Network


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

Hotspot


Dive into the research topics where Jarrod P. Holmes is active.

Publication


Featured researches published by Jarrod P. Holmes.


Cancer | 2012

Clinical trial results of the HER-2/neu (E75) vaccine to prevent breast cancer recurrence in high-risk patients: from US Military Cancer Institute Clinical Trials Group Study I-01 and I-02.

Elizabeth A. Mittendorf; Guy T. Clifton; Jarrod P. Holmes; Kevin S. Clive; Ritesh Patil; Linda C. Benavides; Jeremy D. Gates; Alan K. Sears; Alexander Stojadinovic; Sathibalan Ponniah; George E. Peoples

The authors conducted exploratory phase 1‐2 clinical trials vaccinating breast cancer patients with E75, a human leukocyte antigen (HLA) A2/A3–restricted HER‐2/neu (HER2) peptide, and granulocyte‐macrophage colony‐stimulating factor. The vaccine is given as adjuvant therapy to prevent disease recurrence. They previously reported that the vaccine is safe and effective in stimulating expansion of E75‐specific cytotoxic T cells. Here, they report 24‐month landmark analyses of disease‐free survival (DFS).


Annals of Oncology | 2014

Final report of the phase I/II clinical trial of the E75 (nelipepimut-S) vaccine with booster inoculations to prevent disease recurrence in high-risk breast cancer patients

Elizabeth A. Mittendorf; G. T. Clifton; Jarrod P. Holmes; E. Schneble; D. van Echo; Sathibalan Ponniah; G. E. Peoples

BACKGROUND E75 (nelipepimut-S) is a human leukocyte antigen (HLA)-A2/A3-restricted immunogenic peptide derived from the HER2 protein. We have conducted phase I/II clinical trials vaccinating breast cancer patients with nelipepimut-S and granulocyte-macrophage colony-stimulating factor (GM-CSF) in the adjuvant setting to prevent disease recurrence. All patients have completed 60 months follow-up, and here, we report the final analyses. PATIENTS AND METHODS The studies were conducted as dose escalation/schedule optimization trials enrolling node-positive and high-risk node-negative patients with tumors expressing any degree of HER2 (immunohistochemistry 1-3+). HLA-A2/3+ patients were vaccinated; others were followed prospectively as controls. Local and systemic toxicity was monitored. Clinical recurrences were documented, and disease-free survival (DFS) was analyzed by Kaplan-Meier curves; groups were compared using log-rank tests. RESULTS Of 195 enrolled patients, 187 were assessable: 108 (57.8%) in the vaccinated group (VG) and 79 (42.2%) in the control group (CG). The groups were well matched for clinicopathologic characteristics. Toxicities were minimal. Five-year DFS was 89.7% in the VG versus 80.2% in the CG (P = 0.08). Due to trial design, 65% of patients received less than the optimal vaccine dose. Five-year DFS was 94.6% in optimally dosed patients (P = 0.05 versus the CG) and 87.1% in suboptimally dosed patients. A voluntary booster program was initiated, and among the 21 patients that were optimally boosted, there was only one recurrence (DFS = 95.2%). CONCLUSION The E75 vaccine is safe and appears to have clinical efficacy. A phase III trial evaluating the optimal dose and including booster inoculations has been initiated. CLINICAL TRIALS NCT00841399, NCT00584789.BACKGROUND E75 (nelipepimut-S) is a human leukocyte antigen (HLA)-A2/A3-restricted immunogenic peptide derived from the HER2 protein. We have conducted phase I/II clinical trials vaccinating breast cancer patients with nelipepimut-S and granulocyte-macrophage colony-stimulating factor (GM-CSF) in the adjuvant setting to prevent disease recurrence. All patients have completed 60 months follow-up, and here, we report the final analyses. PATIENTS AND METHODS The studies were conducted as dose escalation/schedule optimization trials enrolling node-positive and high-risk node-negative patients with tumors expressing any degree of HER2 (immunohistochemistry 1-3+). HLA-A2/3+ patients were vaccinated; others were followed prospectively as controls. Local and systemic toxicity was monitored. Clinical recurrences were documented, and disease-free survival (DFS) was analyzed by Kaplan-Meier curves; groups were compared using log-rank tests. RESULTS Of 195 enrolled patients, 187 were assessable: 108 (57.8%) in the vaccinated group (VG) and 79 (42.2%) in the control group (CG). The groups were well matched for clinicopathologic characteristics. Toxicities were minimal. Five-year DFS was 89.7% in the VG versus 80.2% in the CG (P = 0.08). Due to trial design, 65% of patients received less than the optimal vaccine dose. Five-year DFS was 94.6% in optimally dosed patients (P = 0.05 versus the CG) and 87.1% in suboptimally dosed patients. A voluntary booster program was initiated, and among the 21 patients that were optimally boosted, there was only one recurrence (DFS = 95.2%). CONCLUSION The E75 vaccine is safe and appears to have clinical efficacy. A phase III trial evaluating the optimal dose and including booster inoculations has been initiated. CLINICAL TRIALS NCT00841399, NCT00584789.


Cancer | 2010

Results of the first phase 1 clinical trial of the HER-2/neu peptide (GP2) vaccine in disease-free breast cancer patients: United States Military Cancer Institute Clinical Trials Group Study I-04

Mark G. Carmichael; Linda C. Benavides; Jarrod P. Holmes; Jeremy D. Gates; Elizabeth A. Mittendorf; Sathibalan Ponniah; George E. Peoples

HER‐2/neu, overexpressed in breast cancer, is a source of immunogenic peptides that include GP2 and E75. Phase 2 testing of E75 as an adjuvant vaccine has suggested a clinical benefit. GP2, derived from the transmembrane portion of HER‐2/neu, has differing binding characteristics and may be more immunogenic than E75. Results of the first phase 1 trial of GP2 peptide vaccine are presented.


Cancer | 2008

Optimal dose and schedule of an HER-2/neu (E75) peptide vaccine to prevent breast cancer recurrence: from US Military Cancer Institute Clinical Trials Group Study I-01 and I-02.

Jarrod P. Holmes; Jeremy D. Gates; Linda C. Benavides; Matthew T. Hueman; Mark G. Carmichael; Ritesh Patil; Dianna Craig; Elizabeth A. Mittendorf; Alexander Stojadinovic; Sathibalan Ponniah; George E. Peoples

E75, a HER‐2/neu‐derived peptide, was administered as a preventive vaccine with granulocyte‐macrophage–colony‐stimulating factor (GM‐CSF) in disease‐free lymph node‐positive (NP) and lymph node‐negative (NN) breast cancer (BCa) patients. The optimal biologic dose (OBD) was determined based on toxicity and immunologic response.


Expert Review of Vaccines | 2010

Use of GM-CSF as an adjuvant with cancer vaccines: Beneficial or detrimental?

Kevin S. Clive; Josh Tyler; G. Travis Clifton; Jarrod P. Holmes; Elizabeth A. Mittendorf; Sathibalan Ponniah; George E. Peoples

Granulocyte–macrophage colony-stimulating factor (GM-CSF) has been utilized in the clinical management of multiple disease processes. Most recently, GM-CSF has been incorporated into the treatment of malignancies as a sole therapy, as well as a vaccine adjuvant. While the benefits of GM-CSF in this arena have been promising, recent reports have suggested the potential for GM-CSF to induce immune suppression and, thus, negatively impact outcomes in the management of cancer patients. The purpose of this review is to critically evaluate these reports, while considering the most recent clinical data on immunotherapies. We aim to demonstrate the utility of this adjuvant, elucidate those instances in which GM-CSF may induce immune suppression and identify potential explanations for these recent findings.


Cancer | 2011

Use of booster inoculations to sustain the clinical effect of an adjuvant breast cancer vaccine

Jarrod P. Holmes; Guy T. Clifton; Ritesh Patil; Linda C. Benavides; Jeremy D. Gates; Alexander Stojadinovic; Elizabeth A. Mittendorf; Sathibalan Ponniah; George E. Peoples

The authors are conducting clinical trials of the HER‐2/neu E75‐peptide vaccine in clinically disease‐free breast cancer (BC) patients. Their phase 1‐2 trials revealed that the E75 + granulocyte‐macrophage colony‐stimulating factor (GM‐CSF) vaccine is safe and effective in stimulating clonal expansion of E75‐specific CD8+ T cells. They assessed the need for and response to a booster after completion of primary vaccination series.


Expert Opinion on Biological Therapy | 2011

AE37: a novel T-cell-eliciting vaccine for breast cancer

Alan K. Sears; Sonia A. Perez; Guy T. Clifton; Linda C. Benavides; Jeremy D. Gates; Kevin S. Clive; Jarrod P. Holmes; Nathan M. Shumway; David C. Van Echo; M. G. Carmichael; Sathibalan Ponniah; Constantin N. Baxevanis; Elizabeth A. Mittendorf; Michael Papamichail; George E. Peoples

Introduction: Immunotherapy, including vaccines targeting the human EGFR2 (HER-2/neu) protein, is an active area of investigation in combatting breast cancer. Several vaccines are currently undergoing clinical trials, most of which are CD8+ T-cell-eliciting vaccines. AE37 is a promising primarily CD4+ T-cell-eliciting HER-2/neu breast cancer vaccine currently in clinical trials. Areas covered: This article reviews preclinical investigations as well as findings from completed and ongoing Phase I and Phase II clinical trials of the AE37 vaccine. Expert opinion: Clinical trials have shown the AE37 vaccine to be safe and capable of generating peptide-specific, durable immune responses. This has been shown in patients with any level of HER-2/neu expression. Early clinical findings suggest there may be benefit to AE37 vaccination in preventing breast cancer recurrence.


Journal of Surgical Oncology | 2012

The GP2 peptide: a HER2/neu-based breast cancer vaccine.

Kevin S. Clive; Josh Tyler; G. Travis Clifton; Jarrod P. Holmes; Sathibalan Ponniah; George E. Peoples; Elizabeth A. Mittendorf

Preclinical studies suggest that GP2, a HER2/neu‐derived peptide, is immunogenic. Subsequent phase I clinical trials demonstrated that GP2‐based vaccines are safe and effective in stimulating peptide‐specific immunity. A GP2 peptide vaccine is currently being evaluated in a phase II efficacy trial enrolling breast cancer patients. This article reviews initial studies characterizing GP2, clinical trials investigating GP2‐based vaccines, and novel immunotherapy strategies incorporating GP2 in combination with other peptides or with the monoclonal antibody trastuzumab. J. Surg. Oncol. 2012; 105:452–458.


Expert Review of Vaccines | 2011

Comparison of different HER2/neu vaccines in adjuvant breast cancer trials: implications for dosing of peptide vaccines

Linda C. Benavides; Alan K. Sears; Jeremy D. Gates; Guy T. Clifton; Kevin S. Clive; Mark G. Carmichael; Jarrod P. Holmes; Elizabeth A. Mittendorf; Sathibalan Ponniah; George E. Peoples

We have performed multiple adjuvant clinical trials using immunogenic peptides from the HER2/neu protein (AE37/E75/GP2) plus (GM-CSF) given intradermally to breast cancer patients. Four trials were performed with similar dose-escalation design with increasing doses of peptide (AE37/E75/GP2) and varying amounts of GM-CSF. Dose reductions (DRs) were made for significant local and/or systemic toxicity by decreasing GM-CSF for subsequent inoculations. Ex vivo and in vivo immunologic responses were used to compare groups. Of 132 patients, 39 required DR (30 for robust local reactions [DR-L]). DR patients, particularly DR-L, had greater immune responses both ex vivo and in vivo. Postvaccine delayed-type hypersensitivity in DR-L patients compared with all others was larger for E75 (p = 0.001), AE37 (p = 0.077) and GP2 (p = 0.076). All three peptide vaccines were safe and well-tolerated. These findings have led to a clinically relevant optimal vaccine dosing strategy, which may be applicable to other peptide-based cancer vaccines.


Vaccine | 2010

Circulating regulatory T cells (CD4+CD25+FOXP3+) decrease in breast cancer patients after vaccination with a modified MHC class II HER2/neu (AE37) peptide.

Jeremy D. Gates; Guy T. Clifton; Linda C. Benavides; Alan K. Sears; Mark G. Carmichael; Matthew T. Hueman; Jarrod P. Holmes; Yusuf Jama; Mohamed Mursal; Athina Zacharia; Kathy Ciano; Steven Khoo; Alexander Stojadinovic; Sathibalan Ponniah; George E. Peoples

Regulatory T cells (T(Reg)), CD4(+)CD25(+)FOXP3(+), are implicated in suppressing tumor immune responses. We analyzed peripheral blood lymphocytes (PBL) from breast cancer patients receiving a modified HLA class II HER2/neu peptide (AE37) vaccine for T(Reg) cells and correlated their levels with vaccine-specific immune responses. The mean CD4(+)CD25(+)FOXP3(+) T(Reg) cells decreased in patients with vaccination with no significant difference in serum TGF-β levels. IFN-γ ELISPOT and DTH increased after vaccination with a good correlation between T(Reg) cell reduction and size of DTH to AE37. The T(Reg) cell reduction and associated immune response suggest that AE37 may be clinically useful.

Collaboration


Dive into the Jarrod P. Holmes's collaboration.

Top Co-Authors

Avatar

Sathibalan Ponniah

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Elizabeth A. Mittendorf

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

George E. Peoples

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Guy T. Clifton

San Antonio Military Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ritesh Patil

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Mark G. Carmichael

Walter Reed Army Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alan K. Sears

San Antonio Military Medical Center

View shared research outputs
Top Co-Authors

Avatar

Matthew T. Hueman

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Alexander Stojadinovic

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Ge Peoples

San Antonio Military Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge