76 results on '"Mark R. Gilbert"'
Search Results
2. Hippocampal Avoidance During Whole-Brain Radiotherapy Plus Memantine for Patients With Brain Metastases: Phase III Trial NRG Oncology CC001
- Author
-
Steven J. Chmura, Kiran Devisetty, Baldassarre Stea, Jeffrey S. Wefel, Lisa A. Kachnic, Paul D. Brown, Minesh P. Mehta, David R. Grosshans, Vinai Gondi, Wenyin Shi, Joseph Bovi, Cliff G. Robinson, Vijayananda Kundapur, Bethany Anderson, Tammie L.S. Benzinger, Deborah Watkins Bruner, Deepak Khuntia, David Roberge, Andre Konski, Kenneth Y. Usuki, Jing Li, Snehal Deshmukh, Terri Armstrong, Nadia N. Laack, Wolfgang A. Tomé, Harold Yoon, Sunjay Shah, Tim J. Kruser, Mark R. Gilbert, and Stephanie L. Pugh
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Hippocampus ,law.invention ,Antiparkinson Agents ,03 medical and health sciences ,Cognition ,0302 clinical medicine ,Randomized controlled trial ,Memantine ,law ,Internal medicine ,medicine ,Humans ,Progression-free survival ,Radiation Injuries ,Proportional Hazards Models ,Brain Neoplasms ,business.industry ,Radiotherapy Planning, Computer-Assisted ,Chemoradiotherapy ,Middle Aged ,Progression-Free Survival ,Radiation therapy ,Clinical trial ,030220 oncology & carcinogenesis ,Toxicity ,Quality of Life ,Female ,Radiotherapy, Intensity-Modulated ,Cognition Disorders ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
PURPOSE Radiation dose to the neuroregenerative zone of the hippocampus has been found to be associated with cognitive toxicity. Hippocampal avoidance (HA) using intensity-modulated radiotherapy during whole-brain radiotherapy (WBRT) is hypothesized to preserve cognition. METHODS This phase III trial enrolled adult patients with brain metastases to HA-WBRT plus memantine or WBRT plus memantine. The primary end point was time to cognitive function failure, defined as decline using the reliable change index on at least one of the cognitive tests. Secondary end points included overall survival (OS), intracranial progression-free survival (PFS), toxicity, and patient-reported symptom burden. RESULTS Between July 2015 and March 2018, 518 patients were randomly assigned. Median follow-up for alive patients was 7.9 months. Risk of cognitive failure was significantly lower after HA-WBRT plus memantine versus WBRT plus memantine (adjusted hazard ratio, 0.74; 95% CI, 0.58 to 0.95; P = .02). This difference was attributable to less deterioration in executive function at 4 months (23.3% v 40.4%; P = .01) and learning and memory at 6 months (11.5% v 24.7% [ P = .049] and 16.4% v 33.3% [ P = .02], respectively). Treatment arms did not differ significantly in OS, intracranial PFS, or toxicity. At 6 months, using all data, patients who received HA-WBRT plus memantine reported less fatigue ( P = .04), less difficulty with remembering things ( P = .01), and less difficulty with speaking ( P = .049) and using imputed data, less interference of neurologic symptoms in daily activities ( P = .008) and fewer cognitive symptoms ( P = .01). CONCLUSION HA-WBRT plus memantine better preserves cognitive function and patient-reported symptoms, with no difference in intracranial PFS and OS, and should be considered a standard of care for patients with good performance status who plan to receive WBRT for brain metastases with no metastases in the HA region.
- Published
- 2020
3. ENLIGHT: Pancancer response prediction to targeted and immunotherapies via tumor transcriptomics
- Author
-
Ranit Aharonov, Gal Dinstag, Eldad Shulman, Efrat Elis, Doreen Ben-Zvi, Omer Tirosh, Danh-Tai Hoang, Sanju SInha, Andrea B. Apolo, William L. Dahut, Stan Lipkowitz, Raanan Berger, Razelle Kurzrock, Antonios Papanicolau-Sengos, Fatima Karzai, Padma Sheila Rajagopal, Mark R. Gilbert, Kenneth D. Aldape, Tuvik Beker, and Eytan Ruppin
- Subjects
Cancer Research ,Oncology - Abstract
e13556 Background: Precision oncology is gradually advancing into mainstream clinical practice. Despite the significant recent growth in the number of approved biomarkers for immune and targeted therapies, demonstrating significant survival benefits, eligibility and response rates remain limited in many cases, calling for better predictive biomarkers. Methods: We developed ENLIGHT - a transcriptomics-based computational platform that identifies and utilizes clinically relevant genetic interactions (GIs) to predict a patient’s response to cancer treatments. ENLIGHT markedly extends and improves upon SELECT, a previous GI-based framework for obtaining transcriptomics-based response biomarkers. In this study, we focus on three key translational aspects: (i) the number of drugs for which predictions can be obtained (ii) defining a biomarker-based test for Personalized Medicine (PM) that would identify favorable treatments for an individual; and (iii) Improving clinical trial design by excluding a sub-population of patients likely not to respond to the treatment. A key translational feature of the approach is that it does not require training on treatment response data. Thus, in addition to its ability to predict patients' response to approved and well-studied therapies, it can predict the response to new, unexplored treatments. Results: We first tested Enlight in the PM scenario, analyzing 21 patient cohorts from diverse indications, treated with a variety of targeted and immunotherapies. The ENLIGHT treatment matching score is associated with better response with an aggregate Odds Ratio (OR) of 2.59 (95% confidence interval [1.85, 3.55], p= 3.41 e-8). Applied to the WINTHER trial data, encompassing multiple indications and individualized treatments, ENLIGHT recommendations achieved a highly remarkable OR of 11.15 ([2.3, 54.5], p = 8 e-04), demonstrating ENLIGHT’s strong predictive power across a broad spectrum of treatments and cancer types. Second, using ENLIGHT to exclude patients from clinical trials increases the trial response rate, achieving more than 90% of the response rate attainable under an optimal exclusion strategy. Conclusions: ENLIGHT is a powerful transcriptomics-based precision oncology pipeline developed by Pangea Biomed that broadly predicts response to both extant and novel targeted and immune therapies in translationally oriented, clinical terms, going beyond case-specific biomarkers.
- Published
- 2022
4. Radiation Therapy for Glioblastoma: American Society of Clinical Oncology Clinical Practice Guideline Endorsement of the American Society for Radiation Oncology Guideline
- Author
-
Matthew Mumber, Vinai Gondi, Mark R. Gilbert, Terri S. Armstrong, Nofisat Ismaila, Mary Lovely, Andrew E. Sloan, Minesh P. Mehta, Timothy F. Cloughesy, Susan M. Chang, Evanthia Galanis, Tracy T. Batchelor, Erik P. Sulman, and Christina Tsien
- Subjects
Cancer Research ,medicine.medical_specialty ,Consensus ,Biopsy ,medicine.medical_treatment ,MEDLINE ,03 medical and health sciences ,0302 clinical medicine ,Quality of life ,Temozolomide ,Humans ,Medicine ,Medical physics ,Antineoplastic Agents, Alkylating ,Evidence-Based Medicine ,Performance status ,Brain Neoplasms ,business.industry ,Patient Selection ,Chemoradiotherapy ,Guideline ,Evidence-based medicine ,Dacarbazine ,Radiation therapy ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Dose Fractionation, Radiation ,Cranial Irradiation ,Glioblastoma ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Purpose The American Society for Radiation Oncology (ASTRO) produced an evidence-based guideline on radiation therapy for glioblastoma. Because of its relevance to the ASCO membership, ASCO reviewed the guideline and applied a set of procedures and policies used to critically examine guidelines developed by other organizations. Methods The ASTRO guideline on radiation therapy for glioblastoma was reviewed for developmental rigor by methodologists. An ASCO endorsement panel updated the literature search and reviewed the content and recommendations. Results The ASCO endorsement panel determined that the recommendations from the ASTRO guideline, published in 2016, are clear, thorough, and based on current scientific evidence. ASCO endorsed the ASTRO guideline on radiation therapy for glioblastoma and added qualifying statements. Recommendations Partial-brain fractionated radiotherapy with concurrent and adjuvant temozolomide is the standard of care after biopsy or resection of newly diagnosed glioblastoma in patients up to 70 years of age. Hypofractionated radiotherapy for elderly patients with fair to good performance status is appropriate. The addition of concurrent and adjuvant temozolomide to hypofractionated radiotherapy seems to be safe and efficacious without impairing quality of life for elderly patients with good performance status. Reasonable options for patients with poor performance status include hypofractionated radiotherapy alone, temozolomide alone, or best supportive care. Focal reirradiation represents an option for select patients with recurrent glioblastoma, although this is not supported by prospective randomized evidence. Additional information is available at www.asco.org/glioblastoma-radiotherapy-endorsement and www.asco.org/guidelineswiki .
- Published
- 2017
5. First-in-human dose escalation and food effect study of oral ONC206 in adults with recurrent primary CNS neoplasms
- Author
-
Jing Wu, Abhik Ray-Chaudhury, Mark Raffeld, Kenneth Aldape, Joshua E. Allen, Eric Burton, Brett Theeler, Marta Penas-Prado, Kareem A. Zaghloul, Jinkyu Jung, Heather Leeper, Martin Stogniew, Martha Quezado, Ying Yuan, Mark R. Gilbert, Terri Armstrong, and Varun V. Prabhu
- Subjects
Surgical resection ,Oncology ,Cancer Research ,medicine.medical_specialty ,Adjuvant radiotherapy ,business.industry ,Food Effect Study ,First in human ,Systemic therapy ,Cns neoplasms ,Internal medicine ,Dose escalation ,Medicine ,CNS TUMORS ,business - Abstract
TPS2072 Background: The majority of recurrent CNS tumors lack effective systemic therapy options following surgical resection and adjuvant radiotherapy. ONC201, the founding member of the imipridone class of small molecules, has induced durable tumor regressions in patients with diffuse midline glioma, H3 K27M-mutant (DMG H3K27M). ONC206, the second imipridone to enter clinical development, is a DRD2 antagonist and ClpP agonist that exhibits differentiated receptor pharmacology and gene expression profiles in tumors relative to ONC201. The compound is orally bioavailable, penetrates the blood-brain barrier, and exhibits anti-cancer efficacy without toxicity in several preclinical cancer models with pronounced efficacy in myc-overexpressing CNS tumors. Methods: A first-in-human, open label, dose escalation, and food effect Phase I study of oral ONC206 (NCT04541082) is currently enrolling. Patients must be 18 years or older and diagnosed with a recurrent, primary CNS neoplasm. Eligible diseases include recurrent glioblastoma, WHO Grade 2 and 3 infiltrating glial neoplasms, DMG H3K27M, ependymoma, medulloblastoma, malignant meningiomas, and other rare primary CNS neoplasms. Dose escalation, initially with weekly dosing, will follow a standard 3+3 design. After the MTD is established, a food effect cohort will enroll with a balanced, single-dose, two-arm, two-period, crossover design. The primary endpoint is to determine DLT during the first 28-day cycle. Secondary endpoints will include objective response rate by RANO criteria, overall and progression-free survival, and disease control rate. Exploratory biomarker analyses based on preclinical correlations with efficacy will include DRD2, DRD2 dimer, ClpP, DRD5, c-myc, and n-myc expression. Clinical trial information: NCT04541082.
- Published
- 2021
6. A randomized, double-blind, phase II clinical trial of GI-6301 (yeast-brachyury vaccine) versus placebo in combination with standard of care definitive radiotherapy in locally advanced, unresectable, chordoma
- Author
-
Marijo Bilusic, Renee N. Donahue, Jenn Marte, Peter Joseph DeMaria, Seth M. Steinberg, Mark R. Gilbert, Ravi A. Madan, Julius Strauss, James L. Gulley, Jeffrey Schlom, Christopher R. Heery, and Deric M. Park
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Brachyury ,business.industry ,Locally advanced ,medicine.disease ,Placebo ,Metastasis ,Clinical trial ,Double blind ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Neoplasm ,Chordoma ,business ,030215 immunology - Abstract
11527 Background: Chordoma is a rare neoplasm of the notochord that overexpresses brachyury, a transcription factor associated with epithelial-to-mesenchymal transition, metastasis, poor prognosis, and chemotherapy resistance. GI-6301 is a recombinant yeast-brachyury vaccine shown to demonstrate brachyury-specific immunogenicity, excellent safety profile, and some evidence of clinical activity in patients with chordoma in a previous phase I trial. Radiation therapy (RT) can modulate the tumor to become an immunostimulatory milieu. Preclinical studies have shown a synergistic effect combining RT with vaccine, thus prompting this clinical trial evaluating the combination of GI-6301 with RT in chordoma (NCT02383498). Methods: Adults with locally advanced, unresectable chordoma were treated on a randomized, double-blind, placebo controlled, phase 2 clinical trial. Patients received 3 doses of GI-6301 (80 x 107 yeast cells) vs placebo followed by photon or proton RT, followed by GI-6301 vs placebo until disease progression. Primary outcome was overall response rate (ORR) defined as a complete response or partial response in the irradiated tumor site at 24 months. Immune assays were conducted to evaluate immunogenicity. Due to slow accrual, an unplanned interim analysis was undertaken. Results: 24 pts were randomized and treated between May 2015 and September 2019. Vaccine was well tolerated with no dose reductions or treatment discontinuation. Treatment-related serious adverse events included: nausea/emesis (2); fatigue (2); dehydration (2); diarrhea (1); radiation necrosis (1); stroke (1); sepsis (1). There was no difference in ORR between the two arms. Pre-existing brachyury-specific T-cells were detected in the majority of patients but did not correlate with response to therapy. Most patients developed T-cell responses during therapy, regardless of treatment arm. Conclusions: There was no difference in ORR between the two arms. GI-6301 was well tolerated with toxicities related to RT, not vaccine. The trial was stopped early due to low conditional power for finding a statistical difference at the planned end of accrual, based on findings to date. Future studies will define utility of vaccines targeting brachyury in chordoma. Clinical trial information: NCT02383498 . [Table: see text]
- Published
- 2020
7. Phase I/II study of T-DM1 alone versus T-DM1 and metronomic temozolomide in secondary prevention of HER2-positive breast cancer brain metastases following stereotactic radiosurgery
- Author
-
Nicole D. Houston, DeeDee Smart, Patricia S. Steeg, Alexandra S Zimmer, David Lyden, Stanley Lipkowitz, Mark R. Gilbert, Seth M. Steinberg, Terri Armstrong, Brunilde Gril, Nadia Biassou, Eric Burton, Priscilla Kaliopi Brastianos, and Scott L. Carter
- Subjects
Secondary prevention ,Oncology ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,business.industry ,medicine.medical_treatment ,Low dose ,Radiosurgery ,Phase i ii ,HER2 Positive Breast Cancer ,Internal medicine ,medicine ,business ,medicine.drug - Abstract
TPS2572 Background: We demonstrated that low doses of temozolomide (TMZ) administered in a prophylactic, metronomic fashion significantly prevented development of brain metastases in murine models of breast cancer. Based on these findings, we developed a secondary-prevention clinical trial. Methods: Phase I is a standard 3+3 design: T-DM1 3.6mg/kg IV every 21 days plus TMZ 30, 40 or 50 mg/m2 daily, to identify the maximum tolerated dose (MTD) of the combination, and is completing accrual, with 9 patients accrued, currently on the third and last dose level. Phase II will randomize patients to T-DM1 3.6mg/kg versus T-DM1 3.6mg/kg plus TMZ at recommended phase 2 dose (RP2D), to evaluate if addition of TMZ improves the recurrence-free incidence from distant new brain metastases at one year from 50% to 65%. Patients will undergo radiology guided lumbar puncture at baseline and after 6 weeks of treatment (C3D1) for correlative studies, brain MRI, systemic restaging CTs, and questionnaires for evaluation of symptoms and quality of life. Eligibility: HER2+ breast cancer with brain metastases (up to 10 lesions), treated with SRS and/or resection ≤12 weeks before enrollment, no leptomeningeal metastases, no previous WBRT, ECOG ≤2 and adequate organ and marrow function. Biomarkers, including cell free DNA from CSF, serum and tumor block, exosomal DNA, serum markers for neuroinflammation, and patient reported outcomes, will be analyzed in an exploratory fashion. Target accrual: up to 18 patients in phase I and 98 in phase II. Clinical trial information: NCT03190967 .
- Published
- 2020
8. A first-in-human phase I single-agent dose-escalation, food effect and dose expansion study of oral ONC206 in recurrent and rare primary central nervous system neoplasms
- Author
-
Brett Theeler, Kareem A. Zaghloul, Jing Wu, Wolfgang Oster, Mark R. Gilbert, Marta Penas-Prado, Abhik Ray-Chaudhury, Mark Raffeld, Eric Burton, Varun V. Prabhu, Martin Stogniew, Jinkyu Jung, Kenneth Aldape, Joshua E. Allen, Javier A Gonzalez, Edjah K. Nduom, Heather Leeper, Martha Quezado, Ying Yuan, and Terri Armstrong
- Subjects
Cancer Research ,FOOD EFFECT ,business.industry ,Central nervous system ,First in human ,Pharmacology ,Blockade ,medicine.anatomical_structure ,Oncology ,Dopamine receptor D2 ,Dose escalation ,Medicine ,Single agent ,business - Abstract
TPS2576 Background: Blockade of the dopamine receptor D2 (DRD2) has emerged as a therapeutic target in neuro-oncology. ONC201, a first-generation imipridone that antagonizes DRD2, has demonstrated clinical activity in Diffuse Midline Gliomas, H3K27M-mutant (DMGs). Treatment options beyond surgical resection and radiation therapy are limited for most recurrent and rare primary CNS neoplasms. DRD2 blockade holds promise as a therapeutic target in multiple primary CNS cancers. ONC206 is a next generation imipridone and a chemical analog of ONC201 that possesses favorable drug properties such as oral bioavailability, robust stability, and blood-brain barrier penetrance. ONC206 exhibits enhanced allosteric inhibition of DRD2 and nanomolar affinity/potency as a DRD2 antagonist with complete antagonism and specificity for DRD2. ONC206 demonstrated broad spectrum anti-cancer efficacy in vitro across most solid tumor types tested in a panel of > 1000 human cancer cell lines with nervous system tumors emerging as most responsive. In addition to its broad-spectrum activity in vitro, ONC206 efficacy has been demonstrated in xenograft mouse models. Methods: This is an open label, dose escalation, and food effect Phase I study of ONC206at the National Cancer Institute, Neuro-Oncology Branch. Adult patients 18 years and older with recurrent, primary CNS neoplasms will initially be accrued to the dose escalation portion of the study and evaluated for toxicity. Eligible diseases include recurrent glioblastoma, WHO Grade 2 and 3 infiltrating glial neoplasms, and rare primary CNS neoplasms in the NCI-CONNECT program: DMGs, ependymomas, medulloblastomas, and other rare CNS tumor types. The primary endpoint is to determine DLT during the first cycle (28 days). Dose escalation will proceed according to a standard 3+3 design. Both once weekly and more frequent dosing of ONC206 will be explored in the dose escalation scheme. After the MTD is established, food effect will be determined in a dedicated cohort using a balanced, single-dose, two-arm, two-period crossover design. Secondary endpoints will include objective response rate by RANO criteria, overall and progression-free survival, and disease control rate. Exploratory analysis of DRD2 and DRD5 expression, DRD2 dimerization, expression of MYC and N-MYC in tumor tissue in relation to clinical outcomes will also be performed.
- Published
- 2020
9. Phase I trial of TG02 plus dose-dense or metronomic temozolomide for recurrent anaplastic astrocytoma and glioblastoma in adults
- Author
-
Marta Penas-Prado, Tracy Lawhon, Orwa Aboud, Jing Wu, Yu-Ting Su, Christine Bryla, Christine Siegel, Ramya Antony, Terri Armstrong, Ying Yuan, Ann McCoy, Nancy Garren, Brett Theeler, Ewa Grajkowska, Lisa Boris, Christine Cordova, and Mark R. Gilbert
- Subjects
Cancer Research ,Temozolomide ,business.industry ,medicine.disease ,Survival pathways ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,business ,030215 immunology ,Anaplastic astrocytoma ,medicine.drug ,Glioblastoma - Abstract
2031 Background: Therapies targeting multiple survival pathways simultaneously may be more effective for high-grade gliomas, a disease highly resistant to treatment. Our preclinical studies have shown potent anti-glioma effects of TG02 and synergy with temozolomide (TMZ) through modulation of transcription and cellular metabolism. A phase I/II trial was launched to test the combination of TG02 and TMZ in recurrent malignant gliomas and herein we report the phase I results. Methods: Adults with recurrent high-grade astrocytoma, KPS ≥ 60, normal organ function, ≤ 2 prior relapses were enrolled. The primary endpoint was dose limiting toxicity (DLT) from the start of the combined treatment to 4 weeks after in each arm. Bayesian optimal interval (BOIN) design was employed to determine the maximum tolerated dose (MTD) with the target DLT rate of 35% and the toxicity profile of the combination of TG02 (starting dose 200mg orally on days 1, 12, 15, and 26) and TMZ, either as a dose-dense (DD; 125mg/m2/d, 7on/7off, Arm 1) or metronomic (MN; 50mg/m2/d, Arm 2) dosing schedule on a 28-day cycle. Results: Forty patients were enrolled; 38 were evaluable; 70% male; overall median age 50.7; median KPS 90. Of 18 evaluable patients in Arm 1 (DD TMZ), at TG02 dose level 200mg, 1/6 had a DLT: Gr3 diarrhea. At TG02 dose level 250mg, 3/12 had DLTs: Gr4 neutropenia for over 5 days, Gr3 elevated ALT, and Gr3 fatigue. Of 20 evaluable patients in Arm 2 (MN TMZ), at TG02 dose level 200mg, 1/6 had a DLT: recurrent Gr3 neutropenia. At TG02 dose level 250mg, 5/12 had a DLT: Gr3 elevated ALT, Gr3 fatigue, and Gr4 neutropenia. At TG02 dose level 300mg,1 out of 2 had a DLT: Gr4 febrile neutropenia, Gr4 elevated ALT, Gr4 elevated AST, which resulted in hospitalization. Therefore, the TG02 dose level of 250mg was declared as the MTD in both Arm 1 and Arm 2. Conclusions: The combination of TG02 at the MTD of 250mg with DD or MN TMZ has a tolerable toxicity profile. Cohort expansion continues at the MTD in both arms to conduct pharmacokinetics and pharmacogenetics to better elucidate the toxicity profile. Objective responses have been observed, suggesting activity of this regimen and supporting continued investigation with the phase II randomized component. Clinical trial information: NCT02942264.
- Published
- 2019
10. Randomized Trial of Radiation Therapy Plus Procarbazine, Lomustine, and Vincristine Chemotherapy for Supratentorial Adult Low-Grade Glioma: Initial Results of RTOG 9802
- Author
-
Mark R. Gilbert, Paul D. Brown, Minesh P. Mehta, Edward G. Shaw, Meihua Wang, Stephen W. Coons, Jan C. Buckner, Keith J. Stelzer, David Brachman, and Geoffrey R. Barger
- Subjects
Adult ,Oncology ,Cancer Research ,medicine.medical_specialty ,Vincristine ,Time Factors ,Adolescent ,medicine.medical_treatment ,Kaplan-Meier Estimate ,Procarbazine ,Risk Assessment ,Disease-Free Survival ,Young Adult ,Lomustine ,Risk Factors ,Internal medicine ,Glioma ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Aged ,Proportional Hazards Models ,Aged, 80 and over ,Chemotherapy ,Chi-Square Distribution ,business.industry ,Hazard ratio ,Supratentorial Neoplasms ,Chemoradiotherapy, Adjuvant ,Middle Aged ,medicine.disease ,Surgery ,Radiation therapy ,Treatment Outcome ,Multivariate Analysis ,Neoplasm Grading ,business ,Chemoradiotherapy ,medicine.drug - Abstract
Purpose A prior Radiation Therapy Oncology Group (RTOG) clinical trial in anaplastic oligodendroglioma suggested a progression-free survival benefit for procarbazine, lomustine, and vincristine (PCV) chemotherapy in addition to radiation therapy (RT), as have smaller trials in low-grade glioma (LGG). Patients and Methods Eligibility criteria included supratentorial WHO grade 2 LGG, age 18 to 39 years with subtotal resection/biopsy, or age ≥ 40 years with any extent resection. Patients were randomly assigned to RT alone or RT followed by six cycles of PCV. Survival was compared by using the modified Wilcoxon and log-rank tests. Results In all, 251 patients were accrued from 1998 to 2002. Median overall survival (OS) time and 5-year OS rates for RT versus RT + PCV were 7.5 years versus not reached and 63% versus 72%, respectively (hazard ratio [HR]; 0.72; 95% CI, 0.47 to 1.10; P = .33; log-rank P = .13). Median progression-free survival (PFS) time and 5-year PFS rates for RT versus RT + PCV were 4.4 years versus not reached and 46% versus 63%, respectively (HR, 0.6; 95% CI, 0.41 to 0.86; P = .06; log-rank P = .005). OS and PFS were similar for all patients between years 0 and 2. After 2 years, OS and PFS curves separated significantly, favoring RT + PCV. For 2-year survivors (n = 211), the probability of OS for an additional 5 years was 74% with RT + PCV versus 59% with RT alone (HR, 0.52; 95% CI, 0.30 to 0.90; log-rank P = .02). Conclusion PFS but not OS was improved for adult patients with LGG receiving RT + PCV versus RT alone. On post hoc analysis, for 2-year survivors, the addition of PCV to RT conferred a survival advantage, suggesting a delayed benefit for chemotherapy.
- Published
- 2012
11. Immunologic Escape After Prolonged Progression-Free Survival With Epidermal Growth Factor Receptor Variant III Peptide Vaccination in Patients With Newly Diagnosed Glioblastoma
- Author
-
Robert J. Schmittling, James E. Herndon, John H. Sampson, Mark R. Gilbert, Duane Mitchell, Darell D. Bigner, Allan H. Friedman, Weiming Shi, Roger E. McLendon, David A. Reardon, Raymond Sawaya, Henry S. Friedman, James J. Vredenburgh, Amy B. Heimberger, Kenneth Aldape, and Gary E. Archer
- Subjects
Male ,Cancer Research ,Time Factors ,Gene mutation ,medicine.disease_cause ,Multicenter Studies as Topic ,Prospective Studies ,Epidermal growth factor receptor ,DNA Modification Methylases ,Regulation of gene expression ,Mutation ,biology ,Brain Neoplasms ,Middle Aged ,Prognosis ,Immunohistochemistry ,Magnetic Resonance Imaging ,Dacarbazine ,ErbB Receptors ,Europe ,Gene Expression Regulation, Neoplastic ,Oncology ,Chemotherapy, Adjuvant ,DNA methylation ,Female ,medicine.drug ,Adult ,Injections, Intradermal ,Enzyme-Linked Immunosorbent Assay ,Cancer Vaccines ,Disease-Free Survival ,Drug Administration Schedule ,Clinical Trials, Phase II as Topic ,Bias ,Predictive Value of Tests ,Original Reports ,Temozolomide ,medicine ,Humans ,Antineoplastic Agents, Alkylating ,Aged ,business.industry ,Tumor Suppressor Proteins ,DNA Methylation ,Survival Analysis ,United States ,DNA Repair Enzymes ,Sample Size ,Immunology ,biology.protein ,Radiotherapy, Adjuvant ,Glioblastoma ,business - Abstract
Purpose Immunologic targeting of tumor-specific gene mutations may allow precise eradication of neoplastic cells without toxicity. Epidermal growth factor receptor variant III (EGFRvIII) is a constitutively activated and immunogenic mutation not expressed in normal tissues but widely expressed in glioblastoma multiforme (GBM) and other neoplasms. Patients and Methods A phase II, multicenter trial was undertaken to assess the immunogenicity of an EGFRvIII-targeted peptide vaccine and to estimate the progression-free survival (PFS) and overall survival (OS) of vaccinated patients with newly diagnosed EGFRvIII-expressing GBM with minimal residual disease. Intradermal vaccinations were given until toxicity or tumor progression was observed. Sample size was calculated to differentiate between PFS rates of 20% and 40% 6 months after vaccination. Results There were no symptomatic autoimmune reactions. The 6-month PFS rate after vaccination was 67% (95% CI, 40% to 83%) and after diagnosis was 94% (95% CI, 67% to 99%; n = 18). The median OS was 26.0 months (95% CI, 21.0 to 47.7 months). After adjustment for age and Karnofsky performance status, the OS of vaccinated patients was greater than that observed in a control group matched for eligibility criteria, prognostic factors, and temozolomide treatment (hazard ratio, 5.3; P = .0013; n = 17). The development of specific antibody (P = .025) or delayed-type hypersensitivity (P = .03) responses to EGFRvIII had a significant effect on OS. At recurrence, 82% (95% CI, 48% to 97%) of patients had lost EGFRvIII expression (P < .001). Conclusion EGFRvIII-targeted vaccination in patients with GBM warrants investigation in a phase III, randomized trial.
- Published
- 2010
12. NRG BN002: Phase I study of checkpoint inhibitors anti-CTLA-4, anti-PD-1, the combination in patients with newly diagnosed glioblastoma
- Author
-
Minhee Won, Jing Wu, John de Groot, Alfredo Voloschin, Lisa Rogers, Fabio M. Iwamoto, Igor J. Barani, Mark R. Gilbert, Minesh P. Mehta, Raju Raval, Peixin Zhang, Kenneth Aldape, Andrew E. Sloan, and Patrick Y. Wen
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,animal diseases ,Immune checkpoint inhibitors ,chemical and pharmacologic phenomena ,Disease ,Newly diagnosed ,biochemical phenomena, metabolism, and nutrition ,Anti ctla 4 ,medicine.disease ,nervous system diseases ,Phase i study ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Immunity ,030220 oncology & carcinogenesis ,Cancer research ,bacteria ,Medicine ,In patient ,business ,Glioblastoma - Abstract
2053Background: Glioblastoma (GBM) remains an incurable disease that is associated with impaired immunity. Recently, immune checkpoint inhibitors (ICIs) have demonstrated efficacy in several solid ...
- Published
- 2018
13. Phase I factorial study of temozolomide plus memantine, mefloquine, and metformin as post-radiation adjuvant therapy for newly diagnosed glioblastoma
- Author
-
Kenneth Aldape, Vinay K. Puduvalli, Aaron Gerald Mammoser, John de Groot, Barbara O’Brien, Marta Penas-Prado, Kenneth R. Hess, Monica Loghin, Ivo W. Tremont-Lukats, Isaac Melguizo-Gavilanes, Stefania Maraka, Morris D. Groves, W. K. Alfred Yung, Erik P. Sulman, Charles A. Conrad, and Mark R. Gilbert
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Post-radiation ,Temozolomide ,business.industry ,Mefloquine ,Memantine ,medicine.disease ,nervous system diseases ,Metformin ,Internal medicine ,Adjuvant therapy ,Medicine ,business ,Repurposing ,medicine.drug ,Glioblastoma - Abstract
2044Background: Repurposing non-cancer drugs may represent a new source of novel therapies for glioblastoma (GBM). Memantine, mefloquine, and metformin have putative anticancer activity potentially...
- Published
- 2018
14. Polymorphisms of LIG4, BTBD2, HMGA2, and RTEL1 Genes Involved in the Double-Strand Break Repair Pathway Predict Glioblastoma Survival
- Author
-
Sanjay Shete, Richard S. Houlston, Terri S. Armstrong, Spyros Tsavachidis, Kenneth Aldape, Margaret Wrensch, Georgina Armstrong, Mark R. Gilbert, Beatrice Melin, George A. Alexiou, Michael E. Scheurer, Melissa L. Bondy, Yanhong Liu, Ulrika Andersson, John K. Wiencke, Fu Wen Liang, Yuanyuan Xiao, Fay J. Hosking, Carol J. Etzel, and Lindsay B. Robertson
- Subjects
Male ,Cancer Research ,Time Factors ,DNA Ligases ,DNA Repair ,DNA repair ,Kaplan-Meier Estimate ,LIG4 ,Bioinformatics ,Polymorphism, Single Nucleotide ,Risk Assessment ,DNA Ligase ATP ,HMGA2 ,Risk Factors ,Glioma ,Original Reports ,Odds Ratio ,medicine ,Humans ,DNA Breaks, Double-Stranded ,Genetic Predisposition to Disease ,Survivors ,Gene ,Proportional Hazards Models ,Regulation of gene expression ,Chi-Square Distribution ,biology ,Brain Neoplasms ,Proportional hazards model ,Decision Trees ,HMGA2 Protein ,Age Factors ,DNA Helicases ,Middle Aged ,medicine.disease ,Double Strand Break Repair ,Gene Expression Regulation, Neoplastic ,Treatment Outcome ,Oncology ,biology.protein ,Cancer research ,Female ,Carrier Proteins ,Glioblastoma - Abstract
Purpose Glioblastoma (GBM) is the most common and aggressive type of glioma and has the poorest survival. However, a small percentage of patients with GBM survive well beyond the established median. Therefore, identifying the genetic variants that influence this small number of unusually long-term survivors may provide important insight into tumor biology and treatment. Patients and Methods Among 590 patients with primary GBM, we evaluated associations of survival with the 100 top-ranking glioma susceptibility single nucleotide polymorphisms from our previous genome-wide association study using Cox regression models. We also compared differences in genetic variation between short-term survivors (STS; ≤ 12 months) and long-term survivors (LTS; ≥ 36 months), and explored classification and regression tree analysis for survival data. We tested results using two independent series totaling 543 GBMs. Results We identified LIG4 rs7325927 and BTBD2 rs11670188 as predictors of STS in GBM and CCDC26 rs10464870 and rs891835, HMGA2 rs1563834, and RTEL1 rs2297440 as predictors of LTS. Further survival tree analysis revealed that patients ≥ 50 years old with LIG4 rs7325927 (V) had the worst survival (median survival time, 1.2 years) and exhibited the highest risk of death (hazard ratio, 17.53; 95% CI, 4.27 to 71.97) compared with younger patients with combined RTEL1 rs2297440 (V) and HMGA2 rs1563834 (V) genotypes (median survival time, 7.8 years). Conclusion Polymorphisms in the LIG4, BTBD2, HMGA2, and RTEL1 genes, which are involved in the double-strand break repair pathway, are associated with GBM survival.
- Published
- 2010
15. Correlation of O6-Methylguanine Methyltransferase (MGMT) Promoter Methylation With Clinical Outcomes in Glioblastoma and Clinical Strategies to Modulate MGMT Activity
- Author
-
Monika E. Hegi, Wolfgang Wick, Minesh P. Mehta, Roger Stupp, Michael Weller, Lili Liu, James G. Herman, and Mark R. Gilbert
- Subjects
Cancer Research ,Methyltransferase ,Maximum Tolerated Dose ,DNA repair ,Medical Oncology ,O(6)-Methylguanine-DNA Methyltransferase ,Glioma ,Humans ,Medicine ,Gene silencing ,Gene Silencing ,Promoter Regions, Genetic ,Antineoplastic Agents, Alkylating ,neoplasms ,Temozolomide ,Brain Neoplasms ,business.industry ,Cancer ,Methylation ,DNA Methylation ,medicine.disease ,digestive system diseases ,Treatment Outcome ,Models, Chemical ,Oncology ,Immunology ,DNA methylation ,Cancer research ,CpG Islands ,RNA Interference ,Glioblastoma ,business ,medicine.drug - Abstract
Resistance to alkylating agents via direct DNA repair by O6-methylguanine methyltransferase (MGMT) remains a significant barrier to the successful treatment of patients with malignant glioma. The relative expression of MGMT in the tumor may determine response to alkylating agents, and epigenetic silencing of the MGMT gene by promoter methylation plays an important role in regulating MGMT expression in gliomas. MGMT promoter methylation is correlated with improved progression-free and overall survival in patients treated with alkylating agents. Strategies to overcome MGMT-mediated chemoresistance are being actively investigated. These include treatment with nontoxic pseudosubstrate inhibitors of MGMT, such as O6-benzylguanine, or RNA interference-mediated gene silencing of MGMT. However, systemic application of MGMT inhibitors is limited by an increase in hematologic toxicity. Another strategy is to deplete MGMT activity in tumor tissue using a dose-dense temozolomide schedule. These alternative schedules are well tolerated; however, it remains unclear whether they are more effective than the standard dosing regimen or whether they effectively deplete MGMT activity in tumor tissue. Of note, not all patients with glioblastoma having MGMT promoter methylation respond to alkylating agents, and even those who respond will inevitably experience relapse. Herein we review the data supporting MGMT as a major mechanism of chemotherapy resistance in malignant gliomas and describe ongoing studies that are testing resistance-modulating strategies.
- Published
- 2008
16. Phase II Trial of Tipifarnib in Patients With Recurrent Malignant Glioma Either Receiving or Not Receiving Enzyme-Inducing Antiepileptic Drugs: A North American Brain Tumor Consortium Study
- Author
-
Susan M. Chang, Lauren E. Abrey, Jeffrey J. Raizer, Mark R. Gilbert, Frank S. Lieberman, Minesh P. Mehta, Morris D. Groves, John G. Kuhn, Kathleen R. Lamborn, Michael D. Prados, Lisa M. DeAngelis, David Schiff, Patrick Y. Wen, Larry Junck, W. K. Alfred Yung, Karen Fink, H. Ian Robins, Kenneth Aldape, John Tim Wright, and Timothy F. Cloughesy
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Population ,Phases of clinical research ,Antineoplastic Agents ,Quinolones ,Disease-Free Survival ,Drug Administration Schedule ,Internal medicine ,Glioma ,medicine ,Humans ,education ,Aged ,education.field_of_study ,Epilepsy ,Brain Neoplasms ,business.industry ,Incidence (epidemiology) ,Middle Aged ,medicine.disease ,Rash ,Surgery ,Clinical trial ,Treatment Outcome ,Enzyme Induction ,North America ,Toxicity ,Anticonvulsants ,Female ,Tipifarnib ,Neoplasm Recurrence, Local ,medicine.symptom ,Glioblastoma ,business ,medicine.drug - Abstract
Purpose A phase II study was undertaken in patients with recurrent malignant glioma to determine the efficacy and safety of tipifarnib, a farnesyltransferase inhibitor, dosed at the respective maximum-tolerated dose (MTD) for patients receiving and not receiving enzyme-inducing antiepileptic drugs (EIAEDs). Because tipifarnib undergoes extensive hepatic metabolism, MTD is doubled in patients on EIAEDs. The population included 67 patients with glioblastoma multiforme (GBM) and an exploratory group of 22 patients with anaplastic glioma (AG). Patients and Methods Patients received tipifarnib (300 and 600 mg bid for 21 days every 4 weeks in non-EIAED and EIAED patients, respectively). All patients were assessable for efficacy and safety. Results Two AG patients (9.1%) and eight GBM patients (11.9%) had progression-free survival (PFS) more than 6 months. Among the latter eight GBM patients, six of 36 patients (16.7%; 95% CI, 7% to 32%) were not receiving EIAEDs and two of 31 patients (6.5%; 95% CI, 1% to 20%) were receiving EIAEDs. Four patients had partial responses in group A GBM and one patient had a partial response group B GBM. An exploratory comparison of PFS between GBM groups A and B was statistically significant (P = .01). Patients not receiving EIAEDs had a higher incidence and increased severity of hematologic events. However, the incidence and severity of rash (the previously determined dose-limiting toxicity in patients receiving EIAEDs) seemed similar in EIAED and non-EIAED subgroups. Conclusion Tipifarnib (300 mg bid for 21 days every 4 weeks) shows modest evidence of activity in patients with recurrent GBM who are not receiving EIAEDs and is generally well tolerated in this population.
- Published
- 2006
17. Diffusely Infiltrative Low-Grade Gliomas in Adults
- Author
-
Frederick F. Lang and Mark R. Gilbert
- Subjects
Adult ,Cancer Research ,medicine.medical_specialty ,Pathology ,medicine.diagnostic_test ,business.industry ,Optimal treatment ,Evidenced based ,Neurooncology ,Magnetic resonance imaging ,Glioma ,Prognosis ,medicine.disease ,law.invention ,Central Nervous System Neoplasms ,Clinical trial ,Oncology ,Randomized controlled trial ,law ,medicine ,Humans ,Radiology ,business ,Diffusely infiltrative - Abstract
Diffusely infiltrating low-grade gliomas (LGGs) include astrocytomas, oligodendrogliomas, and mixed oligoastrocytomas (WHO grade 2). Due to the routine use of magnetic resonance imaging, there is an increasing need to formulate treatment guidelines for patients with LGGs. However, there is little consensus about the optimal treatment strategy for diffusely infiltrative LGGs, and the clinical management of LGGs is one of the most controversial areas in neurooncology. Although the standard of care has not been established, several randomized trials are beginning to provide some answers. Furthermore, laboratory correlative studies are defining subsets of LGG that may identify patients with better prognoses and increased chance of responding to therapy. This article reviews the most recent data regarding the treatment of LGG, emphasizing evidenced based approaches from current clinical trials.
- Published
- 2006
18. Phase II Trial of Radiosurgery for One to Three Newly Diagnosed Brain Metastases From Renal Cell Carcinoma, Melanoma, and Sarcoma: An Eastern Cooperative Oncology Group Study (E 6397)
- Author
-
Hillard M. Lazarus, Henry N. Wagner, Rafael Manon, Jonathan P.S. Knisely, Anne O'Neill, Minesh P. Mehta, Maria Werner-Wasik, and Mark R. Gilbert
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Time Factors ,medicine.medical_treatment ,Radiosurgery ,Cognition ,Renal cell carcinoma ,Internal medicine ,medicine ,Carcinoma ,Humans ,Prospective Studies ,Carcinoma, Renal Cell ,Melanoma ,Survival rate ,Survival analysis ,Neoplasm Staging ,Performance status ,Brain Neoplasms ,business.industry ,Brain ,Sarcoma ,medicine.disease ,Survival Analysis ,Kidney Neoplasms ,Survival Rate ,Radiation therapy ,Treatment Outcome ,Female ,Neoplasm Recurrence, Local ,business ,Follow-Up Studies - Abstract
Purpose Long-term brain metastases survivors are at risk for neurologic morbidity after whole-brain radiotherapy (WBRT). Retrospective radiosurgery (RS) reports found no survival difference when compared with WBRT. Before RS alone was evaluated with delayed WBRT in a phase III trial, the feasibility of RS alone was tested prospectively. Patients and Methods Patients with renal cell carcinoma, melanoma, or sarcoma; one to three brain metastases; and performance status of 0 to 2 were enrolled. Exclusion criteria were leptomeningeal disease; metastases in medulla, pons, or midbrain; or liver metastases. On the basis of tumor size, patients received 24, 18, or 15 Gy RS. At recurrence, management was discretionary. The primary end point was 3- and 6-month intracranial progression. Results Between July 1998 and August 2003, 36 patients were accrued; 31 were eligible. Median follow-up was 32.7 months and the median survival was 8.3 months (95% CI, 7.4 to 12.2). Three- and 6-month intracranial failure with RS alone was 25.8% and 48.3%. Failure within and outside the RS volume, when in-field and distant intracranial failures were scored independently, was 19.3% and 16.2% (3 months) and 32.2% and 32.2% (6 months), respectively. Approximately 38% of patients experienced death attributable to neurologic cause. There were three grade 3 toxicities related to RS. Conclusion Intracranial failure rates without WBRT were 25.8% and 48.3% at 3 and 6 months, respectively. Delaying WBRT may be appropriate for some subgroups of patients with radioresistant tumors, but routine avoidance of WBRT should be approached judiciously.
- Published
- 2005
19. Reply to F. Felix et al and M.F. Fay et al
- Author
-
Timothy F. Cloughesy, James Perry, Wolfgang Wick, O. L. Chinot, David A. Reardon, Roger Stupp, Monika E. Hegi, Patrick Roth, L. Burt Nabors, Minesh P. Mehta, Stephanie L. Pugh, Thierry Gorlia, Mark R. Gilbert, Michael Weller, Caroline Happold, and University of Zurich
- Subjects
Cancer Research ,business.industry ,MEDLINE ,610 Medicine & health ,10040 Clinic for Neurology ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,10032 Clinic for Oncology and Hematology ,Medicine ,2730 Oncology ,1306 Cancer Research ,business ,Humanities ,030217 neurology & neurosurgery - Published
- 2016
20. Polymorphisms risk modeling for vascular toxicity in patients with glioblastoma treated on NRG Oncology/RTOG 0825
- Author
-
Erik P. Sulman, Ying Yuan, Renke Zhou, D. Brachman, Stephanie L. Pugh, Steven Wilson, Kevin Judy, H. Ian Robins, Yanhong Liu, Terri S. Armstrong, Mark R. Gilbert, Michael E. Scheurer, Elizabeth Vera, Merideth M Wendland, G.K. Hunter, James D. Bearden, Susan L. McGovern, and Melissa L. Bondy
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Bevacizumab ,business.industry ,Vascular toxicity ,medicine.disease ,Thrombosis ,Internal medicine ,Toxicity ,medicine ,In patient ,business ,medicine.drug ,Glioblastoma - Abstract
2049Background: Vascular toxicities, including hypertension and thrombosis, are commonly associated with anti-angiogenic agents, such as Bevacizumab. Risk factors for toxicity have yet to be identi...
- Published
- 2016
21. An independently validated nomogram for individualized estimation of survival among patients with newly diagnosed glioblastoma: NRG oncology/RTOG 0525 and 0825
- Author
-
Andrew E Sloan, Erik P. Sulman, Minesh P. Mehta, Mitchell Machtay, Deborah T. Blumenthal, Haley Gittleman, Daniel Lim, Michael W. Kattan, Michael A. Vogelbaum, Mark R. Gilbert, Peixin Zhang, Emad Youssef, Andrew B. Lassman, Simon S. Lo, Jill S. Barnholtz-Sloan, Tony J. C. Wang, Devin Tian, Marta Penas-Prado, and Arnab Chakravarti
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Karnofsky Performance Status ,Proportional hazards model ,business.industry ,medicine.medical_treatment ,Recursive partitioning ,Newly diagnosed ,Nomogram ,medicine.disease ,Clinical trial ,Radiation therapy ,Internal medicine ,medicine ,business ,Glioblastoma - Abstract
2007Background: Glioblastoma (GBM) is the most common primary malignant brain tumor. Nomograms are often used for individualized estimation of prognosis. This approach has been applied to GBM only once before. This study aimed to build and independently validate a nomogram to estimate individualized survival probabilities for patients with newly diagnosed GBM, using data from two independent NRG Oncology/Radiation Therapy Oncology Group (RTOG) clinical trials. Methods: This analysis included information on 799 (0525) and 555 (0825) eligible and randomized patients with newly diagnosed GBM and contained the following variables: age at diagnosis, race, gender, Karnofsky Performance Status (KPS), extent of resection, O6-methylguanine-DNA methyltransferase (MGMT) methylation status, and survival (months). Survival was assessed using Cox proportional hazards (CPH) regression, random survival forests (RSF), and recursive partitioning analysis (RPA), with adjustment for known prognostic factors. The models were ...
- Published
- 2016
22. Investigation of risk factors associated with fatigue in glioma patients
- Author
-
Anita Mahajan, Jeffrey S. Wefel, Alvina A. Acquaye, Renke Zhou, Ann M. Berger, Ghislain Breton, Catherine M. Sullaway, Elizabeth Vera, Michael E. Scheurer, Mark R. Gilbert, and Terri S. Armstrong
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Clinical variables ,business.industry ,Disease trajectory ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,Glioma ,medicine ,Physical therapy ,business ,030215 immunology - Abstract
2018Background: Fatigue is a consistently reported severe symptom among patients with gliomas throughout the disease trajectory. Clinical variables and genomic pathways associated with fatigue in g...
- Published
- 2016
23. Sequences of taxol and cisplatin: a phase I and pharmacologic study
- Author
-
B G Lubejko, Eric K. Rowinsky, Louise B. Grochow, David S. Ettinger, Arlene A. Forastiere, Dennis A. Noe, B Clark, William P. McGuire, S E Sartorius, and Mark R. Gilbert
- Subjects
Adult ,Male ,Cancer Research ,Neutropenia ,Paclitaxel ,medicine.medical_treatment ,Pharmacology ,Microtubules ,Asymptomatic ,Drug Administration Schedule ,chemistry.chemical_compound ,Alkaloids ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Adverse effect ,Aged ,Cisplatin ,Chemotherapy ,business.industry ,Peripheral Nervous System Diseases ,Arrhythmias, Cardiac ,Middle Aged ,medicine.disease ,Antineoplastic Agents, Phytogenic ,Oncology ,chemistry ,Absolute neutrophil count ,Vomiting ,Drug Evaluation ,Female ,medicine.symptom ,business ,medicine.drug - Abstract
Untreated and minimally pretreated solid tumor patients received alternating sequences of taxol and cisplatin. Sequential dose escalation of each agent using taxol doses of 110 or 135 mg/m2 and cisplatin doses of 50 or 75 mg/m2 resulted in four dosage permutations that induced grades 3 and 4 neutropenia in 72% to 84% and 50% to 53% of courses, respectively. Neutropenia was brief, and hospitalization for neutropenia and fever was required in 13% to 24% of courses. However, further escalation of taxol to 170 or 200 mg/m2 induced grade 4 neutropenia in 79% to 82% of courses. At the highest taxol-cisplatin dose level (200 mg/m2 to 75 mg/m2), the mean neutrophil count nadir was 98/microL, and hospitalization for neutropenia and fever was required in 64% of courses. The sequence of cisplatin before taxol, which has less antitumor activity in vitro, induced more profound neutropenia than the alternate sequence. Pharmacologic studies indicated that this difference was probably due to 25% lower taxol clearance rates when cisplatin preceded taxol. Although neurotoxicity was initially thought to be a potentially serious effect of the combination, mild to modest neurotoxicity occurred in only 27% of patients. Adverse effects also included myalgias, alopecia, vomiting, diarrhea, bradycardia, and asymptomatic ventricular tachycardia. Objective responses were noted in melanoma, as well as non-small-cell lung, ovarian, breast, head and neck, colon, and pancreatic carcinomas. Based on these results, the sequence of taxol before cisplatin at doses of 135 and 75 mg/m2, respectively, is recommended for phase II/III trials, with escalation of taxol to 170 mg/m2 if treatment is well tolerated.
- Published
- 1991
24. Results of NRG oncology/RTOG 9813: A phase III randomized study of radiation therapy (RT) and temozolomide (TMZ) versus RT and nitrosourea (NU) therapy for anaplastic astrocytoma (AA)
- Author
-
David Schiff, Kenneth Aldape, David Brachman, Kurt A. Jaeckle, Susan M. Chang, Helen A. Shih, Christopher J. Schultz, Carol A. Dolinskas, Jean-Paul Bahary, J. Gregory Cairncross, Paul D. Brown, Minesh P. Mehta, Geoffrey R. Barger, Mark R. Gilbert, H. Ian Robins, Peixin Zhang, Marta Penas-Prado, Arnab Chakravarti, Karl Belanger, and Maria Werner-Wasik
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Nitrosourea ,Temozolomide ,business.industry ,medicine.medical_treatment ,medicine.disease ,Anaplastic glioma ,nervous system diseases ,law.invention ,Radiation therapy ,chemistry.chemical_compound ,Randomized controlled trial ,chemistry ,law ,Internal medicine ,medicine ,Overall survival ,business ,neoplasms ,medicine.drug ,Anaplastic astrocytoma ,Glioblastoma - Abstract
2002 Background: TMZ is efficacious in recurrent anaplastic glioma and newly diagnosed glioblastoma. The primary objective was to compare the overall survival (OS) of patients with AA treated with ...
- Published
- 2015
25. Phase I study of vorinostat combined with isotretinoin and temozolomide in adults with recurrent malignant gliomas
- Author
-
Kathy Hunter, Victor A. Levin, Charles A. Conrad, Vinay K. Puduvalli, Monica Loghin, John de Groot, W. K. Alfred Yung, Marta Penas-Prado, Howard Colman, Kenneth R. Hess, Mark R. Gilbert, and Morris D. Groves
- Subjects
Cancer Research ,Temozolomide ,business.industry ,medicine.drug_class ,fungi ,Histone deacetylase inhibitor ,food and beverages ,Pharmacology ,Phase i study ,Oncology ,medicine ,Cancer research ,business ,Vorinostat ,Isotretinoin ,medicine.drug - Abstract
2039 Background: Vorinostat (Vor), a histone deacetylase inhibitor, has shown preliminary activity in recurrent GBM. Preclinical studies demonstrated that Vor can overcome resistance to isotretinoi...
- Published
- 2015
26. A phase I trial everolimus and sorafenib in patients with recurrent high-grade gliomas: Brain Tumor Treatment Collaborative trial 09-01
- Author
-
Monica Loghin, Ivo W. Tremont-Lukats, Jeffrey Raizer, W. K. Alfred Yung, Sean Grimm, Mark R. Gilbert, Tobias Walbert, Nicholas Avgeropoulos, and Marta Penas-Prado
- Subjects
Sorafenib ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Everolimus ,Bevacizumab ,business.industry ,Brain tumor ,Treatment options ,medicine.disease ,Internal medicine ,medicine ,In patient ,business ,medicine.drug - Abstract
2061 Background: Limited treatment options exist for patients with recurrent Malignant Gliomas (MG), especially after failing Bevacizumab (BEV). Signal transduction pathways involved in gliomagenes...
- Published
- 2015
27. A randomized phase II trial of standard dose bevacizumab versus low dose bevacizumab plus lomustine (CCNU) in adults with recurrent glioblastoma
- Author
-
Diane D. Liu, Marta Penas-Prado, Stan Xiaosi Han, Barbara O’Brien, Shiao-Pei Weathers, Ivo W. Tremont-Lukats, Charles A. Conrad, Monica Loghin, W. K. Alfred Yung, John de Groot, Mark R. Gilbert, and Vinay K. Puduvalli
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Bevacizumab ,business.industry ,Recurrent glioblastoma ,Low dose ,Antiangiogenic therapy ,food and beverages ,Vascular permeability ,Lomustine ,Internal medicine ,medicine ,High doses ,Nuclear medicine ,business ,medicine.drug - Abstract
2005 Background: Antiangiogenic therapy can rapidly reduce vascular permeability, but high doses of bevacizumab (BEV) may induce selective pressure to promote resistance. This trial evaluated the e...
- Published
- 2015
28. Brain Tumor Trials Collaborative Bayesian Adaptive Randomized Phase II trial of bevacizumab plus vorinostat versus bevacizumab alone in adults with recurrent glioblastoma (BTTC-1102)
- Author
-
Fabio M. Iwamoto, Jing Wu, Mark R. Gilbert, Terri S. Armstrong, W. K. Alfred Yung, Tobias Walbert, Marc C. Chamberlain, Morris D. Groves, David M. Peereboom, Pierre Giglio, Ryan Merrell, Nina Paleologos, Nicholas Avgeropoulos, Karen Fink, Vinay K. Puduvalli, Ying Yuan, Jeffrey Raizer, and Howard Colman
- Subjects
Response rate (survey) ,Oncology ,Cancer Research ,medicine.medical_specialty ,Bevacizumab ,business.industry ,Recurrent glioblastoma ,Brain tumor ,food and beverages ,medicine.disease ,Antiangiogenic agents ,Internal medicine ,medicine ,Adaptive resistance ,business ,Vorinostat ,medicine.drug - Abstract
2012 Background: Bevacizumab (Bev) is approved in the US for patients (pts) with recurrent glioblastoma based on response rate. However, adaptive resistance to antiangiogenic agents can result in t...
- Published
- 2015
29. Minding the Ps and Qs: Perseverance and Quality Studies Lead to Major Advances in Patients With Anaplastic Oligodendroglioma
- Author
-
Mark R. Gilbert
- Subjects
Oncology ,Clinical Trials as Topic ,Cancer Research ,medicine.medical_specialty ,Vincristine ,Chemotherapy ,Temozolomide ,Brain Neoplasms ,business.industry ,medicine.medical_treatment ,Oligodendroglioma ,Lomustine ,Procarbazine ,Combined Modality Therapy ,Surgery ,Clinical trial ,Radiation therapy ,Regimen ,Research Design ,Internal medicine ,medicine ,Humans ,business ,medicine.drug - Abstract
Almost 6 years ago, the results of two similar studies that described the outcomes for patients with anaplastic oligodendroglial (AO) tumors were published in Journal of Clinical Oncology. These studies were launched in 1994 and 1995 and built on the observations from the late 1980s and early 1990s that some patients with AO had dramatic and durable responses to chemotherapy. The Radiation Therapy Oncology Group (RTOG), in collaboration with four other North American–based cooperative groups, launched the RTOG 9402 trial. This phase III clinical trial compared the standard of care, external beam radiation, to preradiation chemotherapy using up to four cycles of intensive procarbazine, lomustine (CCNU), and vincristine (the iPCV regimen). Within 1 year of the initiation of the RTOG trial, the European Organisation for Research and Treatment of Cancer (EORTC) launched a study of patients with the same tumor type, but the experimental arm differed in that radiation was followed by up to six cycles of standard-dose PCV chemotherapy. When the results of these two trials were published in 2006, they both demonstrated a significant improvement in progression-free survival for patients who received the combination regimens, but there was no difference in overall survival. However, in both studies, approximately 80% of the patients who were randomly assigned to the radiation-only arms were treated with chemotherapy at tumor relapse (in contrast, only 50% of patients who were given up-front chemotherapy received chemotherapy salvage treatment). The conclusions in 2006 were that either treatment approach was acceptable, given that the patients who received only radiation as first-line treatment would then benefit from chemotherapy as second-line treatment. The editorial that accompanied these publications highlighted the need for patient outcomes measures such as neurocognitive and symptom burden consequences that might help determine whether early or late chemotherapy should be the standard of care. In other words, are the early toxicities from the chemotherapy offset by the preservation of function that results from the delay in tumor progression? These trials did not directly address these issues. However, both studies attempted to prospectively collect tumor tissues, although at the time that the studies were launched, no specific molecular markers were known. In 1998, the association of allelic loss of the short arm (p) of chromosome 1 and the long arm (q) of chromosome 19 with some AOs was published. Retrospective series strongly suggested that these changes were highly prognostic; subsequently, the status of 1p/19q was determined in the available tumor samples from both the RTOG and EORTC studies. The 2006 publications analyzed the survival outcomes, stratifying by the 1p/19q loss, and confirmed the prognostic importance of these chromosome changes, but survival was not improved by early chemotherapy in either the patients with 1p/19q loss or those without. However, in 2006, for the group with 1p/19q loss, the median survival had not been reached. As evidenced by the two articles that accompany this editorial, 6 years later the story has dramatically changed. Additional tumor samples were obtained from the original study participants and long-term follow-up data were obtained for both studies, now more than 17 years after their initiation. Remarkably, once again the two trials were complementary. Now with this long-term follow-up, there is clear evidence that for patients with AOs that contain the 1p/19q loss, early chemotherapy with radiation offers a significant improvement in overall survival compared with early radiation even with salvage chemotherapy at tumor relapse. This survival benefit was not found for patients with tumors that did not have the chromosomal deletions, highlighting the clinical importance of this molecular marker. These results have several important implications. These studies establish a new standard of care for patients with AO tumors that harbor the 1p/19q loss. No longer is radiation considered an adequate treatment for this patient population. As a result, the international cooperative group study of newly diagnosed AO with 1p/19q loss, CODEL, which compared radiation therapy with the combination of radiation and chemotherapy (temozolomide) and included an exploratory arm with chemotherapy (temozolomide alone), was halted because the control arm (radiation alone) is no longer viable. There are now ongoing discussions about a new trial design, with considerations ranging from comparing the PCV combination (used in these clinical trials) with temozolomide, an oral agent with a better toxicity profile and proven efficacy in glioblastoma. Further complicating the study design are the reports of tumor response with chemotherapy (PCV or temozolomide) alone, generating the question of whether to evaluate chemotherapy alone as first-line therapy, reserving a radiation-containing regimen for relapse. JOURNAL OF CLINICAL ONCOLOGY E D I T O R I A L VOLUME 31 NUMBER 3 JANUARY 2
- Published
- 2013
30. Venous thromboembolism (VTE) and glioblastoma
- Author
-
Jimin Wu, Courtney C. Webre, Jacob Mandel, Shlomit Yust-Katz, Tushar Pawar, Ying Yuan, Terri S. Armstrong, Mark R. Gilbert, and Harshad Ladha
- Subjects
Cancer Research ,medicine.medical_specialty ,Oncology ,business.industry ,Internal medicine ,medicine ,cardiovascular diseases ,Intensive care medicine ,medicine.disease ,business ,Venous thromboembolism ,Glioblastoma - Abstract
2033 Background: The risk of VTE is very high for patients with brain tumors, with Glioblastoma (GB) considered one of the most at risk cancers. The aim of this study is to describe the occurrence ...
- Published
- 2014
31. Phase III study of radiation therapy (RT) with or without procarbazine, CCNU, and vincristine (PCV) in low-grade glioma: RTOG 9802 with Alliance, ECOG, and SWOG
- Author
-
Christopher J. Schultz, Harold Kim, Keith J. Stelzer, Barbara Fisher, Stephen W. Coons, Paul D. Brown, Minesh P. Mehta, Walter J. Curran, Albert Murtha, Edward G. Shaw, John H. Suh, Mark R. Gilbert, Stephanie L. Pugh, David Brachman, Jan C. Buckner, Geoffrey R. Barger, Jean-Paul Bahary, Dennis E. Bullard, and Peter Ricci
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Vincristine ,business.industry ,medicine.medical_treatment ,Procarbazine ,Radiation therapy ,Early results ,Internal medicine ,medicine ,Low-Grade Glioma ,business ,Nuclear medicine ,Clin oncol ,medicine.drug - Abstract
2000 Background: Early results of R9802 (Shaw et al: J Clin Oncol. 2012; 30(25):3065-70) demonstrated that PCV given with RT at the time of initial diagnosis prolongs progression-free survival (PFS...
- Published
- 2014
32. Anaplastic Oligodendroglial Tumors: A Tale of Two Trials
- Author
-
Frederick F. Lang and Mark R. Gilbert
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,business.industry ,medicine.medical_treatment ,Lomustine ,Procarbazine ,medicine.disease ,Radiation therapy ,Internal medicine ,PCV Regimen ,medicine ,Oligodendroglial Tumor ,Oligodendroglioma ,business ,medicine.drug ,Anaplastic astrocytoma - Abstract
The treatment of patients with anaplastic gliomas with an oligodendroglial component, either pure oligodendroglioma or mixed oligoastrocytoma, remains controversial. In the early 1990s, it was recognized that some patients with these tumors demonstrated dramatic responses to systemic chemotherapy regimens, particularly the combination of procarbazine, lomustine, and vincristine (PCV), whereas patients with other types of glial malignancies within the same WHO grade 3 classification (ie, anaplastic astrocytoma) showed only modest and often short-lived treatment responses. This observation prompted two well-designed and complementary phase III trials for anaplastic oligodendroglial tumors, which are both reported in this issue. The Radiation Therapy Oncology Group (RTOG), in collaboration with four other North American– based cooperative groups, initiated RTOG 9402. Patients with histologically confirmed anaplastic oligodendroglioma or anaplastic mixed oligoastrocytoma were enrolled onto this clinical trial and were randomly assigned to receive either conventional external-beam radiation therapy or up to four cycles of the PCV combination chemotherapy regimen followed by external-beam radiation. Within 1 year of the initiation of the RTOG trial, the European Organisation for Research and Treatment of Cancer launched a study that randomly assigned patients with the same anaplastic oligodendroglial tumors to receive either external-beam radiation alone or external-beam radiation followed by six cycles of the combination PCV regimen. Both studies required central pathology review to standardize and confirm the histologic diagnosis. In addition, each study requested tumor tissue specimens be provided for future correlative studies. Both studies were successful in obtaining tissue samples from the majority of the enrolled patients, which proved to have a critical impact on the results from both trials. Remarkably, both studies showed nearly identical results. They demonstrated no overall survival benefit when chemotherapy was added either before or after radiation in the initial treatment of anaplastic oligodendroglial tumors, despite the established chemotherapy sensitivity of these neoplasms. However, in both studies, the vast majority of patients who were randomly assigned to the radiationonly arm did receive chemotherapy off protocol at the time of tumor relapse. Therefore, a more accurate conclusion is that no survival benefit was noted with early chemotherapy compared with salvage regimens administered at the time of relapse or recurrence. However, both studies did demonstrate a difference in the time to progression, which strongly favored the group receiving treatment with both chemotherapy and radiation. Does this mean that early chemotherapy is the better treatment approach? To answer this question, additional information is required that is currently not available. Specifically, these studies do not provide information regarding the impact of tumor progression on neurologic function and quality of life. If early chemotherapy, with the resultant prolongation of tumor control, delays neurologic worsening, then increasing time to progression has important clinical implications. However, the PCV regimen used by both studies was associated with a high rate of toxicity, particularly myelosuppression. Although we do not know the direct impact of chemotherapy-associated toxicities on neurologic function or quality of life, at least one patient had a treatmentassociated death. Therefore, an analysis of the risk to benefit of the two approaches, early chemotherapy versus treatment at relapse, requires assessment of the functional status of the patients over the course of each trial. Without this analysis, it is difficult to decide whether delaying progression of disease with early chemotherapy is of any clinical benefit when the attendant risk of toxicity is considered. In this context, recent studies suggest that single-agent temozolomide may have similar activity as PCV in anaplastic oligodendroglial tumors but with a better toxicity profile. Therefore, the use of temozolomide would potentially alter the risk-to-benefit analysis of the early versus late use of chemotherapy. However, to date, a randomized clinical trial has not been reported that examines the efficacy of temozolomide plus radiation compared with radiation alone in this patient population. The other critical component of these phase III trials is that they provide prospective validation of the predictive value of the allelic loss of heterozygosity (LOH) of the 1p and 19q chromosomes for oligodendroglial tumors. Previous reports in the literature examining this issue were either small trials or retrospective series that, although quite compelling, did not confirm the importance of this molecular finding. In both studies, patients with loss of 1p and 19q survived significantly longer than patients without the chromosomal changes. However, despite the reports of the association of 1p/19q LOH with response to chemotherapy, in these trials, the early addition of chemotherapy did not improve survival, although there was a significant impact on progressionfree survival. This indicates that 1p/19q LOH is a marker for good prognosis and suggests that it may also predict response to treatment, either radiation or chemotherapy. In fact, this molecular profile with its profound association with outcome has stimulated JOURNAL OF CLINICAL ONCOLOGY E D I T O R I A L VOLUME 24 NUMBER 18 JUNE 2
- Published
- 2006
33. Neurocognitive function (NCF) outcomes in patients with glioblastoma (GBM) enrolled in RTOG 0825
- Author
-
Jeffrey S. Wefel, Stephanie L. Pugh, Ian R. Crocker, H. Ian Robins, David Brachman, Minesh P. Mehta, R. Jeffrey Lee, Ritsuko Komaki, Merideth M Wendland, Mark R. Gilbert, Terri S. Armstrong, and Minhee Won
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Newly diagnosed ,medicine.disease ,Surgery ,Internal medicine ,Overall survival ,medicine ,In patient ,business ,Neurocognitive ,Glioblastoma - Abstract
2004 Background: RTOG 0825 evaluated overall survival (OS) and progression-free survival (PFS) differences in patients with newly diagnosed GBM treated with standard chemoradiation, maintenance temozolomide and placebo (Arm 1) or bevacizumab (Arm 2). While OS was equivalent, PFS was longer in Arm 2. Longitudinal NCF testing was performed to evaluate clinical benefit. Methods: NCF was evaluatedat baseline and while on study and progression free with the Hopkins Verbal Learning Test-Revised (HVLT-R), Trail Making Test (TMT) and Controlled Oral Word Association (COWA). Change in NCF from baseline was categorized using the reliable change index. Differences between treatment arms were compared at follow-up evaluations. Additionally, baseline (B) and early changes (EC) (B to week 10) in NCF were examined as prognostic factors. Results: 542 patients consented and 507 randomized patients participated, with test completion rates at weeks 0 (B), 10, 22, and 34 of 94-97, 69-73, 59-64, and 53-58%, respectively. Mean test performance at B was equivalent between arms and ranged from -0.8 to -4.8 SDs below healthy population norms with global NCF on a composite variable at -2.0 SDs. There were no statistically significant between arm differences in frequency of improvement through week 34. Decline on COWA (verbal measure of executive function) at week 34 relative to baseline was more common (16.1 vs 5.7%) in patients in Arm 1 (p
- Published
- 2013
34. Survival outcome of early versus delayed bevacizumab treatment in patients with recurrent glioblastoma
- Author
-
W. K. Alfred Yung, Vinay K. Puduvalli, John de Groot, Jacob Mandel, Mark R. Gilbert, Charles A. Conrad, and Mohamed Ali Hamza
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Bevacizumab ,business.industry ,Recurrent glioblastoma ,Survival outcome ,Surgery ,Internal medicine ,medicine ,In patient ,business ,medicine.drug - Abstract
2042 Background: Bevacizumab (BEV) is widely used for treatment of patients with recurrent glioblastoma (GB). Differences in outcome between early versus delayed BEV treatment of recurrent GB are not well defined. We examined the relationship between the time of start of BEV treatment and outcomes in patients with recurrent GB. Methods: In this retrospective chart review derived from our longitudinal database, we identified patients with recurrent GB between 2001 and 2011, who were treated with BEV alone or BEV-containing regimens. Data was analyzed to determine overall survival (OS) from time of diagnosis and progression free survival (PFS) from time of BEV start. Early BEV was defined as start of BEV treatment at first recurrence, while delayed BEV was defined as start of treatment at second recurrence or later. Results: A total of 298 patients with recurrent GB who received BEV were identified, of whom 149 patients received early BEV, 134 patients received delayed BEV, and 15 patients who were excluded because they received BEV upfront. There were no significant differences in the age, sex, performance status and extent of resection between patients treated with early BEV and those treated with delayed BEV. The median time from diagnosis to first recurrence was more than 6 months (mos.) for both groups (6.5 mos. for early BEV and 7.6 mos. for delayed BEV, p = 0.01). The median time from diagnosis to start of BEV was 7.9 mos. for patients with early BEV and 15.6 mos. for patients with delayed BEV (p
- Published
- 2013
35. Phase II trial of the phosphatidyinositol-3 kinase (PI3K) inhibitor BKM120 in recurrent glioblastoma (GBM)
- Author
-
Kathleen R. Lamborn, Shakti Ramkissoon, Timothy F. Cloughesy, Susan M. Chang, Howard Colman, David A. Reardon, Antonio Omuro, Cristian Massacesi, Patrick Y. Wen, Michael D. Prados, Tracy T. Batchelor, Lisa M. DeAngelis, Ingo K. Mellinghoff, Keith L. Ligon, Emmanuelle DiTomaso, W. K. Alfred Yung, Mikael L. Rinne, Mark R. Gilbert, and Andrew S. Chi
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,biology ,business.industry ,Phases of clinical research ,Clinical trial ,medicine.anatomical_structure ,Oncology ,In vivo ,Cancer research ,medicine ,biology.protein ,PTEN ,Immunohistochemistry ,Bone marrow ,U87 ,business ,PI3K/AKT/mTOR pathway - Abstract
2015 Background: The PI3K pathway is activated in most GBMs and represents a potential therapeutic target. BKM120 is an oral, pan-Class I PI3K inhibitor that enters the brain at therapeutic concentrations demonstrated to inhibit PI3K pathway, and potently inhibits the growth of U87 GBM tumors and human glioma tumor spheres in vitro and in vivo. Methods: The Ivy Foundation Early Phase Clinical Trials Consortium is conducting a phase II study of BKM120 in recurrent GBM patients with activation of the PI3K pathway (mutation, homozygous deletion or loss of IHC of PTEN, PIK3CA or PIK3RI mutations, or detectable pAKT). Additional eligibility criteria included radiologic progression, 1st or 2nd relapse, > 18 yrs, KPS > 60, adequate bone marrow and organ function, controlled blood glucose, and no enzyme-inducing antiepileptic drugs. Patients received BKM120 100mg daily. The study consisted of 2 parts conducted concurrently. Part 1 involved up to 15 patients who received BKM120 daily for 8-12 days prior to surgery for recurrent disease. Patients underwent FDG PET, pharmacokinetic (PK) studies, and tumor was obtained for drug concentrations and pharmacodynamic effects. Part 2 consisted of up to 50 patients with unresectable GBM treated with BKM120. The primary endpoint for Part 2 was 6-month progression-free survival (p0 =15%; p1= 32%). Results: To date 7 patients have been enrolled into Part 1, 33 into part 2. There were 5 women and 35 men. Median age was 54 yrs (29-68). Treatment was fairly well-tolerated. Major grades 3/4 toxicities were asymptomatic lipase elevation (5), fatigue (3), hyperglycemia (3), rash (3) elevated AST (1), and depression (1). Analysis of tumor from Part 1 showed reduction of pAkt by IHC. Genotyping of tumor specimens is ongoing. To date 33 patients had positive pAkt, 21 had PTEN loss by IHC. Of the first 19 patients who underwent whole exome sequencing, 3 had PIK3Ca mutations and 6 had PTEN mutations. Conclusions: BKM120 is generally well tolerated in patients with recurrent GBM and achieves adequate tumor concentration to inhibit pAkt. Updated PK and efficacy data and correlation of the latter with tumor genotype will be presented. Clinical trial information: NCT01339052.
- Published
- 2013
36. Comparative impact of treatment on patient reported outcomes (PROs) in patients with glioblastoma (GBM) enrolled in RTOG 0825
- Author
-
R. Jeffrey Lee, Jeffrey S. Wefel, David Brachman, Ritsuko Komaki, Merideth M Wendland, Stephanie L. Pugh, H. Ian Robins, Ian R. Crocker, Minesh P. Mehta, Minhee Won, Terri S. Armstrong, and Mark R. Gilbert
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Newly diagnosed ,medicine.disease ,Surgery ,Internal medicine ,medicine ,In patient ,Progression-free survival ,business ,Glioblastoma - Abstract
2003 Background: RTOG 0825 tested if adding bevicizumab (BEV) to standard chemoradiation improves survival (OS) or progression free survival (PFS) in newly diagnosed GBM. While OS was equivalent, PFS was longer with Bev (Arm 2) than with placebo (Arm 1). Patients completed quality of life and symptom PROs to evaluate clinical benefit. Methods: The M.D. Anderson Symptom Inventory-Brain Tumor Module (MDASI-BT) and the EORTC core Quality of life Questionnaire and brain tumor module (EORTC QLQ-C30/BN20), were completed by pts at baseline (B) and longitudinally (wk 6, 10, 22, 34, and 46) while on study. The difference between treatment arms were compared from B with evaluation in subsequent weeks in those pts without disease progression; and early change (EC) (baseline to wk 10) between those with and without progression as predictors for OS and PFS. Results: 542pts consented to participate on this trial, and 507 randomized pts participated, with completion of forms by 94% at baseline, 75% at wk 10, 61% at wk 22, and 58% at wk 34. There was a trend for all MDASI-BT symptom groupings to be worse in Arm 2, with significant findings in treatment symptoms at wk 22 and wk 34; both affective and generalized disease symptoms were also significantly worse. For EORTCQLQ30/BN20, differential effects varied at each time point, with no persistent differences. For the MDASI-BT, B neurologic symptom grouping and EC in cognitive symptoms were prognostic for both OS and PFS. For the EORTC QLQ30/BN20, global QOL and motor dysfunction at B as well as EC in communication and leg weakness were prognostic for OS; whereas physical function at B and EC in headaches, seizures, and weak legs were prognostic for PFS. Conclusions: The longitudinal collection of PROs in this phase III trial revealed important treatment-related differences as there was overall more deterioration in symptoms and QOL in Arm 2 as compared to Arm 1, with persistent significant differences in treatment associated symptoms. B and EC tumor associated symptoms on both PRO tools were prognostic for OS and PFS. Longitudinal modeling is ongoing to further assess for differential impact of treatment on symptoms and QOL. Support: U10 CA21661, U10 CA37422 and Genentech, Inc. Clinical trial information: NCT00884741.
- Published
- 2013
37. RTOG 0825: Phase III double-blind placebo-controlled trial evaluating bevacizumab (Bev) in patients (Pts) with newly diagnosed glioblastoma (GBM)
- Author
-
David Schiff, Kenneth Aldape, James J. Dignam, Robert Jeraj, Minhee Won, Arnab Chakravarti, Deborah T. Blumenthal, Ritsuko Komaki, Maria Werner-Wasik, Terri S. Armstrong, David Brachman, Kurt A. Jaeckle, Howard Colman, Erik P. Sulman, James N. Atkins, Mark R. Gilbert, Paul D. Brown, Minesh P. Mehta, Jeffrey S. Wefel, and Michael A. Vogelbaum
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,Bevacizumab ,business.industry ,Placebo-controlled study ,Newly diagnosed ,Gene signature ,Placebo ,medicine.disease ,Double blind ,Internal medicine ,medicine ,business ,Nuclear medicine ,medicine.drug ,Glioblastoma - Abstract
1 Background: Chemoradiation (CRT) with temozolomide (TMZ/RT→TMZ) is the standard of care for newly diagnosed GBM. This trial determined if the addition of Bev to standard CRT improves survival (OS) or progression-free survival (PFS) in newly diagnosed GBM. Methods: This phase III trial was conducted by the RTOG, NCCTG, and ECOG. Neurologically stable pts > 18 yrs with KPS ≥ 60, and > 1cm3 tumor tissue block, were randomized to Arm 1: standard CRT + placebo or Arm 2: standard CRT plus Bev (10 mg/kg iv q 2wks). Experimental treatment began at wk 4 of radiation then thru 6-12 cycles of maintenance chemotherapy. Protocol specified co-primary endpoints were OS and PFS, with significance levels of .023 and .002, respectively. At progression, treatment was unblinded and pts allowed to crossover or continue Bev. Symptom, QOL and neurocognitive (NCF) testing was performed in the majority of pts. Secondary analyses evaluated impact of MGMT methylation (meth) and prognostic 9 gene signature status. Results: From 978 registered pts, 637 were randomized. Inadequate tissue (n=105) and blood on imaging (n=40) were key reasons for non-randomization. No difference was found between arms for OS (median 16.1 vs. 15.7 mo, p = 0.11). PFS was extended for Arm 2 (7.3 vs. 10.7 mo, p = 0.004). Pts with MGMT meth had superior OS (23.2 vs. 14.3 mo, p < 0.001) and PFS (14.1 vs. 8.2 mo, p < 0.001). Neither the 9 gene signature nor MGMT predicted selective benefit for Bev treatment, but best prognosis pts (MGMT meth, favorable 9-gene), had a worse survival trend with Bev (15.7 vs 25 mo p = 0.08). To date, 128 pts were unblinded on Arm 1 (salvage Bev in 86) and 87 pts on Arm 2 (continued Bev in 39). Increased grade ≥ 3 toxicity was seen with Bev, mostly neutropenia, hypertension, and DVT/PE. Conclusions: The addition of Bev for newly diagnosed GBM did not improve OS, did improve PFS but did not reach the significance criterion. MGMT and 9 gene profile did not identify selective benefit, but risk subset results suggested strongly against the upfront use of Bev in the best prognosis pts. Full interpretation of the PFS results incorporating symptom burden, QOL, and NCF is ongoing. Support: NCI U10 CA 21661, U10 CA37422, and Genentech. Clinical trial information: NCT00884741.
- Published
- 2013
38. BTTC08-01: A phase II study of bevacizumab and erlotinib after radiation therapy and temozolomide in patients with newly diagnosed glioblastoma (GBM) without MGMT promoter methylation
- Author
-
Pierre Giglio, Monica Loghin, Charles A. Conrad, W. K. Alfred Yung, Kenneth Aldape, Marc C. Chamberlain, Ivo W. Tremont-Lukats, Sean Grimm, Brian Vaillant, Mark R. Gilbert, Jeremy Rudnick, Nicholas A. Vick, John de Groot, Morris D. Groves, Vinay K. Puduvalli, Ryan Merrell, Jeffrey Raizer, Nina Paleologos, Jethro Hu, and Surasak Phuphanich
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Temozolomide ,Bevacizumab ,business.industry ,medicine.medical_treatment ,Brain tumor ,Phases of clinical research ,medicine.disease ,Radiation therapy ,Internal medicine ,medicine ,In patient ,Erlotinib ,business ,neoplasms ,medicine.drug ,Glioblastoma - Abstract
2019^ Background: Patients (pts) with GBM with unmethylated MGMT have a worse prognosis than those with methylated MGMT. Novel approaches for this poor risk group are warranted. The Brain Tumor Trials Collaborative (BTTC) performed a phase II trial evaluating standard chemoradiation followed by bevacizumab and erlotinib in patients with MGMT unmethylated GBM. EGFR and VEGFR are upregulated during radiation suggesting that this combination could be more effective than post-radiation adjuvant temozolomide (TMZ). Methods: After informed consent, adult patients with supratentorial GBM, KPS ≥ 70 and > 1 cm2 tumor block for MGMT promoter analysis were screened. Only tumors with confirmed unmethylated MGMT promoter were enrolled. All patients received RT + TMZ and then approximately 4 weeks after RT they received bevacizumab 10 mg/kg every 2 weeks and erlotinib 150 mg/day, continuously. One cycle was 4 weeks; evaluation by MRI was every 2 cycles. Treatment continued until disease progression or intolerable adverse events. Results: 115 patients were screened; 48 were enrolled (2 unevaluable: 1 for an infratentoral GBM and 1 withdrew after 7 days of treatment) with 29 men, 17 women. Median age was 56 yrs (29-75); median KPS was 90 (70-90). The median number of cycles was 8 (2-38) with 4 patients remaining on trial at the time of analysis. Objective responses: 4 CR, 12 PR and 30 SD. Median PFS is 7.3 months (95% CI (6.4, 11)) and median OS 14.2 months (95% CI (10.7, not reached)). There were no unexpected toxicities; grade 3/4 rate < 5%. Conclusions: Adjuvant bevacizumab and erlotinib in GBM with unmethylated MGMT is well tolerated. Preliminary efficacy data is comparable with outcomes in similar unmethylated MGMT patient populations from the EORTC/NCIC and RTOG 0525 studies. Tissue correlation is being performed. Clinical trial information: NCT00720356.
- Published
- 2013
39. Molecular predictors of outcome and response to bevacizumab (BEV) based on analysis of RTOG 0825, a phase III trial comparing chemoradiation (CRT) with and without BEV in patients with newly diagnosed glioblastoma (GBM)
- Author
-
Maria Werner-Wasik, Arnab Chakravarti, Kenneth Aldape, James J. Dignam, Mark R. Gilbert, Ritsuko Komaki, Erik P. Sulman, Robert Jeraj, Deborah T. Blumenthal, James N. Atkins, David Schiff, Howard Colman, Michael A. Vogelbaum, Robert B. Jenkins, Minhee Won, Paul D. Brown, Minesh P. Mehta, David Brachman, and Kurt A. Jaeckle
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Formalin fixed paraffin embedded ,Bevacizumab ,business.industry ,Newly diagnosed ,medicine.disease ,Gene expression profiling ,Internal medicine ,medicine ,TaqMan ,In patient ,Nuclear medicine ,business ,Treatment Arm ,Glioblastoma ,medicine.drug - Abstract
LBA2010 Background: RTOG 0825 evaluated the addition of BEV to standard CRT in the treatment of GBM and included molecular stratification that assessed the degree of mesenchymal (MES) gene enrichment. We investigated the ability of the MES signature to predict response to BEV. Methods: Sufficient FFPE tissue for molecular analysis was available for 650 registered, eligible patients. TaqMan PCR was performed prospectively using the molecular stratifier on all patients and an expanded 43 member MES set on 234 cases. A subset of specimens was subjected to whole genome expression profiling (GEP). Predictive models were evaluated for their ability to predict survival (overall, OS, and progression-free, PFS) in the training cohort of the BEV arm after adjusting for prognostic factors and treatment arm. Unsupervised clustering of GEP data was used to identify molecular subsets and gene set enrichment analysis (GSEA) performed to evaluate for MES enrichment. Results: We observed a significant association between increasing MES signature and worse PFS and OS in the BEV arm (p=0.036 and p=0.032, respectively). Based on the association between high MES expression and poor outcome in the BEV arm, we sought to optimize a predictor using an expanded set of MES genes. Unbiased gene selection from a total of 43 genes followed by radial basis machine modeling identified a 10-gene predictor of outcome in the BEV arm (p
- Published
- 2013
40. Textiloma mimicking recurrent glioblastoma: A case report
- Author
-
Jackson Hamilton, Gregory N. Fuller, Pedro Garciarena, Dawid Schellingerhout, Raymond Sawaya, Mark Anderson, and Mark R. Gilbert
- Subjects
Cancer Research ,medicine.medical_specialty ,Oncology ,business.industry ,Hemostasis ,Recurrent glioblastoma ,medicine ,Radiology ,Dissection (medical) ,business ,medicine.disease ,Surgery - Abstract
e13044 Background: Although uncommon, materials used for dissection or to achieve hemostasis during neurosurgical procedures can cause an inflammatory reaction called textiloma that can mimic recurrent tumor. We report a case where advanced magnetic resonance (MR) imaging suggested tumor, further mimicking previous reports of clinical and conventional MR patterns of tumor progression. Methods: A 61 year-old woman underwent an initial resection of a temporal lobe glioblastoma, followed by conventional chemoradiation, although maintenance temozolomide was stopped secondary to myelotoxicity. A nodular enhancing mass near to the surgical cavity was noted 10 months after diagnosis, leading to further evaluation with advanced MR imaging. Results: Brain MRI with advanced imaging studies including MR Spectroscopy (MRS), and perfusion imaging by dynamic contract-enhanced (DCE), and dynamic contrast susceptibility (DSC) were performed. There was progressive enlargement of an enhancing nodule in the wall of the resection cavity. MRS demonstrated an elevated choline-to-creatine ratio (approximately 2:1), while the DSC imaging demonstrated a mild amount of leak with a corrected cerebral blood volume value of about 1.93, and negative enhancement integral value 1.67. DCE demonstrated mild vascularity and mild-to-moderate leak, with an elevated peak enhancement integral value of ~22%. These imaging findings suggested recurrent GBM rather than post-treatment change. The mass was resected. Pathologic review showed both textiloma and radiation necrosis. Recurrent glioblastoma was not identified. Conclusions: Although very unusual, textiloma should be considered in the differential diagnosis of any growing mass lesion close to a surgical bed. Reparative granulomata show many of the hallmarks of cancer, including neovascularization, rapid growth and high lipid turnover, making it difficult to distinguish such lesions from true neoplasm, even with advanced imaging, without resorting to histology. Advanced imaging performs best in distinguishing conventional post-treatment change from tumor recurrence. This case highlights the major influence of surgical technique on postoperative imaging appearance.
- Published
- 2013
41. Phase I lead-in to a 2x2x2 factorial trial of dose-dense temozolomide, memantine, mefloquine, and metformin as postradiation adjuvant therapy of glioblastoma (GBM)
- Author
-
Mark R. Gilbert, Isaac Melguizo, Kenneth R. Hess, Charles A. Conrad, Kenneth Aldape, Aaron Mammoser, Erik P. Sulman, Ivo W. Tremont-Lukats, Monica Loghin, Marta Penas-Prado, W. K. Alfred Yung, John de Groot, Vinay K. Puduvalli, and Morris D. Groves
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,Mefloquine ,business.industry ,Memantine ,Pharmacology ,medicine.disease ,Metformin ,Unmet needs ,Internal medicine ,medicine ,Adjuvant therapy ,Lead (electronics) ,business ,medicine.drug ,Glioblastoma - Abstract
TPS2106 Background: Treatment for GBM is an area of unmet need. Despite optimal therapy, survival is poor and 2nd line therapies are scarce. There is an urgent need to find better treatments for recurrence, but also more effective 1st line therapies. Dose-dense temozolomide (ddTMZ) using the 7/14-day regimen has shown promising preliminary results in combination with cytostatic agents. Memantine (MEM) is a glutamate receptor (NMDA) blocker with antiproliferation properties and possibly neuroprotective effect. Mefloquine (MFQ) induces autophagy and apoptosis. Metformin (MFM) has mTOR inhibitor properties. Methods: Trial Design: Phase I/II trial to evaluate adjuvant ddTMZ with MEM MFQ and MFM. Primary objective Phase I: MTDs of ddTMZ with MEM/MFQ/MFM, 3+3 design. MTDs will be the recommended Phase II doses for a subsequent randomized factorial Phase II trial (ddTMZ alone and single, double and triple combinations). Accrual of about 55 eligible patients was calculated for Phase I (48-144). Clinical trial registry number is NCT01430351. Treatment planned: Patients accrued sequentially to ddTMZ with 1, 2, or 3 drugs. Arm 1 (ddTMZ alone) will be enrolled in Phase II only. Patients were first accrued to 1-drug Arms 2-4. Once MTDs were determined, accrual started to 2-drug Arms 5-7. Once completed, accrual to Arm 8 will start. Arms 2-4 were started at a predetermined target dose, and deescalated if excessive toxicity. Treatment in Arms 5-8 will be escalated for each drug to reach MTDs of Arms 2-4. Major eligibility criteria: Adults (≥ 18) with supratentorial GBM, KPS ≥60, adequate bone marrow and organ function. Post chemoradiation MRI ≤14 days before enrollment on stable/decreasing steroids and no progression; registration ≤5 weeks of chemoradiation. Patients on MFQ: no EIAED, EKG without prolonged QTc or arrhythmia. Pregnancy not allowed; adequate contraception required. Informed consent in keeping with IRB policies. Current enrollment: To date, 49 patients started treatment and 18 are still active. Enrollment to Arms 2-4 and 6 is completed and MTDs determined. Accrual is ongoing in Arms 5 and 7, and pending in Arm 8. Clinical trial information: NCT01430351.
- Published
- 2013
42. Randomized phase II 8-arm factorial study of adjuvant dose-dense (dd) temozolomide (TMZ) with permutations of thalidomide (Thal), isotretinoin (CRA), and/or celecoxib (Cel) for newly diagnosed glioblastoma (GBM)
- Author
-
Kenneth Aldape, Morris D. Groves, Charles A. Conrad, Lore W. Lagrone, Monica Loghin, Pierre Giglio, Howard Colman, W. K. Alfred Yung, Victor A. Levin, Mark R. Gilbert, Kenneth R. Hess, Michael J. Fisch, Vinay K. Puduvalli, and John de Groot
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,business.industry ,medicine.medical_treatment ,Brain tumor ,Phases of clinical research ,Pharmacology ,medicine.disease ,Thalidomide ,Internal medicine ,medicine ,Celecoxib ,business ,Adjuvant ,Isotretinoin ,Pseudoprogression ,medicine.drug - Abstract
2003 Background: Concurrent radiation and TMZ followed by 6-12 months of adjuvant TMZ (d 1-5 of a 28d cycle) is the current standard of care for patients with newly diagnosed GBM. The addition of cytostatic or signal transduction agents may enhance efficacy without a significant increase in toxicity. A phase I trial (Neuro-oncology 2009) established the safety of ddTMZ with 2 or 3 of the cytostatic agents. Methods: This randomized phase II study was conducted by the Brain Tumor Trials Collaborative (BTTC) and the MDACC CCOP. The primary objectives: determine if specific cytostatic agents added to ddTMZ alters outcomes (PFS, OS) and compare triplet with doublet therapy. Eligibility criteria: centrally confirmed newly diagnosed GBM, age ≥18, KPS≥60, stable or improved after chemoradiation (pseudoprogression allowed), adequate hematologic, renal and hepatic function. Pts were randomly assigned to 12 treatment cycles (28 d/cycle) in 8 arms: ddTMZ alone (150 mg/m2/day, 7-d on, 7-d off) or TMZ-containing doublet, triplet and quadruplet combinations with Thal, CRA, or Cel. Results: The study enrolled 155 eligible patients from 11/2005 to 9/2010 to the 8 arms of the factorial design. Median age was 53 (18-84) and median KPS, 90 (60-100). Compared with TMZ alone, the TMZ+CRA doublet had worse PFS (10.5, 6.5 mo; p=0.043) and OS (21.2, 11.7 mo; p=0.037). Trends were also seen for worse outcome (PFS, OS) for CRA-containing regimens, improved OS for Cel containing arms and no impact of Thal. A strong trend for OS improvement was seen for triplet compared with doublet regimens (20.1, 17.0 mo; p=0.15), but no difference for PFS. Treatment was well tolerated with expected high rates of grade 3/4 lymphopenia, and overall a modest toxicity rate. Conclusions: The results indicate that the addition of CRA to ddTMZ may be detrimental in patients with newly diagnosed GBM. This study demonstrated the utility of the factorial design in efficiently testing drug combinations, the impact of individual agents in these combinations as well as doublet vs. triplet regimens and supports its utility in testing combinations of targeted agents.
- Published
- 2012
43. Impact of duration of bevacizumab (Bev) treatment in the prognosis of adults with recurrent malignant gliomas
- Author
-
Morris D. Groves, W. K. Alfred Yung, John de Groot, Mark R. Gilbert, Mohamed Ali Hamza, Charles A. Conrad, and Vinay K. Puduvalli
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Bevacizumab ,business.industry ,Recurrent glioblastoma ,Internal medicine ,Standard treatment ,medicine ,business ,medicine.drug ,Surgery - Abstract
2064 Background: Bev is the standard treatment for patients with recurrent glioblastoma (GB) but is also used in treating recurrent anaplastic gliomas (AG). Differences in outcome between these groups and optimal duration of treatment with Bev in pts with recurrent malignant gliomas are not well defined. We examined the relationship between the duration of Bev treatment and the outcome in pts with GB and AG. Methods: In this retrospective chart and data review derived from our longitudinal database, we identified pts with recurrent AG and GB who were treated with Bev alone or Bev-containing regimens between 2005 and 2009; the data was analyzed to determine the overall survival (OS) and the progression free survival (PFS). Results: A total of 261 patients with recurrent malignant gliomas (196 with GB and 65 with AG) were identified. There was no significant difference between the median length of treatment between AG and GB (5.81±0.66 months vs. 6.77±0.52 months, p=0.32). PFS6 was 34.2% (95% CI, 27.8-41.3) for patients with GB and 44.2% (95% CI, 32.5-56.7) for patients with AG. Patients with GB who were treated ≥6 months had a significantly higher OS (29.13 months vs. 20.16 months, p= 0.001) compared to those treated
- Published
- 2012
44. Differences in outcome due to bevacizumab (BEV) discontinuation versus BEV failure in adults with glioblastoma
- Author
-
Vinay K. Puduvalli, W. K. Alfred Yung, Mohamed Ali Hamza, Mark Anderson, and Mark R. Gilbert
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Bevacizumab ,business.industry ,Recurrent glioblastoma ,Salvage therapy ,medicine.disease ,Surgery ,Discontinuation ,Tumor progression ,Internal medicine ,medicine ,business ,medicine.drug ,Glioblastoma - Abstract
2030 Background: BEV is approved for use in recurrent glioblastoma. Patients (pts) who benefit from BEV therapy are often treated until tumor progression but fail to respond to salvage therapy suggesting that BEV may alter tumor biology. In a subset of pts who benefit from BEV, treatment is discontinued for reasons other than disease progression; the characteristics and outcomes of this subset are poorly defined. Methods: In this IRB approved retrospective study, our neuro-oncology longitudinal database was screened for pts treated with BEV for ≥ 6 months (mo) between 2005-2010 and 18 pts were identified in whom BEV was discontinued for reasons other than disease progression (BEV-D group). A cohort of 72 pts who received BEV until treatment failure due to progression was used as comparator (BEV-F group). Results: In the BEV-D group, 5 pts completed a planned treatment course and 13 stopped BEV due to toxicity; in this group, progression free survival at 12 mo (PFS12) was 83.3% (95% CI, 56.8-94.3) and median time to progression (TTP) 27.6 mo. Median TTP after BEV discontinuation was 7.0 mo. In contrast, in the BEV-F group, PFS12 was 24.6% (95% CI, 13.9-36.2) and median PFS 9.7 mo. Length of BEV therapy was not significantly different between the groups with a median time to discontinuation of 10.2 and 12 mo. In 12/18 pts in the BEV-D group who subsequently had tumor recurrence a predominantly local pattern of progression was seen unlike those in the BEV-F group who had more infiltrative or distant failures. Salvage therapy yielded a PFS-6 of 28.6% (95% CI, 4.1-61.2) with a median PFS of 17.1 wk compared with 6.8% (95% CI, 1.2-19.8) and 9 wk in the comparator group. Conclusions: Among pts who benefit from BEV therapy, the BEV-D group did not experience an immediate progression suggesting continued benefit after BEV cessation. This cohort also had a less invasive pattern of recurrence and a possibly improved response to salvage therapy compared with BEV-F group. Our results suggest that planned cessation of BEV therapy could potentially change patterns of progression and response to subsequent therapy. This strategy warrants further evaluation in prospective studies given the absence of effective salvage therapy after BEV failure.
- Published
- 2012
45. Bevacizumab versus bevacizumab plus vorinostat in adults with recurrent malignant glioma: Results of a phase I part of a phase I/II trial
- Author
-
W. K. Alfred Yung, Morris D. Groves, Sandra Ictech, Charles A. Conrad, Kenneth R. Hess, Jing Wu, Mark R. Gilbert, Carolyn Loch, Vinay K. Puduvalli, and John de Groot
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Bevacizumab ,business.industry ,Phases of clinical research ,medicine.disease ,Phase i ii ,Hypoxia-inducible factors ,Downregulation and upregulation ,Glioma ,Internal medicine ,medicine ,Histone deacetylase ,business ,Vorinostat ,medicine.drug - Abstract
2029^ Background: Antiangiogenic therapy using b evacizumab (Bev) has shown promising activity against recurrent glioblastoma (GBM). However, most patients have disease progression after striking but transient responses. Salvage therapies have been uniformly ineffective, suggesting development of resistance mechanisms including upregulation of other proangiogenic factors and increased activity of hypoxia inducible factors (HIF)-1a. Vorinostat, a histone deacetylase (HDAC) inhibitor has single agent activity against recurrent GBM and downregulates HIF-1a and other proangiogenic and invasive factors. We hypothesized that HDAC inhibition combined with Bev would result in improved clinical outcome. We report the results of the Phase I portion of the study preceding the initiation of the 2-arm adaptive randomized Phase II study. Methods: Adults with recurrent malignant glioma, KPS ≥ 60, normal hepatic, renal and marrow organ function and no prior exposure to Bev or Vorinostat were enrolled to the combination therapy after the confirmation of recurrent GBM. A conventional 3+3 Phase I design was used to determine the maximum tolerated dose (MTD) and the toxicity profile of the combination of Bev and Vorinostat. The starting dose was Bev at 10mg/kg administered on days 1 and 15 intravenously and Vorinostat 400 mg/day orally on days 1 to 7, and days 15 to 21 with each cycle being 28 days. Results: A total of 6 patients were enrolled and all 6 patients were evaluable. Three patients were enrolled in the first cohort at the starting dose of the combination and completed the first cycle. One patient experienced a grade 3 ALT elevation and grade 3 hyperglycemia, which were designated as possibly related to vorinostat and constituting a dose-limiting toxicity (DLT). No grade 4 toxicities were noted. The cohort was expanded by 3 more patients with none of these patients experienced a DLT in the first cycle. The starting dose level of Bev and v orinostat was declared the Phase II dose. Conclusions: Combination of Bev (10 mg/kg q 2 weeks) and of vorinostat (400 mg on days 1 to 7 and 15 to 21) has tolerable toxicity profile. This will be followed by a multicenter Bayesian adaptive randomized Phase II study.
- Published
- 2012
46. A revised RTOG recursive partitioning analysis (RPA) model for glioblastoma based upon multiplatform biomarker profiles
- Author
-
Alexander C. Klimowicz, Minesh P. Mehta, Monika E. Hegi, Arnab Chakravarti, Walter J. Curran, Maria Werner-Wasik, M. Bredel, Roger Stupp, Kenneth Aldape, Meihua Wang, Erik P. Sulman, Anthony M. Magliocco, Christopher J. Schultz, Mark R. Gilbert, and Anita Mahajan
- Subjects
Radiation therapy ,Cancer Research ,Clinical variables ,Oncology ,business.industry ,medicine.medical_treatment ,medicine ,Recursive partitioning ,Computational biology ,Bioinformatics ,medicine.disease ,business ,Glioblastoma - Abstract
2001 Background: The Radiation Therapy Oncology Group (RTOG) recursive partitioning analysis (RPA) model, which relies on clinical variables, has been used worldwide to establish distinct prognostic classes of patients (pts) with malignant glioma as well as eligibility criteria for clinical trials. In the present study, we have updated the RPA to include additional molecular variables, specifically for glioblastoma (GBM) patients treated in the temozolomide (TMZ)-era, to make the model more relevant, contemporary, and discriminatory. Methods: The dataset utilized was from RTOG 0525, a phase III study examining radiation (RT) with concurrent TMZ, followed by adjuvant standard dose vs. dose-dense TMZ in pts with newly-diagnosed GBM. 162 pts from RTOG 0525 had available tissues for profiling of key signaling molecules using the AQUA platform. Results: pAKT, c-met, and MGMT protein were each found to be significantly associated with adverse outcome on multivariate analysis. These variables were combined with clinical and genetic biomarkers (e.g., MGMT promoter methylation, IDH1 mutation, mRNA profiling) previously found to be of significance (MCP model, ASCO, 2011) to generate an even more robust, discriminatory RTOG RPA model. The explained variation for these three classification models was found to be 41.7 (Current RPA), 19 (MCP), and 14.9% (Clinical RPA), respectively, with higher values indicating better separation of prognostic groups (see table). Conclusions: The current RTOG RPA classification model, based upon incorporation of multi-platform biomarker analysis, holds promise for RT+TMZ-treated GBM patients; further validation of this model is planned. Financial Support: NCI grants U10 CA21661, U10 CA37422, U24 CA114734, 1RC2CA148190, 1RC2CA148190, RO1CA108633, and BTFC grant. [Table: see text]
- Published
- 2012
47. IDH1 status and survival benefit from surgical resection of enhancing and nonenhancing tumor in malignant astrocytomas
- Author
-
Jason Beiko, Frederick F. Lang, Mark R. Gilbert, Kenneth Aldape, Ian E. McCutcheon, Sujit S. Prabhu, Raymond Sawaya, Ganesh Rao, Daniel P. Cahill, Jeffrey S. Weinberg, and Dima Suki
- Subjects
Surgical resection ,Cancer Research ,medicine.medical_specialty ,Pathology ,IDH1 ,business.industry ,Who grade ,medicine.disease ,Resection ,Survival benefit ,Oncology ,medicine ,Radiology ,business ,Glioblastoma ,Anaplastic astrocytoma - Abstract
2019 Background: The value of maximal safe resection for malignant astrocytic gliomas (AA, WHO Grade III anaplastic astrocytoma and GBM, WHO Grade IV glioblastoma) has sometimes been controversial, because of confounding between measures of surgical resection and other prognostic factors. IDH1 gene mutations are associated with improved survival in glioma patients, and are thought to identify tumors with a distinct molecular evolutionary origin. We sought to determine the prognostic impact of surgical resection on survival after controlling for IDH1 status in malignant astrocytomas. Methods: Clinical parameters including preoperative and postoperative MRI-based tumor volume were recorded prospectively on 407 malignant astrocytoma patients – AA (n=157) and GBM (n=250). IDH1 status was assessed by sequencing and R132H-specific immunohistochemistry. Results: The measures of surgical resection associated with longer survival differed between IDH1 wild-type and mutant tumors. In multivariate analyses of IDH1 wild-type tumors (controlling for age, Karnofsky performance score, tumor location, and tumor grade), residual postoperative enhancement was associated with a median survival of 9.9 mo vs. 17.4 mo with no enhancement (HR=1.73, 95% CI, 1.19-2.52, p=.004). Residual non-enhancing disease, however, was not associated with survival (scored as continuous volumetric cc, 95% CI 0.99-1.01, p=.608). These results are consistent with prior studies of GBM, which are largely IDH1 wild-type lesions (Lacroix et al., J Neurosurg 95:190-8, 2001). In contrast, in IDH1 mutant tumors, both residual enhancing (HR=7.93, 95%CI 1.14-55.22, p=.037) and non-enhancing (HR=1.03, 95% CI 1.01-1.05, p=.005) postoperative tumor burden were associated with worse survival. Conclusions: These data suggest surgical resection in malignant astrocytic gliomas may be individualized based on IDH1 genotype. IDH1 mutant tumors have a better baseline overall prognosis, therefore more aggressive surgery and tolerance of temporary peri-operative neurologic deficits can be weighed in an attempt to gain the additional survival benefit that appears to be associated with reducing non-enhancing tumor burden.
- Published
- 2012
48. Lenalidomide and irinotecan in adults with recurrent malignant gliomas: Phase I results of the phase I/II trial
- Author
-
Charles A. Conrad, Sandra Ictech, Morris D. Groves, Monica Loghin, Vinay K. Puduvalli, W. K. Alfred Yung, Victor A. Levin, Sanghee Kang, John de Groot, Mark R. Gilbert, and Kenneth R. Hess
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Treatment options ,Surgery ,Irinotecan ,Phase i ii ,Internal medicine ,medicine ,Progression-free survival ,business ,medicine.drug ,Lenalidomide - Abstract
2036 Background: Patients with recurrent malignant gliomas have limited treatment options. We previously reported promising results using 6 month progression free survival [PFS6] endpoint combining irinotecan and thalidomide compared to historical controls. In this study, we tested the tolerability and efficacy of Irinotecan combined with Lenalidomide, a more potent thalidomide analogue with unique antiangiogenic and immunomodulatory properties. Methods: This phase I portion of the phase I/II study aimed to determine the maximum tolerated doses (MTD) of irinotecan and lenalidomide. Eligible patients were adults (>=18 y) with recurrent WHO Grade 3 or 4 gliomas who were not on EIAED, had failed prior radiation therapy, had a KPS>=60, had adequate marrow, renal and hepatic organ function, no major medical illnesses and no other concurrent malignancies. Each cycle was designated as 4 weeks in duration. Results: Two of the 4 patients enrolled at the starting dose level of Lenalidomide 10 mg/day on days 1-21 and irinotecan 200 mg/m2 q2 weeks developed rash as dose limiting toxicity (DLT). The trial was restarted with no change in irinotecan dose but with a lowered Lenalidomide dose of 7.5 mg/day for cycle 1 with escalation to 10 mg/day for cycle 2 and beyond. Of 3 eligible patients enrolled, no DLTs were noted even after cycle 2. The dose was hence escalated to Lenalidomide 10 mg/day with unchanged irinotecan dose. Only 1/6 patients experienced a DLT (pulmonary embolism); however, it was noted that 4/6 patients required dose reduction of irinotecan to 150 mg/m2 after cycle 1. One patient died during cycle 2 of unknown causes (autopsy declined) without reports of preceding toxicities. Non DLT toxicities included neutropenia, leukopenia, hypokalemia, diarrhea, fatigue and nausea/vomiting. Lenalidomide pharmacokinetic data, obtained by serial blood draws initially after one dose of the drug on day 0 and subsequently after irinotecan and lenalidomide dose on day 1 to examine drug interactions, will be presented. Conclusions: Based on these results, the maximum tolerated dose was Lenalidomide 10 mg/day on days 1-21 and irinotecan 150 mg/m2 q 2 weeks every 28 days which will be used in the phase II study that will open shortly.
- Published
- 2012
49. RTOG 0525: Molecular correlates from a randomized phase III trial of newly diagnosed glioblastoma
- Author
-
David W. Andrews, Mark R. Gilbert, Lindsey Heathcock, Erik P. Sulman, G. Jones, Robert B. Jenkins, Meihua Wang, Monika E. Hegi, Lihong Long, Minesh P. Mehta, Christopher J. Schultz, Arnab Chakravarti, Kenneth Aldape, Howard Colman, and Kristin Diefes
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,IDH1 ,Training set ,Microarray ,business.industry ,Newly diagnosed ,medicine.disease ,Clinical trial ,Risk groups ,Internal medicine ,medicine ,Biomarker (medicine) ,business ,Glioblastoma - Abstract
LBA2000 Background: Formalin-fixed, paraffin embedded GBM tumor tissue, adequate for conventional MGMT methylation analyses, was required for entry onto the RTOG 0525 clinical trial. Methods: Four prognostic biomarkers were evaluated on a training set of 220 retrospectively obtained GBM samples, consisting of IDH1 mutation, the glioma-CpG island methylator phenotype (G-CIMP), a microarray-based mRNA panel and a novel MGMT promoter methylation assay. For each biomarker, 2 (IDH1 and mRNA) or 3 subgroups (G-CIMP and MGMT) were defined based on associations with overall survival. All combinations (36 possible) of each of the 4 biomarker-derived subgroups were then defined and compared with survival data and then consolidated into 4 risk groups. Once created in the training set, this model was applied to the RTOG 0525 samples (n=763) for external validation. Results: Application of the molecular risk classification to RTOG 0525 samples (left table) showed a highly significant survival association (p [Table: see text]
- Published
- 2011
50. Phase I study of aflibercept (VEGF Trap) and temozolomide in newly diagnosed, high-grade glioma
- Author
-
Patrick Y. Wen, Lisa M. DeAngelis, Tracy T. Batchelor, Xiaobu Ye, Stuart A. Grossman, Alice P. Chen, J. F. De Groot, J. Drappatz, Mark R. Gilbert, M. Prados, Antonio Omuro, T. Cloughesy, W. K. A. Yung, Susan M. Chang, Frank S. Lieberman, Joy D. Fisher, and K. Lamborn
- Subjects
Oncology ,Placental growth factor ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,business.industry ,medicine.medical_treatment ,Brain tumor ,medicine.disease ,Surgery ,Radiation therapy ,Vascular endothelial growth factor A ,Regimen ,Concomitant ,Internal medicine ,Medicine ,business ,Aflibercept ,medicine.drug - Abstract
2043 Background: Anti-vascular endothelial growth factor (VEGF) therapy has shown promise in the treatment of high-grade gliomas (HGG). Aflibercept is a recombinant human fusion protein that acts as a soluble decoy receptor for VEGFA, VEGF-B and placental growth factor (PlGF), depleting circulating levels of these growth factors. Methods: The Adult Brain Tumor Consortium (ABTC) conducted a phase I trial of aflibercept and temozolomide (TMZ) in patients with newly diagnosed high-grade gliomas (HGG). Three cohorts were examined: Cohort 1: Aflibercept with radiotherapy and concomitant temozolomide; Cohort 2: Aflibercept and adjuvant temozolomide using the 5/28 regimen; and Cohort 3: Aflibercept and adjuvant temozolomide using the 21/28 day regimen. Eligibility criteria included histologically proven newly-diagnosed glioblastoma (GBM) or anaplastic glioma (AG), > 18 yrs old, KPS > 60, adequate bone marrow reserve and organ function. Patients initially received 2 mg/kg of aflibercept every 2 weeks. If no dose-...
- Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.