Network


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

Hotspot


Dive into the research topics where Stephen J. Schuster is active.

Publication


Featured researches published by Stephen J. Schuster.


The New England Journal of Medicine | 2015

PD-1 Blockade with Nivolumab in Relapsed or Refractory Hodgkin's Lymphoma

Stephen M. Ansell; Alexander M. Lesokhin; Ivan Borrello; Ahmad Halwani; Emma C. Scott; Martin Gutierrez; Stephen J. Schuster; Michael Millenson; Deepika Cattry; Gordon J. Freeman; Scott J. Rodig; Bjoern Chapuy; Azra H. Ligon; Lili Zhu; Joseph F. Grosso; Su Y oung Kim; John M. Timmerman; Margaret A. Shipp; Philippe Armand

BACKGROUND Preclinical studies suggest that Reed-Sternberg cells exploit the programmed death 1 (PD-1) pathway to evade immune detection. In classic Hodgkins lymphoma, alterations in chromosome 9p24.1 increase the abundance of the PD-1 ligands, PD-L1 and PD-L2, and promote their induction through Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling. We hypothesized that nivolumab, a PD-1-blocking antibody, could inhibit tumor immune evasion in patients with relapsed or refractory Hodgkins lymphoma. METHODS In this ongoing study, 23 patients with relapsed or refractory Hodgkins lymphoma that had already been heavily treated received nivolumab (at a dose of 3 mg per kilogram of body weight) every 2 weeks until they had a complete response, tumor progression, or excessive toxic effects. Study objectives were measurement of safety and efficacy and assessment of the PDL1 and PDL2 (also called CD274 and PDCD1LG2, respectively) loci and PD-L1 and PD-L2 protein expression. RESULTS Of the 23 study patients, 78% were enrolled in the study after a relapse following autologous stem-cell transplantation and 78% after a relapse following the receipt of brentuximab vedotin. Drug-related adverse events of any grade and of grade 3 occurred in 78% and 22% of patients, respectively. An objective response was reported in 20 patients (87%), including 17% with a complete response and 70% with a partial response; the remaining 3 patients (13%) had stable disease. The rate of progression-free survival at 24 weeks was 86%; 11 patients were continuing to participate in the study. Reasons for discontinuation included stem-cell transplantation (in 6 patients), disease progression (in 4 patients), and drug toxicity (in 2 patients). Analyses of pretreatment tumor specimens from 10 patients revealed copy-number gains in PDL1 and PDL2 and increased expression of these ligands. Reed-Sternberg cells showed nuclear positivity of phosphorylated STAT3, indicative of active JAK-STAT signaling. CONCLUSIONS Nivolumab had substantial therapeutic activity and an acceptable safety profile in patients with previously heavily treated relapsed or refractory Hodgkins lymphoma. (Funded by Bristol-Myers Squibb and others; ClinicalTrials.gov number, NCT01592370.).


The New England Journal of Medicine | 2014

Ibrutinib versus ofatumumab in previously treated chronic lymphoid leukemia

John C. Byrd; Jennifer R. Brown; Susan O'Brien; Jaqueline C. Barrientos; Neil E. Kay; Nashitha Reddy; Steven Coutre; Constantine S. Tam; Stephen P. Mulligan; Ulrich Jaeger; S Devereux; Paul M. Barr; Richard R. Furman; Thomas J. Kipps; Florence Cymbalista; Christopher Pocock; Patrick Thornton; Federico Caligaris-Cappio; Tadeusz Robak; J. Delgado; Stephen J. Schuster; Marco Montillo; Anna Schuh; S. de Vos; Devinder Gill; Adrian Bloor; Claire Dearden; Carol Moreno; J. J. Jones; Alvina D. Chu

BACKGROUND In patients with chronic lymphoid leukemia (CLL) or small lymphocytic lymphoma (SLL), a short duration of response to therapy or adverse cytogenetic abnormalities are associated with a poor outcome. We evaluated the efficacy of ibrutinib, a covalent inhibitor of Brutons tyrosine kinase, in patients at risk for a poor outcome. METHODS In this multicenter, open-label, phase 3 study, we randomly assigned 391 patients with relapsed or refractory CLL or SLL to receive daily ibrutinib or the anti-CD20 antibody ofatumumab. The primary end point was the duration of progression-free survival, with the duration of overall survival and the overall response rate as secondary end points. RESULTS At a median follow-up of 9.4 months, ibrutinib significantly improved progression-free survival; the median duration was not reached in the ibrutinib group (with a rate of progression-free survival of 88% at 6 months), as compared with a median of 8.1 months in the ofatumumab group (hazard ratio for progression or death in the ibrutinib group, 0.22; P<0.001). Ibrutinib also significantly improved overall survival (hazard ratio for death, 0.43; P=0.005). At 12 months, the overall survival rate was 90% in the ibrutinib group and 81% in the ofatumumab group. The overall response rate was significantly higher in the ibrutinib group than in the ofatumumab group (42.6% vs. 4.1%, P<0.001). An additional 20% of ibrutinib-treated patients had a partial response with lymphocytosis. Similar effects were observed regardless of whether patients had a chromosome 17p13.1 deletion or resistance to purine analogues. The most frequent nonhematologic adverse events were diarrhea, fatigue, pyrexia, and nausea in the ibrutinib group and fatigue, infusion-related reactions, and cough in the ofatumumab group. CONCLUSIONS Ibrutinib, as compared with ofatumumab, significantly improved progression-free survival, overall survival, and response rate among patients with previously treated CLL or SLL. (Funded by Pharmacyclics and Janssen; RESONATE ClinicalTrials.gov number, NCT01578707.).


The New England Journal of Medicine | 2014

PI3Kδ inhibition by idelalisib in patients with relapsed indolent lymphoma

Ajay K. Gopal; Brad S. Kahl; Sven de Vos; Nina D. Wagner-Johnston; Stephen J. Schuster; Wojciech Jurczak; Ian W. Flinn; Christopher R. Flowers; Peter Martin; Andreas Viardot; Kristie A. Blum; Andre Goy; Andrew Davies; Pier Luigi Zinzani; Martin Dreyling; Dave Johnson; Langdon L. Miller; Leanne Holes; Daniel Li; Roger Dansey; Wayne R. Godfrey; Gilles Salles

BACKGROUND Phosphatidylinositol-3-kinase delta (PI3Kδ) mediates B-cell receptor signaling and microenvironmental support signals that promote the growth and survival of malignant B lymphocytes. In a phase 1 study, idelalisib, an orally active selective PI3Kδ inhibitor, showed antitumor activity in patients with previously treated indolent non-Hodgkins lymphomas. METHODS In this single-group, open-label, phase 2 study, 125 patients with indolent non-Hodgkins lymphomas who had not had a response to rituximab and an alkylating agent or had had a relapse within 6 months after receipt of those therapies were administered idelalisib, 150 mg twice daily, until the disease progressed or the patient withdrew from the study. The primary end point was the overall rate of response; secondary end points included the duration of response, progression-free survival, and safety. RESULTS The median age of the patients was 64 years (range, 33 to 87); patients had received a median of four prior therapies (range, 2 to 12). Subtypes of indolent non-Hodgkins lymphoma included follicular lymphoma (72 patients), small lymphocytic lymphoma (28), marginal-zone lymphoma (15), and lymphoplasmacytic lymphoma with or without Waldenströms macroglobulinemia (10). The response rate was 57% (71 of 125 patients), with 6% meeting the criteria for a complete response. The median time to a response was 1.9 months, the median duration of response was 12.5 months, and the median progression-free survival was 11 months. Similar response rates were observed across all subtypes of indolent non-Hodgkins lymphoma, though the numbers were small for some categories. The most common adverse events of grade 3 or higher were neutropenia (in 27% of the patients), elevations in aminotransferase levels (in 13%), diarrhea (in 13%), and pneumonia (in 7%). CONCLUSIONS In this single-group study, idelalisib showed antitumor activity with an acceptable safety profile in patients with indolent non-Hodgkins lymphoma who had received extensive prior treatment. (Funded by Gilead Sciences and others; ClinicalTrials.gov number, NCT01282424.).


Transplantation | 2001

Reduction in immunosuppression as initial therapy for posttransplant lymphoproliferative disorder: analysis of prognostic variables and long-term follow-up of 42 adult patients.

Donald E. Tsai; Christine Hardy; John E. Tomaszewski; Robert M. Kotloff; Kimberly M. Oltoff; Bradley G. Somer; Stephen J. Schuster; David L. Porter; Kathleen T. Montone; Edward A. Stadtmauer

Background. Posttransplant lymphoproliferative disorder (PTLD) is an Epstein-Barr virus–associated malignancy that occurs in the setting of pharmacologic immunosuppression after organ transplantation. With the increased use of organ transplantation and intensive immunosuppression, this disease is becoming more common. We explore reduction in immunosuppression as an initial therapy for PTLD. Methods. We analyzed our organ transplant patient database to identify patients with biopsy-proven PTLD who were initially treated with reduction of their immunosuppressive medications with or without surgical resection of all known disease. Results. Forty-two adult patients were included in this study. Thirty patients were treated with reduction in immunosuppression alone. Twelve patients were treated with both reduction in immunosuppression and surgical resection of all known disease. Thirty-one of 42 patients (73.8%) achieved a complete remission. Of those patients who were treated with reduction in immunosuppression alone, 19 of 30 (63%) responded with a median time to documentation of response of 3.6 weeks. Multivariable analysis showed that elevated lactate dehydrogenase (LDH) ratio, organ dysfunction, and multi-organ involvement by PTLD were independent prognostic factors for lack of response to reduction in immunosuppression. In patients with none of these poor prognostic factors, 16 of 18 (89%) responded to reduction in immunosuppression in contrast to three of five (60%) with one risk factor and zero of seven (0%) with two to three factors present. The analysis also showed that increased age, elevated LDH ratio, severe organ dysfunction, presence of B symptoms (fever, night sweats, and weight loss), and multi-organ involvement by PTLD at the time of diagnosis are independent prognostic indicators for poor survival. With median follow-up of 147 weeks, 55% of patients are alive with 50% in complete remission. Conclusions. Reduction in immunosuppression is an effective initial therapy for PTLD. Clinical prognostic factors may allow clinicians to identify which patients are likely to respond to reduction in immuno- suppression.


Journal of Clinical Oncology | 2016

Nivolumab in Patients With Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study

Alexander M. Lesokhin; Stephen M. Ansell; Philippe Armand; Emma C. Scott; Ahmad Halwani; Martin Gutierrez; Michael Millenson; Adam D. Cohen; Stephen J. Schuster; Daniel Lebovic; Madhav V. Dhodapkar; David Avigan; Bjoern Chapuy; Azra H. Ligon; Gordon J. Freeman; Scott J. Rodig; Deepika Cattry; Lili Zhu; Joseph F. Grosso; M. Brigid Bradley Garelik; Margaret A. Shipp; Ivan Borrello; John M. Timmerman

PURPOSE Cancer cells can exploit the programmed death-1 (PD-1) immune checkpoint pathway to avoid immune surveillance by modulating T-lymphocyte activity. In part, this may occur through overexpression of PD-1 and PD-1 pathway ligands (PD-L1 and PD-L2) in the tumor microenvironment. PD-1 blockade has produced significant antitumor activity in solid tumors, and similar evidence has emerged in hematologic malignancies. METHODS In this phase I, open-label, dose-escalation, cohort-expansion study, patients with relapsed or refractory B-cell lymphoma, T-cell lymphoma, and multiple myeloma received the anti-PD-1 monoclonal antibody nivolumab at doses of 1 or 3 mg/kg every 2 weeks. This study aimed to evaluate the safety and efficacy of nivolumab and to assess PD-L1/PD-L2 locus integrity and protein expression. RESULTS Eighty-one patients were treated (follicular lymphoma, n = 10; diffuse large B-cell lymphoma, n = 11; other B-cell lymphomas, n = 10; mycosis fungoides, n = 13; peripheral T-cell lymphoma, n = 5; other T-cell lymphomas, n = 5; multiple myeloma, n = 27). Patients had received a median of three (range, one to 12) prior systemic treatments. Drug-related adverse events occurred in 51 (63%) patients, and most were grade 1 or 2. Objective response rates were 40%, 36%, 15%, and 40% among patients with follicular lymphoma, diffuse large B-cell lymphoma, mycosis fungoides, and peripheral T-cell lymphoma, respectively. Median time of follow-up observation was 66.6 weeks (range, 1.6 to 132.0+ weeks). Durations of response in individual patients ranged from 6.0 to 81.6+ weeks. CONCLUSION Nivolumab was well tolerated and exhibited antitumor activity in extensively pretreated patients with relapsed or refractory B- and T-cell lymphomas. Additional studies of nivolumab in these diseases are ongoing.


Clinical Cancer Research | 2004

Epratuzumab, a Humanized Anti-CD22 Antibody, in Aggressive Non-Hodgkin’s Lymphoma: Phase I/II Clinical Trial Results

John P. Leonard; Morton Coleman; Jamie Ketas; Amy Chadburn; Richard R. Furman; Michael W. Schuster; Eric J. Feldman; Michelle Ashe; Stephen J. Schuster; William A. Wegener; Hans J. Hansen; Heather Ziccardi; Michael Eschenberg; Urte Gayko; Scott Z. Fields; Alessandra Cesano; David M. Goldenberg

Purpose: We conducted a single-center, dose-escalation study evaluating the safety, pharmacokinetics, and efficacy of epratuzumab, an anti-CD22 humanized monoclonal antibody, in patients with aggressive non-Hodgkin’s lymphoma. Experimental Design: Epratuzumab was administered once weekly for 4 weeks at 120-1000-mg/m2 doses to 56 patients [most (n = 35) with diffuse large B-cell lymphoma]. Results: Patients were heavily pretreated (median, 4 prior therapies), 25% received prior high-dose chemotherapy with stem cell transplant, and 84% had bulky disease (≥5 cm). Epratuzumab was well tolerated, with no dose-limiting toxicity. Most (95%) infusions were completed within 1 h. The mean serum half-life was 23.9 days. Across all dose levels and histologies, objective responses (ORs) were observed in five patients (10%; 95% confidence interval, 3–21%), including three complete responses. In patients with diffuse large B-cell lymphoma, 15% had ORs. Overall, 11 (20%) patients experienced some tumor mass reduction. Median duration of OR was 26.3 weeks, and median time to progression for responders was 35 weeks. Two responses are ongoing at ≥34 months, including one rituximab-refractory patient. Conclusions: These data demonstrate that epratuzumab has a good safety profile and exerts antitumor activity in aggressive non-Hodgkin’s lymphoma at doses of ≥240 mg/m2, thus warranting further evaluation in this clinical setting.


American Journal of Transplantation | 2006

Treatment of PTLD with Rituximab or Chemotherapy

Rebecca Elstrom; Charalambos Andreadis; Nicole A. Aqui; V. Ahya; Roy D. Bloom; Susan C. Brozena; K. Olthoff; Stephen J. Schuster; Sunita D. Nasta; Edward A. Stadtmauer; Donald E. Tsai

Information regarding treatment of post‐transplant lymphoproliferative disease (PTLD) beyond reduction in immunosuppression (RI) is limited. We retrospectively evaluated patients receiving rituximab and/or chemotherapy for PTLD for response, time to treatment failure (TTF) and overall survival (OS). Thirty‐five patients met inclusion criteria. Twenty‐two underwent rituximab treatment, with overall response rate (ORR) 68%. Median TTF was not reached at 19 months and estimated OS was 31 months. In univariable analysis, Epstein‐Barr virus (EBV) positivity predicted response and TTF. LDH elevation predicted shorter OS. No patient died of rituximab toxicity and all patients who progressed underwent further treatment with chemotherapy. Twenty‐three patients received chemotherapy. ORR was 74%, median TTF was 10.5 months and estimated OS was 42 months. Prognostic factors for response included stage, LDH and allograft involvement by tumor. These factors and lack of complete response (CR) predicted poor survival. Twenty‐six percent of the patients receiving chemotherapy died of toxicity. Rituximab and chemotherapy are effective in patients with PTLD who fail or do not tolerate RI. While rituximab is well tolerated, toxicity of chemotherapy is marked. PTLD patients requiring therapy beyond RI should be considered for rituximab, especially with EBV‐positive disease. Chemotherapy should be reserved for patients who fail rituximab, have EBV‐negative tumors or need a rapid response.


Journal of Clinical Oncology | 2011

Vaccination With Patient-Specific Tumor-Derived Antigen in First Remission Improves Disease-Free Survival in Follicular Lymphoma

Stephen J. Schuster; Sattva S. Neelapu; Barry L. Gause; John E. Janik; Franco M. Muggia; Jon P. Gockerman; Jane N. Winter; Christopher R. Flowers; Daniel A. Nikcevich; Eduardo M. Sotomayor; Dean McGaughey; Elaine S. Jaffe; Elise A. Chong; Craig W. Reynolds; Donald A. Berry; Carlos F. Santos; Mihaela Popa; Amy M. McCord; Larry W. Kwak

PURPOSE Vaccination with hybridoma-derived autologous tumor immunoglobulin (Ig) idiotype (Id) conjugated to keyhole limpet hemocyanin (KLH) and administered with granulocyte-monocyte colony-stimulating factor (GM-CSF) induces follicular lymphoma (FL) -specific immune responses. To determine the clinical benefit of this vaccine, we conducted a double-blind multicenter controlled phase III trial. PATIENTS AND METHODS Treatment-naive patients with advanced stage FL achieving complete response (CR) or CR unconfirmed (CRu) after chemotherapy were randomly assigned two to one to receive either Id vaccine (Id-KLH + GM-CSF) or control (KLH + GM-CSF). Primary efficacy end points were disease-free survival (DFS) for all randomly assigned patients and DFS for randomly assigned patients receiving at least one dose of Id vaccine or control. RESULTS Of 234 patients enrolled, 177 (81%) achieved CR/CRu after chemotherapy and were randomly assigned. For 177 randomly assigned patients, including 60 patients not vaccinated because of relapse (n = 55) or other reasons (n = 5), median DFS between Id-vaccine and control arms was 23.0 versus 20.6 months, respectively (hazard ratio [HR], 0.81; 95% CI, 0.56 to 1.16; P = .256). For 117 patients who received Id vaccine (n = 76) or control (n = 41), median DFS after randomization was 44.2 months for Id-vaccine arm versus 30.6 months for control arm (HR, 0.62; 95% CI, 0.39 to 0.99; P = .047) at median follow-up of 56.6 months (range, 12.6 to 89.3 months). In an unplanned subgroup analysis, median DFS was significantly prolonged for patients receiving IgM-Id (52.9 v 28.7 months; P = .001) but not IgG-Id vaccine (35.1 v 32.4 months; P = .807) compared with isotype-matched control-treated patients. CONCLUSION Vaccination with patient-specific hybridoma-derived Id vaccine after chemotherapy-induced CR/CRu may prolong DFS in patients with FL. Vaccine isotype may affect clinical outcome and explain differing results between this and other controlled Id-vaccine trials.


Cancer | 2008

Durable complete responses from therapy with combined epratuzumab and rituximab: final results from an international multicenter, phase 2 study in recurrent, indolent, non-Hodgkin lymphoma.

John P. Leonard; Stephen J. Schuster; Christos Emmanouilides; Felix Couture; Nick Teoh; William A. Wegener; Morton Coleman; David M. Goldenberg

In this international, multicenter trial, the authors evaluated rituximab (anti‐CD20) plus epratuzumab (anti‐CD22) in patients with postchemotherapy relapsed/refractory, indolent non‐Hodgkin lymphoma (NHL), including long‐term efficacy.


British Journal of Haematology | 1992

Cellular sites of extrarenal and renal erythropoietin production in anaemic rats

Stephen J. Schuster; Stephen T. Koury; M. Bohrer; S. Salceda; J. Card

The cellular origins of erythropoietin were investigated in the rat using a probe derived from a cloned rat erythropoietin cDNA. In anaemic‐hypoxic rat liver, in situ hybridization detected erythropoietin mRNA primarily in hepatocytes and less frequently in nonparenchymal sinusoidal or perisinusoidal liver cells. An RNase protection assay was used to compare the erythropoietin mRNA contents of separated rat liver cell fractions and also suggested that hepatocytes are the major source of extrarenal erythropoietin with nonparenchymal liver cells contributing less than 1% to total hepatic erythropoietin production. In kidney, in situ hybridization localized erythropoietin mRNA in nonepithelial cells, as yet of undefined lineage, in the cortical and outer medullary interstitium. These results indicate that, in the rat, the primary sources of erythropoietin in liver and kidney are different types of cells.

Collaboration


Dive into the Stephen J. Schuster's collaboration.

Top Co-Authors

Avatar

Sunita D. Nasta

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Jakub Svoboda

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

David L. Porter

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Donald E. Tsai

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Anthony R. Mato

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Selina M. Luger

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Alison W. Loren

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Elise A. Chong

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge