Network


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

Hotspot


Dive into the research topics where Corey A. Carter is active.

Publication


Featured researches published by Corey A. Carter.


Journal of Clinical Oncology | 2015

Molecular Profiling and Targeted Therapy for Advanced Thoracic Malignancies: A Biomarker-Derived, Multiarm, Multihistology Phase II Basket Trial

Ariel Lopez-Chavez; Anish Thomas; Arun Rajan; Mark Raffeld; Betsy Morrow; Ronan J. Kelly; Corey A. Carter; Udayan Guha; Keith Killian; Christopher Lau; Zied Abdullaev; Liqiang Xi; Svetlana Pack; Paul S. Meltzer; Christopher L. Corless; Alan Sandler; Carol Beadling; Andrea Warrick; David J. Liewehr; Seth M. Steinberg; Arlene Berman; Austin Doyle; Eva Szabo; Yisong Wang; Giuseppe Giaccone

PURPOSE We conducted a basket clinical trial to assess the feasibility of such a design strategy and to independently evaluate the effects of multiple targeted agents against specific molecular aberrations in multiple histologic subtypes concurrently. PATIENTS AND METHODS We enrolled patients with advanced non-small-cell lung cancer (NSCLC), small-cell lung cancer, and thymic malignancies who underwent genomic characterization of oncogenic drivers. Patients were enrolled onto a not-otherwise-specified arm and treated with standard-of-care therapies or one of the following five biomarker-matched treatment groups: erlotinib for EGFR mutations; selumetinib for KRAS, NRAS, HRAS, or BRAF mutations; MK2206 for PIK3CA, AKT, or PTEN mutations; lapatinib for ERBB2 mutations or amplifications; and sunitinib for KIT or PDGFRA mutations or amplification. RESULTS Six hundred forty-seven patients were enrolled, and 88% had their tumors tested for at least one gene. EGFR mutation frequency was 22.1% in NSCLC, and erlotinib achieved a response rate of 60% (95% CI, 32.3% to 83.7%). KRAS mutation frequency was 24.9% in NSCLC, and selumetinib failed to achieve its primary end point, with a response rate of 11% (95% CI, 0% to 48%). Completion of accrual to all other arms was not feasible. In NSCLC, patients with EGFR mutations had the longest median survival (3.51 years; 95% CI, 2.89 to 5.5 years), followed by those with ALK rearrangements (2.94 years; 95% CI, 1.66 to 4.61 years), those with KRAS mutations (2.3 years; 95% CI, 2.3 to 2.17 years), those with other genetic abnormalities (2.17 years; 95% CI, 1.3 to 2.74 years), and those without an actionable mutation (1.85 years; 95% CI, 1.61 to 2.13 years). CONCLUSION This basket trial design was not feasible for many of the arms with rare mutations, but it allowed the study of the genetics of less common malignancies.


Clinical Cancer Research | 2012

A Phase I Combination Study of Olaparib with Cisplatin and Gemcitabine in Adults with Solid Tumors

Arun Rajan; Corey A. Carter; Ronan J. Kelly; Martin Gutierrez; Shivaani Kummar; Eva Szabo; Mary Ann Yancey; Jiuping Ji; Baskar Mannargudi; Sukyung Woo; Shawn D. Spencer; William D. Figg; Giuseppe Giaccone

Purpose: To determine the safety and tolerability of olaparib with cisplatin and gemcitabine, establish the maximum tolerated dose (MTD), and evaluate the pharmacodynamic and pharmacokinetic profile of the combination. Experimental Design: We conducted a phase I study of olaparib with cisplatin and gemcitabine in patients with advanced solid tumors. Treatment at dose level 1 (DL1) consisted of olaparib 100 mg orally every 12 hours on days 1 to 4, gemcitabine 500 mg/m2 on days 3 and 10, and cisplatin 60 mg/m2 on day 3. PAR levels were measured in peripheral blood mononuclear cells (PBMC). Results: Dose-limiting toxicities (DLT) in two of three patients at DL1 included thrombocytopenia and febrile neutropenia. The protocol was amended to enroll patients treated with ≤2 prior severely myelosuppressive chemotherapy regimens and treated with olaparib 100 mg once daily on days 1 to 4 (DL−1). No DLTs were seen in six patients at DL−1. Because of persistent thrombocytopenia and neutropenia following a return to DL1, patients received 100 mg olaparib every 12 hours on day 1 only. No hematologic DLTs were observed; nonhematologic DLTs included gastrointestinal bleed, syncope, and hypoxia. Of 21 patients evaluable for response, two had partial response. Olaparib inhibited PARP in PBMCs and tumor tissue, although PAR levels were less effectively inhibited when olaparib was used for a short duration. Conclusions: Olaparib in combination with cisplatin and gemcitabine is associated with myelosuppression even at relatively low doses. Modified schedules of olaparib in chemotherapy naive patients will have to be explored with standard doses of chemotherapy. Clin Cancer Res; 18(8); 2344–51. ©2012 AACR.


Journal of Clinical Oncology | 2003

Temporarily Deferred Therapy (watchful waiting) for Men Younger Than 70 Years and With Low-Risk Localized Prostate Cancer in the Prostate-Specific Antigen Era

Corey A. Carter; Timothy F. Donahue; Leon Sun; Hongyu Wu; David G. McLeod; Christopher L. Amling; Raymond S. Lance; John P. Foley; Wade J. Sexton; Leo Kusuda; Andrew Chung; Douglas W. Soderdahl; Stephen Jackman; Judd W. Moul

Purpose: Watchful waiting (WW) is an acceptable strategy for managing prostate cancer (PC) in older men. Prostate-specific antigen (PSA) testing has resulted in a stage migration, with diagnoses made in younger men. An analysis of the Department of Defense Center for Prostate Disease Research Database was undertaken to document younger men with low- or intermediate-grade PC who initially chose WW. Patients and Methods: We identified men choosing WW who were diagnosed between January 1991 and January 2002, were 70 years or younger, had a Gleason score ≤ 6 with no Gleason pattern 4, had no more than three positive cores on biopsy, and whose clinical stage was ≤ T2 and PSA level was ≤ 20. We analyzed their likelihood of remaining on WW, the factors associated with secondary treatment, and the influence of comorbidities. Results: Three hundred thirteen men were identified. Median follow-up time was 3.8 years. Median age was65.4 years (range, 41 to 70 years). Ninety-eight patients remained on WW; 215 proceeded...


Lancet Oncology | 2015

Sunitinib in patients with chemotherapy-refractory thymoma and thymic carcinoma: an open-label phase 2 trial

Anish Thomas; Arun Rajan; Arlene Berman; Yusuke Tomita; Christina Brzezniak; Min Jung Lee; Sunmin Lee; Alexander Ling; Aaron John Spittler; Corey A. Carter; Udayan Guha; Yisong Wang; Eva Szabo; Paul S. Meltzer; Seth M. Steinberg; Jane B. Trepel; Patrick J. Loehrer; Giuseppe Giaccone

BACKGROUND No standard treatments are available for advanced thymic epithelial tumours after failure of platinum-based chemotherapy. We investigated the activity of sunitinib, an orally administered tyrosine kinase inhibitor. METHODS Between May 15, 2012, and Oct 2, 2013, we did an open-label phase 2 trial in patients with histologically confirmed chemotherapy-refractory thymic epithelial tumours. Patients were eligible if they had disease progression after at least one previous regimen of platinum-containing chemotherapy, an Eastern Cooperative Oncology Group performance status of two or lower, measurable disease, and adequate organ function. Patients received 50 mg of sunitinib orally once a day, in 6-week cycles (ie, 4 weeks of treatment followed by 2 weeks without treatment), until tumour progression or unacceptable toxic effects arose. The primary endpoint was investigator-assessed best tumour response at any point, which we analysed separately in thymoma and thymic carcinoma cohorts. Patients who had received at least one cycle of treatment and had their disease reassessed were included in the analyses of response. The trial was registered with ClinicalTrials.gov, number NCT01621568. FINDINGS 41 patients were enrolled, 25 with thymic carcinoma and 16 with thymoma. One patient with thymic carcinoma was deemed ineligible after enrolment and did not receive protocol treatment. Of patients who received treatment, one individual with thymic carcinoma was not assessable because she died. Median follow-up on trial was 17 months (IQR 14.0-18.4). Of 23 assessable patients with thymic carcinoma, six (26%, 90% CI 12.1-45.3, 95% CI 10.2-48.4) had partial responses, 15 (65%, 95% CI 42.7-83.6) achieved stable disease, and two (9%, 1.1-28.0) had progressive disease. Of 16 patients with thymoma, one (6%, 95% CI 0.2-30.2) had a partial response, 12 (75%, 47.6-92.7) had stable disease, and three (19%, 4.1-45.7) had progressive disease. The most common grade 3 and 4 treatment-related adverse events were lymphocytopenia (eight [20%] of 40 patients), fatigue (eight [20%]), and oral mucositis (eight [20%]). Five (13%) patients had decreases in left-ventricular ejection fraction, of which three (8%) were grade 3 events. Three (8%) patients died during treatment, including one individual who died of cardiac arrest that was possibly treatment-related. INTERPRETATION Sunitinib is active in previously treated patients with thymic carcinoma. Further studies are needed to identify potential biomarkers of activity. FUNDING National Cancer Institute (Cancer Therapy Evaluation Program).


Annals of Oncology | 2013

A phase I/II study of sepantronium bromide (YM155, survivin suppressor) with paclitaxel and carboplatin in patients with advanced non-small-cell lung cancer

Ronan J. Kelly; A. Thomas; Arun Rajan; G. Chun; A. Lopez-Chavez; Eva Szabo; Shawn D. Spencer; Corey A. Carter; U. Guha; S. Khozin; S. Poondru; C. van Sant; Anne Keating; Seth M. Steinberg; William D. Figg; Giuseppe Giaccone

BACKGROUND This phase I/II study examined the safety and efficacy of Sepantronium Bromide (S), a small-molecule selective survivin suppressant, administered in combination with carboplatin (C) and paclitaxel (P). PATIENTS AND METHODS Forty-one patients were treated on study. Twenty-two patients received escalating doses of S (3.6-12 mg/m(2)) and 19 with untreated stage IV non-small-cell lung cancer (NSCLC) were treated with the maximum tolerated dose of 10 mg/m(2) in combination with standard doses of C (AUC6) and P (200 mg/m(2)) for six cycles. S was administered as a continuous intravenous infusion (CIVI) over 72 h in 21-day treatment cycles. Study end points included safety and toxic effect, response rate, progression-free and overall survival (PFS and OS), as well as exploratory pharmacodynamic correlates. RESULTS Treatment with S was well tolerated, and toxic effects were mostly hematological in the phase II study. Two (11%) partial responses were observed with a median PFS of 5.7 months and median OS 16.1 months. Pharmacodynamic analysis did not demonstrate an association with response. CONCLUSION The combination of S (10 mg/m(2)/day 72-h CIVI) administered with C and P every 3 weeks exhibited a favorable safety profile but failed to demonstrate an improvement in response rate in advanced NSCLC. CLINICAL TRIAL NUMBER NCT01100931.


Lancet Oncology | 2014

Cixutumumab for patients with recurrent or refractory advanced thymic epithelial tumours: a multicentre, open-label, phase 2 trial.

Arun Rajan; Corey A. Carter; Arlene Berman; Liang Cao; Ronan J. Kelly; Anish Thomas; Sean Khozin; Ariel Lopez Chavez; Isabella Bergagnini; Barbara Scepura; Eva Szabo; Min Jung Lee; Jane B. Trepel; Sarah K. Browne; Lindsey B. Rosen; Yunkai Yu; Seth M. Steinberg; Helen X. Chen; Gregory J. Riely; Giuseppe Giaccone

BACKGROUND No standard treatment exists for refractory or relapsed advanced thymic epithelial tumours. We investigated the efficacy of cixutumumab, a fully human IgG1 monoclonal antibody targeting the insulin-like growth factor 1 receptor in thymic epithelial tumours after failure of previous chemotherapy. METHODS Between Aug 25, 2009, and March 27, 2012, we did a multicentre, open-label, phase 2 trial in patients aged 18 years or older with histologically confirmed recurrent or refractory thymic epithelial tumours. We enrolled individuals who had progressed after at least one previous regimen of platinum-containing chemotherapy, had an Eastern Cooperative Oncology Group performance status of 0 or 1, and had measurable disease and adequate organ function. Eligible patients received intravenous cixutumumab (20 mg/kg) every 3 weeks until disease progression or development of intolerable toxic effects. The primary endpoint was the frequency of response, analysed on an intention-to-treat basis. We also did pharmacodynamic studies. This trial is registered with ClinicalTrials.gov, number NCT00965250. FINDINGS 49 patients were enrolled (37 with thymomas and 12 with thymic carcinomas) who received a median of eight cycles of cixutumumab (range 1-46). At the final actuarial analysis when follow-up data were updated (Nov 30, 2012), median potential follow-up (from on-study date to most current follow-up date) was 24·0 months (IQR 17·3-36·9). In the thymoma cohort, five (14%) of 37 patients (95% CI 5-29) achieved a partial response, 28 had stable disease, and four had progressive disease. In the thymic carcinoma cohort, none of 12 patients (95% CI 0-26) had a partial response, five had stable disease, and seven had progressive disease. The most common grade 3-4 adverse events in both cohorts combined were hyperglycaemia (five [10%]), lipase elevation (three [6%]), and weight loss, tumour pain, and hyperuricaemia (two each [4%]). Nine (24%) of 37 patients with thymoma developed autoimmune conditions during treatment (five were new-onset disorders), the most common of which was pure red-cell aplasia. Two (4%) patients died; one was attributed to disease progression and the other to disease-related complications (respiratory failure, myositis, and an acute coronary event), which could have been precipitated by treatment with cixutumumab. INTERPRETATION Cixutumumab monotherapy is well-tolerated and active in relapsed thymoma. Development of autoimmunity during treatment needs further investigation. FUNDING Division of Cancer Treatment and Diagnosis at the National Cancer Institute (National Institutes of Health), ImClone Systems.


Journal of Clinical Oncology | 2010

Personalizing Therapy in an Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitor–Resistant Non–Small-Cell Lung Cancer Using PF-00299804 and Trastuzumab

Ronan J. Kelly; Corey A. Carter; Giuseppe Giaccone

A 50-year-old African American male never-smoker was diagnosed with stage IV non–small-cell lung cancer (NSCLC) in November 2007. His baseline computed tomography (CT) demonstrated bilateral lung nodules and biopsy confirmed a poorly differentiated tumor favoring large-cell carcinoma staining positive for cytokeratin 7 and thyroid transcription factor-1. He was originally treated with eight cycles of standard first-line chemotherapy consisting of carboplatin, paclitaxel, and bevacizumab from January 2008 to May 2008, achieving a partial response. Unfortunately, disease progression was documented in June 2008, and the patient commenced second-line erlotinib. Tumor analysis at that time demonstrated immunohistochemistry (IHC) 3 for epidermal growth factor receptor (EGFR) protein overexpression and fluorescent in situ hybridization (FISH) revealed a nonamplified but high polysomy count of 4 EGFR copies in 40% of cells (Fig 1). Erlotinib was discontinued in July 2008 due to disease progression after only 6 weeks of therapy. He presented to the National Cancer Institute in October 2008 for enrollment in a clinical trial involving a second generation irreversible pan–human epidermal growth factor receptor (HER) tyrosine kinase inhibitor (TKI; PF-00299804). He met all the eligibility criteria and had an excellent Eastern Cooperative Oncology Group (ECOG) performance status of 1. Molecular analysis revealed a K-Ras wild type, and HER2 (IHC 2 and FISH)–positive tumor (Table 1, Fig 1). No EGFR or HER2 mutations were detected. He was commenced on PF-00299804 in December 2008 and had a partial response (70% measurable response on CT scan) after 4 weeks of 45 mg orally once daily with 21 days per cycle (Fig 2). Of particular interest was a notable reduction in the patient’s soluble extracellular domain HER2 levels (Fig 3). The patient subsequently progressed after five cycles of PF00299804 and was taken off study in April 2009. Radiological progression also correlated with a rise in serum HER2 levels (Fig 3). Based on the tumors molecular pattern (Table 1) and his excellent performance status the patient was started on fourth-line single agent trastuzumab in June 2009. After 6 weeks of weekly therapy, vinorelbine was added to trastuzumab (August 2009) after radiological progression on the targeted agent. After an additional 6 weeks of


Lancet Oncology | 2015

Safety and activity of RRx-001 in patients with advanced cancer: a first-in-human, open-label, dose-escalation phase 1 study

Tony Reid; Bryan Oronsky; Jan Scicinski; Curt L Scribner; Susan J. Knox; Shoucheng Ning; Donna M. Peehl; Ron Korn; Meaghan Stirn; Corey A. Carter; Arnold Oronsky; Michael J Taylor; William L. Fitch; Pedro Cabrales; Michelle M. Kim; Howard A. Burris; Christopher D. Lao; Nacer D. Abrouk; Gary Fanger; Jeffrey R. Infante

BACKGROUND Epigenetic alterations have been strongly associated with tumour formation and resistance to chemotherapeutic drugs, and epigenetic modifications are an attractive target in cancer research. RRx-001 is activated by hypoxia and induces the generation of reactive oxygen and nitrogen species that can epigenetically modulate DNA methylation, histone deacetylation, and lysine demethylation. The aim of this phase 1 study was to assess the safety, tolerability, and pharmacokinetics of RRx-001. METHODS In this open-label, dose-escalation, phase 1 study, we recruited adult patients (aged >18 years) with histologically or cytologically confirmed diagnosis of advanced, malignant, incurable solid tumours from University of California at San Diego, CA, USA, and Sarah Cannon Research Institute, Nashville, TN, USA. Key eligibility criteria included evaluable disease, Eastern Cooperative Group performance status of 2 or less, an estimated life expectancy of at least 12 weeks, adequate laboratory parameters, discontinuation of all previous antineoplastic therapies at least 6 weeks before intervention, and no residual side-effects from previous therapies. Patients were assigned to receive intravenous infusions of RRx-001 at increasing doses (10 mg/m(2), 16·7 mg/m(2), 24·6 mg/m(2), 33 mg/m(2), 55 mg/m(2), and 83 mg/m(2)) either once or twice-weekly for at least 4 weeks, with at least three patients per dose cohort and allowing a 2-week observation period before dose escalation. Samples for safety and pharmacokinetics analysis, including standard chemistry and haematological panels, were taken on each treatment day. The primary objective was to assess safety, tolerability, and dose-limiting toxic effects of RRx-001, to determine single-dose pharmacokinetics, and to identify a recommended dose for phase 2 trials. All analyses were done per protocol. Accrual is complete and follow-up is still on-going. This trial is registered with ClinicalTrials.gov, number NCT01359982. FINDINGS Between Oct 10, 2011, and March 18, 2013, we enrolled 25 patients and treated six patients in the 10 mg/m(2) cohort, three patients in the 16·7 mg/m(2) cohort, three patients in the 24·6 mg/m(2) cohort, four patients in the 33 mg/m(2) cohort, three patients in the 55 mg/m(2), and six patients in the 83 mg/m(2) cohort. Pain at the injection site, mostly grade 1 and grade 2, was the most common adverse event related to treatment, experienced by 21 (84%) patients. Other common drug-related adverse events included arm swelling or oedema (eight [32%] patients), and vein hardening (seven [28%] patients). No dose-limiting toxicities were observed. Time constraints related to management of infusion pain from RRx-001 resulted in a maximally feasible dose of 83 mg/m(2). Of the 21 evaluable patients, one (5%) patient had a partial response, 14 (67%) patients had stable disease, and six (29%) patients had progressive disease; all responses were across a variety of tumour types. Four patients who had received RRx-001 were subsequently rechallenged with a treatment that they had become refractory to; all four responded to the rechallenge. INTERPRETATION RRx-001 is a well-tolerated novel compound without clinically significant toxic effects at the tested doses. Preliminary evidence of activity is promising and, on the basis of all findings, a dose of 16·7 mg/m(2) was recommended as the targeted dose for phase 2 trials. FUNDING EpicentRx (formerly RadioRx).


