Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Michael D. Prados is active.

Publication


Featured researches published by Michael D. Prados.


Journal of Clinical Oncology | 2009

Bevacizumab Alone and in Combination With Irinotecan in Recurrent Glioblastoma

Henry S. Friedman; Michael D. Prados; Patrick Y. Wen; Tom Mikkelsen; David Schiff; Lauren E. Abrey; W. K. Alfred Yung; Nina Paleologos; Martin K. Nicholas; Randy L. Jensen; James J. Vredenburgh; Jane Huang; Maoxia Zheng; Timothy F. Cloughesy

PURPOSE We evaluated the efficacy of bevacizumab, alone and in combination with irinotecan, in patients with recurrent glioblastoma in a phase II, multicenter, open-label, noncomparative trial. PATIENTS AND METHODS One hundred sixty-seven patients were randomly assigned to receive bevacizumab 10 mg/kg alone or in combination with irinotecan 340 mg/m(2) or 125 mg/m(2) (with or without concomitant enzyme-inducing antiepileptic drugs, respectively) once every 2 weeks. Primary end points were 6-month progression-free survival and objective response rate, as determined by independent radiology review. Secondary end points included safety and overall survival. RESULTS In the bevacizumab-alone and the bevacizumab-plus-irinotecan groups, estimated 6-month progression-free survival rates were 42.6% and 50.3%, respectively; objective response rates were 28.2% and 37.8%, respectively; and median overall survival times were 9.2 months and 8.7 months, respectively. There was a trend for patients who were taking corticosteroids at baseline to take stable or decreasing doses over time. Of the patients treated with bevacizumab alone or bevacizumab plus irinotecan, 46.4% and 65.8%, respectively, experienced grade > or = 3 adverse events, the most common of which were hypertension (8.3%) and convulsion (6.0%) in the bevacizumab-alone group and convulsion (13.9%), neutropenia (8.9%), and fatigue (8.9%) in the bevacizumab-plus-irinotecan group. Intracranial hemorrhage was noted in two patients (2.4%) in the bevacizumab-alone group (grade 1) and in three patients (3.8%) patients in the bevacizumab-plus-irinotecan group (grades 1, 2, and 4, respectively). CONCLUSION Bevacizumab, alone or in combination with irinotecan, was well tolerated and active in recurrent glioblastoma.


Cell | 2013

The somatic genomic landscape of glioblastoma.

Cameron Brennan; Roel G.W. Verhaak; Aaron McKenna; Benito Campos; Houtan Noushmehr; Sofie R. Salama; Siyuan Zheng; Debyani Chakravarty; J. Zachary Sanborn; Samuel H. Berman; Rameen Beroukhim; Brady Bernard; Chang-Jiun Wu; Giannicola Genovese; Ilya Shmulevich; Jill S. Barnholtz-Sloan; Lihua Zou; Rahulsimham Vegesna; Sachet A. Shukla; Giovanni Ciriello; W.K. Yung; Wei Zhang; Carrie Sougnez; Tom Mikkelsen; Kenneth D. Aldape; Darell D. Bigner; Erwin G. Van Meir; Michael D. Prados; Andrew E. Sloan; Keith L. Black

We describe the landscape of somatic genomic alterations based on multidimensional and comprehensive characterization of more than 500 glioblastoma tumors (GBMs). We identify several novel mutated genes as well as complex rearrangements of signature receptors, including EGFR and PDGFRA. TERT promoter mutations are shown to correlate with elevated mRNA expression, supporting a role in telomerase reactivation. Correlative analyses confirm that the survival advantage of the proneural subtype is conferred by the G-CIMP phenotype, and MGMT DNA methylation may be a predictive biomarker for treatment response only in classical subtype GBM. Integrative analysis of genomic and proteomic profiles challenges the notion of therapeutic inhibition of a pathway as an alternative to inhibition of the target itself. These data will facilitate the discovery of therapeutic and diagnostic target candidates, the validation of research and clinical observations and the generation of unanticipated hypotheses that can advance our molecular understanding of this lethal cancer.


British Journal of Cancer | 2000

A phase II study of temozolomide vs. procarbazine in patients with glioblastoma multiforme at first relapse.

W. K. Yung; Robert E. Albright; Jeffrey J. Olson; R Fredericks; Karen Fink; Michael D. Prados; M. Brada; A. M. Spence; Raymond J. Hohl; William R. Shapiro; Michael J. Glantz; Harry S. Greenberg; R G Selker; Nicholas A. Vick; R Rampling; Henry S. Friedman; Peter C. Phillips; J Bruner; Nancy Yue; D Osoba; S Zaknoen; V A Levin

A randomized, multicentre, open-label, phase II study compared temozolomide (TMZ), an oral second-generation alkylating agent, and procarbazine (PCB) in 225 patients with glioblastoma multiforme at first relapse. Primary objectives were to determine progression-free survival (PFS) at 6 months and safety for TMZ and PCB in adult patients who failed conventional treatment. Secondary objectives were to assess overall survival and health-related quality of life (HRQL). TMZ was given orally at 200 mg/m2/day or 150 mg/m2/day (prior chemotherapy) for 5 days, repeated every 28 days. PCB was given orally at 150 mg/m2/day or 125 mg/m2/day (prior chemotherapy) for 28 days, repeated every 56 days. HRQL was assessed using the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-C30 [+3]) and the Brain Cancer Module 20 (BCM20). The 6-month PFS rate for patients who received TMZ was 21%, which met the protocol objective. The 6-month PFS rate for those who received PCB was 8% (P = 0.008, for the comparison). Overall PFS significantly improved with TMZ, with a median PFS of 12.4 weeks in the TMZ group and 8.32 weeks in the PCB group (P = 0.0063). The 6-month overall survival rate for TMZ patients was 60% vs. 44% for PCB patients (P = 0.019). Freedom from disease progression was associated with maintenance of HRQL, regardless of treatment received. TMZ had an acceptable safety profile; most adverse events were mild or moderate in severity.


Journal of Clinical Oncology | 1999

Outcomes and Prognostic Factors in Recurrent Glioma Patients Enrolled Onto Phase II Clinical Trials

Eric T. Wong; Kenneth R. Hess; Mary Jo Gleason; Kurt A. Jaeckle; Athanassios P. Kyritsis; Michael D. Prados; Victor A. Levin; W. K. Alfred Yung

PURPOSE To determine aggregate outcomes and prognostic covariates in patients with recurrent glioma enrolled onto phase II chemotherapy trials. PATIENTS AND METHODS Patients from eight consecutive phase II trials included 225 with recurrent glioblastoma multiforme (GBM) and 150 with recurrent anaplastic astrocytoma (AA). Their median age was 45 years (range, 15 to 82 years) and their median Karnofsky performance score was 80 (range, 60 to 100). Prognostic covariates were analyzed with respect to tumor response, progression-free survival (PFS), and overall survival (OS) by multivariate logistic and Cox proportional hazards regression analyses. RESULTS Overall, 34 (9%) had complete or partial response, whereas 80 (21%) were alive and progression-free at 6 months (APF6). The median PFS was 10 weeks and median OS was 30 weeks. Histology was a robust prognostic factor across all outcomes. GBM patients had significantly poorer outcomes than AA patients. The APF6 proportion was 15% for GBM and 31% for AA, whereas the median PFS was 9 weeks for GBM and 13 weeks for AA. Results were also significantly poorer for patients with more than two prior surgeries or chemotherapy regimens. CONCLUSION Histology is a dominant factor in determining outcome in patients with recurrent glioma enrolled onto phase II trials. Future trials should be designed with separate histology strata.


Journal of Clinical Oncology | 2008

Role of Extent of Resection in the Long-Term Outcome of Low-Grade Hemispheric Gliomas

Justin S. Smith; Edward F. Chang; Kathleen R. Lamborn; Susan M. Chang; Michael D. Prados; Soonmee Cha; Tarik Tihan; Scott R. VandenBerg; Michael W. McDermott; Mitchel S. Berger

PURPOSE The prognostic role of extent of resection (EOR) of low-grade gliomas (LGGs) is a major controversy. We designed a retrospective study to assess the influence of EOR on long-term outcomes of LGGs. PATIENTS AND METHODS The study population (N = 216) included adults undergoing initial resection of hemispheric LGG. Region-of-interest analysis was performed to measure tumor volumes based on fluid-attenuated inversion-recovery (FLAIR) imaging. RESULTS Median preoperative and postoperative tumor volumes and EOR were 36.6 cm(3) (range, 0.7 to 246.1 cm(3)), 3.7 cm(3) (range, 0 to 197.8 cm(3)) and 88.0% (range, 5% to 100%), respectively. There was no operative mortality. New postoperative deficits were noted in 36 patients (17%); however, all but four had complete recovery. There were 34 deaths (16%; median follow-up, 4.4 years). Progression and malignant progression were identified in 95 (44%) and 44 (20%) cases, respectively. Patients with at least 90% EOR had 5- and 8-year overall survival (OS) rates of 97% and 91%, respectively, whereas patients with less than 90% EOR had 5- and 8-year OS rates of 76% and 60%, respectively. After adjusting each measure of tumor burden for age, Karnofsky performance score (KPS), tumor location, and tumor subtype, OS was predicted by EOR (hazard ratio [HR] = 0.972; 95% CI, 0.960 to 0.983; P < .001), log preoperative tumor volume (HR = 4.442; 95% CI, 1.601 to 12.320; P = .004), and postoperative tumor volume (HR = 1.010; 95% CI, 1.001 to 1.019; P = .03), progression-free survival was predicted by log preoperative tumor volume (HR = 2.711; 95% CI, 1.590 to 4.623; P <or= .001) and postoperative tumor volume (HR = 1.007; 95% CI, 1.001 to 1.014; P = .035), and malignant progression-free survival was predicted by EOR (HR = 0.983; 95% CI, 0.972 to 0.995; P = .005) and log preoperative tumor volume (HR = 3.826; 95% CI, 1.632 to 8.969; P = .002). CONCLUSION Improved outcome among adult patients with hemispheric LGG is predicted by greater EOR.


The New England Journal of Medicine | 2015

Glioma Groups Based on 1p/19q, IDH, and TERT Promoter Mutations in Tumors

Jeanette E. Eckel-Passow; Daniel H. Lachance; Annette M. Molinaro; Kyle M. Walsh; Paul A. Decker; Hugues Sicotte; Melike Pekmezci; Terri Rice; Matt L. Kosel; Ivan Smirnov; Gobinda Sarkar; Alissa Caron; Thomas M. Kollmeyer; Corinne Praska; Anisha R. Chada; Chandralekha Halder; Helen M. Hansen; Lucie McCoy; Paige M. Bracci; Roxanne Marshall; Shichun Zheng; Gerald F. Reis; Alexander R. Pico; Brian Patrick O’Neill; Jan C. Buckner; Caterina Giannini; Jason T. Huse; Arie Perry; Tarik Tihan; Mitchell S. Berger

BACKGROUND The prediction of clinical behavior, response to therapy, and outcome of infiltrative glioma is challenging. On the basis of previous studies of tumor biology, we defined five glioma molecular groups with the use of three alterations: mutations in the TERT promoter, mutations in IDH, and codeletion of chromosome arms 1p and 19q (1p/19q codeletion). We tested the hypothesis that within groups based on these features, tumors would have similar clinical variables, acquired somatic alterations, and germline variants. METHODS We scored tumors as negative or positive for each of these markers in 1087 gliomas and compared acquired alterations and patient characteristics among the five primary molecular groups. Using 11,590 controls, we assessed associations between these groups and known glioma germline variants. RESULTS Among 615 grade II or III gliomas, 29% had all three alterations (i.e., were triple-positive), 5% had TERT and IDH mutations, 45% had only IDH mutations, 7% were triple-negative, and 10% had only TERT mutations; 5% had other combinations. Among 472 grade IV gliomas, less than 1% were triple-positive, 2% had TERT and IDH mutations, 7% had only IDH mutations, 17% were triple-negative, and 74% had only TERT mutations. The mean age at diagnosis was lowest (37 years) among patients who had gliomas with only IDH mutations and was highest (59 years) among patients who had gliomas with only TERT mutations. The molecular groups were independently associated with overall survival among patients with grade II or III gliomas but not among patients with grade IV gliomas. The molecular groups were associated with specific germline variants. CONCLUSIONS Gliomas were classified into five principal groups on the basis of three tumor markers. The groups had different ages at onset, overall survival, and associations with germline variants, which implies that they are characterized by distinct mechanisms of pathogenesis. (Funded by the National Institutes of Health and others.).


