Network


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

Hotspot


Dive into the research topics where Matthew M. Cooney is active.

Publication


Featured researches published by Matthew M. Cooney.


The New England Journal of Medicine | 2015

Chemohormonal Therapy in Metastatic Hormone-Sensitive Prostate Cancer

Christopher Sweeney; Yu-Hui Chen; Michael A. Carducci; Glenn Liu; David F. Jarrard; Mario A. Eisenberger; Yu Ning Wong; Noah M. Hahn; Manish Kohli; Matthew M. Cooney; Robert Dreicer; Nicholas J. Vogelzang; Joel Picus; Daniel H. Shevrin; Maha Hussain; Jorge A. Garcia; Robert S. DiPaola

BACKGROUND Androgen-deprivation therapy (ADT) has been the backbone of treatment for metastatic prostate cancer since the 1940s. We assessed whether concomitant treatment with ADT plus docetaxel would result in longer overall survival than that with ADT alone. METHODS We assigned men with metastatic, hormone-sensitive prostate cancer to receive either ADT plus docetaxel (at a dose of 75 mg per square meter of body-surface area every 3 weeks for six cycles) or ADT alone. The primary objective was to test the hypothesis that the median overall survival would be 33.3% longer among patients receiving docetaxel added to ADT early during therapy than among patients receiving ADT alone. RESULTS A total of 790 patients (median age, 63 years) underwent randomization. After a median follow-up of 28.9 months, the median overall survival was 13.6 months longer with ADT plus docetaxel (combination therapy) than with ADT alone (57.6 months vs. 44.0 months; hazard ratio for death in the combination group, 0.61; 95% confidence interval [CI], 0.47 to 0.80; P<0.001). The median time to biochemical, symptomatic, or radiographic progression was 20.2 months in the combination group, as compared with 11.7 months in the ADT-alone group (hazard ratio, 0.61; 95% CI, 0.51 to 0.72; P<0.001). The rate of a prostate-specific antigen level of less than 0.2 ng per milliliter at 12 months was 27.7% in the combination group versus 16.8% in the ADT-alone group (P<0.001). In the combination group, the rate of grade 3 or 4 febrile neutropenia was 6.2%, the rate of grade 3 or 4 infection with neutropenia was 2.3%, and the rate of grade 3 sensory neuropathy and of grade 3 motor neuropathy was 0.5%. CONCLUSIONS Six cycles of docetaxel at the beginning of ADT for metastatic prostate cancer resulted in significantly longer overall survival than that with ADT alone. (Funded by the National Cancer Institute and others; ClinicalTrials.gov number, NCT00309985.).


Journal of Clinical Oncology | 2009

Phase II Study of Sorafenib in Patients With Metastatic or Recurrent Sarcomas

Robert G. Maki; David R. D'Adamo; Mary Louise Keohan; Michael Saulle; Scott M. Schuetze; Samir D. Undevia; Michael B. Livingston; Matthew M. Cooney; Martee L. Hensley; Monica M. Mita; Chris H. Takimoto; Andrew S. Kraft; Anthony Elias; Bruce Brockstein; Nathalie E. Blachère; Mark A. Edgar; Lawrence H. Schwartz; Li Xuan Qin; Cristina R. Antonescu; Gary K. Schwartz

PURPOSE Since activity of sorafenib was observed in sarcoma patients in a phase I study, we performed a multicenter phase II study of daily oral sorafenib in patients with recurrent or metastatic sarcoma. PATIENTS AND METHODS We employed a multiarm study design, each representing a sarcoma subtype with its own Simon optimal two-stage design. In each arm, 12 patients who received 0 to 1 prior lines of therapy were treated (0 to 3 for angiosarcoma and malignant peripheral-nerve sheath tumor). If at least one Response Evaluation Criteria in Solid Tumors (RECIST) was observed, 25 further patients with that sarcoma subtype were accrued. Results Between October 2005 and November 2007, 145 patients were treated; 144 were eligible for toxicity and 122 for response. Median age was 55 years; female-male ratio was 1.8:1. The median number of cycles was 3. Five of 37 patients with angiosarcoma had a partial response (response rate, 14%). This was the only arm to meet the RECIST response rate primary end point. Median progression-free survival was 3.2 months; median overall survival was 14.3 months. Adverse events (typically dermatological) necessitated dose reduction for 61% of patients. Statistical modeling in this limited patient cohort indicated sorafenib toxicity was correlated inversely to patient height. There was no correlation between phosphorylated extracellular signal regulated kinase expression and response in six patients with angiosarcoma with paired pre- and post-therapy biopsies. CONCLUSION As a single agent, sorafenib has activity against angiosarcoma and minimal activity against other sarcomas. Further evaluation of sorafenib in these and possibly other sarcoma subtypes appears warranted, presumably in combination with cytotoxic or kinase-specific agents.


The Lancet | 2016

Olaratumab and doxorubicin versus doxorubicin alone for treatment of soft-tissue sarcoma: an open-label phase 1b and randomised phase 2 trial

William D. Tap; Robin L. Jones; Brian A. Van Tine; Bartosz Chmielowski; Anthony Elias; Douglas Adkins; Mark Agulnik; Matthew M. Cooney; Michael B. Livingston; Gregory K. Pennock; Meera Hameed; Gaurav D. Shah; Amy Qin; Ashwin Shahir; Damien M. Cronier; Robert L. Ilaria; Ilaria Conti; Jan Cosaert; Gary K. Schwartz

BACKGROUND Treatment with doxorubicin is a present standard of care for patients with metastatic soft-tissue sarcoma and median overall survival for those treated is 12-16 months, but few, if any, novel treatments or chemotherapy combinations have been able to improve these poor outcomes. Olaratumab is a human antiplatelet-derived growth factor receptor α monoclonal antibody that has antitumour activity in human sarcoma xenografts. We aimed to assess the efficacy of olaratumab plus doxorubicin in patients with advanced or metastatic soft-tissue sarcoma. METHODS We did an open-label phase 1b and randomised phase 2 study of doxorubicin plus olaratumab treatment in patients with unresectable or metastatic soft-tissue sarcoma at 16 clinical sites in the USA. For both the phase 1b and phase 2 parts of the study, eligible patients were aged 18 years or older and had a histologically confirmed diagnosis of locally advanced or metastatic soft-tissue sarcoma not previously treated with an anthracycline, an Eastern Cooperative Oncology Group (ECOG) performance status of 0-2, and available tumour tissue to determine PDGFRα expression by immunohistochemistry. In the phase 2 part of the study, patients were randomly assigned in a 1:1 ratio to receive either olaratumab (15 mg/kg) intravenously on day 1 and day 8 plus doxorubicin (75 mg/m(2)) or doxorubicin alone (75 mg/m(2)) on day 1 of each 21-day cycle for up to eight cycles. Randomisation was dynamic and used the minimisation randomisation technique. The phase 1b primary endpoint was safety and the phase 2 primary endpoint was progression-free survival using a two-sided α level of 0.2 and statistical power of 0.8. This study was registered with ClinicalTrials.gov, number NCT01185964. FINDINGS 15 patients were enrolled and treated with olaratumab plus doxorubicin in the phase 1b study, and 133 patients were randomised (66 to olaratumab plus doxorubicin; 67 to doxorubicin alone) in the phase 2 trial, 129 (97%) of whom received at least one dose of study treatment (64 received olaratumab plus doxorubicin, 65 received doxorubicin). Median progression-free survival in phase 2 was 6.6 months (95% CI 4.1-8.3) with olaratumab plus doxorubicin and 4.1 months (2.8-5.4) with doxorubicin (stratified hazard ratio [HR] 0.67; 0.44-1.02, p=0.0615). Median overall survival was 26.5 months (20.9-31.7) with olaratumab plus doxorubicin and 14.7 months (9.2-17.1) with doxorubicin (stratified HR 0.46, 0.30-0.71, p=0.0003). The objective response rate was 18.2% (9.8-29.6) with olaratumab plus doxorubicin and 11.9% (5.3-22.2) with doxorubicin (p=0.3421). Steady state olaratumab serum concentrations were reached during cycle 3 with mean maximum and trough concentrations ranging from 419 μg/mL (geometric coefficient of variation in percentage [CV%] 26.2) to 487 μg/mL (CV% 33.0) and from 123 μg/mL (CV% 31.2) to 156 μg/mL (CV% 38.0), respectively. Adverse events that were more frequent with olaratumab plus doxorubicin versus doxorubicin alone included neutropenia (37 [58%] vs 23 [35%]), mucositis (34 [53%] vs 23 [35%]), nausea (47 [73%] vs 34 [52%]), vomiting (29 [45%] vs 12 [18%]), and diarrhoea (22 [34%] vs 15 [23%]). Febrile neutropenia of grade 3 or higher was similar in both groups (olaratumab plus doxorubicin: eight [13%] of 64 patients vs doxorubicin: nine [14%] of 65 patients). INTERPRETATION This study of olaratumab with doxorubicin in patients with advanced soft-tissue sarcoma met its predefined primary endpoint for progression-free survival and achieved a highly significant improvement of 11.8 months in median overall survival, suggesting a potential shift in the treatment of soft-tissue sarcoma. FUNDING Eli Lilly and Company.


Thyroid | 2009

A Phase II Trial of Fosbretabulin in Advanced Anaplastic Thyroid Carcinoma and Correlation of Baseline Serum-Soluble Intracellular Adhesion Molecule-1 with Outcome

Colin J. Mooney; Govardhanan Nagaiah; Pingfu Fu; Jay Wasman; Matthew M. Cooney; Panos S. Savvides; Joseph A. Bokar; Afshin Dowlati; Ding Wang; Sanjiv S. Agarwala; Susan M. Flick; Paul Hartman; José Ortiz; Pierre Lavertu; Scot C. Remick

BACKGROUND Fosbretabulin is a novel vascular-disrupting agent that has antitumor activity against anaplastic thyroid cancer (ATC) cell lines, xenografts, and demonstrable efficacy in a phase I trial. This phase II study determined the efficacy and safety of fosbretabulin in patients with advanced ATC and whether fosbretabulin altered the natural history of ATC by virtue of doubling the median survival. A secondary aim evaluated the prognostic value of serum soluble intracellular adhesion molecule-1 (sICAM). METHODS Twenty-six patients received fosbretabulin 45 mg/m(2) as a 10-minute intravenous infusion on days 1, 8, and 15 of a 28-day cycle. sICAM levels were obtained at baseline, over the first two cycles, and end of therapy. Treatment was continued until disease progression. RESULTS Fosbretabulin was well tolerated; grade 3 toxicity was observed in nine patients (35%), and grade 4 toxicity in one (4%). QTc prolongation delayed treatment in four causing one to stop treatment. Median survival was 4.7 months with 34% and 23% alive at 6 and 12 months, respectively. Median duration of stable disease in seven patients was 12.3 months (range, 4.4-37.9 months). Baseline serum sICAM levels were measured in 24 patients with a median 253.5 ng/mL. There was a significant difference in event-free survival among tertiles of baseline sICAM levels (p < 0.009). CONCLUSIONS There were no objective responses seen with single-agent fosbretabulin as administered in this trial, and we did not observe a doubling of survival as our primary endpoint. This is among the largest prospective trials ever conducted for ATC. Fosbretabulin has an acceptable safety profile in patients with advanced ATC, and one-third survived more than 6 months. Despite a small sample size, low baseline sICAM levels were predictive of event-free survival. Further prospective validation of sICAM as a therapeutic biomarker and exploring combination regimens with fosbretabulin are warranted.


Clinical Cancer Research | 2004

Cardiovascular Safety Profile of Combretastatin A4 Phosphate in a Single-Dose Phase I Study in Patients with Advanced Cancer

Matthew M. Cooney; Tomas Radivoyevitch; Afshin Dowlati; Beth Overmoyer; Nathan Levitan; Kelly Robertson; Sandra L. Levine; Kathleen R. DeCaro; Carol Buchter; Anne L. Taylor; Bruce S. Stambler; Scot C. Remick

Purpose: The purpose of our study was to review and determine the cardiovascular safety profile of combretastatin A4 phosphate (CA4P) in a Phase I study in 25 patients with advanced solid tumors. Experimental Design: CA4P was administered in a dose-escalating fashion starting at 18 mg/m2 i.v. every 21 days, and the maximal dosage was 90 mg/m2. Continuous evaluation included bedside blood pressure and pulse monitoring, 12-lead electrocardiogram (ECG) at fixed time points for measured QT interval determination, determination of the corrected QT interval (QTc) using Bazett’s formula QTc = QT/(R-R interval)1/2, and chart review. Pharmacodynamic correlations of CA4P dose, CA4P/CA4 area under the curve, and Cmax versus heart rate (HR), blood pressure, QT, and QTc intervals, over the first 4 h postdosing were analyzed. Results: After CA4P administration, there were significant increases in QTc interval at the 3-h and 4-h time points [27.2 ms (P < 0.0001) and 30.8 ms (P < 0.0001), respectively] and HR at the 3- and 4-h time points [13.2 beats per minute (bpm; P < 0.01) and 15.1 bpm (P < 0.001), respectively]. Three of 25 patients had prolonged QTc intervals at baseline, whereas 15 (60%) of 25 and 18 (75%) of 24 patients had prolonged QTc intervals at 3 and 4 h. The slope of HR and QTc increasing as a function of time during the first 4 h was correlated to dose (in milligrams) of CA4P (P = 0.01 and r = 0.49 for HR, P = 0.005 and r = 0.55 for QTc) and to CA4 area under the curve (P = 0.04 and r = 0.41 for HR, P = 0.02 and r = 0.44 for QTc); blood pressure and uncorrected QTc interval dose-response correlations were not significant. Two patients had ECG changes consistent with an acute coronary syndrome within 24 h of CA4P infusion. Conclusions: CA4P prolongs the QTc interval. There was a temporal relationship with the CA4P infusion and with ECG changes consistent with an acute coronary syndrome in two patients. It is advisable that future trials with CA4P have eligibility guidelines limiting patients with known coronary artery disease or those with multiple coronary artery disease risk factors until more experience is gained regarding potential cardiovascular toxicity with this agent.


Journal of Clinical Oncology | 2007

Hypertension, Proteinuria, and Antagonism of Vascular Endothelial Growth Factor Signaling: Clinical Toxicity, Therapeutic Target, or Novel Biomarker?

Willem J. van Heeckeren; Jose Ortiz; Matthew M. Cooney; Scot C. Remick

There are three US Food and Drug Administration–approved angiogenesis inhibitors for the treatment of cancer that specifically target vascular endothelial growth factor (VEGF) signaling. Bevacizumab (monoclonal antibody to VEGF) has been shown to confer a survival advantage when used in combination with cytotoxic chemotherapy in patients with colorectal cancer and non– small-cell lung cancer. 1,2 Sorafenib and sunitinib are orally bioavailable, small-molecule tyrosine kinase inhibitors that target the intracellular tyrosine kinase domain of the VEGF receptor (VEGFR) among other tyrosine kinase targets. Sorafenib has been shown to increase progression-free survival in patients with renal cell carcinoma. 3 Sunitinib has been shown to increase progressionfree survival in patients with renal cell carcinoma and GI stromal tumors. 4-6 A plethora of new small-molecule tyrosine kinase inhibitors are in preclinical development and early-phase clinical trials that target VEGFR with varying degrees of specificity, including AZD2171, which is the subject of a published report by Drevs et al


Clinical Cancer Research | 2009

A Phase I Study of Sunitinib plus Bevacizumab in Advanced Solid Tumors

Brian I. Rini; Jorge A. Garcia; Matthew M. Cooney; Paul Elson; Allison Janine Tyler; Kristi Beatty; Joseph A. Bokar; Tarek Mekhail; Ronald M. Bukowski; G. Thomas Budd; Pierre L. Triozzi; Ernest C. Borden; Percy Ivy; Helen X. Chen; Afshin Dolwati; Robert Dreicer

Purpose: Bevacizumab is an antibody against vascular endothelial growth factor; sunitinib is an inhibitor of vascular endothelial growth factor and related receptors. The safety and maximum tolerated dose of sunitinib plus bevacizumab was assessed in this phase I trial. Experimental Design: Patients with advanced solid tumors were treated on a 3+3 trial design. Patients received sunitinib daily (starting dose level, 25 mg) for 4 weeks on followed by 2 weeks off and bevacizumab (starting dose level, 5 mg/kg) on days 1, 15, and 29 of a 42-day cycle. Dose-limiting toxicities during the first 6-week cycle were used to determine the maximum tolerated dose. Results: Thirty-eight patients were enrolled. Patients received a median of 3 cycles of treatment (range, 1-17+). There was one dose-limiting toxicity (grade 4 hypertension) at 37.5 mg sunitinib and 5 mg/kg bevacizumab. Grade 3 or greater toxicity was observed in 87% of patients including hypertension (47%), fatigue (24%), thrombocytopenia (18%), proteinuria (13%), and hand-foot syndrome (13%). Dose modifications and delays were common at higher dose levels. No clinical or laboratory evidence of microangiopathic hemolytic anemia was observed. Seven patients had a confirmed Response Evaluation Criteria in Solid Tumors–defined partial response (18%; 95% confidence interval, 8-34%). Nineteen of the 32 patients with a postbaseline scan (59%) had at least some reduction in overall tumor burden (median, 32%; range, 3-73%). Conclusions: The combination of sunitinib and bevacizumab in patients with advanced solid tumors is feasible, albeit with toxicity at higher dose levels and requiring dose modification with continued therapy. Antitumor activity was observed across multiple solid tumors. (Clin Cancer Res 2009;15(19):6277–83)


Journal of Clinical Oncology | 2006

Promise of New Vascular-Disrupting Agents Balanced With Cardiac Toxicity: Is It Time for Oncologists to Get to Know Their Cardiologists?

Willem J. van Heeckeren; Shyam Bhakta; Jose Ortiz; Jeff Duerk; Matthew M. Cooney; Afshin Dowlati; Keith R. McCrae; Scot C. Remick

Willem J. van Heeckeren, Division of Hematology/Oncology, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH Shyam Bhakta and Jose Ortiz, Division of Cardiology, Department of Medicine, CASE School of Medicine, Cleveland, OH Jeff Duerk, Departments of Radiology and Biomedical Engineering, CASE School of Medicine, Cleveland, OH Matthew M. Cooney, Afshin Dowlati, Keith McCrae, and Scot C. Remick, Division of Hematology/Oncology, Department of Medicine, Case Western Reserve University School of Medicine, Developmental Therapeutics Program, CASE Comprehensive Cancer Center, University Hospitals of Cleveland, Cleveland, OH


Journal of Clinical Oncology | 2010

Toxicity of Sunitinib Plus Bevacizumab in Renal Cell Carcinoma

Brian I. Rini; Jorge A. Garcia; Matthew M. Cooney; Paul Elson; Allison Janine Tyler; Kristi Beatty; Joseph A. Bokar; Percy Ivy; Helen X. Chen; Afshin Dowlati; Robert Dreicer

TO THE EDITOR: Inhibition of vascular endothelial growth factor (VEGF), either via antibody-mediated binding of the ligand or small molecule inhibition of the VEGF receptor, has demonstrated clinically relevant benefits in metastatic renal cell carcinoma (RCC) and other solid tumors. Recently, combinations of VEGFinhibiting approaches have been studied in an attempt to maximize VEGF blockade and extend the clinical benefit of these approaches. Sunitinib (Sutent; Pfizer Inc, New York, NY), a small-molecule tyrosine kinase inhibitor of a family of receptors including VEGF receptor and bevacizumab (Avastin; Genentech, South San Francisco, CA), a recombinant humanized monoclonal antibody that binds and neutralizes circulating VEGF, have been previously combined in two separate phase I trials. A phase I trial conducted exclusively in metastatic renal cell carcinoma (RCC) and with fixed bevacizumab dosing at 10 mg/kg intravenously every 2 weeks reported unacceptable toxicity, including thrombotic microangiopathy (TMA). We previously reported a similar phase I trial of this combination in 38 patients with advanced solid tumors, notable for the diverse tumor type as well as lower initial doses of bevacizumab. This trial failed to demonstrate laboratory or clinical evidence of TMA in any patent, including the six RCC patients (median, 8 cycles of therapy; range, 2 to 13), all of whom had undergone nephrectomy, and including all patients treated at the highest dose levels up to 20 months of treatment. Due to increased toxicity seen with chronic dosing at higher dose levels in our study and the TMA seen in the other trial, an expanded cohort of patients with RCC was planned to collect more safety experience at dosing of 37.5 mg sunitinib 4 weeks on followed by 2 weeks off and bevacizumab 5 mg/kg intravenously on days 1, 15, and 29 of a 42-day cycle. Five patients were enrolled in this expanded cohort. Three of five patients demonstrated laboratory evidence of TMA. This included low haptoglobin, thrombocytopenia, and schistocytes on peripheral blood smear at the end of cycles 2 and 4 for two patients, and an isolated single low haptoglobin value for the third patient. None of the patients had any clinical signs or symptoms associated with the laboratory abnormalities. Table 1 depicts the patient characteristics and relevant data for the three cases of TMA and the remainder of the patients with RCC enrolled on the initial study and expanded cohort. The first two TMA patients continued to have haptoglobin levels lower than 20 mg/dL for 2 and 20 weeks after discontinuation of bevacizumab (both patients continued on sunitinib monotherapy). The third patient developed the isolated low haptoglobin while receiving sunitinib monotherapy after bevacizumab had been discontinued 8 weeks prior. The haptoglobin normalized 2 weeks later and remained in normal range with continued sunitinib monotherapy for this patient. All five patients on the expanded cohort discontinued bevacizumab when TMA was observed in the initial two patients. Further accrual of patients with RCC was suspended due to this safety signal.


Current Opinion in Hematology | 2007

Complications from vascular disrupting agents and angiogenesis inhibitors : aberrant control of hemostasis and thrombosis

Willem J. van Heeckeren; Sharon L. Sanborn; Ajita Narayan; Matthew M. Cooney; Keith R. McCrae; Alvin H. Schmaier; Scot C. Remick

Purpose of reviewTo discuss thrombotic and hemorrhagic complications from angiogenesis inhibitors and vascular disrupting agents, pathogenesis, and recommendations for prophylaxis and management of those complications. Recent findingsVenous thromboembolism has been a significant complication of the angiogenesis inhibitors thalidomide and lenalidomide. Prophylaxis with aspirin, low-molecular-weight heparin, or warfarin has been shown to decrease rates of venous thromboembolism in patients treated with these agents. Life-threatening hemorrhage and arterial thromboembolism have been observed in patients using treatments that inhibit the vascular endothelial growth factor signaling pathway. Patients should be screened for arterial thromboembolism and hemorrhage risk prior to using vascular endothelial growth factor signal inhibitors. It is not known how angiogenesis inhibitors and vascular disrupting agents upset normal hemostasis. It is likely that disruption of the function and/or integrity of vascular endothelium leads to an increased risk for thrombosis and/or hemorrhage. SummaryNew angiogenesis inhibitors and vascular disrupting agents have been developed that have significant activity against neoplasms. Potentially life-threatening side effects of hemorrhage and thrombosis have been observed with many of these new agents. As new treatments that disrupt angiogenesis or existing tumor vasculature are developed, attention should be given to these toxicities in clinical practice and clinical trials.

Collaboration


Dive into the Matthew M. Cooney's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Afshin Dowlati

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Scot C. Remick

West Virginia University

View shared research outputs
Top Co-Authors

Avatar

Joseph A. Bokar

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Smitha S. Krishnamurthi

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Joanna M. Brell

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joel Picus

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge