search
Back to results

BIBW 2992 (Afatinib) With or Without Daily Temozolomide in the Treatment of Patients With Recurrent Malignant Glioma

Primary Purpose

Glioma

Status
Completed
Phase
Phase 2
Locations
International
Study Type
Interventional
Intervention
BIBW 2992
TMZ
BIBW 2992 plus TMZ
Sponsored by
Boehringer Ingelheim
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Glioma

Eligibility Criteria

18 Years - undefined (Adult, Older Adult)All SexesDoes not accept healthy volunteers

Inclusion criteria:

Phase I Part:

  1. Histologically-confirmed WHO Grade III or IV malignant glioma that is recurrent after prior chemoradiotherapy. Patients with prior low-grade glioma are eligible if histologic assessment demonstrates transformation to WHO Grade III or IV malignant glioma.
  2. Age at least 18 years at entry
  3. KPS at least 60%
  4. Patients must have recovered from previous surgery and chemotherapy.
  5. Written informed consent that is consistent with local law and ICH-GCP guidelines.

Phase II Part:

  1. Histologically-confirmed WHO Grade IV malignant glioma at first episode of recurrence after prior combined chemoradiotherapy. Patients with prior low-grade glioma are eligible if histologic assessment demonstrates transformation to WHO Grade IV malignant glioma and if prior treatment included temozolomide chemotherapy and radiotherapy.
  2. Bi-dimensionally measurable disease with a minimum measurement of 1 cm (10 mm) in one diameter on Gd MRI performed within 14 days prior to first treatment (Day 1).
  3. Age at least 18 years at entry
  4. KPS at least 70%
  5. Patients must have recovered from previous surgery and chemotherapy.
  6. Written informed consent that is consistent with local law and ICH-GCP guidelines.
  7. Patients receiving corticosteroids have to receive a stable or decreasing dose for at least 14 days before start of study treatment.

Exclusion criteria:

Phase I and Phase II Parts:

  1. Less than 12 weeks between radiotherapy and start of study treatment, unless new enhancing lesion outside of radiation field or radiologically progressive on two consecutive MRI scans at least four weeks apart or biopsy-proven recurrence.
  2. Less than two weeks from surgical resection (one week from prior stereotactic biopsy) or major surgical procedure.
  3. Less than two weeks after previous chemotherapy (6 weeks from nitrosureas).
  4. Treatment with other investigational drugs; participation in another clinical study within the past 2 weeks before start of therapy or concomitantly with this study.
  5. Progressive disease or toxicity =CTCAEv3 Grade 3 to protracted temozolomide dosing (defined as temozolomide administered more than 5 days/28 day cycle).
  6. Active infectious disease requiring intravenous therapy.
  7. Known human immunodeficiency virus (HIV) infection or chronic Hepatitis B or C.
  8. Gastrointestinal disorders that may interfere with the absorption of the study drug or chronic diarrhea.
  9. Serious illness or concomitant non-oncological disease considered by the investigator to be incompatible with the protocol.
  10. Patient is <3 years free of another primary malignancy except: if the other primary malignancy is either not currently clinically significant or does not require active intervention (such as a basal cell skin cancer or a cervical carcinoma in situ). Existence of any other malignant disease is not allowed.
  11. Cardiac left ventricular function with resting ejection fraction <50%.
  12. Absolute neutrophil count (ANC) less than 1500/mm3.
  13. Platelet count less than 100,000/mm3.
  14. Bilirubin greater than 1.5 x upper limit of institutional norm.
  15. Aspartate amino transferase (AST) greater than 3 x upper limit of institutional norm.
  16. Serum creatinine greater than 1.5 x upper limit of institutional norm.
  17. Patients who are sexually active and unwilling to use a medically acceptable method of contraception.
  18. Pregnancy or breast-feeding.
  19. Patients unable to comply with the protocol.
  20. Known pre-existing interstitial lung disease (ILD).

Phase I part only:

1. Less than four weeks from prior treatment with bevacizumab.

Phase II Part only:

  1. Prior EGFR-directed therapy.
  2. Prior bevacizumab therapy.
  3. Patients presenting with second or higher number of episodes of recurrence.
  4. Requirement of treatment with any of the prohibited concomitant medications listed in Section 4.2.2 (Restrictions regarding concomitant treatment).

