Network


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

Hotspot


Dive into the research topics where Tj Vreeland is active.

Publication


Featured researches published by Tj Vreeland.


Expert Review of Clinical Immunology | 2016

Gaining ground on a cure through synergy: combining checkpoint inhibitors with cancer vaccines

Tj Vreeland; Gt Clifton; Garth S. Herbert; Diane F. Hale; Doreen O. Jackson; J. S. Berry; George E. Peoples

ABSTRACT Introduction: The approval of multiple checkpoint inhibitors (CPIs) for the treatment of advanced malignancies has sparked an explosion of research in the field of cancer immunotherapy. Despite the success of these medications, a large number of patients with advanced malignancy do not benefit from therapy. Early research indicates that a therapeutic combination of cancer vaccines with checkpoint inhibitors may lead to synergistic effects and higher response rates than monotherapy. Areas covered: This paper summarizes the previously completed and ongoing research on this exciting combination, including the use of the tumor lysate, particle-loaded dendritic cell (TLPLDC) vaccine combined with checkpoint inhibitors in advanced melanoma. Expert commentary: Increasing experience with CPIs has led to improved understanding of which patients may benefit and it is increasingly clear that the presence of a pre-existing immune response to the tumor, along with tumor-infiltrating lymphocytes, is key to the success of CPIs. One exciting possibility for the future is the addition of a cancer vaccine to CPI therapy, eliciting these crucial T cells, which can then be augmented and protected by the CPI. A number of current and future studies are addressing this very exciting combination therapy.


Expert Review of Clinical Immunology | 2017

Cancer vaccines in colon and rectal cancer over the last decade: lessons learned and future directions

John W. Berry; Tj Vreeland; Alfred F. Trappey; Diane F. Hale; Kaitlin M. Peace; Joshua A. Tyler; Avery Walker; Ramon Brown; Garth S. Herbert; Fia Yi; Doreen O. Jackson; Gt Clifton; George E. Peoples

ABSTRACT Introduction: Great advances have been made in screening for and treatment of colorectal cancer (CRC), but recurrence rates remain high and additional therapies are needed. There is great excitement around the field of immunotherapy and many attempts have been made to bring immunotherapy to CRC through a cancer vaccine. Areas covered: This is a detailed review of the last decade’s significant CRC vaccine trials. Expert commentary: Monotherapy with a CRC vaccine is likely best suited for adjuvant therapy in disease free patients. Vaccine therapy elicits crucial tumor infiltrating lymphocytes, which are lacking in microsatellite-stable tumors, and therefore may be better suited for these patients. The combination of CRC vaccines with checkpoint inhibitors may unlock the potential of immunotherapy for a much broader range of patients. Future studies should focus on vaccine monotherapy in correctly selected patients and combination therapy in more advanced disease.


Cancer Research | 2012

Abstract P5-16-02: Final Results of the Phase I/II Trials of the E75 Adjuvant Breast Cancer Vaccine

Tj Vreeland; Gt Clifton; Diane F. Hale; Alan K. Sears; Ritesh Patil; Jarrod P. Holmes; Sathibalan Ponniah; Elizabeth A. Mittendorf; Ge Peoples

Background: We have completed phase I/II clinical trials vaccinating breast cancer patients (pts) with E75, a HLA-A2/A3-restricted HER2/neu (HER2) peptide vaccine. The vaccine was administered in the adjuvant setting to prevent recurrences in high risk patients rendered disease-free with standard of care therapy. We have previously reported preliminary results indicating that the vaccine (including booster inoculations) is safe and effective in stimulating an anti-tumor immune response. Here, we report the final 5 year results from these trials. Methods: The phase I/II trials were performed as dose-escalation/schedule-optimization trials enrolling node positive and high-risk, node negative breast cancer patients with tumors expressing any level of HER2. HLA-A2/A3+ pts were enrolled into the vaccine group (VG) while HLA-A2/A3- pts were followed prospectively as the untreated control group (CG). The VG pts were given 4–6 monthly intradermal inoculations of E75 with GM-CSF during the primary vaccine series (PVS). In addition, a voluntary booster program was initiated during the trial, with booster inoculations being offered every 6 months after completion of the PVS. Patients were monitored for local and systemic toxicity (graded by NCI Common Terminology Criteria for Adverse Events). In vivo immune response was assessed in the VG by delayed type hypersensitivity (DTH) reactions to both E75 and saline, pre- and post-PVS. VG and CG pts were followed for 60 months (mo) and recurrences were documented. Demographic differences were compared with the Fisher9s exact test and disease-free survival was determined using the Kaplan-Meier method and compared by log-rank test. Results: 195 pts were enrolled, 6 withdrew (2 from VG, 4 from CG), 1 was lost to follow-up prior to vaccination, and 1 was found to be ineligible, leaving 187 evaluable pts; 108 in the VG and 79 in the CG. 53 pts volunteered for the booster program and received at least one booster inoculation. The VG and CG were well-matched with the only statistically significant difference being ER−/PR- status (31.1% in VG vs 17.7% in CG, p = 0.04). Vaccination was well tolerated (maximum local toxicity: 73.1% Grade 1, 26.9% Grade 2, 0% Grade 3; maximum systemic toxicity: 72.2% Grade 1, 15.7% Grade 2, and 2.8% Grade 3). In the VG, pre- to post-PVS E75 DTH significantly increased (mean 3.8 ±1.0 vs 14.8±1.4, p Conclusions: The E75 breast cancer vaccine is safe and well–tolerated. It elicits strong immune responses in vaccinated patients. At the end of the 5 year follow-up period, the E75 vaccine shows a strong trend toward preventing breast cancer recurrence in vaccinated patients. To investigate this vaccine (now known as NeuVax) further, the PRESENT trial, a prospective, randomized, double-blind, placebo-controlled, multi-center phase III registration trial has been initiated and is actively enrolling. Citation Information: Cancer Res 2012;72(24 Suppl):Abstract nr P5-16-02.


