Charles J. Schneider
Christiana Care Health System
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Charles J. Schneider.
Journal of Clinical Oncology | 2011
Quynh-Thu Le; Harold Kim; Charles J. Schneider; Györgyi Muraközy; K. Składowski; Sabine Reinisch; Yuhchyau Chen; Michael Hickey; May Mo; Mon Gy Chen; Dietmar Berger; Richard Lizambri; Michael Henke
PURPOSE Oral mucositis (OM) is a debilitating toxicity of chemoradiotherapy for head and neck cancer (HNC). This randomized, placebo-controlled, double-blind study evaluated the efficacy and safety of palifermin to reduce OM associated with definitive chemoradiotherapy for locally advanced HNC. PATIENTS AND METHODS Patients receiving conventionally fractionated radiotherapy (2.0 Gy/d, 5 days/wk to 70 Gy) with cisplatin (100 mg/m(2) on days 1, 22, and 43) received palifermin (180 μg/kg) or placebo before starting chemoradiotherapy and then once weekly for 7 weeks. The primary end point was the incidence of severe, observable, and functional OM (WHO grade 3 to 4). RESULTS The palifermin (n = 94) and placebo (n = 94) arms were well balanced. The incidence of severe OM was significantly lower for palifermin than for placebo (54% v 69%; P = .041). In the palifermin arm, median time to severe OM was delayed (47 v 35 days), median duration of severe OM was shortened (5 v 26 days), and the incidence of xerostomia grade ≥ 2 was lower (67% v 80%), favoring palifermin; however, the differences were not significant after multiplicity adjustment. Opioid analgesic use, average mouth and throat soreness scores, and chemoradiotherapy compliance were not significantly different between treatment arms. Adverse events were similar between arms (98%, palifermin; 93%, placebo). The most common study drug-related adverse events were rash, flushing, and dysgeusia. After median follow-up of 25.8 months, overall survival and progression-free survival were similar between treatment arms. CONCLUSION Although palifermin reduced severe functional OM, its role in the management of locally advanced HNC during chemoradiotherapy remains to be elucidated.
Journal of Clinical Oncology | 2012
Lawrence N. Shulman; Constance Cirrincione; Donald A. Berry; Heather P. Becker; Edith A. Perez; Ruth O'Regan; Silvana Martino; James N. Atkins; Erica L. Mayer; Charles J. Schneider; Gretchen Kimmick; Larry Norton; Hyman B. Muss; Clifford A. Hudis
PURPOSE The ideal duration of adjuvant chemotherapy for patients with lower risk primary breast cancer is not known. Cancer and Leukemia Group B trial 40101 was conducted using a phase III factorial design to define whether six cycles of a chemotherapy regimen are superior to four cycles. We also sought to determine whether paclitaxel (T) is as efficacious as doxorubicin/cyclophosphamide (AC), but with reduced toxicity. PATIENTS AND METHODS Between 2002 and 2008, the study enrolled women with operable breast cancer and zero to three positive nodes. Patients were randomly assigned to either four or six cycles of either AC or T. Study stratifiers were estrogen receptor/progesterone receptor (ER/PgR), human epidermal growth factor receptor 2 (HER2), and menopausal status. After 2003, all treatment was administered in dose-dense fashion. The primary efficacy end point was relapse-free survival (RFS). RESULTS A total of 3,171 patients were enrolled; 94% were node-negative and 6% had one to three positive nodes. At a median follow-up of 5.3 years, the 4-year RFS was 90.9% and 91.8% for six and four cycles, respectively. The adjusted hazard ratio (HR) of six to four cycles regarding RFS was 1.03 (95% CI, 0.84 to 1.28; P=.77). The 4-year OS was 95.3% and 96.3% for six and four cycles, respectively, with an HR of six to four cycles of 1.12 (95% CI, 0.84 to 1.49; P=.44). There was no interaction between treatment duration and chemotherapy regimen, ER/PgR, or HER2 status on RFS or OS. CONCLUSION For women with resected primary breast cancer and zero to three positive nodes, we found no evidence that extending chemotherapy regimens of AC or single-agent T from four to six cycles improves clinical outcome.
Journal of Clinical Oncology | 2014
Lawrence N. Shulman; Donald A. Berry; Constance Cirrincione; Heather P. Becker; Edith A. Perez; Ruth O'Regan; Silvana Martino; Charles L. Shapiro; Charles J. Schneider; Gretchen Kimmick; Harold J. Burstein; Larry Norton; Hyman B. Muss; Clifford A. Hudis
PURPOSE Optimal adjuvant chemotherapy for early-stage breast cancer balances efficacy and toxicity. We sought to determine whether single-agent paclitaxel (T) was inferior to doxorubicin and cyclophosphamide (AC), when each was administered for four or six cycles of therapy, and whether it offered less toxicity. PATIENTS AND METHODS Patients with operable breast cancer with 0 to 3 positive nodes were enrolled onto the study to address the noninferiority of single-agent T to AC, defined as the one-sided 95% upper-bound CI (UCB) of hazard ratio (HR) of T versus AC less than 1.30 for the primary end point of relapse-free survival (RFS). As a 2 × 2 factorial design, duration of therapy was also addressed and was previously reported. RESULTS With 3,871 patients enrolled onto the trial, a median follow-up period of 6.1 years, and 437 RFS events, we achieved an HR of 1.26 (one sided 95% UCB, 1.48; favoring AC does not allow a conclusion of noninferiority of T with AC; UCB > 1.3). With 266 patient deaths, the HR for overall survival (OS) was 1.27 favoring AC (UCB, 1.56). The estimated absolute advantage of AC at 5 years is 3% for RFS (91 v 88%) and 1% for OS (95 v 94%). All nine treatment-related deaths were patients receiving AC and are included in the analyses of both RFS and OS. Hematologic toxicity was more common in patients treated with AC, and neuropathy was more common in patients treated with T. CONCLUSION This trial did not show noninferiority of T to AC, a conclusion that is unlikely to change with additional events and follow-up. T was less toxic than AC.
Cancer | 2012
K. Kian Ang; Amy Y. Chen; Walter J. Curran; Adam S. Garden; Paul M. Harari; Barbara A. Murphy; Stuart J. Wong; Lisa A. Bellm; Marc D. Schwartz; Jason G. Newman; Douglas Adkins; D. Neil Hayes; Upendra Parvathaneni; David Brachman; Bassam Ghabach; Charles J. Schneider; Michael Greenberg; P.R. Anne
Detailed information about how patients with head and neck carcinoma (HNC) are treated across practice settings does not exist. The authors conducted a prospective, observational study to examine the patterns of care for a series of patients with newly diagnosed HNC in the United States and to test 2 hypotheses: 1) There is no difference in the pattern of care between community and academic settings; and 2) the results of major randomized clinical trials will change the pattern of care in both practice settings within 1 year of publication in peer‐reviewed journals.
Oncologist | 2009
Michael J. Guarino; Charles J. Schneider; Martha A. Hosford; Julie R. Brahmer; Charles M. Rudin; Friedrich Graf Finckenstein; Robyn Philip-Norton; Haolan Lu; Martin R. Weber; David S. Ettinger
PURPOSE To determine the optimal dose of the antiepidermal growth factor receptor (EGFR) monoclonal antibody cetuximab that can be safely administered in combination with a standard daily dose of erlotinib in patients with advanced solid malignancies. PATIENTS AND METHODS Patients with advanced solid malignancies who had failed standard chemotherapies received escalating doses of cetuximab without a loading dose (100, 200, 250 mg/m(2) i.v. weekly) in combination with a fixed dose of erlotinib (150 mg daily orally) until disease progression or unacceptable toxicity. RESULTS Twenty-two patients were treated, including 14 patients (64%) with non-small cell lung cancer. Twenty patients received combination treatment at the highest dose level for a median of 5.5 weeks (range, 1-31 weeks). One dose-limiting toxicity was observed: grade 3 skin rash. Overall, the most common adverse events (any grade, grade 3/4) were consistent with the safety profiles of the individual drugs: acneform rash (100%, 9%), diarrhea (77%, 5%), and hypomagnesemia (59%, 12%). Seven of 18 evaluable patients (38.9%) had stable disease lasting for a median of 16.6 weeks (range, 6.1-25.1 weeks). CONCLUSION Dual EGFR inhibition with cetuximab and erlotinib is feasible; the observed toxicities were manageable and consistent with the safety profiles of the individual drugs. The recommended doses for phase II studies are 250 mg/m(2) i.v. weekly for cetuximab and 150 mg daily orally for erlotinib.
