BIBW 2992 (Afatinib) vs Gemcitabine-cisplatin in 1st Line Non-small Cell Lung Cancer (NSCLC)

December 13, 2018 updated by: Boehringer Ingelheim

LUX-Lung 6: A Randomized, Open-label, Phase III Study of BIBW 2992 Versus Chemotherapy as First-line Treatment for Patients With Stage IIIB or IV Adenocarcinoma of the Lung Harbouring an EGFR Activating Mutation

To investigate the efficacy and safety of BIBW 2992 compared to standard first-line chemotherapy in patients with stage IIIB or IV adenocarcinoma of the lung harbouring an EGFR activating mutation

Study Overview

Study Type

Interventional

Enrollment (Actual)

364

Phase

  • Phase 3

Contacts and Locations

This section provides the contact details for those conducting the study, and information on where this study is being conducted.

Study Locations

      • Beijing, China, 100071
        • 307 Hospital of PLA
      • Beijing, China, 100730
        • Peking Union Medical College Hospital
      • Beijing, China, 100020
        • Beijing Chao-Yang Hospital
      • Beijing, China, 101149
        • Beijing Chest Hospital
      • Changchun, China, 130021
        • First Hospital of Jilin University
      • Changsha, China, 410008
        • Xiangya Hospital, Central South University
      • Changsha, China
        • Hunan Province Tumor Hospital
      • Chengdu, China, 610041
        • West China Hospital
      • Fuzhou, China, 350014
        • Fujian Provincial Tumor Hospital
      • Guangzhou, China, 510080
        • Guangdong General Hospital
      • Guangzhou, China, 510120
        • Guangzhou Institute of Respiratory Disease
      • Guangzhou, China, 510515
        • NanFang Hosptial
      • Haerbin, China, 150081
        • The Third Affiliated Hospital of Harbin Medical University
      • Hangzhou, China, 310022
        • Zhejiang Cancer Hospital
      • HongShan, China
        • Hubei Cancer Hospital
      • Kunming, China, 650118
        • Yunnan Provincial Tumor Hospital
      • Linyi, China, 276001
        • Lin Yi Tumor Hospital
      • Nan Ning, China
        • The Affiliated Cancer Hospital, Guangxi Medical University
      • Nanjing, China, 210009
        • Jiangsu Cancer Hospital
      • Nanjing, China, 210002
        • the 81th Hospital of PLA
      • Qingdao, China, 266101
        • The affiliated hospital of medicalcollege qingdao university
      • Shanghai, China, 200030
        • Shanghai Chest Hospital
      • Shanghai, China, 200433
        • Shanghai Pulmonary Hospital
      • Shanghai, China, 200032
        • Zhongshan Hospital Fudan University
      • Shanghai, China, 200003
        • Shanghai Changzheng Hospital
      • Shanghai, China, 200443
        • Changhai Hospital
      • Shenyang, China, 110001
        • The First Hospital of Chinese Medical University
      • Shijiazhuang, China
        • Hebei Provincial Tumor Hospital
      • Xi'An, China, 710038
        • Tangdu Hospital
      • Yangzhou, China, 225001
        • Northern Jiangsu People's Hospital
      • Busan, Korea, Republic of, 602-702
        • Kosin University Gospel Hospital
      • Cheongju, Korea, Republic of, 361711
        • Chungbuk National University Hospital
      • Daegu, Korea, Republic of, 705-717
        • Yeungnam University Medical Center
      • Seoul, Korea, Republic of, 143-729
        • Konkuk University Medical Center
      • Seoul, Korea, Republic of, 152-703
        • Korea University Guro Hospital
      • Songkla, Thailand, 90110
        • Songklanagarind Hospital

Participation Criteria

Researchers look for people who fit a certain description, called eligibility criteria. Some examples of these criteria are a person's general health condition or prior treatments.

Eligibility Criteria

Ages Eligible for Study

18 years and older (Adult, Older Adult)

Accepts Healthy Volunteers

No

Genders Eligible for Study

All

Description

Inclusion criteria:

  1. pathologically confirmed diagnosis of stage IIIB or stage IV adenocarcinoma of the Lung
  2. EGFR(Epidermal Growth Factor Receptor) mutation detected by central laboratory analysis of tumor biopsy material
  3. Measurable disease according to Response Evaluation Criteria in Solid Tumours (RECIST)1.1
  4. Eastern Cooperative Oncology Group (ECOG) score of 0 or 1.
  5. Age>=18 years
  6. life expectancy of at least three months
  7. Written informed consent that is consistent with International Conference on Harmonisation-Good Clinical Practice (ICH-GCP) guidelines.

Exclusion criteria:

  1. Prior chemotherapy for relapsed and/or metastatic NSCLC.
  2. Prior treatment with EGFR targeting small molecules or antibodies.
  3. Radiotherapy or surgery(other than biopsy) within 4 weeks prior to randomization
  4. Active brain metastases
  5. Any other current malignancy or malignancy diagnosed within the past 5 years
  6. Known pre-existing interstitial lung disease
  7. Significant or recent acute gastrointestinal disorders with diarrhoea as a a major symptoms.
  8. History or presence of clinically relevant cardiovascular abnormalities
  9. Cardiac left ventricular function with resting ejection fraction of less than 50%.
  10. Any other concomitant serious illness or organ system dysfunction which in the opinion of the investigator would either compromise patient safety or interfere with the evaluation of the safety of the test drug.
  11. Absolute neutrophil count(ANC)<1500/mm3
  12. Platelet count<100,000/mm3
  13. Creatinine clearance<60ml/min or serum creatinine>1.5 times Upper Limit of Normal (ULN).
  14. Bilirubin>1.5 times ULN
  15. Aspartate Amino Transferase (AST) or Alanine Amino Transferase (ALT) > 3 times ULN
  16. Women of childbearing potential, or men who are able to father a child, unwilling to use a medically acceptable method of contraception during the trial
  17. Pregnancy of breast-feeding
  18. Patients unable to comply with the protocol
  19. Active hepatitis B infection, active hepatitis C infection or known HIV(Human Immunodeficiency Virus) carrier.
  20. Known or suspected active drug or alcohol abuse.
  21. requirement for treatment with any of the prohibited concomitant medications listed in section 4.2.2
  22. Any contraindications for therapy with gemcitabine/cisplatin
  23. Known hypersensitivity to BIBW2992 or the excipient of any of the trial drugs
  24. Use of any investigational drug within 4 weeks of randomization.

Study Plan

This section provides details of the study plan, including how the study is designed and what the study is measuring.

How is the study designed?

Design Details

  • Primary Purpose: Treatment
  • Allocation: Randomized
  • Interventional Model: Parallel Assignment
  • Masking: None (Open Label)

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Experimental: Arm A BIBW 2992
Patients receive a tablet of BIBW 2992 daily until progression or unacceptable toxicity
starting dose is 40 mg, in the event of no or minimal drug-related adverse events after one course, the dose will be increased to 50mg. in the event of certain drug related Adverse Event (AE), dose reduction will be increments of 10 mg, with the lowest dose being 20mg.
Active Comparator: Arm B Chemotherapy
Patients receive Gemcitabine and Cisplatin, maximum is 6 courses
Gemcitabine d1,8, Cisplatin d1, 21 days as a course, up to 6 courses.

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Progression-free Survival
Time Frame: Tumour assessment were performed at screening, week 6, 12, 18, 24, 30, 36, 42, 48 and then every 12 weeks until progression or death whichever occurs first up to week 168

The primary endpoint was progression-free survival (PFS) as assessed by central independent review according to Response Evaluation Criteria in Solid Tumours (RECIST) version 1.1. Progression-free survival was defined as the time from randomisation to disease progression or death whichever occurs earlier. A pre-defined set of censoring rules were used for patients who did not have a PFS.

Only data collected up until the analysis cut-off date (27 December 2013) were considered. Median time results from unstratified Kaplan-Meier estimates.

Tumour assessment were performed at screening, week 6, 12, 18, 24, 30, 36, 42, 48 and then every 12 weeks until progression or death whichever occurs first up to week 168

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Objective Response (OR)
Time Frame: Tumour assessment were performed at screening, week 6, 12, 18, 24, 30, 36, 42, 48 and then every 12 weeks until progression or death whichever occurs first up to week 374

OR is defined as a best overall response of complete response (CR) or partial response (PR) assessed by central independent review according to RECIST version 1.1 and will be presented as the percentage of patients with OR.

CR is defined as the disappearance of all target lesions and non-target lesions and no new lesions.

PR is defined as at least a 30% decrease in the sum of longest diameter (LD) of target lesions taking as reference the baseline sum LD, non-Progressive Disease or non evaluation of all non-target lesions and no new lesions.

(Exact 95% Confidence interval by Clopper and Pearson.)

Tumour assessment were performed at screening, week 6, 12, 18, 24, 30, 36, 42, 48 and then every 12 weeks until progression or death whichever occurs first up to week 374
Disease Control (DC)
Time Frame: Tumour assessment were performed at screening, week 6, 12, 18, 24, 30, 36, 42, 48 and then every 12 weeks until progression or death whichever occurs first up to week 374
DC is defined as a patient with objective response (OR) or stable disease (SD) assessed by central independent review according to RECIST version 1.1 and will be presented as the percentage of patients with DC.
Tumour assessment were performed at screening, week 6, 12, 18, 24, 30, 36, 42, 48 and then every 12 weeks until progression or death whichever occurs first up to week 374
Overall Survival (OS)
Time Frame: From randomisation up to 374 weeks

OS is defined as the time from randomisation to death. For patients who had not died until 7 December 2017, the date they were last known to be alive was derived from patient status records, the trial completion record, radiological imaging assessments, the study treatment termination record, and the randomisation date.

Median time results from unstratified Kaplan-Meier estimates.

From randomisation up to 374 weeks
Time to Objective Response (OR)
Time Frame: Tumour assessment were performed at screening, week 6, 12, 18, 24, 30, 36, 42, 48 and then every 12 weeks until progression or death whichever occurs first up to week 374

OR is defined as a best of overall response of complete response (CR) or partial response (PR) assessed by central independent review according to RECIST version 1.1.

For patients with an objective response, time to objective response was defined as the time from randomisation to the first objective response.

Outcome data are the percentage of patients with OR by each scheduled tumour assessment.

Tumour assessment were performed at screening, week 6, 12, 18, 24, 30, 36, 42, 48 and then every 12 weeks until progression or death whichever occurs first up to week 374
Duration of Objective Response
Time Frame: Tumour assessment were performed at screening, week 6, 12, 18, 24, 30, 36, 42, 48 and then every 12 weeks until progression or death whichever occurs first up to week 374

OR is defined as a best of overall response of complete response (CR) or partial response (PR) assessed by central independent review according to RECIST version 1.1.

For patients with an objective response, duration of objective response was defined as the time from the first objective response to disease progression or death whichever occurs earlier. A pre-defined set of censoring rules were used for patients who did not progress/die. The Median values are Kaplan-Meier estimates.

Tumour assessment were performed at screening, week 6, 12, 18, 24, 30, 36, 42, 48 and then every 12 weeks until progression or death whichever occurs first up to week 374
Duration of Disease Control
Time Frame: Tumour assessment were performed at screening, week 6, 12, 18, 24, 30, 36, 42, 48 and then every 12 weeks until progression or death whichever occurs first up to week 374
For patients with disease control, duration of disease control was defined as the time from randomisation to progression or death whichever occurs first. A pre-defined set of censoring rules were used for patients who did not progress/die. The Median values are Kaplan-Meier estimates.
Tumour assessment were performed at screening, week 6, 12, 18, 24, 30, 36, 42, 48 and then every 12 weeks until progression or death whichever occurs first up to week 374
Tumour Shrinkage
Time Frame: Tumour assessment were performed at screening, week 6, 12, 18, 24, 30, 36, 42, 48 and then every 12 weeks until progression or death whichever occurs first up to week 374

Tumour shrinkage is calculated as the minimum post-baseline sum of longest diameters of target lesions (longest for non-nodal lesions, short axis for nodal lesions) (SLD), as assessed by central independent review. The mean of these minimum values are presented after adjusting for baseline sum of longest diameters and EGFR mutation category. Only data collected up until the analysis cut-off date (27 Dec 2013) were considered. A negative value means the smallest post-baseline SLD was smaller than baseline (decreased since baseline); a positive value means tumor size increased since baseline.

The means are adjusted for baseline sum of lesions and EGFR mutation category.

Tumour assessment were performed at screening, week 6, 12, 18, 24, 30, 36, 42, 48 and then every 12 weeks until progression or death whichever occurs first up to week 374
Change From Baseline in Body Weight
Time Frame: Baseline and throughout the study (every 3 weeks) until progression or death (whichever occurs first) up to 374 weeks.
The change from baseline to the lowest and the last body weight recorded or during the the study.
Baseline and throughout the study (every 3 weeks) until progression or death (whichever occurs first) up to 374 weeks.
Change From Baseline in Eastern Cooperative Oncology Group (ECOG) Performance Status (PS)
Time Frame: Baseline and throughout the study (every 3 weeks) until progression or death (whichever occurs first) up to 374 weeks.

The last ECOG performance score category recorded during the study. Outcome data are the percentage of patients with an shift of ECOG performance status from baseline to the last ECOG performance status.

ECOG PS measured on 6 point scale to assess participant's performance status. 0=Fully active, able to carry on all pre-disease activities without restriction;

  1. Restricted in physically strenuous activity, but ambulatory and able to carry out light or sedentary work;
  2. Ambulatory (>50 percent of waking hours), capable of all selfcare, unable to carry out any work activities;
  3. Capable of only limited self care, confined to bed or chair more then 50 percent of waking hours;
  4. Completely disabled, cannot carry on any selfcare, totally confined to bed or chair;
  5. Dead
Baseline and throughout the study (every 3 weeks) until progression or death (whichever occurs first) up to 374 weeks.
Health Related Quality of Life (HRQOL): Time of Deterioration in Coughing
Time Frame: Baseline and throughout the study (every 3 weeks) until progression or death (whichever occurs first) up to 374 weeks.

HRQOL as measured by standardised questionnaires (EuropeanOrganisation for Research and Treatment of Cancer (EORTC)) quality of life questionaires (OLQ-C30) and its lung cancer module (QLQ-LC13). Analysis for cough: QLQ-LC13, question 1.

Time to deterioration was defined as the time from randomisation to a score increase (i.e. worsened) of at least 10 points from baseline (0 to 100 point scale). Patients without deterioration (including those with disease progression) were censored at the date of the last available HRQOL assessment; patients without post-baseline assessments were censored at the date of randomisation. Patients were considered as having deteriorated at the time of death. The median is Kaplan-Meier estimates.

Baseline and throughout the study (every 3 weeks) until progression or death (whichever occurs first) up to 374 weeks.
Health Related Quality of Life (HRQOL): Time of Deterioration in Dyspnoea
Time Frame: Baseline and throughout the study (every 3 weeks) until progression or death (whichever occurs first) up to 374 weeks.

HRQOL as measured by OLQ-C30 and its lung cancer module (QLQ-LC13). Analysis for dyspnoea: composite of QLQ-LC13, questions 3 to 5; individual item from QLQ-C30, question 8.

Time to deterioration was defined as the time from randomisation to a score increase (i.e. worsened) of at least 10 points from baseline (0 to 100 point scale). Patients without deterioration (including those with disease progression) were censored at the date of the last available HRQOL assessment; patients without post-baseline assessments were censored at the date of randomisation. Patients were considered as having deteriorated at the time of death. The median is Kaplan-Meier estimates.

Baseline and throughout the study (every 3 weeks) until progression or death (whichever occurs first) up to 374 weeks.
Health Related Quality of Life (HRQOL): Time of Deterioration in Pain
Time Frame: Baseline and throughout the study (every 3 weeks) until progression or death (whichever occurs first) up to 374 weeks.

HRQOL as measured by OLQ-C30 and its lung cancer module QLQ-LC13. Analysis for pain: composite of QLQ-C30, questions 9 and 19; individual items from QLQ-LC13, questions 10, 11 and 12.

Time to deterioration was defined as the time from randomisation to a score increase (i.e. worsened) of at least 10 points from baseline (0 to 100 point scale). Patients without deterioration (including those with disease progression) were censored at the date of the last available HRQOL assessment; patients without post-baseline assessments were censored at the date of randomisation. Patients were considered as having deteriorated at the time of death. The median is Kaplan-Meier estimates.

Baseline and throughout the study (every 3 weeks) until progression or death (whichever occurs first) up to 374 weeks.
Pharmacokinetics of Afatinib at Day 22
Time Frame: Day 22 (course 2, visit 1)
Outcome data are the trough plasma concentrations of afatinib at day 22 after multiple daily dosing of 40mg afatinib and after dose escalation to 50mg or dose reduction to 30mg afatinib (no patient dose reduced to 20mg during the Pharmacokinetics (PK) assessment period).
Day 22 (course 2, visit 1)
Pharmacokinetics of Afatinib at Day 29
Time Frame: Day 29 (course 2, visit 2)
Outcome data are the trough plasma concentrations of afatinib at day 29 after multiple daily dosing of 40mg afatinib and after dose escalation to 50mg or dose reduction to 30mg afatinib (no patient dose reduced to 20mg during the PK assessment period).
Day 29 (course 2, visit 2)
Pharmacokinetics of Afatinib at Day 43
Time Frame: Day 43 (course 3, visit 1)
Outcome data are the trough plasma concentrations of afatinib at day 43 after multiple daily dosing of 40mg afatinib and after dose escalation to 50mg or dose reduction to 30mg afatinib (no patient dose reduced to 20mg during the PK assessment period).
Day 43 (course 3, visit 1)
Safety of Afatinib as Indicated by Intensity and Incidence of Adverse Events
Time Frame: From first administration of study medication up to 28 days after the last administration of study medication up to 374 weeks.
Safety of Afatinib as indicated by intensity and incidence of adverse events graded according to the US National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. Presented as the percentage of patients with an Adverse Events during the on-treatment period by highest CTCAE grade.
From first administration of study medication up to 28 days after the last administration of study medication up to 374 weeks.
Changes in Safety Laboratory Parameters
Time Frame: From first administration of study medication up to 28 days after the last administration of study medication up to 374 weeks.

Outcome data presented are the percentage of patients by worst CTCAE grade (only Grades 2 to 4 presented) on treatment for the following laboratory parameters: Potassium, Aspartate Aminotransferase (AST), Alanine Aminotransferase (ALT), Creatine and Creatine Kinase.

For Potassium; only CTCAE grades resulting from hypokalemia (low values) are presented.

From first administration of study medication up to 28 days after the last administration of study medication up to 374 weeks.

Collaborators and Investigators

This is where you will find people and organizations involved with this study.

Publications and helpful links

The person responsible for entering information about the study voluntarily provides these publications. These may be about anything related to the study.

General Publications

Helpful Links

Study record dates

These dates track the progress of study record and summary results submissions to ClinicalTrials.gov. Study records and reported results are reviewed by the National Library of Medicine (NLM) to make sure they meet specific quality control standards before being posted on the public website.

Study Major Dates

Study Start (Actual)

April 19, 2010

Primary Completion (Actual)

November 23, 2017

Study Completion (Actual)

November 26, 2017

Study Registration Dates

First Submitted

April 21, 2010

First Submitted That Met QC Criteria

May 11, 2010

First Posted (Estimate)

May 12, 2010

Study Record Updates

Last Update Posted (Actual)

December 14, 2018

Last Update Submitted That Met QC Criteria

December 13, 2018

Last Verified

December 1, 2018

More Information

This information was retrieved directly from the website clinicaltrials.gov without any changes. If you have any requests to change, remove or update your study details, please contact register@clinicaltrials.gov. As soon as a change is implemented on clinicaltrials.gov, this will be updated automatically on our website as well.

Clinical Trials on Adenocarcinoma

Clinical Trials on BIBW 2992

3
Subscribe