.5 or 0.3 times the human daily exposure (based on AUC) at 400 mg/day
or 800 mg/day, respectively, and 0.4 times the daily exposure in children (based on AUC) at
340 mg/m 2 /day. The no observed effect level (NOEL) was 15 mg/kg/day. The renal
adenoma/carcinoma, the urinary bladder and urethra papilloma, the small intestine adenocarcinomas,
the parathyroid glands adenomas, the benign and malignant medullary tumours of the adrenal glands
and the non-glandular stomach papillomas/carcinomas were noted at 60 mg/kg/day, representing
approximately 1.7 or 1 times the human daily exposure (based on AUC) at 400 mg/day or 800 mg/day,
respectively, and 1.2 times the daily exposure in children (based on AUC) at 340 mg/m 2 /day. The no
observed effect level (NOEL) was 30 mg/kg/day.
The mechanism and relevance of these findings in the rat carcinogenicity study for humans are not yet
clarified.
Non-neoplastic lesions not identified in earlier preclinical studies were the cardiovascular system,
pancreas, endocrine organs and teeth. The most important changes included cardiac hypertrophy and
dilatation, leading to signs of cardiac insufficiency in some animals.
27
6.
PHARMACEUTICAL PARTICULARS
6.1 List of excipients
Capsule content: Cellulose microcrystalline
Crospovidone
Magnesium stearate
Silica colloidal, anhydrous
Capsule shell:
Gelatin
Iron oxide, yellow (E172)
Titanium dioxide (E171)
Printing ink:
Iron oxide, red (E172)
Shellac
Soya lecithin
6.2 Incompatibilities
Not applicable.
6.3 Shelf life
2 years
6.4 Special precautions for storage
Do not store above 30C.
Store in the original package in order to protect from moisture.
6.5 Nature and contents of container
PVC/aluminium blisters
Packs containing 30 capsules.
6.6 Special precautions for disposal
No special requirements.
7.
MARKETING AUTHORISATION HOLDER
Novartis Europharm Limited
Wimblehurst Road
Horsham
West Sussex, RH12 5AB
United Kingdom
8.
MARKETING AUTHORISATION NUMBER(S)
EU/1/01/198/001
28
9.
DATE OF FIRST AUTHORISATION/RENEWAL OF THE AUTHORISATION
Date of first authorisation: 07.11.2001
Date of first renewal: 07.11.2006
10. DATE OF REVISION OF THE TEXT
Detailed information on this product is available on the website of the European Medicines Agency
http://www.ema.europa.eu
29
1.
NAME OF THE MEDICINAL PRODUCT
Glivec 100 mg hard capsules
2.
QUALITATIVE AND QUANTITATIVE COMPOSITION
Each capsule contains 100 mg imatinib (as mesilate).
For a full list of excipients, see section 6.1.
3.
PHARMACEUTICAL FORM
Hard capsule
White to yellow powder in an orange to greyish-orange opaque capsule, marked “NVR SI”.
4.
CLINICAL PARTICULARS
4.1 Therapeutic indications
Glivec is indicated for the treatment of
adult and paediatric patients with newly diagnosed Philadelphia chromosome (bcr-abl) positive
(Ph+) chronic myeloid leukaemia (CML) for whom bone marrow transplantation is not
considered as the first line of treatment.
adult and paediatric patients with Ph+ CML in chronic phase after failure of interferon-alpha
therapy, or in accelerated phase or blast crisis.
adult patients with newly diagnosed Philadelphia chromosome positive acute lymphoblastic
leukaemia (Ph+ ALL) integrated with chemotherapy.
adult patients with relapsed or refractory Ph+ ALL as monotherapy.
adult patients with myelodysplastic/myeloproliferative diseases (MDS/MPD) associated with
platelet-derived growth factor receptor (PDGFR) gene re-arrangements.
adult patients with advanced hypereosinophilic syndrome (HES) and/or chronic eosinophilic
leukaemia (CEL) with FIP1L1-PDGFR rearrangement.
The effect of Glivec on the outcome of bone marrow transplantation has not been determined.
Glivec is indicated for
the treatment of adult patients with Kit (CD 117) positive unresectable and/or metastatic
malignant gastrointestinal stromal tumours (GIST).
the adjuvant treatment of adult patients who are at significant risk of relapse following resection
of Kit (CD117)-positive GIST. Patients who have a low or very low risk of recurrence should
not receive adjuvant treatment.
the treatment of adult patients with unresectable dermatofibrosarcoma protuberans (DFSP) and
adult patients with recurrent and/or metastatic DFSP who are not eligible for surgery.
In adult and paediatric patients, the effectiveness of Glivec is based on overall haematological and
cytogenetic response rates and progression-free survival in CML, on haematological and cytogenetic
response rates in Ph+ ALL, MDS/MPD, on haematological response rates in HES/CEL and on
objective response rates in adult patients with unresectable and/or metastatic GIST and DFSP and on
recurrence-free survival in adjuvant GIST. The experience with Glivec in patients with MDS/MPD
associated with PDGFR gene re-arrangements is very limited (see section 5.1). Except in newly
diagnosed chronic phase CML, there are no controlled trials demonstrating a clinical benefit or
increased survival for these diseases.
30
4.2 Posology and method of administration
Therapy should be initiated by a physician experienced in the treatment of patients with
haematological malignancies and malignant sarcomas, as appropriate.
The prescribed dose should be administered orally with a meal and a large glass of water to minimise
the risk of gastrointestinal irritations. Doses of 400 mg or 600 mg should be administered once daily,
whereas a daily dose of 800 mg should be administered as 400 mg twice a day, in the morning and in
the evening. For patients (children) unable to swallow the capsules, their content may be diluted in a
glass of either still water or apple juice. Since studies in animals have shown reproductive toxicity, and
the potential risk for the human foetus is unknown, women of child-bearing potential who open
capsules should be advised to handle the contents with caution and avoid skin-eye contact or
inhalation (see section 4.6). Hands should be washed immediately after handling open capsules.
Posology for CML in adult patients
The recommended dosage of Glivec is 400 mg/day for patients in chronic phase CML. Chronic phase
CML is defined when all of the following criteria are met: blasts < 15% in blood and bone marrow,
peripheral blood basophils < 20%, platelets > 100 x 10 9 /l.
The recommended dosage of Glivec is 600 mg/day for patients in accelerated phase. Accelerated
phase is defined by the presence of any of the following: blasts  15% but < 30% in blood or bone
marrow, blasts plus promyelocytes  30% in blood or bone marrow (providing < 30% blasts),
peripheral blood basophils  20%, platelets < 100 x 10 9 /l unrelated to therapy.
The recommended dose of Glivec is 600 mg/day for patients in blast crisis. Blast crisis is defined as
blasts  30% in blood or bone marrow or extramedullary disease other than hepatosplenomegaly.
Treatment duration: In clinical trials, treatment with Glivec was continued until disease progression.
The effect of stopping treatment after the achievement of a complete cytogenetic response has not
been investigated.
Dose increases from 400 mg to 600 mg or 800 mg in patients with chronic phase disease, or from
600 mg to a maximum of 800 mg (given as 400 mg twice daily) in patients with accelerated phase or
blast crisis may be considered in the absence of severe adverse drug reaction and severe non-
leukaemia-related neutropenia or thrombocytopenia in the following circumstances: disease
progression (at any time); failure to achieve a satisfactory haematological response after at least
3 months of treatment; failure to achieve a cytogenetic response after 12 months of treatment; or loss
of a previously achieved haematological and/or cytogenetic response. Patients should be monitored
closely following dose escalation given the potential for an increased incidence of adverse reactions at
higher dosages.
Posology for CML in children
Dosing for children should be on the basis of body surface area (mg/m 2 ). The dose of 340 mg/m 2 daily
is recommended for children with chronic phase CML and advanced phase CML (not to exceed the
total dose of 800 mg). Treatment can be given as a once daily dose or alternatively the daily dose may
be split into two administrations – one in the morning and one in the evening. The dose
recommendation is currently based on a small number of paediatric patients (see sections 5.1 and 5.2).
There is no experience with the treatment of children below 2 years of age.
Dose increases from 340 mg/m 2 daily to 570 mg/m 2 daily (not to exceed the total dose of 800 mg) may
be considered in children in the absence of severe adverse drug reaction and severe non-leukaemia-
related neutropenia or thrombocytopenia in the following circumstances: disease progression (at any
time); failure to achieve a satisfactory haematological response after at least 3 months of treatment;
failure to achieve a cytogenetic response after 12 months of treatment; or loss of a previously achieved
haematological and/or cytogenetic response. Patients should be monitored closely following dose
escalation given the potential for an increased incidence of adverse reactions at higher dosages.
31
Posology for Ph+ ALL
The recommended dose of Glivec is 600 mg/day for patients with Ph+ ALL. Haematological experts
in the management of this disease should supervise the therapy throughout all phases of care.
Treatment schedule: On the basis of the existing data, Glivec has been shown to be effective and safe
when administered at 600 mg/day in combination with chemotherapy in the induction phase, the
consolidation and maintenance phases of chemotherapy (see section 5.1) for adult patients with newly
diagnosed Ph+ ALL. The duration of Glivec therapy can vary with the treatment programme selected,
but generally longer exposures to Glivec have yielded better results.
For adult patients with relapsed or refractory Ph+ALL Glivec monotherapy at 600 mg/day is safe,
effective and can be given until disease progression occurs.
Posology for MDS/MPD
The recommended dose of Glivec is 400 mg/day for patients with MDS/MPD.
Treatment duration: In the only clinical trial performed up to now, treatment with Glivec was
continued until disease progression (see section 5.1). At the time of analysis, the treatment duration
was a median of 47 months (24 days - 60 months).
Posology for HES/CEL
The recommended dose of Glivec is 100 mg/day for patients with HES/CEL.
Dose increase from 100 mg to 400 mg may be considered in the absence of adverse drug reactions if
assessments demonstrate an insufficient response to therapy.
Treatment should be continued as long as the patient continues to benefit.
Posology for GIST
The recommended dose of Glivec is 400 mg/day for patients with unresectable and/or metastatic
malignant GIST.
Limited data exist on the effect of dose increases from 400 mg to 600 mg or 800 mg in patients
progressing at the lower dose (see section 5.1).
Treatment duration: In clinical trials in GIST patients, treatment with Glivec was continued until
disease progression. At the time of analysis, the treatment duration was a median of 7 months (7 days
to 13 months). The effect of stopping treatment after achieving a response has not been investigated.
The recommended dose of Glivec is 400 mg/day for the adjuvant treatment of adult patients following
resection of GIST. Optimal treatment duration is not yet established. Length of treatment in the
clinical trial supporting this indication was 12 months.
Posology for DFSP
The recommended dose of Glivec is 800 mg/day for patients with DFSP.
Dose adjustment for adverse reactions
Non-haematological adverse reactions
If a severe non-haematological adverse reaction develops with Glivec use, treatment must be withheld
until the event has resolved. Thereafter, treatment can be resumed as appropriate depending on the
initial severity of the event.
If elevations in bilirubin > 3 x institutional upper limit of normal (IULN) or in liver transaminases
> 5 x IULN occur, Glivec should be withheld until bilirubin levels have returned to < 1.5 x IULN and
transaminase levels to < 2.5 x IULN. Treatment with Glivec may then be continued at a reduced daily
dose. In adults the dose should be reduced from 400 to 300 mg or from 600 to 400 mg, or from
800 mg to 600 mg, and in children from 340 to 260 mg/m 2 /day.
32
Haematological adverse reactions
Dose reduction or treatment interruption for severe neutropenia and thrombocytopenia are
recommended as indicated in the table below.
Dose adjustments for neutropenia and thrombocytopenia:
HES/CEL (starting dose
100 mg)
ANC < 1.0 x 10 9 /l
and/or
platelets < 50 x 10 9 /l
1. Stop Glivec until ANC  1.5 x 10 9 /l and
platelets  75 x 10 9 /l.
2. Resume treatment with Glivec at previous
dose (i.e. before severe adverse reaction).
Chronic phase CML,
MDS/MPD and GIST
(starting dose 400 mg)
HES/CEL
(at dose 400 mg)
ANC < 1.0 x 10 9 /l
and/or
platelets < 50 x 10 9 /l
1. Stop Glivec until ANC  1.5 x 10 9 /l and
platelets  75 x 10 9 /l.
2. Resume treatment with Glivec at previous
dose (i.e. before severe adverse reaction).
3. In the event of recurrence of ANC
< 1.0 x 10 9 /l and/or platelets < 50 x 10 9 /l,
repeat step 1 and resume Glivec at
reduced dose of 300 mg.
Paediatric chronic phase
CML
(at dose 340 mg/m 2 )
ANC < 1.0 x 10 9 /l
and/or
platelets < 50 x 10 9 /l
1. Stop Glivec until ANC  1.5 x 10 9 /l and
platelets  75 x 10 9 /l.
2. Resume treatment with Glivec at previous
dose (i.e. before severe adverse reaction).
3. In the event of recurrence of ANC
< 1.0 x10 9 /l and/or platelets < 50 x10 9 /l,
repeat step 1 and resume Glivec at
reduced dose of 260 mg/m 2 .
Accelerated phase CML
and blast crisis and Ph+
ALL (starting dose
600 mg)
a ANC < 0.5 x 10 9 /l
and/or
platelets < 10 x 10 9 /l
1. Check whether cytopenia is related to
leukaemia (marrow aspirate or biopsy).
2. If cytopenia is unrelated to leukaemia,
reduce dose of Glivec to 400 mg.
3. If cytopenia persists for 2 weeks, reduce
further to 300 mg.
4. If cytopenia persists for 4 weeks and is
still unrelated to leukaemia, stop Glivec
until ANC  1 x 10 9 /l and platelets
 20 x 10 9 /l, then resume treatment at
300 mg.
Paediatric accelerated
phase CML and blast
crisis (starting dose
340 mg/m 2 )
a ANC < 0.5 x 10 9 /l
and/or
platelets < 10 x 10 9 /l
1. Check whether cytopenia is related to
leukaemia (marrow aspirate or biopsy).
2. If cytopenia is unrelated to leukaemia,
reduce dose of Glivec to 260 mg/m 2 .
3. If cytopenia persists for 2 weeks, reduce
further to 200 mg/m 2 .
4. If cytopenia persists for 4 weeks and is
still unrelated to leukaemia, stop Glivec
until ANC  1 x 10 9 /l and platelets
 20 x 10 9 /l, then resume treatment at
200 mg/m 2 .
33
DFSP
(at dose 800 mg)
ANC < 1.0 x 10 9 /l
and/or
platelets < 50 x 10 9 /l
1. Stop Glivec until ANC  1.5 x 10 9 /l and
platelets  75 x 10 9 /l.
2. Resume treatment with Glivec at 600 mg.
3. In the event of recurrence of ANC
< 1.0 x 10 9 /l and/or platelets < 50 x 10 9 /l,
repeat step 1 and resume Glivec at
reduced dose of 400 mg.
ANC = absolute neutrophil count
a occurring after at least 1 month of treatment
Paediatric use: There is no experience in children with CML below 2 years of age (see section 5.1).
There is limited experience in children with Ph+ ALL and very limited experience in children with
MDS/MPD and DFSP. There is no experience in children or adolescents with GIST and HES/CEL.
Hepatic insufficiency: Imatinib is mainly metabolised through the liver. Patients with mild, moderate
or severe liver dysfunction should be given the minimum recommended dose of 400 mg daily. The
dose can be reduced if not tolerated (see sections 4.4, 4.8 and 5.2).
Liver dysfunction classification:
Liver dysfunction Liver function tests
Mild Total bilirubin: = 1.5 ULN
AST: >ULN (can be normal or <ULN if total bilirubin is
>ULN)
Moderate Total bilirubin: >1.5–3.0 ULN
AST: any
Severe Total bilirubin: >3–10 ULN
AST: any
ULN = upper limit of normal for the institution
AST = aspartate aminotransferase
Renal insufficiency: Since the renal clearance of imatinib is negligible, a decrease in free imatinib
clearance is not expected in patients with renal insufficiency. Patients with mild or moderate renal
dysfunction (creatinine clearance = 20–59 ml/min) should be given the minimum recommended dose
of 400 mg daily as starting dose. Although very limited information is available, patients with severe
renal dysfunction (creatinine clearance = < 20 ml/min) or on dialysis could also start at the same dose
of 400 mg. However, in these patients caution is recommended. The dose can be reduced if not
tolerated, or increased for lack of efficacy (see sections 4.4 and 5.2).
Elderly patients: Imatinib pharmacokinetics have not been specifically studied in the elderly. No
significant age-related pharmacokinetic differences have been observed in adult patients in clinical
trials which included over 20% of patients age 65 and older. No specific dose recommendation is
necessary in the elderly.
4.3 Contraindications
Hypersensitivity to the active substance or to any of the excipients.
4.4 Special warnings and precautions for use
When Glivec is co-administered with other medicinal products, there is a potential for drug
interactions (see section 4.5).
Concomitant use of imatinib and medicinal products that induce CYP3A4 (e.g. dexamethasone,
phenytoin, carbamazepine, rifampicin, phenobarbital or Hypericum perforatum , also known as St.
34
John’s Wort) may significantly reduce exposure to Glivec, potentially increasing the risk of
therapeutic failure. Therefore, concomitant use of strong CYP3A4 inducers and imatinib should be
avoided (see section 4.5).
Clinical cases of hypothyroidism have been reported in thyroidectomy patients undergoing
levothyroxine replacement during treatment with Glivec (see section 4.5). TSH levels should be
closely monitored in such patients.
Metabolism of Glivec is mainly hepatic, and only 13% of excretion is through the kidneys. In patients
with hepatic dysfunction (mild, moderate or severe), peripheral blood counts and liver enzymes should
be carefully monitored (see sections 4.2, 4.8 and 5.2). It should be noted that GIST patients may have
hepatic metastases which could lead to hepatic impairment.
Cases of liver injury, including hepatic failure and hepatic necrosis, have been observed with imatinib.
When imatinib is combined with high dose chemotherapy regimens, an increase in serious hepatic
reactions has been detected. Hepatic function should be carefully monitored in circumstances where
imatinib is combined with chemotherapy regimens also known to be associated with hepatic
dysfunction (see section 4.5 and 4.8).
Occurrences of severe fluid retention (pleural effusion, oedema, pulmonary oedema, ascites,
superficial oedema) have been reported in approximately 2.5% of newly diagnosed CML patients
taking Glivec. Therefore, it is highly recommended that patients be weighed regularly. An unexpected
rapid weight gain should be carefully investigated and if necessary appropriate supportive care and
therapeutic measures should be undertaken. In clinical trials, there was an increased incidence of these
events in elderly patients and those with a prior history of cardiac disease. Therefore, caution should
be exercised in patients with cardiac dysfunction.
Patients with cardiac disease or risk factors for cardiac failure should be monitored carefully, and any
patient with signs or symptoms consistent with cardiac failure should be evaluated and treated.
In patients with hypereosinophilic syndrome (HES) and cardiac involvement, isolated cases of
cardiogenic shock/left ventricular dysfunction have been associated with the initiation of imatinib
therapy. The condition was reported to be reversible with the administration of systemic steroids,
circulatory support measures and temporarily withholding imatinib. As cardiac adverse events have
been reported uncommonly with imatinib, a careful assessment of the benefit/risk of imatinib therapy
should be considered in the HES/CEL population before treatment initiation.
Myelodysplastic/myeloproliferative diseases with PDGFR gene re-arrangements could be associated
with high eosinophil levels. Evaluation by a cardiology specialist, performance of an echocardiogram
and determination of serum troponin should therefore be considered in patients with HES/CEL, and in
patients with MDS/MPD associated with high eosinophil levels before imatinib is administered. If
either is abnormal, follow-up with a cardiology specialist and the prophylactic use of systemic steroids
(1–2 mg/kg) for one to two weeks concomitantly with imatinib should be considered at the initiation
of therapy.
In the study in patients with unresectable and/or metastatic GIST, both gastrointestinal and intra-
tumoural haemorrhages were reported (see section 4.8). Based on the available data, no predisposing
factors (e.g. tumour size, tumour location, coagulation disorders) have been identified that place
patients with GIST at a higher risk of either type of haemorrhage. Since increased vascularity and
propensity for bleeding is a part of the nature and clinical course of GIST, standard practices and
procedures for the monitoring and management of haemorrhage in all patients should be applied.
Due to the possible occurrence of tumour lysis syndrome (TLS), correction of clinically significant
dehydration and treatment of high uric acid levels are recommended prior to initiation of Glivec (see
section 4.8).
35
Laboratory tests
Complete blood counts must be performed regularly during therapy with Glivec. Treatment of CML
patients with Glivec has been associated with neutropenia or thrombocytopenia. However, the
occurrence of these cytopenias is likely to be related to the stage of the disease being treated and they
were more frequent in patients with accelerated phase CML or blast crisis as compared to patients with
chronic phase CML. Treatment with Glivec may be interrupted or the dose may be reduced, as
recommended in section 4.2.
Liver function (transaminases, bilirubin, alkaline phosphatase) should be monitored regularly in
patients receiving Glivec.
In patients with impaired renal function, imatinib plasma exposure seems to be higher than that in
patients with normal renal function, probably due to an elevated plasma level of alpha-acid
glycoprotein (AGP), an imatinib-binding protein, in these patients. Patients with renal impairment
should be given the minimum starting dose. Patients with severe renal impairment should be treated
with caution. The dose can be reduced if not tolerated (see section 4.2 and 5.2).
Children and adolescents
There have been case reports of growth retardation occurring in children and pre-adolescents receiving
imatinib. The long-term effects of prolonged treatment with imatinib on growth in children are
unknown. Therefore, close monitoring of growth in children under imatinib treatment is recommended
(see section 4.8).
4.5 Interaction with other medicinal products and other forms of interaction
Active substances that may increase imatinib plasma concentrations:
Substances that inhibit the cytochrome P450 isoenzyme CYP3A4 activity (e.g. ketoconazole,
itraconazole, erythromycin, clarithromycin) could decrease metabolism and increase imatinib
concentrations. There was a significant increase in exposure to imatinib (the mean C max and AUC of
imatinib rose by 26% and 40%, respectively) in healthy subjects when it was co-administered with a
single dose of ketoconazole (a CYP3A4 inhibitor). Caution should be taken when administering
Glivec with inhibitors of the CYP3A4 family.