Sites / Locations

  • 1200.36.0016 Boehringer Ingelheim Investigational Site
  • 1200.36.0012 Boehringer Ingelheim Investigational Site
  • 1200.36.0005 Boehringer Ingelheim Investigational Site
  • 1200.36.0014 Boehringer Ingelheim Investigational Site
  • 1200.36.0019 Boehringer Ingelheim Investigational Site
  • 1200.36.0023 Boehringer Ingelheim Investigational Site
  • 1200.36.0008 Boehringer Ingelheim Investigational Site
  • 1200.36.0002 Boehringer Ingelheim Investigational Site
  • 1200.36.0003 Boehringer Ingelheim Investigational Site
  • 1200.36.0009 Boehringer Ingelheim Investigational Site
  • 1200.36.0001 Boehringer Ingelheim Investigational Site
  • 1200.36.0007 Boehringer Ingelheim Investigational Site
  • 1200.36.0020 Boehringer Ingelheim Investigational Site
  • 1200.36.0017 Boehringer Ingelheim Investigational Site
  • 1200.36.0010 Boehringer Ingelheim Investigational Site
  • 1200.36.0011 Boehringer Ingelheim Investigational Site
  • 1200.36.0022 Boehringer Ingelheim Investigational Site
  • 1200.36.1005 Boehringer Ingelheim Investigational Site
  • 1200.36.1010 Boehringer Ingelheim Investigational Site
  • 1200.36.1009 Boehringer Ingelheim Investigational Site
  • 1200.36.1011 Boehringer Ingelheim Investigational Site
  • 1200.36.1008 Boehringer Ingelheim Investigational Site
  • 1200.36.1001 Boehringer Ingelheim Investigational Site
  • 1200.36.1003 Boehringer Ingelheim Investigational Site
  • 1200.36.1004 Boehringer Ingelheim Investigational Site
  • 1200.36.1007 Boehringer Ingelheim Investigational Site
  • 1200.36.1002 Boehringer Ingelheim Investigational Site
  • 1200.36.1006 Boehringer Ingelheim Investigational Site

Arms of the Study

Arm 1

Arm 2

Arm 3

Arm Type

Experimental

Active Comparator

Experimental

Arm Label

BIBW 2992

TMZ

BIBW 2992 plus TMZ

Arm Description

BIBW 2992 once daily

TMZ 21/28 days

BIBW 2992 once daily plus TMZ 21/28 days

Outcomes

Primary Outcome Measures

Number of Participants With DLT- Phase I
Number of Participants With Dose Limiting Toxicities (DLT) - Phase I Part
Progression-free Survival (PFS-6) at Six Months - Phase II
PFS-6 is defined as probability of patients surviving to six months after randomization without progression. Disease progression was evaluated by an independent review committee and by the investigators, independently. The evaluation by the independent review committee was used for the primary outcome measure. The measurement "Number" the estimated PFS-6 value from the Kaplan-Meier curve of PFS.

Secondary Outcome Measures

Objective Tumor Response in Phase I
Objective Tumor Response is defined as complete response (CR) and partial response (PR) according to the MacDonald criteria assessed by central independent review.
Objective Tumor Response in Phase II
Objective Tumor Response is defined as complete response (CR) and partial response (PR) according to the MacDonald criteria assessed by central independent review. Only data collected until cut-off date July 15, 2016 were considered.
Progression-free Survival (PFS)- Phase II Part
Progression-free survival was defined as the duration between randomization and the date of the first of the two following events: progression or death.
AUCτ,ss for Afatinib
Area under the plasma concentration-time curve of afatinib after multiple administration (AUCτ,ss) of 50 mg afatinib in presence and absence of 75 mg/m² temozolomide (TMZ).
Cmax,ss for Afatinib
maximum measured plasma concentration of afatinib after multiple administration of 50 mg afatinib in presence and absence of 75 mg/m² temozolomide.
Tmax,ss for Afatinib
time from dosing to the maximum plasma concentration of afatinib after multiple administration of 50 mg afatinib in presence and absence of 75 mg/m² temozolomide
AUC (0-8) for Temozolomide
Area under the plasma concentration-time curve over the time interval from zero to 08h (AUC (0-8)) of temozolomide in presence and absence of afatinib.
Cmax for Temozolomide
maximum measured plasma concentration following the first dose of uniform intervals τ (Cmax) of temozolomide in presence and absence of afatinib.
Tmax for Temozolomide
time from dosing to the maximum plasma concentration following the first dose of uniform intervals τ (tmax) of temozolomide in presence and absence of afatinib.
t1/2 for Temozolomide
terminal half-life (t1/2) of temozolomide in presence and absence of afatinib
Phase II - Trough Plasma Concentration of Afatinib
Trough plasma concentration of afatinib after multiple administration of 40 mg afatinib administered as monotherapy or in combination with 75 mg/m² temozolomide
Number of Participants With EGFRvIII Assessed by IHC Test.
Number of participants with the epidermal growth factor receptor variant III (EGFRvIII) assessed by IHC test for the evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for EGFRvIII by immunohistochemistry (IHC) test.
Number of Participants With MGMT Marker Assessed by IHC Test.
Number of participants with MGMT marker assessed by IHC test for evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for O6-methylguanine-DNA methyltransferase (MGMT) by immunohistochemistry (IHC) test.
Number of Participants With EGFR Marker Assessed by IHC Test.
Number of participants with EGFR marker assessed by IHC test for evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for Epidermal Growth Factor Receptor (EGFR) by immunohistochemistry (IHC) test
Number of Participants With PTEN Marker Assessed by IHC Test.
Number of participants with PTEN marker assessed by IHC test for evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for Phosphatase and Tensin Homologue - a tumor suppressor gene/protein (PTEN) by immunohistochemistry (IHC) test.
Number of Participants With PAKT Marker Assessed by IHC Test.
Number of participants with PAKT marker assessed by IHC test for evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for Serinethreonine kinase (PAKT) by immunohistochemistry (IHC) test.
Number of Participants With EGFR Assessed by FISH
Number of participants with EGFR assessed by FISH for evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for EGFR by fluorescent in situ hybridization (FISH).
Number of Participants With PTEN Assessed by FISH
Number of participants with PTEN assessed by FISH for evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for PTEN by fluorescent in situ hybridization (FISH).
Number of Participants With Chromosomes (CEP7) Assessed by FISH
Number of participants with Chromosomes (CEP7) assessed by FISH for evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for Chromosomes (CEP7) by fluorescent in situ hybridization (FISH).
Number of Participants With Chromosomes (CEP10) Assessed by FISH
Number of participants with Chromosomes (CEP10) assessed by FISH for evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for Chromosomes (CEP10) by fluorescent in situ hybridization (FISH).
Number of Participants With Investigator Defined Drug-Related AEs, AEs Leading to Discontinuation of Trial Drug, All Serious Adverse Events (AE) and Other Significant AEs - Phase I
Safety was assessed based on number of participants with investigator defined drug-related AEs, AEs leading to discontinuation of trial drug, All Serious Adverse events (AE) and other significant AEs (according to International Conference on Harmonisation (ICH) E3).
Number of Participants With Adverse Events (AEs) Based on Intensity and Incidence of AE's - Phase I
Safety of afatinib as indicated by number of participants with adverse events based on intensity and incidence of AE's, especially skin reactions (rash, acne), gastrointestinal (GI) (Vomiting, nausea, diarrhea) and neurological
Number of Participants With Adverse Events, Graded According CTCAE - Phase I
Safety of Afatinib assesed based on Number of participants with adverse events, graded according to United States National Cancer Institute Common terminology Criteria for Adverse Events (US NCI CTCAE) Version 3.0. The CTCAE grades are: 1 (mild AE), 2 (moderate AE), 3 (severe AE), 4 (life-threatening or disabling AE), 5 (death related to AE).
Causes of Death - Phase I
Cause of the death reported during on treatment was due to disease progression.
Number of Participants With Investigator Defined Drug-Related AEs, AE Leading to Dose Reduction, AEs Leading to Discontinuation of Trial Drug and All SAE- Phase II
Safety was assessed based on number of participants with investigator defined drug-related AEs, AE leading to dose reduction, Adverse events (AEs) leading to discontinuation of trial drug and All Serious Adverse events (SAE).
Number of Participants With Adverse Events (AEs) Based on Intensity and Incidence of AE's - Phase II
Safety of afatinib as indicated by number of participants with adverse events based on intensity and incidence of AE's, especially skin reactions (rash, acne), gastrointestinal (GI) (Vomiting, nausea, diarrhea) and neurological.
Number of Participants With Adverse Events, Graded According CTCAE - Phase II
Safety of Afatinib assessed based on the number of participants with adverse events, graded according to United States National Cancer Institute Common terminology Criteria for Adverse Events (US NCI CTCAE) Version 3.0. The CTCAE grades are: 1 (mild AE), 2 (moderate AE), 3 (severe AE), 4 (life-threatening or disabling AE), 5 (death related to AE).
Causes of Death - Phase II
Causes of death during on treatment.
Number of Participants With Clinically Relevant Abnormalities for Decreased Cardiac Left Ventricular Function - Phase II
Number of participants with Clinically Relevant Abnormalities for decreased Cardiac left ventricular function.

Full Information

First Posted
July 31, 2008
Last Updated
July 10, 2017
Sponsor
Boehringer Ingelheim
search

1. Study Identification

Unique Protocol Identification Number
NCT00727506
Brief Title
BIBW 2992 (Afatinib) With or Without Daily Temozolomide in the Treatment of Patients With Recurrent Malignant Glioma
Official Title
Phase I/II Trial of BIBW 2992 (Afatinib) in Treating Patients With Recurrent Glioblastoma Multiforme
Study Type
Interventional

2. Study Status

Record Verification Date
July 2017
Overall Recruitment Status
Completed
Study Start Date
July 14, 2008 (Actual)
Primary Completion Date
May 12, 2011 (Actual)
Study Completion Date
May 25, 2016 (Actual)

3. Sponsor/Collaborators

Responsible Party, by Official Title
Sponsor
Name of the Sponsor
Boehringer Ingelheim

4. Oversight

5. Study Description

Brief Summary
Phase I Part: To determine the maximum tolerated dose (MTD) and pharmacokinetics of BIBW 2992 administered in combination with TMZ in patients with recurrent malignant gliomas (WHO Grade III and IV). Phase II Part: To estimate the efficacy and safety of BIBW 2992 monotherapy and BIBW 2992 / TMZ combination therapy compared to TMZ monotherapy (three treatment arms) in patients with recurrent GBM. To evaluate molecular determinants of response to BIBW 2992.

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
Glioma

7. Study Design

Primary Purpose
Treatment
Study Phase
Phase 2
Interventional Study Model
Parallel Assignment
Masking
None (Open Label)
Allocation
Randomized
Enrollment
151 (Actual)

8. Arms, Groups, and Interventions

Arm Title
BIBW 2992
Arm Type
Experimental
Arm Description
BIBW 2992 once daily
Arm Title
TMZ
Arm Type
Active Comparator
Arm Description
TMZ 21/28 days
Arm Title
BIBW 2992 plus TMZ
Arm Type
Experimental
Arm Description
BIBW 2992 once daily plus TMZ 21/28 days
Intervention Type
Drug
Intervention Name(s)
BIBW 2992
Intervention Description
BIBW 2992 once daily
Intervention Type
Drug
Intervention Name(s)
TMZ
Intervention Description
TMZ 21/28
Intervention Type
Drug
Intervention Name(s)
BIBW 2992 plus TMZ
Intervention Description
BIBW 2992 once daily plus TMZ 21/28 days
Primary Outcome Measure Information:
Title
Number of Participants With DLT- Phase I
Description
Number of Participants With Dose Limiting Toxicities (DLT) - Phase I Part
Time Frame
From randomization till data cut-off (10 Jun 2009), with a mean treatment duration of 51 days
Title
Progression-free Survival (PFS-6) at Six Months - Phase II
Description
PFS-6 is defined as probability of patients surviving to six months after randomization without progression. Disease progression was evaluated by an independent review committee and by the investigators, independently. The evaluation by the independent review committee was used for the primary outcome measure. The measurement "Number" the estimated PFS-6 value from the Kaplan-Meier curve of PFS.
Time Frame
At six months after randomization
Secondary Outcome Measure Information:
Title
Objective Tumor Response in Phase I
Description
Objective Tumor Response is defined as complete response (CR) and partial response (PR) according to the MacDonald criteria assessed by central independent review.
Time Frame
From treatment start until the date of first documented progression or data cutoff at May 12, 2011, whichever came first, with a mean treatment duration of 69.7 days.
Title
Objective Tumor Response in Phase II
Description
Objective Tumor Response is defined as complete response (CR) and partial response (PR) according to the MacDonald criteria assessed by central independent review. Only data collected until cut-off date July 15, 2016 were considered.
Time Frame
From randomization to until the date of first documented progression or data cutoff on July 15, 2016, whichever came first, with a mean treatment duration of 110.0 days
Title
Progression-free Survival (PFS)- Phase II Part
Description
Progression-free survival was defined as the duration between randomization and the date of the first of the two following events: progression or death.
Time Frame
from date of randomization until the date of first documented progression or death by any cause, whichever came first, assessed up to 9 Months.
Title
AUCτ,ss for Afatinib
Description
Area under the plasma concentration-time curve of afatinib after multiple administration (AUCτ,ss) of 50 mg afatinib in presence and absence of 75 mg/m² temozolomide (TMZ).
Time Frame
Before (-0.05 h) the drug administration and 0.5, 1, 1.5, 2, 3, 4, 6, 8 h and 24 h after drug administration on Day 15 (in presence of temozolomide) and Day 28 (in absence of temozolomide) of treatment Cycle 1
Title
Cmax,ss for Afatinib
Description
maximum measured plasma concentration of afatinib after multiple administration of 50 mg afatinib in presence and absence of 75 mg/m² temozolomide.
Time Frame
Before (-0.05 h) the drug administration and 0.5, 1, 1.5, 2, 3, 4, 6, 8 h and 24h after drug administration on Day 15 (in presence of temozolomide) and Day 28 (in absence of temozolomide) of treatment Cycle 1
Title
Tmax,ss for Afatinib
Description
time from dosing to the maximum plasma concentration of afatinib after multiple administration of 50 mg afatinib in presence and absence of 75 mg/m² temozolomide
Time Frame
Before (-0.05 h) the drug administration and 0.5, 1, 1.5, 2, 3, 4, 6, 8 h and 24h after drug administration on Day 15 (in presence of temozolomide) and Day 28 (in absence of temozolomide) of treatment Cycle 1
Title
AUC (0-8) for Temozolomide
Description
Area under the plasma concentration-time curve over the time interval from zero to 08h (AUC (0-8)) of temozolomide in presence and absence of afatinib.
Time Frame
Before (-0.05 h) the first drug administration and 0.5, 1, 1.5, 2, 3, 4, 6, 8 h after drug administration on Day 1 (in absence of afatinib) and Day 15 (in presence of afatinib) of treatment Cycle 1
Title
Cmax for Temozolomide
Description
maximum measured plasma concentration following the first dose of uniform intervals τ (Cmax) of temozolomide in presence and absence of afatinib.
Time Frame
Before (-0.05 h) the first drug administration and 0.5, 1, 1.5, 2, 3, 4, 6, 8 h after drug administration on Day 1 (in absence of afatinib) and Day 15 (in presence of afatinib) of treatment Cycle 1
Title
Tmax for Temozolomide
Description
time from dosing to the maximum plasma concentration following the first dose of uniform intervals τ (tmax) of temozolomide in presence and absence of afatinib.
Time Frame
Before (-0.05 h) the first drug administration and 0.5, 1, 1.5, 2, 3, 4, 6, 8 h after drug administration on Day 1 (in absence of afatinib) and Day 15 (in presence of afatinib) of treatment Cycle 1
Title
t1/2 for Temozolomide
Description
terminal half-life (t1/2) of temozolomide in presence and absence of afatinib
Time Frame
Before (-0.05 h) the first drug administration and 0.5, 1, 1.5, 2, 3, 4, 6, 8 h after drug administration on Day 1 (in absence of afatinib) and Day 15 (in presence of afatinib) of treatment Cycle 1
Title
Phase II - Trough Plasma Concentration of Afatinib
Description
Trough plasma concentration of afatinib after multiple administration of 40 mg afatinib administered as monotherapy or in combination with 75 mg/m² temozolomide
Time Frame
Before (-0.05 h) the drug administration of afatinib on Day 15 of Cycle 2 & 3
Title
Number of Participants With EGFRvIII Assessed by IHC Test.
Description
Number of participants with the epidermal growth factor receptor variant III (EGFRvIII) assessed by IHC test for the evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for EGFRvIII by immunohistochemistry (IHC) test.
Time Frame
Baseline (during screening)
Title
Number of Participants With MGMT Marker Assessed by IHC Test.
Description
Number of participants with MGMT marker assessed by IHC test for evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for O6-methylguanine-DNA methyltransferase (MGMT) by immunohistochemistry (IHC) test.
Time Frame
Baseline (during screening)
Title
Number of Participants With EGFR Marker Assessed by IHC Test.
Description
Number of participants with EGFR marker assessed by IHC test for evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for Epidermal Growth Factor Receptor (EGFR) by immunohistochemistry (IHC) test
Time Frame
Baseline (during screening)
Title
Number of Participants With PTEN Marker Assessed by IHC Test.
Description
Number of participants with PTEN marker assessed by IHC test for evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for Phosphatase and Tensin Homologue - a tumor suppressor gene/protein (PTEN) by immunohistochemistry (IHC) test.
Time Frame
Baseline (during screening)
Title
Number of Participants With PAKT Marker Assessed by IHC Test.
Description
Number of participants with PAKT marker assessed by IHC test for evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for Serinethreonine kinase (PAKT) by immunohistochemistry (IHC) test.
Time Frame
Baseline (during screening)
Title
Number of Participants With EGFR Assessed by FISH
Description
Number of participants with EGFR assessed by FISH for evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for EGFR by fluorescent in situ hybridization (FISH).
Time Frame
Baseline (during screening)
Title
Number of Participants With PTEN Assessed by FISH
Description
Number of participants with PTEN assessed by FISH for evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for PTEN by fluorescent in situ hybridization (FISH).
Time Frame
Baseline (during screening)
Title
Number of Participants With Chromosomes (CEP7) Assessed by FISH
Description
Number of participants with Chromosomes (CEP7) assessed by FISH for evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for Chromosomes (CEP7) by fluorescent in situ hybridization (FISH).
Time Frame
Baseline (during screening)
Title
Number of Participants With Chromosomes (CEP10) Assessed by FISH
Description
Number of participants with Chromosomes (CEP10) assessed by FISH for evaluation of molecular determinants of response to afatinib. Archival tumor samples from enrolled patients were collected and analyzed for Chromosomes (CEP10) by fluorescent in situ hybridization (FISH).
Time Frame
Baseline (during screening)
Title
Number of Participants With Investigator Defined Drug-Related AEs, AEs Leading to Discontinuation of Trial Drug, All Serious Adverse Events (AE) and Other Significant AEs - Phase I
Description
Safety was assessed based on number of participants with investigator defined drug-related AEs, AEs leading to discontinuation of trial drug, All Serious Adverse events (AE) and other significant AEs (according to International Conference on Harmonisation (ICH) E3).
Time Frame
From first administration of treatment until 28 days after last drug administration, up to 491 days.
Title
Number of Participants With Adverse Events (AEs) Based on Intensity and Incidence of AE's - Phase I
Description
Safety of afatinib as indicated by number of participants with adverse events based on intensity and incidence of AE's, especially skin reactions (rash, acne), gastrointestinal (GI) (Vomiting, nausea, diarrhea) and neurological
Time Frame
From first administration of treatment until 28 days after last drug administration, up to 491 days.
Title
Number of Participants With Adverse Events, Graded According CTCAE - Phase I
Description
Safety of Afatinib assesed based on Number of participants with adverse events, graded according to United States National Cancer Institute Common terminology Criteria for Adverse Events (US NCI CTCAE) Version 3.0. The CTCAE grades are: 1 (mild AE), 2 (moderate AE), 3 (severe AE), 4 (life-threatening or disabling AE), 5 (death related to AE).
Time Frame
From first administration of treatment until 28 days after last drug administration, up to 491 days.
Title
Causes of Death - Phase I
Description
Cause of the death reported during on treatment was due to disease progression.
Time Frame
From first administration of treatment until 28 days after last drug administration, up to 491 days.
Title
Number of Participants With Investigator Defined Drug-Related AEs, AE Leading to Dose Reduction, AEs Leading to Discontinuation of Trial Drug and All SAE- Phase II
Description
Safety was assessed based on number of participants with investigator defined drug-related AEs, AE leading to dose reduction, Adverse events (AEs) leading to discontinuation of trial drug and All Serious Adverse events (SAE).
Time Frame
From first administration of treatment until 28 days after last drug administration, up to 518 days.
Title
Number of Participants With Adverse Events (AEs) Based on Intensity and Incidence of AE's - Phase II
Description
Safety of afatinib as indicated by number of participants with adverse events based on intensity and incidence of AE's, especially skin reactions (rash, acne), gastrointestinal (GI) (Vomiting, nausea, diarrhea) and neurological.
Time Frame
From first administration of treatment until 28 days after last drug administration, up to 518 days.
Title
Number of Participants With Adverse Events, Graded According CTCAE - Phase II
Description
Safety of Afatinib assessed based on the number of participants with adverse events, graded according to United States National Cancer Institute Common terminology Criteria for Adverse Events (US NCI CTCAE) Version 3.0. The CTCAE grades are: 1 (mild AE), 2 (moderate AE), 3 (severe AE), 4 (life-threatening or disabling AE), 5 (death related to AE).
Time Frame
From first administration of treatment until 28 days after last drug administration, up to 518 days.
Title
Causes of Death - Phase II
Description
Causes of death during on treatment.
Time Frame
From first administration of treatment until 28 days after last drug administration, up to 518 days.
Title
Number of Participants With Clinically Relevant Abnormalities for Decreased Cardiac Left Ventricular Function - Phase II
Description
Number of participants with Clinically Relevant Abnormalities for decreased Cardiac left ventricular function.
Time Frame
From first administration of treatment until 28 days after last drug administration, up to 518 days.

10. Eligibility

Sex
All
Minimum Age & Unit of Time
18 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
Inclusion criteria: Phase I Part: Histologically-confirmed WHO Grade III or IV malignant glioma that is recurrent after prior chemoradiotherapy. Patients with prior low-grade glioma are eligible if histologic assessment demonstrates transformation to WHO Grade III or IV malignant glioma. Age at least 18 years at entry KPS at least 60% Patients must have recovered from previous surgery and chemotherapy. Written informed consent that is consistent with local law and ICH-GCP guidelines. Phase II Part: Histologically-confirmed WHO Grade IV malignant glioma at first episode of recurrence after prior combined chemoradiotherapy. Patients with prior low-grade glioma are eligible if histologic assessment demonstrates transformation to WHO Grade IV malignant glioma and if prior treatment included temozolomide chemotherapy and radiotherapy. Bi-dimensionally measurable disease with a minimum measurement of 1 cm (10 mm) in one diameter on Gd MRI performed within 14 days prior to first treatment (Day 1). Age at least 18 years at entry KPS at least 70% Patients must have recovered from previous surgery and chemotherapy. Written informed consent that is consistent with local law and ICH-GCP guidelines. Patients receiving corticosteroids have to receive a stable or decreasing dose for at least 14 days before start of study treatment. Exclusion criteria: Phase I and Phase II Parts: Less than 12 weeks between radiotherapy and start of study treatment, unless new enhancing lesion outside of radiation field or radiologically progressive on two consecutive MRI scans at least four weeks apart or biopsy-proven recurrence. Less than two weeks from surgical resection (one week from prior stereotactic biopsy) or major surgical procedure. Less than two weeks after previous chemotherapy (6 weeks from nitrosureas). Treatment with other investigational drugs; participation in another clinical study within the past 2 weeks before start of therapy or concomitantly with this study. Progressive disease or toxicity =CTCAEv3 Grade 3 to protracted temozolomide dosing (defined as temozolomide administered more than 5 days/28 day cycle). Active infectious disease requiring intravenous therapy. Known human immunodeficiency virus (HIV) infection or chronic Hepatitis B or C. Gastrointestinal disorders that may interfere with the absorption of the study drug or chronic diarrhea. Serious illness or concomitant non-oncological disease considered by the investigator to be incompatible with the protocol. Patient is <3 years free of another primary malignancy except: if the other primary malignancy is either not currently clinically significant or does not require active intervention (such as a basal cell skin cancer or a cervical carcinoma in situ). Existence of any other malignant disease is not allowed. Cardiac left ventricular function with resting ejection fraction <50%. Absolute neutrophil count (ANC) less than 1500/mm3. Platelet count less than 100,000/mm3. Bilirubin greater than 1.5 x upper limit of institutional norm. Aspartate amino transferase (AST) greater than 3 x upper limit of institutional norm. Serum creatinine greater than 1.5 x upper limit of institutional norm. Patients who are sexually active and unwilling to use a medically acceptable method of contraception. Pregnancy or breast-feeding. Patients unable to comply with the protocol. Known pre-existing interstitial lung disease (ILD). Phase I part only: 1. Less than four weeks from prior treatment with bevacizumab. Phase II Part only: Prior EGFR-directed therapy. Prior bevacizumab therapy. Patients presenting with second or higher number of episodes of recurrence. Requirement of treatment with any of the prohibited concomitant medications listed in Section 4.2.2 (Restrictions regarding concomitant treatment).
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Boehringer Ingelheim
Organizational Affiliation
Boehringer Ingelheim
Official's Role
Study Chair
Facility Information:
Facility Name
1200.36.0016 Boehringer Ingelheim Investigational Site
City
Birmingham
State/Province
Alabama
Country
United States
Facility Name
1200.36.0012 Boehringer Ingelheim Investigational Site
City
Phoenix
State/Province
Arizona
Country
United States
Facility Name
1200.36.0005 Boehringer Ingelheim Investigational Site
City
Duarte
State/Province
California
Country
United States
Facility Name
1200.36.0014 Boehringer Ingelheim Investigational Site
City
Los Angeles
State/Province
California
Country
United States
Facility Name
1200.36.0019 Boehringer Ingelheim Investigational Site
City
Orlando
State/Province
Florida
Country
United States
Facility Name
1200.36.0023 Boehringer Ingelheim Investigational Site
City
Atlanta
State/Province
Georgia
Country
United States
Facility Name
1200.36.0008 Boehringer Ingelheim Investigational Site
City
Louisville
State/Province
Kentucky
Country
United States
Facility Name
1200.36.0002 Boehringer Ingelheim Investigational Site
City
Boston
State/Province
Massachusetts
Country
United States
Facility Name
1200.36.0003 Boehringer Ingelheim Investigational Site
City
Detroit
State/Province
Michigan
Country
United States
Facility Name
1200.36.0009 Boehringer Ingelheim Investigational Site
City
New York
State/Province
New York
Country
United States
Facility Name
1200.36.0001 Boehringer Ingelheim Investigational Site
City
Durham
State/Province
North Carolina
Country
United States
Facility Name
1200.36.0007 Boehringer Ingelheim Investigational Site
City
Charleston
State/Province
South Carolina
Country
United States
Facility Name
1200.36.0020 Boehringer Ingelheim Investigational Site
City
Memphis
State/Province
Tennessee
Country
United States
Facility Name
1200.36.0017 Boehringer Ingelheim Investigational Site
City
Dallas
State/Province
Texas
Country
United States
Facility Name
1200.36.0010 Boehringer Ingelheim Investigational Site
City
Houston
State/Province
Texas
Country
United States
Facility Name
1200.36.0011 Boehringer Ingelheim Investigational Site
City
Charlottesville
State/Province
Virginia
Country
United States
Facility Name
1200.36.0022 Boehringer Ingelheim Investigational Site
City
Seattle
State/Province
Washington
Country
United States
Facility Name
1200.36.1005 Boehringer Ingelheim Investigational Site
City
Calgary
State/Province
Alberta
Country
Canada
Facility Name
1200.36.1010 Boehringer Ingelheim Investigational Site
City
Winnipeg
State/Province
Manitoba
Country
Canada
Facility Name
1200.36.1009 Boehringer Ingelheim Investigational Site
City
Moncton
State/Province
New Brunswick
Country
Canada
Facility Name
1200.36.1011 Boehringer Ingelheim Investigational Site
City
Halifax
State/Province
Nova Scotia
Country
Canada
Facility Name
1200.36.1008 Boehringer Ingelheim Investigational Site
City
Hamilton
State/Province
Ontario
Country
Canada
Facility Name
1200.36.1001 Boehringer Ingelheim Investigational Site
City
Kingston
State/Province
Ontario
Country
Canada
Facility Name
1200.36.1003 Boehringer Ingelheim Investigational Site
City
Toronto
State/Province
Ontario
Country
Canada
Facility Name
1200.36.1004 Boehringer Ingelheim Investigational Site
City
Toronto
State/Province
Ontario
Country
Canada
Facility Name
1200.36.1007 Boehringer Ingelheim Investigational Site
City
Fleurimont
State/Province
Quebec
Country
Canada
Facility Name
1200.36.1002 Boehringer Ingelheim Investigational Site
City
Montreal
State/Province
Quebec
Country
Canada
Facility Name
1200.36.1006 Boehringer Ingelheim Investigational Site
City
Quebec
Country
Canada

12. IPD Sharing Statement

Links:
URL
http://trials.boehringer-ingelheim.com
Description
Related Info
URL
http://trials.boehringer-ingelheim.com/content/dam/internet/opu/clinicaltrial/com_EN/results/1200/1200.36_U11-2804-01-DS.pdf
Description
Related Info
URL
http://trials.boehringer-ingelheim.com/content/dam/internet/opu/clinicaltrial/com_EN/results/1200/1200.36_Literature.pdf
Description
Related Info

Learn more about this trial

BIBW 2992 (Afatinib) With or Without Daily Temozolomide in the Treatment of Patients With Recurrent Malignant Glioma

We'll reach out to this number within 24 hrs