Testing Addition of an Anti-cancer Drug, Vorasidenib to Temozolomide, After Radiation for Advanced Brain Cancer

Study Purpose

This phase III trial compares the effect of vorasidenib to placebo in combination with usual treatment, temozolomide, in treating patients with newly diagnosed grade 3 astrocytoma after radiation. Temozolomide is in a class of medications called alkylating agents. It works by damaging the cell's deoxyribonucleic acid and may kill tumor cells and slow down or stop tumor growth. Vorasidenib citrate blocks the proteins made by the mutated IDH1 and IDH2 genes, which may help keep tumor cells from growing. It is a type of enzyme inhibitor and a type of targeted therapy. Adding vorasidenib to the usual treatment, temozolomide, may be more effective than temozolomide alone in treating patients with newly diagnosed grade 3 astrocytoma after radiation therapy.

Recruitment Criteria

Accepts Healthy Volunteers

Healthy volunteers are participants who do not have a disease or condition, or related conditions or symptoms

No
Study Type

An interventional clinical study is where participants are assigned to receive one or more interventions (or no intervention) so that researchers can evaluate the effects of the interventions on biomedical or health-related outcomes.


An observational clinical study is where participants identified as belonging to study groups are assessed for biomedical or health outcomes.


Searching Both is inclusive of interventional and observational studies.

Interventional
Eligible Ages 12 Years and Over
Gender All
More Inclusion & Exclusion Criteria

Inclusion Criteria:

  • - STEP 0: Histologic diagnosis of astrocytoma, IDH-mutant (central nervous system [CNS] WHO grade 3) - STEP 0: Available diagnostic slides (hematoxylin and eosin staining method [H&E] and immunohistochemical stains for central review) - STEP 0: Tissue available for central biomarker testing (CDKN2A/B and1p/19q co-deletion [all patients], and IDH1/IDH2 [if needed]) - STEP 1: Centrally-confirmed diagnosis of astrocytoma, IDH-mutant (CNS WHO grade 3) - STEP 1: Presence of IDH1 p.
R132 or IDH2 p.172 mutation, confirmed by central review of immunohistochemical stain or molecular testing results, with central confirmation of equivocal results.
  • - STEP 1: Absence of CDKN2A/B homozygous deletion by central testing.
  • - STEP 1: Absence of whole arm 1p/19q co-deletion (i.e. intact 1p/19q) by central testing.
  • - STEP 1: No evidence of spinal or leptomeningeal disease.
  • - STEP 1: No prior chemotherapy, cranial irradiation, IDH-inhibitor therapy, radiotherapy, vaccine therapy, small-molecule therapy, or laser ablation.
  • - STEP 1: Prior diagnostic surgery/resection/biopsy ≤ 6 months of registration.
  • - STEP 1: Planned radiotherapy and adjuvant chemotherapy.
  • - STEP 1: Age ≥ 12 years.
  • - STEP 1: Eastern Cooperative Oncology Group (ECOG) performance status ≤ 2 (or Karnofsky performance status [KPS] ≥ 60%) - STEP 1: Absolute neutrophil count (ANC) ≥ 1,500/mm^3.
  • - STEP 1: Hemoglobin ≥ 9 g/dL.
  • - STEP 1: Platelet count ≥ 100,000/mm^3.
  • - STEP 1: Total bilirubin ≤ 1.5 x upper limit of normal (ULN) * For patients with Gilbert syndrome, total bilirubin ≤ 1.0 x ULN.
  • - STEP 1: Aspartate aminotransferase (AST)(serum glutamic oxaloacetic transaminase [SGOT])/alanine aminotransferase (ALT)(serum glutamic pyruvic transaminase [SGPT]) ≤ 2.5 x ULN.
  • - STEP 1: Alkaline phosphatase ≤ 2.5 x ULN.
  • - STEP 1: Creatinine ≤ 2.0 x ULN or calculated (calc.
) creatinine clearance > 40 mL/min. * For patients ≥ 18 years of age, calculated using the Cockcroft-Gault equation. For patients < 18 years of age, calculated using the Bedside Schwartz method:
  • - Age: 10 to < 13 years; Maximum Serum Creatinine (mg/dL): 1.2 (male) 1.2 (female) - Age: 13 to < 16 years; Maximum Serum Creatinine (mg/dL): 1.5 (male) 1.4 (female) - Age: ≥ 16 years; Maximum Serum Creatinine (mg/dL): 1.7(male) 1.4 (female) - STEP 1: Not pregnant and not nursing, because this study involves agents whose genotoxic, mutagenic, and teratogenic effects on the developing fetus and newborn are unknown.
* Therefore, for women of childbearing potential only, a negative pregnancy test done ≤ 14 days prior to registration is required.
  • - STEP 1: Women and men of reproductive potential should agree to abstain from sexual intercourse or use two highly effective methods of birth control, at least one of which must be a barrier method, throughout their participation in this study and for at least 90 days after the last dose of vorasidenib.
Reproductive status and discussions about birth control measures should be documented in the patient's record. Abstinence is acceptable only as true abstinence when this is in line with the preferred and usual lifestyle of the patient; periodic abstinence (e.g. calendar, ovulation, symptothermal, post-ovulation methods) and withdrawal are not acceptable methods of birth control. Highly effective forms of birth control are defined as hormonal oral contraceptives, injectables, patches, intrauterine devices, intrauterine hormone release systems, bilateral tubal ligation, condoms with spermicide, or male partner sterilization.
  • - STEP 1: No severe or intercurrent illness, no active infection that requires systemic anti-infective therapy, and no active infection with an unexplained fever > 38.5°C within 7 days prior to registration.
  • - STEP 1: Patients with a prior or concurrent malignancy whose natural history or treatment does not have the potential to interfere with the safety or efficacy assessment of the investigational regimen are eligible for this trial.
  • - STEP 1: Patients must be able to tolerate or undergo an MRI.
  • - STEP 1: Patients with known HIV infection on effective anti-retroviral therapy with undetectable viral load within 6 months prior to registration are eligible for this trial.
  • - STEP 1: For patients with evidence of chronic hepatitis B virus (HBV) infection, the HBV viral load must be undetectable on suppressive therapy, if indicated.
  • - STEP 1: Patients with a history of hepatitis C virus (HCV) infection must have been treated and cured.
For patients with HCV infection who are currently on treatment, they are eligible if they have an undetectable HCV viral load.
  • - STEP 1: No significant active cardiac disease within 6 months prior to registration, including New York Heart Association Functional Classification class III or IV congestive heart failure, myocardial infarction, unstable angina, and/or stroke.
To be eligible for this trial, patients should be class 2B or better.
  • - STEP 1: No history of significant (grade ≥ 2) intratumoral or peri-tumoral hemorrhage.
  • - STEP 1: No known active inflammatory gastrointestinal disease, chronic diarrhea, prior gastric resection or lap band dysphagia, short-gut syndrome, gastroparesis, or other condition causing an inability to swallow oral formulations of agents.
Gastroesophageal reflux disease under medical treatment is allowed (assuming no drug interaction potential)
  • - STEP 1: No known hypersensitivity to any of the components of vorasidenib or temozolomide.
  • - STEP 1: No other acute or chronic medical condition or laboratory abnormality that may increase the risk associated with study participation or protocol therapy administration or may interfere with the interpretation of study results and, in the judgment of the Investigator, would make the patient inappropriate for entry into this study.
  • - STEP 1: No concurrent use of other investigational agents.
  • - STEP 1: No concurrent use of alternating tumor treating field (TTField) therapy.
  • - STEP 1: No concurrent use of therapeutic doses of steroids for glioma.
Concurrent use of physiologic doses of steroids (defined as equivalent of ≤ 10 mg prednisone daily) for medical conditions unrelated to glioma is allowed. Corticosteroids administered for reasons related to glioma should be used in the smallest dose possible to control symptoms of cerebral edema and mass effect and discontinued whenever possible.
  • - STEP 1: No concurrent use of warfarin sodium or any other Coumadin-derivative anticoagulant.
Patients must be off Coumadin-derivative anticoagulants for at least 7 days prior to registration. Low molecular weight heparin (LMWH) and factor Xa inhibitors are allowed.
  • - STEP 1: No concurrent use of strong and moderate CYP1A2 inhibitors, moderate CYP1A2 inducers, or CYP3A substrates where a minimal concentration change can reduce efficacy.
Patients should be transferred to other medications prior to registration.

Exclusion Criteria:

-

Trial Details

Trial ID:

This trial id was obtained from ClinicalTrials.gov, a service of the U.S. National Institutes of Health, providing information on publicly and privately supported clinical studies of human participants with locations in all 50 States and in 196 countries.

NCT07215910
Phase

Phase 1: Studies that emphasize safety and how the drug is metabolized and excreted in humans.

Phase 2: Studies that gather preliminary data on effectiveness (whether the drug works in people who have a certain disease or condition) and additional safety data.

Phase 3: Studies that gather more information about safety and effectiveness by studying different populations and different dosages and by using the drug in combination with other drugs.

Phase 4: Studies occurring after FDA has approved a drug for marketing, efficacy, or optimal use.

Phase 3
Lead Sponsor

The sponsor is the organization or person who oversees the clinical study and is responsible for analyzing the study data.

Alliance for Clinical Trials in Oncology
Principal Investigator

The person who is responsible for the scientific and technical direction of the entire clinical study.

Ugonma N Chukwueke, MDRifaquat M. Rahman, MDPatrick Y Wen, MD
Principal Investigator Affiliation Alliance for Clinical Trials in OncologyAlliance for Clinical Trials in OncologyAlliance for Clinical Trials in Oncology
Agency Class

Category of organization(s) involved as sponsor (and collaborator) supporting the trial.

Other, NIH
Overall Status Not yet recruiting
Countries
Conditions

The disease, disorder, syndrome, illness, or injury that is being studied.

Astrocytoma, IDH-Mutant, Grade 3
Additional Details

The primary and secondary objectives of the study: PRIMARY OBJECTIVE:

  • I. To determine if vorasidenib citrate (vorasidenib) and adjuvant temozolomide following radiation therapy improves progression-free survival (PFS) per blinded independent review in patients with newly-diagnosed IDH-mutant astrocytoma (World Health Organization [WHO] grade 3) compared to placebo given with adjuvant temozolomide following radiation therapy.
SECONDARY OBJECTIVES:
  • I. To evaluate the safety and tolerability of vorasidenib versus (vs.#46;) placebo in combination with temozolomide, following radiation therapy.
  • II. To evaluate PFS associated with vorasidenib vs.#46; placebo in combination with temozolomide following radiation therapy, defined by local institutional review.
  • III. To evaluate the efficacy of vorasidenib vs.#46; placebo in combination with adjuvant temozolomide, following radiation therapy, based on overall survival (OS).
  • IV. To evaluate the efficacy of vorasidenib vs.#46; placebo in combination with temozolomide, following radiation therapy, based on objective response rate (ORR), complete response (CR) + partial response (PR), time to response, time to CR+PR, duration of response, and duration of CR+PR, with response assessed per the blinded independent review using the Response Assessment in Neuro-Oncology (RANO) 2.0 criteria in patients with measurable tumor at baseline.
  • V. To evaluate the efficacy of vorasidenib vs.#46; placebo in combination with temozolomide, following radiation therapy, based on ORR, CR+PR, time to response, time to CR+PR, duration of response, and duration of CR+PR, with response assessed per local institutional review or investigator using the RANO 2.0 criteria.
  • VI. To evaluate the efficacy of vorasidenib vs.#46; placebo in combination with temozolomide, following radiation therapy, based on time to next intervention.
  • VII. To evaluate vorasidenib vs.#46; placebo in combination with temozolomide, following radiation, with respect to health-related quality of life (HRQoL) as assessed by the Functional Assessment of Cancer Therapy-Brain (FACT-Br) and symptom burden as assessed by the MD Anderson Symptom Inventory-Brain Tumor (MDASI-BT).
EXPLORATORY OBJECTIVES:
  • I. To correlate tumor genotype with PFS.
  • II. To evaluate the effect of the addition of vorasidenib to adjuvant temozolomide on seizure control.
OUTLINE: Patients are randomized 1:1 to 1 of 2 arms. ARM I (CONTROL): Patients receive intensity-modulated radiation therapy (IMRT)/volume modulated arc therapy (VMAT) or pencil beam scanning (PBS) or intensity-modulated proton therapy (IMPT) once daily (QD) on Monday-Friday for 33 fractions. Starting 4 weeks after radiotherapy, patients receive temozolomide orally (PO) QD on days 1-5 and placebo PO QD on days 1-28 of each cycle. Cycles of combination treatment repeat every 28 days for up to 12 months and placebo alone repeats every 28 days in the absence of disease progression or unacceptable toxicity. Additionally, patients undergo blood sample collection and magnetic resonance imaging (MRI) throughout the study. ARM II (EXPERIMENTAL): Patients receive IMRT/VMAT or PBS or IMPT QD on Monday-Friday for 33 fractions. Starting 4 weeks after radiotherapy, patients receive temozolomide PO QD on days 1-5 and vorasidenib PO QD on days 1-28 of each cycle. Cycles of combination treatment repeat every 28 days for up to 12 months and vorasidenib alone repeats every 28 days in the absence of disease progression or unacceptable toxicity. Additionally, patients undergo blood sample collection and MRI throughout the study. After completion of study treatment, patients are followed up every 3 months for the first 2 years, every 4 months for the next 2 years, and then every 6 months for up to 10 years.

Arms & Interventions

Arms

Placebo Comparator: Arm I (temozolomide, placebo)

Patients receive IMRT/VMAT or PBS or IMPT QD on Monday-Friday for 33 fractions. Starting 4 weeks after radiotherapy, patients receive temozolomide PO QD on days 1-5 and placebo PO QD on days 1-28 of each cycle. Cycles of combination treatment repeat every 28 days for up to 12 months and placebo alone repeats every 28 days in the absence of disease progression or unacceptable toxicity. Additionally, patients undergo blood sample collection and MRI throughout the study.

Experimental: Arm II (temozolomide, vorasidenib)

Patients receive IMRT/VMAT or PBS or IMPT QD on Monday-Friday for 33 fractions. Starting 4 weeks after radiotherapy, patients receive temozolomide PO QD on days 1-5 and vorasidenib PO QD on days 1-28 of each cycle. Cycles of combination treatment repeat every 28 days for up to 12 months and vorasidenib alone repeats every 28 days in the absence of disease progression or unacceptable toxicity. Additionally, patients undergo blood sample collection and MRI throughout the study.

Interventions

Radiation: - Intensity-Modulated Radiation Therapy

Undergo IMRT/VMAT

Radiation: - Volume Modulated Arc Therapy

Undergo IMRT/VMAT

Radiation: - Pencil Beam Scanning

Undergo PBS

Procedure: - Intensity-Modulated Proton Therapy

Undergo IMPT

Drug: - Temozolomide

Given PO

Drug: - Vorasidenib

Given PO

Drug: - Placebo Administration

Given PO

Procedure: - Biospecimen Collection

Undergo blood sample collection

Procedure: - Magnetic Resonance Imaging

Undergo MRI

Other: - Questionnaire Administration

Ancillary Studies

Contact Information

This trial has no sites locations listed at this time. If you are interested in learning more, you can contact the trial's primary contact:

Alexandra Alexandra LeVasseur

[email protected]

773-834-4518

For additional contact information, you can also visit the trial on clinicaltrials.gov.

Stay Informed & Connected