Active substances that may decrease imatinib plasma concentrations:
Substances that are inducers of CYP3A4 activity could increase metabolism and decrease imatinib
plasma concentrations. Co-medications which induce CYP3A4 (e.g. dexamethasone, phenytoin,
carbamazepine, rifampicin, phenobarbital, fosphenytoin, primidone or Hypericum perforatum , also
known as St. John’s Wort) may significantly reduce exposure to Glivec, potentially increasing the risk
of therapeutic failure. Pretreatment with multiple doses of rifampicin 600 mg followed by a single
400 mg dose of Glivec resulted in decrease in C max and AUC (0-∞) by at least 54% and 74%, of the
respective values without rifampicin treatment. Similar results were observed in patients with
malignant gliomas treated with Glivec while taking enzyme-inducing anti-epileptic drugs (EIAEDs)
such as carbamazepine, oxcarbazepine and phenytoin. The plasma AUC for imatinib decreased by
73% compared to patients not on EIAEDs. Concomitant use of rifampicin or other strong CYP3A4
inducers and imatinib should be avoided.
Active substances that may have their plasma concentration altered by Glivec
Imatinib increases the mean C max and AUC of simvastatin (CYP3A4 substrate) 2- and 3.5-fold,
respectively, indicating an inhibition of the CYP3A4 by imatinib. Therefore, caution is recommended
when administering Glivec with CYP3A4 substrates with a narrow therapeutic window (e.g.
cyclosporin or pimozide). Glivec may increase plasma concentration of other CYP3A4 metabolised
drugs (e.g. triazolo-benzodiazepines, dihydropyridine calcium channel blockers, certain HMG-CoA
reductase inhibitors, i.e. statins, etc.).
Because warfarin is metabolised by CYP2C9, patients who require anticoagulation should receive
low-molecular-weight or standard heparin.
36
In vitro Glivec inhibits the cytochrome P450 isoenzyme CYP2D6 activity at concentrations similar to
those that affect CYP3A4 activity. Imatinib at 400 mg twice daily had an inhibitory effect on
CYP2D6-mediated metoprolol metabolism, with metoprolol C max and AUC being increased by
approximately 23% (90%CI [1.16-1.30]). Dose adjustments do not seem to be necessary when
imatinib is co-administrated with CYP2D6 substrates, however caution is advised for CYP2D6
substrates with a narrow therapeutic window such as metoprolol. In patients treated with metoprolol
clinical monitoring should be considered.
In vitro , Glivec inhibits paracetamol O-glucuronidation (Ki value of 58.5 micromol/l at therapeutic
levels).
Caution should therefore be exercised when using Glivec and paracetamol concomitantly, especially
with high doses of paracetamol.
In thyroidectomy patients receiving levothyroxine, the plasma exposure to levothyroxine may be
decreased when Glivec is co-administered (see section 4.4). Caution is therefore recommended.
However, the mechanism of the observed interaction is presently unknown.
In Ph+ ALL patients, there is clinical experience of co-administering Glivec with chemotherapy (see
section 5.1), but drug-drug interactions between imatinib and chemotherapy regimens are not well
characterised. Imatinib adverse events, i.e. hepatotoxicity, myelosuppression or others, may increase
and it has been reported that concomitant use with L-asparaginase could be associated with increased
hepatotoxicity (see section 4.8). Therefore, the use of Glivec in combination requires special
precaution.
4.6 Fertility, pregnancy and lactation
Pregnancy
There are no adequate data on the use of imatinib in pregnant women. Studies in animals have
however shown reproductive toxicity (see section 5.3) and the potential risk for the foetus is unknown.
Glivec should not be used during pregnancy unless clearly necessary. If it is used during pregnancy,
the patient must be informed of the potential risk to the foetus. Women of childbearing potential must
be advised to use effective contraception during treatment.
Breast-feeding
There is limited information on imatinib distribution on human milk. Studies in two breast-feeding
women revealed that both imatinib and its active metabolite can be distributed into human milk. The
milk plasma ratio studied in a single patient was determined to be 0.5 for imatinib and 0.9 for the
metabolite, suggesting greater distribution of the metabolite into the milk. Considering the combined
concentration of imatinib and the metabolite and the maximum daily milk intake by infants, the total
exposure would be expected to be low (~10% of a therapeutic dose). However, since the effects of
low-dose exposure of the infant to imatinib are unknown, women taking imatinib should not breast-
feed.
4.7 Effects on ability to drive and use machines
No studies on the effects on the ability to drive and use machines have been performed. However,
patients should be advised that they may experience undesirable effects such as dizziness or blurred
vision during treatment with imatinib. Therefore, caution should be recommended when driving a car
or operating machinery.
37
4.8 Undesirable effects
Patients with advanced stages of malignancies may have numerous confounding medical conditions
that make causality of adverse reactions difficult to assess due to the variety of symptoms related to
the underlying disease, its progression, and the co-administration of numerous medicinal products.
In clinical trials in CML, drug discontinuation for drug-related adverse reactions was observed in 2.4%
of newly diagnosed patients, 4% of patients in late chronic phase after failure of interferon therapy,
4% of patients in accelerated phase after failure of interferon therapy and 5% of blast crisis patients
after failure of interferon therapy. In GIST the study drug was discontinued for drug-related adverse
reactions in 4% of patients.
The adverse reactions were similar in all indications, with two exceptions. There was more
myelosuppression seen in CML patients than in GIST, which is probably due to the underlying
disease. In the study in patients with unresectable and/or metastatic GIST, 7 (5%) patients experienced
CTC grade 3/4 GI bleeds (3 patients), intra-tumoural bleeds (3 patients) or both (1 patient). GI tumour
sites may have been the source of the GI bleeds (see section 4.4). GI and tumoural bleeding may be
serious and sometimes fatal. The most commonly reported ( 10%) drug-related adverse reactions in
both settings were mild nausea, vomiting, diarrhoea, abdominal pain, fatigue, myalgia, muscle cramps
and rash. Superficial oedemas were a common finding in all studies and were described primarily as
periorbital or lower limb oedemas. However, these oedemas were rarely severe and may be managed
with diuretics, other supportive measures, or by reducing the dose of Glivec.
When imatinib was combined with high dose chemotherapy in Ph+ ALL patients, transient liver
toxicity in the form of transaminase elevation and hyperbilirubinaemia were observed.
Miscellaneous adverse reactions such as pleural effusion, ascites, pulmonary oedema and rapid weight
gain with or without superficial oedema may be collectively described as “fluid retention”. These
reactions can usually be managed by withholding Glivec temporarily and with diuretics and other
appropriate supportive care measures. However, some of these reactions may be serious or life-
threatening and several patients with blast crisis died with a complex clinical history of pleural
effusion, congestive heart failure and renal failure. There were no special safety findings in paediatric
clinical trials.
Adverse reactions
Adverse reactions reported as more than an isolated case are listed below, by system organ class and
by frequency. Frequency categories are defined using the following convention: very common
(≥1/10), common (≥1/100 to <1/10), uncommon (≥1/1,000 to <1/100), rare (≥1/10,000 to <1/1,000),
very rare (<1/10,000), not known (cannot be estimated from the available data).
Within each frequency grouping, undesirable effects are presented in order of frequency, the most
frequent first.
38
Adverse reactions and their frequencies reported in Table 1 are based on the main registration studies.
Table 1
Adverse reactions in clinical studies
Infections and infestati ons
Uncommon: Herpes zoster, herpes simplex, nasopharyngitis, pneumonia 1 , sinusitis, cellulitis,
upper respiratory tract infection, influenza, urinary tract infection, gastroenteritis,
sepsis
Rare: Fungal infection
Neoplasm benign, mali gnant and unspecified (including cysts and polyps)
Rare: Tumour lysis syndrome
Blood and lymphatic sy stem disorders
Very common: Neutropenia, thrombocytopenia, anaemia
Common: Pancytopenia, febrile neutropenia
Uncommon: Thrombocythaemia, lymphopenia, bone marrow depression, eosinophilia,
lymphadenopathy
Rare: Haemolytic anaemia
Metabolism and nutrit ion disorders
Common:
Anorexia
Uncommon:
Hypokalaemia, increased appetite, hypophosphataemia, decreased appetite,
dehydration, gout, hyperuricaemia, hypercalcaemia, hyperglycaemia,
hyponatraemia
Rare:
Hyperkalaemia, hypomagnesaemia
Psychiatric disorders
Common: Insomnia
Uncommon: Depression, libido decreased, anxiety
Rare: Confusional state
Nervous system disord ers
Very common:
Headache 2
Common:
Dizziness, paraesthesia, taste disturbance, hypoaesthesia
Uncommon:
Migraine, somnolence, syncope, peripheral neuropathy, memory impairment,
sciatica, restless leg syndrome, tremor, cerebral haemorrhage
Rare:
Increased intracranial pressure, convulsions, optic neuritis
Eye disorders
Common: Eyelid oedema, lacrimation increased, conjunctival haemorrhage, conjunctivitis, dry
eye, blurred vision
Uncommon: Eye irritation, eye pain, orbital oedema, scleral haemorrhage, retinal haemorrhage,
blepharitis, macular oedema
Rare: Cataract, glaucoma, papilloedema
Ear and labyrinth diso rders
Uncommon:
Vertigo, tinnitus, hearing loss
Cardiac disorders
Uncommon:
Palpitations, tachycardia, cardiac failure congestive 3 , pulmonary oedema
Rare:
Arrhythmia, atrial fibrillation, cardiac arrest, myocardial infarction, angina pectoris,
pericardial effusion
Vascular disorders 4
Common: Flushing, haemorrhage
Uncommon: Hypertension, haematoma, peripheral coldness, hypotension, Raynaud’s
phenomenon
Respiratory, thoracic a nd mediastinal disorders
Common:
Dyspnoea, epistaxis, cough
Uncommon:
Pleural effusion 5 , pharyngolaryngeal pain, pharyngitis
Rare:
Pleuritic pain, pulmonary fibrosis, pulmonary hypertension, pulmonary
haemorrhage
39
 
Gastrointestinal disord ers
Very common: Nausea, diarrhoea, vomiting, dyspepsia, abdominal pain 6
Common: Flatulence, abdominal distension, gastro-oesophageal reflux, constipation, dry
mouth, gastritis
Uncommon: Stomatitis, mouth ulceration, gastrointestinal haemorrhage 7 , eructation, melaena,
oesophagitis, ascites, gastric ulcer, haematemesis, cheilitis, dysphagia, pancreatitis
Rare: Colitis, ileus, inflammatory bowel disease
Hepatobiliary disorder s
Common: Increased hepatic enzymes
Uncommon: Hyperbilirubinaemia, hepatitis, jaundice
Rare: Hepatic failure 8 , hepatic necrosis
Skin and subcutaneous tissue disorders
Very common: Periorbital oedema, dermatitis/eczema/rash
Common: Pruritus, face oedema, dry skin, erythema, alopecia, night sweats, photosensitivity
reaction
Uncommon: Rash pustular, contusion, sweating increased, urticaria, ecchymosis, increased
tendency to bruise, hypotrichosis, skin hypopigmentation, dermatitis exfoliative,
onychoclasis, folliculitis, petechiae, psoriasis, purpura, skin hyperpigmentation,
bullous eruptions
Rare: Acute febrile neutrophilic dermatosis (Sweet’s syndrome), nail discolouration,
angioneurotic oedema, rash vesicular, erythema multiforme, leucocytoclastic
vasculitis, Stevens-Johnson syndrome, acute generalised exanthematous pustulosis
(AGEP)
Musculoskeletal and co nnective tissue disorders
Very common: Muscle spasm and cramps, musculoskeletal pain including myalgia, arthralgia, bone
pain 9
Common: Joint swelling
Uncommon: Joint and muscle stiffness
Rare: Muscular weakness, arthritis, rhabdomyolysis/myopathy
Renal and urinary diso rders
Uncommon: Renal pain, haematuria, renal failure acute, urinary frequency increased
Reproductive system a nd breast disorders
Uncommon: Gynaecomastia, erectile dysfunction, menorrhagia, menstruation irregular, sexual
dysfunction, nipple pain, breast enlargement, scrotal oedema
Rare: Haemorrhagic corpus luteum/haemorrhagic ovarian cyst
General disorders and administration site conditions
Very common:
Fluid retention and oedema, fatigue
Common:
Weakness, pyrexia, anasarca, chills, rigors
Uncommon:
Chest pain, malaise
40
 
Investigations
Very common:
Weight increased
Common :
Weight decreased
Uncommon :
Blood creatinine increased, blood creatine phosphokinase increased, blood lactate
dehydrogenase increased, blood alkaline phosphatase increased
Rare:
Blood amylase increased
1 Pneumonia was reported most commonly in patients with transformed CML and in patients with
GIST.
2 Headache was the most common in GIST patients.
3 On a patient-year basis, cardiac events including congestive heart failure were more commonly
observed in patients with transformed CML than in patients with chronic CML.
4 Flushing was most common in GIST patients and bleeding (haematoma, haemorrhage) was
most common in patients with GIST and with transformed CML (CML-AP and CML-BC).
5 Pleural effusion was reported more commonly in patients with GIST and in patients with
transformed CML (CML-AP and CML-BC) than in patients with chronic CML.
6+7 Abdominal pain and gastrointestinal haemorrhage were most commonly observed in GIST
patients.
8
Musculoskeletal pain and related events were more commonly observed in patients with CML
than in GIST patients.
41
9
Some fatal cases of hepatic failure and of hepatic necrosis have been reported.
 
The following types of reactions have been reported mainly from post-marketing experience with
Glivec. This includes spontaneous case reports as well as serious adverse events from ongoing studies,
the expanded access programmes, clinical pharmacology studies and exploratory studies in
unapproved indications. Because these reactions are reported from a population of uncertain size, it is
not always possible to reliably estimate their frequency or establish a causal relationship to imatinib
exposure.
Table 2
Adverse reactions from post-marketing reports
Neoplasm benign, mali gnant and unspecified (including cysts and polyps)
Not known: Tumour haemorrhage/tumour necrosis
Immune system disord ers
Not known: Anaphylactic shock
Nervous system disord ers
Not known:
Cerebral oedema
Eye disorders
Not known:
Vitreous haemorrhage
Cardiac disorders
Not known:
Pericarditis, cardiac tamponade
Vascular disorders
Not known: Thrombosis/embolism
Respiratory, thoracic a nd mediastinal disorders
Not known: Acute respiratory failure 1 , interstitial lung disease
Gastrointestinal disord ers
Not known: Ileus/intestinal obstruction, gastrointestinal perforation, diverticulitis
Skin and subcutaneous tissue disorders
Not known: Palmoplantar erythrodysesthesia syndrome
Not known: Lichenoid keratosis, lichen planus
Not known: Toxic epidermal necrolysis
Musculoskeletal and co nnective tissue disorders
Not known:
Avascular necrosis/hip necrosis
Not known:
Growth retardation in children
1
Fatal cases have been reported in patients with advanced disease, severe infections, severe
neutropenia and other serious concomitant conditions.
Laboratory test abnormalities
Haematology
In CML, cytopenias, particularly neutropenia and thrombocytopenia, have been a consistent finding in
all studies, with the suggestion of a higher frequency at high doses  750 mg (phase I study).
However, the occurrence of cytopenias was also clearly dependent on the stage of the disease, the
frequency of grade 3 or 4 neutropenias (ANC < 1.0 x 10 9 /l) and thrombocytopenias (platelet count
< 50 x 10 9 /l) being between 4 and 6 times higher in blast crisis and accelerated phase (59–64% and
44–63% for neutropenia and thrombocytopenia, respectively) as compared to newly diagnosed
patients in chronic phase CML (16.7% neutropenia and 8.9% thrombocytopenia). In newly diagnosed
chronic phase CML grade 4 neutropenia (ANC < 0.5 x 10 9 /l) and thrombocytopenia (platelet count
< 10 x 10 9 /l) were observed in 3.6% and < 1% of patients, respectively. The median duration of the
neutropenic and thrombocytopenic episodes usually ranged from 2 to 3 weeks, and from 3 to 4 weeks,
respectively. These events can usually be managed with either a reduction of the dose or an
interruption of treatment with Glivec, but can in rare cases lead to permanent discontinuation of
treatment. In paediatric CML patients the most frequent toxicities observed were grade 3 or 4
cytopenias involving neutropenia, thrombocytopenia and anaemia. These generally occur within the
first several months of therapy.
42
 
In the study in patients with unresectable and/or metastatic GIST, grade 3 and 4 anaemia was reported
in 5.4% and 0.7% of patients, respectively, and may have been related to gastrointestinal or intra-
tumoural bleeding in at least some of these patients. Grade 3 and 4 neutropenia was seen in 7.5% and
2.7% of patients, respectively, and grade 3 thrombocytopenia in 0.7% of patients. No patient
developed grade 4 thrombocytopenia. The decreases in white blood cell (WBC) and neutrophil counts
occurred mainly during the first six weeks of therapy, with values remaining relatively stable
thereafter.
Biochemistry
Severe elevation of transaminases (<5%) or bilirubin (<1%) was seen in CML patients and was
usually managed with dose reduction or interruption (the median duration of these episodes was
approximately one week). Treatment was discontinued permanently because of liver laboratory
abnormalities in less than 1% of CML patients. In GIST patients (study B2222), 6.8% of grade 3 or 4
ALT (alanine aminotransferase) elevations and 4.8% of grade 3 or 4 AST (aspartate aminotransferase)
elevations were observed. Bilirubin elevation was below 3%.
There have been cases of cytolytic and cholestatic hepatitis and hepatic failure; in some of them
outcome was fatal, including one patient on high dose paracetamol.
4.9 Overdose
Experience with doses higher than the recommended therapeutic dose is limited. Isolated cases of
Glivec overdose have been reported spontaneously and in the literature. In the event of overdose the
patient should be observed and appropriate symptomatic treatment given. Generally the reported
outcome in these cases was “improved” or “recovered”. Events that have been reported at different
dose ranges are as follows:
Adult population
1200 to 1600 mg (duration varying between 1 to 10 days): Nausea, vomiting, diarrhoea, rash,
erythema, oedema, swelling, fatigue, muscle spasms, thrombocytopenia, pancytopenia, abdominal
pain, headache, decreased appetite.
1800 to 3200 mg (as high as 3200 mg daily for 6 days): Weakness, myalgia, increased creatine
phosphokinase, increased bilirubin, gastrointestinal pain.
6400 mg (single dose): One case reported in the literature of one patient who experienced nausea,
vomiting, abdominal pain, pyrexia, facial swelling, decreased neutrophil count, increased
transaminases.
8 to 10 g (single dose): Vomiting and gastrointestinal pain have been reported.
Paediatric population
One 3-year-old male exposed to a single dose of 400 mg experienced vomiting, diarrhoea and anorexia
and another 3-year-old male exposed to a single dose of 980 mg dose experienced decreased white
blood cell count and diarrhoea.
In the event of overdose, the patient should be observed and appropriate supportive treatment given.
5.
PHARMACOLOGICAL PROPERTIES
5.1 Pharmacodynamic properties
Pharmacotherapeutic group: protein-tyrosine kinase inhibitor, ATC code: L01XE01
Imatinib is a protein-tyrosine kinase inhibitor which potently inhibits the Bcr-Abl tyrosine kinase at
the in vitro , cellular and in vivo levels. The compound selectively inhibits proliferation and induces
apoptosis in Bcr-Abl positive cell lines as well as fresh leukaemic cells from Philadelphia chromosome
positive CML and acute lymphoblastic leukaemia (ALL) patients.
43
In vivo the compound shows anti-tumour activity as a single agent in animal models using Bcr-Abl
positive tumour cells.
Imatinib is also an inhibitor of the receptor tyrosine kinases for platelet-derived growth factor (PDGF),
PDGF-R, and stem cell factor (SCF), c-Kit, and inhibits PDGF- and SCF-mediated cellular events. In
vitro , imatinib inhibits proliferation and induces apoptosis in gastrointestinal stromal tumour (GIST)
cells, which express an activating kit mutation. Constitutive activation of the PDGF receptor or the
Abl protein tyrosine kinases as a consequence of fusion to diverse partner proteins or constitutive
production of PDGF have been implicated in the pathogenesis of MDS/MPD, HES/CEL and DFSP.
Imatinib inhibits signalling and proliferation of cells driven by dysregulated PDGFR and Abl kinase
activity.
Clinical studies in chronic myeloid leukaemia
The effectiveness of Glivec is based on overall haematological and cytogenetic response rates and
progression-free survival. Except in newly diagnosed chronic phase CML, there are no controlled
trials demonstrating a clinical benefit, such as improvement in disease-related symptoms or increased
survival.
Three large, international, open-label, non-controlled phase II studies were conducted in patients with
Philadelphia chromosome positive (Ph+) CML in advanced, blast or accelerated phase disease, other
Ph+ leukaemias or with CML in the chronic phase but failing prior interferon-alpha (IFN) therapy.
One large, open-label, multicentre, international randomised phase III study has been conducted in
patients with newly diagnosed Ph+ CML. In addition, children have been treated in two phase I
studies and one phase II study.
In all clinical studies 38–40% of patients were  60 years of age and 10–12% of patients were
 70 years of age.
Chronic phase, newly diagnosed: This phase III study in adult patients compared treatment with either
single-agent Glivec or a combination of interferon-alpha (IFN) plus cytarabine (Ara-C). Patients
showing lack of response (lack of complete haematological response (CHR) at 6 months, increasing
WBC, no major cytogenetic response (MCyR) at 24 months), loss of response (loss of CHR or MCyR)
or severe intolerance to treatment were allowed to cross over to the alternative treatment arm. In the
Glivec arm, patients were treated with 400 mg daily. In the IFN arm, patients were treated with a
target dose of IFN of 5 MIU/m 2 /day subcutaneously in combination with subcutaneous Ara-C
20 mg/m 2 /day for 10 days/month.
A total of 1,106 patients were randomised, 553 to each arm. Baseline characteristics were well
balanced between the two arms. Median age was 51 years (range 18–70 years), with 21.9% of patients
≥ 60 years of age. There were 59% males and 41% females; 89.9% caucasian and 4.7% black patients.
Seven years after the last patient had been recruited, the median duration of first-line treatment was 82
and 8 months in the Glivec and IFN arms, respectively. The median duration of second-line treatment
with Glivec was 64 months. Overall, in patients receiving first-line Glivec, the average daily dose
delivered was 406 ± 76 mg. The primary efficacy endpoint of the study is progression-free survival.
Progression was defined as any of the following events: progression to accelerated phase or blast
crisis, death, loss of CHR or MCyR, or in patients not achieving a CHR an increasing WBC despite
appropriate therapeutic management. Major cytogenetic response, haematological response, molecular
response (evaluation of minimal residual disease), time to accelerated phase or blast crisis and survival
are main secondary endpoints. Response data are shown in Table 3.
44
Table 3
Response in newly diagnosed CML Study (84-month data)
Glivec
IFN+Ara-C
(Best response rates)
n=553
n=553
Haematological response
CHR rate n (%)
534 (96.6%)*
313 (56.6%)*
[95% CI]
[94.7%, 97.9%]
[52.4%, 60.8%]
Cytogenetic response
Major response n (%)
490 (88.6%)*
129 (23.3%)*
[95% CI]
[85.7%, 91.1%]
[19.9%, 27.1%]
Complete CyR n (%)
456 (82.5%)*
64 (11.6%)*
Partial CyR n (%)
34 (6.1%)
65 (11.8%)
Molecular response **
Major response at 12 months (%)
153/305=50.2%
8/83=9.6%
Major response at 24 months (%)
73/104=70.2%
3/12=25%
Major response at 84 months (%)
102/116=87.9%
3/4=75%
* p<0.001, Fischer’s exact test
** molecular response percentages are based on available samples
Haematological response criteria (all responses to be confirmed after 4 weeks):
WBC < 10 x 10 9 /l, platelet < 450 x 10 9 /l, myelocyte+metamyelocyte < 5% in blood, no blasts and
promyelocytes in blood, basophils < 20%, no extramedullary involvement
Cytogenetic response criteria: complete (0% Ph+ metaphases), partial (1–35%), minor (36–65%) or
minimal (66–95%). A major response (0–35%) combines both complete and partial responses.
Major molecular response criteria : in the peripheral blood reduction of ≥ 3 logarithms in the
amount of Bcr-Abl transcripts (measured by real-time quantitative reverse transcriptase PCR assay)
over a standardised baseline.
Rates of complete haematological response, major cytogenetic response and complete cytogenetic
response on first-line treatment were estimated using the Kaplan-Meier approach, for which non-
responses were censored at the date of last examination. Using this approach, the estimated cumulative
response rates for first-line treatment with Glivec improved from 12 months of therapy to 84 months
of therapy as follows: CHR from 96.4% to 98.4% and CCyR from 69.5% to 87.2%, respectively.
With 7 years follow-up, there were 93 (16.8%) progression events in the Glivec arm: 37 (6.7%)
involving progression to accelerated phase/blast crisis, 31 (5.6%) loss of MCyR, 15 (2.7%) loss of
CHR or increase in WBC, and 10 (1.8%) CML unrelated deaths. In contrast, there were 165 (29.8%)
events in the IFN+Ara-C arm, of which 130 occurred during first-line treatment with IFN+Ara-C.
The estimated rate of patients free of progression to accelerated phase or blast crisis at 84 months was
significantly higher in the Glivec arm compared to the IFN arm (92.5% versus 85.1%, p<0.001). The
annual rate of progression to accelerated phase or blast crisis decreased with time on therapy and was
less than 1% annually in the fourth and fifth years. The estimated rate of progression-free survival at
84 months was 81.2% in the Glivec arm and 60.6% in the control arm (p<0.001). The yearly rates of
progression of any type for Glivec also decreased over time.
A total of 71 (12.8%) and 85 (15.4%) patients died in the Glivec and IFN+Ara-C groups, respectively.
At 84 months the estimated overall survival is 86.4% (83, 90) vs. 83.3% (80, 87) in the randomised
Glivec and the IFN+Ara-C groups, respectively (p=0.073, log-rank test). This time-to-event endpoint
is strongly affected by the high crossover rate from IFN+Ara-C to Glivec. The effect of Glivec
treatment on survival in chronic phase, newly diagnosed CML has been further examined in a
retrospective analysis of the above reported Glivec data with the primary data from another Phase III
study using IFN+Ara-C (n=325) in an identical regimen. In this retrospective analysis, the superiority
45
 
of Glivec over IFN+Ara-C in overall survival was demonstrated (p<0.001); within 42 months, 47
(8.5%) Glivec patients and 63 (19.4%) IFN+Ara-C patients had died.
The degree of cytogenetic response and molecular response had a clear effect on long-term outcomes
in patients on Glivec. Whereas an estimated 96% (93%) of patients with CCyR (PCyR) at 12 months
were free of progression to accelerated phase/blast crisis at 84 months, only 81% of patients without
MCyR at 12 months were free of progression to advanced CML at 84 months (p<0.001 overall,
p=0.25 between CCyR and PCyR). For patients with reduction in Bcr-Abl transcripts of at least
3 logarithms at 12 months, the probability of remaining free from progression to accelerated
phase/blast crisis was 99% at 84 months. Similar findings were found based on a 18-months landmark
analysis.
In this study, dose escalations were allowed from 400 mg daily to 600 mg daily, then from 600 mg
daily to 800 mg daily. After 42 months of follow-up, 11 patients experienced a confirmed loss (within
4 weeks) of their cytogenetic response. Of these 11 patients, 4 patients escalated up to 800 mg daily, 2
of whom regained a cytogenetic response (1 partial and 1 complete, the latter also achieving a
molecular response), while of the 7 patients who did not escalate the dose, only one regained a
complete cytogenetic response. The percentage of some adverse reactions was higher in the 40 patients
in whom the dose was increased to 800 mg daily compared to the population of patients before dose
increase (n=551). The more frequent adverse reactions included gastrointestinal haemorrhages,
conjunctivitis and elevation of transaminases or bilirubin. Other adverse reactions were reported with
lower or equal frequency.
Chronic phase, Interferon failure: 532 adult patients were treated at a starting dose of 400 mg. The
patients were distributed in three main categories: haematological failure (29%), cytogenetic failure
(35%), or intolerance to interferon (36%). Patients had received a median of 14 months of prior IFN
therapy at doses  25 x 10 6 IU/week and were all in late chronic phase, with a median time from
diagnosis of 32 months. The primary efficacy variable of the study was the rate of major cytogenetic
response (complete plus partial response, 0 to 35% Ph+ metaphases in the bone marrow).
In this study 65% of the patients achieved a major cytogenetic response that was complete in 53%
(confirmed 43%) of patients (Table 4). A complete haematological response was achieved in 95% of
patients.
Accelerated phase : 235 adult patients with accelerated phase disease were enrolled. The first
77 patients were started at 400 mg, the protocol was subsequently amended to allow higher dosing and
the remaining 158 patients were started at 600 mg.
The primary efficacy variable was the rate of haematological response, reported as either complete
haematological response, no evidence of leukaemia (i.e. clearance of blasts from the marrow and the
blood, but without a full peripheral blood recovery as for complete responses), or return to chronic
phase CML. A confirmed haematological response was achieved in 71.5% of patients (Table 4).
Importantly, 27.7% of patients also achieved a major cytogenetic response, which was complete in
20.4% (confirmed 16%) of patients. For the patients treated at 600 mg, the current estimates for
median progression-free-survival and overall survival were 22.9 and 42.5 months, respectively.
Myeloid blast crisis: 260 patients with myeloid blast crisis were enrolled. 95 (37%) had received prior
chemotherapy for treatment of either accelerated phase or blast crisis (“pretreated patients”) whereas
165 (63%) had not (“untreated patients”). The first 37 patients were started at 400 mg, the protocol
was subsequently amended to allow higher dosing and the remaining 223 patients were started at
600 mg.
The primary efficacy variable was the rate of haematological response, reported as either complete
haematological response, no evidence of leukaemia, or return to chronic phase CML using the same
criteria as for the study in accelerated phase. In this study, 31% of patients achieved a haematological
response (36% in previously untreated patients and 22% in previously treated patients). The rate of
response was also higher in the patients treated at 600 mg (33%) as compared to the patients treated at
46
400 mg (16%, p=0.0220). The current estimate of the median survival of the previously untreated and
treated patients was 7.7 and 4.7 months, respectively.
Lymphoid blast crisis : a limited number of patients were enrolled in phase I studies (n=10). The rate of
haematological response was 70% with a duration of 2 3 months.
Table 4
Response in adult CML studies
Study 0110
37-month data
Chronic phase,
IFN failure
(n=532)
Study 0109
40.5-month data
Accelerated phase
(n=235)
Study 0102
38-month data
Myeloid blast
crisis
(n=260)
% of patients (CI 95% )
Haematological response 1
95% (92.3–96.3)
71% (65.3–77.2)
31% (25.2–36.8)
Complete haematological
response (CHR)
95%
42%
8%
No evidence of leukaemia
(NEL)
Not applicable
12%
5%
Return to chronic phase (RTC)
Not applicable
17%
18%
Major cytogenetic response 2
65% (61.2–69.5)
28% (22.0–33.9)
15% (11.2–20.4)
Complete
53%
20%
7%
(Confirmed 3 ) [95% CI]
(43%) [38.6–47.2] (16%) [11.3–21.0]
(2%) [0.6–4.4]
Partial
12%
7%
8%
1 Haematological response criteria (all responses to be confirmed after 4 weeks):
CHR: Study 0110 [WBC < 10 x 10 9 /l, platelets < 450 x 10 9 /l, myelocyte+metamyelocyte < 5% in
blood, no blasts and promyelocytes in blood, basophils < 20%, no extramedullary
involvement] and in studies 0102 and 0109 [ANC  1.5 x 10 9 /l, platelets  100 x 10 9 /l, no
blood blasts, BM blasts < 5% and no extramedullary disease]
NEL Same criteria as for CHR but ANC  1 x 10 9 /l and platelets  20 x 10 9 /l (0102 and 0109
only)
RTC < 15% blasts BM and PB, < 30% blasts+promyelocytes in BM and PB, < 20% basophils in
PB, no extramedullary disease other than spleen and liver (only for 0102 and 0109).
BM = bone marrow, PB = peripheral blood
2 Cytogenetic response criteria:
A major response combines both complete and partial responses: complete (0% Ph+ metaphases),
partial (1–35%)
3 Complete cytogenetic response confirmed by a second bone marrow cytogenetic evaluation
performed at least one month after the initial bone marrow study.
Paediatric patients : A total of 26 paediatric patients of age < 18 years with either chronic phase CML
(n=11) or CML in blast crisis or Ph+ acute leukaemias (n=15) were enrolled in a dose-escalation phase
I trial. This was a population of heavily pretreated patients, as 46% had received prior BMT and 73% a
prior multi-agent chemotherapy. Patients were treated at doses of Glivec of 260 mg/m 2 /day (n=5),
340 mg/m 2 /day (n=9), 440 mg/m 2 /day (n=7) and 570 mg/m 2 /day (n=5). Out of 9 patients with chronic
phase CML and cytogenetic data available, 4 (44%) and 3 (33%) achieved a complete and partial
cytogenetic response, respectively, for a rate of MCyR of 77%.
A total of 51 paediatric patients with newly diagnosed and untreated CML in chronic phase have been
enrolled in an open-label, multicentre, single-arm phase II trial. Patients were treated with Glivec
340 mg/m 2 /day, with no interruptions in the absence of dose limiting toxicity. Glivec treatment
induces a rapid response in newly diagnosed paediatric CML patients with a CHR of 78% after
8 weeks of therapy. The high rate of CHR is accompanied by the development of a complete
cytogenetic response (CCyR) of 65% which is comparable to the results observed in adults.
Additionally, partial cytogenetic response (PCyR) was observed in 16% for a MCyR of 81%. The
47
 
majority of patients who achieved a CCyR developed the CCyR between months 3 and 10 with a
median time to response based on the Kaplan-Meier estimate of 5.6 months.
The European Medicines Agency has waived the obligation to submit the results of studies with
Glivec in all subsets of the paediatric population in Philadelphia chromosome (bcr-abl translocation)-
positive chronic myeloid leukaemia (see section 4.2 for information on paediatric use).
Clinical studies in Ph+ ALL
Newly diagnosed Ph+ ALL : In a controlled study (ADE10) of imatinib versus chemotherapy induction
in 55 newly diagnosed patients aged 55 years and over, imatinib used as single agent induced a
significantly higher rate of complete haematological response than chemotherapy (96.3% vs. 50%;
p=0.0001). When salvage therapy with imatinib was administered in patients who did not respond or
who responded poorly to chemotherapy, it resulted in 9 patients (81.8%) out of 11 achieving a
complete haematological response. This clinical effect was associated with a higher reduction in bcr-
abl transcripts in the imatinib-treated patients than in the chemotherapy arm after 2 weeks of therapy
(p=0.02). All patients received imatinib and consolidation chemotherapy (see Table 5) after induction
and the levels of bcr-abl transcripts were identical in the two arms at 8 weeks. As expected on the
basis of the study design, no difference was observed in remission duration, disease-free survival or
overall survival, although patients with complete molecular response and remaining in minimal
residual disease had a better outcome in terms of both remission duration (p=0.01) and disease-free
survival (p=0.02).
The results observed in a population of 211 newly diagnosed Ph+ ALL patients in four uncontrolled
clinical studies (AAU02, ADE04, AJP01 and AUS01) are consistent with the results described above.
Imatinib in combination with chemotherapy induction (see Table 5) resulted in a complete
haematological response rate of 93% (147 out of 158 evaluable patients) and in a major cytogenetic
response rate of 90% (19 out of 21 evaluable patients). The complete molecular response rate was 48%
(49 out of 102 evaluable patients). Disease-free survival (DFS) and overall survival (OS) constantly
exceeded 1 year and were superior to historical control (DFS p<0.001; OS p<0.0001) in two studies
(AJP01 and AUS01).
48
Table 5
Chemotherapy regimen used in combination with imatinib
Study ADE10
Prephase DEX 10 mg/m 2 oral, days 1-5; CP 200 mg/m 2 i.v., days 3, 4, 5; MTX
12 mg intrathecal, day 1
Remission induction DEX 10 mg/m 2 oral, days 6-7, 13-16; VCR 1 mg i.v., days 7, 14; IDA
8 mg/m 2 i.v. (0.5 h), days 7, 8, 14, 15; CP 500 mg/m 2 i.v.(1 h) day 1; Ara-
C 60 mg/m 2 i.v., days 22-25, 29-32
Consolidation
therapy I, III, V
MTX 500 mg/m 2 i.v. (24 h), days 1, 15; 6-MP 25 mg/m 2 oral, days 1-20
Consolidation
therapy II, IV
Ara-C 75 mg/m 2 i.v. (1 h), days 1-5; VM26 60 mg/m 2 i.v. (1 h), days 1-5
Study AAU02
Induction therapy ( de
novo Ph+ ALL)
Daunorubicin 30 mg/m 2 i.v., days 1-3, 15-16; VCR 2 mg total dose i.v.,
days 1, 8, 15, 22; CP 750 mg/m 2 i.v., days 1, 8; prednisone 60 mg/m 2
oral, days 1-7, 15-21; IDA 9 mg/m 2 oral, days 1-28; MTX 15 mg
intrathecal, days 1, 8, 15, 22; Ara-C 40 mg intrathecal, days 1, 8, 15, 22;
methylprednisolone 40 mg intrathecal, days 1, 8, 15, 22
Consolidation ( de
novo Ph+ ALL)
Ara-C 1,000 mg/m 2 /12 h i.v.(3 h), days 1-4; mitoxantrone 10 mg/m 2 i.v.
days 3-5; MTX 15 mg intrathecal, day 1; methylprednisolone 40 mg
intrathecal, day 1
Study ADE04
Prephase
DEX 10 mg/m 2 oral, days 1-5; CP 200 mg/m 2 i.v., days 3-5; MTX 15 mg
intrathecal, day 1
Induction therapy I
DEX 10 mg/m 2 oral, days 1-5; VCR 2 mg i.v., days 6, 13, 20;
daunorubicin 45 mg/m 2 i.v., days 6-7, 13-14
Induction therapy II
CP 1 g/m 2 i.v. (1 h), days 26, 46; Ara-C 75 mg/m 2 i.v. (1 h), days 28-31,
35-38, 42-45; 6-MP 60 mg/m 2 oral, days 26-46
Consolidation
therapy
DEX 10 mg/m 2 oral, days 1-5; vindesine 3 mg/m 2 i.v., day 1; MTX
1.5 g/m 2 i.v. (24 h), day 1; etoposide 250 mg/m 2 i.v. (1 h) days 4-5; Ara-
C 2x 2 g/m 2 i.v. (3 h, q 12 h), day 5
Study AJP01
Induction therapy
CP 1.2 g/m 2 i.v. (3 h), day 1; daunorubicin 60 mg/m 2 i.v. (1 h), days 1-3;
vincristine 1.3 mg/m 2 i.v., days 1, 8, 15, 21; prednisolone 60 mg/m 2 /day
oral
Consolidation
therapy
Alternating chemotherapy course: high dose chemotherapy with MTX
1 g/m 2 i.v. (24 h), day 1, and Ara-C 2 g/m 2 i.v. (q 12 h), days 2-3, for
4 cycles
Maintenance
VCR 1.3 g/m 2 i.v., day 1; prednisolone 60 mg/m 2 oral, days 1-5
Study AUS01
Hyper-CVAD regimen: CP 300 mg/m 2 i.v. (3 h, q 12 h), days 1-3;
vincristine 2 mg i.v., days 4, 11; doxorubicine 50 mg/m 2 i.v. (24 h),
day 4; DEX 40 mg/day on days 1-4 and 11-14, alternated with MTX
1 g/m 2 i.v. (24 h), day 1, Ara-C 1 g/m 2 i.v. (2 h, q 12 h), days 2-3 (total of
8 courses)
Maintenance VCR 2 mg i.v. monthly for 13 months; prednisolone 200 mg oral, 5 days
per month for 13 months
All treatment regimens include administration of steroids for CNS prophylaxis.
Ara-C: cytosine arabinoside; CP: cyclophosphamide; DEX: dexamethasone; MTX: methotrexate;
6-MP: 6-mercaptopurine VM26: Teniposide; VCR: vincristine; IDA: idarubicine; i.v.:
intravenous
49
Induction-
consolidation therapy
Relapsed/refractory Ph+ ALL: When imatinib was used as single agent in patients with
relapsed/refractory Ph+ ALL, it resulted, in the 53 out of 411 patients evaluable for response, in a
haematological response rate of 30% (9% complete) and a major cytogenetic response rate of 23%.
(Of note, out of the 411 patients, 353 were treated in an expanded access program without primary
response data collected.) The median time to progression in the overall population of 411 patients with
relapsed/refractory Ph+ ALL ranged from 2.6 to 3.1 months, and median overall survival in the
401 evaluable patients ranged from 4.9 to 9 months. The data was similar when re-analysed to include
only those patients age 55 or older.
Clinical studies in MDS/MPD
Experience with Glivec in this indication is very limited and is based on haematological and
cytogenetic response rates. There are no controlled trials demonstrating a clinical benefit or increased
survival. One open label, multicentre, phase II clinical trial (study B2225) was conducted testing
Glivec in diverse populations of patients suffering from life-threatening diseases associated with Abl,
Kit or PDGFR protein tyrosine kinases. This study included 7 patients with MDS/MPD who were
treated with Glivec 400 mg daily. Three patients presented a complete haematological response (CHR)
and one patient experienced a partial haematological response (PHR). At the time of the original
analysis, three of the four patients with detected PDGFR gene rearrangements developed
haematological response (2 CHR and 1 PHR). The age of these patients ranged from 20 to 72 years. In
addition a further 24 patients with MDS/MPD were reported in 13 publications. 21 patients were
treated with Glivec 400 mg daily, while the other 3 patients received lower doses. In eleven patients
PDGFR gene rearrangements was detected, 9 of them achieved a CHR and 1 PHR. The age of these
patients ranged from 2 to 79 years. In a recent publication updated information from 6 of these
11 patients revealed that all these patients remained in cytogenetic remission (range 32-38 months).
The same publication reported long term follow-up data from 12 MDS/MPD patients with PDGFR
gene rearrangements (5 patients from study B2225). These patients received Glivec for a median of
47 months (range 24 days – 60 months). In 6 of these patients follow-up now exceeds 4 years. Eleven
patients achieved rapid CHR; ten had complete resolution of cytogenetic abnormalities and a decrease
or disappearance of fusion transcripts as measured by RT-PCR. Haematological and cytogenetic
responses have been sustained for a median of 49 months (range 19-60) and 47 months (range 16-59),
respectively. The overall survival is 65 months since diagnosis (range 25-234). Glivec administration
to patients without the genetic translocation generally results in no improvement.
Clinical studies in HES/CEL
One open-label, multicentre, phase II clinical trial (study B2225) was conducted testing Glivec in
diverse populations of patients suffering from life-threatening diseases associated with Abl, Kit or
PDGFR protein tyrosine kinases. In this study, 14 patients with HES/CEL were treated with 100 mg to
1,000 mg of Glivec daily. A further 162 patients with HES/CEL, reported in 35 published case reports
and case series received Glivec at doses from 75 mg to 800 mg daily. Cytogenetic abnormalities were
evaluated in 117 of the total population of 176 patients. In 61 of these 117 patients FIP1L1-PDGFRα
fusion kinase was identified. An additional four HES patients were found to be FIP1L1-PDGFRα-
positive in other 3 published reports. All 65 FIP1L1-PDGFRα fusion kinase positive patients achieved
a CHR sustained for months (range from 1+ to 44+ months censored at the time of the reporting). As
reported in a recent publication 21 of these 65 patients also achieved complete molecular remission
with a median follow-up of 28 months (range 13-67 months). The age of these patients ranged from 25
to 72 years. Additionally, improvements in symptomatology and other organ dysfunction
abnormalities were reported by the investigators in the case reports. Improvements were reported in
cardiac, nervous, skin/subcutaneous tissue, respiratory/thoracic/mediastinal,
musculoskeletal/connective tissue/vascular, and gastrointestinal organ systems.
Clinical studies in unresectable and/or metastatic GIST
One phase II, open-label, randomised, uncontrolled multinational study was conducted in patients with
unresectable or metastatic malignant gastrointestinal stromal tumours (GIST). In this study
147 patients were enrolled and randomised to receive either 400 mg or 600 mg orally once daily for up
to 36 months. These patients ranged in age from 18 to 83 years old and had a pathologic diagnosis of
Kit-positive malignant GIST that was unresectable and/or metastatic. Immunohistochemistry was
50
routinely performed with Kit antibody (A-4502, rabbit polyclonal antiserum, 1:100; DAKO
Corporation, Carpinteria, CA) according to analysis by an avidin-biotin-peroxidase complex method
after antigen retrieval.
The primary evidence of efficacy was based on objective response rates. Tumours were required to be
measurable in at least one site of disease, and response characterisation based on Southwestern
Oncology Group (SWOG) criteria. Results are provided in Table 6.
Table 6
Best tumour response in trial STIB2222 (GIST)
Best response
All doses (n=147)
400 mg (n=73)
600 mg (n=74)
n (%)
Complete response
1(0.7)
Partial response
98 (66.7)
Progressive disease
23 (15.6)
Not evaluable
5 (3.4)
Unknown
2 (1.4)
There were no differences in response rates between the two dose groups. A significant number of
patients who had stable disease at the time of the interim analysis achieved a partial response with
longer treatment (median follow-up 31 months). Median time to response was 13 weeks (95% C.I. 12–
23). Median time to treatment failure in responders was 122 weeks (95% C.I 106–147), while in the
overall study population it was 84 weeks (95% C.I 71–109). The median overall survival has not been
reached. The Kaplan-Meier estimate for survival after 36-month follow-up is 68%.
In two clinical studies (study B2222 and an intergroup study S0033) the daily dose of Glivec was
escalated to 800 mg in patients progressing at the lower daily doses of 400 mg or 600 mg. The daily
dose was escalated to 800 mg in a total of 103 patients; 6 patients achieved a partial response and 21
stabilisation of their disease after dose escalation for an overall clinical benefit of 26%. From the
safety data available, escalating the dose to 800 mg daily in patients progressing at lower doses of
400 mg or 600 mg daily does not seem to affect the safety profile of Glivec.
Clinical study in adjuvant GIST
In the adjuvant setting, Glivec was investigated in a multicentre, double-blind, long-term, placebo-
controlled phase III study (Z9001) involving 773 patients. The ages of these patients ranged from 18
to 91 years. Patients were included who had a histological diagnosis of primary GIST expressing Kit
protein by immunochemistry and a tumour size ≥ 3 cm in maximum dimension, with complete gross
resection of primary GIST within 14-70 days prior to registration. After resection of primary GIST,
patients were randomised to one of the two arms: Glivec at 400 mg/day or matching placebo for one
year.
The primary endpoint of the study was recurrence-free survival (RFS), defined as the time from date
of randomisation to the date of recurrence or death from any cause.
Glivec significantly prolonged RFS, with 75% of patients being recurrence-free at 38 months in the
Glivec group vs. 20 months in the placebo group (95% CIs, [30 - non-estimable]; [14 - non-estimable],
respectively); (hazard ratio = 0.398 [0.259-0.610], p<0.0001). At one year the overall RFS was
significantly better for Glivec (97.7%) vs. placebo (82.3%), (p<0.0001). The risk of recurrence was
thus reduced by approximately 89% as compared with placebo (hazard ratio = 0.113 [0.049-0.264]).
The risk of recurrence in patients after surgery of their primary GIST was retrospectively assessed
based on the following prognostic factors: tumour size, mitotic index, tumour location. Mitotic index
data were available for 556 of the 773 intention-to-treat (ITT) population. The results of subgroup
analyses according to the United States National Institutes of Health (NIH) and the Armed Forces
51
Stable disease
18 (12.2)
 
Institute of Pathology (AFIP) risk classifications are shown in Table 7. No benefit was observed in the
low and very low risk groups. No overall survival benefit has been observed.
Table 7
Summary of Z9001 trial RFS analyses by NIH and AFIP risk classifications
Risk
criteria
Risk Level
% of
patients
No. of events /
No. of patients
Overall hazard
ratio (95%CI)*
RFS rates (%)
12 month
24 month
Glivec vs placebo
Glivec vs
placebo
Glivec vs
placebo
Low
29.5
0/86 vs. 2/90
N.E.
100 vs. 98.7 100 vs. 95.5
NIH
Intermediate
25.7
4/75 vs. 6/78
0.59 (0.17; 2.10) 100 vs. 94.8 97.8 vs. 89.5
High
44.8
21/140 vs. 51/127
0.29 (0.18; 0.49) 94.8 vs. 64.0 80.7 vs. 46.6
Very Low
20.7
0/52 vs. 2/63
N.E.
100 vs. 98.1 100 vs. 93.0
AFIP
Low
25.0
2/70 vs. 0/69
N.E.
100 vs. 100
97.8 vs. 100
Moderate
24.6
2/70 vs. 11/67
0.16 (0.03; 0.70) 97.9 vs. 90.8 97.9 vs. 73.3
High
29.7
16/84 vs. 39/81
0.27 (0.15; 0.48) 98.7 vs. 56.1 79.9 vs. 41.5
* Full follow-up period; NE – Not estimable
Clinical studies in DFSP
One phase II, open label, multicentre clinical trial (study B2225) was conducted including 12 patients
with DFSP treated with Glivec 800 mg daily. The age of the DFSP patients ranged from 23 to
75 years; DFSP was metastatic, locally recurrent following initial resective surgery and not considered
amenable to further resective surgery at the time of study entry. The primary evidence of efficacy was
based on objective response rates. Out of the 12 patients enrolled, 9 responded, one completely and 8
partially. Three of the partial responders were subsequently rendered disease free by surgery. The
median duration of therapy in study B2225 was 6.2 months, with a maximum duration of 24.3 months.
A further 6 DFSP patients treated with Glivec were reported in 5 published case reports, their ages
ranging from 18 months to 49 years. The adult patients reported in the published literature were
treated with either 400 mg (4 cases) or 800 mg (1 case) Glivec daily. The paediatric patient received
400 mg/m 2 /daily, subsequently increased to 520 mg/m 2 /daily. 5 patients responded, 3 completely and
2 partially. The median duration of therapy in the published literature ranged between 4 weeks and
more than 20 months. The translocation t(17:22)[(q22:q13)], or its gene product, was present in nearly
all responders to Glivec treatment.
5.2 Pharmacokinetic properties
Pharmacokinetics of Glivec
The pharmacokinetics of Glivec have been evaluated over a dosage range of 25 to 1,000 mg. Plasma
pharmacokinetic profiles were analysed on day 1 and on either day 7 or day 28, by which time plasma
concentrations had reached steady state.
Absorption
Mean absolute bioavailability for the capsule formulation is 98%. There was high between-patient
variability in plasma imatinib AUC levels after an oral dose. When given with a high-fat meal, the rate
of absorption of imatinib was minimally reduced (11% decrease in C max and prolongation of t max by
1.5 h), with a small reduction in AUC (7.4%) compared to fasting conditions. The effect of prior
gastrointestinal surgery on drug absorption has not been investigated.
Distribution
At clinically relevant concentrations of imatinib, binding to plasma proteins was approximately 95%
on the basis of in vitro experiments, mostly to albumin and alpha-acid-glycoprotein, with little binding
to lipoprotein.
52
 
Metabolism
The main circulating metabolite in humans is the N-demethylated piperazine derivative, which shows
similar in vitro potency to the parent. The plasma AUC for this metabolite was found to be only 16%
of the AUC for imatinib. The plasma protein binding of the N-demethylated metabolite is similar to
that of the parent compound.
Imatinib and the N-demethyl metabolite together accounted for about 65% of the circulating
radioactivity (AUC (0-48h) ). The remaining circulating radioactivity consisted of a number of minor
metabolites.
The in vitro results showed that CYP3A4 was the major human P450 enzyme catalysing the
biotransformation of imatinib. Of a panel of potential comedications (acetaminophen, aciclovir,
allopurinol, amphotericin, cytarabine, erythromycin, fluconazole, hydroxyurea, norfloxacin, penicillin
V) only erythromycin (IC 50 50 µM) and fluconazole (IC 50 118 µM) showed inhibition of imatinib
metabolism which could have clinical relevance.
Imatinib was shown in vitro to be a competitive inhibitor of marker substrates for CYP2C9, CYP2D6
and CYP3A4/5. K i values in human liver microsomes were 27, 7.5 and 7.9 mol/l, respectively.
Maximal plasma concentrations of imatinib in patients are 2–4 mol/l, consequently an inhibition of
CYP2D6 and/or CYP3A4/5-mediated metabolism of co-administered drugs is possible. Imatinib did
not interfere with the biotransformation of 5-fluorouracil, but it inhibited paclitaxel metabolism as a
result of competitive inhibition of CYP2C8 (K i = 34.7 µM). This K i value is far higher than the
expected plasma levels of imatinib in patients, consequently no interaction is expected upon co-
administration of either 5-fluorouracil or paclitaxel and imatinib.
Elimination
Based on the recovery of compound(s) after an oral 14 C-labelled dose of imatinib, approximately 81%
of the dose was recovered within 7 days in faeces (68% of dose) and urine (13% of dose). Unchanged
imatinib accounted for 25% of the dose (5% urine, 20% faeces), the remainder being metabolites.
Plasma pharmacokinetics
Following oral administration in healthy volunteers, the t ½ was approximately 18 h, suggesting that
once-daily dosing is appropriate. The increase in mean AUC with increasing dose was linear and dose
proportional in the range of 25–1,000 mg imatinib after oral administration. There was no change in
the kinetics of imatinib on repeated dosing, and accumulation was 1.5–2.5-fold at steady state when
dosed once daily.
Pharmacokinetics in GIST patients
In patients with GIST steady-state exposure was 1.5-fold higher than that observed for CML patients
for the same dosage (400 mg daily). Based on preliminary population pharmacokinetic analysis in
GIST patients, there were three variables (albumin, WBC and bilirubin) found to have a statistically
significant relationship with imatinib pharmacokinetics. Decreased values of albumin caused a
reduced clearance (CL/f); and higher levels of WBC led to a reduction of CL/f. However, these
associations are not sufficiently pronounced to warrant dose adjustment. In this patient population, the
presence of hepatic metastases could potentially lead to hepatic insufficiency and reduced metabolism.
Population pharmacokinetics
Based on population pharmacokinetic analysis in CML patients, there was a small effect of age on the
volume of distribution (12% increase in patients > 65 years old). This change is not thought to be
clinically significant. The effect of bodyweight on the clearance of imatinib is such that for a patient
weighing 50 kg the mean clearance is expected to be 8.5 l/h, while for a patient weighing 100 kg the
clearance will rise to 11.8 l/h. These changes are not considered sufficient to warrant dose adjustment
based on kg bodyweight. There is no effect of gender on the kinetics of imatinib.
53
Pharmacokinetics in children
As in adult patients, imatinib was rapidly absorbed after oral administration in paediatric patients in
both phase I and phase II studies. Dosing in children at 260 and 340 mg/m 2 /day achieved the same
exposure, respectively, as doses of 400 mg and 600 mg in adult patients. The comparison of AUC (0-24)
on day 8 and day 1 at the 340 mg/m 2 /day dose level revealed a 1.7-fold drug accumulation after
repeated once-daily dosing.
Organ function impairment
Imatinib and its metabolites are not excreted via the kidney to a significant extent. Patients with mild
and moderate impairment of renal function appear to have a higher plasma exposure than patients with
normal renal function. The increase is approximately 1.5- to 2-fold, corresponding to a 1.5-fold
elevation of plasma AGP, to which imatinib binds strongly. The free drug clearance of imatinib is
probably similar between patients with renal impairment and those with normal renal function, since
renal excretion represents only a minor elimination pathway for imatinib (see sections 4.2 and 4.4).
Although the results of pharmacokinetic analysis showed that there is considerable inter-subject
variation, the mean exposure to imatinib did not increase in patients with varying degrees of liver
dysfunction as compared to patients with normal liver function (see sections 4.2, 4.4 and 4.8).
5.3 Preclinical safety data
The preclinical safety profile of imatinib was assessed in rats, dogs, monkeys and rabbits.
Multiple dose toxicity studies revealed mild to moderate haematological changes in rats, dogs and
monkeys, accompanied by bone marrow changes in rats and dogs.
The liver was a target organ in rats and dogs. Mild to moderate increases in transaminases and slight
decreases in cholesterol, triglycerides, total protein and albumin levels were observed in both species.
No histopathological changes were seen in rat liver. Severe liver toxicity was observed in dogs treated
for 2 weeks, with elevated liver enzymes, hepatocellular necrosis, bile duct necrosis, and bile duct
hyperplasia.
Renal toxicity was observed in monkeys treated for 2 weeks, with focal mineralisation and dilation of
the renal tubules and tubular nephrosis. Increased blood urea nitrogen (BUN) and creatinine were
observed in several of these animals. In rats, hyperplasia of the transitional epithelium in the renal
papilla and in the urinary bladder was observed at doses  6 mg/kg in the 13-week study, without
changes in serum or urinary parameters. An increased rate of opportunistic infections was observed
with chronic imatinib treatment.
In a 39-week monkey study, no NOAEL (no observed adverse effect level) was established at the
lowest dose of 15 mg/kg, approximately one-third the maximum human dose of 800 mg based on
body surface. Treatment resulted in worsening of normally suppressed malarial infections in these
animals.
Imatinib was not considered genotoxic when tested in an in vitro bacterial cell assay (Ames test), an in
vitro mammalian cell assay (mouse lymphoma) and an in vivo rat micronucleus test. Positive
genotoxic effects were obtained for imatinib in an in vitro mammalian cell assay (Chinese hamster
ovary) for clastogenicity (chromosome aberration) in the presence of metabolic activation. Two
intermediates of the manufacturing process, which are also present in the final product, are positive for
mutagenesis in the Ames assay. One of these intermediates was also positive in the mouse lymphoma
assay.
In a study of fertility, in male rats dosed for 70 days prior to mating, testicular and epididymal weights
and percent motile sperm were decreased at 60 mg/kg, approximately equal to the maximum clinical
dose of 800 mg/day, based on body surface area. This was not seen at doses ≤ 20 mg/kg. A slight to
moderate reduction in spermatogenesis was also observed in the dog at oral doses  30 mg/kg. When
54
female rats were dosed 14 days prior to mating and through to gestational day 6, there was no effect on
mating or on number of pregnant females. At a dose of 60 mg/kg, female rats had significant post-
implantation foetal loss and a reduced number of live foetuses. This was not seen at doses ≤ 20 mg/kg.
In an oral pre- and postnatal development study in rats, red vaginal discharge was noted in the
45 mg/kg/day group on either day 14 or day 15 of gestation. At the same dose, the number of stillborn
pups as well as those dying between postpartum days 0 and 4 was increased. In the F 1 offspring, at the
same dose level, mean body weights were reduced from birth until terminal sacrifice and the number
of litters achieving criterion for preputial separation was slightly decreased. F 1 fertility was not
affected, while an increased number of resorptions and a decreased number of viable foetuses was
noted at 45 mg/kg/day. The no observed effect level (NOEL) for both the maternal animals and the F 1
generation was 15 mg/kg/day (one quarter of the maximum human dose of 800 mg).
Imatinib was teratogenic in rats when administered during organogenesis at doses ≥ 100 mg/kg,
approximately equal to the maximum clinical dose of 800 mg/day, based on body surface area.
Teratogenic effects included exencephaly or encephalocele, absent/reduced frontal and absent parietal
bones. These effects were not seen at doses ≤ 30 mg/kg.
In the 2-year rat carcinogenicity study administration of imatinib at 15, 30 and 60 mg/kg/day resulted
in a statistically significant reduction in the longevity of males at 60 mg/kg/day and females at
≥30 mg/kg/day. Histopathological examination of decedents revealed cardiomyopathy (both sexes),
chronic progressive nephropathy (females) and preputial gland papilloma as principal causes of death
or reasons for sacrifice. Target organs for neoplastic changes were the kidneys, urinary bladder,
urethra, preputial and clitoral gland, small intestine, parathyroid glands, adrenal glands and non-
glandular stomach.
Papilloma/carcinoma of the preputial/clitoral gland were noted from 30mg/kg/day onwards,
representing approximately 0.5 or 0.3 times the human daily exposure (based on AUC) at 400 mg/day
or 800 mg/day, respectively, and 0.4 times the daily exposure in children (based on AUC) at
340 mg/m 2 /day. The no observed effect level (NOEL) was 15 mg/kg/day. The renal
adenoma/carcinoma, the urinary bladder and urethra papilloma, the small intestine adenocarcinomas,
the parathyroid glands adenomas, the benign and malignant medullary tumours of the adrenal glands
and the non-glandular stomach papillomas/carcinomas were noted at 60 mg/kg/day, representing
approximately 1.7 or 1 times the human daily exposure (based on AUC) at 400 mg/day or 800 mg/day,
respectively, and 1.2 times the daily exposure in children (based on AUC) at 340 mg/m 2 /day. The no
observed effect level (NOEL) was 30 mg/kg/day.
The mechanism and relevance of these findings in the rat carcinogenicity study for humans are not yet
clarified.
Non-neoplastic lesions not identified in earlier preclinical studies were the cardiovascular system,
pancreas, endocrine organs and teeth. The most important changes included cardiac hypertrophy and
dilatation, leading to signs of cardiac insufficiency in some animals.
55
6.
PHARMACEUTICAL PARTICULARS
6.1 List of excipients
Capsule content: Cellulose microcrystalline
Crospovidone
Magnesium stearate
Silica colloidal, anhydrous
Capsule shell:
Gelatin
Iron oxide, red (E172)
Iron oxide, yellow (E172)
Titanium dioxide (E171)
Printing ink:
Iron oxide, red (E172)
Shellac
Soya lecithin
6.2 Incompatibilities
Not applicable.
6.3 Shelf life
2 years
6.4 Special precautions for storage
Do not store above 30C.
Store in the original package in order to protect from moisture.
6.5 Nature and contents of container
PVC/aluminium blisters
Packs containing 24, 48, 96, 120 and 180 capsules.
Not all pack sizes may be marketed.
6.6 Special precautions for disposal
No special requirements.
7.
MARKETING AUTHORISATION HOLDER
Novartis Europharm Limited
Wimblehurst Road
Horsham
West Sussex, RH12 5AB
United Kingdom
56
8.
MARKETING AUTHORISATION NUMBER(S)
EU/1/01/198/002-006
9.
DATE OF FIRST AUTHORISATION/RENEWAL OF THE AUTHORISATION
Date of first authorisation: 07.11.2001
Date of first renewal: 07.11.2006
10. DATE OF REVISION OF THE TEXT
Detailed information on this product is available on the website of the European Medicines Agency
http://www.ema.europa.eu
57
1.
MARKETING AUTHORISATION HOLDER
Novartis Europharm Limited
Wimblehurst Road
Horsham
West Sussex, RH12 5AB
United Kingdom
84
8.
MARKETING AUTHORISATION NUMBER(S)
EU/1/01/198/007
EU/1/01/198/008
EU/1/01/198/011
EU/1/01/198/012
9.
DATE OF FIRST AUTHORISATION/RENEWAL OF THE AUTHORISATION
Date of first authorisation: 11.11.2003
Date of first renewal: 07.11.2006
10. DATE OF REVISION OF THE TEXT
Detailed information on this product is available on the website of the European Medicines Agency
http://www.ema.europa.eu
85
1.
FURTHER INFORMATION
What Glivec contains
-
The active substance is imatinib mesilate. Each tablet of Glivec contains 400 mg imatinib
mesilate.
-
The other ingredients are microcrystalline cellulose, crospovidone, hypromellose, magnesium
stearate and anhydrous colloidal silica.
-
The tablet coating is made of red iron oxide (E172), yellow iron oxide (E172), macrogol, talc
and hypromellose.
What Glivec looks like and contents of the pack
Glivec 400 mg film-coated tablets are very dark yellow to brownish-orange oval tablets. They have
“NVR” on one side and “SL” on the other side.
They are supplied in packs containing 10, 30 or 90 tablets, but these may not all be available in your
country.
Marketing Authorisation Holder
Novartis Europharm Limited
Wimblehurst Road
Horsham
West Sussex, RH12 5AB
United Kingdom
160
Manufacturer
Novartis Pharma GmbH
Roonstrasse 25
D-90429 Nuremberg
Germany
For any information about this medicine, please contact the local representative of the Marketing
Authorisation Holder.
België/Belgique/Belgien
Novartis Pharma N.V.
Tél/Tel: +32 2 246 16 11
Luxembourg/Luxemburg
Novartis Pharma GmbH
Tél/Tel: +49 911 273 0
България
Novartis Pharma Services Inc.
Тел.: +359 2 489 98 28
Magyarország
Novartis Hungária Kft. Pharma
Tel.: +36 1 457 65 00
Česká republika
Novartis s.r.o.
Tel: +420 225 775 111
Malta
Novartis Pharma Services Inc.
Tel: +356 2298 3217
Danmark
Novartis Healthcare A/S
Tlf: +45 39 16 84 00
Nederland
Novartis Pharma B.V.
Tel: +31 26 37 82 111
Deutschland
Novartis Pharma GmbH
Tel: +49 911 273 0
Norge
Novartis Norge AS
Tlf: +47 23 05 20 00
Eesti
Novartis Pharma Services Inc.
Tel: +372 66 30 810
Österreich
Novartis Pharma GmbH
Tel: +43 1 86 6570
Ελλά
Novartis (Hellas) A.E.B.E.
Τηλ: +30 210 281 17 12
Polska
Novartis Poland Sp. z o.o.
Tel.: +48 22 375 4888
España
Novartis Farmacéutica, S.A.
Tel: +34 93 306 42 00
Portugal
Novartis Farma - Produtos Farmacêuticos, S.A.
Tel: +351 21 000 8600
France
Novartis Pharma S.A.S.
Tél: +33 1 55 47 66 00
România
Novartis Pharma Services Romania SRL
Tel: +40 21 31299 01
Ireland
Novartis Ireland Limited
Tel: +353 1 260 12 55
Slovenija
Novartis Pharma Services Inc.
Tel: +386 1 300 75 50
Ísland
Vistor hf.
Sími: +354 535 7000
Slovenská republika
Novartis Slovakia s.r.o.
Tel: +421 2 5542 5439
161
Italia
Novartis Farma S.p.A.
Tel: +39 02 96 54 1
Suomi/Finland
Novartis Finland Oy
Puh/Tel: +358 (0)10 6133 200
Κύπρς
Novartis Pharma Services Inc.
Τηλ: +357 22 690 690
Sverige
Novartis Sverige AB
Tel: +46 8 732 32 00
Latvija
Novartis Pharma Services Inc.
Tel: +371 67 887 070
United Kingdom
Novartis Pharmaceuticals UK Ltd.
Tel: +44 1276 698370
Lietuva
Novartis Pharma Services Inc.
Tel: +370 5 269 16 50
This leaflet was last approved in
Detailed information on this medicine is available on the European Medicines Agency website:
http://www.ema.europa.eu
162


Source: European Medicines Agency



- Please bookmark this page (add it to your favorites).
- If you wish to link to this page, you can do so by referring to the URL address below this line.



https://theodora.com/drugs/eu/glivec.html

Copyright © 1995-2021 ITA all rights reserved.