Gary I. Cohen
Greater Baltimore Medical Center
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Gary I. Cohen.
Journal of Clinical Oncology | 1999
Martee L. Hensley; Lynn M. Schuchter; Celeste Lindley; Neal J. Meropol; Gary I. Cohen; Gail Broder; William J. Gradishar; Daniel M. Green; Robert Langdon; R. Brian Mitchell; Robert S. Negrin; Ted P. Szatrowski; J. Tate Thigpen; Daniel VonHoff; Todd H. Wasserman; David G. Pfister
PURPOSE Because toxicities associated with chemotherapy and radiotherapy can adversely affect short- and long-term patient quality of life, can limit the dose and duration of treatment, and may be life-threatening, specific agents designed to ameliorate or eliminate certain chemotherapy and radiotherapy toxicities have been developed. Variability in interpretation of the available data pertaining to the efficacy of the three United States Food and Drug Administration-approved agents that have potential chemotherapy- and radiotherapy-protectant activity-dexrazoxane, mesna, and amifostine-and questions about the role of these protectant agents in cancer care led to concern about the appropriate use of these agents. The American Society of Clinical Oncology sought to establish evidence-based, clinical practice guidelines for the use of dexrazoxane, mesna, and amifostine in patients who are not enrolled on clinical treatment trials. METHODS A multidisciplinary Expert Panel reviewed the clinical data regarding the activity of dexrazoxane, mesna, and amifostine. A computerized literature search was performed using MEDLINE. In addition to reports collected by individual Panel members, all articles published in the English-speaking literature from June 1997 through December 1998 were collected for review by the Panel chairpersons, and appropriate articles were distributed to the entire Panel for review. Guidelines for use, levels of evidence, and grades of recommendation were reviewed and approved by the Panel. Outcomes considered in evaluating the benefit of a chemotherapy- or radiotherapy-protectant agent included amelioration of short- and long-term chemotherapy- or radiotherapy-related toxicities, risk of tumor protection by the agent, toxicity of the protectant agent itself, quality of life, and economic impact. To the extent that these data were available, the Panel placed the greatest value on lesser toxicity that did not carry a concomitant risk of tumor protection. RESULTS AND CONCLUSION Mesna: (1) Mesna, dosed as detailed in these guidelines, is recommended to decrease the incidence of standard-dose ifosfamide-associated urothelial toxicity. (2) There is insufficient evidence on which to base a guideline for the use of mesna to prevent urothelial toxicity with ifosfamide doses that exceed 2.5 g/m(2)/d. (3) Either mesna or forced saline diuresis is recommended to decrease the incidence of urothelial toxicity associated with high-dose cyclophosphamide use in the stem-cell transplantation setting. Dexrazoxane: (1) The use of dexrazoxane is not routinely recommended for patients with metastatic breast cancer who receive initial doxorubicin-based chemotherapy. (2) The use of dexrazoxane may be considered for patients with metastatic breast cancer who have received a cumulative dosage of 300 mg/m(2) or greater of doxorubicin in the metastatic setting and who may benefit from continued doxorubicin-containing therapy. (3) The use of dexrazoxane in the adjuvant setting is not recommended outside of a clinical trial. (4) The use of dexrazoxane can be considered in adult patients who have received more than 300 mg/m(2) of doxorubicin-based therapy for tumors other than breast cancer, although caution should be used in settings in which doxorubicin-based therapy has been shown to improve survival because of concerns of tumor protection by dexrazoxane. (5) There is insufficient evidence to make a guideline for the use of dexrazoxane in the treatment of pediatric malignancies, with epirubicin-based regimens, or with high-dose anthracycline-containing regimens. Similarly, there is insufficient evidence on which to base a guideline for the use of dexrazoxane in patients with cardiac risk factors or underlying cardiac disease. (6) Patients receiving dexrazoxane should continue to be monitored for cardiac toxicity. Amifostine: (1) Amifostine may be considered for the reduction of nephrotoxicity in patients receiving cisplatin-based chemoth
Breast Journal | 2003
Sarah Spagnola; James Zabora; Karlynn BrintzenhofeSzoc; Craig M. Hooker; Gary I. Cohen; Frank Baker
Abstract: Despite improved overall survival rates, the diagnosis of breast cancer continues to generate fear and turmoil in the lives of many women. All phases related to diagnosis, treatment, and recovery create challenges and problems that patients and survivors must face. Clearly, at the time of diagnosis and during the first phases of treatment, patients experience uncertainty, confusion, and distress. Quality of life (QOL) can be negatively affected by inadequate information, complex decisions, and adverse events related to cancer therapies. As treatment continues, concerns related to physical functioning, body image, mood, sexuality, family, and vocational pursuits quickly emerge. Adjuvant treatments generate additional physiological assaults that further affect body image, sexuality, and family. As women move beyond treatment, the role of patient shifts to that of survivor, with a need for continued focus on overall QOL issues. Throughout this continuum, QOL is a critical factor that must be evaluated and monitored. The Satisfaction with Life Domains Scale for Breast Cancer (SLDS‐BC) is a reliable and valid scale that presents a critical opportunity to assess QOL throughout the various phases of patient care. A principal component factor analysis with a varimax rotation identified the following five QOL factors, explaining 70.8% of the variance: social functioning, physical functioning, internal locus of control, spirituality, and communication with medical providers. Cronbachs alpha for the entire scale was 0.93. Test‐retest produced rs for each factor ranging from 0.45 to 0.91, with an overall r = 0.70. Concurrent and divergent validity were assessed through the Functional Assessment of Cancer Therapies for Breast Cancer (FACT‐B) and the Brief Symptom Inventory (BSI). Significant negative correlations (p < 0.01) were found between the SLDS‐BC and the FACT‐B as well as the BSI. These results indicated strong concurrent and divergent validity. The SLDS‐BC clearly offers a user‐friendly format that can briefly and rapidly assess QOL across the breast cancer continuum of care.
Journal of Clinical Oncology | 2015
David H. Lawson; Sandra J. Lee; Fengmin Zhao; Ahmad A. Tarhini; Kim Margolin; Marc S. Ernstoff; Michael B. Atkins; Gary I. Cohen; Theresa L. Whiteside; Lisa H. Butterfield; John M. Kirkwood
PURPOSE We conducted a double-blind, placebo-controlled trial to evaluate the effect of granulocyte-macrophage colony-stimulating factor (GM-CSF) and peptide vaccination (PV) on relapse-free survival (RFS) and overall survival (OS) in patients with resected high-risk melanoma. PATIENTS AND METHODS Patients with completely resected stage IV or high-risk stage III melanoma were grouped by human leukocyte antigen (HLA) -A2 status. HLA-A2-positive patients were randomly assigned to receive GM-CSF, PV, both, or placebo; HLA-A2-negative patients, GM-CSF or placebo. Treatment lasted for 1 year or until recurrence. Efficacy analyses were conducted in the intent-to-treat population. RESULTS A total of 815 patients were enrolled. There were no significant improvements in OS (stratified log-rank P = .528; hazard ratio, 0.94; 95% repeated CI, 0.77 to 1.15) or RFS (P = .131; hazard ratio, 0.88; 95% CI, 0.74 to 1.04) in the patients assigned to GM-CSF (n = 408) versus those assigned to placebo (n = 407). The median OS times with GM-CSF versus placebo treatments were 69.6 months (95% CI, 53.4 to 83.5 months) versus 59.3 months (95% CI, 44.4 to 77.3 months); the 5-year OS probability rates were 52.3% (95% CI, 47.3% to 57.1%) versus 49.4% (95% CI, 44.3% to 54.3%), respectively. The median RFS times with GM-CSF versus placebo were 11.4 months (95% CI, 9.4 to 14.8 months) versus 8.8 months (95% CI, 7.5 to 11.2 months); the 5-year RFS probability rates were 31.2% (95% CI, 26.7% to 35.9%) versus 27.0% (95% CI, 22.7% to 31.5%), respectively. Exploratory analyses showed a trend toward improved OS in GM-CSF-treated patients with resected visceral metastases. When survival in HLA-A2-positive patients who received PV versus placebo was compared, RFS and OS were not significantly different. Treatment-related grade 3 or greater adverse events were similar between GM-CSF and placebo groups. CONCLUSION Neither adjuvant GM-CSF nor PV significantly improved RFS or OS in patients with high-risk resected melanoma. Exploratory analyses suggest that GM-CSF may be beneficial in patients with resected visceral metastases; this observation requires prospective validation.
Cancer Investigation | 2001
Barbara Poniatowski; Patricia Grimm; Gary I. Cohen
Menopause is a natural biologic event in the reproductive cycle of the middle-aged woman (1–3). This event can be delineated into three phases. The premenopausal phase is defined as having menstruated within the last 3 months; the menopausal phase is defined as having last menstruated within the past 3–12 months, and the postmenopausal phase is defined as absent menstruation for at least 12 months (1–4). The Massachusetts Women’s Health Study (MWHS) provides stable estimates of parameters for a natural menopause. This longitudinal 5year study of 2570 women identified the average age of menopause as 50, with a range of 45–55 (5,6). Physical and environmental factors may affect the age of menopause. These factors include nutrition, smoking, reproductive history, and socioeconomic status (4). Malnutrition, smoking, and lower height and weight contribute to early menopause and a shorter premenopausal period (4,5). During menopause, biological fertility and endocrine ovarian activity ceases with a marked decline in
Cancer | 2011
Tomasz P. Srokowski; James Liebmann; Manuel R. Modiano; Gary I. Cohen; Barbara Pro; Jorge Romaguera; Christine Kuepfer; Jack W. Singer; Luis Fayad
Pixantrone dimaleate (pixantrone) has been shown to have antitumor activity in leukemia and lymphoma in vitro models and to lack delayed cardiotoxicity associated with mitoxantrone in animal models. FND‐R, a combination regimen of fludarabine, mitoxantrone, dexamethasone, and rituximab, has been shown to be an effective regimen for low‐grade lymphomas.
Supportive Care in Cancer | 1999
Burton Needles; Eduardo Miranda; Francisco M. Garcia Rodriguez; Luis Baez Diaz; Jesse Spector; Johnny Craig; Gary I. Cohen; Steven Krasnow; J. Brogden; Michael Ames
Abstract The objectives of this study were to compare the efficacy and safety of orally administered ondansetron 8 mg b.i.d., 24 mg q.d., and 32 mg q.d. in the prevention of nausea and vomiting associated with high-dose cisplatin-based chemotherapy (cisplatin ≥50 mg/m2). This was a randomized, parallel-group, double-blind study conducted in North America. It was planned that all patients would receive one of the following orally administered ondansetron treatments 30 min before starting cisplatin dosing (administered over ≤3 h): 8 mg b.i.d. with 8 h between doses (124 patients), 24 mg q.d. (116 patients), and 32 mg q.d. (117 patients). Use of prophylactic corticosteroids was not permitted. During the 24-h study period, the highest complete response rate (no emesis, rescue antiemetic therapy, or withdrawal) occurred in patients who received ondansetron 24 mg q.d.: 76/115 or 66%, as against 68/124 (55%) after ondansetron 8 mg b.i.d. and 64/117 (55%) after ondansetron 32 mg q.d. Complete control of nausea (no nausea, no rescue, no withdrawal) occurred in more patients in the ondansetron 24 mg q.d. group (64/114, 56%) than in the ondansetron 8 mg b.i.d. group (43/121, 36%) or in the ondansetron 32 mg group (55/117, 50%). These results demonstrate that following highly emetogenic cisplatin-based chemotherapy (≥50 mg/m2), oral ondansetron 24 mg q.d. is more effective than 8 mg b.i.d. for overall control of nausea, and at least as effective if not more effective in the control of acute vomiting than 8 mg b.i.d. or 32 mg q.d. Ondansetron 24 mg q.d. was well tolerated, and no new or unexpected adverse events were identified.
Journal of Clinical Oncology | 2017
Sanjiv S. Agarwala; Sandra J. Lee; Wai-Ki Yip; Uma N. M. Rao; Ahmad A. Tarhini; Gary I. Cohen; Douglas S. Reintgen; Terry Evans; Joanna Brell; Mark R. Albertini; Michael B. Atkins; Shaker R. Dakhil; Robert M. Conry; Jeffrey A. Sosman; Lawrence E. Flaherty; Vernon K. Sondak; William E. Carson; Michael Smylie; Alberto S. Pappo; Richard F. Kefford; John M. Kirkwood
Purpose To test the efficacy of 4 weeks of intravenous (IV) induction with high-dose interferon (IFN) as part of the Eastern Cooperative Oncology Group regimen compared with observation (OBS) in patients with surgically resected intermediate-risk melanoma. Patients and Methods In this intergroup international trial, eligible patients had surgically resected cutaneous melanoma in the following categories: (1) T2bN0, (2) T3a-bN0, (3) T4a-bN0, and (4) T1-4N1a-2a (microscopic). Patients were randomly assigned to receive IFN α-2b at 20 MU/m2/d IV for 5 days (Monday to Friday) every week for 4 weeks (IFN) or OBS. Stratification factors were pathologic lymph node status, lymph node staging procedure, Breslow depth, ulceration of the primary lesion, and disease stage. The primary end point was relapse-free survival. Secondary end points included overall survival, toxicity, and quality of life. Results A total of 1,150 patients were randomly assigned. At a median follow-up of 7 years, the 5-year relapse-free survival rate was 0.70 (95% CI, 0.66 to 0.74) for OBS and 0.70, (95% CI, 0.66 to 0.74) for IFN ( P = .964). The 5-year overall survival rate was 0.83 (95% CI, 0.79 to 0.86) for OBS and 0.83 (95% CI, 0.80 to 0.86) for IFN ( P = .558). Treatment-related grade 3 and higher toxicity was 4.6% versus 57.9% for OBS and IFN, respectively ( P < .001). Quality of life was worse for the treated group. Conclusion Four weeks of IV induction as part of the Eastern Cooperative Oncology Group high-dose IFN regimen is not better than OBS alone for patients with intermediate-risk melanoma as defined in this trial.
Clinical Cancer Research | 2017
Lisa H. Butterfield; Fengmin Zhao; Sandra J. Lee; Ahmad A. Tarhini; Kim Margolin; Richard L. White; Michael B. Atkins; Gary I. Cohen; Theresa L. Whiteside; John M. Kirkwood; David H. Lawson
Purpose: E4697 was a multicenter intergroup randomized placebo-controlled phase III trial of adjuvant GM-CSF and/or a multiepitope melanoma peptide vaccine for patients with completely resected, high-risk stage III/IV melanoma. Experimental Design: A total of 815 patients were enrolled from December 1999 to October 2006 into this six-arm study. GM-CSF was chosen to promote the numbers and functions of dendritic cells (DC). The melanoma antigen peptide vaccine (Tyrosinase368-376 (370D), gp100209-217 (210M), MART-127-35) in montanide was designed to promote melanoma-specific CD8+ T-cell responses. Results: Although the overall RFS and OS were not significantly improved with the vaccine or GM-CSF when compared with placebo, immunomodulatory effects were observed in peripheral blood and served as important correlates to this therapeutic study. Peripheral blood was examined to evaluate the impact of GM-CSF and/or the peptide vaccine on peripheral blood immunity and to investigate potential predictive or prognostic biomarkers. A total of 11.3% of unvaccinated patients and 27.1% of vaccinated patients developed peptide-specific CD8+ T-cell responses. HLA-A2+ patients who had any peptide-specific CD8+ T-cell response at day +43 tended to have poorer OS in univariate analysis. Patients receiving GM-CSF had significant reduction in percentages of circulating myeloid dendritic cells (mDC) and plasmacytoid DC (pDC) at day +43. In a subset of patients who received GM-CSF, circulating myeloid-derived suppressor cells (MDSC), and anti-GM-CSF–neutralizing antibodies (Nabs) were also modulated. The majority of patients developed anti-GM-CSF Nabs, which correlated with improved RFS and OS. Conclusions: The assessment of cellular and humoral responses identified counterintuitive immune system changes correlating with clinical outcome. Clin Cancer Res; 23(17); 5034–43. ©2017 AACR.
Cancer | 2017
Kevin Kalinsky; Sandra J. Lee; Krista M. Rubin; Donald P. Lawrence; Anthony J. Iafrarte; Darell R. Borger; Kim Margolin; Mario M. Leitao; Ahmad A. Tarhini; Henry B. Koon; Andrew L. Pecora; Anthony J. Jaslowski; Gary I. Cohen; Timothy M. Kuzel; Christopher D. Lao; John M. Kirkwood
KIT‐directed tyrosine kinase inhibitors such as imatinib have demonstrated benefits in KIT‐mutant (KIT+) mucosal, acral, vulvovaginal, and chronically sun‐damaged (CSD) melanoma. Dasatinib has superior preclinical activity in comparison with other tyrosine kinase inhibitors against cells with the most common KIT mutation, exon 11L576P. The ECOG‐ACRIN E2607 trial assessed dasatinib in patients with these melanoma subtypes.
Journal of Clinical Apheresis | 1998
Barbara Poniatowski; Gary I. Cohen; Margaret Tillett; Ellen Carr; Kurt Gunter; Susan Erickson; Rebecca Haley; Clifford F. Melick
The purpose of this in vitro study was to determine whether the Gemini PC‐2TX infusion pump could safely deliver peripheral stem cells (PSC) for an autologous PSC transplant. For purposes of hypothesis testing, it was assumed that there would be no significant difference in CD34+ cell counts and colony‐forming units‐granulocyte, macrophage (CFU‐GM) when the PSCs were administered by an IMED PC‐2TX infusion pump as opposed to an intravenous push method. The American Red Cross collected 50‐ml samples of PSCs from four donors by apheresis. These cells were tested for CD34+ using flow cytometry and for functional progenitor cells using a CFU‐GM assay. The cells were cryopreserved after testing. For our study, samples were tested simultaneously at a single facility. Each sample was individually thawed and a baseline thaw sample collected; 10 ml of the donor specimen was pushed through a syringe into a specimen container (intravenous push sample). The remainder of the specimen was infused through the IMED Gemini PC‐2TX pump into a specimen container (intravenous pump sample). All samples were assayed for CD34+ cell counts and CFU‐GM. Data analyses were conducted using the t‐test for paired samples, with values of P < 0.05 considered significant. Results failed to demonstrate a statistically significant difference between the CD34+ or CFU‐GM results of the intravenous push and intravenous pump specimens. Additionally, we failed to find a statistically significant difference when we compared the intravenous push and the intravenous pump specimens with the baseline thaw sample. The results of this study support the hypothesis that the Gemini PC‐2TX infusion pump can safely deliver PSCs for the purposes of stem cell transplantation. J. Cell. Apheresis 13:23–27, 1998.