Erlotinib in Treating Patients With Recurrent Malignant Glioma or Recurrent or Progressive Meningioma

July 14, 2017 updated by: National Cancer Institute (NCI)

A Phase I/II Trial of OSI-774 in Patients With Recurrent Malignant Gliomas and Malignant Gliomas Post Radiation Therapy

Phase I/II trial to study the effectiveness of erlotinib in treating patients who have recurrent malignant glioma or recurrent or progressive meningioma. Erlotinib may stop the growth of tumor cells by blocking the enzymes necessary for tumor cell growth.

Study Overview

Detailed Description

OBJECTIVES:

Phase 1 I. Determine the maximum tolerated dose of erlotinib in patients with recurrent malignant glioma or recurrent or progressive meningioma.

II. Determine the safety profile of this drug in these patients. III. Determine the pharmacokinetics of this drug in these patients.

Phase 2 I. Determine the 6-month progression-free survival (recurrent malignant glioma) II.12-month survival of patients treated with this drug (stable glioblastoma post radiation therapy)

Phase 2 - Secondary Recurrent Malignant Glioma I. Objective Tumor Response rate associated with erlotinib therapy in recurrent or progressive malignant glioma.

III. 12-month survival of patients treated with this drug Determine the safety profile of this drug in these patients. IV.. Determine the pharmacokinetics of this drug in these patients

OUTLINE: This is a dose-escalation, multicenter study. Patients are stratified according to study phase (I vs II), concurrent enzyme-inducing antiepileptic drugs (EIAEDs) (yes vs no), histology (recurrent GBM vs recurrent anaplastic glioma vs recurrent meningioma vs stable GBM), preoperative candidacy (yes vs no), and concurrent steroids (yes vs no).

Phase I: Patients concurrently receiving EIAEDs receive oral erlotinib once daily. Courses repeat every 4 weeks in the absence of disease progression or unacceptable toxicity.

Cohorts of 3-6 patients receive escalating doses of erlotinib until the maximum tolerated dose (MTD) is determined. The MTD is the dose preceding that at which at least 2 of 3 or 2 of 6 patients experience dose-limiting toxicity.

Phase II: Once the MTD is determined, additional patients concurrently receiving EIAEDs are treated with erlotinib as above at the phase II dose. Patients not concurrently receiving EIAEDs are treated with erlotinib as above at a predetermined dose.

Patients are followed for survival.

Study Type

Interventional

Enrollment (Actual)

136

Phase

  • Phase 2
  • Phase 1

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

    • California
      • Los Angeles, California, United States, 90095
        • University of California Los Angeles
      • San Francisco, California, United States, 94115
        • University of California San Francisco
    • Maryland
      • Bethesda, Maryland, United States, 20814
        • National Cancer Institute Neuro-Oncology Branch
    • New York
      • New York, New York, United States, 10065
        • Memorial Sloan Kettering Cancer Center
    • Pennsylvania
      • Pittsburgh, Pennsylvania, United States, 15232
        • University of Pittsburgh
    • Texas
      • Dallas, Texas, United States, 75235
        • University of Texas Southwestern Medical Center
    • Wisconsin
      • Madison, Wisconsin, United States, 53792
        • University of Wisconsin

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:

  • One of the following diagnoses:

    • Histologically confirmed intracranial malignant glioma

      • Glioblastoma multiforme (GBM), anaplastic astrocytoma, anaplastic oligodendroglioma, anaplastic mixed oligoastrocytoma, or malignant astrocytoma not otherwise specified
      • Original histology of low-grade glioma allowed provided a subsequent histology of malignant glioma is confirmed
    • Histologically or radiographically confirmed recurrent or progressive benign or malignant meningioma
  • Progressive disease or tumor recurrence on MRI or CT scan

    • Phase I: No more than 3 prior relapses and no more than 2 prior chemotherapy* or biologic therapy regimens
    • Phase II: No more than 2 prior relapses and no more than 2 prior chemotherapy* or biologic therapy regimens
  • Patients with progressive disease must have failed prior radiotherapy* that was completed at least 4 weeks ago

    • Patients with progressive disease between 4 and 12 weeks after completion of external beam radiotherapy must have clear evidence of progression on MRI
    • Patients with GBM who have completed external beam radiotherapy and do not show progression are eligible
    • Patients with progressive disease after interstitial brachytherapy or stereotactic radiosurgery must have confirmed true progression rather than radiation necrosis based upon positron-emission tomography, thallium scanning, MRI, or surgical documentation
  • Measurable or evaluable disease
  • Performance status - Karnofsky 60-100%
  • More than 8 weeks
  • WBC at least 3,000/mm^3
  • Absolute neutrophil count at least 1,500/mm^3
  • Platelet count at least 100,000/mm^3
  • Hemoglobin at least 10 mg/dL (transfusion allowed)
  • Bilirubin less than 1.5 times upper limit of normal (ULN)
  • SGOT less than 1.5 times ULN
  • Creatinine less than 1.5 mg/dL
  • None of the following ophthalmic abnormalities:

    • Abnormalities of the cornea (e.g., dry eye syndrome or Sjögren's syndrome)
    • Congenital abnormality (e.g., Fuch's dystrophy)
    • Abnormal slit-lamp examination using a vital dye (e.g., fluorescein or Bengal-Rose)
    • Abnormal corneal sensitivity test (Schirmer test or similar tear production test)
  • Patients found to have dry eyes on examination but have an otherwise normal examination allowed
  • No active infection
  • No other serious concurrent medical illness
  • No other malignancy within the past 3 years except nonmelanoma skin cancer or carcinoma in situ of the cervix
  • No other disease that would obscure toxicity or dangerously alter drug metabolism
  • No significant medical illness that would preclude study participation
  • Not pregnant or nursing
  • Negative pregnancy test
  • Fertile patients must use effective barrier contraception during and for 12 weeks after study participation
  • See Disease Characteristics
  • At least 1 week since prior thalidomide
  • At least 1 week since prior interferon
  • At least 4 weeks since prior SU5416 or other experimental biologic agents
  • See Disease Characteristics
  • No prior chemotherapy (including polifeprosan 20 with carmustine implant [Gliadel wafers]) for patients with stable GBM
  • At least 2 weeks since prior vincristine
  • At least 3 weeks since prior procarbazine
  • At least 6 weeks since prior nitrosoureas
  • At least 1 week since prior tamoxifen
  • See Disease Characteristics
  • Recovered from prior radiotherapy
  • No more than 6 weeks since prior external beam radiotherapy for patients with GBM without evidence of progression
  • Recovered from prior surgery
  • Recovered from prior therapy
  • At least 1 week since prior noncytotoxic agents (e.g., isotretinoin) except radiosensitizers
  • At least 4 weeks since prior cytotoxic therapy
  • At least 4 weeks since prior tipifarnib or imatinib mesylate
  • No prior erlotinib or other epidermal growth factor receptor inhibitors
  • No concurrent combination antiretroviral therapy for HIV-positive patients

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: Non-Randomized
  • Interventional Model: Parallel Assignment
  • Masking: None (Open Label)

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
Experimental: Phase 1 Dose Escalation

Phase I: Patients concurrently receiving EIAEDs receive oral erlotinib once daily. Courses repeat every 4 weeks in the absence of disease progression or unacceptable toxicity.

Cohorts of 3-6 patients receive escalating doses of erlotinib until the maximum tolerated dose (MTD) is determined. The MTD is the dose preceding that at which at least 2 of 3 or 2 of 6 patients experience dose-limiting toxicity.

erlotinib hydrochloride given orally

Other: pharmacological study.

correlative studies
given orally
Other Names:
  • OSI-774
  • erlotinib
  • CP-358,774
Experimental: Phase 2 recurrent malignant gliomas and nonprogressive GBM

Phase II: Patients not concurrently receiving EIAEDs are treated with erlotinib as above at a predetermined dose (150mg/day.

patients requiring surgery treated 7 days prior to tumor removal (150mg/day)

PK analysis and effects of erlotinib on epidermal growth factor receptor (EGFR)

erlotinib hydrochloride given orally

Other: pharmacological study, laboratory biomarker analysis.

correlative studies
correlative studies
given orally
Other Names:
  • OSI-774
  • erlotinib
  • CP-358,774

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Number of Dose Limiting Toxicity (DLT) Each Dose Level Phase I
Time Frame: 28 days
DLT Definition: any grade 3 thrombocytopenia and grade 4 anemia and neutropenia; any non-hematologic grade 3 toxicity; failure to recover from toxicities to be eligible for re-treatment with erlotinib within 2 weeks of the last dose of erlotinib.
28 days
Define Maximum Tolerated Dose (MTD) of Erlotinib by Phase 1 Cohorts
Time Frame: cycle 1 - 28 days
standard 3+3 dose escalation design 3 patients in each dose level, observed for 28 days before enrollment to next level. if none of the patients experienced DLT dose escalated, if 1 of 3 experienced DLT 3 more enrolled at that level, if none of the 3 additional pts had DLT escalate to next level, if one or more of the additional pts experienced DLT, the MTD was exceeded and 3 more patients were treated at the next lower dose (if only 3 pts treated at the lower dose). The MTD is the dose at which 0/3 or 1/6 patients have experienced a DLT with the next higher dose having at least 2/3 or 2/6 patients encountering DLT.
cycle 1 - 28 days
6 Months Progression-free Survival in Recurrent Malignant Gliomas (Phase II)
Time Frame: 6 months
Progression: 25% increase in the sum of products of all measurable lesions over smallest sum observed (over BL if no decrease), OR clear worsening of any evaluable disease, OR appearance of any new lesion/site, OR failure to return for evaluation due to death or deteriorating condition (unless clearly unrelated to this cancer).
6 months

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Percent of Participants With a Grade 3 or 4 Adverse Events Phase 1
Time Frame: 1 year
CTCAE
1 year
1 Year Survival - Phase II Newly Diagnosed GBM Post RT
Time Frame: At 1 year
12 month survival for newly diagnosed stable GBM post RT treated with erlotinib post RT
At 1 year
Overall Survival Newly Diagnosed GBM Post RT
Time Frame: 2 years
Overall Survival defined as Time from Start of treatment to time of death due to any cause
2 years
Response Rate (Complete or Partial Response) Graded Using Modified RECIST Criteria Phase II
Time Frame: At 1 year

Measurable: Bidimensionally measurable lesions w/ clearly defined margins by MRI

Evaluable: Unidimensionally measurable lesions, masses w/margins not clearly defined.

Complete Response (CR): Complete disappearance of all measurable/evaluable disease. No new lesions. No evidence of non-evaluable disease. Patients on minimal/no steroids.

Partial Response (PR): >/= to 50% decrease under baseline in the sum of products of perpendicular diameters of all measurable lesions. No progression of evaluable disease. Responders must be on same/decreasing doses of dexamethasone.

Stable/No Response: Does not qualify for CR, PR, or progression.

Progression: 25% increase in the sum of products of all measurable lesions over smallest sum observed (over BL if no decrease), OR clear worsening of any evaluable disease, OR appearance of any new lesion/site, OR failure to return for evaluation due to death or deteriorating condition (unless clearly unrelated to this cancer).

At 1 year
Percent of Patients With One or More Grade 3-5 Toxicity Described Based on the CTC Severity Grading Phase II
Time Frame: Up to 1 year
Summarized by descriptive statistics.
Up to 1 year
Time of Peak Plasma Concentration Per Dose Level Phase I (on Anticonvulsants) -
Time Frame: baseline, 1,2,4,6,8,12,24h post administration day 1 cycle 1.
plasma concentrations relative to erlotiniab administration and sample time and dose level
baseline, 1,2,4,6,8,12,24h post administration day 1 cycle 1.
Peak Plasma Concentration Per Dose Level Phase I (on Anticonvulsants) -
Time Frame: baseline, 1,2,4,6,8,12,24h post administration day 1 cycle 1. One sample on day 8 cycle 1 and day 1 of cycle 2,3 and 5
plasma concentrations relative to erlotiniab administration and sample time and dose level
baseline, 1,2,4,6,8,12,24h post administration day 1 cycle 1. One sample on day 8 cycle 1 and day 1 of cycle 2,3 and 5
Estimation of the Area Under the Curve Per Dose Level Phase I (on Anticonvulsants) -
Time Frame: baseline, 1,2,4,6,8,12,24h post administration day 1 cycle 1. One sample on day 8 cycle 1 and day 1 of cycle 2,3 and 5
plasma concentrations relative to erlotiniab administration and sample time and dose level
baseline, 1,2,4,6,8,12,24h post administration day 1 cycle 1. One sample on day 8 cycle 1 and day 1 of cycle 2,3 and 5
Trough Level Per Dose Level Phase I (on Anticonvulsants) -
Time Frame: cycle 1 day eight
plasma concentrations relative to erlotiniab administration and sample time and dose level
cycle 1 day eight
Peak Plasma Concentration Level for Recurrent Patients Not on Enzyme-inducing Antiepileptic Drugs - Phase 2 - Dose 150mg -
Time Frame: baseline, 1,2,4,6,8,12,24h post administration day 1 cycle 1. One sample on day 8 cycle 1 and day 1 of cycle 2,3 and 5
plasma concentrations relative to erlotiniab administration and sample time and dose level
baseline, 1,2,4,6,8,12,24h post administration day 1 cycle 1. One sample on day 8 cycle 1 and day 1 of cycle 2,3 and 5
Time to Peak Plasma Concentration for Recurrent Patients Not on Enzyme-inducing Antiepileptic Drugs - Phase 2 - Dose 150mg -
Time Frame: baseline, 1,2,4,6,8,12,24h post administration day 1 cycle 1. One sample on day 8 cycle 1 and day 1 of cycle 2,3 and 5
plasma concentrations relative to erlotiniab administration and sample time and dose level
baseline, 1,2,4,6,8,12,24h post administration day 1 cycle 1. One sample on day 8 cycle 1 and day 1 of cycle 2,3 and 5
Estimation of Area Under the Curve for Recurrent Patients Not on Enzyme-inducing Antiepileptic Drugs - Phase 2 - Dose 150mg-
Time Frame: baseline, 1,2,4,6,8,12,24h post administration day 1 cycle 1. One sample on day 8 cycle 1 and day 1 of cycle 2,3 and 5
plasma concentrations relative to erlotiniab administration and sample time and dose level
baseline, 1,2,4,6,8,12,24h post administration day 1 cycle 1. One sample on day 8 cycle 1 and day 1 of cycle 2,3 and 5
Trough Level for Recurrent Patients Not on Enzyme-inducing Antiepileptic Drugs - Phase 2 - Dose 150mg -
Time Frame: One sample on day 8 cycle 1
plasma concentrations relative to erlotiniab administration and sample time and dose level
One sample on day 8 cycle 1
Pharmacokinetics (Plasma) Level for Recurrent Patients Not on Enzyme-inducing Antiepileptic Drugs
Time Frame: Pre-surgery and time of resection
Drug administered 6 days prior to surgery
Pre-surgery and time of resection
Pharmacokinetics (Tissue) Level for Recurrent Patients Not on Enzyme-inducing Antiepileptic Drugs
Time Frame: Pre-surgery and time of resection
Drug administered 6 days prior to surgery
Pre-surgery and time of resection

Collaborators and Investigators

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

Investigators

  • Principal Investigator: Lauren Abrey, MD, National Cancer Institute (NCI)

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.

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

August 1, 2002

Primary Completion (Actual)

December 1, 2010

Study Completion (Actual)

December 1, 2010

Study Registration Dates

First Submitted

September 6, 2002

First Submitted That Met QC Criteria

January 26, 2003

First Posted (Estimate)

January 27, 2003

Study Record Updates

Last Update Posted (Actual)

August 17, 2017

Last Update Submitted That Met QC Criteria

July 14, 2017

Last Verified

July 1, 2017

More Information

Terms related to this study

Plan for Individual participant data (IPD)

Plan to Share Individual Participant Data (IPD)?

NO

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 Adult Anaplastic Astrocytoma

Clinical Trials on laboratory biomarker analysis

3
Subscribe