Journal of Clinical Oncology | 2003
D. Bruce Panasuk; Thomas Bauer; Allen L. Davies; Charles J. Schneider; Cynthia Flynn
A 21-year-old white female presented with a 9-month history of progressively worsening vague chest pressure and discomfort, dyspnea, anorexia, and a 20-lb weight loss. Her medical history was unremarkable, and physical examination was normal, aside from a tachycardia of 120 beats per minute. Computed tomography (CT) of the chest revealed a large, somewhat lobulated, homogeneous anterior mediastinal mass measuring approximately 13 10 8 cm (Fig 1). Despite its size, the mass seemed resectable, without definite invasion of adjacent structures. CT-directed fine-needle aspiration biopsy was performed, and histopathologic examination of the biopsy specimen showed a neoplasm with prominent myxoid differentiation without evidence of lymphoma or thymoma. Given this tentative diagnosis of a soft tissue sarcoma, the resectable appearance of the mass on CT, and a negative metastatic work-up, surgical resection via median sternotomy was undertaken. At the time of operation, the mass appeared larger than reported by the preoperative CT scan and was found to be densely adherent to the pericardium, the mediastinal pleura, and the left lung. Complete surgical resection with negative margins was achieved by wide en bloc resection of these structures along with the tumor (Fig 2). The patient’s recovery was uneventful. Final pathologic examination revealed a high-grade malignant neoplasm with clear-cut features of skeletal muscle differentiation. An area of focal residual thymus was evident (Fig 3, upper left corner) along with the high-grade malignant neoplasm with large atypical cells (Fig 3, lower portion of slide). High-power magnification revealed large tumor cells with prominent round to oval nuclei and intensely eosinophilic cytoplasm—so-called myoblast strap cells. Cross striations in the myoblasts are not seen in this view, however (Fig 4). The tumor was immunoreactive for common muscle actin and desmin, was negative for smooth muscle actin and keratin (CAM 5.2 and AE1/AE3), but demonstrated convincing nuclear positivity for myogen. Final diagnosis was a primary anterior mediastinal rhabdomyosarcoma, anatomically related to, and likely arising from, the thymus. Primary mediastinal rhabdomyosarcomas unassociated with germ cell, teratomatous, or malignant epithelial components are extremely rare. They are thought to arise from the thymus; the ability of thymic tissue to differentiate toward myoid tissue has been reported previously. These tumors tend to occur in the anterior mediastinum of young adults and demonstrate large size and local invasion at the time of diagnosis. Their biologic behavior is aggressive, usually characterized by rapid recurrence and dissemination, even following resection. Because of the limited information regarding treatment of these unusual tumors, therapy must be extrapolated from the management of pediatric rhabdomyosarcomas and applied to these patients. This patient is currently receiving combination chemotherapy
Clinical Cancer Research | 2017
Jhanelle E. Gray; Rebecca S. Heist; Alexander Starodub; D. Ross Camidge; Ebenezer A. Kio; Gregory A. Masters; W. Thomas Purcell; Michael J. Guarino; Jamal Ghazi Misleh; Charles J. Schneider; Bryan J. Schneider; Allyson J. Ocean; Tirrell Johnson; Leena Gandhi; Kevin Kalinsky; Ronald J. Scheff; Wells A. Messersmith; Serengulam V. Govindan; Pius Maliakal; Boyd Mudenda; William A. Wegener; Robert M. Sharkey; David M. Goldenberg
Purpose: We evaluated a Trop-2–targeting antibody conjugated with SN-38 in metastatic small cell lung cancer (mSCLC) patients. Experimental Design: Sacituzumab govitecan was studied in patients with pretreated (median, 2; range, 1–7) mSCLC who received either 8 or 10 mg/kg i.v. on days 1 and 8 of 21-day cycles. The primary endpoints were safety and objective response rate (ORR); duration of response, progression-free survival (PFS), and overall survival (OS) were secondary endpoints. Results: Sixty percent of patients showed tumor shrinkage from baseline CTs. On an intention-to-treat basis (N = 50), the ORR was 14% (17% for the 10-mg/kg group); the median response duration, 5.7 months; the clinical benefit rate (CBR ≥4 months), 34%; median PFS, 3.7 months; and median OS, 7.5 months. There was a suggested improvement in PR, CBR, and PFS with sacituzumab govitecan in second-line patients who were sensitive to first-line therapy, but no difference between first-line chemosensitive versus chemoresistant patients in the overall population. There was a statistically significant higher OS in those patients who received prior topotecan versus no topotecan therapy in a small subgroup. Grade ≥3 adverse events included neutropenia (34%), fatigue (13%), diarrhea (9%), and anemia (6%). Trop-2 tumor staining was not required for patient selection. No antibodies to the drug conjugate or its components were detected on serial blood collections. Conclusions: Sacituzumab govitecan appears to have a safe and effective therapeutic profile in heavily pretreated mSCLC patients, including those who are chemosensitive or chemoresistant to first-line chemotherapy. Additional studies as a monotherapy or combination therapy are warranted. Clin Cancer Res; 23(19); 5711–9. ©2017 AACR.
Cancer Research | 2017
Jhanelle E. Gray; Rebecca S. Heist; Alexander Starodub; D. Ross Camidge; Ebenezer A. Kio; Gregory A. Masters; W. Thomas Purcell; Michael J. Guarino; Jamal Ghazi Misleh; Charles J. Schneider; Bryan J. Schneider; Allyson J. Ocean; Tirrell Johnson; Leena Gandhi; Kevin Kalinsky; Serengulam V. Govindan; Pius Maliakal; Boyd Mudenda; William A. Wegener; Robert M. Sharkey; David M. Goldenberg
Introduction: Sacituzumab govitecan (IMMU-132) is an antibody-drug conjugate (ADC) delivering SN-38, a topoisomerase-1 inhibitor, to mSCLC cells expressing Trop-2. We are studying the safety/tolerability and efficacy of IMMU-132 in patients with relapsed/refractory mSCLC who had received a platinum-containing first-line regimen. Experimental Procedure: Patients received 8 or 10 mg/kg IV IMMU-132 on days 1 and 8 of repeated 21-day cycles. Objective tumor response (ORR) was determined by RECISTv1.1 in patients receiving at least one treatment cycle, and progression-free survival (PFS) and overall survival (OS) in all patients by Kaplan-Meier methods. (ClinicalTrials.gov, NCT01631552) Summary of New Unpublished Data: A total of 53 patients (23/30 M/F, median 63 years old) with 1-7 (median 2) prior lines of therapy were enrolled between November, 2013 and June, 2016. Immunohistochemistry of evaluable archival tumor specimens for Trop-2 expression (N=26) showed 92% positivity (61% moderately to strongly positive). They received up to 32 treatment cycles (median 5); the most frequent Grade >3 adverse events were neutropenia (34%), fatigue (13%) and diarrhea (9%). Four patients did not complete one cycle of treatment. In the other 49 patients (14 at 8 mg/kg, 35 at 10 mg/kg), there were 7 confirmed PRs and 21 SDs as best response. The resulting ORR rate was 14% (7/49), with a median duration and time-to-progression of a response of 4.0 and 7.6 mos, respectively, and the clinical benefit rate (PR+SD >4 mos) was 35% (17/49). The ORR rate was similar in pts who were sensitive (>3 mos response) or resistant ( Conclusion: These interim results demonstrate encouraging activity in patients with late-stage mSCLC having a high expression of Trop-2. Even after failing 1st-line platinum chemotherapy or 2nd-line topotecan therapy, IMMU-132 showed promising activity, and has a manageable toxicity profile. IMMU-132 given at 10 mg/kg on day 1 and 8 of a 3-week cycle was selected for further clinical evaluation in this population. Citation Format: Jhanelle E. Gray, Rebecca S. Heist, Alexander N. Starodub, D. Ross Camidge, Ebenezer Kio, Gregory Masters, W. Thomas Purcell, Michael J. Guarino, Jamal Misleh, Charles J. Schneider, Bryan J. Schneider, Allyson J. Ocean, Tirrell Johnson, Leena Gandhi, Kevin Kalinsky, Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, David M. Goldenberg. Phase 2 study of sacituzumab govitecan (IMMU-132), an anti-Trop-2/SN-38 antibody-drug conjugate (ADC), in patients with pretreated metastatic small-cell lung cancer (mSCLC) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr CT155. doi:10.1158/1538-7445.AM2017-CT155
Oral Oncology | 2018
Joshua D. Palmer; Charles J. Schneider; Neil G. Hockstein; Alexandra L. Hanlon; Jordan Silberg; Jon Strasser; Elizabeth Mauer; Michael Dzeda; Robert L. Witt; Adam Raben
OBJECTIVES The objective of this study was to investigate the safety, tolerability and preliminary efficacy of radiotherapy plus cetuximab in high risk CSCC patients. MATERIALS AND METHODS Patients with high-risk CSCC diagnosed between 2006 and 2013 were analyzed. Patients were divided into two groups: radiotherapy alone versus radiotherapy plus cetuximab. Among 68 patients meeting study criteria, we identified 29 treated with cetuximab plus RT and 39 with RT alone. Primary analysis examined disease-free and overall survival, freedom from local and distant recurrence in the propensity score matched cohort. Propensity score analysis was performed with weighted factors including: Charlson Comorbidity Index score, age. KPS, primary location, T and N stage, recurrent status, margin status, LVSI, PNI and grade. Toxicity was assessed using the CTCAE v4.0. RESULTS Median follow-up for living patients was 30 months. Patients in the cetuximab group were more likely to have advanced N stage, positive margins and recurrent disease. After propensity score matching the groups were well balanced. Six patients experienced ≥ grade 3 acute toxicity in the cetuximab group. The 1-year, 2-year and 5-year progression free survival (PFS) for patients in the cetuximab group were 86%, 72% and 66%, respectively. The 1-year, 2-year and 5-year overall survival (OS) for patients in the cetuximab group was 98%, 80% and 80%, respectively. CONCLUSIONS Although limited by small numbers, the combination of cetuximab and radiotherapy in CSCC appears well tolerated there were more long-term survivors and less distant metastasis in the cetuximab group. These promising finding warrant further studies.
JAMA Oncology | 2018
Robert L. Ferris; Nabil F. Saba; Barbara J. Gitlitz; Robert I. Haddad; Ammar Sukari; Prakash Neupane; John C. Morris; Krzysztof Misiukiewicz; Julie E. Bauman; Moon Jung Fenton; Antonio Jimeno; Douglas Adkins; Charles J. Schneider; Assuntina G. Sacco; Keisuke Shirai; Daniel W. Bowles; Michael K. Gibson; T. Nwizu; Raphael Gottardo; Kristi Manjarrez; Gregory N. Dietsch; James Kyle Bryan; Robert M. Hershberg; Ezra E.W. Cohen
Importance Immunotherapy for recurrent and/or metastatic (R/M) squamous cell carcinoma of the head and neck (SCCHN) is promising. The toll-like receptor 8 (TLR8) agonist motolimod may stimulate innate and adaptive immunity. Objective To determine whether motolimod improves outcomes for R/M SCCHN when combined with standard therapy. Design, Setting, and Participants The Active8 study was a multicenter, randomized, double-blind, placebo-controlled clinical trial enrolling adult patients (age ≥18 years) with histologically confirmed R/M SCCHN of the oral cavity, oropharynx, hypopharynx, or larynx between October 2013 and August 2015. Follow-up ended September 2016. Analysis for the present report was conducted between June 2016 and December 2017. Interventions Combination treatment with platinum (carboplatin or cisplatin), fluorouracil, cetuximab (the EXTREME regimen), and either placebo or motolimod, each administered intravenously every 3 weeks. Patients received a maximum of 6 chemotherapy cycles, after which patients received weekly cetuximab with either placebo or motolimod every 4 weeks. Main Outcomes and Measures Progression-free survival (PFS) as determined by independent central review using immune-related RECIST (Response Evaluation Criteria in Solid Tumors). Key secondary end points included overall survival (OS) and safety. Results Of 195 patients enrolled, 85% were men (n = 166); 82% were white (n = 159); median age was 58 years (range 23-81 years). Median PFS was 6.1 vs 5.9 months (hazard ratio [HR], 0.99; 1-sided 90% CI, 0.00-1.22; P = .47), and median OS was 13.5 vs 11.3 months (HR, 0.95; 1-sided 90% CI, 0.00-1.22; P = .40) for motolimod vs placebo. Increased incidence of injection site reactions, pyrexia, chills, anemia, and acneiform rash were noted with motolimod. Of 83 cases oropharyngeal cancer, 52 (63%) were human papillomavirus (HPV) positive. In a prespecified subgroup analysis of HPV-positive participants, motolimod vs placebo resulted in significantly longer PFS (7.8 vs 5.9 months; HR, 0.58; 1-sided 90% CI, 0.00-0.90; P = .046) and OS (15.2 vs 12.6 months; HR, 0.41; 1-sided 90% CI, 0.00-0.77; P = .03). In an exploratory analysis, patients with injection site reactions had longer PFS and OS (median PFS, 7.1 vs 5.9 months; HR, 0.69; 1-sided 90% CI, 0.00-0.93; P = .06; and median OS, 18.7 vs 12.6; HR, 0.56; 1-sided 90% CI, 0.00-0.81; P = .02). Conclusions and Relevance Adding motolimod to the EXTREME regimen was well tolerated but did not improve PFS or OS in the intent-to-treat population. Significant benefit was observed in HPV-positive patients and those with injection site reactions, suggesting that TLR8 stimulation may benefit subset- and biomarker-selected patients. Trial Registration ClinicalTrials.gov identifier: NCT01836029.