Cancer Cell | 2010

A Hierarchy of Self-Renewing Tumor-Initiating Cell Types in Glioblastoma

Ruihuan Chen; Merry Nishimura; Stephanie M. Bumbaca; Samir Kharbanda; William F. Forrest; Ian Kasman; Joan M. Greve; Robert Soriano; Laurie L. Gilmour; Celina Sanchez Rivers; Zora Modrusan; Serban Nacu; Steve Guerrero; Kyle A. Edgar; Jeffrey Wallin; Katrin Lamszus; Manfred Westphal; Susanne Heim; C. David James; Scott R. VandenBerg; Joseph F. Costello; Scott Moorefield; Cynthia Cowdrey; Michael D. Prados; Heidi S. Phillips

The neural stem cell marker CD133 is reported to identify cells within glioblastoma (GBM) that can initiate neurosphere growth and tumor formation; however, instances of CD133(-) cells exhibiting similar properties have also been reported. Here, we show that some PTEN-deficient GBM tumors produce a series of CD133(+) and CD133(-) self-renewing tumor-initiating cell types and provide evidence that these cell types constitute a lineage hierarchy. Our results show that the capacities for self-renewal and tumor initiation in GBM need not be restricted to a uniform population of stemlike cells, but can be shared by a lineage of self-renewing cell types expressing a range of markers of forebrain lineage.


Nature Genetics | 2009

Variants in the CDKN2B and RTEL1 regions are associated with high-grade glioma susceptibility.

Margaret Wrensch; Robert B. Jenkins; Jeffrey S. Chang; Ru Fang Yeh; Yuanyuan Xiao; Paul A. Decker; Karla V. Ballman; Mitchel S. Berger; Jan C. Buckner; Susan M. Chang; Caterina Giannini; Chandralekha Halder; Thomas M. Kollmeyer; Matthew L. Kosel; Daniel H. Lachance; Lucie McCoy; Brian Patrick O'Neill; Joe Patoka; Alexander R. Pico; Michael D. Prados; Charles P. Quesenberry; Terri Rice; Amanda L. Rynearson; Ivan Smirnov; Tarik Tihan; Joseph L. Wiemels; Ping Yang; John K. Wiencke

The causes of glioblastoma and other gliomas remain obscure. To discover new candidate genes influencing glioma susceptibility, we conducted a principal component–adjusted genome-wide association study (GWAS) of 275,895 autosomal variants among 692 adult high-grade glioma cases (622 from the San Francisco Adult Glioma Study (AGS) and 70 from the Cancer Genome Atlas (TCGA)) and 3,992 controls (602 from AGS and 3,390 from Illumina iControlDB (iControls)). For replication, we analyzed the 13 SNPs with P < 10−6 using independent data from 176 high-grade glioma cases and 174 controls from the Mayo Clinic. On 9p21, rs1412829 near CDKN2B had discovery P = 3.4 × 10−8, replication P = 0.0038 and combined P = 1.85 × 10−10. On 20q13.3, rs6010620 intronic to RTEL1 had discovery P = 1.5 × 10−7, replication P = 0.00035 and combined P = 3.40 × 10−9. For both SNPs, the direction of association was the same in discovery and replication phases.


Clinical Cancer Research | 2006

Phase I/II Study of Imatinib Mesylate for Recurrent Malignant Gliomas: North American Brain Tumor Consortium Study 99-08

Patrick Y. Wen; W. K. Alfred Yung; Kathleen R. Lamborn; Patricia L M Dahia; Yanfeng Wang; Bin Peng; Lauren E. Abrey; Jeffrey Raizer; Timothy F. Cloughesy; Karen Fink; Mark R. Gilbert; Susan M. Chang; Larry Junck; David Schiff; Frank S. Lieberman; Howard A. Fine; Minesh P. Mehta; H. Ian Robins; Lisa M. DeAngelis; Morris D. Groves; Vinay K. Puduvalli; Victor A. Levin; Charles A. Conrad; Elizabeth A. Maher; Kenneth D. Aldape; Michael Hayes; Merrill J. Egorin; Renaud Capdeville; Richard S. Kaplan; Anthony J. Murgo

