Network


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

Hotspot


Dive into the research topics where Sarah Bernstein is active.

Publication


Featured researches published by Sarah Bernstein.


Journal of Immunotherapy | 1999

A phase I vaccine trial with peptides reflecting ras oncogene mutations of solid tumors.

Samir N. Khleif; Scott I. Abrams; J. Michael Hamilton; Elke S. Bergmann-Leitner; Alice Chen; Anne Bastian; Sarah Bernstein; Yoomie Chung; Carmen J. Allegra; Jeffrey Schlom

Mutations in the ras genes occur in 20% of all human cancers. These genes, in turn, produce mutated proteins that are unique to cancer cells, rendering them distinguishable from normal cells by the immune system. Thus, mutated Ras proteins may form potential targets for immune therapy. We conducted a phase I/pilot clinical trial in patients with advanced cancers to test the toxicity and the ability to induce an immune response by vaccination with 13-mer mutated Ras peptides reflecting codon 12 mutations. These peptides corresponded to each of the patients own tumor Ras mutation. Patients were vaccinated monthly x3 subcutaneously with the specific Ras peptide along with Detox adjuvant (RiBi ImmunoChem Research, Inc., Hamilton, MT, U.S.A.) at one of five different peptide dose levels (100, 500, 1,000, 1,500, and 5,000 micrograms). Three out of 10 evaluable patients generated a mutant Ras specific CD4+ and/or CD8+ T-cell immune response. The CD8+ cytotoxic cells specific for Gly to Val mutation at codon 12 were capable of lysing an HLA-A2-matched tumor cell line carrying the corresponding mutant but not the wild-type ras gene. The treatment has been well tolerated with no evidence of serious acute or delayed systemic side effects on any of the five dose levels. We demonstrated that we can generate in cancer patients specific T-lymphocyte responses that detect single amino acid differences in Ras oncoproteins. Neither the immune responses nor the minor side effects seen were found to be dose dependent. This approach may provide a unique opportunity for generating a tumor-directed therapy. Also, in vitro stimulation of these cells with the corresponding peptide generated specific T-cell lines that could be used for adoptive immune therapy.


Journal of Translational Medicine | 2014

The immunological and clinical effects of mutated ras peptide vaccine in combination with IL-2, GM-CSF, or both in patients with solid tumors

Osama E. Rahma; J M Hamilton; Malgorzata Wojtowicz; Omar Dakheel; Sarah Bernstein; David J. Liewehr; Seth M. Steinberg; Samir N. Khleif

BackgroundMutant Ras oncogenes produce proteins that are unique to cancer cells and represent attractive targets for vaccine therapy. We have shown previously that vaccinating cancer patients with mutant ras peptides is feasible and capable of inducing a specific immune response against the relevant mutant proteins. Here, we tested the mutant ras peptide vaccine administered in combination with low dose interleukin-2 (IL-2) or/and granulocyte-macrophage colony-stimulating factor (GM-CSF) in order to enhance the vaccine immune response.Methods5000μg of the corresponding mutant ras peptide was given subcutaneously (SQ) along with IL-2 (Arm A), GM-CSF (Arm B) or both (Arm C). IL-2 was given SQ at 6.0 million IU/m2/day starting at day 5, 5 days/week for 2 weeks. GM-CSF was given SQ in a dose of 100μg/day one day prior to each ras peptide vaccination for 4 days. Vaccines were repeated every 5 weeks on arm A and C, and every 4 weeks on arm B, for a maximum of 15 cycles or until disease progression.ResultsWe treated 53 advanced cancer patients (38 with colorectal, 11 with pancreatic, 1 with common bile duct and 3 with lung) on 3 different arms (16 on arm A, 18 on arm B, and 19 on arm C). The median progression free survival (PFS) and overall survival (OS) was 3.6 and 16.9 months, respectively, for all patients evaluable for clinical response (n = 48). There was no difference in PFS or OS between the three arms (P = 0.73 and 0.99, respectively). Most adverse events were grade 1-2 toxicities and resolved spontaneously. The vaccine induced an immune response to the relevant ras peptide in a total of 20 out of 37 evaluable patients (54%) by ELISPOT, proliferative assay, or both. While 92.3% of patients on arm B had a positive immune response, only 31% of patients on arm A and 36% of patients on arm C had positive immune responses (P = 0.003, Fisher’s exact test).ConclusionsThe reported data showed that IL-2 might have a negative effect on the specific immune response induced by the relevant mutant ras vaccine in patients with advanced cancer. This observation deserves further investigations.Trial registrationNCI97C0141


Journal of Translational Medicine | 2010

A pilot clinical trial testing mutant von Hippel- Lindau peptide as a novel immune therapy in metastatic Renal Cell Carcinoma

Osama E. Rahma; Ed Ashtar; Ramy Ibrahim; Antoun Toubaji; B. Gause; V. E. Herrin; W. Marston Linehan; Seth M. Steinberg; Frank Grollman; George Grimes; Sarah Bernstein; Jay A. Berzofsky; Samir N. Khleif

BackgroundDue to the lack of specific tumor antigens, the majority of tested cancer vaccines for renal cell carcinoma (RCC) are based on tumor cell lysate. The identification of the von Hippel-Lindau (VHL) gene mutations in RCC patients provided the potential for developing a novel targeted vaccine for RCC. In this pilot study, we tested the feasibility of vaccinating advanced RCC patients with the corresponding mutant VHL peptides.MethodsSix patients with advanced RCC and mutated VHL genes were vaccinated with the relevant VHL peptides. Patients were injected with the peptide mixed with Montanide subcutaneously (SQ) every 4 weeks until disease progression or until the utilization of all available peptide stock.ResultsFour out of five evaluable patients (80%) generated specific immune responses against the corresponding mutant VHL peptides. The vaccine was well tolerated. No grade III or IV toxicities occurred. The median overall survival (OS) and median progression-free survival (PFS) were 30.5 and 6.5 months, respectively.ConclusionsThe vaccine demonstrated safety and proved efficacy in generating specific immune response to the mutant VHL peptide. Despite the fact that the preparation of these custom-made vaccines is time consuming, the utilization of VHL as a vaccine target presents a promising approach because of the lack of other specific targets for RCC. Accordingly, developing mutant VHL peptides as vaccines for RCC warrants further investigation in larger trials. Trial registration: 98C0139


Cancer Research | 2010

Abstract 2414: Comparable effect of p53 peptide vaccine in adjuvant or pulsed on dendritic cells in ovarian cancer patients: A gynecologic oncology group study

Osama E. Rahma; Ed Achtar; Malgorzata Czystowska; Marta Szajnik; Eva Wieckowski; Sarah Bernstein; V. E. Herrin; Seth M. Steinberg; Maria J. Merino; William E. Gooding; Carmen Visus; Albert B. DeLeo; Jay A. Berzofsky; Theresa L. Whiteside; Judith K. Wolf; Jeffrey Bell; Samir N. Khleif

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC Purpose: Peptide antigens have been administered by different approaches in cancer vaccine therapy, including direct injection or pulsed onto dendritic cells; however, the optimal delivery method is still debatable. In this study, we describe the immune response elicited by the wild-type (wt) p53 vaccine using two approaches. Experimental Design: Twenty-one HLA-A2.1 patients with stage III, IV, or recurrent ovarian cancer overexpressing the p53 protein with no evidence of disease were treated in two cohorts. Arm A received subcutaneous wt p53:264-272 peptide admixed with Montanide and GM-CSF. Arm B received wt p53:264-272 peptide-pulsed dendritic cells intravenously. Interleukin-2 (IL-2) was administered in both cohorts in alternative cycles. The immunologic response was assessed by ELISPOT and tetramer assays. Results: Nine of 13 patients (69%) in arm A and 5 of 6 patients (83%) in arm B had an immunologic response. The vaccine caused no serious systemic side effects. IL-2 administration resulted in grade 3 and 4 toxicities in both arms and directly induced expansion of T regulatory cells. The median overall survival was 40.8 and 29.6 months for arm A and B, respectively; the median progression-free survival was 4.2 and. 8.7 months, respectively. Conclusion: We demonstrated that using either vaccination approach could generate comparable specific immune responses against the wt p53 peptide with minimal toxicity. Accordingly, our findings support the utilization of the less demanding subcutaneous method. Furthermore, as IL-2 may add toxicity and induce T regulatory cells, which can suppress the immune response, therefore, it may not be needed in future trials. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2414.


Cancer Immunology, Immunotherapy | 2008

Pilot study of mutant ras peptide-based vaccine as an adjuvant treatment in pancreatic and colorectal cancers

Antoun Toubaji; Moujahed Achtar; Maurizio Provenzano; V. E. Herrin; Robert Behrens; Michael Hamilton; Sarah Bernstein; David Venzon; B. Gause; Francesco M. Marincola; Samir N. Khleif


Cancer Immunology, Immunotherapy | 2012

A gynecologic oncology group phase II trial of two p53 peptide vaccine approaches: subcutaneous injection and intravenous pulsed dendritic cells in high recurrence risk ovarian cancer patients

Osama E. Rahma; Ed Ashtar; Malgorzata Czystowska; Marta Szajnik; Eva Wieckowski; Sarah Bernstein; V. E. Herrin; Mortada A. Shams; Seth M. Steinberg; Maria J. Merino; William E. Gooding; Carmen Visus; Albert B. DeLeo; Judith K. Wolf; Jeffrey Bell; Jay A. Berzofsky; Theresa L. Whiteside; Samir N. Khleif


Journal of Translational Medicine | 2014

Pre-immature dendritic cells (PIDC) pulsed with HPV16 E6 or E7 peptide are capable of eliciting specific immune response in patients with advanced cervical cancer.

Osama E. Rahma; V. E. Herrin; Rami A Ibrahim; Anton Toubaji; Sarah Bernstein; Omar Dakheel; Seth M. Steinberg; Rasha Abu Eid; Mikayel Mkrtichyan; Jay A. Berzofsky; Samir N. Khleif


Journal of Clinical Oncology | 2004

Mutant P53 vaccination of patients with advanced cancers generates specific immunological responses

Ramy Ibrahim; M. S. Achtar; V. E. Herrin; B. Gause; Sarah Bernstein; T. B. Brent-Steel; C. M. Maruffi; Jay A. Berzofsky; Samir N. Khleif


Journal of Clinical Oncology | 2005

Pre-immature dendritic cells pulsed with human papillomavirus 16 E7 peptide vaccine in advanced cervical cancer

M. S. Achtar; Ramy Ibrahim; V. E. Herrin; B. Gause; Seth M. Steinberg; F. Grollman; Osama E. Rahma; Sarah Bernstein; C. M. Maruffi; Jay A. Berzofsky; Samir N. Khleif


Journal of Clinical Oncology | 2005

Autologous PBMCs pulsed with HPV16 E6 as an immunotherapeutic agent for patients with advanced cervical cancer

Ramy Ibrahim; M. S. Achtar; V. E. Herrin; B. Gause; Sarah Bernstein; F. Grollman; D. Contoise; Seth M. Steinberg; Jay A. Berzofsky; Samir N. Khleif

Collaboration


Dive into the Sarah Bernstein's collaboration.

Top Co-Authors

Avatar

Samir N. Khleif

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

V. E. Herrin

University of Mississippi Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jay A. Berzofsky

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Seth M. Steinberg

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ramy Ibrahim

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Antoun Toubaji

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Michael Hamilton

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carmen Visus

University of Pittsburgh

View shared research outputs
Researchain Logo
Decentralizing Knowledge