20-01-2021
20-01-2021
דבכנ ת/חקור ,ה/אפור
לע ךעידוהל החמש םרפואינ לש הרושיא היוותה תפסות
רישכתל :םי
Revlimid 2.5 mg Hard Capsule
דימילבר
2.5
ג"מ
החישק היילבט
Revlimid 5 mg Hard Capsule
דימילבר
5
ג"מ החישק היילבט
Revlimid 7.5 mg Hard Capsule
דימילבר
7.5
ג"מ החישק היילבט
Revlimid 10 mg Hard Capsule
דימילבר
10
ג"מ החישק היילבט
Revlimid 15 mg Hard Capsule
דימילבר
15
ג"מ החישק היילבט
Revlimid 20 mg Hard Capsule
דימילבר
20
ג"מ החישק היילבט
Revlimid 25 mg Hard Capsule
דימילבר
25
ג"מ החישק היילבט
םירמוחה
םיליעפה
:םתומכו
Revlimid 2.5 mg: each hard capsule contains 2.5 mg Lenalidomide.
Revlimid 5 mg: each hard capsule contains 5 mg Lenalidomide.
Revlimid 7.5 mg: each hard capsule contains 7.5 mg Lenalidomide.
Revlimid 10 mg: each hard capsule contains 10 mg Lenalidomide.
Revlimid 15 mg: each hard capsule contains 15 mg Lenalidomide.
Revlimid 20 mg: each hard capsule contains 20 mg Lenalidomide.
Revlimid 25 mg: each hard capsule contains 25 mg Lenalidomide.
:השדחה היוותהה חסונ
Multiple Myeloma
Revlimid is indicated for:
The maintenance treatment of adult patients with newly diagnosed multiple myeloma (MM) who
have undergone autologous stem cell transplantation.
Previously untreated multiple myeloma in adult patients who are not eligible for transplant.
In combination with dexamethasone treatment of multiple myeloma patients who have received
at least one prior therapy.
Myelodysplastic Syndromes
REVLIMID is indicated for patients with transfusion-dependent anemia due to low- or intermediate-
1-risk
myelodysplastic syndromes (MDS) associated with a deletion 5q cytogenetic abnormality with or
without additional cytogenetic abnormalities.
Revlimid 7.5 mg is not indicated for treatment in MDS.
Mantle Cell Lymphoma
REVLIMID is indicated for the treatment of adult patients with relapsed and/or refractory mantle
cell lymphoma (MCL).
ףסונב .הרמחה םניאש םיפסונ םייוניש ןולעב .הרמחה םיווהמה םייונישה םיניוצמ וז העדוהב
טסקט ,יתחת וקב ןמוסמ ףסוותהש טסקט .הצוח וקב ןמוסמ רסוהש
םינוכדעה םיירקיעה :םיאבה םיפיעסב ושענ אפורל ןולעב
5.
WARNINGS AND PRECAUTIONS
…….
5.2 Reproductive Risk and Special Prescribing Requirements (Revlimid RMP-PPP)
…….
Prescriptions for women of childbearing potential can be for a maximum duration of 4 weeks, and prescriptions
for all other patients can be for a maximum duration of 12 weeks.
5.3 Other warnings and precautions of use
Hematologic Toxicity
REVLIMID can cause significant neutropenia and thrombocytopenia. REVLIMID can cause significant neutropenia
and thrombocytopenia. Monitor patients with neutropenia for signs of infection. Advise patients to observe for bleeding
or bruising, especially with use of concomitant medication that may increase risk of bleeding. Patients taking
REVLIMID should have their complete blood counts assessed periodically as described below [see Dosage and
Administration (2.1, 2.2, 2.3)]. Patients should be advised to promptly report febrile episodes and dose reductions may
be required. Patients and physicians are advised to be observant for signs and symptoms of bleeding, including
petechiae and epistaxes, especially in patients receiving concomitant medicinal products susceptible to induce
bleeding.
Patients taking REVLIMID in combination with dexamethasone or as REVLIMID maintenance therapy for MM should
have their complete blood counts (CBC) assessed every 7 days (weekly) for the first 2 cycles, on Days 1 and 15 of
Cycle 3, and every 28 days (4 weeks) thereafter. A dose interruption and/or dose reduction may be required [see
Dosage and Administration (2.1)]. In the MM maintenance therapy trials, Grade 3 or 4 neutropenia was reported in up
to 59% of REVLIMID treated patients and Grade 3 or 4 thrombocytopenia in up to 38% of REVLIMID-treated patients
[see Adverse Reactions (6.1)].
…….
myelomaPatients taking REVLIMID for MCL should have their complete blood counts monitored weekly for the first
cycle (28 days), every 2 weeks during cycles 2-4, and then monthly thereafter. Patients may require dose interruption
and/or dose reduction. In the MCL trial, Grade 3 or 4 neutropenia was reported in 43% of the patients. Grade 3 or 4
thrombocytopenia was reported in 28% of the patients.
Neutropenia and thrombocytopenia
NDMM
Newly diagnosed multiple myeloma in: patients who have undergone ASCT treated with lenalidomide maintenance
The adverse reactions from CALGB 100104 included events reported post-high dose melphalan and ASCT
(HDM/ASCT) as well as events from the maintenance treatment period. A second analysis identified events that
occurred after the start of maintenance treatment. In IFM 2005-02, the adverse reactions were from the maintenance
treatment period only.
Overall, grade 4 neutropenia was observed at a higher frequency in the lenalidomide maintenance arms compared to
the placebo maintenance arms in the 2 studies evaluating lenalidomide maintenance in NDMM patients who have
undergone ASCT (32.1% vs 26.7% [16.1% vs 1.8% after the start of maintenance treatment] in CALGB 100104 and
16.4% vs 0.7% in IFM 2005-02, respectively). Treatment-emergent AEs of neutropenia leading to lenalidomide
discontinuation were reported in 2.2% of patients in CALGB 100104 and 2.4% of patients in IFM 2005-02,
respectively. Grade 4 febrile neutropenia was reported at similar frequencies in the lenalidomide maintenance arms
compared to placebo maintenance arms in both studies (0.4% vs 0.5% [0.4% vs 0.5% after the start of maintenance
treatment] in CALGB 100104 and 0.3% vs 0% in IFM 2005-02, respectively). Patients should be advised to promptly
report febrile episodes, a treatment interruption and/or dose reductions may be required (see section 4.2).
Grade 3 or 4 thrombocytopenia was observed at a higher frequency in the lenalidomide maintenance arms compared
to the placebo maintenance arms in studies evaluating lenalidomide maintenance in NDMM patients who have
undergone ASCT (37.5% vs 30.3% [17.9% vs 4.1% after the start of maintenance treatment] in CALGB 100104 and
13.0% vs 2.9% in IFM 2005-02, respectively). Patients and physicians are advised to be observant for signs and
symptoms of bleeding, including petechiae and epistaxes, especially in patients receiving concomitant medicinal
products susceptible to induce bleeding (see section 4.8, Haemorrhagic disorders).
…….
Myelodysplastic Syndromes patients
Patients taking REVLIMID for MDS should have their complete blood counts monitored weekly for the first 8 weeks
and at least monthly thereafter.
Grade 3 or 4 hematologic toxicity was seen in 80% of patients enrolled in the MDS study. In the 48% of patients who
developed Grade 3 or 4 neutropenia, the median time to onset was 42 days (range, 14-411 days), and the median
time to documented recovery was 17 days (range, 2-170 days). In the 54% of patients who developed Grade 3 or 4
thrombocytopenia, the median time to onset was 28 days (range, 8-290 days), and the median time to documented
recovery was 22 days (range, 5-224 days) [see Boxed Warning and Dosage and Administration (2.2)].
…….
Infection with or without Neutropenia
Patients with multiple myeloma are prone to develop infections including pneumonia. A higher rate of infections was
observed with lenalidomide in combination with dexamethasone than with MPT. in patients with NDMM who are not
eligible for transplant, and with lenalidomide maintenance compared to placebo in patients with NDMM who had
undergone ASCT.
…….
1.1
5.4 Venous and Arterial Thromboembolism
Venous thromboembolic events (deep
In patients with multiple myeloma, the combination of lenalidomide with dexamethasone is associated with an
increased risk of venous thromboembolism (predominantly deep vein thrombosis and pulmonary embolism) and
arterial was seen to a lesser extent with lenalidomide in combination with melphalan and prednisone.
In patients with multiple myeloma, myelodysplastic syndromes and mantle cell lymphoma, treatment with lenalidomide
monotherapy was associated with a lower risk of venous thromboembolism (predominantly deep vein thrombosis are
increasedand pulmonary embolism) than in patients with multiple myeloma treated with REVLIMID. A
significantlylenalidomide in combination therapy
In patients with multiple myeloma, the combination of lenalidomide with dexamethasone is associated with an
increased risk of DVT (7.4%) and of PE (3.7%) occurred in patients with multiple myeloma who were treated with
REVLIMID and dexamethasone therapy compared to patients treated in the placebo and dexamethasone group (3.1%
arterial thromboembolism (predominantly myocardial infarction and 0.9%) in a clinical trials with varying use of
anticoagulant therapies. [see Boxed Warning and Adverse Reactions (6.1)].
Venous thromboembolism cerebrovascular event) and was seen to a lesser extent with lenalidomide in combination
with melphalan and prednisone. The risk of ATE is lower in newly diagnosed patients with multiple myeloma
andtreated with lenalidomide monotherapy than in patients with multiple myeloma treated with lenalidomide in
myelodysplastic syndromes.
…….
Consequently, patients with known risk factors for thromboembolism – including prior thrombosis – should be closely
monitored. Action should be taken to try to minimize all modifiable risk factors (e.g. smoking, hypertension, and
hyperlipidaemia). Concomitant administration of erythropoietic agents or previous history of thromboembolic events
may also increase thrombotic risk in these patients. Therefore, erythropoietic agents, or other agents that may
increase the risk of thrombosis, such as hormone replacement therapy, should be used with caution in multiple
myeloma patients receiving lenalidomide with dexamethasone. A haemoglobin concentration above 12 g/dl should
lead to discontinuation of erythropoietic agents.
Patients and physicians are advised to be observant for the signs and symptoms of thromboembolism. Patients
should be instructed to seek medical care if they develop symptoms such as shortness of breath, chest pain, arm or
leg swelling. Prophylactic antithrombotic medicines should be recommended, especially in patients with additional
thrombotic risk factors. The decision to take antithrombotic prophylactic measures should be made after careful
assessment of an individual patient’s underlying risk factors.
If the patient experiences any thromboembolic events, treatment must be discontinued and standard anticoagulation
therapy started. Once the patient has been stabilised on the anticoagulation treatment and any complications of the
thromboembolic event have been managed, the lenalidomide treatment may be restarted at the original dose
dependent upon a benefit risk assessment. The patient should continue anticoagulation therapy during the course of
lenalidomide treatment.
…….
5.6 Second Primary Malignancies
An increase of second primary malignancies (SPM) has been observed in clinical trials in previously treated myeloma
patients receiving lenalidomide/dexamethasone (3.98 per 100 person-years) compared to controls (1.38 per 100
person-years). Non-invasive SPM comprise basal cell or squamous cell skin cancers. Most of the invasive SPMs were
solid tumour malignancies.
…….
The increased risk of secondary primary malignancies associated with lenalidomide is relevant also in the context of
NDMM after stem cell transplantation. Though this risk is not yet fully characterized, it should be kept in mind when
considering and using Revlimid in this setting.
The incidence rate of hematologic malignancies, most notably AML, MDS and B-cell malignancies (including
Hodgkin’s lymphoma), was 1.31 per 100 person-years for the lenalidomide arms and 0.58 per 100 person-years for
the placebo arms (1.02 per 100 person-years for patients exposed to lenalidomide after ASCT and 0.60 per 100
person-years for patients not-exposed to lenalidomide after ASCT). The incidence rate of solid tumour SPMs was 1.36
per 100 person-years for the lenalidomide arms and 1.05 per 100 person-years for the placebo arms (1.26 per 100
person-years for patients exposed to lenalidomide after ASCT and 0.60 per 100 person-years for patients not-exposed
to lenalidomide after ASCT).
The risk of occurrence of hematologic SPM must be taken into account before initiating treatment with lenalidomide
either in combination with melphalan or immediately following high-dose melphalan and ASCT. Physicians should
carefully evaluate patients before and during treatment using standard cancer screening for occurrence of SPM and
institute treatment as indicated.
…….
5.8 Allergic Reactionsand severe skin reactions
…….
Severe cutaneous reactions including SJS, and TEN and DRESS have been reported with the use of lenalidomide.
Patients should be advised of the signs and symptoms of these reactions by their prescribers and should be told to
seek medical attention immediately if they develop these symptoms. Lenalidomide must be discontinued for exfoliative
or bullous rash, or if SJS, TEN or DRESS is suspected, and should not be resumed following discontinuation for these
reactions. Interruption or discontinuation of lenalidomide should be considered for other forms of skin reaction
depending on severity. Patients with a history of severe rash associated with thalidomide treatment should not receive
lenalidomide.
…….
Peripheral neuropathy
Lenalidomide is structurally related to thalidomide, which is known to induce severe peripheral neuropathy. There was
no increase in peripheral neuropathy observed with long term use of lenalidomide for the treatment of newly
diagnosed multiple myeloma.
…….
6.1 Clinical Trials Experience in Multiple Myeloma
…….
Tabulated summary for monotherapy in MM
…….
Table 1. ADRs reported in clinical trials in patients with multiple myeloma treated with lenalidomide
maintenance therapy
System Organ Class/Preferred
Term
All ADRs/Frequency
Grade 3-4 ADRs/Frequency
Infections and Infestations
Very Common
Pneumonias
◊, a
, Upper respiratory
tract infection, Neutropenic infection,
Bronchitis
, Influenza
Gastroenteritis
, Sinusitis,
Nasopharyngitis, Rhinitis
Common
Infection
, Urinary tract infection
Lower respiratory tract infection, Lung
infection
Very Common
Pneumonias
◊, a
, Neutropenic infection
Common
Sepsis
◊, b
, Bacteraemia, Lung
infection
, Lower respiratory tract
infection bacterial, Bronchitis
Influenza
, Gastroenteritis
, Herpes
zoster
, Infection
Neoplasms Benign, Malignant and
Unspecified (incl cysts and
polyps)
Common
Myelodysplastic syndrome
Blood and Lymphatic System
Disorders
Very Common
Neutropenia
, Febrile neutropenia
Thrombocytopenia
, Anemia,
Leucopenia
, Lymphopenia
Very Common
Neutropenia
, Febrile neutropenia
Thrombocytopenia
, Anemia,
Leucopenia
, Lymphopenia
Common
Pancytopenia
Metabolism and Nutrition
Disorders
Very Common
Hypokalaemia
Common
Hypokalaemia, Dehydration
Nervous System Disorders
Very Common
Paraesthesia
Common
Common
Headache
System Organ Class/Preferred
Term
All ADRs/Frequency
Grade 3-4 ADRs/Frequency
Peripheral neuropathy
Vascular Disorders
Common
Pulmonary embolism
Common
Deep vein thrombosis
,◊,d
Respiratory, Thoracic and
Mediastinal Disorders
Very Common
Cough
Common
Dyspnoea
, Rhinorrhoea
Common
Dyspnoea
Gastrointestinal Disorders
Very Common
Diarrhoea, Constipation, Abdominal
pain, Nausea
Common
Vomiting, Abdominal pain upper
Common
Diarrhoea, Vomiting, Nausea
Hepatobiliary Disorders
Very Common
Abnormal liver function tests
Common
Abnormal liver function tests
Skin and Subcutaneous Tissue
Disorders
Very Common
Rash, Dry skin
Common
Rash, Pruritus
Musculoskeletal and Connective
Tissue Disorders
Very Common
Muscle spasms
Common
Myalgia, Musculoskeletal pain
General Disorders and
Administration Site Conditions
Very Common
Fatigue, Asthenia, Pyrexia
Common
Fatigue, Asthenia
Adverse reactions reported as serious in clinical trials in patients with NDMM who had undergone ASCT
Applies to serious adverse drug reactions only
“Pneumonias” combined AE term includes the following PTs: Bronchopneumonia, Lobar pneumonia, Pneumocystis
jiroveci pneumonia, Pneumonia, Pneumonia klebsiella, Pneumonia legionella, Pneumonia mycoplasmal, Pneumonia
pneumococcal, Pneumonia streptococcal, Pneumonia viral, Lung disorder, Pneumonitis
“Sepsis” combined AE term includes the following PTs: Bacterial sepsis, Pneumococcal sepsis, Septic shock,
Staphylococcal sepsis
“Peripheral neuropathy” combined AE term includes the following preferred terms (PTs): Neuropathy peripheral,
Peripheral sensory neuropathy, Polyneuropathy
“Deep vein thrombosis” combined AE term includes the following PTs: Deep vein thrombosis, Thrombosis, Venous
thrombosis
Tabulated summary for combination therapy in MM
…….
Table 12: ADRs reported in clinical studies in patients with multiple myeloma treated with lenalidomide in
combination with dexamethasone, or with melphalan and prednisone
System Organ
Class
/ Preferred Term
All ADRs/Frequency
Grade 3−4 ADRs/Frequency
Infections
and Infestations
Very Common
Pneumonia
, Upper respiratory tract infection
Bacterial, viral and fungal infections (including
opportunistic infections)
, Nasopharyngitis,
Pharyngitis, Bronchitis
Common
Sepsis
, Sinusitis
Common
Pneumonia
, Bacterial, viral and
fungal infections (including
opportunistic infections
Cellulitis
, Sepsis
, Bronchitis
Metabolism and
Nutrition Disorders
Very Common
Hypokalaemia
, Hyperglycaemia,
Hypocalcaemia
, Decreased appetite, Weight
decreased
Common
Hypomagnesaemia, Hyperuricaemia,
Dehydration
, Hypercalcaemia
Common
Hypokalaemia
, Hyperglycaemia
Hypocalcaemia, Diabetes
mellitus
, Hypophosphataemia,
Hyponatraemia
, Hyperuricaemia,
Gout, Decreased appetite,
Weight decreased
System Organ
Class
/ Preferred Term
All ADRs/Frequency
Grade 3−4 ADRs/Frequency
Musculoskeletal
and Connective
Tissue Disorders
Very Common
Muscle spasms, Bone pain
Musculoskeletal and connective tissue pain and
discomfort (including back pain
),, Arthralgia
Common
Muscular weakness, Joint swelling, Myalgia
Common
Muscular weakness, Bone pain
Musculoskeletal and connective
tissue pain and discomfort
(including back pain
Uncommon
Joint swelling
◊ Adverse reactions reported as serious in clinical trials in patients with multiple myeloma treated with lenalidomide in
combination with dexamethasone, or with melphalan and prednisone
+ Applies to serious adverse drug reactions only
* Squamous skin cancer was reported in clinical trials in previously treated myeloma patients with
lenalidomide/dexamethasone compared to controls
** Squamous cell carcinoma of skin was reported in a clinical trial in newly diagnosed myeloma patients with
lenalidomide/dexamethasone compared to controls
…….
6.3 Clinical Trials Experience in Mantle Cell Lymphoma
…….
Table 5:
ADRs reported in clinical trials in patients with mantle cell lymphoma treated with lenalidomide
System Organ
Class
/ Preferred Term
All ADRs/Frequency
Grade 3−4 ADRs/Frequency
Cardiac
Disorders
Common
Acute mMyocardial infarction (including
acute)
, Cardiac failure
Adverse events reported as serious in mantle cell lymphoma clinical trials
…….
Table 56: ADRs reported in post-marketing use in patients treated with lenalidomide
System Organ Class
/ Preferred Term
All ADRs/Frequency
Grade 3−4 ADRs/Frequency
Immune System
Disorders
Not Known
Solid organ transplant rejection
Skin and
Subcutaneous Tissue
Disorders
Uncommon
Angioedema
Rare
Stevens-Johnson Syndrome, Toxic
epidermal necrolysis
Not Known
Leukocytoclastic vasculitis, Drug
Reaction with Eosinophilia and
Systemic Symptoms
…….
7.
USE IN SPECIFIC POPULATIONS
8.1 Pregnancy
Pregnancy Category X [see Boxed Warnings and Contraindications (4.1)] exposure registry
There is a pregnancy exposure registry that monitors pregnancy outcomes in females exposed to REVLIMID during
pregnancy as well as female partners of male patients who are exposed to REVLIMID. This registry is also used to
understand the root cause for the pregnancy.
…….
The estimated background risk of major birth defects and miscarriage for the indicated population is unknown.
…….
Data
Animal data
…….
Following daily oral administration of lenalidomide from Gestation Day 7 through Gestation Day 20 in pregnant rabbits,
fetal plasma lenalidomide concentrations were approximately 20-40% of the maternal Cmax. Following a single oral
dose to pregnant rats, lenalidomide was detected in fetal plasma and tissues; concentrations of radioactivity in fetal
tissues were generally lower than those in maternal tissues. These data indicated that lenalidomide crossed the
placenta.
8.2 Nursing mothers Lactation
It is not known whether this drug is excreted in human milk.Risk summary
There is no information regarding the presence of lenalidomide in human milk, the effects of REVLIMID on the
breastfed infant, or the effects of REVLIMID on milk production. Because many drugs are excreted in human milk and
because of the potential for adverse reactions in nursing infants from lenalidomide, a decision should be made
whether to discontinue nursing or to discontinue the drug, taking into account the importance of the drug to the mother
advice women not to breastfeed during treatment with REVLIMID.
8.3 Females and males of reproductive potential
Pregnancy Testing
REVLIMID can cause fetal harm when administered during pregnancy [see Use in Specific Populations (8.1)]. Verify
the pregnancy status of females of reproductive potential prior to initiating REVLIMID therapy and during therapy.
Advise females of reproductive potential that they must avoid pregnancy 4 weeks before therapy, while taking
REVLIMID, during dose interruptions and for at least 4 weeks after completing therapy.
Females of reproductive potential must have 2 negative pregnancy tests before initiating REVLIMID. The first test
should be performed within 10-14 days, and the second test within 24 hours prior to prescribing REVLIMID. Once
treatment has started and during dose interruptions, pregnancy testing for females of reproductive potential should
occur weekly during the first 4 weeks of use, then pregnancy testing should be repeated every 4 weeks in females
with regular menstrual cycles. If menstrual cycles are irregular, the pregnancy testing should occur every 2 weeks.
Pregnancy testing and counseling should be performed if a patient misses her period or if there is any abnormality in
her menstrual bleeding. REVLIMID treatment must be discontinued during this evaluation.
Contraception
Females
Females of reproductive potential must commit either to abstain continuously from heterosexual sexual intercourse or
to use 2 methods of reliable birth control simultaneously: one highly effective form of contraception – tubal ligation,
IUD, hormonal (birth control pills, injections, hormonal patches, vaginal rings, or implants), or partner’s vasectomy,
and 1 additional effective contraceptive method – male latex or synthetic condom, diaphragm, or cervical cap.
Contraception must begin 4 weeks prior to initiating treatment with REVLIMID, during therapy, during dose
interruptions, and continuing for 4 weeks following discontinuation of REVLIMID
therapy. Reliable contraception is indicated even where there has been a history of infertility, unless due to
hysterectomy. Females of reproductive potential should be referred to a qualified provider of contraceptive methods, if
needed.
1.1
Males
Lenalidomide is present in the semen of males who take REVLIMID. Therefore, males must always use a latex or
synthetic condom during any sexual contact with females of reproductive potential while taking REVLIMID and for up
to 4 weeks after discontinuing REVLIMID, even if they have undergone a successful vasectomy.
Male patients taking REVLIMID must not donate sperm.
1.2
8.4 Pediatric use
Revlimid should not be used in children and adolescents from birth to less than 18 years because of safety concerns.
Safety and effectiveness have not been established in pediatric patients.
8.45 Geriatric use
REVLIMID has been usedMM in multiple myeloma (MM) clinical trials in combination: Overall, of the 1613 patients up
to 91 years of age. in the NDMM study
Newly diagnosed multiple myeloma
In patients with newly diagnosed multiple myeloma aged 75 years and older who received lenalidomide, there was a
higher incidence of serious adverse reactions and adverse reactions that led to study treatment discontinuation
Patients with newly diagnosed multiple myeloma aged, 94% (1521 /1613) were 65 years of age or older, while 35%
(561/1613) were over 75 years of age. The percentage of patients over age 75 years and older should be carefully
assessed beforewas similar between study arms (Rd Continuous: 33%; Rd18: 34%; MPT: 33%). Overall, across all
treatment is considered.
In clinical trials of newly diagnosed multiple myelomaarms, the frequency in transplant non eligible patients,
lenalidomide combined therapymost of the AE categories (eg, all AEs, grade 3/4 AEs, serious AEs) was less tolerated
higher in patients older (> 75 years of age) than in younger (≤ 75 years of age compared to the younger population.
These patients discontinued at a higher rate due to intolerance () subjects. Grade 3 or 4 adverse events and serious
adverse events), when compared to patients < 75years.AEs in the General Disorders and Administration Site
Conditions body system were consistently reported at a higher
Multiple myeloma withfrequency (with a difference of at least 5%) in older subjects than in younger subjects across all
treatment arms. Grade 3 or 4 TEAEs in the Infections and Infestations, Cardiac Disorders (including cardiac failure
and congestive cardiac failure), Skin and Subcutaneous Tissue Disorders, and Renal and Urinary Disorders (including
renal failure) body systems were also reported slightly, but consistently, more frequently (<5% difference), in older
subjects than in younger subjects across all treatment arms. For other body systems (e.g., Blood and Lymphatic
System Disorders, Infections and Infestations, Cardiac Disorders, Vascular Disorders), there was a less consistent
trend for increased frequency of grade 3/4 AEs in older vs younger subjects across all treatment arms Serious AEs
were generally reported at a higher frequency in the older subjects than in the younger subjects across all treatment
arms.
MM maintenance therapy: Overall, 10% (106/1018) of patients were 65 years of age or older, while no patients were
over 75 years of age. Grade 3 or 4 AEs were higher in the REVLIMID arm (more than 5% higher) in the patients 65
years of age or older versus younger patients. The frequency of Grade 3 or 4 AEs in the Blood and Lymphatic System
Disorders were higher in the REVLIMID arm (more than 5% higher) in the patients 65 years of age or older versus
younger patients. There were not a sufficient number of patients 65 years of age or older in REVLIMID maintenance
studies who experienced either a serious AE, or discontinued therapy due to an AE to determine whether elderly
patients respond relative to safety differently from younger patients.
MM after at least one prior therapy:
…….
Of the 134 patients with MCL enrolled in the MCL trial, 63% were age 65 and over, while 22% of patients were age 75
and over. The overall frequency of adverse events was similar in patients over 65 years of age and in younger
patients (98% vs. 100%). The overall incidence of grade 3 and 4 adverse events was also similar in these 2 patient
groups (79% vs. 78%, respectively). The frequency of serious adverse events was higher in patients over 65 years of
age than in younger patients (55% vs. 41%). No differences in efficacy were observed between patients over 65 years
of age and younger patients.
…….
אפורל ןולעה ב"צמ יפכ
רשואש
לע
ידי
דרשמ
תואירבה
.ילארשיה
ןולעה
אפורל
דרשמל חלשנ
תואירבה
ךרוצל
תאלעה
תופורתה רגאמל
רתאבש
דרשמ
תואירבה
ולבקל ןתינו
ספדומ
לע
ידי
היינפ
םרפואינ :םושירה לעבל
מ"עב
חולישה 'חר
.ד.ת ,
7063
חתפ
,הווקת
:לט
03-9373737
,הכרבב
ךלוו זוע
ו היצלוגר להנמ הנוממ חקור
םרפואינ
קיפיטנייס
מ"עב
FULL PRESCRIBING INFORMATION
NAME OF THE MEDICINAL PRODUCT
Revlimid 2.5 mg, hard capsules
Revlimid 5 mg, hard capsules
Revlimid 7.5 mg, hard capsules
Revlimid 10 mg, hard capsules
Revlimid 15 mg, hard capsules
Revlimid 20 mg, hard capsules
Revlimid 25 mg, hard capsules
QUALITATIVE AND QUANTITATIVE COMPOSITION
Revlimid 2.5 mg hard capsules
Each capsule contains 2.5 mg of lenalidomide.
Excipient(s) with known effect
Each capsule contains 73.5 mg of lactose (as anhydrous lactose).
Revlimid 5 mg hard capsules
Each capsule contains 5 mg of lenalidomide.
Excipient(s) with known effect
Each capsule contains 147 mg of lactose (as anhydrous lactose).
Revlimid 7.5 mg hard capsules
Each capsule contains 7.5 mg of lenalidomide.
Excipient(s) with known effect
Each capsule contains 144.5 mg of lactose (as anhydrous lactose).
Revlimid 10 mg hard capsules
Each capsule contains 10 mg of lenalidomide.
Excipient(s) with known effect
Each capsule contains 294 mg of lactose (as anhydrous lactose).
Revlimid 15 mg hard capsules
Each capsule contains 15 mg of lenalidomide.
Excipient(s) with known effect
Each capsule contains 289 mg of lactose (as anhydrous lactose).
Revlimid 20 mg hard capsules
Each capsule contains 20 mg of lenalidomide.
Excipient(s) with known effect
Each capsule contains 244.5 mg of lactose (as anhydrous lactose).
Revlimid 25 mg hard capsules
Each capsule contains 25 mg of lenalidomide.
Excipient(s) with known effect
Each capsule contains 200 mg of lactose (as anhydrous lactose).
PHARMACEUTICAL FORM
Please refer to section 3.
Because of the embryo-fetal risk, the marketing of Revlimid is subject to a risk management plan
(RMP), including the following:
Prescribers guide. Prescribers must be certified with the REVLIMID RMP program by
enrolling and complying with the RMP requirements.
Patients brochure. patient must sign a Patient-Physician agreement form and comply with the
RMP requirements. In particular, female patients of reproductive potential who are not pregnant
must comply with the pregnancy testing and contraception and males must comply with
contraception requirements.
Pharmacies must be certified with the REVLIMID RMP program, must only dispense to
patients who are authorized to receive REVLIMID and comply with RMP requirements.
Please ensure you are familiar with this important information and explain to the patient the need
to review the brochures before starting treatment.
WARNING: EMBRYO FETAL TOXICITY, HEMATOLOGIC TOXICITY, and
VENOUS and ARTERIAL THROMBOEMBOLISM
Do not use REVLIMID during pregnancy. Lenalidomide, a thalidomide analogue, caused
limb abnormalities in a developmental monkey study. Thalidomide is a known human
teratogen that causes severe life-threatening human birth defects. If lenalidomide is used
during pregnancy, it may cause birth defects or death to a developing baby. In women of
childbearing potential, obtain a negative pregnancy test before starting REVLIMID
treatment. Women of childbearing potential must use 2 reliable forms of contraception
simultaneously or continuously abstain from heterosexual sex 4 weeks before starting
treatment, during (including dose interruptions) and for 4 weeks following discontinuation
of REVLIMID treatment [see Warnings and Precautions (5.1 )]].
Hematologic Toxicity (Neutropenia and Thrombocytopenia
REVLIMID can cause significant neutropenia and thrombocytopenia. Eighty percent of
patients with del 5q myelodysplastic syndromes had to have a dose delay/reduction during
the major study. Thirty-four percent of patients had to have a second dose delay/reduction.
Grade 3 or 4 hematologic toxicity was seen in 80% of patients enrolled in the study.
Patients on therapy for del 5q myelodysplastic syndromes should have their complete
blood counts monitored weekly for the first 8 weeks of therapy and at least monthly
thereafter. Patients may require dose interruption and/or reduction. Patients may require
use of blood product support and/or growth factors [see Dosage and Administration (2.2)].
Venous and Arterial Thromboembolism
REVLIMID has demonstrated a significantly increased risk of deep vein thrombosis (DVT)
and pulmonary embolism (PE), as well as risk of myocardial infarction, and stroke in
patients with multiple myeloma who were treated with REVLIMID and dexamethasone
therapy. Monitor for and advise patients about signs and symptoms of thromboembolism.
Advise patients to seek immediate medical care if they develop symptoms such as shortness
of breath, chest pain, or arm or leg swelling. Thromboprophylasix is recommended and
the choice of regimen should be based on an assessment of an individual patient’s
underlying risk factors [see Warnings and Precautions (5.4)].
1.
INDICATIONS AND USAGE
1.1
Multiple Myeloma
Revlimid is indicated for:
The maintenance treatment of adult patients with newly diagnosed multiple myeloma (MM) who have
undergone autologous stem cell transplantation.
previously untreated multiple myeloma in adult patients who are not eligible for transplant.
in combination with dexamethasone treatment of multiple myeloma patients who have
received at least one prior therapy.
1.2
Myelodysplastic Syndromes
REVLIMID is indicated for patients with transfusion-dependent anemia due to low- or
intermediate-1-risk myelodysplastic syndromes (MDS) associated with a deletion 5q cytogenetic
abnormality with or without additional cytogenetic abnormalities.
Revlimid 7.5 mg is not indicated for treatment in MDS.
1.3
Mantle Cell Lymphoma
REVLIMID is indicated for the treatment of adult patients with relapsed and/or refractory mantle
cell lymphoma (MCL).
2.
DOSAGE AND ADMINISTRATION
REVLIMID should be taken orally at about the same time each day, either with or without food.
REVLIMID hard capsules should be swallowed whole with water. The hard capsules should not
be opened, broken, or chewed.
2.1 Newly Diagnosed Multiple Myeloma
(NDMM)
Lenalidomide maintenance in patients who have undergone autologous stem cell transplantation
(ASCT)
Lenalidomide maintenance should be initiated after adequate haematologic recovery following
ASCT in patients without evidence of progression. Lenalidomide must not be started if the
Absolute Neutrophil Count (ANC) is < 1.0 x 10
/L, and/or platelet counts are < 75 x 10
Recommended dose
The recommended starting dose is lenalidomide 10 mg orally once daily continuously (on days 1
to 28 of repeated 28-day cycles) given until disease progression or intolerance. After 3 cycles of
lenalidomide maintenance, the dose can be increased to 15 mg orally once daily if tolerated.
Dose reduction steps
Starting dose (10 mg)
If dose increased (15 mg)
Dose level -1
5 mg
10 mg
Dose level -2
5 mg (days 1-21 every 28 days)
5 mg
Dose level -3
Not applicable
5 mg (days 1-21 every 28 days)
Do not dose below 5 mg (days 1-21 every 28 days)
After 3 cycles of lenalidomide maintenance, the dose can be increased to 15 mg orally once daily if tolerated.
Thrombocytopenia
When platelets
Recommended course
Fall to < 30 x 10
Interrupt lenalidomide treatment
Return to ≥ 30 x 10
Resume lenalidomide at dose level -1 once
daily
For each subsequent drop below 30 x 10
Interrupt lenalidomide treatment
Return to ≥ 30 x 10
Resume lenalidomide at next lower dose
level once daily
Neutropenia
When neutrophils
Recommended course
Fall to < 0.5 x 10
Interrupt lenalidomide treatment
Return to ≥ 0.5 x 10
Resume lenalidomide at dose level -1 once
daily
For each subsequent drop below < 0.5 x 10
Interrupt lenalidomide treatment
Return to ≥ 0.5 x 10
Resume lenalidomide at next lower dose
level once daily
At the physician’s discretion, if neutropenia is the only toxicity at any dose level, add granulocyte colony stimulating factor (G-CSF) and
maintain the dose level of lenalidomide.
Lenalidomide in combination with dexamethasone until disease progression in
patients who are
not eligible for transplant
Lenalidomide treatment must not be started if the ANC is < 1.0 x 10
/L, and/or platelet counts are
< 50 x 10
Recommended dose
The recommended starting dose of lenalidomide is 25 mg orally once daily on days 1-21 of
repeated 28-day cycles. The recommended dose of dexamethasone is 40 mg orally once daily on
days 1, 8, 15 and 22 of repeated 28-day cycles. Patients may continue lenalidomide and
dexamethasone therapy until disease progression or intolerance.
Dosing is continued or modified based upon clinical and laboratory findings. For patients ≥75
years of age, the starting dose of dexamethasone is 20 mg/day on days 1, 8, 15 and 22 of each 28-
day treatment cycle. The recommended dose of lenalidomide for patients suffering from
moderate renal impairment is 10 mg once daily.
Recommended dose adjustments during treatment and restart of treatment:
Dose adjustments, as summarised below, are recommended to manage grade 3 or 4
thrombocytopenia, neutropenia, or other grade 3 or 4 toxicity judged to be related to
lenalidomide.
Dose reduction steps
Lenalidomide
Dexamethasone
Starting dose
25 mg
40 mg
Dose level -1
20 mg
20 mg
Dose level -2
15 mg
12 mg
Dose level -3
10 mg
8 mg
Dose level- 4
5 mg
4 mg
Dose level -5
2.5 mg
Thrombocytopenia
When platelets
Recommended course
Fall to < 25 x 10
Stop lenalidomide dosing for
remainder of cycleª
Return to ≥ 50 x 10
Decrease by one dose level when
dosing resumed at next cycle
If Dose Limiting Toxicity (DLT) occurs on > day15 of a cycle, lenalidomide dosing will be interrupted for at least the remainder of the current
28-day cycle.
Neutropenia
When neutrophils
Recommended course
First fall to < 0.5 x 10
Interrupt lenalidomide treatment
Return to ≥ 1 x 10
/L when neutropenia is
the only observed toxicity
Resume lenalidomide at Starting dose
once daily
Return to ≥ 0.5 x 10
/L when dose-
dependent haematological toxicities other
than neutropenia are observed
Resume lenalidomide at Dose level -1
once daily
For each subsequent drop below
< 0.5 x 10
Interrupt lenalidomide treatment
Return to ≥ 0.5 x 10
Resume lenalidomide at next lower
dose level once daily.
In case of neutropenia, the use of growth factors in patient management should be considered.
If the dose of lenalidomide was reduced for a hematologic DLT, the dose of lenalidomide may
be re-introduced to the next higher dose level (up to the starting dose) at the discretion of the
treating physician if continued lenalidomide / dexamethasone therapy resulted in improved bone
marrow function (no DLT for at least 2 consecutive cycles and an ANC ≥1,5 x 10
/L with a
platelet count ≥ 100,
x 10
/L at the beginning of a new cycle at the current dose level).
Lenalidomide in combination with melphalan and prednisone followed by maintenance
monotherapy in patients who are
not eligible for transplant
Lenalidomide treatment must not be started if the ANC is < 1.5 x 10
/L, and/or platelet counts
are < 75 x 10
Recommended dose
The recommended starting dose is lenalidomide
mg/day orally on days 1-21 of repeated
28-day cycles for up to 9 cycles, melphalan 0.18 mg/kg orally on days 1-4 of repeated 28
cycles, prednisone 2 mg/kg orally on days 1-4 of repeated 28-day cycles. Patients who
complete
cycles or who are unable to complete the combination therapy due
intolerance
are treated with lenalidomide alone, 10 mg/day orally on days 1-21 of repeated
28-day
cycles
given until disease
progression.
Dosing is continued or modified based upon clinical and
laboratory findings.
Recommended dose adjustments during treatment and restart of treatment:
Dose adjustments, as summarised below, are recommended to manage grade 3 or 4
thrombocytopenia, neutropenia, or other grade 3 or 4 toxicity judged to be related to
lenalidomide.
Dose reduction steps
Lenalidomide
Melphalan
Prednisone
Starting dose
10 mgª
0.18 mg/kg
2 mg/kg
Dose level -1
7.5 mg
0.14 mg/kg
1 mg/kg
Dose level -2
5 mg
0.10 mg/kg
0.5 mg/kg
Dose level -3
2.5 mg
0.25 mg/kg
If neutropenia is the only toxicity at any dose level, add granulocyte colony stimulating factor (G-CSF) and maintain the dose level of
lenalidomide
Thrombocytopenia
When platelets
Recommended course
First fall to < 25 x 10
Interrupt lenalidomide treatment
Return to ≥ 25 x 10
Resume lenalidomide and melphalan
at Dose level -1
For each subsequent drop below
30 x 10
Interrupt lenalidomide treatment
Return to ≥ 30 x 10
Resume lenalidomide at next lower
dose level (Dose level -2 or -3) once
daily.
Neutropenia
When neutrophils
Recommended course
First fall to < 0.5 x 10
/Lª
Interrupt lenalidomide treatment
Return to ≥ 0.5 x 10
/L when neutropenia
is the only observed toxicity
Resume lenalidomide at Starting dose
once daily
Return to ≥ 0.5 x 10
/L when dose-
dependent haematological toxicities other
than neutropenia are observed
Resume lenalidomide at Dose level -1
once daily
For each subsequent drop below
< 0.5 x 10
Interrupt lenalidomide treatment
Return to ≥ 0.5 x 10
Resume lenalidomide at next lower
dose level once daily.
ªIf the subject has not been receiving G-CSF therapy, initiate G-CSF therapy. On Day 1 of next cycle, continue G-CSF as needed and maintain
dose of melphalan if neutropenia was the only DLT. Otherwise, decrease by one dose level at start of next cycle.
In case of neutropenia, the use of growth factors in patient management should be considered.
For patients older than 75 years of age treated with lenalidomide in combination with
dexamethasone, the starting dose of dexamethasone is 20 mg/day on days 1, 8, 15 and 22 of each
28-day treatment cycle.
No dose adjustment is proposed for patients older than 75 years treated with lenalidomide in
combination with melphalan and prednisone.
2.2
Multiple Myeloma with at least one prior therapy
The recommended starting dose of REVLIMID is 25 mg once daily on days 1-21 of repeated 28-
day cycles. The recommended dose of dexamethasone is 40 mg once daily on days 1-4, 9-12,
and 17-20 of each 28-day cycle for the first 4 cycles of therapy and then 40 mg once daily orally
on days 1-4 every 28 days. Treatment is continued or modified based upon clinical and
laboratory findings.
Prescribing physicians should carefully evaluate which dose of dexamethasone to use, taking
into account the condition and disease status of the patient.
Lenalidomide treatment must not be started if the ANC < 1,000/mcL, and/or platelet counts <
75,000/ mcL or, dependent on bone marrow infiltration by plasma cells, platelet counts <
30,000/mcL.
Dose Adjustments for Hematologic Toxicities During Multiple Myeloma Treatment
Dose modification guidelines, as summarized below, are recommended to manage Grade 3 or 4
neutropenia or thrombocytopenia or other Grade 3 or 4 toxicity judged to be related to
REVLIMID.
Platelet counts
Thrombocytopenia in MM
When Platelets
Recommended Course
Fall to <30,000/mcL
Interrupt REVLIMID
treatment, follow CBC weekly
Return to ≥30,000/mcL
Restart REVLIMID at 15 mg
daily
For each subsequent drop <30,000/mcL
Interrupt REVLIMID
treatment
Return to ≥30,000/mcL
Resume REVLIMID at 5 mg
less than the previous dose. Do
not dose below 5 mg daily
Absolute Neutrophil counts (ANC)
Neutropenia in MM
When Neutrophils
Recommended Course
Fall to <1000/mcL
Interrupt REVLIMID
treatment, add G-CSF, follow
CBC weekly
Return to ≥1,000/mcL and neutropenia is the only
toxicity
Resume REVLIMID at 25
mg daily
Return to ≥1,000/mcL and if other toxicity
Resume REVLIMID at 15
mg daily
For each subsequent drop <1,000/mcL
Interrupt REVLIMID
treatment
Return to ≥1,000/mcL
Resume REVLIMID at 5 mg
less than the previous dose.
Do not dose below 5 mg daily
In case of neutropenia, the physician should consider the use of growth factors in patient
management.
Starting Dose Adjustment for Renal Impairment in MM:
See Section 2.5
2.3
Myelodysplastic Syndromes
The recommended starting dose of REVLIMID is 10 mg daily. Treatment is continued or
modified based upon clinical and laboratory findings.
Dose Adjustments for Hematologic Toxicities During MDS Treatment
Patients who are dosed initially at 10 mg and who experience thrombocytopenia should have
their dosage adjusted as follows:
Platelet counts
If thrombocytopenia develops WITHIN 4 weeks of starting treatment at 10 mg daily in
MDS
If baseline ≥100,000/mcL
When Platelets
Recommended Course
Fall to <50,000/mcL
Interrupt REVLIMID treatment
Return to ≥50,000/mcL
Resume REVLIMID at 5 mg daily
If baseline <100,000/mcL
When Platelets
Recommended Course
Fall to 50% of the baseline value
Interrupt REVLIMID treatment
If baseline ≥60,000/mcL and
returns to ≥50,000/mcL
Resume REVLIMID at 5 mg daily
If baseline <60,000/mcL and
returns to ≥30,000/mcL
Resume REVLIMID at 5 mg daily
If thrombocytopenia develops AFTER 4 weeks of starting treatment at 10 mg daily in MDS
When Platelets
Recommended Course
<30,000/mcL or <50,000/mcL
with platelet transfusions
Interrupt REVLIMID treatment
Return to ≥30,000/mcL
(without hemostatic failure)
Resume REVLIMID at 5 mg daily
Patients who experience thrombocytopenia at 5 mg daily should have their dosage adjusted as
follows:
If thrombocytopenia develops during treatment at 5 mg daily in MDS
When Platelets
Recommended Course
<30,000/mcL or <50,000/mcL
with platelet transfusions
Interrupt REVLIMID treatment
Return to ≥30,000/mcL
(without hemostatic failure)
Resume REVLIMID at 2.5 mg
daily
Patients who are dosed initially at 10 mg and experience neutropenia should have their dosage
adjusted as follows:
Absolute Neutrophil counts (ANC)
If neutropenia develops WITHIN 4 weeks of starting treatment at 10 mg daily in MDS
If baseline ANC ≥1,000/mcL
When Neutrophils
Recommended Course
Fall to <750/mcL
Interrupt REVLIMID treatment
Return to ≥1,000/mcL
Resume REVLIMID at 5 mg
daily
If baseline ANC <1,000/mcL
When Neutrophils
Recommended Course
Fall to <500/mcL
Interrupt REVLIMID treatment
Return to ≥500/mcL
Resume REVLIMID at 5 mg
daily
If neutropenia develops AFTER 4 weeks of starting treatment at 10 mg daily in MDS
When Neutrophils
Recommended Course
<500/mcL for ≥7 days or <500/mcL
associated with fever (≥38.5°C)
Interrupt REVLIMID treatment
Return to ≥500/mcL
Resume REVLIMID at 5 mg
daily
Patients who experience neutropenia at 5 mg daily should have their dosage adjusted as follows:
If neutropenia develops during treatment at 5 mg daily in MDS
When Neutrophils
Recommended Course
<500/mcL for ≥7 days or <500/mcL
associated with fever (≥38.5°C)
Interrupt REVLIMID
treatment
Return to ≥500/mcL
Resume REVLIMID at 2.5
mg daily
Discontinuation of lenalidomide
Patients without at least a minor erythroid response within 4 months of therapy initiation,
demonstrated by at least a 50% reduction in transfusion requirements or, if not transfused, a
1g/dl rise in haemoglobin, should discontinue lenalidomide treatment.
Starting Dose Adjustment for Renal Impairment in MDS:
See Section 2.5
2.4
Mantle Cell Lymphoma
The recommended starting dose of REVLIMID is 25 mg/day orally on days 1-21 of repeated
28-day cycles.
Dosing is continued or modified based upon clinical and laboratory findings.
Dose reduction steps
Starting dose
25 mg once daily on days 1-21, every 28 days
Dose Level -1
20 mg once daily on days 1-21, every 28 days
Dose Level -2
15 mg once daily on days 1-21, every 28 days
Dose Level -3
10 mg once daily on days 1-21, every 28 days
Dose Level -4
5 mg once daily on days 1-21, every 28 days
Dose Level -5
2.5 mg once daily on days 1-21, every 28 days
5 mg every other day on days 1-21, every 28 days
Thrombocytopenia
When Platelets
Recommended Course
Fall to <50 x 10
Interrupt REVLIMID
treatment and conduct
Complete Blood Count (CBC)
at least every 7 days
Return to ≥ 60 x 10
Resume lenalidomide at next
lower level (Dose Level -1)
For each subsequent drop below 50 x 10
Return to ≥60 x 10
Interrupt REVLIMID
treatment and conduct the
CBC at least every 7 days
Resume REVLIMID at next
lower level (Dose Level -2, -3,
-4 or -5). Do not dose below
Dose Level -5
Neutropenia
When Neutrophils
Recommended Course
Fall to <1 x 10
for at least 7 days
Falls to <1 x 10
/L with an associated fever (body
temperature ≥ 38.5°C)
Falls to <0.5 x 10
Interrupt REVLIMID
treatment and conduct the
CBC at least every 7 days
Return to ≥1 x 10
Resume REVLIMID at next
lower dose level (Dose Level
– 1)
For each subsequent drop below 1 x 10
/L for at least
7 days or drop to < 1 x 10
/L with associated fever
(body temperature ≥ 38.5°C) or drop to < 0.5 x 10
Returns to ≥1 x 10
Interrupt REVLIMID
treatment
Resume REVLIMID at next
lower dose level (Dose Level
-2, -3, -4, -5). Do not dose
below Dose Level -5
Tumour flare reaction
REVLIMID may be continued in patients with Grade 1 or 2 tumour flare reaction (TFR) without
interruption or modification, at the physician’s discretion. In patients with Grade 3 or 4 TFR,
withhold treatment with REVLIMID until TFR resolves to ≤ Grade 1 and patients may be treated
for management of symptoms per the guidance for treatment of Grade 1 and 2 TFR.
Starting Dose Adjustment for Renal Impairment in MCL:
See Section 2.5.
2.5 Starting Dose for Renal Impairment in MM, MDS or MCL
The recommendations for starting doses for patients with renal impairment are shown in the
following table.
Starting Dose Adjustments for Patients with Renal Impairment
Renal Function
(Cockcroft-Gault)
Dose in
Revlimid
combination
therapy for
MM and for
MCL
Dose in Revlimid
maintenance therapy
following auto-HSCT
for MM and for MDS
CLcr 30to 60 mL/min
10 mg once
daily
5 mg once daily
CLcr < 30 mL/min
(not requiring
dialysis)
7.5 mg
once daily
2.5 mg once daily
CLcr < 30 mL/min
(requiring dialysis)
5 mg
once daily. On
dialysis days,
administer the
dose following
dialysis.
2.5 mg once daily. On
dialysis days,
administer the dose
following dialysis.
REVLIMID Maintenance Therapy Following Auto-HSCT for MM and for MCL and
MDS:
Base subsequent REVLIMID dose increase or decrease on individual patient treatment
tolerance [see Dosage and Administration (2.1- 2.3)].
All patients
For other grade 3 or 4 toxicities judged to be related to lenalidomide, treatment should be
stopped and only restarted at next lower dose level when toxicity has resolved to ≤ grade 2
depending on the physician’s discretion.
Lenalidomide interruption or discontinuation should be considered for grade 2 or 3 skin rash.
Lenalidomide must be discontinued for angioedema, grade 4 rash, exfoliative or bullous rash, or
if Stevens-Johnson syndrome (SJS), toxic epidermal necrolysis (TEN) or Drug Reaction with
Eosinophilia and Systemic Symptoms (DRESS) is suspected, and should not be resumed
following discontinuation from these reactions.
3.
DOSAGE FORMS AND STRENGTHS
REVLIMID 2.5 mg, 5 mg, 7.5mg, 10 mg, 15 mg, 20 mg and 25 mg hard capsules will be
supplied through the Revlimid RMP-PPP.
REVLIMID is available in the following hard capsule strengths:
2.5 mg: White and blue-green opaque hard capsules imprinted “REV” on one half and “2.5 mg”
on the other half in black ink
5 mg: White opaque hard capsules imprinted “REV” on one half and “5 mg” on the other half
in black ink
7.5mg: Pale yellow/white hard capsules marked “REV 7.5 mg”
10 mg: Blue/green and pale yellow opaque hard capsules imprinted “REV” on one half and “10
mg” on the other half in black ink
15 mg: Powder blue and white opaque hard capsules imprinted “REV” on one half and “15 mg”
on the other half in black ink
20 mg: Powder blue and blue-green opaque hard capsules imprinted “REV” on one half and “20
mg” on the other half in black ink
25 mg: White opaque hard capsules imprinted “REV” on one half and “25 mg” on the other half
in black ink
4.
CONTRAINDICATIONS
4.1 Pregnancy
REVLIMID can cause fetal harm when administered to a pregnant female. Limb abnormalities
were seen in the offspring of monkeys that were dosed with lenalidomide during organogenesis.
This effect was seen at all doses tested. Due to the results of this developmental monkey study,
and lenalidomide’s structural similarities to thalidomide, a known human teratogen, lenalidomide
is contraindicated in pregnant women and women capable of becoming pregnant [
see Boxed
Warning
.
Females of childbearing potential may be treated with lenalidomide provided adequate
precautions are taken to avoid pregnancy.
If hormonal or IUD contraception is medically contraindicated, two other effective or highly
effective methods may be used.
Females of childbearing potential being treated with REVLIMID must have pregnancy testing
(sensitivity of at least 25 mIU/mL). The test should be performed prior to beginning therapy within
3 days prior to prescribing REVLIMID and then monthly thereafter (including dose interruptions).
Pregnancy
testing
should
be
performed
also
4
weeks
following
discontinuation
of
REVLIMID therapy.
Pregnancy testing and counseling must be performed if a patient misses her period or if there is
any abnormality in menstrual bleeding. If pregnancy occurs, REVLIMID must be immediately
discontinued. Under these conditions, the patient should be referred to an obstetrician/gynecologist
experienced in reproductive toxicity for further evaluation and counseling.
If this drug is used during pregnancy or if the patient becomes pregnant while taking this drug, the
patient should be apprised of the potential hazard to the fetus [
see Warnings and Precautions (5.1,
5.2), Use in Special Populations (8.1), (8.5)
4.2
Allergic Reactions
REVLIMID is contraindicated in patients who have demonstrated hypersensitivity (e.g.,
angioedema, Stevens-Johnson syndrome, toxic epidermal necrolysis) to lenalidomide [
see
Warnings and Precautions (5.8)
.
5.
WARNINGS AND PRECAUTIONS
5.1 Embryo-Fetal Toxicity
REVLIMID
thalidomide
analogue
contraindicated
during
pregnancy.
Thalidomide is a known human teratogen that causes life-threatening human birth defects or
embryo-fetal death [
see Use in Specific Populations
(8.1)
]. An embryo-fetal development study in
monkeys indicates that lenalidomide produced malformations in the offspring of female monkeys
who received the drug during pregnancy, similar to birth defects observed in humans following
exposure to thalidomide during pregnancy.
If REVLIMID is used during pregnancy, it may cause birth defects or death to a developing baby.
Females of childbearing potential must be advised to avoid pregnancy while on REVLIMID. Two
reliable forms of contraception should be used simultaneously during therapy, during dose
interruptions and for at least 4 weeks following discontinuation of therapy.
There are no adequate and well-controlled studies in pregnant females.
REVLIMID can be prescribed only in agreement with RMP limitations.
5.2
Reproductive Risk and
Special Prescribing Requirements (Revlimid RMP-PPP)
Revlimid can be prescribed and dispensed only if following the Revlimid Risk Management
Program. All patients must follow the Revlimid Risk Minimization Program in order to receive
Revlimid (refer to black box warning section)
Female Patients:
Two effective contraception methods must be used by female patients of childbearing potential for
at least 4 weeks before beginning REVLIMID therapy, during therapy, during dose interruptions
and for 4 weeks following discontinuation of REVLIMID therapy unless continuous abstinence
from heterosexual sexual contact is the chosen method. Reliable contraception is indicated even
where there has been a history of infertility, unless due to hysterectomy, a bilateral oophorectomy,
because the patient has been postmenopausal naturally for at least 24 consecutive months or in any
other case indicated in Revlimid RMP. Females of childbearing potential should be referred to a
qualified provider of contraceptive methods, if needed. Sexually mature females who have not
undergone
hysterectomy,
have
bilateral
oophorectomy,
have
been
postmenopausal naturally for at least 24 consecutive months (i.e., who have had menses at some
time in the preceding 24 consecutive months) or in any other case indicated in the Revlimid RMP,
are considered to be females of childbearing potential.
Cessation of menses due to anti-cancer therapy, do not exclude the potential to become
pregnant.
Two reliable forms of contraception must be used simultaneously unless continuous abstinence
from heterosexual sexual contact is the chosen method.
Females of childbearing potential must have a negative pregnancy test (sensitivity of at least 25
mIU/mL) before starting the therapy, and then monthly thereafter (including dose interruptions
and including 4 weeks following discontinuation of REVLIMID therapy). The test should be
performed within 3 days prior to prescribing REVLIMID. A prescription for REVLIMID for a
female of childbearing potential must not be issued by the prescriber until a negative pregnancy
test has been verified by the prescriber.
Pregnancy testing and counseling should be performed if a patient misses her period or if there is
any abnormality in her pregnancy test or in her menstrual bleeding. REVLIMID therapy must be
discontinued during this evaluation.
Pregnancy test results should be verified by the prescriber prior to dispensing
any
prescription.
If pregnancy does occur during treatment, REVLIMID must be discontinued immediately.
Any suspected fetal exposure to REVLIMID must be reported to the attending physician and
Neopharm.
patient
should
referred
obstetrician/gynecologist
experienced
reproductive toxicity for further evaluation and counseling.
Do not breastfeed during therapy (including dose interruptions).
Patients should not donate blood while taking REVLIMID, during any breaks (discontinuations)
in your therapy, and for 4 weeks following discontinuation of REVLIMID therapy.
Male Patients:
Clinical data has demonstrated the presence of lenalidomide in human semen. Therefore, males
receiving REVLIMID must always use a latex/ polyurethane condom during any sexual contact
with females of childbearing potential, even if they have undergone a successful vasectomy. In the
case of a male patient with an allergy to latex or polyurethane, at least one highly effective form
of contraception should be used by any female sexual partner. Contraception should be started in
this partner at least 4 weeks prior to the start of a sexual relationship with the patient, and continued
throughout
REVLIMID
therapy
including
dose
interruptions
weeks
following
discontinuation of therapy.
Patients should not donate blood and semen or sperm while taking REVLIMID, during any breaks
(discontinuations) in your therapy, and for 4 weeks following discontinuation of REVLIMID
therapy.
Once treatment has started and during dose interruptions
, pregnancy testing for females of
childbearing potential should be performed every 4 weeks.
Pregnancy testing should be performed also 4 weeks following discontinuation of
REVLIMID therapy
Prescriptions for women of childbearing potential can be for a maximum duration of 4 weeks,
and prescriptions for all other patients can be for a maximum duration of 12 weeks.
5.3 Other warnings and precautions of use
Hematologic Toxicity
REVLIMID can cause significant neutropenia and thrombocytopenia. Monitor patients with
neutropenia for signs of infection. Advise patients to observe for bleeding or bruising, especially
with use of concomitant medication that may increase risk of bleeding. Patients taking
REVLIMID should have their complete blood counts assessed periodically as described below
[see Dosage and Administration (2.1, 2.2, 2.3)]. Patients should be advised to promptly report
febrile episodes and dose reductions may be required. Patients and physicians are advised to be
observant for signs and symptoms of bleeding, including petechiae and epistaxes, especially in
patients receiving concomitant medicinal products susceptible to induce bleeding.
Patients taking REVLIMID in combination with dexamethasone or as REVLIMID maintenance
therapy for MM should have their complete blood counts (CBC) assessed every 7 days (weekly)
for the first 2 cycles, on Days 1 and 15 of Cycle 3, and every 28 days (4 weeks) thereafter. A
dose interruption and/or dose reduction may be required [see Dosage and Administration (2.1)].
In the MM maintenance therapy trials, Grade 3 or 4 neutropenia was reported in up to 59% of
REVLIMID treated patients and Grade 3 or 4 thrombocytopenia in up to 38% of REVLIMID-
treated patients [see Adverse Reactions (6.1)].
Patients taking REVLIMID for MDS should have their complete blood counts monitored weekly
for the first 8 weeks and at least monthly thereafter.
Patients taking REVLIMID for MCL should have their complete blood counts monitored weekly
for the first cycle (28 days), every 2 weeks during cycles 2-4, and then monthly thereafter.
Patients may require dose interruption and/or dose reduction.
In the MCL trial, Grade 3 or 4
neutropenia was reported in 43% of the patients. Grade 3 or 4 thrombocytopenia was reported in
28% of the patients.
Neutropenia and thrombocytopenia
NDMM
Newly diagnosed multiple myeloma: patients who have undergone ASCT treated with
lenalidomide maintenance
The adverse reactions from CALGB 100104 included events reported post-high dose melphalan
and ASCT (HDM/ASCT) as well as events from the maintenance treatment period. A second
analysis identified events that occurred after the start of maintenance treatment. In IFM 2005-02,
the adverse reactions were from the maintenance treatment period only.
Overall, grade 4 neutropenia was observed at a higher frequency in the lenalidomide
maintenance arms compared to the placebo maintenance arms in the 2 studies evaluating
lenalidomide maintenance in NDMM patients who have undergone ASCT (32.1% vs 26.7%
[16.1% vs 1.8% after the start of maintenance treatment] in CALGB 100104 and 16.4% vs 0.7%
in IFM 2005-02, respectively). Treatment-emergent AEs of neutropenia leading to lenalidomide
discontinuation were reported in 2.2% of patients in CALGB 100104 and 2.4% of patients in
IFM 2005-02, respectively. Grade 4 febrile neutropenia was reported at similar frequencies in the
lenalidomide maintenance arms compared to placebo maintenance arms in both studies (0.4% vs
0.5% [0.4% vs 0.5% after the start of maintenance treatment] in CALGB 100104 and 0.3% vs
0% in IFM 2005-02, respectively). Patients should be advised to promptly report febrile
episodes, a treatment interruption and/or dose reductions may be required (see section 4.2).
Grade 3 or 4 thrombocytopenia was observed at a higher frequency in the lenalidomide
maintenance arms compared to the placebo maintenance arms in studies evaluating lenalidomide
maintenance in NDMM patients who have undergone ASCT (37.5% vs 30.3% [17.9% vs 4.1%
after the start of maintenance treatment] in CALGB 100104 and 13.0% vs 2.9% in IFM 2005-02,
respectively). Patients and physicians are advised to be observant for signs and symptoms of
bleeding, including petechiae and epistaxes, especially in patients receiving concomitant
medicinal products susceptible to induce bleeding (see section 4.8, Haemorrhagic disorders).
Newly diagnosed multiple myeloma: patients who are not eligible for transplant treated with
lenalidomide in combination with low dose dexamethasone
Grade 4 neutropenia was observed in the lenalidomide arms in combination with low dose
dexamethasone to a lesser extent than in the comparator arm (8.5% in the Rd [continuous
treatment] and Rd18 [treatment for 18 four-week cycles] compared with 15% in the
melphalan/prednisone/thalidomide arm). Grade 4 febrile neutropenia episodes were consistent
with the comparator arm (0.6 % in the Rd and Rd18 lenalidomide/dexamethasone-treated
patients compared with 0.7% in the melphalan/prednisone/thalidomide arm.
Grade 3 or 4 thrombocytopenia was observed to a lesser extent in the Rd and Rd18 arms than in
the comparator arm (8.1% vs 11.1%, respectively).
Newly diagnosed multiple myeloma: patients who are not eligible for transplant treated with
lenalidomide in combination with melphalan and prednisone
The combination of lenalidomide with melphalan and prednisone in clinical trials of newly
diagnosed multiple myeloma patients is associated with a higher incidence of grade 4
neutropenia (34.1% in melphalan, prednisone and lenalidomide arm followed by lenalidomide
(MPR+R) and melphalan, prednisone and lenalidomide followed by placebo (MPR+p) treated
patients compared with 7.8% in MPp+p-treated patients;). Grade 4 febrile neutropenia episodes
were observed infrequently (1.7% in MPR+R/MPR+p treated patients compared to 0.0 % in
MPp+p treated patients;).
The combination of lenalidomide with melphalan and prednisone in multiple myeloma patients is
associated with a higher incidence of grade 3 and grade 4 thrombocytopenia (40.4% in
MPR+R/MPR+p treated patients, compared with 13.7% in MPp+p-treated patients).
Multiple myeloma: patients with at least one prior therapy
In the pooled MM trials Grade 3 and 4 hematologic toxicities were more frequent in patients
treated with the combination of REVLIMID and dexamethasone than in patients treated with
dexamethasone alone [
see Adverse Reactions (6.1)
Myelodysplastic Syndromes patients
Patients taking REVLIMID for MDS should have their complete blood counts monitored weekly
for the first 8 weeks and at least monthly thereafter.
Grade 3 or 4 hematologic toxicity was seen in 80% of patients enrolled in the MDS study. In the
48% of patients who developed Grade 3 or 4 neutropenia, the median time to onset was 42 days
(range, 14-411 days), and the median time to documented recovery was 17 days (range, 2-170
days). In the 54% of patients who developed Grade 3 or 4 thrombocytopenia, the median time to
onset was 28 days (range, 8-290 days), and the median time to documented recovery was 22 days
(range, 5-224 days) [see Boxed Warning and Dosage and Administration (2.2)].
Mantle cell lymphoma patients
Lenalidomide treatment in mantle cell lymphoma patients is associated with a higher incidence
of grade 3 and 4 neutropenia compared with patients on the control arm.
Tumour flare reaction and tumour lysis syndrome
Because lenalidomide has anti-neoplastic activity the complications of tumour lysis syndrome
(TLS) may occur. TLS and tumour flare reaction (TFR) have uncommonly been observed in
patients with lymphomas, who were treated with lenalidomide. Fatal instances of TLS have been
reported during treatment with lenalidomide. The patients at risk of TLS and TFR are those with
high tumour burden prior to treatment. Caution should be practiced when introducing these
patients to lenalidomide. These patients should be monitored closely, especially during the first
cycle or dose-escalation, and appropriate precautions taken. There have been rare reports of TLS
in patients with MM treated with lenalidomide, and no reports in patients with MDS treated with
lenalidomide.
Tumour burden
Mantle cell lymphoma
Lenalidomide is not recommended for the treatment of patients with high tumour burden if
alternative treatment options are available.
Early death
In study MCL-002 there was overall an apparent increase in early (within 20 weeks) deaths.
Patients with high tumour burden at baseline are at increased risk of early death, there were
16/81 (20%) early deaths in the lenalidomide arm and 2/28 (7%) early deaths in the control arm.
Within 52 weeks corresponding figures were 32/81 (40%) and 6/28 (21%) (See section 5.1).
Adverse events
In study MCL-002, during treatment cycle 1, 11/81 (14%) patients with high tumour burden
were withdrawn from therapy in the lenalidomide arm vs. 1/28 (4%) in the control group. The
main reason for treatment withdrawal for patients with high tumour burden during treatment
cycle 1 in the lenalidomide arm was adverse events, 7/11 (64%).
Patients with high tumour burden should therefore be closely monitored for adverse reactions
(see Section 4.8) including signs of tumour flare reaction (TFR). Please refer to section 4.2 for
dose adjustments for TFR.
High tumour burden was defined as at least one lesion ≥5 cm in diameter or 3 lesions ≥3 cm.
Tumour flare reaction
Mantle cell lymphoma
Careful monitoring and evaluation for TFR is recommended. Patients with high mantle cell
lymphoma International Prognostic Index (MIPI) at diagnosis or bulky disease (at least one
lesion that is ≥ 7 cm in the longest diameter) at baseline may be at risk of TFR. Tumour flare
reaction may mimic progression of disease (PD). Patients in studies MCL-002 and MCL-001 that
experienced Grade 1 and 2 TFR were treated with corticosteroids, non-steroidal anti-
inflammatory drugs (NSAIDs) and/or narcotic analgesics for management of TFR symptoms.
The decision to take therapeutic measures for TFR should be made after careful clinical
assessment of the individual patient (see section 4.2).
Infection with or without Neutropenia
Patients with multiple myeloma are prone to develop infections including pneumonia. A higher
rate of infections was observed with lenalidomide in combination with dexamethasone than with
MPT in patients with NDMM who are not eligible for transplant, and with lenalidomide
maintenance compared to placebo in patients with NDMM who had undergone ASCT.
Grade ≥ 3 infections occurred within the context of neutropenia in less than one-third of the
patients. Patients with known risk factors for infections should be closely monitored. All patients
should be advised to seek medical attention promptly at the first sign of infection (e.g., cough,
fever, etc.) thereby allowing for early management to reduce severity.
Viral reactivation
Cases of viral reactivation have been reported in patients receiving lenalidomide, including
serious cases of herpes zoster or hepatitis B virus (HBV) reactivation.
Some of the cases of viral reactivation had a fatal outcome.
Some of the cases of herpes zoster reactivation resulted in disseminated herpes zoster, meningitis
herpes zoster or ophthalmic herpes zoster requiring a temporary hold or permanent
discontinuation of the treatment with lenalidomide and adequate antiviral treatment.
Reactivation of hepatitis B has been reported rarely in patients receiving lenalidomide who have
previously been infected with the hepatitis B virus (HBV). Some of these cases have progressed
to acute hepatic failure resulting in discontinuation of lenalidomide and adequate antiviral
treatment. Hepatitis B virus status should be established before initiating treatment with
lenalidomide. For patients who test positive for HBV infection, consultation with a physician
with expertise in the treatment of hepatitis B is recommended. Caution should be exercised when
lenalidomide is used in patients previously infected with HBV, including patients who are anti-
HBc positive but HBsAg negative. These patients should be closely monitored for signs and
symptoms of active HBV infection throughout therapy.
Venous and Arterial Thromboembolism
In patients with multiple myeloma, the combination of lenalidomide with dexamethasone is
associated with an increased risk of venous thromboembolism (predominantly deep vein
thrombosis and pulmonary embolism) and was seen to a lesser extent with lenalidomide in
combination with melphalan and prednisone.
In patients with multiple myeloma, myelodysplastic syndromes and mantle cell lymphoma,
treatment with lenalidomide monotherapy was associated with a lower risk of venous
thromboembolism (predominantly deep vein thrombosis and pulmonary embolism) than in
patients with multiple myeloma treated with lenalidomide in combination therapy
In patients with multiple myeloma, the combination of lenalidomide with dexamethasone is
associated with an increased risk of arterial thromboembolism (predominantly myocardial
infarction and cerebrovascular event) and was seen to a lesser extent with lenalidomide in
combination with melphalan and prednisone. The risk of ATE is lower in patients with multiple
myeloma treated with lenalidomide monotherapy than in patients with multiple myeloma treated
with lenalidomide in combination therapy.
Consequently, patients with known risk factors for thromboembolism – including prior
thrombosis – should be closely monitored. Action should be taken to try to minimize all
modifiable risk factors (e.g. smoking, hypertension, and hyperlipidaemia). Concomitant
administration of erythropoietic agents or previous history of thromboembolic events may also
increase thrombotic risk in these patients. Therefore, erythropoietic agents, or other agents that
may increase the risk of thrombosis, such as hormone replacement therapy, should be used with
caution in multiple myeloma patients receiving lenalidomide with dexamethasone. A
haemoglobin concentration above 12 g/dl should lead to discontinuation of erythropoietic agents.
Patients and physicians are advised to be observant for the signs and symptoms of
thromboembolism. Patients should be instructed to seek medical care if they develop symptoms
such as shortness of breath, chest pain, arm or leg swelling. Prophylactic antithrombotic
medicines should be recommended, especially in patients with additional thrombotic risk factors.
The decision to take antithrombotic prophylactic measures should be made after careful
assessment of an individual patient’s underlying risk factors.
If the patient experiences any thromboembolic events, treatment must be discontinued, and
standard anticoagulation therapy started. Once the patient has been stabilised on the
anticoagulation treatment and any complications of the thromboembolic event have been
managed, the lenalidomide treatment may be restarted at the original dose dependent upon a
benefit risk assessment. The patient should continue anticoagulation therapy during the course of
lenalidomide treatment.
Second Primary Malignancies
An increase of second primary malignancies (SPM) has been observed in clinical trials in
previously treated myeloma patients receiving lenalidomide/dexamethasone (3.98 per 100
person-years) compared to controls (1.38 per 100 person-years). Non-invasive SPM comprise
basal cell or squamous cell skin cancers. Most of the invasive SPMs were solid tumour
malignancies.
In clinical trials of newly diagnosed multiple myeloma patients not eligible for transplant, a 4.9-
fold increase in incidence rate of hematologic SPM (cases of AML, MDS) has been observed in
patients receiving lenalidomide in combination with melphalan and prednisone until progression
(1.75 per 100 person-years) compared with melphalan in combination with prednisone (0.36 per
100 person-years).
A 2.12-fold increase in incidence rate of solid tumor SPM has been observed in patients
receiving lenalidomide (9 cycles) in combination with melphalan and prednisone (1.57 per 100
person-years) compared with melphalan in combination with prednisone (0.74 per 100 person-
years).
In patients receiving lenalidomide in combination with dexamethasone until progression or for
18 months, the hematologic SPM incidence rate (0.16 per 100 person-years) was not increased as
compared to thalidomide in combination with melphalan and prednisone (0.79 per 100 person-
years).
A 1.3-fold increase in incidence rate of solid tumor SPM has been observed in patients receiving
lenalidomide in combination with dexamethasone until progression or for 18 months (1.58 per
100 person-years) compared to thalidomide in combination with melphalan and prednisone (1.19
per 100 person-years).
The increased risk of secondary primary malignancies associated with lenalidomide is relevant
also in the context of NDMM after stem cell transplantation. Though this risk is not yet fully
characterized, it should be kept in mind when considering and using Revlimid in this setting.
The incidence rate of hematologic malignancies, most notably AML, MDS and B-cell
malignancies (including Hodgkin’s lymphoma), was 1.31 per 100 person-years for the
lenalidomide arms and 0.58 per 100 person-years for the placebo arms (1.02 per 100 person-
years for patients exposed to lenalidomide after ASCT and 0.60 per 100 person-years for patients
not-exposed to lenalidomide after ASCT). The incidence rate of solid tumour SPMs was 1.36 per
100 person-years for the lenalidomide arms and 1.05 per 100 person-years for the placebo arms
(1.26 per 100 person-years for patients exposed to lenalidomide after ASCT and 0.60 per 100
person-years for patients not-exposed to lenalidomide after ASCT).
The risk of occurrence of hematologic SPM must be taken into account before initiating treatment
with lenalidomide either in combination with melphalan or immediately following high-dose
melphalan and ASCT. Physicians should carefully evaluate patients before and during treatment
using standard cancer screening for occurrence of SPM and institute treatment as indicated.
Hepatotoxicity
Hepatic failure, including fatal cases, has occurred in patients treated with lenalidomide in
combination with dexamethasone: acute hepatic failure, toxic hepatitis, cytolytic hepatitis,
cholestatic hepatitis, and mixed cytolytic/cholestatic hepatitis have been reported. In clinical
trials, 15% of patients experienced hepatotoxicity (with hepatocellular, cholestatic and mixed
characteristics); 2% of patients with multiple myeloma and 1% of patients with myelodysplasia
had serious hepatotoxicity events. The mechanism of drug-induced hepatotoxicity is unknown.
Pre-existing viral liver disease, elevated baseline liver enzymes, and possibly treatment with
antibiotics and older age might be risk factors. Monitor liver enzymes periodically. Stop
Revlimid upon elevation of liver enzymes. After return to baseline values, treatment at a lower
dose may be considered.
Lenalidomide is excreted by the kidneys. It is important to dose adjust patients with renal
impairment in order to avoid plasma levels which may increase the risk for higher
haematological side effects or hepatotoxicity. Monitoring of liver function is recommended,
particularly when there is a history of or concurrent viral liver infection or when lenalidomide is
combined with medications known to be associated with liver dysfunction.
Allergic and severe skin reactions
Severe cutaneous reactions including SJS, and TEN and DRESS have been reported with the use
of lenalidomide. Patients should be advised of the signs and symptoms of these reactions by their
prescribers and should be told to seek medical attention immediately if they develop these
symptoms. Lenalidomide must be discontinued for exfoliative or bullous rash, or if SJS, TEN or
DRESS is suspected, and should not be resumed following discontinuation for these reactions.
Interruption or discontinuation of lenalidomide should be considered for other forms of skin
reaction depending on severity. Patients with a history of severe rash associated with thalidomide
treatment should not receive lenalidomide.
REVLIMID hard capsules contain lactose. Risk-benefit of REVLIMID treatment should be
evaluated in patients with lactose intolerance.
Newly diagnosed multiple myeloma patients
There was a higher rate of intolerance (grade 3 or 4 adverse events, serious adverse events,
discontinuation) in patients with age > 75 years, ISS stage III, ECOG PS≤2 or CLcr<60 mL/min
when lenalidomide is given in combination. Patients should be carefully assessed for their ability
to tolerate lenalidomide in combination, with consideration to age, ISS stage III, ECOG PS≤2 or
CLcr<60 mL/min.
Cataract
Cataract has been reported with a higher frequency in patients receiving lenalidomide in
combination with dexamethasone particularly when used for a prolonged time. Regular
monitoring of visual ability is recommended.
Peripheral neuropathy
Lenalidomide is structurally related to thalidomide, which is known to induce severe peripheral
neuropathy. There was no increase in peripheral neuropathy observed with long term use of
lenalidomide for the treatment of newly diagnosed multiple myeloma.
6 ADVERSE REACTIONS
The following adverse reactions are described in detail in other sections of the prescribing
information:
Embryo-Fetal Toxicity [
see Boxed Warnings, Warnings and Precautions (5.1, 5.2)
Neutropenia and thrombocytopenia [
see Boxed Warnings, Other Warnings and
Precautions of use (5.3)
Venous and arterial thromboembolism [
see Other Warnings and Precautions of use
(5.3)]
Second Primary Malignancies [
see Other Warnings and Precautions of use (5.3)
Hepatotoxicity [
see Other
Warnings
and Precautions of use (5.3)
Allergic and severe skin reactions [
see Other Warnings and Precautions of use (5.3)
Cataract [
see Other Warnings and Precautions of use (5.3)
Peripheral Neuropathy [
see Other Warnings and Precautions of use (5.3)
Because clinical trials are conducted under widely varying conditions, adverse reaction rates
observed in the clinical trials of a drug cannot be directly compared to rates in the clinical trials
of another drug and may not reflect the rates observed in practice.
6.1 Clinical Trials Experience in Multiple Myeloma
Summary of the safety profile
Newly diagnosed multiple myeloma: patients who have undergone ASCT treated with
lenalidomide maintenance
A conservative approach was applied to determine the adverse reactions from CALGB 100104.
The adverse reactions described in Table 1 included events reported post-HDM/ASCT as well as
events from the maintenance treatment period. A second analysis that identified events that
occurred after the start of maintenance treatment suggests that the frequencies described in Table
1 may be higher than actually observed during the maintenance treatment period. In IFM 2005-
02, the adverse reactions were from the maintenance treatment period only.
The serious adverse reactions observed more frequently (≥5%) with lenalidomide maintenance
than placebo were:
Pneumonias (10.6%; combined term) from IFM 2005-02
Lung infection (9.4% [9.4% after the start of maintenance treatment]) from CALGB
100104
In the IFM 2005-02 study, the adverse reactions observed more frequently with lenalidomide
maintenance than placebo were neutropenia (60.8%), bronchitis (47.4%), diarrhoea (38.9%),
nasopharyngitis (34.8%), muscle spasms (33.4%), leucopenia (31.7%), asthenia (29.7%), cough
(27.3%), thrombocytopenia (23.5%), gastroenteritis (22.5%) and pyrexia (20.5%).
In the CALGB 100104 study, the adverse reactions observed more frequently with lenalidomide
maintenance than placebo were neutropenia (79.0% [71.9% after the start of maintenance
treatment]), thrombocytopenia (72.3% [61.6%]), diarrhoea (54.5% [46.4%]), rash (31.7%
[25.0%]), upper respiratory tract infection (26.8% [26.8%]), fatigue (22.8% [17.9%]), leucopenia
(22.8% [18.8%]) and anemia (21.0% [13.8%]).
Summary of the safety profile in newly diagnosed multiple myeloma:patients who are not eligible
for transplant treated with lenalidomide in combination with low dose dexamethasone:
The serious adverse reactions observed more frequently (≥5%) with lenalidomide in combination
with low dose dexamethasone (Rd and Rd18) than with melphalan, prednisone and thalidomide
(MPT) were:
Pneumonia (9.8%)
Renal failure (including acute) (6.3%)
The adverse reactions observed more frequently with Rd or Rd18 than MPT were: diarrhoea
(45.5%), fatigue (32.8%), back pain (32.0%), asthenia (28.2%), insomnia (27.6%), rash (24.3%),
decreased appetite (23.1%), cough (22.7%), pyrexia (21.4%), and muscle spasms (20.5%).
Summary of the safety profile in newly diagnosed multiple myeloma: who are not eligible for
transplant treated with lenalidomide in combination with melphalan and prednisone:
The serious adverse reactions observed more frequently (≥5%) with melphalan prednisone, and
lenalidomide followed by lenalidomide maintenance (MPR+R) or melphalan prednisone, and
lenalidomide followed by placebo (MPR+p) than melphalan, prednisone and placebo followed
by placebo (MPp+p) were:
Febrile neutropenia (6.0%)
Anemia (5.3%)
The adverse reactions observed more frequently with MPR+R or MPR+ p than MPp+p were:
neutropenia (83.3%), anemia (70.7%), thrombocytopenia (70.0%), leukopenia (38.8%),
constipation (34.0%), diarrhoea (33.3%), rash (28.9%), pyrexia (27.0%), peripheral oedema
(25.0%), cough (24.0%), decreased appetite (23.7%), and asthenia (22.0%).
Summary of the safety profile in multiple myeloma: patients with at least one prior therapy
In two Phase III placebo-controlled studies, 353 patients with multiple myeloma were exposed to
the lenalidomide/dexamethasone combination and 351 to the placebo/dexamethasone
combination.
The most serious adverse reactions observed more frequently in lenalidomide/dexamethasone
than placebo/dexamethasone combination were:
Venous thromboembolism (deep vein thrombosis, pulmonary embolism)
Grade 4 neutropenia.
The observed adverse reactions which occurred more frequently with lenalidomide and
dexamethasone than placebo and dexamethasone in pooled multiple myeloma clinical trials
(MM-009 and MM-010) were fatigue (43.9%), neutropenia (42.2%), constipation (40.5%),
diarrhoea (38.5%), muscle cramp (33.4%), anemia (31.4%), thrombocytopenia (21.5%), and rash
(21.2%).
Tabulated summary of adverse reactions
The adverse reactions observed in patients treated for multiple myeloma are listed below by
system organ class and frequency. Within each frequency grouping, adverse reactions are
presented in order of decreasing seriousness. Frequencies are defined as: very common (≥ 1/10);
common (≥ 1/100 to < 1/10); uncommon (≥ 1/1,000 to < 1/100); rare (≥ 1/10,000 to < 1/1,000);
very rare (< 1/10,000), not known (cannot be estimated from the available data).
Adverse reactions have been included under the appropriate category in the table below
according to the highest frequency observed in any of the main clinical trials.
Tabulated summary for monotherapy in MM
The following table is derived from data gathered during NDMM studies in patients who have
undergone ASCT treated with lenalidomide maintenance. The data were not adjusted according
to the longer duration of treatment in the lenalidomide-containing arms continued until disease
progression versus the placebo arms in the pivotal multiple myeloma studies (see section 5.1).
Table 1. ADRs reported in clinical trials in patients with multiple myeloma treated with
lenalidomide maintenance therapy
System Organ Class/Preferred
Term
All ADRs/Frequency
Grade 3-4 ADRs/Frequency
Infections and Infestations
Very Common
Pneumonias
◊, a
, Upper
respiratory tract infection,
Neutropenic infection,
Bronchitis
, Influenza
Gastroenteritis
, Sinusitis,
Nasopharyngitis, Rhinitis
Common
Infection
, Urinary tract
infection
*, Lower respiratory
tract infection, Lung infection
Very Common
Pneumonias
◊, a
, Neutropenic
infection
Common
Sepsis
◊, b
, Bacteraemia, Lung
infection
, Lower respiratory
tract infection bacterial,
Bronchitis
, Influenza
Gastroenteritis
, Herpes zoster
Infection
Neoplasms Benign, Malignant
and Unspecified (incl cysts
and polyps)
Common
Myelodysplastic syndrome
Blood and Lymphatic System
Disorders
Very Common
Neutropenia
, Febrile
neutropenia
Thrombocytopenia
Anemia
Leucopenia
, Lymphopenia
Very Common
Neutropenia
, Febrile
neutropenia
Thrombocytopenia
nemia
Leucopenia
, Lymphopenia
Common
Pancytopenia
Metabolism and Nutrition
Disorders
Very Common
Hypokalaemia
Common
Hypokalaemia, Dehydration
Nervous System Disorders
Very Common
Paraesthesia
Common
Peripheral neuropathy
Common
Headache
Vascular Disorders
Common
Pulmonary embolism
Common
Deep vein thrombosis
,◊,d
Respiratory, Thoracic and
Mediastinal Disorders
Very Common
Cough
Common
Dyspnoea
, Rhinorrhoea
Common
Dyspnoea
Gastrointestinal Disorders
Very Common
Diarrhoea, Constipation,
Abdominal pain, Nausea
Common
Vomiting, Abdominal pain
upper
Common
Diarrhoea, Vomiting, Nausea
Hepatobiliary Disorders
Very Common
Abnormal liver function tests
Common
Abnormal liver function tests
Skin and Subcutaneous Tissue
Disorders
Very Common
Rash, Dry skin
Common
Rash, Pruritus
Musculoskeletal and
Connective Tissue Disorders
Very Common
Muscle spasms
System Organ Class/Preferred
Term
All ADRs/Frequency
Grade 3-4 ADRs/Frequency
Common
Myalgia, Musculoskeletal pain
General Disorders and
Administration Site
Conditions
Very Common
Fatigue, Asthenia, Pyrexia
Common
Fatigue, Asthenia
Adverse reactions reported as serious in clinical trials in patients with NDMM who had undergone ASCT
Applies to serious adverse drug reactions only
“Pneumonias” combined AE term includes the following PTs: Bronchopneumonia, Lobar pneumonia, Pneumocystis jiroveci pneumonia,
Pneumonia, Pneumonia klebsiella, Pneumonia legionella, Pneumonia mycoplasmal, Pneumonia pneumococcal, Pneumonia streptococcal,
Pneumonia viral, Lung disorder, Pneumonitis
“Sepsis” combined AE term includes the following PTs: Bacterial sepsis, Pneumococcal sepsis, Septic shock, Staphylococcal sepsis
“Peripheral neuropathy” combined AE term includes the following preferred terms (PTs): Neuropathy peripheral, Peripheral sensory
neuropathy, Polyneuropathy
“Deep vein thrombosis” combined AE term includes the following PTs: Deep vein thrombosis, Thrombosis, Venous thrombosis
Tabulated summary for combination therapy in MM
The following table is derived from data gathered during the multiple myeloma studies with
combination therapy. The data were not adjusted according to the greater duration of treatment in
the lenalidomide/dexamethasone versus the placebo/dexamethasone arms in the pivotal multiple
myeloma studies.
Table 2:
ADRs reported in clinical studies in patients with multiple myeloma treated with
lenalidomide in combination with dexamethasone, or with melphalan and
prednisone
System Organ
Class
/ Preferred Term
All ADRs/Frequency
Grade 3−4 ADRs/Frequency
Infections
and Infestations
Very Common
Pneumonia
, Upper respiratory tract
infection
, Bacterial, viral and fungal
infections (including opportunistic
infections)
, Nasopharyngitis, Pharyngitis,
Bronchitis
Common
Sepsis
, Sinusitis
Common
Pneumonia
, Bacterial, viral
and fungal infections
(including opportunistic
infections
), Cellulitis
Sepsis
, Bronchitis
Neoplasms
Benign,
Malignant and
Unspecified (incl
cysts and polyps)
Uncommon
Basal cell carcinoma
Squamous skin cancer
◊,
Common
Acute myeloid leukaemia
Myelodysplastic syndrome
Squamous cell carcinoma of
skin
◊,
Uncommon
T-cell type acute leukaemia
Basal cell carcinoma
, Tumour
lysis syndrome
System Organ
Class
/ Preferred Term
All ADRs/Frequency
Grade 3−4 ADRs/Frequency
Blood and
Lymphatic
System Disorders
Very Common
Neutropenia
, Thrombocytopenia
Anaemia
, Haemorrhagic disorder,
Leucopenias
Common
Febrile neutropenia
, Pancytopenia
Uncommon
Haemolysis, Autoimmune haemolytic
anaemia, Haemolytic anaemia
Very Common
Neutropenia
Thrombocytopenia
Anaemia
, Leucopenias
Common
Febrile neutropenia
Pancytopenia
, Haemolytic
anaemia
Uncommon
Hypercoagulation,
Coagulopathy
Immune System
Disorders
Uncommon
Hypersensitivity
Endocrine
Disorders
Common
Hypothyroidism
Metabolism and
Nutrition
Disorders
Very Common
Hypokalaemia
, Hyperglycaemia,
Hypocalcaemia
, Decreased appetite, Weight
decreased
Common
Hypomagnesaemia, Hyperuricaemia,
Dehydration
, Hypercalcaemia
Common
Hypokalaemia
Hyperglycaemia
Hypocalcaemia, Diabetes
mellitus
Hypophosphataemia,
Hyponatraemia
Hyperuricaemia, Gout,
Decreased appetite, Weight
decreased
Psychiatric
Disorders
Very Common
Depression, Insomnia
Uncommon
Loss of libido
Common
Depression, Insomnia
Nervous System
Disorders
Very Common
Peripheral neuropathies (excluding motor
neuropathy), Dizziness, Tremor, Dysgeusia,
Headache
Common
Ataxia, Balance impaired
Common
Cerebrovascular accident
Dizziness, Syncope
Uncommon
Intracranial haemorrhage,
Transient ischaemic attack,
Cerebral ischemia
Eye Disorders
Very Common
Cataracts, Blurred vision
Common
Reduced visual acuity
Common
Cataract
Uncommon
Blindness
Ear and
Labyrinth
Disorders
Common
Deafness (Including Hypoacusis), Tinnitus
System Organ
Class
/ Preferred Term
All ADRs/Frequency
Grade 3−4 ADRs/Frequency
Cardiac
Disorders
Common
Atrial fibrillation
, Bradycardia
Uncommon
Arrhythmia, QT prolongation, Atrial flutter,
Ventricular extrasystoles
Common
Myocardial infarction
(including acute)
, Atrial
fibrillation
, Congestive
cardiac failure
, Tachycardia,
Cardiac failure
, Myocardial
ischemia
Vascular
Disorders
Very Common
Venous thromboembolic events,
predominantly deep vein thrombosis and
pulmonary embolism
Common
Hypotension
, Hypertension, Ecchymosis
Very Common
Venous thromboembolic
events, predominantly deep
vein thrombosis and
pulmonary embolism
Common
Vasculitis
Uncommon
Ischemia, Peripheral ischemia,
Intracranial venous sinus
thrombosis
Respiratory,
Thoracic
and Mediastinal
Disorders
Very Common
Dyspnoea
, Epistaxis
Common
Respiratory distress
Dyspnoea
Gastrointestinal
Disorders
Very Common
Diarrhoea
, Constipation
, Abdominal pain
Nausea, Vomiting, Dyspepsia
Common
Gastrointestinal haemorrhage (including
rectal haemorrhage, haemorrhoidal
haemorrhage, peptic ulcer haemorrhage and
gingival bleeding), Dry mouth, Stomatitis,
Dysphagia
Uncommon
Colitis, Caecitis
Common
Diarrhoea
, Constipation
Abdominal pain
, Nausea,
Vomiting
Hepatobiliary
Disorders
Common
Abnormal liver function tests
Uncommon
Hepatic failure
Common
Cholestasis
, Abnormal liver
function tests
Uncommon
Hepatic failure
Skin and
Subcutaneous
Tissue Disorders
Very Common
Rashes, Pruritus
Common
Urticaria, Hyperhidrosis, Dry skin, Skin
hyperpigmentation, Eczema, Erythema
Uncommon
Skin discolouration, Photosensitivity
reaction
Common
Rashes
System Organ
Class
/ Preferred Term
All ADRs/Frequency
Grade 3−4 ADRs/Frequency
Musculoskeletal
and Connective
Tissue Disorders
Very Common
Muscle spasms, Bone pain
Musculoskeletal and connective tissue pain
and discomfort (including back pain
Arthralgia
Common
Muscular weakness, Joint swelling
, Myalgia
Common
Muscular weakness, Bone
pain
, Musculoskeletal and
connective tissue pain and
discomfort (including back
pain
Uncommon
Joint swelling
Renal and
Urinary
Disorders
Very Common
Renal failure (including acute)
Common
Haematuria, Urinary retention,
Urinary incontinence
Uncommon
Acquired Fanconi syndrome
Uncommon
Renal tubular necrosis
Reproductive
System and
Breast Disorders
Common
Erectile dysfunction
General
Disorders
and
Administration
Site Conditions
Very Common
Fatigue
, Oedema (including peripheral
oedema), Pyrexia
, Asthenia, Influenza like
illness syndrome (including pyrexia, cough,
myalgia, musculoskeletal pain, headache and
rigors)
Common
Chest pain, Lethargy
Common
Fatigue
, Pyrexia
, Asthenia
Investigations
Common
C-reactive protein increased
Injury, Poisoning
and Procedural
Complications
Common
Fall, Contusion
◊ Adverse reactions reported as serious in clinical trials in patients with multiple myeloma treated with lenalidomide in combination with
dexamethasone, or with melphalan and prednisone
+ Applies to serious adverse drug reactions only
* Squamous skin cancer was reported in clinical trials in previously treated myeloma patients with lenalidomide/dexamethasone compared to
controls
** Squamous cell carcinoma of skin was reported in a clinical trial in newly diagnosed myeloma patients with lenalidomide/dexamethasone
compared to controls
Other Adverse Reactions After At Least One Prior Therapy for Multiple Myeloma
In these studies, the following adverse drug reactions (ADRs) not described in other MM tables
(or the postmarketing experience table) that occurred at ≥1% rate and of at least twice of the
placebo percentage rate were reported:
Blood and lymphatic system disorders:
autoimmune hemolytic anemia
Cardiac disorders:
angina pectoris
Endocrine disorders:
hirsutism
Eye disorders:
blindness, ocular hypertension,
Gastrointestinal disorders:
glossodynia
General disorders and administration site conditions:
malaise
Investigations:
liver function tests abnormal, alanine aminotransferase increased
Psychiatric disorders:
mood swings, hallucination
Respiratory, thoracic and mediastinal disorders:
hoarseness
Skin and subcutaneous tissue disorders:
exanthem
6.2 Clinical Trials Experience in Myelodysplastic Syndromes
A total of 148 patients received at least 1 dose of 10 mg REVLIMID in the del 5q MDS clinical
study. At least one adverse event was reported in all of the 148 patients who were treated with
the 10 mg starting dose of REVLIMID. The most frequently reported adverse events were
related to blood and lymphatic system disorders, skin and subcutaneous tissue disorders,
gastrointestinal disorders, and general disorders and administrative site conditions
.
Thrombocytopenia (61.5%; 91/148) and neutropenia (58.8%; 87/148) were the most frequently
reported adverse events. The next most common adverse events observed were diarrhea (48.6%;
72/148), pruritus (41.9%; 62/148), rash (35.8%; 53/148) and fatigue (31.1%; 46/148). Table 3
summarizes the adverse events that were reported in ≥ 5% of the REVLIMID treated patients in
the del 5q MDS clinical study. Table 3 summarizes the most frequently observed Grade 3 and
Grade 4 adverse reactions regardless of relationship to treatment with REVLIMID. In the single-
arm studies conducted, it is often not possible to distinguish adverse events that are drug-related
and those that reflect the patient’s underlying disease.
Table 3: Summary of Adverse Events Reported in ≥5% of the
REVLIMID Treated Patients in del 5q MDS Clinical Study
10 mg Overall
System organ class/Preferred term
[a]
(N=148)
Patients with at least one adverse event
(100.0)
Blood and Lymphatic System Disorders
Thrombocytopenia
(61.5)
Neutropenia
(58.8)
Anemia
(11.5)
Leukopenia
(8.1)
Febrile Neutropenia
(5.4)
Skin and Subcutaneous Tissue Disorders
Pruritus
62 (41.9)
Rash
(35.8)
Dry Skin
(14.2)
Contusion
(8.1)
Night Sweats
(8.1)
Sweating Increased
(6.8)
Ecchymosis
(5.4)
Erythema
(5.4)
Gastrointestinal Disorders
Diarrhea
(48.6)
Constipation
(23.6)
Nausea
(23.6)
Abdominal Pain
(12.2)
Vomiting
(10.1)
Abdominal Pain Upper
(8.1)
Dry Mouth
(6.8)
Loose Stools
(6.1)
Respiratory, Thoracic and Mediastinal Disorders
Nasopharyngitis
(23.0)
Cough
(19.6)
Dyspnea
(16.9)
Pharyngitis
(15.5)
Epistaxis
(14.9)
Dyspnea Exertional
(6.8)
Rhinitis
(6.8)
Bronchitis
(6.1)
General Disorders and Administration Site Conditions
Fatigue
(31.1)
Pyrexia
(20.9)
Edema Peripheral
(20.3)
Asthenia
(14.9)
Edema
(10.1)
Pain
(6.8)
Rigors
(6.1)
Chest Pain
(5.4)
Musculoskeletal and Connective Tissue Disorders
Arthralgia
(21.6)
Back Pain
(20.9)
Muscle Cramp
(18.2)
Pain in Limb
(10.8)
Myalgia
(8.8)
Peripheral Swelling
(8.1)
Nervous System Disorders
Dizziness
(19.6)
Headache
(19.6)
Hypoesthesia
(6.8)
Dysgeusia
(6.1)
Peripheral Neuropathy
(5.4)
Infections and Infestations
Upper Respiratory Tract Infection
(14.9)
Pneumonia
(11.5)
Urinary Tract Infection
(10.8)
Sinusitis
(8.1)
Cellulitis
(5.4)
Metabolism and Nutrition Disorders
Hypokalemia
(10.8)
Anorexia
(10.1)
Hypomagnesemia
(6.1)
Investigations
Alanine Aminotransferase Increased
(8.1)
Psychiatric Disorders
Insomnia
(10.1)
Depression
(5.4)
Renal and Urinary Disorders
Dysuria 10 (6.8)
Vascular Disorders
Hypertension 9 ( 6.1)
Endocrine Disorders
Acquired Hypothyroidism 10 (6.8)
Cardiac Disorders
Palpitations 8 (5.4)
System organ classes and preferred terms are coded using the MedDRA
dictionary. System organ classes
and preferred terms are listed in descending order of frequency for the Overall
column. A patient with multiple
occurrences of an AE is counted only once in the AE category.
Table 4: Most Frequently Observed Grade 3 and 4 Adverse Events [1]
Regardless of Relationship to Study Drug Treatment
10 mg
Preferred term
[2]
(N=148)
Patients with at least one Grade 3/4 AE
(88.5)
Neutropenia
(53.4)
Thrombocytopenia
(50.0)
Pneumonia
(7.4)
Rash
(6.8)
Anemia
(6.1)
Leukopenia
(5.4)
Fatigue
(4.7)
Dyspnea
(4.7)
Back Pain
(4.7)
Febrile Neutropenia
(4.1)
Nausea
(4.1)
Diarrhea
(3.4)
Pyrexia
(3.4)
Sepsis
(2.7)
Dizziness
(2.7)
Granulocytopenia
(2.0)
Chest Pain
(2.0)
Pulmonary Embolism
(2.0)
Respiratory Distress
(2.0)
Pruritus
(2.0)
Pancytopenia
(2.0)
Muscle Cramp
(2.0)
Respiratory Tract Infection
(1.4)
Upper Respiratory Tract Infection
(1.4)
Asthenia
(1.4)
Multi-organ Failure
(1.4)
Epistaxis
(1.4)
Hypoxia
(1.4)
Pleural Effusion
(1.4)
Pneumonitis
(1.4)
Pulmonary Hypertension
(1.4)
Vomiting
(1.4)
Sweating Increased
(1.4)
Arthralgia
(1.4)
Pain in Limb
(1.4)
Headache
(1.4)
Syncope
(1.4)
[1] Adverse events with frequency ≥1% in the 10 mg Overall group. Grade 3 and 4 are based on National Cancer
Institute Common Toxicity Criteria version 2.
[2] Preferred Terms are coded using the MedDRA dictionary. A patient with multiple occurrences of an AE is
counted only once in the Preferred Term category.
In other clinical studies of REVLIMID in MDS patients, the following serious adverse events
(regardless of relationship to study drug treatment) not described in Table 6 or 7 were reported:
Blood and lymphatic system disorders:
warm type hemolytic anemia, splenic infarction, bone
marrow depression, coagulopathy, hemolysis, hemolytic anemia, refractory anemia
Cardiac disorders:
cardiac failure congestive, atrial fibrillation, angina pectoris, cardiac arrest,
cardiac failure, cardio-respiratory arrest, cardiomyopathy, myocardial infarction, myocardial
ischemia, atrial fibrillation aggravated, bradycardia, cardiogenic shock, pulmonary edema,
supraventricular arrhythmia, tachyarrhythmia, ventricular dysfunction
Ear and labyrinth disorders:
vertigo
Endocrine disorders:
Basedow’s disease
Gastrointestinal disorders:
gastrointestinal hemorrhage, colitis ischemic, intestinal perforation,
rectal hemorrhage, colonic polyp, diverticulitis, dysphagia, gastritis, gastroenteritis,
gastroesophageal reflux disease, obstructive inguinal hernia, irritable bowel syndrome, melena,
pancreatitis due to biliary obstruction, pancreatitis, perirectal abscess, small intestinal
obstruction, upper gastrointestinal hemorrhage
General disorders and administration site conditions:
disease progression, fall, gait abnormal,
intermittent pyrexia, nodule, rigors, sudden death
Hepatobiliary disorders:
hyperbilirubinemia, cholecystitis, acute cholecystitis, hepatic failure
Immune system disorders:
hypersensitivity
Infections and infestations
infection bacteremia, central line infection, clostridial infection, ear
infection,
Enterobacter
sepsis, fungal infection, herpes viral infection NOS, influenza, kidney
infection,
Klebsiella
sepsis, lobar pneumonia, localized infection, oral infection,
Pseudomonas
infection, septic shock, sinusitis acute, sinusitis,
Staphylococcal
infection, urosepsis
Injury, poisoning and procedural complications:
femur fracture, transfusion reaction, cervical
vertebral fracture, femoral neck fracture, fractured pelvis, hip fracture, overdose, post procedural
hemorrhage, rib fracture, road traffic accident, spinal compression fracture
Investigations:
blood creatinine increased, hemoglobin decreased, liver function tests abnormal,
troponin I increased
Metabolism and nutrition disorders:
dehydration, gout, hypernatremia, hypoglycemia
Musculoskeletal and connective tissue disorders:
arthritis, arthritis aggravated, gouty arthritis,
neck pain, chondrocalcinosis pyrophosphate
Neoplasms benign, malignant and unspecified:
acute leukemia, acute myeloid leukemia,
bronchoalveolar carcinoma, lung cancer metastatic, lymphoma, prostate cancer metastatic
Nervous system disorders:
cerebrovascular accident, aphasia, cerebellar infarction, cerebral
infarction, depressed level of consciousness, dysarthria, migraine, spinal cord compression,
subarachnoid hemorrhage, transient ischemic attack
Psychiatric disorders:
confusional state
Renal and urinary disorders:
renal failure, hematuria, renal failure acute, azotemia, calculus
ureteric, renal mass
Reproductive system and breast disorders:
pelvic pain
Respiratory, thoracic and mediastinal disorders:
bronchitis, chronic obstructive airways
disease exacerbated, respiratory failure, dyspnea exacerbated, interstitial lung disease, lung
infiltration, wheezing
Skin and subcutaneous tissue disorders:
acute febrile neutrophilic dermatosis
Vascular system disorders:
deep vein thrombosis, hypotension, aortic disorder, ischemia,
thrombophlebitis superficial, thrombosis
6.3 Clinical Trials Experience in Mantle Cell Lymphoma
The overall safety profile of Revlimid in patients with mantle cell lymphoma is based on data
from 254 patients from a Phase II randomised controlled study MCL-002.
Additionally, ADRs from supportive study MCL-001 have been included in table 4.
The serious adverse reactions observed more frequently in Study MCL-002 (with a difference of
at least 2 percentage points) in the lenalidomide arm compared with the control arm were
Neutropenia (3.6%)
Pulmonary embolism (3.6%)
Diarrhoea (3.6%)
The most frequently observed adverse reactions which occurred more frequently in the
lenalidomide arm compared with the control arm in Study MCL-002 were neutropenia (50.9%),
anemia
(28.7%), diarrhoea (22.8%), fatigue (21.0%), constipation (17.4%), pyrexia (16.8%), and
rash (including dermatitis allergic) (16.2%).
In study MCL-002 there was overall an apparent increase in early (within 20 weeks) deaths.
Patients with high tumour burden at baseline are at increased risk of early death, 16/81 (20%) early
deaths in the lenalidomide arm and 2/28 (7%) early deaths in the control arm. Within 52 weeks
corresponding figures were 32/81 (39.5%) and 6/28 (21%).
During treatment cycle 1, 11/81 (14%) patients with high tumour burden were withdrawn from
therapy in the lenalidomide arm vs. 1/28 (4%) in the control group. The main reason for treatment
withdrawal for patients with high tumour burden during treatment cycle 1 in the lenalidomide arm
was adverse events, 7/11 (64%). High tumour burden was defined as at least one lesion ≥5 cm in
diameter or 3 lesions ≥3 cm.
Table 5:
ADRs reported in clinical trials in patients with mantle cell lymphoma treated
with lenalidomide
System Organ
Class
/ Preferred
Term
All ADRs/Frequency
Grade 3−4 ADRs/Frequency
Infections and
Infestations
Very Common
Bacterial, viral and fungal
infections (including opportunistic
infections)
, Nasopharyngitis,
Pneumonia
Common
Sinusitis
Common
Bacterial, viral and fungal infections
(including opportunistic infections)
Pneumonia
Neoplasms
Benign,
Malignant and
Unspecified
(incl cysts and
polyps)
Common
Tumour flare reaction
Common
Tumour flare reaction, Squamous skin
cancer
◊,
Basal cell carcinoma
Blood and
Lymphatic
System
Disorders
Very Common
Thrombocytopenia, Neutropenia
Leucopenias
, Anemia
Common
Febrile neutropenia
Very Common
Thrombocytopenia, Neutropenia
Anemia
Common
Febrile neutropenia
, Leucopenias
Metabolism
and Nutrition
Disorders
Very
Common
Decreased appetite, Weight
decreased, Hypokalaemia
Common
Dehydration
Common
Dehydration
, Hyponatraemia,
Hypocalcaemia
Psychiatric
Disorders
Common
Insomnia
Nervous
System
Disorders
Common
Dysgeuesia, Headache, neuropathy
peripheral
Common
Peripheral sensory neuropathy,
Lethargy
Ear and
Labyrinth
Disorders
Common
Vertigo
Cardiac
Disorders
Common
Myocardial infarction (including
acute)
, Cardiac failure
Vascular
Disorders
Common
Hypotension
Common
Deep vein thrombosis
, pulmonary
embolism
, Hypotension
Respiratory,
Thoracic and
Mediastinal
Disorders
Very
Common
Dyspnoea
Common
Dyspnoea
System Organ
Class
/ Preferred
Term
All ADRs/Frequency
Grade 3−4 ADRs/Frequency
Gastrointestina
l Disorders
Very
Common
Diarrhoea
, Nausea
, Vomiting
Constipation
Common
Abdominal pain
Common
Diarrhoea
, Abdominal pain
Constipation
Skin and
Subcutaneous
Tissue
Disorders
Very
Common
Rashes (including dermatitis
allergic), Pruritus
Common
Night sweats, Dry skin
Common
Rashes
Musculoskeleta
l and
Connective
Tissue
Disorders
Very
Common
Muscle spasms, Back pain
Common
Arthralgia, Pain in extremity,
Muscular weakness
Common
Back pain, Muscular weakness
Arthralgia, Pain in extremity
Renal and
Urinary
Disorders
Common
Renal failure
General
Disorders and
Administration
Site Conditions
Very
Common
Fatigue, Asthenia
, Peripheral
oedema, Influenza like illness
syndrome (including pyrexia
cough)
Common
Chills
Common
Pyrexia
, Asthenia
, Fatigue
Adverse events reported as serious in mantle cell lymphoma clinical trials
Algorithm applied for mantle cell lymphoma:
Mantle cell lymphoma controlled Phase II study
All treatment-emergent adverse events with ≥ 5% of subjects in lenalidomide arm and at least 2% difference
in proportion between lenalidomide and control arm
All treatment-emergent grade 3 or 4 adverse events in ≥1% of subjects in lenalidomide arm and at least 1.0%
difference in proportion between lenalidomide and control arm
All Serious treatment-emergent adverse events in ≥1% of subjects in lenalidomide arm and at least 1.0%
difference in proportion between lenalidomide and control arm
Mantle cell lymphoma single arm Phase II study
All treatment-emergent adverse events with ≥ 5% of subjects
All grade 3 or 4 treatment-emergent adverse events reported in 2 or more subjects
All Serious treatment-emergent adverse events reported in 2 or more subjects
Tabulated summary of post-marketing adverse reactions
In addition to the above adverse reactions identified from the pivotal clinical trials, the following
table is derived from data gathered from post-marketing data.
Table 6: ADRs reported in post-marketing use in patients treated with lenalidomide
System Organ Class/
Preferred Term
All ADRs/Frequency
Grade 3−4
ADRs/Frequency
Infections and
Infestations
Not Known
Viral infections, including herpes
zoster and hepatitis B virus
reactivation
Not Known
Viral infections, including
herpes zoster and hepatitis B
virus reactivation
Neoplasms Benign,
Malignant and
Unspecified (incl
cysts and polyps)
Rare
Tumour lysis syndrome
Blood and
Lymphatic System
Disorders
Not Known
Acquired haemophilia
Immune System
Disorders
Not Known
Solid organ transplant rejection
Endocrine Disorders
Common
Hyperthyroidism
Respiratory,
Thoracic and
Mediastinal
Disorders
Not Known
Interstitial pneumonitis
Gastrointestinal
Disorders
Not Known
Pancreatitis, Gastrointestinal
perforation (including
diverticular, intestinal and
large intestine perforations)
Hepatobiliary
Disorders
Not Known
Acute hepatic failure, Hepatitis toxic,
Cytolytic hepatitis, Cholestatic
hepatitis, Mixed cytolytic/cholestatic
hepatitis
Not Known
Acute hepatic failure,
Hepatitis toxic^
Skin and
Subcutaneous Tissue
Disorders
Uncommon
Angioedema
Rare
Stevens-Johnson Syndrome,
Toxic epidermal necrolysis
Not Known
Leukocytoclastic vasculitis,
Drug Reaction with
Eosinophilia and Systemic
Symptoms
Reporting of suspected adverse reactions
Reporting suspected adverse reactions after authorisation of the medicinal product is
important. It allows continued monitoring of the benefit/risk balance of the medicinal
product. Any suspected adverse events should be reported to the Ministry of Health
according to the National Regulation by using an online form
/https://sideeffects.health.gov.il and additionally emailed to the Registration Holder’s
Patient Safety Unit at: drugsafety@neopharmgroup.com
7.
DRUG INTERACTIONS
7.1 Digoxin
When digoxin was co-administered with multiple doses of REVLIMID (10 mg/day) the digoxin
and AUC
0-∞
were increased by 14%. Periodic monitoring of digoxin plasma levels, in
accordance with clinical judgment and based on standard clinical practice in patients receiving
this medication, is recommended during administration of REVLIMID.
7.2
Warfarin
Co-administration of multiple dose REVLIMID (10 mg) with single dose warfarin (25 mg) had
no effect on the pharmacokinetics of total lenalidomide or R- and S-warfarin. Expected changes
in laboratory assessments of PT and INR were observed after warfarin administration, but these
changes were not affected by concomitant REVLIMID administration. It is not known whether
there is an interaction between dexamethasone and warfarin. Close monitoring of PT and INR is
recommended in multiple myeloma patients taking concomitant warfarin.
7.3 Concomitant Therapies That May Increase the Risk of Thrombosis
Erythropoietic agents, or other agents that may increase the risk of thrombosis, such as estrogen
containing therapies, should be used with caution after making a benefit-risk assessment in
patients receiving REVLIMID [
see Warnings and Precautions (5.4)
8.
USE IN SPECIFIC POPULATIONS
8.1 Pregnancy
Pregnancy exposure registry
There is a pregnancy exposure registry that monitors pregnancy outcomes in females exposed to
REVLIMID during pregnancy as well as female partners of male patients who are exposed to
REVLIMID. This registry is also used to understand the root cause for the pregnancy.
Risk Summary
Based on the mechanism of action [see Clinical Pharmacology (12.1)] and findings from animal
studies [see Data], REVLIMID can cause embryo-fetal harm when administered to a pregnant
female and is contraindicated during pregnancy [see Boxed Warning, Contraindications (4.1),
and Use in Specific Populations (5.1)].
REVLIMID is a thalidomide analogue. Thalidomide is a human teratogen, inducing a high
frequency of severe and life-threatening birth defects such as
amelia (absence of limbs),
phocomelia (short limbs), hypoplasticity of the bones, absence of bones, external ear abnormalities
(including anotia, micropinna, small or absent external auditory
canals), facial palsy, eye
abnormalities (anophthalmos, microphthalmos), and congenital heart defects. Alimentary tract,
urinary tract, and genital malformations have also been documented and mortality at or shortly
after birth has been reported in about 40% of infants.
Lenalidomide caused thalidomide-type limb defects in monkey offspring. If this drug is used
during pregnancy, or if the patient becomes pregnant while taking this drug, the patient should be
apprised of the potential hazard to a fetus.
If pregnancy does occur during treatment, immediately discontinue the drug. Under these
conditions, refer patient to an obstetrician/gynecologist experienced in reproductive toxicity for
further evaluation and counseling. Any suspected fetal exposure to REVLIMID must be reported
to the attending physician and Neopharm.
The estimated background risk of major birth defects and miscarriage for the indicated
population is unknown.
Data
Animal data
embryo-fetal
developmental
toxicity
study
monkeys,
teratogenicity,
including
thalidomide-like limb defects, occurred in offspring when pregnant monkeys received oral
lenalidomide during organogenesis. Exposure (AUC) in monkeys at the lowest dose was 0.17
times the human exposure at the maximum recommended human dose (MRHD) of 25 mg. Similar
studies in pregnant rabbits and rats at 20 times and 200 times the MRHD respectively, produced
embryo lethality in rabbits and no adverse reproductive effects in rats.
pre-
post-natal
development
study
rats,
animals
received
lenalidomide
from
organogenesis through lactation. The study revealed a few adverse effects on the offspring of
female rats treated with lenalidomide at doses up to 500 mg/kg (approximately 200 times the
human dose of 25 mg based on body surface area). The male offspring exhibited slightly delayed
sexual maturation and the female offspring had slightly lower body weight gains during gestation
when bred to male offspring. As with thalidomide, the rat model may not adequately address the
full spectrum of potential human embryo-fetal developmental effects for lenalidomide.
Following daily oral administration of lenalidomide from Gestation Day 7 through Gestation Day
20 in pregnant rabbits, fetal plasma lenalidomide concentrations were approximately 20-40% of
the maternal Cmax. Following a single oral dose to pregnant rats, lenalidomide was detected in
fetal plasma and tissues; concentrations of radioactivity in fetal tissues were generally lower than
those in maternal tissues. These data indicated that lenalidomide crossed the placenta.
8.2 Lactation
Risk summary
There is no information regarding the presence of lenalidomide in human milk, the effects of
REVLIMID on the breastfed infant, or the effects of REVLIMID on milk production. Because
many drugs are excreted in human milk and because of the potential for adverse reactions in
nursing infants from lenalidomide advice women not to breastfeed during treatment with
REVLIMID.
8.3 Females and males of reproductive potential
Pregnancy Testing
REVLIMID can cause fetal harm when administered during pregnancy [see Use in Specific
Populations (8.1)]. Verify the pregnancy status of females of reproductive potential prior to
initiating REVLIMID therapy and during therapy. Advise females of reproductive potential that
they must avoid pregnancy 4 weeks before therapy, while taking REVLIMID, during dose
interruptions and for at least 4 weeks after completing therapy.
Females of reproductive potential must have 2 negative pregnancy tests before initiating
REVLIMID. The first test should be performed within 10-14 days, and the second test within 24
hours prior to prescribing REVLIMID. Once treatment has started and during dose interruptions,
pregnancy testing for females of reproductive potential should occur weekly during the first 4
weeks of use, then pregnancy testing should be repeated every 4 weeks in females with regular
menstrual cycles. If menstrual cycles are irregular, the pregnancy testing should occur every 2
weeks. Pregnancy testing and counseling should be performed if a patient misses her period or if
there is any abnormality in her menstrual bleeding. REVLIMID treatment must be discontinued
during this evaluation.
Contraception
Females
Females of reproductive potential must commit either to abstain continuously from heterosexual
sexual intercourse or to use 2 methods of reliable birth control simultaneously: one highly
effective form of contraception – tubal ligation, IUD, hormonal (birth control pills, injections,
hormonal patches, vaginal rings, or implants), or partner’s vasectomy, and 1 additional effective
contraceptive method – male latex or synthetic condom, diaphragm, or cervical cap.
Contraception must begin 4 weeks prior to initiating treatment with REVLIMID, during therapy,
during dose interruptions, and continuing for 4 weeks following discontinuation of REVLIMID
therapy. Reliable contraception is indicated even where there has been a history of infertility,
unless due to hysterectomy. Females of reproductive potential should be referred to a qualified
provider of contraceptive methods, if needed.
Males
Lenalidomide is present in the semen of males who take REVLIMID. Therefore, males must
always use a latex or synthetic condom during any sexual contact with females of reproductive
potential while taking REVLIMID and for up to 4 weeks after discontinuing REVLIMID, even if
they have undergone a successful vasectomy.
Male patients taking REVLIMID must not donate sperm.
8.4 Pediatric use
Safety and effectiveness have not been established in pediatric patients.
8.5 Geriatric use
MM in combination: Overall, of the 1613 patients in the NDMM study who received study
treatment, 94% (1521 /1613) were 65 years of age or older, while 35% (561/1613) were over 75
years of age. The percentage of patients over age 75 was similar between study arms (Rd
Continuous: 33%; Rd18: 34%; MPT: 33%). Overall, across all treatment arms, the frequency in
most of the AE categories (eg, all AEs, grade 3/4 AEs, serious AEs) was higher in older (> 75
years of age) than in younger (≤ 75 years of age) subjects. Grade 3 or 4 AEs in the General
Disorders and Administration Site Conditions body system were consistently reported at a higher
frequency (with a difference of at least 5%) in older subjects than in younger subjects across all
treatment arms. Grade 3 or 4 TEAEs in the Infections and Infestations, Cardiac Disorders
(including cardiac failure and congestive cardiac failure), Skin and Subcutaneous Tissue
Disorders, and Renal and Urinary Disorders (including renal failure) body systems were also
reported slightly, but consistently, more frequently (<5% difference), in older subjects than in
younger subjects across all treatment arms. For other body systems (e.g., Blood and Lymphatic
System Disorders, Infections and Infestations, Cardiac Disorders, Vascular Disorders), there was
a less consistent trend for increased frequency of grade 3/4 AEs in older vs younger subjects
across all treatment arms Serious AEs were generally reported at a higher frequency in the older
subjects than in the younger subjects across all treatment arms.
MM maintenance therapy: Overall, 10% (106/1018) of patients were 65 years of age or older,
while no patients were over 75 years of age. Grade 3 or 4 AEs were higher in the REVLIMID
arm (more than 5% higher) in the patients 65 years of age or older versus younger patients. The
frequency of Grade 3 or 4 AEs in the Blood and Lymphatic System Disorders were higher in the
REVLIMID arm (more than 5% higher) in the patients 65 years of age or older versus younger
patients. There were not a sufficient number of patients 65 years of age or older in REVLIMID
maintenance studies who experienced either a serious AE, or discontinued therapy due to an AE
to determine whether elderly patients respond relative to safety differently from younger
patients.
MM after at least one prior therapy: Of the 703 MM patients who received study treatment in
Studies 1 and 2, 45% were age 65 or over while 12% of patients were age 75 and over. The
percentage of patients age 65 or over was not significantly different between the
REVLIMID/dexamethasone and placebo/dexamethasone groups. Of the 353 patients who
received REVLIMID/dexamethasone, 46% were age 65 and over. In both studies, patients > 65
years of age were more likely than patients ≤ 65 years of age to experience DVT, pulmonary
embolism, atrial fibrillation, and renal failure following use of REVLIMID. No differences in
efficacy were observed between patients over 65 years of age and younger patients.
Of the 148 patients with del 5q MDS enrolled in the major study, 38% were age 65 and over, while
33% were age 75 and over. Although the overall frequency of adverse events (100%) was the same
in patients over 65 years of age as in younger patients, the frequency of serious adverse events was
higher in patients over 65 years of age than in younger patients (54% vs. 33%). A greater
proportion of patients over 65 years of age discontinued from the clinical studies because of
adverse events than the proportion of younger patients (27% vs.16%). No differences in efficacy
were observed between patients over 65 years of age and younger patients.
Of the 134 patients with MCL enrolled in the MCL trial, 63% were age 65 and over, while 22%
of patients were age 75 and over. The overall frequency of adverse events was similar in patients
over 65 years of age and in younger patients (98% vs. 100%). The overall incidence of grade 3
and 4 adverse events was also similar in these 2 patient groups (79% vs. 78%, respectively). The
frequency of serious adverse events was higher in patients over 65 years of age than in younger
patients (55% vs. 41%). No differences in efficacy were observed between patients over 65 years
of age and younger patients.
Because elderly patients are more likely to have decreased renal function, care should be taken in
dose selection and it would be prudent to monitor renal function.
8.6 Renal Impairment
Adjust the starting dose of REVLIMID based on the creatinine clearance value and for patients
on dialysis [
see Dosage and Administration (2.4)
.
9.
OVERDOSAGE
There is no specific experience in the management of lenalidomide overdose in patients;
although in dose-ranging studies, some patients were exposed to up to 150 mg and in single-dose
studies, some patients were exposed to up to 400 mg.
In studies, the dose-limiting toxicity was essentially hematological. In the event of overdose,
supportive care is advised.
10.
DESCRIPTION
REVLIMID, a thalidomide analogue, is an immunomodulatory agent with antiangiogenic and
antineoplastic properties. The chemical name is 3-(4-amino-1-oxo 1,3-dihydro-2
H
-isoindol-2-yl)
piperidine-2,6-dione and it has the following chemical structure:
3-(4-amino-1-oxo 1,3-dihydro-2
H
-isoindol-2-yl) piperidine-2,6-dione
The empirical formula for lenalidomide is C
and the gram molecular weight is 259.3.
Lenalidomide is an off-white to pale-yellow solid powder. It is soluble in organic solvent/water
mixtures, and buffered aqueous solvents. Lenalidomide is more soluble in organic solvents and
low pH solutions. Solubility was significantly lower in less acidic buffers, ranging from about
0.4 to 0.5 mg/ml. Lenalidomide has an asymmetric carbon atom and can exist as the optically
active forms S(-) and R(+), and is produced as a racemic mixture with a net optical rotation of
zero.
REVLIMID is available in 2.5 mg, 5 mg, 7.5mg, 10 mg, 15 mg, 20 mg and 25 mg hard capsules
for oral administration. Each hard capsule contains lenalidomide as the active ingredient and the
following inactive ingredients: lactose anhydrous, microcrystalline cellulose, croscarmellose
sodium, and magnesium stearate. The 5 mg and 25 mg hard capsule shell contains gelatin,
titanium dioxide and black ink. The 2.5 mg and 10 mg hard capsule shell contains gelatin, FD&C
blue #2, yellow iron oxide, titanium dioxide and black ink. The 15 mg hard capsule shell
contains gelatin, FD&C blue #2, titanium dioxide and black ink. The 20 mg hard capsule shell
contains gelatin, FD&C blue #2, yellow iron oxide, titanium dioxide and black ink.
11.
CLINICAL PHARMACOLOGY
11.1 Mechanism of Action
Lenalidomide is an analogue of thalidomide with immunomodulatory, antiangiogenic, and
antineoplastic properties. Lenalidomide inhibits proliferation and induces apoptosis of certain
hematopoietic tumor cells including multiple myeloma, mantle cell lymphoma, and del (5q)
myelodysplastic syndromes
in vitro
. Lenalidomide causes a delay in tumor growth in some
in vivo
nonclinical hematopoietic tumor models including multiple myeloma.
Immunomodulatory
properties of lenalidomide include activation of T cells and natural killer (NK) cells, increased
numbers of NKT cells, and inhibition of pro-inflammatory cytokines (e.g., TNF-α and IL-6) by
monocytes. In multiple myeloma cells, the combination of lenalidomide and dexamethasone
synergizes the inhibition of cell proliferation and the induction of apoptosis.
Lenalidomide binds directly to cereblon, a component of a cullin ring E3 ubiquitin ligase enzyme
complex that includes deoxyribonucleic acid (DNA) damage-binding protein 1(DDB1), cullin 4
(CUL4), and regulator of cullins 1 (Roc1). In the presence of lenalidomide, cereblon binds
substrate proteins Aiolos and Ikaros which are lymphoid transcriptional factors, leading to their
ubiquitination and subsequent degradation resulting in cytoxic and immunomodulatory effects.
11.2 Pharmacodynamics
The effect of lenalidomide on the QTc interval was evaluated in 60 healthy male subjects in a
randomised, thorough QT study with placebo and positive controls. At a dose two times the
maximum recommended dose, lenalidomide does not prolong the QTc interval to any clinically
relevant extent. The largest upper bound of the 2-sided 90% CI for the mean differences between
lenalidomide and placebo was below 10 ms.
11.3 Pharmacokinetics
Absorption
Lenalidomide is rapidly absorbed following oral administration. Following single and multiple
doses of REVLIMID in patients with MM or MDS the maximum plasma concentrations
occurred between 0.5 and 6 hours post-dose. The single and multiple dose pharmacokinetic
disposition of lenalidomide is linear with AUC and C
values increasing proportionally with
dose. Multiple dosing at the recommended dose-regimen does not result in medicinal product
accumulation.
Systemic exposure (AUC) of lenalidomide in MM and MDS patients with normal or mild renal
function (CLcr
60 mL/min) is approximately 60% higher as compared to young healthy male
subjects.
Administration of a single 25 mg dose of REVLIMID with a high-fat meal in healthy subjects
reduces the extent of absorption, with an approximate 20% decrease in AUC and 50% decrease
in C
. In the trials where the efficacy and safety were established for REVLIMID, the
medicinal product was administered without regard to food intake. REVLIMID can be
administered with or without food.
Population pharmacokinetic analyses show that the oral absorption rate of lenalidomide in
patients with MCL is similar to that observed in patients with MM or MDS.
Distribution
In vitro (
C)-lenalidomide binding to plasma proteins is approximately 30%.
Lenalidomide is present in semen at 2 hours (1379 ng/ejaculate) and 24 hours (35 ng/ejaculate)
after the administration of REVLIMID 25 mg daily.
Elimination
Metabolism
Lenalidomide undergoes limited metabolism. Unchanged lenalidomide is the predominant
circulating component in humans. Two identified metabolites are hydroxy-lenalidomide and N-
acetyl-lenalidomide; each constitutes less than 5% of parent levels in circulation.
Excretion
Elimination is primarily renal. Following a single oral administration of [
C]-lenalidomide (25
mg) to healthy subjects, approximately 90% and 4% of the radioactive dose is eliminated within
ten days in urine and feces, respectively. Approximately 82% of the radioactive dose is excreted
as lenalidomide in the urine within 24 hours. Hydroxy-lenalidomide and N-acetyl-lenalidomide
represent 4.59% and 1.83% of the excreted dose, respectively. The renal clearance of
lenalidomide exceeds the glomerular filtration rate.
Specific Populations
Renal Impairment:
Eight subjects with mild renal impairment (creatinine clearance (CLcr) 50 to
79 mL/min calculated using Cockcroft-Gault), 9 subjects with moderate renal impairment (CLcr
30 to 49 mL/min), 4 subjects with severe renal impairment (CLcr < 30 mL/min), and 6 patients
with end stage renal disease (ESRD) requiring dialysis were administered a single 25 mg dose of
REVLIMID. Three healthy subjects of similar age with normal renal function (CLcr > 80 mL/min)
were also administered a single 25 mg dose of REVLIMID. As CLcr decreased, half-life increased
and drug clearance decreased linearly. Patients with moderate and severe impairment had a 3-fold
increase in half-life and a 66% to 75% decrease in drug clearance compared to healthy subjects.
Patients on hemodialysis (n=6) had an approximate 4.5-fold increase in half-life and an 80%
decrease in drug clearance compared to healthy subjects. Approximately 30% of the drug in body
was removed during a 4-hour hemodialysis session.
Adjust the starting dose of REVLIMID in patients with renal impairment based on the CLcr
value [see Dosage and Administration (2.4)].
Hepatic impairment:
Mild hepatic impairment (defined as total bilirubin > 1 to 1.5 times upper
limit normal (ULN) or any aspartate transaminase greater than ULN) did not influence the
disposition of lenalidomide. No pharmacokinetic data is available for patients with moderate to
severe hepatic impairment.
Other Intrinsic Factors:
Age (39 to 85 years), body weight (33 to 135 kg), sex, race, and type of
hematological malignancies (MM, MDS or MCL) did not have a clinically relevant effect on
lenalidomide clearance in adult patients.
Drug Interactions
Co-administration of a single dose or multiple doses of dexamethasone (40 mg) had no clinically
relevant effect on the multiple dose pharmacokinetics of REVLIMID (25 mg). Co-administration
of REVLIMID (25 mg) after multiple doses of a P-gp inhibitor such as quinidine (600 mg twice
daily) did not significantly increase the Cmax or AUC of lenalidomide. Co-administration of the
P-gp inhibitor and substrate temsirolimus (25 mg),with REVLIMID (25 mg) did not significantly
alter the pharmacokinetics of lenalidomide, temsirolimus, or sirolimus (metabolite of
temsirolimus). In vitro studies demonstrated that REVLIMID is a substrate of P-glycoprotein (P-
gp). REVLIMID is not a substrate of human breast cancer resistance protein (BCRP), multidrug
resistance protein (MRP) transporters MRP1, MRP2, or MRP3, organic anion transporters
(OAT) OAT1 and OAT3, organic anion transporting polypeptide 1B1 (OATP1B1), organic
cation transporters (OCT) OCT1 and OCT2, multidrug and toxin extrusion protein (MATE)
MATE1, and organic cation transporters novel (OCTN) OCTN1 and OCTN2. Lenalidomide is
not an inhibitor of P-gp, bile salt export pump (BSEP), BCRP, MRP2, OAT1, OAT3, OATP1B1,
OATP1B3, or OCT2. Lenalidomide does not inhibit or induce CYP450 isoenzymes. Also,
lenalidomide does not inhibit bilirubin glucuronidation formation in human liver microsomes
with UGT1A1 genotyped as UGT1A1*1/*1, UGT1A1*1/*28, and UGT1A1*28/*28
12. NONCLINICAL TOXICOLOGY
12.1 Carcinogenesis
Mutagenesis, Impairment of Fertility
Carcinogenicity studies with lenalidomide have not been conducted.
Lenalidomide was not mutagenic in the bacterial reverse mutation assay (Ames test) and did not
induce chromosome aberrations in cultured human peripheral blood lymphocytes, or mutations at
the thymidine kinase (tk) locus of mouse lymphoma L5178Y cells. Lenalidomide did not
increase morphological transformation in Syrian Hamster Embryo assay or induce micronuclei in
the polychromatic erythrocytes of the bone marrow of male rats.
A fertility and early embryonic development study in rats, with administration of lenalidomide
up to 500 mg/kg (approximately 200 times the human dose of 25 mg, based on body surface
area) produced no parental toxicity and no adverse effects on fertility.
12.
2 Reproductive and Developmental Toxicity
Lenalidomide had an embryocidal effect in rabbits at a dose of 50 mg/kg (approximately 120
times the human dose of 10 mg based on body surface area).
In an embryofetal developmental toxicity study in monkeys, teratogenicity, including
thalidomide-like limb defects, occurred in offspring when pregnant monkeys received oral
lenalidomide during organogenesis at doses approximately 0.17 times the maximum
recommended human dose (MRHD) of 25 mg, based on body surface area.
A pre- and post-natal development study in rats revealed few adverse effects on the offspring of
female rats treated with lenalidomide at doses up to 500 mg/kg (approximately 200 times the
human dose of 25 mg based on body surface area). The male offspring exhibited slightly delayed
sexual maturation and the female offspring had slightly lower body weight gains during gestation
when bred to male offspring.
13. CLINICAL STUDIES
13.1 Newly diagnosed multiple myeloma
Lenalidomide efficacy and safety have been evaluated in five phase III studies in newly
diagnosed multiple myeloma.
Newly diagnosed multiple myeloma
Lenalidomide maintenance in patients who have undergone ASCT
The efficacy and safety of lenalidomide maintenance was assessed in two phase 3 multicenter,
randomized, double-blind 2-arm, parallel group, placebo-controlled studies: CALGB 100104 and
IFM 2005-002.
CALGB 100104
Patients between 18 and 70 years of age with active MM requiring treatment and without prior
progression after initial therapy were eligible.
Patients were randomised 1:1 within 90-100 days after ASCT to receive either lenalidomide or
placebo maintenance. The maintenance dose was 10 mg once daily on days 1-28 of repeated 28-
day cycles (increased up to 15 mg once daily after 3 months in the absence of dose-limiting
toxicity), and treatment was continued until disease progression.
primary
efficacy
endpoint
study
progression
free
survival
(PFS)
from
randomisation to the date of progression or death, whichever occurred first; the study was not
powered for the overall survival endpoint. In total 460 patients were randomised: 231 patients to
Lenalidomide and 229 patients to placebo. The demographic and disease-related characteristics
were balanced across both arms.
The study was unblinded upon the recommendations of the data monitoring committee after
surpassing the threshold for a preplanned interim analysis of PFS. After unblinding, patients in the
placebo arm were allowed to cross over to receive lenalidomide before disease progression.
The results of PFS at unblinding, following a preplanned interim analysis, using a cut-off of 17
December 2009 (15.5 months follow up) showed a 62% reduction in risk of disease progression
or death favoring lenalidomide (HR = 0.38; 95% CI 0.27, 0.54; p <0.001). The median overall PFS
was 33.9 months (95% CI NE, NE) in the lenalidomide arm versus 19.0 months (95% CI 16.2,
25.6) in the placebo arm.
The PFS benefit was observed both in the subgroup of patients with CR and in the subgroup of
patients who had not achieved a CR.
Table 7: Summary of overall efficacy data
Lenalidomide
(N = 231)
Placebo
(N = 229)
Investigator-assessed PFS
Median
PFS time, months (95% CI)
56.9
(41.9, 71.7)
29,4
(20.7, 35.5)
HR [95% CI]
; p-valued
0.61
(0.48, 0.76); <0.001
PFS2
e
Median
PFS2 time, months (95% CI)b
80.2
(63.3, 101.8)
52.8
(41.3, 64.0)
HR [95% CI]c ; p-valued
0.61
(0.48, 0.78); <0.001
Overall survival
Median
OS time, months (95% CI)
111.0
(101.8, NE)
84.2
(71.0, 102.7)
8-year survival rate, % (SE)
60.9 (3.78)
44.6 (3.98)
HR [95% CI]
; p-value
0.61
(0.46, 0.81); <0.001
Follow-up
Median
(min, max), months: all surviving patients
81.9
(0.0, 119.8)
81.0
(4.1, 119.5)
CI = confidence interval; HR = hazard ratio; max = maximum; min = minimum; NE = not estimable; OS = overall survival; PFS = progression-
free survival;
The median is based on the Kaplan-Meier estimate.
The 95% CI about the median.
Based on Cox proportional hazards model comparing the hazard functions associated with the indicated treatment arms.
The p-value is based on the unstratified log-rank test of Kaplan-Meier curve differences between the indicated treatment arms.
Exploratory endpoint (PFS2). Lenalidomide received by subjects in the placebo arm who crossed over prior to PD upon study unblinding was
not considered as a second-line therapy.
Median follow-up post-ASCT for all surviving subjects.
Data cuts:
17 Dec 2009 and 01 Feb 2016
IFM 2005-02
Patients aged < 65 years at diagnosis who had undergone ASCT and had achieved at least a stable
disease response at the time of hematologic recovery were eligible. Patients were randomised 1:1
to receive either lenalidomide or placebo maintenance (10 mg once daily on days 1-28 of repeated
28-day cycles increased up to 15 mg once daily after 3 months in the absence of dose-limiting
toxicity) following 2 courses of lenalidomide consolidation (25 mg/day, days 1-21 of a 28-day
cycle). Treatment was to be continued until disease progression.
The primary endpoint was PFS defined from randomisation to the date of progression or death,
whichever occurred first; the study was not powered for the overall survival endpoint. In total 614
patients were randomised: 307 patients to lenalidomide and 307 patients to placebo.
The study was unblinded upon the recommendations of the data monitoring committee after
surpassing the threshold for a preplanned interim analysis of PFS. After unblinding, patients
receiving placebo were not crossed over to lenalidomide therapy prior to progressive disease. The
lenalidomide arm was discontinued, as a proactive safety measure, after observing an imbalance
of SPMs (see Section 4.4).
The results of PFS at unblinding, following a preplanned interim analysis, using a cut-off of 7 July
2010 (31.4 months follow up) showed a 48% reduction in risk of disease progression or death
favoring lenalidomide (HR = 0.52; 95% CI 0.41, 0.66; p <0.001). The median overall PFS was
40.1 months (95% CI 35.7, 42.4) in the lenalidomide arm versus 22.8 months (95% CI 20.7, 27.4)
in the placebo arm.
The PFS benefit was less in the subgroup of patients with CR than in the subgroup of patients who
had not achieved a CR.
The updated PFS, using a cut-off of 1 February 2016 (96.7 months follow up) continues to show
a PFS advantage: HR = 0.57 (95% CI 0.47, 0.68; p < 0.001). The median overall PFS was 44.4
months (39.6, 52.0) in the lenalidomide arm versus 23.8 months (95% CI 21.2, 27.3) in the placebo
arm. For PFS2, the observed HR was 0.80 (95% CI 0.66, 0.98; p = 0.026) for lenalidomide versus
placebo. The median overall PFS2 was 69.9 months (95% CI 58.1, 80.0) in the lenalidomide arm
versus 58.4 months (95% CI 51.1, 65.0) in the placebo arm. For OS, the observed HR was 0.90:
(95% CI 0.72, 1.13; p = 0.355) for lenalidomide versus placebo. The median overall survival time
was 105.9 months (95% CI 88.8, NE) in the lenalidomide arm versus 88.1 months (95% CI 80.7,
108.4) in the placebo arm.
Lenalidomide in combination with dexamethasone in patients
who are not candidates for stem
cell transplantation:
The safety and efficacy of lenalidomide was assessed in a Phase III, multicenter, randomised, open-
label, 3-arm study (MM-020) of
patients who were
at least 65 years of age or older or, if younger
than 65 years of age, were not candidates for stem cell transplantation because they declined to
undergo stem cell transplantation or stem cell transplantation is not available to the patient due to
cost or other reason. The study (MM-020) compared lenalidomide and dexamethasone (Rd) given
for 2 different durations of time (i.e., until progressive disease [Arm Rd] or for up to eighteen 28-
day cycles [72 weeks, Arm Rd18]) to that of melphalan, prednisone and thalidomide (MPT) for a
maximum of twelve 42-day cycles (72 weeks). Patients
were randomised (1:1:1) to 1 of 3
treatment arms. Patients were stratified at randomisation by age (≤75 versus >75 years), stage (ISS
Stages I and II versus Stage III), and country.
Patients in the Rd and Rd18 arms took lenalidomide 25 mg once daily on days 1 to 21 of 28-day
cycles according to protocol arm. Dexamethasone 40 mg was dosed once daily on days 1, 8, 15,
and 22 of each 28-day cycle. Initial dose and regimen for Rd and Rd18 were adjusted according
to age and renal function. Patients >75 years received a dexamethasone dose of 20 mg once daily
on days 1, 8, 15, and 22 of each 28-day cycle. All patients received prophylactic anticoagulation
(low molecular weight heparin, warfarin, heparin, low-dose aspirin) during the study.
The primary efficacy endpoint in the study was progression free survival (PFS). In total 1623
patients were enrolled into the study, with 535 patients randomised to Rd, 541 patients
randomised to Rd18 and 547 patients randomised to MPT. The demographics and disease-
related baseline characteristics of the patients were well balanced in all 3 arms. In general, study
subjects had advanced-stage disease: of the total study population, 41% had ISS stage III, 9%
had severe renal insufficiency (creatinine clearance [CLcr] < 30 mL/min). The median age was
73 in the 3 arms.
In an updated analysis of PFS, PFS2, OS and DR where the median follow up time for all
surviving subjects was 45.5 months, the results of the study are presented in Table 8:
Table 8: Summary of overall efficacy data
Rd
(N = 541)
Rd18
(N = 540)
MPT
(N = 542)
Investigator-assessed PFS
(months)
Median
PFS time, months (95% CI)
26.0 (20.7,
29.7)
21.0 (19.7,
22.4)
21.9 (19.8,
23.9)
HR [95% CI]
; p-value
Rd vs MPT
0.69 (0.59, 0.80); <0.001
Rd vs Rd18
0.71 (0.61, 0.83); <0.001
Rd18 vs MPT
0.99 (0.86, 1.14); <0.001
PFS2
e
(months)
Median
PFS time, months (95% CI)
42.9 (38.1,
47.4)
40.0 (36.2, 44.2) 35.0 (30.4, 37.8)
HR [95% CI]
; p-value
Rd vs MPT
0.74 (0.63, 0.86); <0.001
Rd vs Rd18
0.92 (0.78, 1.08); 0.316
Rd18 vs MPT
0.80 (0.69, 0.93); 0.004
Overall survival (months)
Median
OS time, months (95% CI)
58.9 (56.0, NE)
56.7 (50.1, NE)
48.5 (44.2,
52.0)
HR [95% CI]
; p-value
Rd vs MPT
0.75 (0.62, 0.90); 0.002
Rd vs Rd18
0.91 (0.75, 1.09); 0.305
Rd18 vs MPT
0.83 (0.69, 0.99); 0.034
Follow-up (months)
Median
(min, max): all patients
40.8 (0.0, 65.9)
40.1 (0.4, 65.7)
38.7 (0.0, 64.2)
Myeloma response
n (%)
81 (15.1)
77 (14.2)
51 (9.3)
VGPR
152 (28.4)
154 (28.5)
103 (18.8)
169 (31.6)
166 (30.7)
187 (34.2)
Overall response: CR, VGPR, or PR
402 (75.1)
397 (73.4)
341 (62.3)
Duration of response
(months)
Median
(95% CI)
35.0 (27.9,
43.4)
22.1 (20.3,
24.0)
22.3 (20.2,
24.9)
AMT = antimyeloma therapy; CI = confidence interval; CR = complete response; d = low-dose dexamethasone; HR = hazard ratio;
IMWG = International Myeloma Working Group; IRAC = Independent Response Adjudication Committee; M = melphalan; max = maximum;
min = minimum; NE = not estimable; OS = overall survival; P = prednisone; PFS = progression-free survival; PR = partial response;
R = lenalidomide; Rd = Rd given until documentation of progressive disease; Rd18 = Rd given for
18 cycles; SE = standard error;
T = thalidomide; VGPR = very good partial response; vs = versus.
The median is based on the Kaplan-Meier estimate.
The 95% CI about the median.
Based on Cox proportional hazards model comparing the hazard functions associated with the indicated treatment arms.
The p-value is based on the unstratified log-rank test of Kaplan-Meier curve differences between the indicated treatment arms.
Exploratory endpoint (PFS2)
The median is the univariate statistic without adjusting for censoring.
Best assessment of adjudicated response during the treatment phase of the study (for definitions of each response category, Data cutoff date =
24 May 2013).
data cut 24 May 2014
Lenalidomide in combination with melphalan and prednisone followed by maintenance
therapy in patients
not eligible for transplant
The safety and efficacy of lenalidomide was assessed in a Phase III multicenter, randomised
double blind 3 arm study (MM-015) of patients who were 65 years or older and had a serum
creatinine < 2.5 mg/dL. The study compared lenalidomide in combination with melphalan and
prednisone
(MPR)
with
without
lenalidomide
maintenance
monotherapy
until
disease
progression, to that of melphalan and prednisone for a maximum of 9 cycles. Patients were
randomised in a 1:1:1 ratio to one of 3 treatment arms. Patients were stratified at randomisation
by age (
75 vs. > 75 years) and stage (ISS; Stages I and II vs. stage III).
This study investigated the use of combination therapy of MPR (melphalan 0.18 mg/kg orally on
days 1-4 of repeated 28-day cycles; prednisone 2 mg/kg orally on days 1-4 of repeated 28-day
cycles; and lenalidomide 10 mg/day orally on days 1-21 of repeated 28-day cycles) for induction
therapy, up to 9 cycles. Patients who completed 9 cycles or who were unable to complete 9 cycles
due to intolerance proceeded to maintenance monotherapy starting with lenalidomide 10 mg orally
on days 1-21 of repeated 28-day cycles until disease progression.
The primary efficacy endpoint in the study was progression free survival (PFS). In total 459
patients were enrolled into the study, with 152 patients randomised to MPR+R, 153 patients
randomised to MPR+p and 154 patients randomised to MPp+p. The demographics and disease-
related baseline characteristics of the patients were well balanced in all 3 arms; notably,
approximately 50% of the patients enrolled in each arm had the following characteristics; ISS
Stage III, and creatinine clearance < 60 mL/min. The median age was 71 in the MPR+R and
MPR+p arms and 72 in the MPp+p arm.
In an analysis of PFS, PFS2, OS using a cut off of April 2013 where the median follow up time
for all surviving subjects was 62.4 months, the results of the study are presented in Table 9:
Table 9: Summary of overall efficacy data
MPR+R
(N = 152)
MPR+p
(N = 153)
MPp +p
(N = 154)
Investigator-assessed PFS
(months)
Median
PFS time, months (95% CI)
27.4 (21.3,
35.0)
14.3 (13.2,
15.7)
13.1 (12.0,
14.8)
HR [95% CI]; p-value
MPR+R vs MPp+p
0.37 (0.27, 0.50); <0.001
MPR+R vs MPR+p
0.47 (0.35, 0.65); <0.001
MPR+p vs MPp +p
0.78 (0.60, 1.01); 0.059
PFS2
(months)
Median
PFS time, months (95% CI)
39.7 (29.2,
48.4)
27.8 (23.1, 33.1) 28.8 (24.3, 33.8)
HR [95% CI]; p-value
MPR+R vs MPp+p
0.70 (0.54, 0.92); 0.009
MPR+R
(N = 152)
MPR+p
(N = 153)
MPp +p
(N = 154)
MPR+R vs MPR+p
0.77 (0.59, 1.02); 0.065
MPR+p vs MPp +p
0.92 (0.71, 1.19); 0.051
Overall survival (months)
Median
OS time, months (95% CI)
55.9 (49.1,
67.5)
51.9 (43.1,
60.6)
53.9 (47.3,
64.2)
HR [95% CI]; p-value
MPR+R vs MPp+p
0.95 (0.70, 1.29); 0.736
MPR+R vs MPR+p
0.88 (0.65, 1.20); 0.43
MPR+p vs MPp +p
1.07 (0.79, 1.45); 0.67
Follow-up (months)
Median (min, max): all patients
48.4 (0.8, 73.8)
46.3 (0.5, 71.9)
50.4 (0.5, 73.3)
Investigator-assessed Myeloma
response n (%)
30 (19.7)
17 (11.1)
9 (5.8)
90 (59.2)
99 ( 64.7)
75 (48.7)
Stable Disease (SD)
24 (15.8)
31 (20.3)
63 (40.9)
Response Not Evaluable (NE)
8 (5.3)
4 (2.6)
7 (4.5)
Investigator-assessed Duration of
response (CR+PR)
(months)
Median
(95% CI)
26.5 (19.4,
35.8)
12.4 (11.2,
13.9)
12.0 (9.4, 14.5)
CI = confidence interval; CR = complete response; HR = Hazard Rate; M = melphalan; NE = not estimable; OS = overall survival; p = placebo; P
= prednisone;
PD = progressive disease; PR = partial response; R = lenalidomide; SD = stable disease; VGPR = very good partial response.
ª The median is based on the Kaplan-Meier estimate
PFS2 (an exploratory endpoint) was defined for all patients (ITT) as time from randomisation to start of 3rd line antimyeloma therapy (AMT) or
death for all randomised patients
Supportive newly diagnosed multiple myeloma studies
An open-label, randomised, multicenter, Phase III study (ECOG E4A03) was conducted in 445
patients
with
newly
diagnosed
multiple
myeloma;
patients
were
randomised
lenalidomide/low
dose
dexamethasone
arm,
were
randomized
lenalidomide/standard dose dexamethasone arm. Patients randomised to the lenalidomide/standard
dose dexamethasone arm received lenalidomide 25 mg/day, days 1 to 21 every 28 days plus
dexamethasone 40 mg/day on days 1 to 4, 9 to 12, and 17 to 20 every 28 days for the first four
cycles.
Patients
randomised
lenalidomide/low
dose
dexamethasone
received
lenalidomide 25 mg/day, days 1 to 21 every 28 days plus low dose dexamethasone – 40 mg/day
on days 1, 8, 15, and 22 every 28 days. In the lenalidomide/low dose dexamethasone group, 20
patients (9.1%) underwent at least one dose interruption compared to 65 patients (29.3%) in the
lenalidomide/standard dose dexamethasone arm.
In a post-hoc analysis, lower mortality was observed in the lenalidomide/low dose dexamethasone
arm 6.8% (15/220) compared to the lenalidomide/standard dose dexamethasone arm 19.3%
(43/223), in the newly diagnosed multiple myeloma patient population, with a median follow up
of 72.3 weeks.
However, with a longer follow-up, the difference in overall survival in favour of lenalidomide/
low dose dexamethasone tends to decrease.
13.2 Multiple myeloma with at least one prior therapy
Two randomised studies (Studies 1 and 2) were conducted to evaluate the efficacy and safety of
REVLIMID. These multicenter, multinational, double-blind, placebo-controlled studies
compared REVLIMID plus oral pulse high-dose dexamethasone therapy to dexamethasone
therapy alone in patients with multiple myeloma who had received at least one prior treatment.
These studies enrolled patients with absolute neutrophil counts (ANC) ≥ 1000/mm
, platelet
counts ≥ 75,000/mm
, serum creatinine ≤ 2.5 mg/dL, serum SGOT/AST or SGPT/ALT ≤ 3 x
upper limit of normal (ULN), and serum direct bilirubin ≤ 2 mg/dL.
In both studies, patients in the REVLIMID/dexamethasone group took 25 mg of REVLIMID
orally once daily on days 1 to 21 and a matching placebo hard capsule once daily on days 22 to
28 of each 28-day cycle. Patients in the placebo/dexamethasone group took 1 placebo hard
capsule on days 1 to 28 of each 28-day cycle. Patients in both treatment groups took 40 mg of
dexamethasone orally once daily on days 1 to 4, 9 to 12, and 17 to 20 of each 28-day cycle for
the first 4 cycles of therapy.
The dose of dexamethasone was reduced to 40 mg orally once daily on days 1 to 4 of each
28-day cycle after the first 4 cycles of therapy. In both studies, treatment was to continue until
disease progression.
In both studies, dose adjustments were allowed based on clinical and laboratory findings.
Sequential dose reductions to 15 mg daily, 10 mg daily and 5 mg daily were allowed for toxicity
see Dosage and Administration (2.2)
.
Table 10 summarizes the baseline patient and disease characteristics in the two studies. In both
studies, baseline demographic and disease-related characteristics were comparable between the
REVLIMID/dexamethasone and placebo/dexamethasone groups.
Table 10: Baseline Demographic and Disease-Related Characteristics – Studies 1 and 2
Study 1
Study 2
REVLIMID/De
x
N=177
Placebo/De
x
N=176
REVLIMID/De
x
N=176
Placebo/De
x
N=175
Patient Characteristics
Age (years)
Median
Min, Max
36, 86
37, 85
33, 84
40, 82
Male
Female
106 (60%)
71 (40%)
104 (59%)
72 (41%)
104 (59%)
72 (41%)
103 (59%)
72 (41%)
Race/Ethnicity
White
Other
141(80%)
36 (20%)
148 (84%)
28 (16%)
172 (98%)
4 (2%)
175(100%)
0 (0%)
ECOG Performance
Status 0-1
157 (89%)
168 (95%)
150 (85%)
144 (82%)
Disease Characteristics
Multiple Myeloma Stage
(Durie-Salmon)
microglobuli
n (mg/L)
≤ 2.5 mg/L
> 2.5 mg/L
52 (29%)
125 (71%)
51 (29%)
125 (71%)
51 (29%)
125 (71%)
48 (27%)
127 (73%)
Number of Prior
Therapies
≥ 2
Types of Prior
Therapies
Stem Cell
Transplantation
Thalidomide
Dexamethasone
Bortezomib
Melphalan
Doxorubicin
The primary efficacy endpoint in both studies was time to progression (TTP). TTP was defined
as the time from randomisation to the first occurrence of progressive disease.
Preplanned interim analyses of both studies showed that the combination of
REVLIMID/dexamethasone was significantly superior to dexamethasone alone for TTP. The
studies were unblinded to allow patients in the placebo/dexamethasone group to receive
treatment with the REVLIMID/dexamethasone combination. For both studies, the extended
follow-up survival data with crossovers were analyzed. In study 1, the median survival time was
39.4 months (95%CI: 32.9, 47.4) in REVLIMID/dexamethasone group and 31.6 months (95%CI:
24.1, 40.9) in placebo/dexamethasone group, with a hazard ratio of 0.79 (95% CI: 0.61-1.03). In
study 2, the median survival time was 37.5 months (95%CI: 29.9, 46.6) in
REVLIMID/dexamethasone group and 30.8 months (95%CI: 23.5, 40.3) in
placebo/dexamethasone group, with a hazard ratio of 0.86 (95% CI: 0.65-1.14).
Table 11: TTP Results in Study 1 and Study 2
Study 1
Study 2
REVLIMID/Dex
N=177
Placebo/Dex
N=176
REVLIMID/Dex
N=176
Placebo/Dex
N=175
TTP
Events n (%)
73 (41)
120 (68)
68 (39)
130 (74)
Median TTP in
months [95% CI]
13.9
[9.5, 18.5]
[3.7, 4.9]
12.1
[9.5, NE]
[3.8, 4.8]
Hazard Ratio
[95% CI]
0.285
[0.210, 0.386]
0.324
[0.240, 0.438]
Log-rank Test p-value
<0.001
<0.001
Response
Complete Response
(CR) n (%)
23 (13)
1 (1)
27 (15)
7 (4)
Partial Response
(RR/PR) n (%)
84 (48)
33 (19)
77 (44)
34 (19)
Overall Response n
107 (61)
34 (19)
104 (59)
41 (23)
p-value
<0.001
<0.001
Odds Ratio [95% CI]
6.38
[3.95, 10.32]
4.72
[2.98, 7.49]
Figure 1: Kaplan-Meier Estimate of Time to Progression — Study 1
Revlimid/Dex
Placebo/Dex
Log Rank p < 0.001
HR (95% CI) = 0.285 (0.210-0.386)
Time to Progression (months)
Proportion of Subjects
Figure 2: Kaplan-Meier Estimate of Time to Progression — Study 2
13.3 Myelodysplastic Syndromes (MDS) with a Deletion 5q Cytogenetic
Abnormality
The efficacy and safety of REVLIMID were evaluated in patients with transfusion-dependent
anemia in low- or intermediate-1- risk MDS with a 5q (q31-33) cytogenetic abnormality in
isolation or with additional cytogenetic abnormalities, at a dose of 10 mg once daily or 10 mg
once daily for 21 days every 28 days in an open-label, single-arm, multi-center study. The major
study was not designed nor powered to prospectively compare the efficacy of the 2 dosing
regimens. Sequential dose reductions to 5 mg daily and 5 mg every other day, as well as dose
delays, were allowed for toxicity [
Dosage and Administration (2.3)
This major study enrolled 148 patients who had RBC transfusion dependent anemia. RBC
transfusion dependence was defined as having received ≥ 2 units of RBCs within 8 weeks prior
to study treatment. The study enrolled patients with absolute neutrophil counts (ANC) ≥
500/mm
, platelet counts ≥ 50,000/mm
, serum creatinine ≤ 2.5 mg/dL, serum SGOT/AST or
SGPT/ALT ≤ 3 x upper limit of normal (ULN), and serum direct bilirubin ≤ 2 mg/dL.
Granulocyte colony-stimulating factor was permitted for patients who developed neutropenia or
fever in association with neutropenia. Baseline patient and disease-related characteristics are
summarized in Table 12.
Table 12: Baseline Demographic and Disease-Related Characteristics in the MDS
Study
Overall
(N=148)
Age (years)
Median
71.0
Min, Max
37.0, 95.0
Gender
n
(%)
Male
(34.5)
Female
(65.5)
Race
n
(%)
White
(96.6)
Other
(3.4)
Duration of MDS (years)
Median
Min, Max
0.1, 20.7
Del 5 (q31-33) Cytogenetic Abnormality
n
(%)
(100.0)
Other cytogenetic abnormalities
(25.2)
IPSS Score
[a]
n
(%)
Low (0)
(37.2)
Intermediate-1 (0.5-1.0)
(43.9)
Intermediate-2 (1.5-2.0)
(4.1)
High (≥2.5)
(1.4)
Missing
(13.5)
FAB Classification
[b]
from central review
n
(%)
(52.0)
RARS
(10.8)
RAEB
(20.3)
CMML
(2.0)
IPSS Risk Category: Low (combined score = 0), Intermediate-1 (combined score = 0.5 to 1.0),
Intermediate-2 (combined score = 1.5 to 2.0), High (combined score ≥ 2.5); Combined score =
(Marrow blast score + Karyotype score + Cytopenia score)
French-American-British (FAB) classification of MDS.
The frequency of RBC transfusion independence was assessed using criteria modified from the
International Working Group (IWG) response criteria for MDS. RBC transfusion independence
was defined as the absence of any RBC transfusion during any consecutive “rolling” 56 days (8
weeks) during the treatment period.
Transfusion independence was seen in 99/148 (67%) patients (95% CI [59, 74]). The median
duration from the date when RBC transfusion independence was first declared (i.e., the last day
of the 56-day RBC transfusion-free period) to the date when an additional transfusion was
received after the 56-day transfusion-free period among the 99 responders was 44 weeks (range
of 0 to >67 weeks). Ninety percent of patients who achieved a transfusion benefit did so by
completion of three months in the study.
RBC transfusion independence rates were unaffected by age or gender.
The dose of REVLIMID was reduced or interrupted at least once due to an adverse event in 118
(79.7%) of the 148 patients; the median time to the first dose reduction or interruption was 21
days (mean, 35.1 days; range, 2-253 days), and the median duration of the first dose interruption
was 22 days (mean, 28.5 days; range, 2-265 days). A second dose reduction or interruption due
to adverse events was required in 50 (33.8%) of the 148 patients. The median interval between
the first and second dose reduction or interruption was 51 days (mean, 59.7 days; range, 15-205
days) and the median duration of the second dose interruption was 21 days (mean, 26 days;
range, 2-148 days).
13.4 Mantle Cell Lymphoma
The efficacy and safety of lenalidomide were evaluated in patients with mantle cell lymphoma in
a phase II, multicenter, randomised open-label study versus single agent of investigator’s choice
in patients who were refractory to their last regimen or had relapsed one to three times (Study
MCL-002).
Patients who were at least 18 years of age with histologically-proven MCL and CT-measurable
disease were enrolled. Patients were required to have received adequate previous treatment with
at least one prior combination chemotherapy regimen. Also, patients had to be ineligible for
intensive chemotherapy and/or transplant at time of inclusion in the study. Patients were
randomised 2:1 to the lenalidomide or the control arm. The investigator’s choice treatment was
selected before randomisation and consisted of monotherapy with either chlorambucil, cytarabine,
rituximab, fludarabine, or gemcitabine.
Lenalidomide was administered orally 25 mg once daily for the first 21 days (D1 to D21) of
repeating 28-day cycles until progression or unacceptable toxicity. Patients with moderate renal
insufficiency were to receive a lower starting dose of lenalidomide 10 mg daily on the same
schedule.
The baseline demographics were comparable between the lenalidomide arm and control arm.
Both patient populations presented a median age of 68.5 years with comparable male to female
ratio. The ECOG performance status was comparable between both groups, as was the number of
prior therapies.
The primary efficacy endpoint in Study MCL-002 was progression-free survival (PFS).
The efficacy results for the Intent-to-Treat (ITT) population were assessed by the Independent
Review Committee (IRC), and are presented in the table 13 below.
Table 13: Summary of efficacy results – study MCL-002, intent-to-treat population
Lenalidomide Arm
Control Arm
N = 170
N = 84
PFS
PFS,
median
[95% CI]
(weeks)
37.6 [24.0, 52.6]
22.7 [15.9, 30.1]
Sequential HR
[95% CI]
0.61 [0.44, 0.84]
Sequential log-rank test, p-value
0.004
Response
, n (%)
Complete response (CR)
8 (4.7)
0 (0.0)
Partial response (PR)
60 (35.3)
9 (10.7)
Stable disease (SD)
50 (29.4)
44 (52.4)
Progressive disease (PD)
34 (20.0)
26 (31.0)
Not done/Missing
18 (10.6)
5 (6.0)
ORR (CR, CRu, PR)
, n (%) [95% CI]
68 (40.0) [32.58, 47.78]
9 (10.7)
[5.02, 19.37]
p-value
< 0.001
CRR (CR, CRu)
, n (%) [95% CI]
8 (4.7) [2.05, 9.06]
0 (0.0) [95.70, 100.00]
p-value
0.043
Duration of Response,
median
[95% CI]
(weeks)
69.6 [41.1, 86.7]
45.1 [36.3, 80.9]
Overall Survival
HR
[95% CI]
0.89 [0.62, 1.28]
Log-rank test, p-value
0.520
CI = confidence interval; CRR = complete response rate; CR = complete response; CRu = complete response unconfirmed; DMC = Data
Monitoring Committee; ITT = intent-to-treat; HR = hazard ratio; KM = Kaplan-Meier; MIPI = Mantle Cell Lymphoma International
Prognostic Index; NA = not applicable; ORR = overall response rate; PD = progressive disease; PFS = progression-free survival; PR= partial
response; SCT = stem cell transplantation; SD = stable disease; SE = standard error.
The median was based on the KM estimate.
Range was calculated as 95% CIs about the median survival time.
The mean and median are the univariate statistics without adjusting for censoring.
The stratification variables included time from diagnosis to first dose (< 3 years and ≥ 3 years), time from last prior systemic anti-lymphoma
therapy to first dose (< 6 months and ≥ 6 months), prior SCT (yes or no), and MIPI at baseline (low, intermediate, and high risk).
Sequential test was based on a weighted mean of a log-rank test statistic using the unstratified log-rank test for sample size increase and the
unstratified log-rank test of the primary analysis. The weights are based on observed events at the time the third DMC meeting was held and
based on the difference between observed and expected events at the time of the primary analysis. The associated sequential HR and the
corresponding 95% CI are presented.
In study MCL-002 in the ITT population, there was an overall apparent increase in deaths within
20 weeks in the lenalidomide arm 22/170 (13%) versus 6/84 (7%) in the control arm. In patients
with high tumour burden, corresponding figures were 16/81 (20%) and 2/28 (7%).
14.
REFERENCES
OSHA Hazardous Drugs.
OSHA
[Accessed on 29 January 2013, from
http://www.osha.gov/SLTC/hazardousdrugs/index.html]
15.
HOW SUPPLIED/STORAGE AND HANDLING
15.1
How Supplied
White and blue-green opaque hard capsules imprinted “REV” on one half and “2.5 mg” on the
other half in black ink:
2.5 mg blisters of 21
White opaque hard capsules imprinted “REV” on one half and “5 mg” on the other half in black
ink:
5 mg blisters of 21
Pale yellow/white hard capsules marked “REV 7.5 mg”
7.5 mg blisters of 21.
Blue/green and pale yellow opaque hard capsules imprinted “REV” on one half and “10 mg” on
the other half in black ink:
10 mg blisters of 21
Powder blue and white opaque hard capsules imprinted “REV” on one half and “15 mg” on the
other half in black ink:
15 mg blisters of 21
Powder blue and blue-green opaque hard capsules imprinted “REV” on one half and “20 mg” on
the other half in black ink.
20 mg blisters of 21
White opaque hard capsules imprinted “REV” on one half and “25 mg” on the other half in black
ink:
25 mg blisters of 21
15.2 Storage
DO NOT STORE ABOVE 25°C.
15.2
Handling and Disposal
Care should be exercised in the handling of REVLIMID. REVLIMID hard capsules should not
be opened or crushed. If powder from REVLIMID contacts the skin, wash the skin immediately
and thoroughly with soap and water. If REVLIMID contacts the mucous membranes, they
should be thoroughly flushed with water. Procedures for the proper handling and disposal of
anticancer drugs should be considered. Several guidelines on the subject have been published.
Dispense no more than a 28-day supply.
Registration No.
Revlimid 2.5mg: 33894
Revlimid 5mg: 31660
Revlimid 7.5mg: 33896
Revlimid 10mg: 31661
Revlimid 15mg: 31662
Revlimid 20mg: 33965
Revlimid 25mg: 31663
Manufacturers
Celgene International Sarl, Boudry, Switzerland
Registration Holder
Neopharm Scientific Ltd. P.O.B 7063, Petach Tiqva 49170
The format of this leaflet has defined by the Ministry of Health; its content has been checked and
approved in 08/2019
.