Purpose: Phase I: To determine the maximum tolerated doses, toxicities, and pharmacokinetics of imatinib mesylate (Gleevec) in patients with malignant gliomas taking enzyme-inducing antiepileptic drugs (EIAED) or not taking EIAED. Phase II: To determine the therapeutic efficacy of imatinib. Experimental Design: Phase I component used an interpatient dose escalation scheme. End points of the phase II component were 6-month progression-free survival and response. Results: Fifty patients enrolled in the phase I component (27 EIAED and 23 non-EIAED). The maximum tolerated dose for non-EIAED patients was 800 mg/d. Dose-limiting toxicities were neutropenia, rash, and elevated alanine aminotransferase. EIAED patients received up to 1,200 mg/d imatinib without developing dose-limiting toxicity. Plasma exposure of imatinib was reduced by ∼68% in EIAED patients compared with non-EIAED patients. Fifty-five non-EIAED patients (34 glioblastoma multiforme and 21 anaplastic glioma) enrolled in the phase II component. Patients initially received 800 mg/d imatinib; 15 anaplastic glioma patients received 600 mg/d after hemorrhages were observed. There were 2 partial response and 6 stable disease among glioblastoma multiforme patients and 0 partial response and 5 stable disease among anaplastic glioma patients. Six-month progression-free survival was 3% for glioblastoma multiforme and 10% for anaplastic glioma patients. Five phase II patients developed intratumoral hemorrhages. Conclusions: Single-agent imatinib has minimal activity in malignant gliomas. CYP3A4 inducers, such as EIAEDs, substantially decreased plasma exposure of imatinib and should be avoided in patients receiving imatinib for chronic myelogenous leukemia and gastrointestinal stromal tumors. The evaluation of the activity of combination regimens incorporating imatinib is under way in phase II trials.


Investigational New Drugs | 2005

Phase II study of CCI-779 in patients with recurrent glioblastoma multiforme

Susan M. Chang; Patrick Y. Wen; Timothy F. Cloughesy; Harry S. Greenberg; David Schiff; Charles A. Conrad; Karen Fink; H. Ian Robins; Lisa De Angelis; Jeffrey Raizer; Kenneth R. Hess; Kenneth D. Aldape; Kathleen R. Lamborn; John G. Kuhn; Janet Dancey; Michael D. Prados

Purpose: Loss of PTEN, which is common in glioblastoma multiforme (GBM), results in activation of the mammalian target of rapapmycin (mTOR), thereby increasing mRNA translation of a number of key proteins required for cell-cycle progression. CCI-779 is an inhibitor of mTOR. The primary objectives of this study were to determine the efficacy of CCI-779 in patients with recurrent GBM and to further assess the toxicity of the drug. Experimental Design: CCI-779 was administered weekly at a dose of 250 mg intravenously for patients on enzyme-inducing anti-epileptic drugs (EIAEDs). Patients not on EIAEDs were initially treated at 250 mg; however, the dose was reduced to 170 mg because of intolerable side effects. Treatment was continued until unacceptable toxicity, tumor progression, or patient withdrawal. The primary endpoint was 6-month progression-free survival. Results: Forty-three patients were enrolled; 29 were not on EIAEDs. The expected toxicity profile of increased lipids, lymphopenia, and stomatitis was seen. There were no grade IV hematological toxicities and no toxic deaths. One patient was progression free at 6 months. Of the patients assessable for response, there were 2 partial responses and 20 with stabilization of disease. The median time to progression was 9 weeks. Conclusions: CCI-779 was well tolerated at this dose schedule; however, there was no evidence of efficacy in patients with recurrent GBM. Despite initial disease stabilization in approximately 50% of patients, the durability of response was short. Because of the low toxicity profile, CCI-779 may merit exploration in combination with other modalities.

Collaboration


Dive into the Michael D. Prados's collaboration.

Top Co-Authors

Avatar

Susan M. Chang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge