Clinical Trials Logo

Clinical Trial Details — Status: Completed

Administrative data

NCT number NCT03914742
Other study ID # ABTC-1801
Secondary ID UM1CA137443
Status Completed
Phase Phase 1/Phase 2
First received
Last updated
Start date January 3, 2020
Est. completion date October 31, 2023

Study information

Verified date February 2024
Source Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
Contact n/a
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial Summary

This phase I/II trial studies the side effects and how well BGB-290 and temozolomide work in treating patients with gliomas (brain tumors) with IDH1/2 mutations that have come back. BGB-290 may stop the growth of tumor cells by blocking some of the enzymes needed for cell growth. Drugs used in chemotherapy, such as temozolomide, work in different ways to stop the growth of tumor cells, either by killing the cells, by stopping them from dividing, or by stopping them from spreading. Giving BGB-290 and temozolomide may work better in treating patients with recurrent gliomas.


Description:

PRIMARY OBJECTIVES: (Phase I) I. Determine the safety and tolerability of the combination of PARP inhibitor BGB-290 (BGB-290) and temozolomide (TMZ) in patients with recurrent IDH1/2 mutant glioma, including the maximum tolerated dose (MTD) and characterization of dose-limiting toxicities (DLTs) in the Phase I portion. (Phase II) II. Determine the overall response rate of BGB-290 with TMZ in patients with recurrent IDH1/2-mutant gliomas that have progressed on TMZ and another alkylator (Arm A) in the Phase II portion. III. Determine the overall response rate of BGB-290 with TMZ in patients with recurrent IDH1/2-mutant glioma that have failed one alkylator with >= 12 months since last treatment (Arm B) in the Phase II portion. PRIMARY OBJECTIVES: I. Determine the safety and tolerability of the combination of PARP inhibitor BGB-290 (BGB-290) and temozolomide (TMZ) in patients with recurrent IDH1/2 mutant glioma, including the maximum tolerated dose (MTD) and characterization of dose-limiting toxicities (DLTs) in the Phase I portion. (Phase I) II. Determine the overall response rate of BGB-290 with TMZ in patients with recurrent IDH1/2-mutant gliomas that have progressed on TMZ and another alkylator (Arm A) in the Phase II portion. (Phase II) III. Determine the overall response rate of BGB-290 with TMZ in patients with recurrent IDH1/2-mutant glioma that have failed one alkylator with >= 12 months since last treatment (Arm B) in the Phase II portion. (Phase II) SECONDARY OBJECTIVES: I. Determine the progression-free survival (PFS) and overall survival (OS) after treatment with BGB-290 and TMZ in recurrent IDH1/2-mutant gliomas in Arms A and B. II. Determine the duration of response to therapy in recurrent IDH1/2-mutant glioma. III. Confirm the safety and tolerability of BGB-290 in combination with TMZ. EXPLORATORY OBJECTIVES: I. Assess tumor response rates, PFS, and OS in patients with World Health Organization (WHO) grade IV glioblastoma (GBM) treated with BGB-290 and TMZ. II. Assess the mutational landscape via whole-exome sequencing (WES). III. Assess gene expression patterns using ribonucleic acid (RNA) sequencing (RNAseq). IV. Assess the methylation profiling with Infinium methylation assays. V. Quantify 2-hydroxyglutarate (2HG) in archival formalin-fixed paraffin-embedded (FFPE) specimens via liquid chromatography mass spectrometry (LC-MS) detection and correlate with treatment response. VI. Correlate response with 2HG levels, somatic alterations, gene expression/methylation patterns in FFPE tumor tissue. VII. Assess tumor tissue BGB-290 levels, 2HG, and PolyADP-ribosylation (PARylation) in a patient subset treated with drug prior to re-resection. VIII. Evaluate changes in tumor growth rate in subjects with non-enhancing glioma based on fluid attenuated inverse recovery (FLAIR) tumor volume measurements of serial MRI exams. IX. Assess if change in tumor growth rate (based on FLAIR tumor volume) in subjects with non-enhancing glioma before and after treatment is associated with progression by Response Assessment in Neuro-Oncology for Low Grade Gliomas (RANO LGG; phase II patients only) or survival. OUTLINE: This is a phase I, dose de-escalation study of temozolomide followed by a phase II study. PHASE I: Patients receive PARP inhibitor BGB-290 orally (PO) twice daily (BID) on days 1-28 and temozolomide PO once daily (QD) on days 1-28, 1-21, 1-14, or 1-7. Courses repeat every 28 days in the absence of disease progression or unacceptable toxicity. SURGICAL PORTION: 10 patients eligible for re-resection at the time of recurrence receive PARP inhibitor BGB-290 PO BID on days 1-6 and QD on day 7 (the morning of surgery). Within 45 days after surgery, patients receive PARP inhibitor BGB-290 PO BID on days 1-28 and temozolomide on the schedule established in Phase I. Courses repeat every 28 days in the absence of disease progression or unacceptable toxicity. PHASE II: Patients receive PARP inhibitor BGB-290 PO BID on days 1-28 and temozolomide PO QD on the schedule established in Phase I. Courses repeat every 28 days in the absence of disease progression or unacceptable toxicity. After completion of study treatment, patients are followed up for 30 days, every 2 months for 2 years, then every 6 months thereafter.


Recruitment information / eligibility

Status Completed
Enrollment 60
Est. completion date October 31, 2023
Est. primary completion date October 31, 2023
Accepts healthy volunteers No
Gender All
Age group 18 Years and older
Eligibility Inclusion Criteria: - PHASE I: Patients must have histologically confirmed WHO grade II-III glioma that is progressive or recurrent following at least one prior chemotherapy regimen plus or minus radiation therapy regimen or (b) Grade IV disease in their recurrent resection or biopsy specimen or (c) Grade IV glioma at initial diagnosis, with recurrent disease. Phase I patients may have failed an unlimited number of prior systemic regimens. - PHASE II: Patients must have histologically confirmed WHO grade II-IV glioma that is progressive or recurrent following therapy: - Arm A patients must have WHO grade II-III glioma and have failed TMZ and another alkylator (e.g., carmustine, lomustine, procarbazine). Patients in Arm A may have failed an unlimited number of prior systemic regimens. Prior radiotherapy (RT) is not required for eligibility. There is no minimum time from the last antineoplastic treatment, except to allow for recovery: three weeks from last dose of TMZ and six weeks from last dose of nitrosourea. - Arm B patients must have WHO grade II-III glioma and have experienced tumor progression after TMZ or another alkylator (maximum one prior chemotherapy regimen), and have gone >= 12 months since last treatment (chemotherapy or RT). Prior radiation therapy (RT) is allowed but not mandated. - GBM Arm patients must have WHO grade IV glioblastoma following radiotherapy (45-60 gray [Gy] in 1.8-2.0 Gy fractions) plus chemotherapy and may have failed an unlimited number of prior systemic regimens. - Surgical portion patients must have histologically confirmed WHO grade II-IV glioma that is progressive or recurrent following therapy and must be undergoing repeat surgery that is clinically indicated as determined by their care providers. Surgical Portion patients may have had an unlimited number of prior therapy regimens. - Recurrence in non-enhancing tumors will be defined as 25% or more increase in bi-dimensional product of FLAIR signal abnormality (measurable disease) per the low-grade glioma (LGG) RANO criteria. Contrast-enhancing tumors with measurable enhancing targets will be defined as recurrent based on standard RANO criteria. - Patients with recurrent glioma < 12 weeks after completion of radiotherapy must have new enhancement outside of the RT field (beyond the high-dose region or 80% isodose line), or evidence of viable tumor on histopathologic sampling. - PHASE I AND PHASE II: Patients must have available at least 3 prior full sets of magnetic resonance imaging (MRI) scans (not including screening), each separated by at least 2 months. - Patients must have IDH1/2-mutant glioma. IDH1/2-mutation status can be confirmed by immunohistochemistry (IHC) or direct deoxyribonucleic acid (DNA) sequencing, provided that it is performed in a Clinical Laboratory Improvement Amendments/College of American Pathologists (CLIA/CAP)-certified laboratory. IDH1/2 mutations must be associated with neomorphic activity of the encoded proteins (i.e. IDH1 R132, IDH2 R172, IDH2 R140, IDH1 R100, IDH1 G97, IDH1 Y139). - Patients must have archival formalin-fixed paraffin-embedded (FFPE) specimens and mutations will be verified centrally, although this will not preclude patients with appropriate documentation of IDH1/2-mutant status from trial enrollment. Patients must have a tumor tissue form indicating availability of archived tissue from a previous surgery, completed and signed by a pathologist; sites must agree to provide this form within 14 days after treatment start. - Patients must have measurable (defined by at least 1 cm x 1 cm) contrast-enhancing disease or measurable abnormal T2/FLAIR hyperintensity indicative of tumor by MRI imaging within 21 days of starting treatment. - Patients must have documented molecular 1p/19q and MGMT testing. If either of these studies has not been performed previously, they can be done prior to enrollment. - Patients must be able to undergo MRI of the brain with gadolinium. Patients must be maintained on a stable or decreasing dose of corticosteroid regimen (no increase for 5 days) prior to this baseline MRI. - Patients must have recovered from severe toxicity of prior therapy. The following intervals from previous treatments are required to be eligible: - 12 weeks from the completion of radiation - 6 weeks from a nitrosourea chemotherapy - 3 weeks from a non-nitrosourea chemotherapy - 4 weeks from any investigational (not Food and Drug Administration [FDA]-approved) agents - 2 weeks from administration of a non-cytotoxic, FDA-approved agent (e.g., erlotinib, hydroxychloroquine, etc.). - Patients must have a Karnofsky performance (KPS) status >= 60% (i.e. the patient must be able to care for himself/herself with occasional help from others). - Absolute neutrophil count >= 1,500/ uL. - Platelets >= 100,000/ uL. - Hemoglobin >= 9 g/dL. - Total bilirubin =< institutional upper limit of normal. - Aspartate aminotransferase (AST) (serum glutamic oxaloacetic transaminase [SGOT])/alanine aminotransferase (ALT) (serum glutamate pyruvate transaminase [SGPT]) =< 4 × institutional upper limit of normal. - Creatinine =< institutional upper limit of normal OR creatinine clearance >= 60 ml/min/1.73m^2 for patients with creatinine levels above institutional normal. - Activated partial thromboplastin time (APTT) or PTT =< 1.5 × institutional upper limit of normal. - Patients must be able to provide written informed consent. - Women of childbearing potential must have a negative serum pregnancy test prior to study start. Women of childbearing potential and men must agree to use adequate contraception (hormonal or barrier method of birth control; abstinence) prior to study entry, for the duration of study participation, and through 4 months after the last dose of study drug. Should a woman become pregnant or suspect she is pregnant while she or her partner is participating in this study, she should inform her treating physician immediately. Men treated or enrolled on this protocol must also agree to use adequate contraception prior to the study, for the duration of study participation, and through 4 months after completion of BGB-290 or temozolomide administration. - Patients must have no concurrent malignancy except curatively treated basal or squamous cell carcinoma of the skin or carcinoma in situ of the cervix, breast, or bladder. Patients with prior malignancies must be disease-free for >= 5 years. - Patients must be able to swallow tablets and capsules. Exclusion Criteria: - Patients receiving any other investigational agents are ineligible. - Patients previously treated with a small molecule inhibitor of mutant IDH1/2 proteins are ineligible. - Patients with a history of allergic reactions attributed to compounds of similar chemical or biologic composition to BGB-290 are ineligible. - Patients who have received bevacizumab within the last 6 months are ineligible. - Patients with a known hypersensitivity to TMZ are ineligible. - Patients who have received a PARP inhibitor previously are excluded. - Patients on enzyme-inducing anti-epileptic drugs (EIAED) are not eligible for treatment on this protocol. Patients may be on non-enzyme inducing anti-epileptic drugs or not be taking any anti-epileptic drugs. Patients previously treated with EIAEDs may be enrolled if they have been off the EIAED for 10 days or more prior to the first dose of BGB-290. - Patients who have not recovered to < Common Terminology Criteria for Adverse Events (CTCAE) grade 2 toxicities apart from alopecia related to prior therapy are ineligible. - Patients with uncontrolled intercurrent illness including, but not limited to, ongoing or active infection, symptomatic congestive heart failure, clinically significant cardiac disease, unstable angina pectoris, cardiac arrhythmia, or psychiatric illness/social situations that would limit compliance with study requirements, are ineligible. - Pregnant women are excluded from this study because the effects of BGB-290 on a fetus are unknown. Because there is an unknown but potential risk for adverse events in nursing infants secondary to treatment of the mother with BGB-290, breastfeeding should be discontinued if the mother is treated with BGB-290. - Human immunodeficiency virus (HIV)-positive patients on combination antiretroviral therapy are ineligible due to potential drug-drug interactions with BGB-290.

Study Design


Intervention

Drug:
PARP Inhibitor BGB-290
Given PO
Temozolomide
Given PO
Procedure:
Therapeutic Conventional Surgery
resection surgery

Locations

Country Name City State
United States Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Baltimore Maryland
United States UAB Comprehensive Cancer Center Birmingham Alabama
United States Dana Farber Cancer Institute Boston Massachusetts
United States University of Virginia Charlottesville Virginia
United States Cleveland Clinic Taussig Cancer Center Cleveland Ohio
United States Josephine Ford Cancer Center at Henry Ford Hospital Detroit Michigan
United States University of California, Los Angeles Los Angeles California
United States Yale University New Haven Connecticut
United States University of Pennsylvania Philadelphia Pennsylvania
United States Hillman Cancer Center at University of Pittsburgh Cancer Institute Pittsburgh Pennsylvania
United States Washington University Saint Louis Missouri
United States Wake Forest University Comprehensive Cancer Center Winston-Salem North Carolina

Sponsors (3)

Lead Sponsor Collaborator
Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins BeiGene, National Cancer Institute (NCI)

Country where clinical trial is conducted

United States, 

Outcome

Type Measure Description Time frame Safety issue
Primary Phase I: Maximum tolerated dose (MTD) defined as the combination regimen that yields a dose limiting toxicity (DLT) rate less than, or equal to 33% up to 28 days
Primary Phase I: Percentage of participants with adverse events Percentage of participants who experience grade 3 or higher toxicities as defined by Common Terminology Criteria for Adverse Events (CTCAE) version 5.0 up to 90 days post treatment
Primary Phase II: Tumor radiographic response Number of participants with response as defined by Response Assessment in Neuro-oncology (RANO) criteria: Complete Response (CR)= no change in size of T1-gadolinium-enhancing (T1-Gd+) disease, stable or reduced T2/FLAIR signal, no new lesion, no corticosteroid use, and stable or improved clinical status; Partial Response (PR)= =50% change in size of T1-Gd+ disease, stable or reduced T2/FLAIR signal, no new lesion, stable or reduced corticosteroid use, and stable or improved clinical status; Stable Disease (SD)= <50% reduction to <25% increase size of T1-Gd+ disease, stable or reduced T2/FLAIR signal, no new lesion, stable or reduced corticosteroid use, and stable or improved clinical status; Progressive Disease (PD)= =25% increase size of T1-Gd+ disease, or increased T2/FLAIR signal, or presence of new lesion, or worsening clinical status. up to 2 years
Secondary Phase II: Progression-free survival (PFS) Time from date of treatment start to date of initial scan indicating progression up to 2 years
Secondary Phase II: Overall survival (OS) Median time from start date of treatment to date of death up to 2 years
Secondary Duration of response Time from date of initial scan indicating complete or partial response to a date of the initial scan deemed tumor progression. Response is defined by RANO criteria as follows: Complete Response (CR)= no change in size of T1-gadolinium-enhancing (T1-Gd+) disease, stable or reduced T2/FLAIR signal, no new lesion, no corticosteroid use, and stable or improved clinical status; Partial Response (PR)= =50% change in size of T1-Gd+ disease, stable or reduced T2/FLAIR signal, no new lesion, stable or reduced corticosteroid use, and stable or improved clinical status; Progressive Disease (PD)= =25% increase size of T1-Gd+ disease, or increased T2/FLAIR signal, or presence of new lesion, or worsening clinical status. up to 2 years
Secondary Percentage of participants with serious or life-threatening adverse events Percentage of participants with serious or life-threatening toxicities as defined by CTCAE version 5.0 up to 2 years
See also
  Status Clinical Trial Phase
Recruiting NCT05577091 - Tris-CAR-T Cell Therapy for Recurrent Glioblastoma Phase 1
Recruiting NCT05284643 - Spectroscopic MRI, Proton Therapy, and Avastin for Recurrent Glioblastoma N/A
Recruiting NCT05039281 - Atezolizumab and Cabozantinib for the Treatment of Recurrent Glioblastoma Phase 1/Phase 2
Recruiting NCT04988750 - Evaluate the Safety and Preliminary Efficacy of the Combination of NaviFUS System With Re-irradiation for rGBM Patients N/A
Recruiting NCT06058988 - Trastuzumab Deruxtecan (T-DXd) for People With Brain Cancer Phase 2
Completed NCT00503204 - Phase I : Cediranib in Combination With Lomustine Chemotherapy in Recurrent Malignant Brain Tumour Phase 1
Completed NCT03216499 - HIF-2 Alpha Inhibitor PT2385 in Treating Patients With Recurrent Glioblastoma Phase 2
Not yet recruiting NCT04717999 - Pilot Study of NKG2D CAR-T in Treating Patients With Recurrent Glioblastoma N/A
Not yet recruiting NCT05540275 - Tislelizumab (One Anti-PD-1 Antibody) Plus Low-dose Bevacizumab for Bevacizumab Refractory Recurrent Glioblastoma Phase 2
Recruiting NCT04528680 - Ultrasound-based Blood-brain Barrier Opening and Albumin-bound Paclitaxel and Carboplatin for Recurrent Glioblastoma Phase 1/Phase 2
Active, not recruiting NCT04044937 - Fluoroethyltyrosine for Evaluation of Intracranial Neoplasms Phase 2
Recruiting NCT04888611 - Neoadjuvant PD-1 Antibody Alone or Combined With DC Vaccines for Recurrent Glioblastoma Phase 2
Completed NCT00390299 - Viral Therapy in Treating Patients With Recurrent Glioblastoma Multiforme Phase 1
Recruiting NCT05463848 - Surgical Pembro +/- Olaparib w TMZ for rGBM Phase 2
Active, not recruiting NCT04479241 - LUMINOS-101: Lerapolturev (PVSRIPO) and Pembrolizumab in Patients With Recurrent Glioblastoma Phase 2
Active, not recruiting NCT00777153 - Cediranib in Combination With Lomustine Chemotherapy in Recurrent Glioblastoma Phase 3
Withdrawn NCT05017610 - Inducing a Hypothyroxinemic State in Patients With Recurrent Glioblastoma or Gliosarcoma Early Phase 1
Recruiting NCT04323046 - Immunotherapy Before and After Surgery for Treatment of Recurrent or Progressive High Grade Glioma in Children and Young Adults Phase 1
Active, not recruiting NCT05324501 - A Study of Intra-tumoral Administered MTX110 in Patients With Recurrent Glioblastoma Phase 1
Withdrawn NCT05666349 - Reirradiation and Niraparib in Patients With Recurrent Glioblastoma Phase 1