Clinical Cancer Research | 2014

A Phase I/II Trial of Belinostat in Combination with Cisplatin, Doxorubicin and Cyclophosphamide in Thymic Epithelial Tumors: A Clinical And Translational Study

Anish Thomas; Arun Rajan; Eva Szabo; Yusuke Tomita; Corey A. Carter; Barbara Scepura; Ariel Lopez-Chavez; Min-Jung Lee; Christophe E. Redon; Ari Frosch; Cody J. Peer; Yuanbin Chen; Richard Piekarz; Seth M. Steinberg; Jane B. Trepel; William D. Figg; David S. Schrump; Giuseppe Giaccone

Purpose: This phase I/II study sought to determine the safety and maximum tolerated dose (MTD) of a novel schedule of belinostat, a histone deacetylase inhibitor (HDAC) administered before and in combination with cisplatin (P), doxorubicin (A), and cyclophosphamide (C) in thymic epithelial tumors (TET). Antitumor activity, pharmacokinetics, and biomarkers of response were also assessed. Experimental Design: Patients with advanced, unresectable TET received increasing doses of belinostat as a continuous intravenous infusion over 48 hours with chemotherapy in 3-week cycles. In phase II, belinostat at the MTD was used. Results: Twenty-six patients were enrolled (thymoma, 12; thymic carcinoma, 14). Dose-limiting toxicities at 2,000 mg/m2 belinostat were grade 3 nausea and diarrhea and grade 4 neutropenia and thrombocytopenia, respectively, in two patients. Twenty-four patients were treated at the MTD of 1,000 mg/m2 with chemotherapy (P, 50 mg/m2 on day 2; A, 25 mg/m2 on days 2 and 3; C, 500 mg/m2 on day 3). Objective response rates in thymoma and thymic carcinoma were 64% (95% confidence interval, 30.8%-89.1%) and 21% (4.7%–50.8%), respectively. Modulation of pharmacodynamic markers of HDAC inhibition and declines in regulatory T cell (Treg) and exhausted CD8+ T-cell populations were observed. Decline in Tregs was associated with response (P = 0.0041) and progression-free survival (P = 0.021). Declines in TIM3+ CD8+ T cells were larger in responders than nonresponders (P = 0.049). Conclusion: This study identified the MTD of belinostat in combination with PAC and indicates that the combination is active and feasible in TETs. Immunomodulatory effects on Tregs and TIM3+ CD8+ T cells warrant further study. Clin Cancer Res; 20(21); 5392–402. ©2014 AACR.


Expert Opinion on Investigational Drugs | 2009

Small-molecule inhibitors of the human epidermal receptor family.

Corey A. Carter; Ronan J. Kelly; Giuseppe Giaccone

Background: Small molecule inhibitors of human epidermal receptors (HER) have become an integral part of the armamentarium available to the medical oncologist in the treatment of solid tumor malignancies. At present, there are two small-molecule inhibitors (erlotinib and lapatinib) approved by the FDA in the USA, and a third inhibitor, gefitinib, is approved in other countries. Objective: To summarize the current standards of care for these new agents in solid tumors, and to discuss ongoing clinical trials; to review the known mechanisms of action of these inhibitors as well as to discuss both the known predictive markers for response and likely mechanisms of resistance. Methods: We reviewed key presentations and recent publications on small-molecule inhibitors targeting the HER family in solid tumors. Conclusions: Recent data have highlighted the importance of mutations and amplifications of receptors within the HER family. Amplification of HER2 often translates into responses in anti-HER2 therapy. Mutations either enhance sensitivity or confer resistance to small-molecule inhibitors. Other mechanisms of resistance are being elucidated which should lead to the ability to predict both responses and resistance to HER family inhibitors and should translate into improvements in patient care.

Collaboration


Dive into the Corey A. Carter's collaboration.

Top Co-Authors

Avatar

Bryan Oronsky

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Tony Reid

University of California

View shared research outputs
Top Co-Authors

Avatar

Jan Scicinski

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Scott Caroen

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Arnold Oronsky

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Christina Brzezniak

Walter Reed National Military Medical Center

View shared research outputs
Top Co-Authors

Avatar

Arun Rajan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Pedro Cabrales

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge