Yondelis 0.25 mg powder for concentrate for solution for infusion/Yondelis 1 mg powder for concentrate for solution for infusion

*
Pharmacy Only: Prescription
  • Company:

    Immedica (distributor)
  • Status:

    No Recent Update
  • Legal Category:

    Product subject to medical prescription which may not be renewed (A)
  • Active Ingredient(s):

    *Additional information is available within the SPC or upon request to the company

Updated on 19 January 2023

File name

Yondelis SPC-Med.ie_en2021-07-22.pdf

Reasons for updating

  • Document format updated

Legal category:Product subject to medical prescription which may not be renewed (A)

Updated on 01 September 2021

File name

Yondelis PIL-Med.ie-en2021-07-22.pdf

Reasons for updating

  • Change to section 4 - possible side effects
  • Change to section 6 - date of revision

Updated on 01 September 2021

File name

Yondelis SPC-Med.ie_en2021-07-22.pdf

Reasons for updating

  • Change to section 4.8 - Undesirable effects
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

New text highlighted in blue

Deleted text highlighted in red

Note: sections only provided where amendments have been made

4.8         Undesirable effects

Summary of the safety profile

Most patients treated with Yondelis can be expected to have adverse reactions of any grade (91% in monotherapy and 99.4% in combination therapy) and less than one third serious adverse reactions of grade 3 or 4 severity (10% in monotherapy and 25% in combination therapy). The most common adverse reactions of any severity grade were neutropenia, nausea, vomiting, increase in AST/ALT, anaemia, fatigue, thrombocytopenia, anorexia and diarrhoea.

Fatal adverse reactions have occurred in 1.9% and 0.90.6% of patients treated with the monotherapy and combination regimens respectively. They were often the result of a combination of events including pancytopenia, febrile neutropenia, some of them with sepsis, hepatic involvement, renal or multiorgan failure and rhabdomyolysis.

Tabulated summary of adverse reactions

System Organ Class

Very Common

Common

Uncommon

Rare

Psychiatric Disorders

Insomnia

Insomnia

 

 

Musculoskeletal and Connective Tissue Disorders

Arthralgia

Back pain

Blood creatine phosphokinase increased

Arthralgia

Myalgia

 

Rhabdomyolysis

 

*   Adverse drug reaction only for Ovarian cancer patients, including data from ET743‑OVA‑301, a randomized phase 3 study of 672 patients who received either trabectedin (1.1 mg/m2) and PLD (30 mg/m2) every 3 weeks or PLD (50 mg/m2) every 4 weeks; and from study ET743-OVC-3006 which enrolled 576 patients who received either PLD (30 mg/m2) followed by trabectedin (1.1 mg/m2) every 3 weeks or PLD alone (50 mg/m2) every 4 weeks.

In the ET743‑OVA‑301 Yondelis+PLD arm, non‑white (mainly Asian) patients had a higher incidence than white patients in grade 3 or 4 adverse reactions (96% versus 87%), and serious adverse reactions (44% versus 23% all grades). The differences were mainly observed in relation with neutropenia (93% versus 66%), anaemia (37% versus 14%) and thrombocytopenia (41% versus 19%). However, the incidences of clinical complications related to haematological toxicity such as severe infections or bleeding, or those leading to death or treatment termination, were similar in both subpopulations.

10.       DATE OF REVISION OF THE TEXT

24/07/202022/07/2021

Updated on 17 September 2020

File name

Yondelis SPC-Med.ie_appr24July2020.pdf

Reasons for updating

  • Change to section 5.1 - Pharmacodynamic properties
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

New text highlighted in blue

Deleted text highlighted in red

Note: sections only provided where amendments have been made

5.1       Pharmacodynamic properties

Section is unchanged up to this point

No data are available comparing Yondelis+PLD to a platinum‑based regimen in platinum‑sensitive patients.

No statistically significant differences were found between treatment arms in global measures of Quality of Life.

The Yondelis+PLD combination in relapsed ovarian cancer also was evaluated in study ET743-OVC-3006, a phase 3 study in which women with ovarian cancer after failure of a second platinum-containing regimen were randomized to Yondelis (1.1 mg/m2) and PLD (30 mg/m2) every 3 weeks or PLD (50 mg/m2) every 4 weeks. Study participants were required to be platinum sensitive (PFI ≥ 6 months) following their first platinum-containing regimen and have a complete or partial response to a second line platinum-based chemotherapy (without PFI restrictions) meaning that these patients could be either platinum-sensitive (PFI ≥ 6 months) or platinum-resistant (PFI < 6 months) following their second platinum-containing regimen. A post hoc analysis determined that 42% of enrolled subjects were platinum-resistant (PFI < 6 months) following their last platinum-containing regimen.

The primary endpoint of study ET743-OVC-3006 was OS and secondary endpoints included PFS and ORR. The study was sized to enrol approximately 670 patients in order to observe 514 deaths to detect a HR of 0.78 for OS with 80% power given a two-sided significance level of 0.05 spread across two planned analyses on OS, at interim (60% or 308/514 deaths) and final analysis (514 deaths). Two early unscheduled futility analyses were performed at the request of the Independent Data Monitoring Committee (IDMC). Following the second futility analysis performed at 45% of planned events (232/514 deaths), the IDMC recommended discontinuing the study due to (1) futility of the primary analysis on OS and (2) excessive risk based on imbalance of adverse events not in favour of Yondelis+PLD. At early termination of the study, 9% (52/572 treated) of subjects stopped treatment, 45% (260/576 randomized) stopped follow-up, and 54% (310/576 randomized) were censored from OS assessment, precluding reliable estimates of PFS and OS endpoints.

No data are available comparing Yondelis+PLD to a platinum-based regimen in platinum-sensitive patients.

10.       DATE OF REVISION OF THE TEXT

12/201924/07/2020

Updated on 17 September 2020

File name

Yondelis PIL-Med.ie_appr24July2020.pdf

Reasons for updating

  • Change to section 6 - date of revision

Free text change information supplied by the pharmaceutical company

Date leaflet was last revised in changed to state: 07/2020

Updated on 02 March 2020

File name

Yondelis SPC-Med.ie-v21-02.03.2020-clean.pdf

Reasons for updating

  • Change to section 4.4 - Special warnings and precautions for use
  • Change to Section 4.8 – Undesirable effects - how to report a side effect
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

New text highlighted in blue

Deleted text highlighted in red

Note: sections only provided where amendments have been made

4.4       Special warnings and precautions for use

Cardiac Dysfunction

It is recommended to monitor patients for clinical cardiac signs or symptoms. It is also recommended to monitor LVEF at baseline and periodically during the treatment; particularly in patients at risk of cardiomyopathy from previous anthracycline exposure or in patients with symptoms of decreasing cardiac function.

Patients should be monitored for cardiac-related adverse events or myocardial dysfunction.

A thorough cardiac assessment including determination of left ventricular ejection fraction (LVEF) by echocardiogram or multigated acquisition scan (MUGA) should be conducted before initiation of trabectedin and at 2 to 3-month intervals thereafter until trabectedin is discontinued.

Patients with LVEF less than the lower limit of normal (LVEF < LLN), prior cumulative anthracycline dose of >300mg/m2, aged > 65 years, or a history of cardiovascular disease (especially in those with cardiac medication) may be at increased risk of cardiac dysfunction at treatment with trabectedin as monotherapy or in combination with doxorubicin.

For patients with Grade 3 or 4 cardiac adverse events indicative of cardiomyopathy or for patients with a LVEF that decreases below the LLN (assessed as either an absolute decrease of LVEF of ≥15% or <LLN with an absolute decrease of ≥5%), trabectedin should be discontinued.

10.       DATE OF REVISION OF THE TEXT

06/201812/2019

Updated on 02 March 2020

File name

Yondelis PIL-Med.ie-v19-02.03.2020.pdf

Reasons for updating

  • Change to section 2 - what you need to know - warnings and precautions
  • Change to section 4 - how to report a side effect
  • Change to section 6 - date of revision

Free text change information supplied by the pharmaceutical company

Warnings and precautions

Talk to your doctor before using Yondelis

Yondelis or its combination with PLD must not be used if you have severe liver, kidney or cardiac damage. Tell your doctor if you know or suspect that you have any liver, kidney or cardiac problems before starting the treatment with Yondelis.

Tell your doctor if you know or suspect that you have any of the following before starting the treatment with Yondelis:

  • Liver or kidney problems.
  •  Cardiac problems or a history of cardiac problems.
  • Left ventricular ejection fraction (LVEF) under the lower limit of normal.
  • Received high anthracycline dose treatment in the past.

Updated on 23 July 2018

File name

Yondelis-PIL-IPHA v18-19.07.2018.pdf

Reasons for updating

  • Change to section 4 - possible side effects
  • Change to section 6 - date of revision

Updated on 23 July 2018

File name

Yondelis SPC-IPHA v20-19.07.2018-clean.pdf

Reasons for updating

  • Change to section 4.4 - Special warnings and precautions for use
  • Change to section 4.8 - Undesirable effects
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

 

Note: sections only provided where amendments have been made

 

4.4       Special warnings and precautions for use

 

Capillary Leak Syndrome (CLS)

 

Cases of Capillary Leak Syndrome (CLS) have been reported with trabectedin (including cases with fatal outcomes).  If symptoms of possible CLS develop, such as unexplained oedema with or without hypotension, the treating physician should reassess serum albumin level. A rapid decline in serum albumin level may be indicative of CLS. If a diagnosis of CLS is confirmed after exclusion of other causes, the treating physician should discontinue trabectedin and initiate CLS treatment according to institutional guidelines (see sections 4.2 and 4.8).

 

4.8       Undesirable effects

 Updates made to this section.

 

10.       DATE OF REVISION OF THE TEXT

 

06/2018

 

Updated on 15 November 2017

Reasons for updating

  • New SPC for new product

Legal category:Product subject to medical prescription which may not be renewed (A)

Updated on 15 November 2017

Reasons for updating

  • Change to section 1 - Name of medicinal product
  • Change to section 4.2 - Posology and method of administration
  • Change to section 4.4 - Special warnings and precautions for use
  • Change to section 5.2 - Pharmacokinetic properties
  • Change to section 9 - Date of first authorisation/renewal of the authorisation
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

New text highlighted in blue

Deleted text highlighted in red

 

1.            NAME OF THE MEDICINAL PRODUCT

Yondelis 0.25 mg

Yondelis 0.25 mg powder for concentrate for solution for infusion.

Yondelis 1 mg

Yondelis 1 mg powder for concentrate for solution for infusion.

 

4.2         Posology and method of administration

Hepatic impairment

 

No studies with the proposed regimen have been conducted in patients with liver dysfunction. Thus, data are not available to recommend a lower starting dose in patients with hepatic impairment. However, sSpecial caution is advised and dose adjustments may be necessary in these patients with hepatic impairment since systemic exposure to trabectedin is probably increased and the risk of hepatotoxicity might be increased. Patients with elevated serum bilirubin levels at baseline must not be treated with Yondelis. Liver function tests should be monitored during treatment with Yondelis as dose adjustments may be indicated (see Table 1 and section 4.4).

 

4.4         Special warnings and precautions for use

Hepatic impairment

 

Patients must meet specific criteria on hepatic function parameters to start treatment with Yondelis. Since the systemic exposure to trabectedin is probably on average approximately doubled (see section 5.2) increased due to hepatic impairment and therefore the risk of hepatotoxicityies might be increased, patients with clinically relevant liver diseases, such as active chronic hepatitis, must be closely monitored and the dose adjusted if needed. Patients with elevated serum bilirubin levels must not be treated with trabectedin (see section 4.2).

 

Liver Function Test (LFT) abnormalities

 

Reversible acute increases in aspartate aminotransferase (AST) and alanine aminotransferase (ALT) have been reported in most patients. Yondelis must not be used in patients with elevated bilirubin. Patients with increases in AST, ALT and alkaline phosphatase between cycles may necessitate dose reduction adjustments (see section 4.2).

 

 

5.2         Pharmacokinetic properties

 

Hepatic impairment

 

Although the population analysis showed no relationship between the serum liver enzymes concentrations and the plasma clearance of trabectedin, systemic exposure to trabectedin may be increased in patients with hepatic impairment; therefore close monitoring of toxicity is warranted.

The effect of hepatic impairment on the pharmacokinetics of trabectedin was assessed in 15 cancer patients at doses ranging from 0.58 to 1.3 mg/m2 administered as 3-hour infusion. The geometric mean dose normalized trabectedin exposure (AUC) increased by 97% (90% CI: 20%, 222%) in 6 patients with moderate hepatic impairment (increased serum bilirubin levels from 1.5 to 3 x ULN and increase of aminotransferases (AST or ALT) < 8 x ULN) following administration of a single trabectedin dose of 0.58 mg/m2 (n=3) or 0.9 mg/m2 (n=3) compared to 9 patients with normal liver function following administration of a single trabectedin dose of 1.3 mg/m2 (see sections 4.2 and 4.4).

 

 

9.            DATE OF FIRST AUTHORISATION/RENEWAL OF THE AUTHORISATION

 

Date of first authorisation: 17 September 2007

Date of latest renewal: 17 03 AugustSeptember 2012

 

10.       DATE OF REVISION OF THE TEXT

 

10/2017

Updated on 14 November 2017

File name

PIL_12831_708.pdf

Reasons for updating

  • New PIL for new product

Updated on 14 November 2017

Reasons for updating

  • Change to section 2 - interactions with other medicines, food or drink
  • Change to section 4 - possible side effects
  • Change to section 6 - date of revision

Updated on 04 September 2017

Reasons for updating

  • Change to section 4.4 - Special warnings and precautions for use
  • Change to Section 4.8 – Undesirable effects - how to report a side effect
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

New text highlighted in blue

Deleted text highlighted in red

 

4.4         Special warnings and precautions for use

Capillary Leak Syndrome (CLS)

 

Cases of Capillary Leak Syndrome (CLS) have been reported with trabectedin. If symptoms of possible CLS develop, such as unexplained edema with or without hypotension, the treating physician should reassess serum albumin level. A rapid decline in serum albumin level may be indicative of CLS. If a diagnosis of CLS is confirmed after exclusion of other causes, the treating physician should discontinue trabectedin and initiate CLS treatment according to institutional guidelines (see sections 4.2 and 4.8).

 

4.8         Undesirable effects

Tabulated summary of adverse reactions

 

The frequencies of the adverse reactions reported below are classified as very common ( 1/10), common ( 1/100 to < 1/10) and uncommon ( 1/1,000 to < 1/100).

 

The table below displays the adverse reactions reported in ≥ 1% of patients treated with the soft tissue sarcoma recommended regimen (1.5 mg/m2, 24 hour infusion every 3 weeks) according to the standard MedDRA (Medical Dictionary for Regulatory Activities) system organ class. Both adverse reactions and laboratory values have been used to provide frequencies. Within each frequency grouping, undesirable effects are presented in order of decreasing seriousness.

 

System Organ Class

Adverse reactions reported in ≥ 1% of patients with soft tissue sarcoma in clinical trials.

Infections and Infestations

Common

Infection

Blood and Lymphatic System Disorders

Very Common

Neutropenia* (Grade 3 = 26%, Grade 4 = 24%), Thrombocytopenia* (Grade 3 = 11%, Grade 4 = 2%), Anaemia* (Grade 3 = 10%, Grade 4 = 3%), Leukopenia*

Common

Febrile neutropenia

Metabolism and Nutrition Disorders

Very Common

Anorexia (Grade 3‑4 < 1%)

Common

Dehydration, Decreased appetite, Hypokalaemia

Psychiatric Disorders

Common

Insomnia

Nervous System Disorders

Very Common

Headache

Common

Peripheral sensory neuropathy, Dysgeusia, Dizziness, Paraesthesia

Vascular Disorders

Common

Hypotension, Flushing

Uncommon

Capillary Leak Syndrome

Respiratory, Thoracic and Mediastinal Disorders

Common

Dyspnoea (Grade 3‑4 = 2%), Cough

Gastrointestinal disorders

Very Common

Vomiting (Grade 3‑4 = 6.5%), Nausea (Grade 3‑4 = 6%), Constipation (Grade 3‑4 < 1%)

Common

Diarrhoea (Grade 3‑4 < 1%), Stomatitis (Grade 3‑4 < 1%), Abdominal pain, Dyspepsia, Upper abdominal pain

Hepatobiliary Disorders

Very Common

Hyperbilirubinemia* (Grade 3 = 1%), Alanine aminotransferase increased* (Grade 3 = 38%, Grade 4 = 3%), Aspartate aminotransferase increased* (Grade 3 = 44%, Grade 4 = 7%), Blood alkaline phosphatase increased*, Gamma‑glutamyltransferase increased*

Skin and Subcutaneous Tissue Disorders

Common

Alopecia

 

Musculoskeletal and Connective Tissue Disorders

Common

Myalgia, Arthralgia, Back pain

 

General Disorders and Administration Site Conditions

Very Common

Fatigue (Grade 3‑4 = 9%), Asthenia (Grade 3‑4 = 1%)

Common

Pyrexia, Oedema, Oedema peripheral, Injection site reaction

Investigations

Very Common

Blood creatine phosphokinase increased* (Grade 3‑4 = 4%), Blood creatinine increased*, Blood albumin decreased*

Common

Weight decreased

 

 

* Derived from laboratory data

 

The table below provides the frequency and severity of undesirable effects considered possibly related to study medicinal product and reported in ≥ 5% of patients with ovarian cancer randomised to receive Yondelis 1.1 mg/m2/PLD 30 mg/m2 in the pivotal trial ET743‑OVA‑301. Both adverse reactions and laboratory values have been used. Within each frequency grouping, undesirable effects are presented in order of decreasing seriousness.

 

 

Adverse reactions reported in ≥ 5% of patients in clinical trial ET743‑OVA‑301

System Organ Class

Frequency

Event

Yondelis+PLD
n=333

All Grades

(%)

Grade 3

(%)

Grade 4

(%)

Blood and lymphatic system disorders

 

Very common

 

Neutropenia*

91.6

29.7

42.3

Leukopenia*

94.9

44.7

17.7

Anaemia*

94.9

12.9

5.7

Thrombocytopenia*

63.7

12.3

10.8

Common

Febrile neutropenia*

6.9

4.5

2.4

Metabolism and nutrition disorders

 

Very common

Anorexia

28.8

2.1

 

Common

Hypokalaemia

6.3

2.1

 

Nervous system disorders

 

Common

 

Headache

6.6

0.3

 

Dysgeusia

5.4

0.3

 

Vascular disorders

Uncommon

 

Capillary Leak Syndrome

 

 

 

Respiratory, thoracic and mediastinal disorders

Common

Dyspnoea

6.6

0.3

 

Gastrointestinal disorders

 

Very common

 

Nausea

70.9

8.7

 

Vomiting

51.7

9.9

0.3

Constipation

20.4

0.9

 

Stomatitis

19.2

0.9

 

Diarrhoea

17.1

2.1

 

Common

 

Abdominal pain

9.3

0.6

 

Dyspepsia

7.5

0.3

 

Hepatobiliary disorders

Very common

Hyperbilirubinaemia*

(25.2)

(0.3)

 

Alanine aminotransferase increased*

96.1

45.6

4.5

Aspartate aminotransferase increased*

89.5

12.0

1.8

Blood alkaline phosphatase increased*

61.3

1.5

 

Skin and subcutaneous tissue disorders

 

Very common

 

Palmar‑plantar erythrodysaesthesia syndrome

24

3.9

 

Alopecia

12

 

 

Common

 

Rash

8.1

 

 

Skin hyperpigmentation

5.4

 

 

General disorders and administration site conditions

Very common

 

Fatigue

42.3

5.7

0.3

Asthenia

15.3

1.2

 

Mucosal inflammation

11.4

2.1

 

Pyrexia

10.2

0.9

 

Investigations 

Common

Blood creatine phosphokinase increased*

22.0

0.9

0.9

 

* Derived from laboratory data

 

Capillary Leak Syndrome: Cases of suspected capillary leak syndrome have been reported with trabectedin. Frequency uncommon.

 

10.       DATE OF REVISION OF THE TEXT

 

06/2017

 

 

Updated on 29 August 2017

Reasons for updating

  • Change to section 2 - what you need to know - warnings and precautions
  • Change to section 4 - possible side effects
  • Change to section 6 - date of revision

Updated on 26 July 2016

Reasons for updating

  • Change to section 4.4 - Special warnings and precautions for use
  • Change to section 4.8 - Undesirable effects
  • Change to section 5.1 - Pharmacodynamic properties
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

New text highlighted in blue

Deleted text highlighted in red

 

4.4         Special warnings and precautions for use

 

Cardiac Dysfunction

 

It is recommended to monitor patients for clinical cardiac signs or symptoms. It is also recommended to monitor LVEF at baseline and periodically during the treatment; particularly in patients at risk of cardiomyopathy from previous anthracycline exposure or in patients with symptoms of decreasing cardiac function.

 

4.8         Undesirable effects

Most patients treated with Yondelis can be expected to have adverse reactions of any grade (91% in monotherapy and 99% in combination therapy) and less than one third serious adverse reactions of grade 3 or 4 severity (10% in monotherapy and 25% in combination therapy). The most common adverse reactions of any severity grade were neutropenia, nausea, vomiting, increases in AST/ALT, anaemia, fatigue, thrombocytopenia, anorexia and diarrhoea.

 

The following reactions have been reported with a frequency below 5% in the combination arm, but are included here for their clinical relevance: neutropenic infection (< 1%), neutropenic sepsis (< 1%), pancytopenia (1.8%), bone marrow failure (1.5%), granulocytopenia (1.5%), dehydration, insomnia, peripheral sensory neuropathy, syncope, left ventricular dysfunction (< 1%), pulmonary embolism (1.2%), pulmonary oedema (< 1%), cough, hepatotoxicity (< 1%), gamma‑glutamyltransferase increased, bilirubin conjugated increased, musculoskeletal pain, myalgia, blood creatinine increased, oedema/peripheral oedema, catheter site reactions.

 

5.         PHARMACOLOGICAL PROPERTIES

 

5.1       Pharmacodynamic properties

 

Results from an expanded access program for patients with STS (study ET743-SAR- 3002) show that among the 903 subjects assessed for OS, the median survival time was 11.9 months (95% CI: 11.2, 13.8). The median survival by histology tumour type was 16.2 months [95% CI: 14.1, 19.5] for subjects with leiomyosarcomas and liposarcomas and 8.4 months [95% CI: 7.1, 10.7] for subjects with other types of sarcomas. The median survival for subjects with liposarcoma was 18.1 months [95% CI: 15.0, 26.4] and for subjects with leiomyosarcoma 16.2 months [95% CI: 11.7, 24.3].

 

Additional efficacy data are available from a randomized active-controlled phase III study of trabectedin vs. dacarbazine (Study ET743-SAR-3007), in patients treated for unresectable or metastatic lipo- or leiomyosarcoma who have been previously treated with at least an anthracycline and ifosfamide containing regimen, or an anthracycline containing regimen and one additional cytotoxic chemotherapy regimen. Patients in the trabectedin arm were required to receive dexamethasone 20 mg intravenous injection prior to each trabectedin infusion. Overall, 384 patients were randomized to the trabectedin group [1.5 mg/m2 once every 3 weeks (q3wk 24-h)] and 193 patients to the dacarbazine group (1 g/m2 once every 3 weeks). The median patient age was 56 years (range 17 to 81), 30% were male, 77% Caucasian, 12% African American and 4% Asian. Patients in the trabectedin and dacarbazine arms received a median of 4 and 2 cycles respectively. The primary efficacy endpoint of the study was OS, which included 381 death events (66% of all randomized patients): 258 (67.2%) deaths in the trabectedin group and 123 (63.7%) deaths in the dacarbazine group (HR 0.927 [95% CI: 0.748, 1.150; p=0.4920]). The final analysis showed no significant difference with a median survival follow-up of 21.2 months resulted in a median of 13.7 months (95% CI: 12.2, 16.0) for the trabectedin arm and 13.1 months [95% CI: 9.1, 16.2] for the dacarbazine arm. The main secondary endpoints are summarized in the table below:

 

Efficacy results from Study ET743SAR3007

 

Endpoints / Study population

Trabectedin

Dacarbazine

Hazard Ratio / Odds Ratio

p value

Primary endpoint

n=384

n=193

 

 

Overall survival,

n (%)

258 (67.2%)

123 (63.7%)

0.927 (0.748-1.150)

0.4920

Secondary endpoints

n=345

n=173

 

 

PFS

(months; 95% CI)

4.2

1.5

0.55 (0.44, 0.70)

<0.0001

ORR,

n (%);

Odds ratio (95% CI)

34 (9.9%)

12 (6.9%)

1.47 (0.72, 3.2)

0.33

DOR

(months; 95% CI)

6.5

4.2

0.47 (0.17, 1.32)

0.14

CBR,

n (%);

Odds ratio (95% CI)

34.2%

18.5%

2.3 (1.45, 3.7)

<0.0002

 

Additional efficacy data are available from a randomized, open-label, multicenter phase II study [JapicCTI-121850] conducted in Japanese patients with translocation-related sarcoma (TRS), most common being myxoid round-cell liposarcoma (n=24), synovial sarcoma (n=18), mesenchymal chondrosarcoma (n=6), and extraskeletal Ewing sarcoma/PNET, alveolar soft part sarcoma, alveolar rhabdomyosarcoma and clear cell sarcoma (n=5 each). The study assessed the efficacy and safety of trabectedin vs. best supportive care (BSC) as second-line or later therapy for patients with advanced TRS unresponsive or intolerant to standard chemotherapy regimen. The patients received the trabectedin dose of 1.2 mg/m2 recommended for Japanese patients [1.2 mg/m2 once every 3 weeks (q3wk 24-h)]. A total of 76 Japanese patients were enrolled in the study, among which 73 patients were included in the final analysis set. The study primary endpoint was PFS, that showed a statistically significant improvement in favour of trabectedin over BSC [HR=0.07; 95% CI: 0.03-0.16; p<0.0001], with a median PFS in the trabectedin group of 5.6 months [95% CI: 4.1-7.5] and in the BSC group of 0.9 months [95% CI: 0.7-1.0]. The secondary endpoints included objective response analysed using the RECIST and Choi criteria. Using the RECIST criteria the ORR among patients treated with trabectedin was 3 (8.1%; 95% CI: 1.7.21.9%) and 0 (0%, 95% CI: 0.0-9.7%) among patients treated with best supportive care, while the CBR was 24 (64.9%, 95% CI: 47.5-79.9%) versus 0 (0%, 95% CI: 0.0-9.7%), respectively. Using the Choi criteria the ORR among patients treated with trabectedin was 4 (10.8%; 95% CI: 3.0-25.4%) and 0 (0%, 95% CI: 0.0-9.7%) among patients treated with best supportive care, while the CBR was 7 (18.9%, 95% CI: 8.0-35.2%) versus 0 (0%, 95% CI: 0.0-9.7%), respectively.

 

10.       DATE OF REVISION OF THE TEXT

 

07/2016

 

Updated on 21 July 2016

Reasons for updating

  • Change to warnings or special precautions for use
  • Change to side-effects
  • Change to date of revision
  • Change to dosage and administration

Updated on 28 January 2016

Reasons for updating

  • Change to section 6.5 - Nature and contents of container

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

Typographical errors  - rectified

6.5 Nature and contents of container

 

 

Yondelis 0.25 mg

Type I colourless glass vial with a butyl rubber stopper covered with an aluminium flip‑off seal containing 0.25 mg of trabectedin.

 

Yondelis 1 mg

 

Type I colourless glass vial with a butyl rubber stopper covered with an aluminium flip-off seal containing 1mg of trabectedin.

Updated on 27 January 2016

Reasons for updating

  • Change to section 1 - Name of medicinal product
  • Change to section 2 - Qualitative and quantitative composition
  • Change to section 4.2 - Posology and method of administration
  • Change to section 4.6 - Pregnancy and lactation
  • Change to Section 4.8 – Undesirable effects - how to report a side effect
  • Change to section 5.3 - Preclinical safety data
  • Change to section 6.5 - Nature and contents of container
  • Change to section 6.6 - Special precautions for disposal and other handling
  • Change to section 8 - MA number
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company


Highlighted changes following approval of:

 

  • The two Yondelis SmPCs 0.25mg and 1mg have been merged into one unique combined SmPC.
  • Results from a preclinical study in pregnant rats have been included in the SmPC
  • As cases of suspected capillary leak syndrome have been reported with Yondelis, this new adverse reaction is now included in SmPC section 4.8. “Undesirable effects”

Please note only the text that has been updated is shown below, refer to SPC for full section approved text.

 

 

1. NAME OF THE MEDICINAL PRODUCT

 

Yondelis 0.25 mg vial

 

Yondelis 0.25 mg powder for concentrate for solution for infusion.

 

Yondelis 1 mg vial

 

Yondelis 1 mg powder for concentrate for solution for infusion.

 

2. QUALITATIVE AND QUANTITATIVE COMPOSITION

 

Yondelis 0.25 mg vial

 

Each vial of powder contains 0.25 mg of trabectedin.

 

One ml of reconstituted solution contains 0.05 mg of trabectedin.

 

Excipients with known effect:

 

Each vial of powder contains 2 mg of potassium and 0.1 g of sucrose.

 

For the full list of excipients, see section 6.1.

 

Yondelis 1 mg vial

 

Each vial of powder contains 1 mg of trabectedin.

 

0.25 mg and 1 mg vials

 

One ml of reconstituted solution contains 0.05 mg of trabectedin.

 

Excipients with known effect:

 

0.25 mg vial

 

Each vial of powder contains 2 mg of potassium and 0.1 g of sucrose.

 

1 mg vial

 

Each vial of powder contains 8 mg of potassium and 0.4 g of sucrose.

 

0.25 mg and 1 mg vials

 

For the full list of excipients, see section 6.1.

 

4.2 Posology and method of administration

 

Older peopleElderly

 

No specific studies in older people have been performed. Overall 20% of the 1,164 patients in the integrated safety analysis of monotherapy clinical trials were over 65 years. Of the 333 patients with ovarian cancer who received trabectedin in combination with PLD, 24% were 65 years of age or older and 6% were over 75 years.  No relevant differences in the safety profile were seen in this patient population. It seems that plasma clearance and distribution volume of trabectedin are not influenced by age. Therefore, dose adjustments based uniquely on age criteria are not routinely recommended.

 

Patients with hHepatic impairment

 

No studies with the proposed regimen have been conducted in patients with liver dysfunction. Thus, data are not available to recommend a lower starting dose in patients with hepatic impairment. However, special caution is advised and dose adjustments may be necessary in these patients since systemic exposure is probably increased and the risk of hepatotoxicity might be increased. Patients with elevated bilirubin must not be treated with Yondelis (see section 4.4).

 

Patients with rRenal impairment

 

Studies including patients with renal insufficiency (creatinine clearance < 30 ml/min for the monotherapy, and < 60 ml/min for the combination regimen) have not been conducted and therefore Yondelis must not be used in this patient population (see section 4.4). Considering the pharmacokinetic characteristics of trabectedin (see section 5.2), no dose adjustments are warranted in patients with mild or moderate renal impairment.

 

4.6 Fertility, pregnancy and lactation

 

Pregnancy

 

No sufficient clinical data on exposed pregnancies are available. However, based on its known mechanism of action, trabectedin may cause serious birth defects when administered during pregnancy. Trabectedin crossed the placenta when administered to pregnant rats. Trabectedin should not be used during pregnancy unless clearly necessary. If it is used If pregnancy occurs during pregnancytreatment, the patient must be informed of the potential risk to the foetus (see section 5.3) and be monitored carefully. If trabectedin is used at the end of pregnancy, potential adverse reactions should be monitored carefully in the newborns.

 

4.8   Undesirable effects

 

Capillary Leak Syndrome: Cases of suspected capillary leak syndrome have been reported with trabectedin. Frequency uncommon.

 

Reporting of suspected adverse reactions

Reporting suspected adverse reactions after authorisation of the medicinal product is important. It allows continued monitoring of the benefit/risk balance of the medicinal product. Healthcare professionals are asked to report any suspected adverse reactions via: the national reporting system listed in Appendix V.

 

5.3 Preclinical safety data

 

Preclinical data indicate that trabectedin has limited effect on the cardiovascular, respiratory and central nervous system at exposures below the therapeutic clinical range, in terms of AUC.

 

The effects of trabectedin on cardiovascular and respiratory function have been investigated in vivo (anesthetised Cynomolgus monkeys). A 1 hour infusion schedule was selected to attain maximum plasma levels (Cmax values) in the range of those observed in the clinic. The plasma trabectedin levels attained were 10.6 ± 5.4 (Cmax), higher than those reached in patients after infusion of 1,500 mg/m2 for 24 (Cmax of 1.8 ± 1.1 ng/ml) and similar to those reached after administration of the same dose by 3 hour infusion (Cmax of 10.8 ± 3.7 ng/ml).

 

Myelosupression and hepatoxicity were identified as the primary toxicity for trabectedin. Findings observed included haematopoietic toxicity (severe leukopenia, anaemia, and lymphoid and bone marrow depletion) as well as increases in liver function tests, hepatocellular degeneration, intestinal epithelial necrosis, and severe local reactions at the injection site. Renal toxicological findings were detected in multi-cycle toxicity studies conducted in monkeys. These findings were secondary to severe local reaction at the administration site, and therefore uncertainly attributable to trabectedin; however, caution must be guaranteed in the interpretation of these renal findings, and treatment-related toxicity cannot be excluded.

 

Trabectedin is genotoxic both in vitro and in vivo. Long‑term carcinogenicity studies have not been performed.

 

Fertility studies with trabectedin were not performed but limited histopathological changes were observed in the gonads in the repeat dose toxicity studies. Considering the nature of the compound (cytotoxic and mutagenic), it is likely to affect the reproductive capacity.

 

Placental transfer of trabectedin and fetal exposure to trabectedin were observed in a study in pregnant rats that received a single i.v. 14C-trabectedin dose at 0.061 mg/kg. Maximum fetal tissue radioactivity concentration was similar to that in maternal plasma or blood.

 

6.5 Nature and contents of container

 

Yondelis 0.25 mg

 

Type I colourless glass vial with a butyl rubber stopper covered with an aluminium flip‑off seal containing

 

Each carton contains one vial.

 

0.25 mg vial

 

0.25 mg of trabectedin.

 

Yondelis1 mg vial

 

Type I colourless glass vial with a butyl rubber stopper covered with an aluminium flip‑off seal containing

 

1 mg of trabectedin.

 

0.25 mg and 1 mg vials

 

Each carton contains one vial.

 

6.6 Special precautions for disposal and other handling

 

Preparation for intravenous infusion

 

0.25 mg vial

 

Yondelis must be reconstituted and further diluted prior to intravenous infusionAppropriate aseptic techniques must be used to prepare the infusion solution (see Instructions for reconstitution and for dilution).

 

When used in combination with PLD the intravenous line should be flushed well with 50 mg/ml (5%) glucose solution for infusion after administration of PLD and before administration of Yondelis. The use of any diluent other than 50 mg/ml (5%) glucose solution for infusion for this line flushing may cause precipitation of PLD (see also PLD Summary of Product Characteristics for specific handling instructions).

 

Instructions for reconstitution

 

Yondelis 0.25 mg

 

Each vial containing 0.25 mg of trabectedin is reconstituted with 5 ml of sterile water for injections. The solution obtained has a concentration of 0.05 mg/ml and is for single‑use only.

 

A syringe is used to inject 5 ml of sterile water for injections into the vial. The vial must be shaken until complete dissolution. The reconstituted solution results in a clear, colourless or slightly yellowish solution, essentially free of visible particles.

 

This reconstituted solution contains 0.05 mg/ml of trabectedin. It requires further dilution and is for single-use only.

 

Preparation for intravenous infusion

 

Yondelis 1 mg vial

 

Each vial containing 1 mg of trabectedin is reconstituted with 20 ml of sterile water for injections. The solution obtained has a concentration of 0.05 mg/ml and is for single‑use only.

 

A syringe is used to inject 20 ml of sterile water for injections into the vial. The vial must be shaken until complete dissolution. The reconstituted solution results in a clear, colourless or slightly yellowish solution, essentially free of visible particles.

 

This reconstituted solution contains 0.05 mg/ml of trabectedin. It requires further dilution and is for single-use only.

 

Yondelis must be reconstituted and further diluted prior to intravenous infusionAppropriate aseptic techniques must be used to prepare the infusion solution (see Instructions for reconstitution and for dilution).

 

When used in combination with PLD the intravenous line should be flushed well with 50 mg/ml (5%) glucose solution for infusion after administration of PLD and before administration of Yondelis. The use of any diluent other than 50 mg/ml (5%) glucose solution for infusion for this line flushing may cause precipitation of PLD (see also PLD Summary of Product Characteristics for specific handling instructions).

 

Instructions for reconstitution

 

Each vial containing 1 mg of trabectedin is reconstituted with 20 ml of sterile water for injections. The solution obtained has a concentration of 0.05 mg/ml and is for single‑use only.

 

A syringe is used to inject 20 ml of sterile water for injections into the vial. The vial must be shaken until complete dissolution. The reconstituted solution results in a clear, colourless or slightly yellowish solution, essentially free of visible particles.

 

0.25 mg and 1 mg vials

 

This reconstituted solution contains 0.05 mg/ml of trabectedin. It requires further dilution and is for single-use only.

 

8. MARKETING AUTHORISATION NUMBER(S)

 

Yondelis 0.25 mg vial

 

EU/1/07/417/001

 

Yondelis 1 mg vial

 

EU/1/07/417/002

 

10.       DATE OF REVISION OF THE TEXT

 

0612/2015

 

 

Updated on 26 January 2016

Reasons for updating

  • Change to side-effects
  • Change to information about pregnancy or lactation
  • Change to date of revision

Updated on 07 July 2015

Reasons for updating

  • Removal of black triangle
  • Change to section 4.2 - Posology and method of administration
  • Change to Section 4.8 – Undesirable effects - how to report a side effect
  • Change to section 5.1 - Pharmacodynamic properties
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

Highlighted changes following approval of the 7th Annual Reassessment (EMEA/H/C/000773/S/0042) and PSUR II (EMEA/H/C/000773/PSUSA/00003001/201409)

 

  

This medicinal product is subject to additional monitoring. This will allow quick identification of new safety information. Healthcare professionals are asked to report any suspected adverse reactions. See section 4.8 for how to report adverse reactions

 

4.2       Posology and method of administration

 

Method of administration

 

Intravenous administration through a central venous line is strongly recommended (see sections 4.4 and 6.6).

 

For instructions on reconstitution and dilution of the medicinal product before administration, see section 6.6.

 

4.8       Undesirable effects

 

Reporting of suspected adverse reactions

 

Reporting suspected adverse reactions after authorisation of the medicinal product is important. It allows continued monitoring of the benefit/risk balance of the medicinal product. Healthcare professionals are asked to report any suspected adverse reactions via; the national reporting system listed in Appendix V.

 

United Kingdom

Yellow Card Scheme

Website: www.mhra.gov.uk/yellowcard

 

Ireland

HPRA Pharmacovigilance

Earlsfort Terrace

IRL - Dublin 2

Tel: +353 1 6764971

Fax: +353 1 6762517

Website: www.hpra.ie

e-mail: medsafety@hpra.ie

 

Malta

ADR Reporting

Website: www.medicinesauthority.gov.mt/adrportal

 

Ísland

til Lyfjastofnunar,  www.lyfjastofnun.is

5.1       Pharmacodynamic properties

 

Paediatric population

 

In SAR-2005 phase I-II study, a total of 50 paediatric patients with rhabdomyosarcoma, Ewing sarcoma or non rhabdomyosarcoma soft tissue sarcoma were enrolled. Eight patients were treated with a dose of 1.3 mg/m2 and 42 with 1.5 mg/m2. Trabectedin was administered as a 24-hour intravenous infusion every 21 days. Forty patients were fully evaluable for response.  One partial response (PR) centrally confirmed was observed: overall RR: 2.5% CI95% (0.1%-13.2%). The PR corresponded to a patient with an alveolar rhabdomyosarcoma. Duration of the response was 6.5 months No responses were observed for Ewing sarcoma and NRSTS, [RR: 0% CI95% (0%-30.9%)]. Three patients achieved stable disease (one with rhabdomyosarcoma after 15 cycles, one with spindle cell sarcoma after 2 cycles, and one with Ewing sarcoma after 4 cycles.

 

Adverse reactions, included reversible elevation of liver enzymes and haematological events; in addition, fever, infection, dehydration and thrombosis/embolism were also reported.

 

This medicinal product has been authorised under “exceptional circumstances”. This means that due to the rarity of soft tissue sarcoma, it has not been possible to obtain complete information on this medicinal product in this indication.

 

The European Medicines Agency will review any new information which may become available every year and this SmPC will be updated as necessary.

 

 

10.       DATE OF REVISION OF THE TEXT

 

04/201406/2015

 

Updated on 02 July 2015

Reasons for updating

  • Change to side-effects
  • Change to date of revision
  • Removal of black triangle

Updated on 19 August 2014

Reasons for updating

  • Change to Section 4.8 – Undesirable effects - how to report a side effect

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

Highlighted changes to include new IE Agency - HPRA reporting information following the Agency name change (formerly IMB) in July 2014

 

  

4.8        Undesirable effects

 

Reporting of suspected adverse reactions

Reporting suspected adverse reactions after authorisation of the medicinal product is important. It allows continued monitoring of the benefit/risk balance of the medicinal product. Healthcare professionals are asked to report any suspected adverse reactions via:

 

United Kingdom

Yellow Card Scheme

Website: www.mhra.gov.uk/yellowcard

 

Ireland

HPRAIMB Pharmacovigilance

Earlsfort Terrace

IRL - Dublin 2

Tel: +353 1 6764971

Fax: +353 1 6762517

Website: www.imbhpra.ie

e-mail: imbpharmacovigilance@imb.iemedsafety@hpra.ie

 

Malta

ADR Reporting

The Medicines Authority

Post-Licensing Directorate

203 Level 3, Rue D'Argens

GŻR-1368 Gżira

Website: www.medicinesauthority.gov.mt

e-mail: postlicensing.medicinesauthority@gov.mt

 

Ísland

til Lyfjastofnunar,  www.lyfjastofnun.is

 

Updated on 11 August 2014

Reasons for updating

  • Addition of information on reporting a side effect.

Updated on 11 June 2014

Reasons for updating

  • Change to section 4.5 - Interaction with other medicinal products and other forms of interaction
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

Highlighted changes following approval of the PSUR (EMEA/H/C/000773/PSUV/0038), to include new information related to CT OVC-1002-1003 (interactions).

 

 

4.5       Interaction with other medicinal products and other forms of interaction

 

Effects of other substances on trabectedin

 

Interaction studies have only been performed in adults.

 

In vivo interaction studies have not been performed. Since trabectedin is metabolised mainly by CYP3A4, co-administration of substances that inhibit this isoenzyme e.g. ketoconazole, fluconazole ritonavir, clarithromycin or aprepitant could decrease metabolism and increase trabectedin concentrations. If such combinations are needed, close monitoring of toxicities is required (see section 4.4). Likewise co‑administration with potent inducers of this enzyme (e.g. rifampicin, phenobarbital, Saint John’s Wort) may decrease the systemic exposure to trabectedin. the concentrations of trabectedin in plasma are likely to be increased in patients who are co-administered drugs that potently inhibit the activity of this isoenzyme. Similarly, the co-administration of trabectedin with potent inducers of CPY3A4 may increase the metabolic clearance of trabectedin. Two in vivo drug-drug interaction phase 1 studies have confirmed trends toward increased and decreased trabectedin exposures when administered with ketoconazole and rifampicin, respectively.

 

When ketoconazole was co-administered with trabectedin, the plasma exposure of trabectedin was increased by approximately 21% for Cmax and 66% for AUC, but no new safety concerns were identified. Close monitoring of toxicities is required in patients receiving trabectedin in combination with potent CYP3A4 inhibitors (e.g. oral ketoconazole, fluconazole, ritonavir, clarithromycin or aprepitant) and such combinations should be avoided if possible. If such combinations are needed, appropriate dose adjustments should be applied in the event of toxicities (see sections 4.2 and 4.4).

 

When rifampicin was co-administered with trabectedin, it resulted in reduced plasma exposure of trabectedin by approximately 22% for Cmax and 31% for AUC. Therefore, the concomitant use of trabectedin with strong CYP3A4 inducers (e.g., rifampicin, phenobarbital, Saint John’s Wort) should be avoided if possible (see section 4.4).

 

Alcohol consumption must be avoided during treatment with trabectedin due to the hepatotoxicity of the medicinal product (see section 4.4).

 

Preclinical data have demonstrated that trabectedin is a substrate to P‑gp. Concomitant administration of inhibitors of P-gp, e.g. cyclosporine and verapamil, may alter trabectedin distribution and/or elimination. The relevance of this interaction e.g. central nervous system (CNS) toxicity has not been established. Caution should be taken in such situations.

 

 

10.       DATE OF REVISION OF THE TEXT

 

03/201404/2014

 

 

Updated on 04 June 2014

Reasons for updating

  • Change to side-effects
  • Change to drug interactions
  • Change to date of revision

Updated on 25 March 2014

Reasons for updating

  • Change to section 4.8 - Undesirable effects
  • Change to section 6.3 - Shelf life
  • Change to section 9 - Date of renewal of authorisation
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

Highlighted changes following approval of the variation procedure IB-0039

 

 

4.8  Undesirable effects

 

Summary of the safety profile

 

Unless otherwise specified, the following safety profile of Yondelis is based on the evaluation in clinical trials of patients treated with the recommended treatment regimens for both indications.

 

Most patients treated with Yondelis can be expected to have adverse reactions of any grade (91% in monotherapy and 99% in combination therapy) and less than one third serious adverse reactions of grade 3 or 4 severity (10% in monotherapy and 25% in combination therapy). The most common adverse reactions of any severity grade were neutropenia, nausea, vomiting, increases in AST/ALT, anemia, fatigue, thrombocytopenia, anorexia and diarrhoea.

 

Fatal adverse reactions have occurred in 1.9% and 0.9% of patients treated with the monotherapy and combination regimens respectively. They were often the result of a combination of events including pancytopenia, febrile neutropenia, some of them with sepsis, hepatic involvement, renal or multiorgan failure and rhabdomyolysis.

 

Tabulated summary of adverse reactions

 

The frequencies of the adverse reactions reported below are classified as very common ( ≥ 1/10), common ( ≥ 1/100 to < 1/10) and uncommon ( ≥ 1/1,000 to < 1/100).

 

The table below displays the adverse reactions reported in ≥ 1% of patients treated with the soft tissue sarcoma recommended regimen (1.5 mg/m2, 24 hour infusion every 3 weeks) according to the standard MedDRA (Medical Dictionary for Regulatory Activities) system organ class. Both adverse reactions and laboratory values have been used to provide frequencies. Within each frequency grouping, undesirable effects are presented in order of decreasing seriousness.

 

System Organ Class

Adverse reactions reported in ≥ 1% of patients with soft tissue sarcoma in clinical trials.

Infections and Infestations

Common

Infection

Blood and Lymphatic System Disorders

Very Common

Neutropenia* (Grade 3 = 26%, Grade 4 = 24%),

Thrombocytopenia* (Grade 3 = 11%, Grade 4 = 2%),

Anaemia* (Grade 3 = 10%, Grade 4 = 3%), Leukopenia*

Common

Febrile neutropenia

Metabolism and Nutrition Disorders

Very Common

Anorexia (Grade 3‑4 < 1%)

Common

Dehydration, Decreased appetite, Hypokalaemia

Psychiatric Disorders

Common

Insomnia

Nervous System Disorders

Very Common

Headache

Common

Peripheral sensory neuropathy, Dysgeusia, Dizziness, Paraesthesia

Vascular Disorders

Common

Hypotension, Flushing

Respiratory, Thoracic and Mediastinal Disorders

Common

Dyspnoea (Grade 3‑4 = 2%), Cough

Gastrointestinal disorders

Very Common

Vomiting (Grade 3‑4 = 6.5%), Nausea (Grade 3‑4 = 6%),

Constipation (Grade 3‑4 < 1%)

Common

Diarrhoea (Grade 3‑4 < 1%), Stomatitis (Grade 3‑4 < 1%), Abdominal pain, Dyspepsia, Upper abdominal pain

Hepatobiliary Disorders

Very Common

Hyperbilirubinemia* (Grade 3 = 1%), Alanine aminotransferase increased* (Grade 3 = 38%, Grade 4 = 3%), Aspartate aminotransferase increased* (Grade 3 = 44%, Grade 4 = 7%), Blood alkaline phosphatase increased*, Gamma‑glutamyltransferase increased*

Skin and Subcutaneous Tissue Disorders

Common

Alopecia

 

Musculoskeletal and Connective Tissue Disorders

Common

Myalgia, Arthralgia, Back pain

 

General Disorders and Administration Site Conditions

Very Common

Fatigue (Grade 3‑4 = 9%), Asthenia (Grade 3‑4 = 1%)

Common

Pyrexia, Oedema, Oedema peripheral, Injection site reaction

Investigations

Very Common

Blood creatine phosphokinase increased* (Grade 3‑4 = 4%), Blood creatinine increased*, Blood albumin decreased*

Common

Weight decreased

 

* Derived from laboratory data

 

The table below provides the frequency and severity of undesirable effects considered possibly related to study medicinal product and reported in ≥ 5% of patients with ovarian cancer randomised to receive Yondelis 1.1 mg/m2/PLD 30 mg/m2 in the pivotal trial ET743‑OVA-301. Both adverse reactions and laboratory values have been used. Within each frequency grouping, undesirable effects are presented in order of decreasing seriousness.

 

Adverse reactions reported in ≥ 5% of patients in clinical trial ET743-OVA-301


System Organ Class

Frequency

Event

Yondelis+PLD
n=333

All Grades

(%)

Grade 3

(%)

Grade 4

(%)

Blood and lymphatic system disorders

 

Very common

 

Neutropenia*

91.6

29.7

42.3

Leukopenia*

94.9

44.7

17.7

Anaemia*

94.9

12.9

5.7

Thrombocytopenia*

63.7

12.3

10.8

Common

Febrile neutropenia*

6.9

4.5

2.4

Metabolism and nutrition disorders

 

Very common

Anorexia

28.8

2.1

 

Common

 

Hypokalaemia

6.3

2.1

 

Nervous system disorders

 

Common

 

Headache

6.6

0.3

 

Dysgeusia

5.4

0.3

 

Respiratory, thoracic and mediastinal disorders

Common

Dyspnoea

6.6

0.3

 

Gastrointestinal disorders

 

Very common

 

Nausea

70.9

8.7

 

Vomiting

51.7

9.9

0.3

Constipation

20.4

0.9

 

Stomatitis

19.2

0.9

 

Diarrhoea

17.1

2.1

 

Common

 

Abdominal pain

9.3

0.6

 

Dyspepsia

7.5

0.3

 

Hepatobiliary disorders

Very common

 

Hyperbilirubinaemia*

(25.2)

(0.3)

 

Alanine aminotransferase increased*

96.1

45.6

4.5

Aspartate aminotransferase increased*

89.5

12.0

1.8

Blood alkaline phosphatase increased*

61.3

1.5

 

Skin and subcutaneous tissue disorders

 

Very common

 

Palmar-plantar erythrodysaesthesia syndrome

24

3.9

 

Alopecia

12

 

 

Common

 

Rash

8.1

 

 

Skin hyperpigmentation

5.4

 

 

General disorders and administration site conditions

 

 

 

Very common

 

Fatigue

42.3

5.7

0.3

Asthenia

15.3

1.2

 

Mucosal inflammation

11.4

2.1

 

Pyrexia

10.2

0.9

 

Investigations

Common

Blood creatine phosphokinase increased*

22.0

0.9

 

0.9

 

* Derived from laboratory data

 

The following reactions have been reported with a frequency below 5% in the combination arm, but are included here for their clinical relevance: neutropenic infection (< 1%), neutropenic sepsis (< 1%), pancytopenia (1.8%), bone marrow failure (1.5%), granulocytopenia (1.5%), dehydration, insomnia, peripheral sensory neuropathy, syncope, left ventricular dysfunction (< 1%), pulmonary embolism (1.2%), pulmonary edema (< 1%), cough, hepatotoxicity (< 1%), gamma-glutamyltransferase increased, bilirubin conjugated increased, musculoskeletal pain, myalgia, blood creatinine increased, oedema/peripheral oedema, catheter site reactions.

 

In the Yondelis+PLD arm, non-white (mainly Asian) patients had a higher incidence than white patients in grade 3 or 4 adverse reactions (96% versus 87%), and serious adverse reactions (44% versus 23% all grades). The differences were mainly observed in relation with neutropenia (93% versus 66%), anaemia (37% versus 14%) and thrombocytopenia (41% versus 19%). However, the incidences of clinical complications related to haematological toxicity such as severe infections or bleeding, or those leading to death or treatment termination, were similar in both subpopulations.

 

Description of selected adverse reactions

 

Most frequent adverse reactions

 

Blood and lymphatic system disorders

 

Neutropenia:

Neutropenia is the most common haematological toxicity. It followed a predictable pattern of rapid onset and reversibility, and was rarely associated with fever or infection. Neutrophil nadirs occurred at a median of 15 days and recovered within a week. The analysis per cycle performed in patients treated with the monotherapy regimen showed neutropenia of grade 3 and 4 in approximately 19% and 8% of cycles respectively.  In this population febrile neutropenia occurred in 2% of patients and in < 1% of cycles.

 

Thrombocytopenia:

Bleeding events associated to thrombocytopenia occurred in < 1% of patients treated with the monotherapy regimen.  The analysis per cycle performed in these patients showed thrombocytopenia of grade 3 and 4 in approximately 3% and < 1% of cycles respectively.

 

Anaemia:

Anaemia occurred in 93% and 94% of patients treated with the monotherapy and combination regimens respectively. The percentages of patients anaemic at baseline were 46% and 35% respectively.  The analysis per cycle performed in patients treated with the monotherapy regimen showed anaemia of grade 3 and 4 in approximately 3% and 1% of cycles respectively.

 

Hepatobiliary disorders

 

AST/ALT increases:

The median time to reach the peak values was 5 days for both AST and ALT. Most of the values had decreased to grade 1 or resolved by day 14‑15 (see section 4.4). The analysis per cycle performed in patients treated with the monotherapy regimen showed grade 3 elevations of AST and ALT in 12% and 20% of cycles respectively. Grade 4 elevations of AST and ALT occurred in 1% and 2% of cycles respectively. Most transaminase elevations improved to grade 1 or to pre-retreatment levels within 15 days, and less than 2% of cycles had recovering times longer than 25 days. ALT and AST increases did not follow a cumulative pattern but showed a tendency towards less severe elevations over time.

 

Hyperbilirubinemia:

Bilirubin peaks approximately a week after onset and resolves approximately two weeks after onset.

 

Liver function tests predicting severe toxicity (meeting Hy´s law) and clinical manifestations of severe hepatic injury were uncommon with a lower than 1% incidence of individual signs and symptoms including jaundice, hepatomegaly or liver pain. Mortality in the presence of hepatic injury occurred in less than 1% of patients in both regimens.

 

Other adverse reactions

 

CPK elevations and rhabdomyolysis: CPK elevations of any grade were observed in 23‑26% of patients in both regimens. CPK increases in association with rhabdomyolysis were reported in less than 1% of patients.

Alopecia: Alopecia was reported in approximately 3% of patients treated with the monotherapy regimen, of which the majority was grade 1 alopecia.

 

Hepatic failure: Rare cases of hepatic failure (including cases with fatal outcomes) have been reported in patients with serious underlying medical conditions treated with trabectedin, both in clinical trials and in post marketing setting. Some potential risk factors that may have contributed to increased trabectedin toxicity observed in these cases were dose management inconsistent with recommended guidelines, potential CYP3A4 interaction due to multiple competing CYP3A4 substrates or CYP3A4 inhibitors, or lack of dexamethasone prophylaxis.

 

Allergic Reactions: During clinical trials, hypersensitivity was reported in 2% of patients receiving trabectedin either alone or in combination with PLD, and most of these cases were Grade 1 or 2 in severity.

 

During post marketing experience, hypersensitivity reactions with very rare occurrence of fatal outcome, have been reported in association with trabectedin administration either alone or in combination with PLD (see sections 4.3 and 4.4).

 

Extravasation and Tissue necrosis: During post-marketing surveillance, a few cases of trabectedin extravasation with subsequent tissue necrosis requiring debridement have been reported (see section 4.4).

 

Septic shock: Cases of septic shock, some of which were fatal, have been uncommonly reported in clinical studies and postmarketing experience, in patients treated either with monotherapy or combination therapy.

 

Reporting of suspected adverse reactions

Reporting suspected adverse reactions after authorisation of the medicinal product is important. It allows continued monitoring of the benefit/risk balance of the medicinal product. Healthcare professionals are asked to report any suspected adverse reactions via:

 

United Kingdom

Yellow Card Scheme

Website: www.mhra.gov.uk/yellowcard

 

Ireland

IMB Pharmacovigilance Section

Irish Medicines Board

Kevin O’Malley House

Earlsfort Centre

Earlsfort Terrace

IRL - Dublin 2

Tel: +353 1 6764971

Fax: +353 1 6762517

Website: www.imb.ie

e-mail: imbpharmacovigilance@imb.ie

 


Malta

ADR Reporting

The Medicines Authority

Post-Licensing Directorate

203 Level 3, Rue D'Argens

GŻR-1368 Gżira

Website: www.medicinesauthority.gov.mt

e-mail: postlicensing.medicinesauthority@gov.mt

 

Ísland

til Lyfjastofnunar,  www.lyfjastofnun.is

 

 

6.3 Shelf life

 

Unopened vials:

 

3660 months.

 

After reconstitution

 

Chemical and physical stability has been demonstrated for 30 hours up to 25ºC.

 

From a microbiological point of view, the reconstituted solution should be diluted and used immediately. If not diluted and used immediately, in‑use storage times and conditions prior to use of the reconstituted product are the responsibility of the user and would normally not be longer than 24 hours at 2ºC to 8ºC, unless reconstitution has taken place in controlled and validated aseptic conditions.

 

After dilution

 

Chemical and physical stability has been demonstrated for 30 hours up to 25ºC.

 

 

9. DATE OF FIRST AUTHORISATION / RENEWAL OF THE AUTHORISATION

 

Date of first authorisation: 17 September 2007

Date of latest renewal: 17 September03 August 2012

 

 

10.       DATE OF REVISION OF THE TEXT

 

06/201303/2014

 

 

Updated on 20 March 2014

Reasons for updating

  • Change to side-effects
  • Change to date of revision

Updated on 06 August 2013

Reasons for updating

  • Addition of black triangle
  • Change to section 4.2 - Posology and method of administration
  • Change to section 4.4 - Special warnings and precautions for use
  • Change to Section 4.8 – Undesirable effects - how to report a side effect
  • Change to section 6.6 - Special precautions for disposal and other handling
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

  

[Black Triangle symbol] This medicinal product is subject to additional monitoring. This will allow quick identification of new safety information. Healthcare professionals are asked to report any suspected adverse reactions. See section 4.8 for how to report adverse reactions

 

4.2       Posology and method of administration

 

Yondelis must be administered under the supervision of a physician experienced in the use of chemotherapy. Its use should be confined to qualified oncologists or other health professionals specialised in the administration of cytotoxic agents.

 

Posology

 

For the treatment of soft tissue sarcoma, the recommended dose is 1.5 mg/m2 body surface area, administered as an intravenous infusion over 24 hours with a three‑week interval between cycles.

 

For the treatment of ovarian cancer Yondelis is administered every three weeks as a 3‑hour infusion at a dose of 1.1 mg/m2, immediately after PLD 30 mg/m2. To minimize the risk of PLD infusion reactions, the initial dose is administered at a rate no greater than 1 mg/minute. If no infusion reaction is observed, subsequent PLD infusions may be administered over a 1‑hour period. (see also PLD Summary of Product Characteristics [SmPC] for specific administration advice).

 

All patients must receive corticosteroids e.g. 20 mg of dexamethasone intravenously 30 minutes prior to PLD (in combination therapy) or Yondelis (in monotherapy); not only as anti‑emetic prophylaxis, but also because it appears to provide hepatoprotective effects. Additional anti‑emetics may be administered as needed.

 

The following criteria are required to allow treatment with Yondelis:

 

-             Absolute neutrophil count (ANC) ≥ 1,500/mm3

-             Platelet count ≥ 100,000/mm3

-             Bilirubin ≤ upper limit of normal (ULN)

-             Alkaline phosphatase ≤ 2.5 x ULN (consider hepatic isoenzymes 5‑nucleotidase or gamma glutamyl transpeptidase (GGT), if the elevation could be osseous in origin).

-             Albumin ≥ 25 g/l.

-             Alanine aminotransferase (ALT) and Aspartate aminotransferase (AST) ≤ 2.5 x ULN

-             Creatinine clearance ≥ 30 ml/min (monotherapy), serum creatinine £ 1.5 mg/dl (£ 132.6 μmol/l) or creatinine clearance ³ 60 ml/min (combination therapy)

-             Creatine phosphokinase (CPK) ≤ 2.5 x ULN

-             Haemoglobin ≥ 9 g/dl

 

The same criteria as above must be met prior to re‑treatment. Otherwise treatment must be delayed for up to 3 weeks until the criteria are met.

 

Additional monitoring of haematological parameters bilirubin, alkaline phosphatase, aminotransferases and CPK should occur weekly during the first two cycles of therapy, and at least once between treatments in subsequent cycles.

 

The same dose should be given for all cycles provided that no grade 3‑4 toxicities are seen and that the patient fulfils the re‑treatment criteria.

 


Dose adjustments during treatment

 

Prior to re‑treatment, patients must fulfil the baseline criteria defined above. If any of the following events occur at any time between cycles, the dose must be reduced one level, according to table 1 below, for subsequent cycles:

 

-             Neutropenia < 500/mm3 lasting for more than 5 days or associated with fever or infection

-             Thrombocytopenia < 25,000/mm3

-             Increase of bilirubin > ULN and/or alkaline phosphatase > 2.5 x ULN

-             Increase of aminotransferases (AST or ALT) > 2.5 x ULN (monotherapy) or > 5 x ULN (combination therapy), which has not recovered by day 21

-             Any other grade 3 or 4 adverse reactions (such as nausea, vomiting, fatigue)

 

Once a dose has been reduced because of toxicity, dose escalation in the subsequent cycles is not recommended. If any of these toxicities reappear in subsequent cycles in a patient exhibiting clinical benefit, the dose may be further reduced (see below). Colony stimulating factors can be administered for haematologic toxicity according to local standard practice.

 

Table 1 Dose modification table for Yondelis (as single agent for soft tissue sarcoma (STS) or

in combination for ovarian cancer) and PLD

 

Soft tissue sarcoma

Ovarian cancer

 

Yondelis

Yondelis

PLD

Starting dose

1.5 mg/m2

1.1 mg/m2

30 mg/m2  

First reduction

1.2 mg/m2

0.9 mg/m2

25 mg/m2  

Second reduction

1 mg/m2

0.75 mg/m2

20 mg/m2  

 

See the PLD SmPC for more detailed information on PLD dose adjustments.

 

In the event that further dose reductions are necessary, treatment discontinuation should be considered.

 

Duration of treatment

 

In clinical trials, there were no pre‑defined limits to the number of cycles administered. Treatment continued whilst clinical benefit was noted. Yondelis has been administered for 6 or more cycles in 29.5% and 52% of patients treated with the monotherapy and combination dose and schedule respectively. The monotherapy and combination regimens have been used for up to 38 and 21 cycles respectively. No cumulative toxicities have been observed in patients treated with multiple cycles.

 

Paediatric population

Yondelis should not be used in children below 18 years with paediatric sarcomas because of efficacy concerns (see 5.1 for results of paediatric sarcoma study).

 

Elderly patientsOlder people

 

No specific studies in elderly patientsolder people have been performed. Overall 20% of the 1,164 patients in the integrated safety analysis of monotherapy clinical trials were over 65 years. Of the 333 patients with ovarian cancer who received trabectedin in combination with PLD, 24% were 65 years of age or older and 6% were over 75 years.  No relevant differences in the safety profile were seen in this patient population. It seems that plasma clearance and distribution volume of trabectedin are not influenced by age. Therefore, dose adjustments based uniquely on age criteria are not routinely recommended.

 


Patients with hepatic impairment

 

No studies with the proposed regimen have been conducted in patients with liver dysfunction. Thus, data are not available to recommend a lower starting dose in patients with hepatic impairment. However, special caution is advised and dose adjustments may be necessary in these patients since systemic exposure is probably increased and the risk of hepatotoxicity might be increased. Patients with elevated bilirubin must not be treated with Yondelis (see section 4.4).

 

Patients with renal impairment

 

Studies including patients with renal insufficiency (creatinine clearance < 30 ml/min for the monotherapy, and < 60 ml/min for the combination regimen) have not been conducted and therefore Yondelis must not be used in this patient population (see section 4.4). Considering the pharmacokinetic characteristics of trabectedin (see section 5.2), no dose adjustments are warranted in patients with mild or moderate renal impairment.

 

Method of administration

 

Intravenous administration through a central venous line is strongly recommended (see section 6.6).

 

For instructions on reconstitution and dilution of the medicinal product before administration, see section 6.6.

 

 

4.4 Special warnings and precautions for use

 

Hepatic impairment

 

Patients must meet specific criteria on hepatic function parameters to start treatment with Yondelis. Since systemic exposure to trabectedin is probably increased due to hepatic impairment and therefore the risk of hepatotoxicity might be increased, patients with clinically relevant liver diseases, such as active chronic hepatitis, must be closely monitored and the dose adjusted if needed.  Patients with elevated bilirubin must not be treated with trabectedin (see section 4.2).

 

Renal impairment

 

Creatinine clearance must be monitored prior to and during treatment.  Yondelis monotherapy and combination regimens must not be used in patients with creatinine clearance < 30 ml/min and < 60 ml/min respectively (see section 4.2).

 

Neutropenia and thrombocytopenia

 

Grades 3 or 4 neutropenia and thrombocytopenia associated with Yondelis therapy have been very commonly reported. A full blood cell count including differential and platelet count must be performed at baseline, weekly for the first two cycles and then once between cycles (see section 4.2). Patients who develop fever should promptly seek medical attention. If this occurs, active supportive therapy should be started immediately.

 

Yondelis should not be administered to patients with baseline neutrophil counts of less than 1,500 cells/mm3 and platelets count of less than 100,000 cells/mm3. If severe neutropenia (ANC < 500 cells/mm3) lasting more than 5 days or associated with fever or infection occurs, dose reduction is recommended (see section 4.2).

 


Nausea and vomiting

 

Anti-emetic prophylaxis with corticosteroids such as dexamethasone must be administered to all patients (see section 4.2).

 

Rhabdomyolysis and severe CPK elevations ( > 5 x ULN)

 

Trabectedin must not be used in patients with CPK > 2.5 x  ULN (see section 4.2). Rhabdomyolysis has been uncommonly reported, usually in association with myelotoxicity, severe liver function test abnormalities and/or renal or multiorgan failure. Therefore, CPK should be closely monitored whenever a patient may be experiencing any of these toxicities or muscle weakness or muscle pain. If rhabdomyolysis occurs, supportive measures such as parenteral hydration, urine alkalinisation and dialysis should be promptly established, as indicated. Treatment with Yondelis should be discontinued until the patient fully recovers.

 

Caution should be taken if medicinal products associated with rhabdomyolysis (e.g. statins), are administered concomitantly with trabectedin, since the risk of rhabdomyolysis may be increased

 

Liver Function Test (LFT) abnormalities

 

Reversible acute increases in aspartate aminotransferase (AST) and alanine aminotransferase (ALT) have been reported in most patients. Yondelis must not be used in patients with elevated bilirubin. Patients with increases in AST, ALT and alkaline phosphatase between cycles may necessitate dose reduction (see section 4.2).

 

Injection site reactions

 

The use of central venous access is strongly recommended (see section 4.2). Patients may develop a potentially severe injection site reaction when trabectedin is administered through a peripheral venous line.

 

Trabectedin extravasation may cause tissue necrosis requiring debridement. There is no specific antidote for extravasation of trabectedin. Extravasation should be managed by local standard practice.

 

Allergic Reactions

 

During postmarketing experience, hypersensitivity reactions with very rare occurrence of fatal outcome, have been reported in association with trabectedin administration either alone or in combination with PLD (see sections 4.3 and 4.8).

 

Others

 

Co‑administration of Yondelis with potent inhibitors of the enzyme CYP3A4 should be avoided (see section 4.5). If this is not possible, close monitoring of toxicities are required and dose reductions of trabectedin should be considered.

 

Caution should be taken if medicinal products associated with hepatotoxicity are administered concomitantly with trabectedin, since the risk of hepatotoxicity may be increased.

 

Concomitant use of trabectedin with phenytoin may reduce phenytoin absorption leading to an exacerbation of convulsions. Combination of trabectedin with phenytoin or live attenuated vaccines is not recommended and with yellow fever vaccine is specifically contraindicated (see section 4.3).

 

The concomitant use of trabectedin with alcohol must be avoided (see section 4.5).

 

Women of childbearing potential must use effective contraception during treatment and 3 months thereafter, and immediately inform the treating physician if a pregnancy occurs (see section 5.3).

Men in fertile age must use effective contraception during treatment and 5 months after treatment  (see section 4.6).

 

This medicine contains potassium, less than 1 mmol (39 mg) per vial, i.e. essentially “potassium‑free”.

 

See also PLD Summary of Product Characteristics for more detailed information on warnings and precautions.

 

 

4.8  Undesirable effects

 

Summary of the safety profile

 

Unless otherwise specified, the following safety profile of Yondelis is based on the evaluation in clinical trials of patients treated with the recommended treatment regimens for both indications.

 

Most patients treated with Yondelis can be expected to have adverse reactions of any grade (91% in monotherapy and 99% in combination therapy) and less than one third serious adverse reactions of grade 3 or 4 severity (10% in monotherapy and 25% in combination therapy). The most common adverse reactions of any severity grade were neutropenia, nausea, vomiting, increases in AST/ALT, anemia, fatigue, thrombocytopenia, anorexia and diarrhoea.

 

Fatal adverse reactions have occurred in 1.9% and 0.9% of patients treated with the monotherapy and combination regimens respectively. They were often the result of a combination of events including pancytopenia, febrile neutropenia, some of them with sepsis, hepatic involvement, renal or multiorgan failure and rhabdomyolysis.

 

Tabulated summary of adverse reactions

 

The frequencies of the adverse reactions reported below are classified as very common ( ≥ 1/10), common ( ≥ 1/100 to < 1/10) and uncommon ( ≥ 1/1,000 to < 1/100).

 

The table below displays the adverse reactions reported in ≥ 1% of patients treated with the soft tissue sarcoma recommended regimen (1.5 mg/m2, 24 hour infusion every 3 weeks) according to the standard MedDRA (Medical Dictionary for Regulatory Activities) system organ class. Both adverse reactions and laboratory values have been used to provide frequencies. Within each frequency grouping, undesirable effects are presented in order of decreasing seriousness.

 

System Organ Class

Adverse reactions reported in ≥ 1% of patients with soft tissue sarcoma in clinical trials.

Infections and Infestations

Common

Infection

Blood and Lymphatic System Disorders

Very Common

Neutropenia* (Grade 3 = 26%, Grade 4 = 24%),

Thrombocytopenia* (Grade 3 = 11%, Grade 4 = 2%),

Anaemia* (Grade 3 = 10%, Grade 4 = 3%), Leukopenia*

Common

Febrile neutropenia

Metabolism and Nutrition Disorders

Very Common

Anorexia (Grade 3‑4 < 1%)

Common

Dehydration, Decreased appetite, Hypokalaemia

Psychiatric Disorders

Common

Insomnia

Nervous System Disorders

Very Common

Headache

Common

Peripheral sensory neuropathy, Dysgeusia, Dizziness, Paraesthesia

Vascular Disorders

Common

Hypotension, Flushing

Respiratory, Thoracic and Mediastinal Disorders

Common

Dyspnoea (Grade 3‑4 = 2%), Cough

Gastrointestinal disorders

Very Common

Vomiting (Grade 3‑4 = 6.5%), Nausea (Grade 3‑4 = 6%),

Constipation (Grade 3‑4 < 1%)

Common

Diarrhoea (Grade 3‑4 < 1%), Stomatitis (Grade 3‑4 < 1%), Abdominal pain, Dyspepsia, Upper abdominal pain

Hepatobiliary Disorders

Very Common

Hyperbilirubinemia* (Grade 3 = 1%), Alanine aminotransferase increased* (Grade 3 = 38%, Grade 4 = 3%), Aspartate aminotransferase increased* (Grade 3 = 44%, Grade 4 = 7%), Blood alkaline phosphatase increased*, Gamma‑glutamyltransferase increased*

Skin and Subcutaneous Tissue Disorders

Common

Alopecia

 

Musculoskeletal and Connective Tissue Disorders

Common

Myalgia, Arthralgia, Back pain

 

General Disorders and Administration Site Conditions

Very Common

Fatigue (Grade 3‑4 = 9%), Asthenia (Grade 3‑4 = 1%)

Common

Pyrexia, Oedema, Oedema peripheral, Injection site reaction

Investigations

Very Common

Blood creatine phosphokinase increased* (Grade 3‑4 = 4%), Blood creatinine increased*, Blood albumin decreased*

Common

Weight decreased

 

* Derived from laboratory data

 

The table below provides the frequency and severity of undesirable effects considered possibly related to study medicinal product and reported in ≥ 5% of patients with ovarian cancer randomised to receive Yondelis 1.1 mg/m2/PLD 30 mg/m2 or PLD 50 mg/m2 in the pivotal trial ET743‑OVA-301. Both adverse reactions and laboratory values have been used. Within each frequency grouping, undesirable effects are presented in order of decreasing seriousness.

 


 

 

Adverse reactions reported in ≥ 5% of patients in clinical trial ET743-OVA-301

System Organ Class

Frequency

Event

Yondelis+PLD
n=333

PLD

n=330

All
Grades

(%)

Grade 3

(%)

Grade 4

(%)

All
Grades

(%)

Grade 3

(%)

Grade 4

(%)

Blood and lymphatic system disorders

 

Very common

 

Neutropenia*

91.6

29.7

42.3

73.5

19.7

9.8

Leukopenia*

94.9

44.7

17.7

81.8

16.0

4.0

Anaemia*

94.9

12.9

5.7

82.1

6.2

2.2

Thrombocytopenia*

63.7

12.3

10.8

27.4

2.5

1.8

Common

Febrile neutropenia*

6.9

4.5

2.4

2.1

1.8

0.3

Metabolism and nutrition disorders

 

Very common

Anorexia

28.8

2.1

 

20

1.5

 

Common

 

Hypokalaemia

6.3

2.1

 

2.1

 

 

Nervous system disorders

 

Common

 

Headache

6.6

0.3

 

2.4

 

 

Dysgeusia

5.4

0.3

 

2.7

 

 

Respiratory, thoracic and mediastinal disorders

Common

Dyspnoea

6.6

0.3

 

3.3

0.3

0.3

Gastrointestinal disorders

 

Very common

 

Nausea

70.9

8.7

 

37.6

2.4

 

Vomiting

51.7

9.9

0.3

23.9

2.1

 

Constipation

20.4

0.9

 

15.5

0.3

 

Stomatitis

19.2

0.9

 

31.2

4.8

0.3

Diarrhoea

17.1

2.1

 

10

1.2

 

Common

 

Abdominal pain

9.3

0.6

 

7

0.9

 

Dyspepsia

7.5

0.3

 

6.1

0.6

 

Hepatobiliary disorders

Very common

 

Hyperbilirubinaemia*

(25.2)

(0.3)

 

(12.9)

(0.3)

 

Alanine aminotransferase increased*

96.1

45.6

4.5

36.0

2.2

 

Aspartate aminotransferase increased*

89.5

12.0

1.8

42.6

1.2

0.3

Blood alkaline phosphatase increased*

61.3

1.5

 

41.8

1.2

 

Skin and subcutaneous tissue disorders

 

Very common

 

Palmar-plantar erythrodysaesthesia syndrome

24

3.9

 

53.6

18.5

1.2

Alopecia

12

 

 

13.3

0.3

 

Common

 

Rash

8.1

 

 

16.1

0.9

 

Skin hyperpigmentation

5.4

 

 

7

 

 

General disorders and administration site conditions

 

 

 

Very common

 

Fatigue

42.3

5.7

0.3

29.7

2.4

0.3

Asthenia

15.3

1.2

 

9.1

0.3

 

Mucosal inflammation

11.4

2.1

 

18.8

5.8

 

Pyrexia

10.2

0.9

 

4.5

0.3

 

Investigations

Common

Blood creatine phosphokinase increased*

22.0

0.9

 

0.9

 

13.7

 

 

 

* Derived from laboratory data

 

The following reactions have been reported with a frequency below 5% in the combination arm, but are included here for their clinical relevance: neutropenic infection (< 1%), neutropenic sepsis (< 1%), pancytopenia (1.8%), bone marrow failure (1.5%), granulocytopenia (1.5%), dehydration, insomnia, peripheral sensory neuropathy, syncope, left ventricular dysfunction (< 1%), pulmonary embolism (1.2%), pulmonary edema (< 1%), cough, hepatotoxicity (< 1%), gamma-glutamyltransferase increased, bilirubin conjugated increased, musculoskeletal pain, myalgia, blood creatinine increased, oedema/peripheral oedema, catheter site reactions.

 

In the Yondelis+PLD arm, non-white (mainly Asian) patients had a higher incidence than white patients in grade 3 or 4 adverse reactions (96% versus 87%), and serious adverse reactions (44% versus 23% all grades). The differences were mainly observed in relation with neutropenia (93% versus 66%), anaemia (37% versus 14%) and thrombocytopenia (41% versus 19%). However, the incidences of clinical complications related to haematological toxicity such as severe infections or bleeding, or those leading to death or treatment termination, were similar in both subpopulations.

 

Description of selected adverse reactions

 

Most frequent adverse reactions

 

Blood and lymphatic system disorders

 

Neutropenia:

Neutropenia is the most common haematological toxicity. It followed a predictable pattern of rapid onset and reversibility, and was rarely associated with fever or infection. Neutrophil nadirs occurred at a median of 15 days and recovered within a week. The analysis per cycle performed in patients treated with the monotherapy regimen showed neutropenia of grade 3 and 4 in approximately 19% and 8% of cycles respectively.  In this population febrile neutropenia occurred in 2% of patients and in < 1% of cycles.

 

Thrombocytopenia:

Bleeding events associated to thrombocytopenia occurred in < 1% of patients treated with the monotherapy regimen.  The analysis per cycle performed in these patients showed thrombocytopenia of grade 3 and 4 in approximately 3% and < 1% of cycles respectively.

 

Anaemia:

Anaemia occurred in 93% and 94% of patients treated with the monotherapy and combination regimens respectively. The percentages of patients anaemic at baseline were 46% and 35% respectively.  The analysis per cycle performed in patients treated with the monotherapy regimen showed anaemia of grade 3 and 4 in approximately 3% and 1% of cycles respectively.

 

Hepatobiliary disorders

 

AST/ALT increases:

The median time to reach the peak values was 5 days for both AST and ALT. Most of the values had decreased to grade 1 or resolved by day 14‑15 (see section 4.4). The analysis per cycle performed in patients treated with the monotherapy regimen showed grade 3 elevations of AST and ALT in 12% and 20% of cycles respectively. Grade 4 elevations of AST and ALT occurred in 1% and 2% of cycles respectively. Most transaminase elevations improved to grade 1 or to pre-retreatment levels within 15 days, and less than 2% of cycles had recovering times longer than 25 days. ALT and AST increases did not follow a cumulative pattern but showed a tendency towards less severe elevations over time.

 

Hyperbilirubinemia:

Bilirubin peaks approximately a week after onset and resolves approximately two weeks after onset.

 

Liver function tests predicting severe toxicity (meeting Hy´s law) and clinical manifestations of severe hepatic injury were uncommon with a lower than 1% incidence of individual signs and symptoms including jaundice, hepatomegaly or liver pain. Mortality in the presence of hepatic injury occurred in less than 1% of patients in both regimens.

 

Other adverse reactions

 

CPK elevations and rhabdomyolysis: CPK elevations of any grade were observed in 23‑26% of patients in both regimens. CPK increases in association with rhabdomyolysis were reported in less than 1% of patients.

 

Alopecia: Alopecia was reported in approximately 3% of patients treated with the monotherapy regimen, of which the majority was grade 1 alopecia.

 

Hepatic failure: Rare cases of hepatic failure (including cases with fatal outcomes) have been reported in patients with serious underlying medical conditions treated with trabectedin, both in clinical trials and in post marketing setting. Some potential risk factors that may have contributed to increased trabectedin toxicity observed in these cases were dose management inconsistent with recommended guidelines, potential CYP3A4 interaction due to multiple competing CYP3A4 substrates or CYP3A4 inhibitors, or lack of dexamethasone prophylaxis.

 

Allergic Reactions: During clinical trials, hypersensitivity was reported in 2% of patients receiving trabectedin either alone or in combination with PLD, and most of these cases were Grade 1 or 2 in severity.

 

During post marketing experience, hypersensitivity reactions with very rare occurrence of fatal outcome, have been reported in association with trabectedin administration either alone or in combination with PLD (see sections 4.3 and 4.4).

 

Extravasation and Tissue necrosis: During post-marketing surveillance, a few cases of trabectedin extravasation with subsequent tissue necrosis requiring debridement have been reported (see section 4.4).

 

Septic shock: Cases of septic shock, some of which were fatal, have been uncommonly reported in clinical studies and postmarketing experience, in patients treated either with monotherapy or combination therapy.

 

Reporting of suspected adverse reactions

Reporting suspected adverse reactions after authorisation of the medicinal product is important. It allows continued monitoring of the benefit/risk balance of the medicinal product. Healthcare professionals are asked to report any suspected adverse reactions via:

 

United Kingdom

Yellow Card Scheme

Website: www.mhra.gov.uk/yellowcard

 

Ireland

Pharmacovigilance Section

Irish Medicines Board

Kevin O’Malley House

Earlsfort Centre

Earlsfort Terrace

IRL - Dublin 2

Tel: +353 1 6764971

Fax: +353 1 6762517

Website: www.imb.ie

e-mail: imbpharmacovigilance@imb.ie

 

Malta

ADR Reporting

The Medicines Authority

Post-Licensing Directorate

203 Level 3, Rue D'Argens

GŻR-1368 Gżira

Website: www.medicinesauthority.gov.mt

e-mail: postlicensing.medicinesauthority@gov.mt

 

Ísland

til Lyfjastofnunar,  www.lyfjastofnun.is

 

Post-marketing experience

 

During post-marketing surveillance few cases of trabectedin extravasation with subsequent tissue necrosis requiring debridement have been reported (see section 4.4).

 

6.6 Special precautions for disposal and other handling

 

Preparation for intravenous infusion

 

0.25 mg vial

 

Yondelis must be reconstituted and further diluted prior to intravenous infusion. Appropriate aseptic techniques must be used to prepare the infusion solution (see Instructions for reconstitution and for dilution).

 

Yondelis must be reconstituted and further diluted prior to infusion. When used in combination with PLD the intravenous line should be flushed well with 50 mg/ml (5%) glucose solution for infusion after administration of PLD and before administration of Yondelis. The use of any diluent other than 50 mg/ml (5%) glucose solution for infusion for this line flushing may cause precipitation of PLD (see also PLD Summary of Product Characteristics for specific handling instructions).

 

Instructions for reconstitution

 

Each vial containing 0.25 mg of trabectedin is reconstituted with 5 ml of sterile water for injections. The solution obtained has a concentration of 0.05 mg/ml and is for single‑use only.

 

A syringe is used to inject 5 ml of sterile water for injections into the vial. The vial must be shaken until complete dissolution. The reconstituted solution results in a clear, colourless or slightly yellowish solution, essentially free of visible particles.

 

Preparation for intravenous infusion

 

1 mg vial

 

Yondelis must be reconstituted and further diluted prior to intravenous infusionAppropriate aseptic techniques must be used to prepare the infusion solution (see Instructions for reconstitution and for dilution).

 

Yondelis must be reconstituted and further diluted prior to infusion. When used in combination with PLD the intravenous line should be flushed well with 50 mg/ml (5%) glucose solution for infusion after administration of PLD and before administration of Yondelis. The use of any diluent other than 50 mg/ml (5%) glucose solution for infusion for this line flushing may cause precipitation of PLD (see also PLD Summary of Product Characteristics for specific handling instructions).

 

Instructions for reconstitution

 

Each vial containing 1 mg of trabectedin is reconstituted with 20 ml of sterile water for injections. The solution obtained has a concentration of 0.05 mg/ml and is for single‑use only.

 

A syringe is used to inject 20 ml of sterile water for injections into the vial. The vial must be shaken until complete dissolution. The reconstituted solution results in a clear, colourless or slightly yellowish solution, essentially free of visible particles.

 

0.25 mg and 1 mg vials

 

This reconstituted solution contains 0.05 mg/ml of trabectedin. It requires further dilution and is for single-use only.

 

Instructions for dilution

 

The reconstituted solution should be diluted with sodium chloride 9 mg/ml (0.9%) solution for infusion or glucose 50 mg/ml (5%) solution for infusion. The required volume should be calculated as follows:

 

Volume (ml) = BSA (m2) x individual dose (mg/m2)

 

 

0.05 mg/ml

 

BSA = Body Surface Area

 

If administration is to be made through a central venous line, the appropriate amount of reconstituted solution should be withdrawn from the vial and added to an infusion bag containing ³ 50 ml of diluent (sodium chloride 9 mg/ml (0.9%) solution for infusion or glucose 50 mg/ml (5%) solution for infusion), the concentration of trabectedin in the infusion solution being ≤ 0.030 mg/ml.

 

If central venous access is not feasible and a peripheral venous line has to be used, the reconstituted solution should be added to an infusion bag containing ³ 1,000 ml of diluent (sodium chloride 9 mg/ml (0.9%) solution for infusion or glucose 50 mg/ml (5%) solution for infusion).

 

Parenteral solutions should be inspected visually for particles prior to administration. Once the infusion is prepared, it should be administered immediately.

 

Instructions for handling and disposal

 

Yondelis is a cytotoxic anticancer medicinal product and, as with other potentially toxic compounds, caution should be exercised during handling. Procedures for proper handling and disposal of cytotoxic medicinal products must be followed. Personnel should be trained in the correct techniques to reconstitute and dilute the medicinal product and should wear protective clothing including mask, goggles and gloves during the reconstitution and dilution. Pregnant staff must be excluded from working with this medicinal product.

 

Accidental contact with the skin, eyes or mucous membranes must be treated immediately with copious amounts of water.

 

No incompatibilities have been observed between Yondelis and type I glass bottles, polyvinylchloride (PVC) and polyethylene (PE) bags and tubing, polyisoprene reservoirs and titanium implantable vascular access systems.

 

Any unused medicinal product or waste material should be disposed of in accordance with local requirements for cytotoxic medicinal products.

 

10.       DATE OF REVISION OF THE TEXT

 

13/12/201206/2013

 

Updated on 31 July 2013

Reasons for updating

  • Change to warnings or special precautions for use
  • Change to side-effects
  • Change to date of revision
  • Addition of black triangle

Updated on 06 March 2013

Reasons for updating

  • Change to section 4.2 - Posology and method of administration
  • Change to section 5.1 - Pharmacodynamic properties
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

New added text highlighted in yellow.  Deleted text marked by strickthrough

4.2       Posology and method of administration

 

Yondelis must be administered under the supervision of a physician experienced in the use of chemotherapy. Its use should be confined to qualified oncologists or other health professionals specialised in the administration of cytotoxic agents.

 

Posology

 

For the treatment of soft tissue sarcoma, the recommended dose is 1.5 mg/m2 body surface area, administered as an intravenous infusion over 24 hours with a three‑week interval between cycles.

 

For the treatment of ovarian cancer Yondelis is administered every three weeks as a 3‑hour infusion at a dose of 1.1 mg/m2, immediately after PLD 30 mg/m2. To minimize the risk of PLD infusion reactions, the initial dose is administered at a rate no greater than 1 mg/minute. If no infusion reaction is observed, subsequent PLD infusions may be administered over a 1‑hour period. (see also PLD Summary of Product Characteristics [SmPC] for specific administration advice).

 

All patients must receive corticosteroids e.g. 20 mg of dexamethasone intravenously 30 minutes prior to PLD (in combination therapy) or Yondelis (in monotherapy); not only as anti‑emetic prophylaxis, but also because it appears to provide hepatoprotective effects. Additional anti‑emetics may be administered as needed.

 

The following criteria are required to allow treatment with Yondelis:

 

-             Absolute neutrophil count (ANC) ≥ 1,500/mm3

-             Platelet count ≥ 100,000/mm3

-             Bilirubin ≤ upper limit of normal (ULN)

-             Alkaline phosphatase ≤ 2.5 x ULN (consider hepatic isoenzymes 5‑nucleotidase or gamma glutamyl transpeptidase (GGT), if the elevation could be osseous in origin).

-             Albumin ≥ 25 g/l.

-             Alanine aminotransferase (ALT) and Aspartate aminotransferase (AST) ≤ 2.5 x ULN

-             Creatinine clearance ≥ 30 ml/min (monotherapy), serum creatinine £ 1.5 mg/dl (£ 132.6 μmol/l) or creatinine clearance ³ 60 ml/min (combination therapy)

-             Creatine phosphokinase (CPK) ≤ 2.5 x ULN

-             Haemoglobin ≥ 9 g/dl

 

The same criteria as above must be met prior to re‑treatment. Otherwise treatment must be delayed for up to 3 weeks until the criteria are met.

 

Additional monitoring of haematological parameters bilirubin, alkaline phosphatase, aminotransferases and CPK should occur weekly during the first two cycles of therapy, and at least once between treatments in subsequent cycles.

 

The same dose should be given for all cycles provided that no grade 3‑4 toxicities are seen and that the patient fulfils the re‑treatment criteria.

 

Dose adjustments during treatment

 

Prior to re‑treatment, patients must fulfil the baseline criteria defined above. If any of the following events occur at any time between cycles, the dose must be reduced one level, according to table 1 below, for subsequent cycles:

 

-             Neutropenia < 500/mm3 lasting for more than 5 days or associated with fever or infection

-             Thrombocytopenia < 25,000/mm3

-             Increase of bilirubin > ULN and/or alkaline phosphatase > 2.5 x ULN

-             Increase of aminotransferases (AST or ALT) > 2.5 x ULN (monotherapy) or > 5 x ULN (combination therapy), which has not recovered by day 21

-             Any other grade 3 or 4 adverse reactions (such as nausea, vomiting, fatigue)

 

Once a dose has been reduced because of toxicity, dose escalation in the subsequent cycles is not recommended. If any of these toxicities reappear in subsequent cycles in a patient exhibiting clinical benefit, the dose may be further reduced (see below). Colony stimulating factors can be administered for haematologic toxicity according to local standard practice.

 

Table 1 Dose modification table for Yondelis (as single agent for soft tissue sarcoma (STS) or

in combination for ovarian cancer) and PLD

 

Soft tissue sarcoma

Ovarian cancer

 

Yondelis

Yondelis

PLD

Starting dose

1.5 mg/m2

1.1 mg/m2

30 mg/m2  

First reduction

1.2 mg/m2

0.9 mg/m2

25 mg/m2  

Second reduction

1 mg/m2

0.75 mg/m2

20 mg/m2  

 

See the PLD SmPC for more detailed information on PLD dose adjustments.

 

In the event that further dose reductions are necessary, treatment discontinuation should be considered.

 

Duration of treatment

 

In clinical trials, there were no pre‑defined limits to the number of cycles administered. Treatment continued whilst clinical benefit was noted. Yondelis has been administered for 6 or more cycles in 29.5% and 52% of patients treated with the monotherapy and combination dose and schedule respectively. The monotherapy and combination regimens have been used for up to 38 and 21 cycles respectively. No cumulative toxicities have been observed in patients treated with multiple cycles.

 

Paediatric population

The safety and efficacy of trabectedin in the paediatric population have not yet been established.   No data are available.Yondelis should not be used in children below 18 years with paediatric sarcomas because of efficacy concerns (see 5.1 for results of paediatric sarcoma study).

 

Elderly patients

 

No specific studies in elderly patients have been performed. Overall 20% of the 1,164 patients in the integrated safety analysis of monotherapy clinical trials were over 65 years. Of the 333 patients with ovarian cancer who received trabectedin in combination with PLD, 24% were 65 years of age or older and 6% were over 75 years.  No relevant differences in the safety profile were seen in this patient population. It seems that plasma clearance and distribution volume of trabectedin are not influenced by age. Therefore, dose adjustments based uniquely on age criteria are not routinely recommended.

 

Patients with hepatic impairment

 

No studies with the proposed regimen have been conducted in patients with liver dysfunction. Thus, data are not available to recommend a lower starting dose in patients with hepatic impairment. However, special caution is advised and dose adjustments may be necessary in these patients since systemic exposure is probably increased and the risk of hepatotoxicity might be increased. Patients with elevated bilirubin must not be treated with Yondelis (see section 4.4).

 

 

Patients with renal impairment

 

Studies including patients with renal insufficiency (creatinine clearance < 30 ml/min for the monotherapy, and < 60 ml/min for the combination regimen) have not been conducted and therefore Yondelis must not be used in this patient population (see section 4.4). Considering the pharmacokinetic characteristics of trabectedin (see section 5.2), no dose adjustments are warranted in patients with mild or moderate renal impairment.

 

Method of administration

 

Intravenous administration through a central venous line is strongly recommended (see section 6.6).

 

For instructions on reconstitution and dilution of the medicinal product before administration, see section 6.6.

 

5.         PHARMACOLOGICAL PROPERTIES

 

5.1       Pharmacodynamic properties

 

Pharmacotherapeutic group: Antineoplastic agent, ATC code: L01CX01.

 

Mechanism of action

 

Trabectedin binds to the minor groove of deoxyribonucleic acid (DNA), bending the helix to the major groove. This binding to DNA triggers a cascade of events affecting several transcription factors, DNA binding proteins, and DNA repair pathways, resulting in perturbation of the cell cycle.

 

Pharmacodynamic effects

 

Trabectedin has been shown to exert antiproliferative in vitro and in vivo activity against a range of human tumour cell lines and experimental tumours, including malignancies such as sarcoma, breast, non‑small cell lung, ovarian and melanoma.

 

Electrocardiogram (ECG) investigations

 

In a placebo‑controlled QT/QTc study, trabectedin did not prolong the QTc interval in patients with advanced solid malignancies.

 

Clinical efficacy

 

The efficacy and safety of trabectedin in soft tissue sarcoma is based in a randomised trial in patients with locally advanced or metastatic lipo- or leiomyosarcoma, whose disease had progressed or relapsed after treatment with at least anthracyclines and ifosfamide. In this trial trabectedin was administered either at 1.5 mg/m2 as a 24‑hour intravenous infusion every 3 weeks or at 0.58 mg/m2 weekly as a 3‑hour intravenous infusion for 3‑weeks of a 4‑week cycle. The protocol specified final time to progression (TTP) analysis showed a 26.6% reduction in the relative risk of progression for patients treated in the 24‑h q3wk group [Hazard Ratio (HR)=0.734, Confidence Interval (CI): 0.554‑0.974]. Median TTP values were 3.7 months (CI: 2.1‑5.4 m) in the 24‑h q3wk group and 2.3 months (CI: 2.0‑3.5 m) in the 3‑h qwk group (p=0.0302). No significant differences were detected in overall survival (OS). Median OS with the 24‑h q3wk regimen was 13.9 months (CI: 12.5‑18.6) and 60.2% of patients were alive at 1 year (CI: 52.0‑68.5%).

 

Additional efficacy data are available from 3 single‑arm Phase II trials with similar populations treated with the same regimen. These trials evaluated a total of 100 patients with lipo- and leiomyosarcoma and 83 patients with other types of sarcoma.

 

Results from an expanded access program for patients with STS (study ET743-SAR- 3002) show that among the 903 subjects assessed for OS, the median survival time was 11.9 months (95% CI: 11.2, 13.8). The median survival by histology tumor type was 16.2 months [95% CI: 14.1, 19.5] for subjects with leiomyosarcomas and liposarcomas and 8.4 months [95% CI: 7.1, 10.7] for subjects with other types of sarcomas. The median survival for subjects with liposarcoma was 18.1 months [95% CI: 15.0, 26.4] and for subjects with leiomyosarcoma 16.2 months [95% CI: 11.7, 24.3].

 

The efficacy of Yondelis/PLD combination in relapsed ovarian cancer is based on ET743‑OVA‑301, a randomized phase 3 study of 672 patients who received either trabectedin (1.1 mg/m2) and PLD (30 mg/m2) every 3 weeks or PLD (50 mg/m2) every 4 weeks. The primary analysis of progression free survival (PFS) was performed in 645 patients with measurable disease and assessed by independent radiology review. Treatment with the combination arm resulted in a 21% risk reduction for disease progression compared to PLD alone (HR=0.79, CI: 0.65‑0.96, p=0.0190). Secondary analyses of PFS and response rate also favoured the combination arm. The results of the main efficacy analyses are summarised in the table below:

 

Efficacy analyses from ET743‑OVA‑301

 

Yondelis+PLD

PLD

Hazard/Odds ratio

p‑value

Progression Free Survival

Independent radiology review,

measurable disease *

n=328

n=317

 

 

Median PFS (95% CI) (months)

7.3 (5.9‑7.9)

5.8 (5.5‑7.1)

0.79 (0.65‑0.96)

0.0190 a

12 months PFS rate (95% CI) (%)

25.8 (19.7‑32.3)

18.5 (12.9‑24.9)

 

 

Independent oncology review,

all randomised

n=336

n=335

 

 

Median PFS (95% CI) (months)

7.4 (6.4‑9.2)

5.6 (4.2‑6.8)

0.72 (0.60‑0.88)

0.0008 a

Overall Survival (Final analysis - n=522 events)

All randomised

n=337

n=335

 

 

Median OS (95% CI) (months)

22.2 (19.3‑25.0)

18.9 (17.1‑21.5)

0.86 (0.72‑1.02)

0.0835 a

Overall survival in platinum‑sensitive population (Final analysis n=316 events)

 

n=218

n=212

 

 

Median OS (95% CI) (months)

27.0 (24.1‑31.4)

24.1 (20.9‑25.9)

0.83 (0.67‑1.04)

0.1056 a

Overall Response Rate (ORR)

Independent radiology review,

all randomised

n=337

n=335

 

 

ORR (95% CI) (%)

27.6 (22.9‑32.7)

18.8 (14.8‑23.4)

1.65 (1.14‑2.37)

0.0080 b

* Primary efficacy analysis

a Log rank test

b Fisher’s test

 

Based on independent oncology review, patients with platinum‑free interval (PFI) < 6 months (35% in Yondelis+PLD and 37% in PLD arm) had similar PFS in the two arms with both showing median PFS of 3.7 months (HR=0.89, CI: 0.67‑1.20). In patients with PFI ³ 6 months (65% in Yondelis+PLD and 63% in PLD arm), median PFS was 9.7 months in the Yondelis+PLD arm compared with 7.2 months in the PLD monotherapy arm (HR=0.66, CI: 0.52‑0.85).

 

In the final analysis, the effect of the Yondelis+PLD combination vs. PLD alone on overall survival was more pronounced in patients with PFI ³ 6 months (platinum‑sensitive population: 27.0 vs. 24.1 months, HR=0.83, CI: 0.67‑1.04) than in those with PFI < 6 months (platinum‑resistant population: 14.2 vs. 12.4 months, HR=0.92, CI: 0. 70‑1.21).

 

The benefit in OS with Yondelis plus PLD was not due to the effect of subsequent therapies, which were well balanced between the two treatment arms.

 

In the multivariate analyses including PFI, treatment effect on overall survival was statistically significant favouring the Yondelis+PLD combination over PLD alone (all randomised: p=0.0285; platinum‑sensitive population: p=0.0319).

 

No data are available comparing Yondelis+PLD to a platinum‑based regimen in platinum‑sensitive patients.

 

No statistically significant differences were found between treatment arms in global measures of Quality of Life.

 

Paediatric population

 

In SAR-2005 phase I-II study, a total of 50 paediatric patients with rhabdomyosarcoma, Ewing sarcoma or non rhabdomyosarcoma soft tissue sarcoma were enrolled. Eight patients were treated with a dose of 1.3 mg/m2 and 42 with 1.5 mg/m2. Trabectedin was administered as a 24-hour intravenous infusion every 21 days. Forty patients were fully evaluable for response.  One partial response (PR) centrally confirmed was observed: overall RR: 2.5% CI95% (0.1%-13.2%). The PR corresponded to a patient with an alveolar rhabdomyosarcoma. Duration of the response was 6.5 months No responses were observed for Ewing sarcoma and NRSTS, [RR: 0% CI95% (0%-30.9%)]. Three patients achieved stable disease (one with rhabdomyosarcoma after 15 cycles, one with spindle cell sarcoma after 2 cycles, and one with Ewing sarcoma after 4 cycles.

 

Adverse reactions, included reversible elevation of liver enzymes and haematological events; in addition, fever, infection, dehydration and thrombosis/embolism were also reported.

 

This medicinal product has been authorised under “exceptional circumstances”. This means that due to the rarity of soft tissue sarcoma, it has not been possible to obtain complete information on this medicinal product in this indication.

 

The European Medicines Agency will review any new information which may become available every year and this SmPC will be updated as necessary.

 

 

10.       DATE OF REVISION OF THE TEXT

 

13/12/2012

 

 

Updated on 01 March 2013

Reasons for updating

  • Change to date of revision
  • Change to dosage and administration

Updated on 24 August 2012

Reasons for updating

  • Change to section 2 - Qualitative and quantitative composition
  • Change to section 4.1 - Therapeutic indications
  • Change to section 4.2 - Posology and method of administration
  • Change to section 4.4 - Special warnings and precautions for use
  • Change to section 4.5 - Interaction with other medicinal products and other forms of interaction
  • Change to section 4.6 - Pregnancy and lactation
  • Change to section 4.7 - Effects on ability to drive and use machines
  • Change to section 4.8 - Undesirable effects
  • Change to section 5.1 - Pharmacodynamic properties
  • Change to section 5.2 - Pharmacokinetic properties
  • Change to section 5.3 - Preclinical safety data
  • Change to section 6.1 - List of excipients
  • Change to section 6.3 - Shelf life
  • Change to section 6.5 - Nature and contents of container
  • Change to section 6.6 - Special precautions for disposal and other handling
  • Change to section 9 - Date of renewal of authorisation
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

Highlighted changes following the MHRA’s notification of Black triangle removal and following approval of the MA renewal procedure

 

 

 

2.       QUALITATIVE AND QUANTITATIVE COMPOSITION

 

0.25 mg vial

 

Each vial of powder contains 0.25 mg of trabectedin.

 

1 mg vial

 

Each vial of powder contains 1 mg of trabectedin

 

0.25 mg and 1 mg vials

 

One1 ml of reconstituted solution contains 0.05 mg of trabectedin.

 

Excipients with known effect:

 

0.25 mg vial

 

Each vial of powder contains 2 mg of potassium and 0.1 g of sucrose.

 

1 mg vial

 

Each vial of powder contains 8 mg of potassium and 0.4 g of sucrose.

 

0.25 mg and 1 mg vials

 

For the full list of excipients, see section 6.1.

 

 

4.         Clinical particulars

 

4.1       Therapeutic indications

 

Yondelis is indicated for the treatment of adult patients with advanced soft tissue sarcoma, after failure of anthracyclines and ifosfamide, or who are unsuited to receive these agents. Efficacy data are based mainly on liposarcoma and leiomyosarcoma patients.

 

Yondelis in combination with pegylated liposomal doxorubicin (PLD) is indicated for the treatment of patients with relapsed platinum‑sensitive ovarian cancer.

 

4.2       Posology and method of administration

 

Yondelis must be administered under the supervision of a physician experienced in the use of chemotherapy. Its use should be confined to qualified oncologists or other health professionals specialised in the administration of cytotoxic agents.

 

Posology

 

For the treatment of soft tissue sarcoma, the recommended dose is 1.5 mg/m2 body surface area, administered as an intravenous infusion over 24 hours with a three‑week interval between cycles.

 

For the treatment of ovarian cancer Yondelis is administered every three weeks as a 3‑hour infusion at a dose of 1.1 mg/m2, immediately after PLD 30 mg/m2. To minimize the risk of PLD infusion reactions, the initial dose is administered at a rate no greater than 1 mg/minute. If no infusion reaction is observed, subsequent PLD infusions may be administered over a 1‑hour period (see. (See also PLD Summary of Product Characteristics [SmPC] for specific administration advice).

 

All patients must receive corticosteroids e.g. 20 mg of dexamethasone intravenously 30 minutes prior to PLD (in combination therapy) or Yondelis (in monotherapy); not only as anti‑emetic prophylaxis, but also because it appears to provide hepatoprotective effects. Additional anti‑emetics may be administered as needed.

 

The following criteria are required to allow treatment with Yondelis:

 

-                 Absolute neutrophil count (ANC) ³ 1,500/mm3

-                 Platelet count ³ 100,000/mm3

-                 Bilirubin £ upper limit of normal (ULN)

-                 Alkaline phosphatase £ 2.5 x ULN (consider hepatic isoenzymes 5‑nucleotidase or gamma glutamyl transpeptidase (GGT), if the elevation could be osseous in origin).

-                 Albumin ³ 25 g/l

-                 Alanine aminotransferase (ALT) and Aspartate aminotransferase (AST) £ 2.5 x ULN

-                 Creatinine clearance ³ 30 ml/min (monotherapy), serum creatinine £ 1.5 mg/dl (£ 132.6 μmol/l) or creatinine clearance ³ 60 ml/min (combination therapy)

-                 Creatine phosphokinase (CPK) £ 2.5 x ULN

-                 Haemoglobin ³ 9 g/dl

 

The same criteria as above must be met prior to re‑treatment. Otherwise treatment must be delayed for up to 3 weeks until the criteria are met.

 

Additional monitoring of haematological parameters bilirubin, alkaline phosphatase, aminotransferases and CPK should occur weekly during the first two cycles of therapy, and at least once between treatments in subsequent cycles.

 

The same dose should be given for all cycles provided that no grade 3‑4 toxicities are seen and that the patient fulfils the re‑treatment criteria.

 

Dose adjustments during treatment

 

Prior to re‑treatment, patients must fulfil the baseline criteria defined above. If any of the following events occur at any time between cycles, the dose must be reduced one level, according to table 1 below, for subsequent cycles:

 

-                 Neutropenia < 500/mm3 lasting for more than 5 days or associated with fever or infection

-                 Thrombocytopenia < 25,000/mm3

-                 Increase of bilirubin > ULN and/or alkaline phosphatase > 2.5 x ULN

-                 Increase of aminotransferases (AST or ALT) > 2.5 x ULN (monotherapy) or > 5 x ULN (combination therapy), which has not recovered by day 21

-                 Any other grade 3 or 4 adverse reactions (such as nausea, vomiting, fatigue)

 

Once a dose has been reduced because of toxicity, dose escalation in the subsequent cycles is not recommended. If any of these toxicities reappear in subsequent cycles in a patient exhibiting clinical benefit, the dose may be further reduced (see below). Colony stimulating factors can be administered for haematologichematologic toxicity according to local standard practice.

 

Table 1 Dose modification table for Yondelis (as single agent for soft tissue sarcoma (STS) or

in combination for ovarian cancer) and PLD

 

 

Soft tissue sarcomaSoft Tissue Sarcoma

Ovarian cancerCancer

 

Yondelis

Yondelis

PLD

Starting dose

1.5 mg/m2

1.1 mg/m2

30 mg/m2  

First reduction

1.2 mg/m2

0.9 mg/m2

25 mg/m2  

Second reduction

1 mg/m2

0.75 mg/m2

20 mg/m2  

 

See the PLD SmPCSPC for more detailed information on PLD dose adjustments.

 

In the event that further dose reductions are necessary, treatment discontinuation should be considered.

 

Duration of treatment

 

In clinical trials, there were no pre‑defined limits to the number of cycles administered. Treatment continued whilst clinical benefit was noted. Yondelis has been administered for 6 or more cycles in 29.5% and 52% of patients treated with the monotherapy and combination dose and schedule respectively. The monotherapy and combination regimens have been used for up to 38 and 21 cycles respectively. No cumulative toxicities have been observed in patients treated with multiple cycles.

 

Paediatric population

 

The safety and efficacy of trabectedin in the paediatric population have not yet been established. No data are available.

 

Elderly patients

 

No specific studies in elderly patients have been performed. Overall 20% of the 1,164 patients in the integrated safety analysis of monotherapy clinical trials were over 65 years. Of the 333 patients with ovarian cancer who received trabectedin in combination with PLD, 24% were 65 years of age or older and 6% were over 75 years. No relevant differences in the safety profile were seen in this patient population. It seems that plasma clearance and distribution volume of trabectedin are not influenced by age. Therefore, dose adjustments based uniquely on age criteria are not routinely recommended.

 

Patients with hepatic impairment

 

No studies with the proposed regimen have been conducted in patients with liver dysfunction. Thus, data are not available to recommend a lower starting dose in patients with hepatic impairment. However, special caution is advised and dose adjustments may be necessary in these patients since systemic exposure is probably increased and the risk of hepatotoxicity might be increased. Patients with elevated bilirubin must not be treated with Yondelis (see section 4.4).

 

Patients with renal impairment

 

Studies including patients with renal insufficiency (creatinine clearance < 30 ml/min for the monotherapy, and < 60 ml/min for the combination regimen) have not been conducted and therefore Yondelis must not be used in this patient population (see section 4.4). Considering the pharmacokinetic characteristics of trabectedin (see section 5.2), no dose adjustments are warranted in patients with mild or moderate renal impairment.

 

Method of administration

 

Intravenous administrationAdministration through a central venous line is strongly recommended (see section 6.6).

 

For instructions on reconstitution and dilution of the medicinal product before administration, see section 6.6.

 

4.4       Special warnings and precautions for use

 

Hepatic impairment

 

Patients must meet specific criteria on hepatic function parameters to start treatment with Yondelis. Since systemic exposure to trabectedin is probably increased due to hepatic impairment and therefore the risk of hepatotoxicity might be increased, patients with clinically relevant liver diseases, such as active chronic hepatitis, must be closely monitored and the dose adjusted if needed. Patients with elevated bilirubin must not be treated with trabectedin (see section 4.2).

 

Renal impairment

 

Creatinine clearance must be monitored prior to and during treatment. Yondelis monotherapy and combination regimens must not be used in patients with creatinine clearance < 30 ml/min and < 60 ml/min respectively (see section 4.2).

 

Neutropenia and thrombocytopenia

 

Grades 3 or 4 neutropenia and thrombocytopenia associated with Yondelis therapy have been very commonly reported. A full blood cell count including differential and platelet count must be performed at baseline, weekly for the first two cycles and then once between cycles (see section 4.2). Patients who develop fever should promptly seek medical attention. If this occurs, active supportive therapy should be started immediately.

 

Yondelis should not be administered to patients with baseline neutrophil counts of less than 1,500 cells/mm3 and platelets count of less than 100,000 cells/mm3. If severe neutropenia (ANC < 500 cells/mm3) lasting more than 5 days or associated with fever or infection occurs, dose reduction is recommended (see section 4.2).

 

Nausea and vomiting

 

Anti‑emetic prophylaxis with corticosteroids such as dexamethasone must be administered to all patients (see section 4.2).

 

Rhabdomyolysis and severe CPK elevations (> 5 x ULN)

 

Trabectedin must not be used in patients with CPK > 2.5 x ULN (see section 4.2). Rhabdomyolysis has been uncommonly reported, usually in association with myelotoxicity, severe liver function test abnormalities and/or renal or multiorgan failure. Therefore, CPK should be closely monitored whenever a patient may be experiencing any of these toxicities or muscle weakness or muscle pain. If rhabdomyolysis occurs, supportive measures such as parenteral hydration, urine alkalinisation and dialysis should be promptly established, as indicated. Treatment with Yondelis should be discontinued until the patient fully recovers.

 

Caution should be taken if medicinal products associated with rhabdomyolysis (e.g. statins), are administered concomitantly with trabectedin, since the risk of rhabdomyolysis may be increased

 

Liver Function Test (LFT) abnormalities

 

Reversible acute increases in aspartate aminotransferase (AST) and alanine aminotransferase (ALT) have been reported in most patients. Yondelis must not be used in patients with elevated bilirubin. Patients with increases in AST, ALT and alkaline phosphatase between cycles may necessitate dose reduction (see section 4.2).

 

Injection site reactions

 

The use of central venous access is strongly recommended (see section 4.2). Patients may develop a potentially severe injection site reaction when trabectedin is administered through a peripheral venous line.

 

Trabectedin extravasation may cause tissue necrosis requiring debridement. There is no specific antidote for extravasation of trabectedin. Extravasation should be managed by local standard practice.

 

Others

 

Co‑administration of Yondelis with potent inhibitors of the enzyme CYP3A4 should be avoided (see section 4.5). If this is not possible, close monitoring of toxicities are required and dose reductions of trabectedin should be considered.

 

Caution should be taken if medicinal products associated with hepatotoxicity are administered concomitantly with trabectedin, since the risk of hepatotoxicity may be increased.

 

Concomitant use of trabectedin with phenytoin may reduce phenytoin absorption leading to an exacerbation of convulsions. Combination of trabectedin with phenytoin or live attenuated vaccines is not recommended and with yellow fever vaccine is specifically contraindicated (see section 4.3).

 

The concomitant use of trabectedin with alcohol must be avoided (see section 4.5).

 

WomenMen in fertile age and women of childbearing potential must use effective contraception during treatment and 3 months thereafter, for women and immediately inform the treating physician if a pregnancy occurs (see section 5.3).

Men in fertile age must use effective contraception during treatment, and 5 months after treatment for men (see section 4.6).

 

This medicine contains potassium, less than 1 mmol (39 mg) per vial, i.e. essentially “potassium‑free”.

 

See also PLD Summary of Product Characteristics for more detailed information on warnings and precautions.

 

4.5       Interaction with other medicinal products and other forms of interaction

 

Effects of other substances on trabectedin

 

In vivo interaction studies have not been performed. Since trabectedin is metabolised mainly by CYP3A4, co‑administration of substances that inhibit this isoenzyme e.g. ketoconazole, fluconazole ritonavir, clarithromycin or aprepitant could decrease metabolism and increase trabectedin concentrations. If such combinations are needed, close monitoring of toxicities is required (see section 4.4). Likewise co‑administration with potent inducers of this enzyme (e.g. rifampicin, phenobarbital, Saint John’s Wort) may decrease the systemic exposure to trabectedin.

 

Alcohol consumption must be avoided during treatment with trabectedin due to the hepatotoxicity of the medicinal product (see section 4.4).

 

Preclinical data have demonstrated that trabectedin is a substrate to P‑gp. Concomitant administration of inhibitors of P‑gp, e.g. cyclosporine and verapamil, may alter trabectedin distribution and/or elimination. The relevance of this interaction e.g. central nervous system (CNS) toxicity has not been established. Caution should be taken in such situations.

 

4.6       Fertility, pregnancy and lactation

 

Pregnancy

 

No sufficient clinical data on exposed pregnancies are available. However, based on its known mechanism of action, trabectedin may cause serious birth defects when administered during pregnancy. Trabectedin should not be used during pregnancy unless clearly necessary. If it is used during pregnancy, the patient must be informed of the potential risk to the foetus (see section 5.3) and be monitored carefully. If trabectedin is used at the end of pregnancy, potential adverse reactions should be monitored carefully in the newborns.

 

Women of childbearing potential

 

Women of childbearing potential must use effective contraception during treatment and 3 months thereafter, and immediately inform the treating physician if a pregnancy occurs (see section 5.3).

 

If pregnancy occurs during treatment the possibility of genetic counselling should be considered.

 

Breast‑feeding

 

It is not known whether trabectedin is excreted in human milk. The excretion of trabectedin in milk has not been studied in animals. Breast‑feeding is contraindicated during treatment and 3 months thereafter (see section 4.3).

 

Fertility

 

Men in fertile age and women of childbearing potential must use effective contraception during treatment and 3 months thereafter for women and immediately inform the treating physician if a pregnancy occurs (see section 5.3) and 5 months after treatment for men (see section 4.4).

 

Trabectedin can have genotoxic effects. Advice on conservation of ovules or sperm should be sought prior to treatment because of the possibility of irreversible infertility due to therapy with Yondelis.

If pregnancy occurs during treatment the possibility of genetic counselling should be considered. Genetic counselling is also recommended for patients wishing to have children after therapy.

 

4.7       Effects on ability to drive and use machines

 

No studies on the effects of the ability to drive and to use machines have been performed. However, fatigue and/or asthenia have been reported in patients receiving trabectedin. Patients who experience any of these adverse reactionsevents during therapy must not drive or operate machines.

 

4.8       Undesirable effects

 

Summary of the safety profile

 

Unless otherwise specified, the following safety profile of Yondelis is based on the evaluation in clinical trials of patients treated with the recommended treatment regimens for both indications.

 

Most patients treated with Yondelis can be expected to have adverse reactions of any grade (91% in monotherapy and 99% in combination therapy) and less than one third serious adverse reactions of grade 3 or 4 severity (10% in monotherapy and 25% in combination therapy). The most common adverse reactions of any severity grade were neutropenia, nausea, vomiting, increases in AST/ALT, anemia, fatigue, thrombocytopenia , anorexia and diarrhoea.

 

Fatal adverse reactions have occurred in 1.9% and 0.9% of patients treated with the monotherapy and combination regimens respectively. They were often the result of a combination of events including pancytopenia, febrile neutropenia, some of them with sepsis, hepatic involvement, renal or multiorgan failure and rhabdomyolysis.

 

Tabulated summary of adverseAdverse reactions

 

The frequencies of the adverse reactions reported below are classified as very common ( 1/10), common ( 1/100 to < 1/10) and uncommon ( 1/1,000 to < 1/100).

 

The table below displays the adverse reactions reported in ≥ 1% of patients treated with the soft tissue sarcoma recommended regimen (1.5 mg/m2, 24 hour infusion every 3 weeks) according to the standard MedDRA (Medical Dictionary for Regulatory Activities) system organ class. Both adverse reactions and laboratory values have been used to provide frequencies. Within each frequency grouping, undesirable effects are presented in order of decreasing seriousness.


 

System Organ Class

Adverse reactions reported in ≥ 1% of patients with soft tissue sarcoma in clinical trials.  at the recommended regimen [1.5 mg/m2, 24 hour infusion every 3 weeks ]

Infections and InfestationsInvestigations

Very Common

Blood creatine phosphokinase increased* (Grade 3‑4 = 4%), Blood creatinine increased*, Blood albumin decreased*

Common

InfectionWeight decreased

Blood and Lymphatic System Disorders

Very Common

Neutropenia* (Grade 3 = 26%, Grade 4 = 24%), Thrombocytopenia* (Grade 3 = 11%, Grade 4 = 2%), Anaemia* (Grade 3 = 10%, Grade 4 = 3%), Leukopenia*

Common

Febrile neutropenia

Metabolism and Nutrition Disorders

Very Common

Anorexia (Grade 3‑4 < 1%)

Common

Dehydration, Decreased appetite, Hypokalaemia

Psychiatric Disorders

Common

Insomnia

Nervous System Disorders

Very Common

Headache

Common

Peripheral sensory neuropathy, Dysgeusia, Dizziness, Paraesthesia

Vascular Disorders

Common

Hypotension, Flushing

Respiratory, Thoracic and Mediastinal Disorders

Common

Dyspnoea (Grade 3‑4 = 2%), Cough

Gastrointestinal disorders

Very Common

Vomiting (Grade 3‑4 = 6.5%), Nausea (Grade 3‑4 = 6%), Constipation (Grade 3‑4 < 1%)

Common

Diarrhoea (Grade 3‑4 < 1%), Stomatitis (Grade 3‑4 < 1%), Abdominal pain, Dyspepsia, Upper abdominal pain

Hepatobiliary Disorders

Very Common

Hyperbilirubinemia* (Grade 3 = 1%), Alanine aminotransferase increased* (Grade 3 = 38%, Grade 4 = 3%), Aspartate aminotransferase increased* (Grade 3 = 44%, Grade 4 = 7%), Blood alkaline phosphatase increased*, Gamma‑glutamyltransferase increased*

Skin and Subcutaneous Tissue Disorders

Common

Alopecia

 

Musculoskeletal and Connective Tissue Disorders

Common

Myalgia, Arthralgia, Back pain

 

Metabolism and Nutrition Disorders

Very Common

Anorexia (Grade 3‑4 < 1%)

Common

Dehydration, Decreased appetite, Hypokalaemia

Infections and Infestations

Common

Infection

Vascular Disorders

Common

Hypotension, Flushing

General Disorders and Administration Site Conditions

Very Common

Fatigue (Grade 3‑4 = 9%), Asthenia (Grade 3‑4 = 1%)

Common

Pyrexia, Oedema, Oedema peripheral, Injection site reaction

InvestigationsHepatobiliary Disorders

Very Common

Blood creatine phosphokinase increased* (Grade 3‑4 = 4%), Blood creatinine increased*, Blood albumin decreased*

Common

Weight decreasedHyperbilirubinemia* (Grade 3 = 1%), Alanine aminotransferase increased* (Grade 3 = 38%, Grade 4 = 3%), Aspartate aminotransferase increased* (Grade 3 = 44%, Grade 4 = 7%), Blood alkaline phosphatase increased*, Gamma‑glutamyltransferase increased*

Psychiatric Disorders

Common

Insomnia

 

* Derived from laboratory data

 

The table below provides the frequency and severity of undesirable effects considered possibly related to study medicinal productdrug and reported in ≥ 5% of patients with ovarian cancer randomised to receive Yondelis 1.1 mg/m2/PLD 30 mg/m2 or PLD 50 mg/m2 in the pivotal trial ET743‑OVA‑301. Both adverse reactions and laboratory values have been used. Within each frequency grouping, undesirable effects are presented in order of decreasing seriousness.

 

Adverse reactions reported in ≥ 5% of patients in clinical trial ET743‑OVA‑301

System Organ Class

Frequency

Event

Yondelis+PLD
n=333

PLD

n=330

All
Grades

(%)

Grade 3

(%)

Grade 4

(%)

All
Grades

(%)

Grade 3

(%)

Grade 4

(%)

Investigations 

Common

Blood creatine phosphokinase increased*

22.0

0.9

0.9

13.7

 

 

Blood and lymphatic system disorders

 

Very common

 

Neutropenia*

91.6

29.7

42.3

73.5

19.7

9.8

Leukopenia*

94.9

44.7

17.7

81.8

16.0

4.0

Anaemia*

94.9

12.9

5.7

82.1

6.2

2.2

Thrombocytopenia*

63.7

12.3

10.8

27.4

2.5

1.8

Common

Febrile neutropenia*

6.9

4.5

2.4

2.1

1.8

0.3

Metabolism and nutrition disorders

 

Very common

Anorexia

28.8

2.1

 

20

1.5

 

Common

Hypokalaemia

6.3

2.1

 

2.1

 

 

Nervous system disorders

 

Common

 

Headache

6.6

0.3

 

2.4

 

 

Dysgeusia

5.4

0.3

 

2.7

 

 

Respiratory, thoracic and mediastinal disorders

Common

Dyspnoea

6.6

0.3

 

3.3

0.3

0.3

Gastrointestinal disorders

 

Very common

 

Nausea

70.9

8.7

 

37.6

2.4

 

Vomiting

51.7

9.9

0.3

23.9

2.1

 

Constipation

20.4

0.9

 

15.5

0.3

 

Stomatitis

19.2

0.9

 

31.2

4.8

0.3

Diarrhoea

17.1

2.1

 

10

1.2

 

Common

 

Abdominal pain

9.3

0.6

 

7

0.9

 

Dyspepsia

7.5

0.3

 

6.1

0.6

 

Hepatobiliary disorders

Very common

Hyperbilirubinaemia*

(25.2)

(0.3)

 

(12.9)

(0.3)

 

Alanine aminotransferase increased*

96.1

45.6

4.5

36.0

2.2

 

Aspartate aminotransferase increased*

89.5

12.0

1.8

42.6

1.2

0.3

Blood alkaline phosphatase increased*

61.3

1.5

 

41.8

1.2

 

Gastrointestinal disorders

 

Very common

 

Nausea

70.9

8.7

 

37.6

2.4

 

Vomiting

51.7

9.9

0.3

23.9

2.1

 

Constipation

20.4

0.9

 

15.5

0.3

 

Stomatitis

19.2

0.9

 

31.2

4.8

0.3

Diarrhoea

17.1

2.1

 

10

1.2

 

Common

 

Abdominal pain

9.3

0.6

 

7

0.9

 

Dyspepsia

7.5

0.3

 

6.1

0.6

 

Skin and subcutaneous tissue disorders

 

Very common

 

Palmar‑plantar erythrodysaesthesia syndrome

24

3.9

 

53.6

18.5

1.2

Alopecia

12

 

 

13.3

0.3

 

Common

 

Rash

8.1

 

 

16.1

0.9

 

Skin hyperpigmentation

5.4

 

 

7

 

 

General disorders and administration site conditions

 

 

 

Very common

 

Fatigue

42.3

5.7

0.3

29.7

2.4

0.3

Asthenia

15.3

1.2

 

9.1

0.3

 

Mucosal inflammation

11.4

2.1

 

18.8

5.8

 

Pyrexia

10.2

0.9

 

4.5

0.3

 

Investigations 

Common

Blood creatine phosphokinase increased*

22.0

0.9

0.9

13.7

 

 

Metabolism and nutrition disorders

 

Very common

Anorexia

28.8

2.1

 

20

1.5

 

Common

Hypokalaemia

6.3

2.1

 

2.1

 

 

General disorders and administration site conditions

 

 

 

Very common

 

Fatigue

42.3

5.7

0.3

29.7

2.4

0.3

Asthenia

15.3

1.2

 

9.1

0.3

 

Mucosal inflammation

11.4

2.1

 

18.8

5.8

 

Pyrexia

10.2

0.9

 

4.5

0.3

 

Hepatobiliary disorders

Very common

Hyperbilirubinaemia*

(25.2)

(0.3)

 

(12.9)

(0.3)

 

Alanine aminotransferase increased*

96.1

45.6

4.5

36.0

2.2

 

Aspartate aminotransferase increased*

89.5

12.0

1.8

42.6

1.2

0.3

Blood alkaline phosphatase increased*

61.3

1.5

 

41.8

1.2

 

 

* Derived from laboratory data

 

The following reactions have been reported with a frequency below 5% in the combination arm, but are included here for their clinical relevance: neutropenic infection (< 1%), neutropenic sepsis (< 1%), pancytopenia (1.8%), bone marrow failure (1.5%), granulocytopenia (1.5%), dehydration, insomnia, peripheral sensory neuropathy, syncope, left ventricular dysfunction (< 1%), pulmonary embolism (1.2%), pulmonary edema (< 1%), cough, hepatotoxicity (< 1%), gamma‑glutamyltransferase increased, bilirubin conjugated increased, musculoskeletal pain, myalgia, blood creatinine increased, oedema/peripheral oedema, catheter site reactions.

 

In the Yondelis+PLD arm, non‑white (mainly Asian) patients had a higher incidence than white patients in grade 3 or 4 adverse reactions (96% versus 87%), and serious adverse reactions (44% versus 23% all grades). The differences were mainly observed in relation with neutropenia (93% versus 66%), anaemia (37% versus 14%) and thrombocytopenia (41% versus 19%). However, the incidences of clinical complications related to haematological toxicity such as severe infections or bleeding, or those leading to death or treatment termination, were similar in both subpopulations.

 

Description of selected adverse reactions

 

Most frequent adverse reactions

 

Blood and lymphaticLymphatic system disorders

 

Neutropenia:

Neutropenia is the most common haematological toxicity. It followed a predictable pattern of rapid onset and reversibility, and was rarely associated with fever or infection. Neutrophil nadirs occurred at a median of 15 days and recovered within a week. The analysis per cycle performed in patients treated with the monotherapy regimen showed neutropenia of grade 3 and 4 in approximately 19% and 8% of cycles respectively. In this population febrile neutropenia occurred in 2% of patients and in < 1% of cycles.

 

Thrombocytopenia:

 Bleeding events associated to thrombocytopenia occurred in < 1% of patients treated with the monotherapy regimen. The analysis per cycle performed in these patients showed thrombocytopenia of grade 3 and 4 in approximately 3% and < 1% of cycles respectively.

 

Anaemia:

Anaemia occurred in 93% and 94% of patients treated with the monotherapy and combination regimens respectively. The percentages of patients anaemic at baseline were 46% and 35% respectively. The analysis per cycle performed in patients treated with the monotherapy regimen showed anaemia of grade 3 and 4 in approximately 3% and 1% of cycles respectively.

 

Hepatobiliary disorders

 

AST/ALT increases:

The median time to reach the peak values was 5 days for both AST and ALT. Most of the values had decreased to grade 1 or resolved by day 14‑15 (see section 4.4). The analysis per cycle performed in patients treated with the monotherapy regimen showed grade 3 elevations of AST and ALT in 12% and 20% of cycles respectively. Grade 4 elevations of AST and ALT occurred in 1% and 2% of cycles respectively. Most transaminase elevations improved to grade 1 or to pre‑retreatment levels within 15 days, and less than 2% of cycles had recovering times longer than 25 days. ALT and AST increases did not follow a cumulative pattern but showed a tendency towards less severe elevations over time.

 

Hyperbilirubinemia:

Bilirubin peaks approximately a week after onset and resolves approximately two weeks after onset.

 

Liver function tests predicting severe toxicity (meeting Hy´s law) and clinical manifestations of severe hepatic injury were uncommon with a lower than 1% incidence of individual signs and symptoms including jaundice, hepatomegaly or liver pain. Mortality in the presence of hepatic injury occurred in less than 1% of patients in both regimens.

 

Other adverse reactions

 

CPK elevations and rhabdomyolysis: CPK elevations of any grade were observed in 23‑26% of patients in both regimens. CPK increases in association with rhabdomyolysis were reported in less than 1% of patients.

 

Alopecia: Alopecia was reported in approximately 3% of patients treated with the monotherapy regimen, of which the majority was grade 1 alopecia.

 

Post‑marketing experience

 

During post‑marketing surveillance few cases of trabectedin extravasation with subsequent tissue necrosis requiring debridement have been reported (see section 4.4).

 

5.         PHARMACOLOGICAL PROPERTIES

 

5.1       Pharmacodynamic properties

 

Pharmacotherapeutic group: Antineoplastic agent, ATC code: L01CX01.

 

Mechanism of action

 

Trabectedin binds to the minor groove of deoxyribonucleic acid (DNA), bending the helix to the major groove. This binding to DNA triggers a cascade of events affecting several transcription factors, DNA binding proteins, and DNA repair pathways, resulting in perturbation of the cell cycle.

 

Pharmacodynamic effects

 

Trabectedin has been shown to exert antiproliferative in vitro and in vivo activity against a range of human tumour cell lines and experimental tumours, including malignancies such as sarcoma, breast, non‑small cell lung, ovarian and melanoma.

 

Electrocardiogram (ECG) investigations

 

In a placebo‑controlled QT/QTc study, trabectedin did not prolong the QTc interval in patients with advanced solid malignancies.

 

Clinical efficacy

 

The efficacy and safety of trabectedin in soft tissue sarcoma is based in a randomised trial in patients with locally advanced or metastatic lipo- or leiomyosarcoma, whose disease had progressed or relapsed after treatment with at least anthracyclines and ifosfamide. In this trial trabectedin was administered either at 1.5 mg/m2 as a 24‑hour intravenous infusion every 3 weeks or at 0.58 mg/m2 weekly as a 3‑hour intravenous infusion for 3‑weeks of a 4‑week cycle. The protocol specified final time to progression (TTP) analysis showed a 26.6% reduction in the relative risk of progression for patients treated in the 24‑h q3wk group [Hazard Ratio (HR)=0.734, Confidence Interval (CI): 0.554‑0.974]. Median TTP values were 3.7 months (CI: 2.1‑5.4 m) in the 24‑h q3wk group and 2.3 months (CI: 2.0‑3.5 m) in the 3‑h qwk group (p=0.0302). No significant differences were detected in overall survival (OS). Median OS with the 24‑h q3wk regimen was 13.9 months (CI: 12.5‑18.6) and 60.2% of patients were alive at 1 year (CI: 52.0‑68.5%).

 

Additional efficacy data are available from 3 single‑arm Phase II trials with similar populations treated with the same regimen. These trials evaluated a total of 100 patients with lipo- and leiomyosarcoma and 83 patients with other types of sarcoma.

 

The efficacy of Yondelis/PLD combination in relapsed ovarian cancer is based on ET743‑OVA‑301, a randomized phase 3 study of 672 patients who received either trabectedin (1.1 mg/m2) and PLD (30 mg/m2) every 3 weeks or PLD (50 mg/m2) every 4 weeks. The primary analysis of progression free survival (PFS) was performed in 645 patients with measurable disease and assessed by independent radiology review. Treatment with the combination arm resulted in a 21% risk reduction for disease progression compared to PLD alone (HR=0.79, CI: 0.65‑0.96, p=0.0190). Secondary analyses of PFS and response rate also favoured the combination arm. The results of the main efficacy analyses are summarised in the table below:


 

Efficacy analyses from ET743‑OVA‑301

 

Yondelis+PLD

PLD

Hazard/Odds ratio

p‑value

Progression Free Survival

Independent radiology review,

measurable disease *

n=328

n=317

 

 

Median PFS (95% CI) (months)

7.3 (5.9‑7.9)

5.8 (5.5‑7.1)

0.79 (0.65‑0.96)

0.0190 a

12 months PFS rate (95% CI) (%)

25.8 (19.7‑32.3)

18.5 (12.9‑24.9)

 

 

Independent oncology review,

all randomised

n=336

n=335

 

 

Median PFS (95% CI) (months)

7.4 (6.4‑9.2)

5.6 (4.2‑6.8)

0.72 (0.60‑0.88)

0.0008 a

Overall Survival (Final analysis - n=522 events)

All randomised

n=337

n=335

 

 

Median OS (95% CI) (months)

22.2 (19.3‑25.0)

18.9 (17.1‑21.5)

0.86 (0.72‑1.02)

0.0835 a

Overall survival in platinum‑sensitive population (Final analysis n=316 events)

 

n=218

n=212

 

 

Median OS (95% CI) (months)

27.0 (24.1‑31.4)

24.1 (20.9‑25.9)

0.83 (0.67‑1.04)

0.1056 a

Overall Response Rate (ORR)

Independent radiology review,

all randomised

n=337

n=335

 

 

ORR (95% CI) (%)

27.6 (22.9‑32.7)

18.8 (14.8‑23.4)

1.65 (1.14‑2.37)

0.0080 b

* Primary efficacy analysis

a Log rank test

b Fisher’s test

 

Based on independent oncology review, patients with platinum‑free interval (PFI) < 6 months (35% in Yondelis+PLD and 37% in PLD arm) had similar PFS in the two arms with both showing median PFS of 3.7 months (HR=0.89, CI: 0.67‑1.20). In patients with PFI ³ 6 months (65% in Yondelis+PLD and 63% in PLD arm), median PFS was 9.7 months in the Yondelis+PLD arm compared with 7.2 months in the PLD monotherapy arm (HR=0.66, CI: 0.52‑0.85).

 

In the final analysis, the effect of the Yondelis+PLD combination vs. PLD alone on overall survival was more pronounced in patients with PFI ³ 6 months (platinum‑sensitive population: 27.0 vs. 24.1 months, HR=0.83, CI: 0.67‑1.04) than in those with PFI < 6 months (platinum‑resistant population: 14.2 vs. 12.4 months, HR=0.92, CI: 0. 70‑1.21).

 

The benefit in OS with Yondelis plus PLD was not due to the effect of subsequent therapies, which were well balanced between the two treatment arms.

 

In the multivariate analyses including PFI, treatment effect on overall survival was statistically significant favouring the Yondelis+PLD combination over PLD alone (all randomised: p=0.0285; platinum‑sensitive population: p=0.0319).

 

No data are available comparing Yondelis+PLD to a platinum‑based regimen in platinum‑sensitive patients.

 

No statistically significant differences were found between treatment arms in global measures of Quality of Life.

 

This medicinal product has been authorised under “exceptional circumstances”. This means that due to the rarity of soft tissue sarcoma, it has not been possible to obtain complete information on this medicinal productYondelis in this indication.

 

The European Medicines Agency will review any new information which may become available every year and this SmPC will be updated as necessary.

 

5.2       Pharmacokinetic properties

 

Distribution

 

Systemic exposure after intravenous administration as a constant rate infusion is dose proportional at doses up to and including 1.8 mg/m2. Trabectedin pharmacokinetic profile is consistent with a multiple‑compartment disposition model.

 

Following intravenous administration, trabectedin demonstrates a high apparent volume of distribution, consistent with extensive tissue and plasma protein binding (94 to 98% of trabectedin in plasma is protein bound). The distribution volume at steady state of trabectedin in human subjects exceeds 5,000 l.

 

Biotransformation

 

Cytochrome P450 3A4 is the major cytochrome P450 isozyme responsible for the oxidative metabolism of trabectedin at clinically relevant concentrations. Other P450 enzymes may contribute to metabolism. Trabectedin does not induce or inhibit major cytochrome P450 enzymes.

 

Elimination

 

Renal elimination of unchanged trabectedin in humans is low (less than 1%). The terminal half‑life is long (population value of the terminal elimination phase: 180‑hr). After a dose of radiolabelled trabectedin administered to cancer patients, faecal mean (SD) recovery of total radioactivity is 58% (17%), and urinary mean (SD) recovery is 5.8% (1.73%). Based on the population estimate for plasma clearance of trabectedin (30.9 l/h) and blood/plasma ratio (0.89), the clearance of trabectedin in whole blood is approximately 35 l/h. This value is approximately one‑half the rate of human hepatic blood flow. Thus the trabectedin extraction ratio can be considered moderate. The inter‑patient variability of the population estimate for plasma clearance of trabectedin was 49% and intra‑patient variability was 28%.

 

A population pharmacokinetic analysis showed that when administered in combination with PLD, the plasma clearance of trabectedin was decreased by 31%; the plasma pharmacokinetics of PLD were not influenced by the concomitant administration of trabectedin.

 

Special populations

 

A population pharmacokinetic analysis indicated that the plasma clearance of trabectedin is not influenced by age (range 19‑83 years), gender, total body weight (range: 36 to 148 kg) or body surface area (range: 0.9 to 2.8 m2). An analysis made on a limited number of patients shows that race and ethnicity are not expected to have clinically significant effects on trabectedin pharmacokinetics.

 

Renal impairment

 

There is no relevant influence of renal function measured by creatinine clearance on trabectedin pharmacokinetics within the range of values ( 30.3 ml/min) present in the patients included in the clinical studies. No data are available in patients with a creatinine clearance of less than 30.3 ml/min. The low recovery (< 9% in all studied patients) of total radioactivity in the urine after a single dose of 14C‑labelled trabectedin indicates that renal impairment has little influence on the elimination of trabectedin or its metabolites.

 

Hepatic impairment

 

Although the population analysis showed no relationship between the serum liver enzymes concentrations and the plasma clearance of trabectedin, systemic exposure to trabectedin may be increased in patients with hepatic impairment; therefore close monitoring of toxicity is warranted.

 

5.3       Preclinical safety data

 

Preclinical data indicate that trabectedin has limited effect on the cardiovascular, respiratory and central nervous system at exposures below the therapeutic clinical range, in terms of AUC.

 

The effects of trabectedin on cardiovascular and respiratory function have been investigated in vivo (anesthetised Cynomolgus monkeys). A 1 hour infusion schedule was selected to attain maximum plasma levels (Cmax values) in the range of those observed in the clinic. The plasma trabectedin levels attained were 10.6 ± 5.4 (Cmax), higher than those reached in patients after infusion of 1,500 mg/m2 for 24 (Cmax of 1.8 ± 1.1 ng/ml) and similar to those reached after administration of the same dose by 3 hour infusion (Cmax of 10.8 ± 3.7 ng/ml).

 

Myelosupression and hepatoxicity were identified as the primary toxicity for trabectedin. Findings observed included haematopoietic toxicity (severe leukopenia, anaemia, and lymphoid and bone marrow depletion) as well as increases in liver function tests, hepatocellular degeneration, intestinal epithelial necrosis, and severe local reactions at the injection site. Renal toxicological findings were detected in multi‑cycle toxicity studies conducted in monkeys. These findings were secondary to severe local reaction at the administration site, and therefore uncertainly attributable to trabectedin; however, caution must be guaranteed in the interpretation of these renal findings, and treatment‑related toxicity cannot be excluded.

 

Trabectedin is genotoxic both in vitro and in vivo. Long‑term carcinogenicity studies have not been performed.

 

Fertility studies with trabectedin were not performed but limited histopathological changes were observed in the gonads in the repeat dose toxicity studies. Considering the nature of the compound (cytotoxic and mutagenic), it is likely to affect the reproductive capacity.

 

 

6.         PHARMACEUTICAL PARTICULARS

 

6.1       List of excipients

 

Sucrose.

 

Potassium dihydrogen phosphate.

 

Phosphoric acid (for pH‑adjustment).

 

Potassium hydroxide (for pH‑adjustment).

 

6.3       Shelf life

 

Unopened vials

 

: 36 months.

 

After reconstitution

 

Chemical, chemical and physical stability has been demonstrated for 30 hours up to 25ºC.

 

From a microbiological point of view, the reconstituted solution should be diluted and used immediately. If not diluted and used immediately, in‑use storage times and conditions prior to use of the reconstituted product are the responsibility of the user and would normally not be longer than 24 hours at 2ºC to 8ºC, unless reconstitution has taken place in controlled and validated aseptic conditions.

 

After dilution

 

Chemical, chemical and physical stability has been demonstrated for 30 hours up to 25ºC.

 

6.5       Nature and contents of container

 

Yondelis is supplied in a Type I colourless glass vial with a bromobutyl rubber stopper covered with an aluminium flip‑off seal containing .

 

0.25 mg vial

 

Each vial contains 0.25 mg of trabectedin.

 

1 mg vial

 

Each vial contains 1 mg of trabectedin.

 

0.25 mg and 1 mg vials

 

Each outer carton contains one vial.

 

6.6       Special precautions for disposal and other handling

 

Preparation for intravenous infusion

 

0.25 mg vial

 

Appropriate aseptic techniques must be used. Yondelis must be reconstituted and further diluted prior to infusion. When used in combination the intravenous line should be flushed well with 50 mg/ml (5%) glucose solution for infusion after administration of PLD and before administration of Yondelis. The use of any diluent other than 50 mg/ml (5%) glucose solution for infusion may cause precipitation of PLD (see. (See also PLD Summary of Product Characteristics for specific handling instructions).

 

Each vial containing 0.25 mg of trabectedin is reconstituted with 5 ml of sterile water for injections. The solution obtained has a concentration of 0.05 mg/ml and is for single‑use only.

 

Instructions for reconstitution

 

Each vial containing 0.25 mg of trabectedin is reconstituted with 5 ml of water for injections. The solution obtained has a concentration of 0.05 mg/ml and is for single‑use only.

 

A syringe is used to inject 5 ml of sterile water for injections into the vial. TheShake the vial must be shaken until complete dissolution. The reconstituted solution results in a clear, colourless or slightly yellowish solution, essentially free of visible particles.

 

1 mg vial

 

Appropriate aseptic techniques must be used. Yondelis must be reconstituted and further diluted prior to infusion. When used in combination the intravenous line should be flushed well with 50 mg/ml (5%) glucose solution for infusion after administration of PLD and before administration of Yondelis. The use of any diluent other than 50 mg/ml (5%) glucose solution for infusion may cause precipitation of PLD (see. (See also PLD Summary of Product Characteristics for specific handling instructions).

 

Each vial containing 1 mg of trabectedin is reconstituted with 20 ml of sterile water for injections. The solution obtained has a concentration of 0.05 mg/ml and is for single‑use only

 

Instructions for reconstitution

 

Each vial containing 1 mg of trabectedin is reconstituted with 20 ml of water for injections. The solution obtained has a concentration of 0.05 mg/ml and is for single‑use only.

 

A syringe is used to inject 20 ml of sterile water for injections into the vial. TheShake the vial must be shaken until complete dissolution. The reconstituted solution results in a clear, colourless or slightly yellowish solution, essentially free of visible particles.

 

0.25 mg and 1 mg vials

 

This reconstituted solution contains 0.05 mg/ml of trabectedin. It requires further dilution and is for single‑use only.

 

Instructions for dilution

 

The reconstituted solution should be diluted with sodium chloride 9 mg/ml (0.9%) solution for infusion or glucose 50 mg/ml (5%) solution for infusion. The required volume should be calculated as follows:

 

Volume (ml) = BSA (m2) x individual dose (mg/m2)

                                    0.05 mg/ml

 

BSA = Body Surface Area

 

If administration is to be made through a central venous line, the appropriate amount of reconstituted solution should be withdrawn from the vial and added to an infusion bag containing ³ 50 ml of diluent (sodium chloride 9 mg/ml (0.9%) solution for infusion or glucose 50 mg/ml (5%) solution for infusion), the concentration of trabectedin in the infusion solution being ≤ 0.030 mg/ml.

 

If central venous access is not feasible and a peripheral venous line has to be used, the reconstituted solution should be added to an infusion bag containing ³ 1,000 ml of diluent (sodium chloride 9 mg/ml (0.9%) solution for infusion or glucose 50 mg/ml (5%) solution for infusion).

 

Parenteral solutions should be inspected visually for particles prior to administration. Once the infusion is prepared, it should be administered immediately.

 

Instructions for handling and disposal

 

Yondelis is a cytotoxic anticancer medicinal product and, as with other potentially toxic compounds, caution should be exercised during handling. Procedures for proper handling and disposal of cytotoxic medicinal products must be followed. Personnel should be trained in the correct techniques to reconstitute and dilute the medicinal product and should wear protective clothing including mask, goggles and gloves during the reconstitution and dilution. Pregnant staff must be excluded from working with this medicinal product.

 

Accidental contact with the skin, eyes or mucous membranes must be treated immediately with copious amounts of water.

 

Any unused medicinal product or waste material should be disposed of in accordance with local requirements for cytotoxic medicinal products.

 

No incompatibilities have been observed between Yondelis and type I glass bottles, polyvinylchloride (PVC) and polyethylene (PE) bags and tubing, polyisoprene reservoirs and titanium implantable vascular access systems.

 

Any unused medicinal product or waste material should be disposed of in accordance with local requirements for cytotoxic medicinal products.

 

 

 

9.         DATE OF FIRST AUTHORISATION/RENEWAL OF THE AUTHORISATION

 

Date of first authorisation: 17 September 2007

Date of latest renewal: 03 August 2012

 

 

10.       DATE OF REVISION OF THE TEXT

 

08/2012

 

Detailed information on this medicinal product is available on the website of the European Medicines Agency http://www.ema.europa.eu.

 

Updated on 17 August 2012

Reasons for updating

  • Change of contraindications
  • Change to storage instructions
  • Change to side-effects
  • Change to information about pregnancy or lactation
  • Change to further information section
  • Change to improve clarity and readability

Updated on 16 February 2012

Reasons for updating

  • Change to section 2 - Qualitative and quantitative composition
  • Change to section 4.2 - Posology and method of administration
  • Change to section 4.3 - Contraindications
  • Change to section 4.5 - Interaction with other medicinal products and other forms of interaction
  • Change to section 4.6 - Pregnancy and lactation
  • Change to section 5.1 - Pharmacodynamic properties
  • Change to section 5.2 - Pharmacokinetic properties
  • Change to section 6.4 - Special precautions for storage
  • Change to section 6.6 - Special precautions for disposal and other handling
  • Change to section 9 - Date of renewal of authorisation
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

New text appears in blue
Deleted text appears in red
Existing text appears in black




2.         QUALITATIVE AND QUANTITATIVE COMPOSITION

 

Each vial contains 0.25 mg of trabectedin.

 

1 ml of reconstituted solution contains 0.05 mg of trabectedin.

 

Excipients with known effect:

 

Each vial contains 2 mg of potassium and 0.1 g of sucrose.

 

For thea full list of excipients, see section 6.1.

 

4.2        Posology and method of administration

 

Yondelis must be administered under the supervision of a physician experienced in the use of chemotherapy. Its use should be confined to qualified oncologists or other health professionals specialised in the administration of cytotoxic agents.

 

Posology

 

For the treatment of soft tissue sarcoma, the recommended dose is 1.5 mg/m2 body surface area, administered as an intravenous infusion over 24 hours with a three‑week interval between cycles.

 

For the treatment of ovarian cancer Yondelis is administered every three weeks as a 3‑hour infusion at a dose of 1.1 mg/m2, immediately after PLD 30 mg/m2. To minimize the risk of PLD infusion reactions, the initial dose is administered at a rate no greater than 1 mg/minute. If no infusion reaction is observed, subsequent PLD infusions may be administered over a 1‑hour period. (See also PLD Summary Product Characteristics for specific administration advice).

 

All patients must receive corticosteroids e.g. 20 mg of dexamethasone intravenously 30 minutes prior to PLD (in combination therapy) or Yondelis (in monotherapy); not only as anti‑emetic prophylaxis, but also because it appears to provide hepatoprotective effects. Additional anti‑emetics may be administered as needed.

 

The following criteria are required to allow treatment with Yondelis:

 

-                 Absolute neutrophil count (ANC) ³ 1,500/mm3

-                 Platelet count ³ 100,000/mm3

-                 Bilirubin £ upper limit of normal (ULN)

-                 Alkaline phosphatase £ 2.5 x ULN (consider hepatic isoenzymes 5‑nucleotidase or GGT, if the elevation could be osseous in origin).

-                 Albumin ³ 25 g/l

-                 Alanine aminotransferase (ALT) and Aspartate aminotransferase (AST) £ 2.5 x ULN

-                 Creatinine clearance ³ 30 ml/min (monotherapy), serum creatinine £ 1.5 mg/dl (£ 132.6 μmol/l) or creatinine clearance ³ 60 ml/min (combination therapy)

-                 Creatine phosphokinase (CPK) £ 2.5 x ULN

-                 Haemoglobin ³ 9 g/dl

 

The same criteria as above must be met prior to re‑treatment. Otherwise treatment must be delayed for up to 3 weeks until the criteria are met.

 

Additional monitoring of haematological parameters bilirubin, alkaline phosphatase, aminotransferases and CPK should occur weekly during the first two cycles of therapy, and at least once between treatments in subsequent cycles.

 

The same dose should be given for all cycles provided that no grade 3‑4 toxicities are seen and that the patient fulfils the re‑treatment criteria.

 

Dose adjustments during treatment

 

Prior to re‑treatment, patients must fulfil the baseline criteria defined above. If any of the following events occur at any time between cycles, the dose must be reduced one level, according to table 1 below, for subsequent cycles:

 

-                 Neutropenia < 500/mm3 lasting for more than 5 days or associated with fever or infection

-                 Thrombocytopenia < 25,000/mm3

-                 Increase of bilirubin > ULN and/or alkaline phosphatase > 2.5 x ULN

-                 Increase of aminotransferases (AST or ALT) > 2.5 x ULN (monotherapy) or > 5 x ULN (combination therapy), which has not recovered by day 21

-                 Any other grade 3 or 4 adverse reactions (such as nausea, vomiting, fatigue)

 

Once a dose has been reduced because of toxicity, dose escalation in the subsequent cycles is not recommended. If any of these toxicities reappear in subsequent cycles in a patient exhibiting clinical benefit, the dose may be further reduced (see below). Colony stimulating factors can be administered for hematologic toxicity according to local standard practice.

 

Table 1 Dose modification table for Yondelis (as single agent for STS or

in combination for ovarian cancer) and PLD

 

Soft Tissue Sarcoma

Ovarian Cancer

 

Yondelis

Yondelis

PLD

Starting dose

1.5 mg/m2

1.1 mg/m2

30 mg/m2  

First reduction

1.2 mg/m2

0.9 mg/m2

25 mg/m2  

Second reduction

1 mg/m2

0.75 mg/m2

20 mg/m2  

 

See the PLD SPC for more detailed information on PLD dose adjustments.

 

In the event that further dose reductions are necessary, treatment discontinuation should be considered.

 

Duration of treatment

 

In clinical trials, there were no pre‑defined limits to the number of cycles administered. Treatment continued whilst clinical benefit was noted. Yondelis has been administered for 6 or more cycles in 29.5% and 52% of patients treated with the monotherapy and combination dose and schedule respectively. The monotherapy and combination regimens have been used for up to 38 and 21 cycles respectively. No cumulative toxicities have been observed in patients treated with multiple cycles.

 

Paediatric populationpatients

 

The safety and efficacy of trabectedin in the paediatric patientspopulation have not yet been established.  Therefore, this medicinal product must not be used in children and adolescents until further data become available.

No data are available.

 

Elderly patients

 

No specific studies in elderly patients have been performed. Overall 20% of the 1164 patients in the integrated safety analysis of monotherapy clinical trials were over 65 years. Of the 333 patients with ovarian cancer who received trabectedin in combination with PLD, 24% were 65 years of age or older and 6% were over 75 years. No relevant differences in the safety profile were seen in this patient population. It seems that plasma clearance and distribution volume of trabectedin are not influenced by age. Therefore, dose adjustments based uniquely on age criteria are not routinely recommended.

 

Patients with hepatic impairmentimpaired hepatic function

 

No studies with the proposed regimen have been conducted in patients with liver dysfunction. Thus, data are not available to recommend a lower starting dose in patients with hepatic impairment. However, special caution is advised and dose adjustments may be necessary in these patients since systemic exposure is probably increased and the risk of hepatotoxicity might be increased. Patients with elevated bilirubin must not be treated with Yondelis (see section 4.4).

 

Patients with renal impairmentimpaired renal function

 

Studies including patients with renal insufficiency (creatinine clearance < 30 ml/min for the monotherapy, and < 60 ml/min for the combination regimen) have not been conducted and therefore Yondelis must not be used in this patient population (see section 4.4). Considering the pharmacokinetic characteristics of trabectedin (see section 5.2), no dose adjustments are warranted in patients with mild or moderate renal impairment.

 

Method of administration

 

Administration through a central venous line is strongly recommended (see section 6.6).

 

For instructions on reconstitution and dilution of the medicinal product before administration, see section 6.6.

 

 

4.3        Contraindications

 

-       Hypersensitivity to trabectedin or to any of the excipients listed in section 6.1

-       Concurrent serious or uncontrolled infection

-       Breast‑feeding (see section 4.6)

-       Combination with yellow fever vaccine (see section 4.4)

 

 

4.5        Interaction with other medicinal products and other forms of interaction

 

Effects of other substances on trabectedin

 

In vivo interaction studies have not been performed. Since trabectedin is metabolised mainly by CYP3A4, co‑administration of substances that inhibit this isoenzyme e.g. ketoconazole, fluconazole ritonavir, clarithromycin or aprepitant could decrease metabolism and increase trabectedin concentrations. If such combinations are needed, close monitoring of toxicities is required (see section 4.4). Likewise co‑administration with potent inducers of this enzyme (e.g. rifampicin, phenorbarbital, Saint John’s Wort) may decrease the systemic exposure to trabectedin.

 

Alcohol consumption must be avoided during treatment with trabectedin due to the hepatotoxicity of the medicinal product (see section 4.4).

 

Preclinical data have demonstrated that trabectedin is a substrate to P‑gp. Concomitant administration of inhibitors of P‑gp, e.g. cyclosporine and verapamil, may alter trabectedin distribution and/or elimination. The relevance of this interaction e.g. CNS toxicity has not been established. Caution should be taken in such situations.

 


4.6        Fertility, pregnancy and lactation

 

Pregnancy

 

No sufficient clinical data on exposed pregnancies are available. However, based on its known mechanism of action, trabectedin may cause serious birth defects when administered during pregnancy. Trabectedin should not be used during pregnancy unless clearly necessary. If it is used during pregnancy, the patient must be informed of the potential risk to the foetus (see section 5.3) and be monitored carefully. If trabectedin is used at the end of pregnancy, potential adverse reactions should be monitored carefully in the newborns.

 

Breast-feeding

 

It is not known whether trabectedin is excreted in human milk. The excretion of trabectedin in milk has not been studied in animals. Breast-feeding is contraindicated during treatment and 3 months thereafter (see section 4.3).

 

Fertility

 

Men in fertile age and women of childbearing potential must use effective contraception during treatment and 3 months thereafter for women and immediately inform the treating physician if a pregnancy occurs (see section 5.3) and 5 months after treatment for men (see section 4.4).

 

Trabectedin can have genotoxic effects. Advice on conservation of sperm should be sought prior to treatment because of the possibility of irreversible infertility due to therapy with Yondelis.

If pregnancy occurs during treatment the possibility of genetic counselling should be considered. Genetic counselling is also recommended for patients wishing to have children after therapy.

 

 

5.1        Pharmacodynamic properties

 

Pharmacotherapeutic group: Antineoplastic agent, ATC code: L01CX01.

 

Mechanism of action

 

Trabectedin binds to the minor groove of DNA, bending the helix to the major groove. This binding to DNA triggers a cascade of events affecting several transcription factors, DNA binding proteins, and DNA repair pathways, resulting in perturbation of the cell cycle.

 

Pharmacodynamic effects

Trabectedin has been shown to exert antiproliferative in vitro and in vivo activity against a range of human tumour cell lines and experimental tumours, including malignancies such as sarcoma, breast, non‑small cell lung, ovarian and melanoma.

ECG investigations

 

In a placebo‑controlled QT/QTc study, trabectedin did not prolong the QTc interval in patients with advanced solid malignancies.

 

Clinical efficacy

 

The efficacy and safety of trabectedin in soft tissue sarcoma is based in a randomised trial in patients with locally advanced or metastatic lipo or leiomyosarcoma, whose disease had progressed or relapsed after treatment with at least anthracyclines and ifosfamide. In this trial trabectedin was administered either at 1.5 mg/m2 as a 24‑hour intravenous infusion every 3 weeks or at 0.58 mg/m2 weekly as a 3‑hour intravenous infusion for 3‑weeks of a 4‑week cycle. The protocol specified final time to progression (TTP) analysis showed a 26.6% reduction in the relative risk of progression for patients treated in the 24‑h q3wk group [Hazard Ratio (HR)=0.734, CI: 0.554‑0.974]. Median TTP values were 3.7 months (CI: 2.1‑5.4 m) in the 24‑h q3wk group and 2.3 months (CI: 2.0‑3.5 m) in the 3‑h qwk group (p=0.0302). No significant differences were detected in overall survival (OS). Median OS with the 24‑h q3wk regimen was 13.9 months (CI: 12.5‑18.6) and 60.2% of patients were alive at 1 year (CI: 52.0‑68.5%).

 

Additional efficacy data are available from 3 single‑arm Phase II trials with similar populations treated with the same regimen. These trials evaluated a total of 100 patients with lipo and leiomyosarcoma and 83 patients with other types of sarcoma.

 

The efficacy of Yondelis/PLD combination in relapsed ovarian cancer is based on ET743‑OVA‑301, a randomized phase 3 study of 672 patients who received either trabectedin (1.1 mg/m2) and PLD (30 mg/m2) every 3 weeks or PLD (50 mg/m2) every 4 weeks. The primary analysis of progression free survival (PFS) was performed in 645 patients with measurable disease and assessed by independent radiology review. Treatment with the combination arm resulted in a 21% risk reduction for disease progression compared to PLD alone (HR=0.79, CI: 0.65‑0.96, p=0.0190). Secondary analyses of PFS and response rate also favoured the combination arm. The results of the main efficacy analyses are summarised in the table below:

 

Efficacy analyses from ET743‑OVA‑301

 

Yondelis+PLD

PLD

Hazard/Odds ratio

p‑value

Progression Free Survival

Independent radiology review,

measurable disease *

n=328

n=317

 

 

Median PFS (95% CI) (months)

7.3 (5.9‑7.9)

5.8 (5.5‑7.1)

0.79 (0.65‑0.96)

0.0190 a

12 months PFS rate (95% CI) (%)

25.8 (19.7‑32.3)

18.5 (12.9‑24.9)

 

 

Independent oncology review,

all randomised

n=336

n=335

 

 

Median PFS (95% CI) (months)

7.4 (6.4‑9.2)

5.6 (4.2‑6.8)

0.72 (0.60‑0.88)

0.0008 a

Overall Survival (FinalInterim analysis - n=522419 events, 38% censoring)

All randomised

n=337

n=335

 

 

Median OS (95% CI) (months)

22.24 (19.34‑25.01)

198.95 (17.14‑212.51)

0.865 (0.702‑1.023)

0.0835920 a

Overall survival in platinum‑sensitive population (Final analysis n=316 events)

 

n=218

n=212

 

 

Median OS (95% CI) (months)

27.0 (24.1‑31.4)

24.1 (20.9‑25.9)

0.83 (0.67‑1.04)

0.1056 a

Overall Response Rate

Independent radiology review,

all randomised

n=337

n=335

 

 

ORR (95% CI) (%)

27.6 (22.9‑32.7)

18.8 (14.8‑23.4)

1.65 (1.14‑2.37)

0.0080 b

* Primary efficacy analysis

a Log rank test

b Fisher’s test

 

 

Based on independent oncology review, patients with platinum‑free interval (PFI) < 6 months (35% in Yondelis+PLD and 37% in PLD arm) had similar PFS in the two arms with both showing median PFS of 3.7 months (HR=0.89, CI: 0.67‑1.20). In patients with PFI ³ 6 months (65% in Yondelis+PLD and 63% in PLD arm), median PFS was 9.7 months in the Yondelis+PLD arm compared with 7.2 months in the PLD monotherapy arm (HR=0.66, CI: 0.52‑0.85).

 

In the finalinterim analysis, the effect of the Yondelis+PLD combination vs. PLD alone on overall survival was more pronounced in patients with PFI ³ 6 months (platinum‑sensitive population: 27.0 vs. 24.13 months, HR=0.832 CI: 0.637‑1.046) than in those with PFI < 6 months (platinum‑resistant population: 14.2 vs. 12.4 months, HR=0.920, CI: 0.7868 1.210).

 

The benefit in OS with Yondelis plus PLD was not due to the effect of subsequent therapies, which were well balanced between the two treatment arms.

In the multivariate analyses including PFI, treatment effect overall survival was statistically significant favouring the Yondelis+PLD combination over PLD alone (all randomisedPFS: p=0.0285;157: platinum‑sensitive population: OS;p=0.0319407).

No data are available comparing Yondelis+PLD to a platinum‑based regimen in platinum‑sensitive patients.

 

No statistically significant differences were found between treatment arms in global measures of Quality of Life.

 

This medicinal product has been authorised under “Eexceptional Ccircumstances”. This means that due to the rarity of soft tissue sarcoma, it has not been possible to obtain complete information on Yondelis in this indication.

 

The European Medicines Agency will review any new information which may become available every year and this SmPC will be updated as necessary.

 

5.2        Pharmacokinetic properties

Distribution

Systemic exposure after intravenous administration as a constant rate infusion is dose proportional at doses up to and including 1.8 mg/m2. Trabectedin pharmacokinetic profile is consistent with a multiple‑compartment disposition model.

 

Following intravenous administration, trabectedin demonstrates a high apparent volume of distribution, consistent with extensive tissue and plasma protein binding (94 to 98% of trabectedin in plasma is protein bound). The distribution volume at steady state of trabectedin in human subjects exceeds 5000 l.

 

Biotransformation

 

Cytochrome P450 3A4 is the major cytochrome P450 isozyme responsible for the oxidative metabolism of trabectedin at clinically relevant concentrations. Other P450 enzymes may contribute to metabolism. Trabectedin does not induce or inhibit major cytochrome P450 enzymes.

 

Elimination

 

Renal elimination of unchanged trabectedin in humans is low (less than 1%). The terminal half‑life is long (population value of the terminal elimination phase: 180‑hr). After a dose of radiolabelled trabectedin administered to cancer patients, faecal mean (SD) recovery of total radioactivity is 58% (17%), and urinary mean (SD) recovery is 5.8% (1.73%). Based on the population estimate for plasma clearance of trabectedin (30.9 l/h) and blood/plasma ratio (0.89), the clearance of trabectedin in whole blood is approximately 35 l/h. This value is approximately one‑half the rate of human hepatic blood flow. Thus the trabectedin extraction ratio can be considered moderate. The inter‑patient variability of the population estimate for plasma clearance of trabectedin was 49% and intra‑patient variability was 28%.

 

A population pharmacokinetic analysis showed that when administered in combination with PLD, the plasma clearance of trabectedin was decreased by 31%; the plasma pharmacokinetics of PLD were not influenced by the concomitant administration of trabectedin.

 

Special populations

 

A population pharmacokinetic analysis indicated that the plasma clearance of trabectedin is not influenced by age (range 19‑83 years), gender, total body weight (range: 36 to 148 kg) or body surface area (range: 0.9 to 2.8 m2). An analysis made on a limited number of patients shows that race and ethnicity are not expected to have clinically significant effects on trabectedin pharmacokinetics.

 

Impaired renal functionRenal iImpairment

There is no relevant influence of renal function measured by creatinine clearance on trabectedin pharmacokinetics within the range of values ( 30.3 ml/min) present in the patients included in the clinical studies. No data are available in patients with a creatinine clearance of less than 30.3 ml/min. The low recovery (< 9% in all studied patients) of total radioactivity in the urine after a single dose of 14C‑labelled trabectedin indicates that renal impairment has little influence on the elimination of trabectedin or its metabolites.

Impaired hHepatic functionIimpairment

Although the population analysis showed no relationship between the serum liver enzymes concentrations and the plasma clearance of trabectedin, systemic exposure to trabectedin may be increased in patients with hepatic impairment; therefore close monitoring of toxicity is warranted.

 

6.4        Special precautions for storage

 

Store in a refrigerator (2ºC ‑ 8ºC).

 

For storage conditions of the reconstituted and dilutedafter reconstitution and dilution of the medicinal product, see section 6.3.

 

6.6        Special precautions for disposal and other handling

 

Preparation for intravenous infusion

 

Appropriate aseptic techniques must be used. Yondelis must be reconstituted and further diluted prior to infusion. When used in combination the intravenous line should be flushed well with 50 mg/ml (5%) glucose solution for infusion after administration of PLD and before administration of Yondelis. The use of any diluent other than 50 mg/ml (5%) glucose solution for infusion may cause precipitation of PLD. (See also PLD Summary Product Characteristics for specific handling instructions)

 

Each vial containing 0.25 mg of trabectedin is reconstituted with 5 ml of sterile water for injections. The solution obtained has a concentration of 0.05 mg/ml and is for single‑use only.

 

Instructions for reconstitution

 

A syringe is used to inject 5 ml of sterile water for injections into the vial. Shake the vial until complete dissolution. The reconstituted solution results in a clear, colourless or slightly yellowish solution, essentially free of visible particles.

 

This reconstituted solution contains 0.05 mg/ml of trabectedin. It requires further dilution and is for single‑use only.

 

Instructions for dilution

 

The reconstituted solution should be diluted with sodium chloride 9 mg/ml (0.9%) solution for infusion or glucose 50 mg/ml (5%) solution for infusion. The required volume should be calculated as follows:

 

Volume (ml) = BSA (m2) x individual dose (mg/m2)

                                                0.05 mg/ml

 

BSA = Body Surface Area

 

If administration is to be made through a central venous line, the appropriate amount of reconstituted solution should be withdrawn from the vial and added to an infusion bag containing ³ 50 ml of diluent (sodium chloride 9 mg/ml (0.9%) solution for infusion or glucose 50 mg/ml (5%) solution for infusion), the concentration of trabectedin in the infusion solution being ≤ 0.030 mg/ml.

 

If central venous access is not feasible and a peripheral venous line has to be used, the reconstituted solution should be added to an infusion bag containing ³ 1,000 ml of diluent (sodium chloride 9 mg/ml (0.9%) solution for infusion or glucose 50 mg/ml (5%) solution for infusion).

 

Parenteral solutions should be inspected visually for particles prior to administration. Once the infusion is prepared, it should be administered immediately.

 

Instructions for handling and disposal

 

Yondelis is a cytotoxic anticancer medicinal product and, as with other potentially toxic compounds, caution should be exercised during handling. Procedures for proper handling and disposal of cytotoxic medicinal products must be followed. Personnel should be trained in the correct techniques to reconstitute and dilute the medicinal product and should wear protective clothing including mask, goggles and gloves during the reconstitution and dilution. Pregnant staff must be excluded from working with this medicinal product.

 

Accidental contact with the skin, eyes or mucous membranes must be treated immediately with copious amounts of water.

 

Any unused medicinal product or waste material should be disposed of in accordance with local requirements for cytotoxic medicinal products.

 

No incompatibilities have been observed between Yondelis and type I glass bottles, polyvinylchloride (PVC) and polyethylene (PE) bags and tubing, polyisoprene reservoirs and titanium implantable vascular access systems.

 

9.         DATE OF FIRST AUTHORISATION/RENEWAL OF THE AUTHORISATION

Date of first authorisation: 17 September 2007

Date of latest renewal:

 


10. DATE OF REVISION OF THE TEXT

 

 

05/201131/01/2012

Updated on 08 February 2012

Reasons for updating

  • Change to further information section
  • Change to information about driving or using machinery
  • Change to date of revision
  • Change to dosage and administration
  • Change of contraindications
  • Change to storage instructions
  • Change to side-effects
  • Change to information about pregnancy or lactation

Updated on 01 June 2011

Reasons for updating

  • Change to section 4.2 - Posology and method of administration
  • Change to section 4.6 - Pregnancy and lactation
  • Change to section 5.1 - Pharmacodynamic properties
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

New text appears in blue
Deleted text appears in red
Existing text appears in black


4.2          Posology and method of administration

Yondelis must be administered under the supervision of a physician experienced in the use of chemotherapy. Its use should be confined to qualified oncologists or other health professionals specialised in the administration of cytotoxic agents.

Posology

 

Yondelis must be administered under the supervision of a physician experienced in the use of chemotherapy. Its use should be confined to qualified oncologists or other health professionals specialised in the administration of cytotoxic agents.

For the treatment of soft tissue sarcoma, the recommended dose is 1.5 mg/m2 body surface area, administered as an intravenous infusion over 24 hours with a three‑week interval between cycles.

For the treatment of ovarian cancer Yondelis is administered every three weeks as a 3‑hour infusion at a dose of 1.1 mg/m2, immediately after PLD 30 mg/m2. To minimize the risk of PLD infusion reactions, the initial dose is administered at a rate no greater than 1 mg/minute. If no infusion reaction is observed, subsequent PLD infusions may be administered over a 1‑hour period. (See also PLD Summary Product Characteristics for specific administration advice).

Administration through a central venous line is strongly recommended (see section 6.6).

All patients must receive corticosteroids e.g. 20 mg of dexamethasone intravenously 30 minutes prior to PLD (in combination therapy) or Yondelis (in monotherapy); not only as anti‑emetic prophylaxis, but also because it appears to provide hepatoprotective effects. Additional anti‑emetics may be administered as needed.

The following criteria are required to allow treatment with Yondelis:

-                 Absolute neutrophil count (ANC) ³ 1,500/mm3

-                 Platelet count ³ 100,000/mm3

-                 Bilirubin £ upper limit of normal (ULN)

-                 Alkaline phosphatase £ 2.5 x ULN (consider hepatic isoenzymes 5‑nucleotidase or GGT, if the elevation could be osseous in origin).

-                 Albumin ³ 25 g/l

-                 Alanine aminotransferase (ALT) and Aspartate aminotransferase (AST) £ 2.5 x ULN

-                 Creatinine clearance ³ 30 ml/min (monotherapy), serum creatinine £ 1.5 mg/dl (£ 132.6 μmol/l) or creatinine clearance ³ 60 ml/min (combination therapy)

-                 Creatine phosphokinase (CPK) £ 2.5 x ULN

-                 Haemoglobin ³ 9 g/dl

 

The same criteria as above must be met prior to re‑treatment. Otherwise treatment must be delayed for up to 3 weeks until the criteria are met.

 

Additional monitoring of haematological parameters bilirubin, alkaline phosphatase, aminotransferases and CPK should occur weekly during the first two cycles of therapy, and at least once between treatments in subsequent cycles.

The same dose should be given for all cycles provided that no grade 3‑4 toxicities are seen and that the patient fulfils the re‑treatment criteria.

Dose adjustments during treatment

Prior to re‑treatment, patients must fulfil the baseline criteria defined above. If any of the following events occur at any time between cycles, the dose must be reduced one level, according to table 1 below, for subsequent cycles:

-                 Neutropenia < 500/mm3 lasting for more than 5 days or associated with fever or infection

-                 Thrombocytopenia < 25,000/mm3

-                 Increase of bilirubin > ULN and/or alkaline phosphatase > 2.5 x ULN

-                 Increase of aminotransferases (AST or ALT) > 2.5 x ULN (monotherapy) or > 5 x ULN (combination therapy), which has not recovered by day 21

-                 Any other grade 3 or 4 adverse reactions (such as nausea, vomiting, fatigue)

 

Once a dose has been reduced because of toxicity, dose escalation in the subsequent cycles is not recommended. If any of these toxicities reappear in subsequent cycles in a patient exhibiting clinical benefit, the dose may be further reduced (see below). Colony stimulating factors can be administered for hematologic toxicity according to local standard practice.

 

Table 1 Dose modification table for Yondelis (as single agent for STS or

in combination for ovarian cancer) and PLD

 

Soft Tissue Sarcoma

Ovarian Cancer

 

Yondelis

Yondelis

PLD

Starting dose

1.5 mg/m2

1.1 mg/m2

30 mg/m2  

First reduction

1.2 mg/m2

0.9 mg/m2

25 mg/m2  

Second reduction

1 mg/m2

0.75 mg/m2

20 mg/m2  

 

See the PLD SPC for more detailed information on PLD dose adjustments.

In the event that further dose reductions are necessary, treatment discontinuation should be considered.

Duration of treatment

In clinical trials, there were no pre‑defined limits to the number of cycles administered. Treatment continued whilst clinical benefit was noted. Yondelis has been administered for 6 or more cycles in 29.5% and 52% of patients treated with the monotherapy and combination dose and schedule respectively. The monotherapy and combination regimens have been used for up to 38 and 21 cycles respectively. No cumulative toxicities have been observed in patients treated with multiple cycles.

Special patient populations

Paediatric patients

The safety and efficacy of trabectedin in paediatric patients have not yet been established. Therefore, this medicinal product must not be used in children and adolescents until further data become available.

Elderly patients

No specific studies in elderly patients have been performed. Overall 20% of the 1164 patients in the integrated safety analysis of monotherapy clinical trials were over 65 years. Of the 333 patients with ovarian cancer who received trabectedin in combination with PLD, 24% were 65 years of age or older and 6% were over 75 years. No relevant differences in the safety profile were seen in this patient population. It seems that plasma clearance and distribution volume of trabectedin are not influenced by age. Therefore, dose adjustments based uniquely on age criteria are not routinely recommended.

Patients with impaired hepatic function

No studies with the proposed regimen have been conducted in patients with liver dysfunction. Thus, data are not available to recommend a lower starting dose in patients with hepatic impairment. However, special caution is advised and dose adjustments may be necessary in these patients since systemic exposure is probably increased and the risk of hepatotoxicity might be increased. Patients with elevated bilirubin must not be treated with Yondelis (see section 4.4).

Patients with impaired renal function

Studies including patients with renal insufficiency (creatinine clearance < 30 ml/min for the monotherapy, and < 60 ml/min for the combination regimen) have not been conducted and therefore Yondelis must not be used in this patient population (see section 4.4). Considering the pharmacokinetic characteristics of trabectedin (see section 5.2), no dose adjustments are warranted in patients with mild or moderate renal impairment.

Method of administration

Administration through a central venous line is strongly recommended (see section 6.6).

For instructions on reconstitution and dilution of the medicinal product before administration, see section 6.6.

4.6          Fertility, pPregnancy and lactation

Pregnancy

No sufficient clinical data on exposed pregnancies are available. However, based on its known mechanism of action, trabectedin may cause serious birth defects when administered during pregnancy. Trabectedin should not be used during pregnancy unless clearly necessary. If it is used during pregnancy, the patient must be informed of the potential risk to the foetus (see section 5.3) and be monitored carefully. If trabectedin is used at the end of pregnancy, potential adverse reactions should be monitored carefully in the newborns.


Breastfeeding

It is not known whether trabectedin is excreted in human milk. The excretion of trabectedin in milk has not been studied in animals. Breastfeeding is contraindicated during treatment and 3 months thereafter (see section 4.3).

Fertility

Men in fertile age and women of childbearing potential must use effective contraception during treatment and 3 months thereafter for women and immediately inform the treating physician if a pregnancy occurs (see section 5.3) and 5 months after treatment for men (see section 4.4).

Trabectedin can have genotoxic effects. Advice on conservation of sperm should be sought prior to treatment because of the possibility of irreversible infertility due to therapy with Yondelis.  If pregnancy occurs during treatment the possibility of genetic counselling should be considered. Genetic counselling is also recommended for patients wishing to have children after therapy.

Lactation

It is not known whether trabectedin is excreted in human milk. The excretion of trabectedin in milk has not been studied in animals. Breast‑feeding is contraindicated during treatment and 3 months thereafter (see section 4.3).

5.1          Pharmacodynamic properties

Pharmacotherapeutic group: Antineoplastic agent, ATC code: L01CX01.

Mechanism of action

Trabectedin binds to the minor groove of DNA, bending the helix to the major groove. This binding to DNA triggers a cascade of events affecting several transcription factors, DNA binding proteins, and DNA repair pathways, resulting in perturbation of the cell cycle. Trabectedin has been shown to exert antiproliferative in vitro and in vivo activity against a range of human tumour cell lines and experimental tumours, including malignancies such as sarcoma, breast, non‑small cell lung, ovarian and melanoma.

Clinical efficacy

The efficacy and safety of trabectedin in soft tissue sarcoma is based in a randomised trial in patients with locally advanced or metastatic lipo or leiomyosarcoma, whose disease had progressed or relapsed after treatment with at least anthracyclines and ifosfamide. In this trial trabectedin was administered either at 1.5 mg/m2 as a 24‑hour intravenous infusion every 3 weeks or at 0.58 mg/m2 weekly as a 3‑hour intravenous infusion for 3‑weeks of a 4‑week cycle. The protocol specified final time to progression (TTP) analysis showed a 26.6% reduction in the relative risk of progression for patients treated in the 24‑h q3wk group [Hazard Ratio (HR)=0.734, CI: 0.554‑0.974]. Median TTP values were 3.7 months (CI: 2.1‑5.4 m) in the 24‑h q3wk group and 2.3 months (CI: 2.0‑3.5 m) in the 3‑h qwk group (p=0.0302). No significant differences were detected in overall survival (OS). Median OS with the 24‑h q3wk regimen was 13.9 months (CI: 12.5‑18.6) and 60.2% of patients were alive at 1 year (CI: 52.0‑68.5%).

Additional efficacy data are available from 3 single‑arm Phase II trials with similar populations treated with the same regimen. These trials evaluated a total of 100 patients with lipo and leiomyosarcoma and 83 patients with other types of sarcoma.

The efficacy of Yondelis/PLD combination in relapsed ovarian cancer is based on ET743‑OVA‑301, a randomized phase 3 study of 672 patients who received either trabectedin (1.1 mg/m2) and PLD (30 mg/m2) every 3 weeks or PLD (50 mg/m2) every 4 weeks. The primary analysis of progression free survival (PFS) was performed in 645 patients with measurable disease and assessed by independent radiology review. Treatment with the combination arm resulted in a 21% risk reduction for disease progression compared to PLD alone (HR=0.79, CI: 0.65-0.96, p=0.0190). Secondary analyses of PFS and response rate also favoured the combination arm. The results of the main efficacy analyses are summarised in the table below:

Efficacy analyses from ET743-OVA-301

 

Yondelis+PLD

PLD

Hazard/Odds ratio

p-value

Progression Free Survival

Independent radiology review,

measurable disease *

n=328

n=317

 

 

Median PFS (95% CI) (months)

7.3 (5.9-7.9)

5.8 (5.5-7.1)

0.79 (0.65-0.96)

0.0190 a

12 months PFS rate (95% CI) (%)

25.8 (19.7-32.3)

18.5 (12.9-24.9)

 

 

Independent oncology review,

all randomised

n=336

n=335

 

 

Median PFS (95% CI) (months)

7.4 (6.4-9.2)

5.6 (4.2-6.8)

0.72 (0.60-0.88)

0.0008 a

Overall Survival (Interim analysis - n=419 events, 38% censoring)

All randomised

n=337

n=335

 

 

Median OS (95% CI) (months)

22.4 (19.4-25.1)

19.5 (17.4-22.1)

0.85 (0.70-1.03)

0.0920 a

Overall Response Rate

Independent radiology review,

all randomised

n=337

n=335

 

 

ORR (95% CI) (%)

27.6 (22.9-32.7)

18.8 (14.8-23.4)

1.65 (1.14-2.37)

0.0080 b

* Primary efficacy analysis

a Log rank test

b Fisher´s test

 

Based on independent oncology review, patients with platinum-free interval (PFI) < 6 months (35% in Yondelis+PLD and 37% in PLD arm) had similar PFS in the two arms with both showing median PFS of 3.7 months (HR=0.89, CI: 0.67-1.20). In patients with PFI ³ 6 months (65% in Yondelis+PLD and 63% in PLD arm), median PFS was 9.7 months in the Yondelis+PLD arm compared with 7.2 months in the PLD monotherapy arm (HR=0.66, CI: 0.52-0.85).

 In the interim analysis, the effect of the Yondelis+PLD combination on overall survival was more pronounced in patients with PFI ³ 6 months (27.0 vs. 24.3 months, HR=0.82, CI: 0.63-1.06) than in those with PFI < 6 months (14.2 vs. 12.4 months, HR=0.90, CI: 0.68-1.20).

In the multivariate analyses including PFI, treatment effect was statistically significant favouring the Yondelis+PLD combination (PFS, p=0.0157; OS, p=0.0407).

No data are available comparing Yondelis+PLD to a platinum-based regimen in platinum-sensitive patients.

No statistically significant differences were found between treatment arms in global measures of Quality of Life.

This medicinal product has been authorised under “Exceptional Circumstances”. This means that due to the rarity of soft tissue sarcoma, it has not been possible to obtain complete information on Yondelis in this indication.

The European Medicines Agency (EMEA) will review any new information which may become available every year and this SmPC will be updated as necessary.

 


10. DATE OF REVISION OF THE TEXT

 

 

28 October 200905/2011

Updated on 01 June 2011

Reasons for updating

  • Change to further information section
  • Change to date of revision

Updated on 17 November 2009

Reasons for updating

  • Change to section 4.1 - Therapeutic indications
  • Change to section 4.2 - Posology and method of administration
  • Change to section 4.4 - Special warnings and precautions for use
  • Change to section 4.8 - Undesirable effects
  • Change to section 5.1 - Pharmacodynamic properties
  • Change to section 5.2 - Pharmacokinetic properties
  • Change to section 6.6 - Special precautions for disposal and other handling
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

New text appears in blue
Deleted text appears in red
Existing text appears in black

4.1 Therapeutic indications

 

Yondelis in combination with pegylated liposomal doxorubicin (PLD) is indicated for the treatment of patients with relapsed platinum-sensitive ovarian cancer.

4.2 Posology and method of administration

For the treatment of soft tissue sarcoma, the recommended dose is 1.5 mg/m2 body surface area, administered as an intravenous infusion over 24 hours with a three‑week interval between cycles.

 

For the treatment of ovarian cancer Yondelis is administered every three weeks as a 3hour infusion at a dose of 1.1 mg/m2, immediately after PLD 30 mg/m2. To minimize the risk of PLD infusion reactions, the initial dose is administered at a rate no greater than 1 mg/minute. If no infusion reaction is observed, subsequent PLD infusions may be administered over a 1hour period. (See also PLD Summary Product Characteristics for specific administration advice).

 

All patients must receive corticosteroids e.g. 20 mg of dexamethasone intravenously 30 minutes prior to PLD (in combination therapy) or Yondelis (in monotherapy); not only as anti‑emetic prophylaxis, but also because it appears to provide hepatoprotective effects. Additional anti‑emetics may be administered as needed.

 

The following criteria are required to allow treatment with Yondelis:

 

-                 Alkaline phosphatase ≤ 2.5 x ULN (consider hepatic isoenzymes 5‑nucleotidase or GGT, if the elevation could be osseous in origin).



- Creatinine clearance ≥ 30 ml/min (monotherapy), serum creatinine ≤ 1.5 mg/dl (≤132.6 μmol/l) or creatinine clearance 
60 ml/min (combination therapy)

- Creatine phosphokinase (CPK) ≤ 2.5
x ULN

Dose adjustments during treatment

 

Prior to re‑treatment, patients must fulfil the baseline criteria defined above. If any of the following events occur at any time between cycles, the dose must be reduced one level, according to table 1 below, to 1.2 mg/m2 for subsequent cycles:

 

-                 Increase of aminotransferases (AST or ALT) > 2.5 x ULN (monotherapy) or > 5 x ULN (combination therapy), which has not recovered by day 21

 

Once a dose has been reduced because of toxicity, dose escalation in the subsequent cycles is not recommended. If any of these toxicities reappear in subsequent cycles in a patient exhibiting clinical benefit, the dose may be further reduced (see below)to 1 mg/m2. Colony stimulating factors can be administered for hematologic toxicity according to local standard practice.

 

Table 1 Dose modification table for Yondelis (as single agent for STS or

in combination for ovarian cancer) and PLD

 

Soft Tissue Sarcoma

Ovarian Cancer

 

Yondelis

Yondelis

PLD

Starting dose

1.5 mg/m2

1.1 mg/m2

30 mg/m2  

First reduction

1.2 mg/m2

0.9 mg/m2

25 mg/m2  

Second reduction

1 mg/m2

0.75 mg/m2

20 mg/m2  

 

See the PLD SPC for more detailed information on PLD dose adjustments.

 

Duration of treatment

 

In clinical trials, there were no pre‑defined limits to the number of cycles administered. Treatment continued whilst clinical benefit was noted. Trabectedin Yondelis has been administered for 6 or more cycles in 168 out of 569 (29.5%) and 52% of patients treated with the monotherapy and combinationproposed dose and schedule respectively. Theis monotherapy and combination regimens haves been used for up to 38 and 21 cycles respectively. No cumulative toxicities have been observed in patients treated with multiple cycles.

 

Elderly patients

 

No specific studies in elderly patients have been performed. Overall 20% of the 1164 patients in the integrated safety analysis of monotherapy clinical trials were over 65 years. Of the 333 patients with ovarian cancer who received trabectedin in combination with PLD, 24% were 65 years of age or older and 6% were over 75 years. No relevant differences in the safety profile were seen in this patient population. It seems that plasma clearance and distribution volume of trabectedin are not influenced by age. Therefore, dose adjustments based uniquely on age criteria are not routinely recommended.

Patients with impaired hepatic function

 

No studies with the proposed regimen have been conducted in patients with liver dysfunction. Thus, data are not available to recommend a lower starting dose in patients with hepatic impairment. However, special caution is advised and dose adjustments may be necessary in these patients since systemic exposure is probably increased and the risk of hepatotoxicity might be increased. Patients with elevated bilirubin must not be treated with Yondelis (see section 4.4).

 

Patients with impaired renal function

 

Studies including patients with severe renal insufficiency (creatinine clearance < 30 ml/min for the monotherapy, and < 60 ml/min for the combination regimen) have not been conducted and therefore Yondelis must not be used in this patient population (see section 4.4). Considering the pharmacokinetic characteristics of trabectedin (see section 5.2), no dose adjustments are warranted in patients with mild or moderate renal impairment.

 

4.4 Special warnings and precautions for use

Renal impairment

 

Creatinine clearance must be monitored prior to and during treatment. Trabectedin Yondelis monotherapy and combination regimens must not be used in patients with creatinine clearance < 30 ml/min and < 60 ml/min respectively (see section 4.2).

 

Neutropenia and thrombocytopenia

 

Grades 3 or 4 neutropenia and thrombocytopenia associated with trabectedin Yondelis therapy have been very commonly reported. A full blood cell count including differential and platelet count must be performed at baseline, weekly for the first two cycles and then once between cycles (see section 4.2). Patients who develop fever should promptly seek medical attention. If this occurs, active supportive therapy should be started immediately.

 

Yondelis should not be administered to patients with baseline neutrophil counts of less than 1,500 cells/mm3 and platelets count of less than 100,000 cells/mm3. If severe neutropenia (ANC < 500 cells/mm3) lasting more than 5 days or associated with fever or infection occurs, dose reduction is recommended (see section 4.2).

 

Nausea and vomiting

 

Anti-emetic prophylaxis with corticosteroids such as dexamethasone must be administered to all patients (see section 4.2).

 

Rhabdomyolysis and severe CPK elevations (> 10 x ULN)

 

Trabectedin must not be used in patients with CPK > 2.5 x ULN (see section 4.2). Rhabdomyolysis has been uncommonly reported, usually in association with myelotoxicity, severe liver function test abnormalities and/or renal or multiorgan failure. Therefore, CPK should be closely monitored whenever a patient may be experiencing any of these toxicities or muscle weakness or muscle pain. If rhabdomyolysis occurs, supportive measures such as parenteral hydration, urine alkalinisation and dialysis should be promptly established, as indicated. Treatment with Yondelis should be discontinued until the patient fully recovers.


Others


 

See also PLD Summary of Product Characteristics for more detailed information on warnings and precautions.


 

4.8       Undesirable effects

Unless otherwise specified, the following safety profile of Yondelis is based on the evaluation in clinical trials of 569 patients treated up to April 2007 with the recommended treatment regimens for both indications. in several cancer types including soft tissue sarcoma, breast cancer, osteosarcoma, ovarian cancer, GIST, melanoma and renal carcinoma.

 

 


Most Approximately 91% of patients treated with Yondelis can be expected to have adverse reactions of any grade (91% in monotherapy and 99% in combination therapy). Around  40% of patients are expected to have and less than one third serious adverse reactions of grade 3 or 4 severity (10% in monotherapy and 25% in combination therapy). The most common adverse reactions of any severity grade were neutropenia, nausea, vomiting, increases in AST/ALT, anemia, fatigue, thrombocytopenia vomiting, anorexia, neutropenia, and increases in AST/ALT. diarrhoea.

 

Fatal adverse reactions have occurred in 1.9% and 0.9% of patients treated with the monotherapy and combination regimens respectively. They were often the result of a combination of events including pancytopenia, febrile neutropenia, some of them with sepsis, hepatic involvement, renal or multiorgan failure and rhabdomyolysis.

 

Adverse reactions


The table belows displays the 
adverse reactions reported in ≥ 1% of patients treated with the soft tissue sarcoma recommended regimen (1.5 mg/m2, 24 hour infusion every 3 weeks) according to the standard MedDRA system organ class. Both adverse reactionsevents and laboratory values have been used to provide frequencies. Within each frequency grouping, undesirable effects are presented in order of decreasing seriousness.


 

System Organ Class

Adverse reactions reported in ≥ 1% of patients in clinical trials at the recommended regimen [1.5 mg/m2, 24 hour infusion every 3 weeks (24‑h q3wk)]

Investigations

Very Common

Blood creatine phosphokinase increased* (Grade 34 = 4%), Blood creatinine increased*, Blood albumin decreased*

Common

Weight decreased

Blood and Lymphatic System Disorders

Very Common

Neutropenia* (Grade 3 = 26%, Grade 4 = 24%), Thrombocytopenia* (Grade 3 = 11%, Grade 4 = 2%), Anaemia* (Grade 3 = 10%, Grade 4 = 3%), Leukopenia*

Common

Febrile neutropenia

Nervous System Disorders

Very Common

Headache

Common

Peripheral sensory neuropathy, Dysgeusia, Dizziness, Paraesthesia

Respiratory, Thoracic and Mediastinal Disorders

Common

Dyspnoea (Grade 34 = 2%), Cough

Gastrointestinal disorders

Very Common

Vomiting (Grade 34 = 6.5%), Nausea (Grade 34 = 6%), Constipation (Grade 34 < 1%)

Common

Diarrhoea (Grade 34 < 1%), Stomatitis (Grade 34 < 1%), Abdominal pain, Dyspepsia, Upper abdominal pain

Skin and Subcutaneous Tissue Disorders

Common

Alopecia

 

Musculoskeletal and Connective Tissue Disorders

Common

Myalgia, Arthralgia, Back pain

 

Metabolism and Nutrition Disorders

Very Common

Anorexia (Grade 34 < 1%)

Common

Dehydration, Decreased appetite, Hypokalaemia

Infections and Infestations

Common

Infection

Vascular Disorders

Common

Hypotension, Flushing

General Disorders and Administration Site Conditions

Very Common

Fatigue (Grade 34 = 9%), Asthenia (Grade 34 = 1%)

Common

Pyrexia, Oedema, Oedema peripheral, Injection site reaction

Hepatobiliary Disorders

Very Common

Hyperbilirubinemia* (Grade 3 = 1%), Alanine aminotransferase increased* (Grade 3 = 38%, Grade 4 = 3%), Aspartate aminotransferase increased* (Grade 3 = 44%, Grade 4 = 7%), Blood alkaline phosphatase increased*, Gamma‑glutamyltransferase increased*

Psychiatric Disorders

Common

Insomnia

 

* Derived from laboratory data

 

The table below provides the frequency and severity of undesirable effects considered possibly related to study drug and reported in ≥ 5% of patients with ovarian cancer randomised to receive Yondelis 1.1 mg/m2/PLD 30 mg/m2 or PLD 50 mg/m2 in the pivotal trial ET743‑OVA-301. Both adverse reactions and laboratory values have been used. Within each frequency grouping, undesirable effects are presented in order of decreasing seriousness.

 

Adverse reactions reported in ≥ 5% of patients in clinical trial ET743-OVA-301

System Organ Class

Frequency

Event

Yondelis+PLD
n=333

PLD

n=330

All
Grades

(%)

Grade 3

(%)

Grade 4

(%)

All
Grades

(%)

Grade 3

(%)

Grade 4

(%)

Investigations 

Common

Blood creatine phosphokinase increased*

22.0

0.9

0.9

13.7

 

 

Blood and lymphatic system disorders

 

Very common

 

Neutropenia*

91.6

29.7

42.3

73.5

19.7

9.8

Leukopenia*

94.9

44.7

17.7

81.8

16.0

4.0

Anaemia*

94.9

12.9

5.7

82.1

6.2

2.2

Thrombocytopenia*

63.7

12.3

10.8

27.4

2.5

1.8

Common

Febrile neutropenia*

6.9

4.5

2.4

2.1

1.8

0.3

Nervous system disorders

 

Common

 

Headache

6.6

0.3

 

2.4

 

 

Dysgeusia

5.4

0.3

 

2.7

 

 

Respiratory, thoracic and mediastinal disorders

Common

Dyspnoea

6.6

0.3

 

3.3

0.3

0.3

Gastrointestinal disorders

 

Very common

 

Nausea

70.9

8.7

 

37.6

2.4

 

Vomiting

51.7

9.9

0.3

23.9

2.1

 

Constipation

20.4

0.9

 

15.5

0.3

 

Stomatitis

19.2

0.9

 

31.2

4.8

0.3

Diarrhoea

17.1

2.1

 

10

1.2

 

Common

 

Abdominal pain

9.3

0.6

 

7

0.9

 

Dyspepsia

7.5

0.3

 

6.1

0.6

 

Skin and subcutaneous tissue disorders

 

Very common

 

Palmar-plantar erythrodysaesthesia syndrome

24

3.9

 

53.6

18.5

1.2

Alopecia

12

 

 

13.3

0.3

 

Common

 

Rash

8.1

 

 

16.1

0.9

 

Skin hyperpigmentation

5.4

 

 

7

 

 

Metabolism and nutrition disorders

 

Very common

Anorexia

28.8

2.1

 

20

1.5

 

Common

Hypokalaemia

6.3

2.1

 

2.1

 

 

General disorders and administration site conditions

 

 

 

Very common

 

Fatigue

42.3

5.7

0.3

29.7

2.4

0.3

Asthenia

15.3

1.2

 

9.1

0.3

 

Mucosal inflammation

11.4

2.1

 

18.8

5.8

 

Pyrexia

10.2

0.9

 

4.5

0.3

 

Hepatobiliary disorders

Very common

Hyperbilirubinaemia*

(25.2)

(0.3)

 

(12.9)

(0.3)

 

Alanine aminotransferase increased*

96.1

45.6

4.5

36.0

2.2

 

Aspartate aminotransferase increased*

89.5

12.0

1.8

42.6

1.2

0.3

Blood alkaline phosphatase increased*

61.3

1.5

 

41.8

1.2

 

 

* Derived from laboratory data

 

The following reactions have been reported with a frequency below 5% in the combination arm, but are included here for their clinical relevance: neutropenic infection (< 1%), neutropenic sepsis (< 1%), pancytopenia (1.8%), bone marrow failure (1.5%), granulocytopenia (1.5%), dehydration, insomnia, peripheral sensory neuropathy, syncope, left ventricular dysfunction (< 1%), pulmonary embolism (1.2%), pulmonary edema (< 1%), cough, hepatotoxicity (< 1%), gamma-glutamyltransferase increased, bilirubin conjugated increased, musculoskeletal pain, myalgia, blood creatinine increased, oedema/peripheral oedema, catheter site reactions.

 

In the Yondelis+PLD arm, non-white (mainly Asian) patients had a higher incidence than white patients in grade 3 or 4 adverse reactions (96% versus 87%), and serious adverse reactions (44% versus 23% all grades). The differences were mainly observed in relation with neutropenia (93% versus 66%), anaemia (37% versus 14%) and thrombocytopenia (41% versus 19%). However, the incidences of clinical complications related to haematological toxicity such as severe infections or bleeding, or those leading to death or treatment termination, were similar in both subpopulations.

 


Most frequent adverse reactions

 

Blood and Lymphatic system disorders

 

Neutropenia: Neutropenia occurred in 77% of patients. Grade 3 and 4 neutropenia occurred in 26% and 24% of patients respectively). Neutropenia is the most common haematological toxicity. It followed a predictable pattern of rapid onset and reversibility, and was rarely associated with fever or infection. Neutrophil nadirs occurred at a median of 15 days and recovered within a week. The analysis per cycle performed in patients treated with the monotherapy regimen showed that neutropenia of grade 3 and 4 occurred in approximately 19% and 8% of cycles respectively. In this population fFebrile neutropenia occurred in 2% of patients and in < 1% of cycles.

 

Neutropenia followed a predictable pattern of rapid onset and reversibility, and was rarely associated with fever or infection.

 

Thrombocytopenia: Bleeding events associated to thrombocytopenia occurred in < 1% of patients treated with the monotherapy regimen. Grade 3 and 4 thrombocytopenia occurred in 11% and 2% of patients respectively. The analysis per cycle performed in these patients showed that thrombocytopenia of grade 3 and 4 occurred in approximately 3% and < 1% of cycles respectively. Bleeding events associated to thrombocytopenia occurred in < 1% of patients.


 

Anaemia: Anaemia occurred in 93% and 94% of patients treated with the monotherapy and combination regimens respectively. The percentages of patients anaemic at baseline werealthough 46% and 35% respectivelyof patients were anaemic at baseline.. Grade 3 and 4 anaemia occurred in 10% and 3% of patients respectively. The analysis per cycle performed in patients treated with the monotherapy regimen showed that anaemia of grade 3 and 4 occurred in approximately 3% and 1% of cycles respectively.


Hepatobiliary disorders

 

AST/ALT increases: The median time to reach the peak values was 5 days for both AST and ALT. Most of the values had decreased to grade 1 or resolved by day 14‑15 (see section 4.4). The analysis per cycle performed in patients treated with the monotherapy regimen showed Ggrade 3 elevations of AST and ALT occurred in 12% and 20% of cycles respectively. Grade 4 elevations of AST and ALT occurred in 1% and 2% of cycles respectively. Most transaminase elevations improved to grade 1 or to pre-retreatment levels within 15 days, and less than 2% of cycles had recovering times longer than 25 days. ALT and AST increases did not follow a cumulative pattern but showed a tendency towards less severe elevations over time.

 

Hyperbilirubinemia: Grades 1 to 2 bilirubin increases were observed in 23% of the patients. Grade 3 hyperbilirubinemia occurred in 1% of patients. Bilirubin peaks approximately a week after onset and resolves approximately two weeks after onset.


Liver function tests predicting severe toxicity (meeting Hy´s law) and cClinical manifestations of severe hepatic injury were uncommon with a lower than 1% incidence of individual signs and symptoms including jaundice, hepatomegaly or liver pain. Mortality in the presence of hepatic injury occurred in less than 1% of patients in both regimens.

 

Other adverse reactions


Nausea, vomiting, diarrhoea and constipation: Nausea and vomiting were reported in 63 and 38.5% of patients respectively. Grade 3‑4 nausea and vomiting were reported in 6% and 6.5% of patients, respectively. Grade 3‑4 diarrhoea and constipation were reported in less than 1% of patients.

 

Stomatitis: Grade 3‑4 mucositis was reported in less than 1% of the patients.

 

Fatigue/Asthenia: Grade 3‑4 fatigue/asthenia occurred in 9 and 1% of patients respectively.

 

Anorexia: Grade 3‑4 anorexia occurred in less than 1% of the patients.


CPK elevations and rhabdomyolysis: CPK elevations of any grade were observed in 23‑2626% of patients in both regimens. Grade 3 or 4 increases of CPK were observed in 4% of patients. CPK increases in association with rhabdomyolysis were reported in less than 1% of patients.

 

Dyspnoea: Grade 3‑4 dyspnoea reported as trabectedin related occurred in 2% of the patients.

 

Alopecia: Alopecia was reported in approximately 3% of all patients treated with the monotherapy regimen, of which the majority was grade 1 alopecia.

5.1     Pharmacodynamic properties

Clinical efficacy

 

The efficacy and safety of trabectedin in soft tissue sarcoma is based in a randomised trial in patients with locally advanced or metastatic liposarcoma or leiomyosarcoma, whose disease had progressed or relapsed after treatment with at least anthracyclines and ifosfamide. In this trial trabectedin was administered either at 1.5 mg/m2 as a 24‑hour intravenous infusion every 3 weeks or at 0.58 mg/m2 weekly as a 3‑hour intravenous infusion for 3‑weeks of a 4‑week cycle. The protocol specified final time to progression (TTP) analysis showed a 26.6% reduction in the relative risk of progression for patients treated in the 24‑h q3wk group [(Hazard Ratio (HR)=0.734, CI: 0.554‑0.974]). Median TTP values were 3.7 months (CI: 2.1‑5.4 m) in the 24‑h q3wk group and 2.3 months (CI: 2.0‑3.5 m) in the 3‑h qwk group (p=0.0302). No significant differences were detected in overall survival (OS). Median OS with the 24‑h q3wk regimen was 13.9 months (CI: 12.5‑18.6) and 60.2% of patients were alive at 1 year (CI: 52.0‑68.5%).


Additional efficacy data are available from 3 single‑arm Phase II trials with similar populations treated with the same regimen. These trials evaluated a total of 100 patients with lipo and leiomyosarcoma and 83 patients with other types of sarcoma.

 

The efficacy of Yondelis/PLD combination in relapsed ovarian cancer is based on ET743‑OVA‑301, a randomized phase 3 study of 672 patients who received either trabectedin (1.1 mg/m2) and PLD (30 mg/m2) every 3 weeks or PLD (50 mg/m2) every 4 weeks. The primary analysis of progression free survival (PFS) was performed in 645 patients with measurable disease and assessed by independent radiology review. Treatment with the combination arm resulted in a 21% risk reduction for disease progression compared to PLD alone (HR=0.79, CI: 0.65-0.96, p=0.0190). Secondary analyses of PFS and response rate also favoured the combination arm. The results of the main efficacy analyses are summarised in the table below:

 

Efficacy analyses from ET743-OVA-301

 

Yondelis+PLD

PLD

Hazard/Odds ratio

p-value

Progression Free Survival

Independent radiology review,

measurable disease *

n=328

n=317

 

 

Median PFS (95% CI) (months)

7.3 (5.9-7.9)

5.8 (5.5-7.1)

0.79 (0.65-0.96)

0.0190 a

12 months PFS rate (95% CI) (%)

25.8 (19.7-32.3)

18.5 (12.9-24.9)

 

 

Independent oncology review,

all randomised

n=336

n=335

 

 

Median PFS (95% CI) (months)

7.4 (6.4-9.2)

5.6 (4.2-6.8)

0.72 (0.60-0.88)

0.0008 a

Overall Survival (Interim analysis - n=419 events, 38% censoring)

All randomised

n=337

n=335

 

 

Median OS (95% CI) (months)

22.4 (19.4-25.1)

19.5 (17.4-22.1)

0.85 (0.70-1.03)

0.0920 a

Overall Response Rate

Independent radiology review,

all randomised

n=337

n=335

 

 

ORR (95% CI) (%)

27.6 (22.9-32.7)

18.8 (14.8-23.4)

1.65 (1.14-2.37)

0.0080 b

* Primary efficacy analysis

a Log rank test

b Fisher´s test


Based on independent oncology review, patients with platinum-free interval (PFI) < 6 months (35% in Yondelis+PLD and 37% in PLD arm) had similar PFS in the two arms with both showing median PFS of 3.7 months (HR=0.89, CI: 0.67-1.20). In patients with PFI  ³ 6 months (65% in Yondelis+PLD and 63% in PLD arm), median PFS was 9.7 months in the Yondelis+PLD arm compared with 7.2 months in the PLD monotherapy arm (HR=0.66, CI: 0.52-0.85).

In the interim analysis, the effect of the Yondelis+PLD combination on overall survival was more pronounced in patients with PFI 
³  6 months (27.0 vs. 24.3 months, HR=0.82, CI: 0.63-1.06) than in those with PFI < 6 months (14.2 vs. 12.4 months, HR=0.90, CI: 0.68-1.20).

No data are available comparing Yondelis+PLD to a platinum-based regimen in platinum-sensitive patients.

 

No statistically significant differences were found between treatment arms in global measures of Quality of Life.


This medicinal product has been authorised under “Exceptional Circumstances”. This means that due to the rarity of soft tissue sarcoma the disease, it has not been possible to obtain complete information on Yondelis in this medicinal product. this indication.

 

5.2       Pharmacokinetic properties


Systemic exposure after intravenous administration as a 24 hour constant rate intravenous infusion is dose proportional at doses up to and including 1.8 mg/m2. Trabectedin pharmacokinetic profile is consistent with a multiple‑compartment disposition model.

 


Renal elimination of unchanged trabectedin in humans is low (less than 1%). The terminal half‑life is long (population value of the terminal elimination phase: 180‑hr). After a dose of radiolabelled trabectedin administered to cancer patients, faecal mean (SD) recovery of total radioactivity is 58% (17%), and urinary mean (SD) recovery is 5.8% (1.73%). Based on the population estimate for plasma clearance of trabectedin (31.5 30.9 l/h) and blood/plasma ratio (0.89), the clearance of trabectedin in whole blood is approximately 35 l/h. This value is approximately one‑half the rate of human hepatic blood flow. Thus the trabectedin extraction ratio can be considered moderate. The inter‑patient variability of the population estimate for plasma clearance of trabectedin was 5149% and intra‑patient variability was 28%.

 

A population pharmacokinetic analysis showed that when administered in combination with PLD, the plasma clearance of trabectedin was decreased by 31%; the plasma pharmacokinetics of PLD were not influenced by the concomitant administration of trabectedin.

 

Special populations

 

A population pharmacokinetic analysis indicated that the plasma clearance of trabectedin is not influenced by age (range 19‑83 years), or gender, total body weight (range: 36 to 148 kg) or body surface area (range: 0.9 to 2.8 m2),. An analysis made on a limited number of patients shows that The effects of race and ethnicity are not expected to have clinically significant effects on trabectedin pharmacokinetics. have not been studied.

 

Impaired renal function

 

There is no relevant influence of renal function measured by creatinine clearance on trabectedin pharmacokinetics within the range of values (≥ 30.3 4.4 ml/min) present in the patients included in the clinical studies. No data are available in patients with a creatinine clearance of less than 30.3 4.4 ml/min. The low recovery (< 9% in all studied patients) of total radioactivity in the urine after a single dose of 14C‑labelled trabectedin indicates that renal impairment has little influence on the elimination of trabectedin or its metabolites.

 

6.6       Special precautions for disposal and other handling

 

Preparation for intravenous infusion

0.25 mg vial

 

Appropriate aseptic techniques must be used. Yondelis must be reconstituted and further diluted prior to infusion. When used in combination the intravenous line should be flushed well with 50 mg/ml (5%) glucose solution for infusion after administration of PLD and before administration of Yondelis. The use of any diluent other than 50 mg/ml (5%) glucose solution for infusion may cause precipitation of PLD. (See also PLD Summary Product Characteristics for specific handling instructions).


1 mg vial

Appropriate aseptic techniques must be used. Yondelis must be reconstituted and further diluted prior to infusion. When used in combination the intravenous line should be flushed well with 50 mg/ml (5%) glucose solution for infusion after administration of PLD and before administration of Yondelis. The use of any diluent other than 50 mg/ml (5%) glucose solution for infusion may cause precipitation of PLD. (See also PLD Summary Product Characteristics for specific handling instructions).

10. DATE OF REVISION OF THE TEXT

 

22 June28 October 2009

 

Updated on 10 November 2009

Reasons for updating

  • Change to, or new use for medicine
  • Change to warnings or special precautions for use
  • Change to side-effects
  • Change to further information section
  • Change to date of revision
  • Change to dosage and administration
  • Changes to therapeutic indications
  • Change of special precautions for disposal

Updated on 04 August 2009

Reasons for updating

  • Change to section 4.4 - Special warnings and precautions for use
  • Change to section 4.5 - Interaction with other medicinal products and other forms of interaction
  • Change to section 4.8 - Undesirable effects
  • Change to section 6.6 - Special precautions for disposal and other handling
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

Section 4.4 Special warning and precautions for use

Injection site reactions

New paragraph added: “Trabectedin extravasation may cause.....local standard practice.”

Others

3rd paragraph:  “(see sections 4.3 and 4.5).” has been replaced by “(see section 4.3)”

4.5 Interaction with other medicinal products and other forms of interaction

Effects of other substances on trabectedin

1st paragraph: “....ritonavir or clarithromycin could....” has been replaced by “....ritonavir,  clarithromycin or aprepitant  could....”

3rd paragraph: “....of Pgp, e.g....” has been replaced by “P-gp, e.g...”

4.8 Undesirable effects

New section/paragraph added to end of section:

 “Post-marketing experience

During....(see section 4.4).”

6.6 Special precautions for disposal and other handling

Paragraph under “BSA = Body Surface Area”

“....infusion), being the...” has been replaced by “infusion), the concentration of trabectedin in the infusion solution being....”

10. DATE OF REVISION OF THE TEXT

22 June 2009

Updated on 29 July 2009

Reasons for updating

  • Change to warnings or special precautions for use
  • Change to side-effects
  • Change to further information section

Updated on 30 April 2009

Reasons for updating

  • Change to section 6.6 - Special precautions for disposal and other handling
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

Section 6.6 - Instructions for dilution - If administration is to be made through a central venous line, the appropriate amount of reconstituted solution should be withdrawn from the vial and added to an infusion bag containing  50 ml of diluent (sodium chloride 9 mg/ml (0.9%) solution for infusion or glucose 50 mg/ml (5%) solution for infusion), being the concentration of trabectedin in the infusion solution 0.030 mg/ml.

(Underlined text has been added to paragraph.  Deletions:   "500" ml changed to "50 ml" and "if administration is to be made through a central venous line removed from the end of the paragraph.)
 
Section 6.6. Instructions for handling and disposal - No incompatibilities have been observed between Yondelis and type I glass bottles, polyvinylchloride (PVC) and polyethylene (PE) bags and tubing, polyisoprene reservoirs and titanium implantable vascular access systems.
 
(Underlined text has been added to paragraph)

Updated on 30 April 2009

Reasons for updating

  • Change to further information section
  • Change to date of revision
  • Change to dosage and administration

Updated on 09 April 2008

Reasons for updating

  • Change to section 6.3 - Shelf life
  • Change to section 10 - Date of revision of the text

Legal category:Product subject to medical prescription which may not be renewed (A)

Free text change information supplied by the pharmaceutical company

Section 6.3 have been changed following the approval for an increase in the shelf life to 36 months
Section 10. 03/2008

Updated on 03 March 2008

Reasons for updating

  • New SPC for new product

Legal category:Product subject to medical prescription which may not be renewed (A)

Updated on 19 February 2008

Reasons for updating

  • New PIL for new product