Cancer Research | 2011

P1-13-02: Long-Term Clinical Benefit of Adjuvant Breast Cancer Vaccine: 5 Year Efficacy of E75 with Multiple Booster Inoculations.

Tj Vreeland; Gt Clifton; Alan K. Sears; Diane F. Hale; Ritesh Patil; Kevin S. Clive; Jarrod P. Holmes; Elizabeth A. Mittendorf; Sathibalan Ponniah; Ge Peoples

Background: We are conducting phase I/II clinical trials vaccinating breast cancer patients with E75, an HLA-A2/A3 restricted HER2/neu (HER2) peptide mixed with GM-CSF. The vaccine has been studied in the adjuvant setting to prevent recurrences in clinically disease-free patients after completion of standard therapy. We have previously reported that the vaccine is safe, effectively stimulates HER2−specific immunity, and appears to improve disease-free survival at 24 months. Here, we report long-term data at a median follow-up of 60 months. Methods: The phase I/II trials were performed as dose escalation/schedule optimization trials enrolling node positive and high-risk, node-negative patients with tumors expressing any level of HER2. Vaccinated patients were given 4–6 monthly inoculations of E75 with GM-CSF immunoadjuvant. Due to waning immunity, a voluntary booster program was initiated, with inoculations every 6 months after completion of the primary vaccine series (PVS). Patients were monitored for local and systemic toxicities, which were graded by the NCI9s Common Terminology Criteria for Adverse Events. Vaccinated patients and controls were followed for 60 months and recurrences were documented. Demographic differences were compared with the Fisher9s exact test and survival was analyzed by the log-rank test. Results: 187 patients were enrolled; 108 in the vaccine group (VG) and 79 in the unvaccinated control group (CG). The vaccine and control groups were well-matched with the only statistically significant difference being ER-/PR- status (31.1% in VG vs 17.7% in CG, p=0.04). Vaccination was well tolerated with primarily grade 1 and grade 2 toxicity in the PVS (Local Toxicity: 85% Grade 1, 15% Grade 2, 0% Grade 3; systemic toxicity: 71% Grade 1, 14% Grade 2, and 3% Grade 3). Fifty-three of the VG patients received at least one booster, with 34 receiving a second booster, 25 a third, 22 a fourth, 12 a fifth, and 9 receiving at least six boosters. Booster inoculations were well-tolerated with only grade 1 and 2 local and systemic toxicities. There were delayed urticarial reactions in 7/53(13%) of the boosted patients occurring at a median of 9 days (5-21 days) after inoculation; these were grade 2 reactions and well-tolerated. After a median follow-up of 60 months, there has been a nonsignificant decrease in recurrences observed in the VG compared to the CG (10.6% vs 20.3%, p=0.098). The hazard ratio is 0.52 in the VG. In patients with immunity maintained with voluntary boosters, there have been even fewer (3.8%) recurrences (p=0.03). Conclusions: The E75 breast cancer vaccine is safe and well-tolerated. With long-term follow-up at 60 months, the E75 vaccine continues to show a strong trend toward preventing breast cancer recurrence in vaccinated patients particularly in patients whose immunity is maintained with booster inoculations. To investigate this further, a phase III trial with prospective boosting is being initiated. Citation Information: Cancer Res 2011;71(24 Suppl):Abstract nr P1-13-02.


Vaccine | 2018

Initial phase I/IIa trial results of an autologous tumor lysate, particle-loaded, dendritic cell (TLPLDC) vaccine in patients with solid tumors

Garth S. Herbert; Tj Vreeland; Guy T. Clifton; Julia M. Greene; Doreen O. Jackson; Mark O. Hardin; Diane F. Hale; John S. Berry; Pauline Nichol; Sook Yin; Xianzhong Yu; Thomas E. Wagner; George E. Peoples

INTRODUCTION Tumor vaccines use various strategies to generate immune responses, commonly targeting generic tumor-associated antigens. The tumor lysate, particle-loaded, dendritic cell (TLPLDC) vaccine is produced from DC loaded with autologous tumor antigens, creating a patient-specific vaccine. Here, we describe initial phase I/IIa trial results. METHODS This trial includes patients with any stage solid tumors, ECOG ≤1, and >4 months life-expectancy. A personalized vaccine is created using 1 mg of tumor and 120 ml blood (to isolate DC). Primary vaccination series (PVS) is four monthly inoculations. Patients are followed per standard of care (SOC). Endpoints include safety and tumor response (RECIST v1.1). RESULTS 44 patients were enrolled and vaccinated consisting of 31 late stage patients with residual/measurable disease, and 13 disease-free patients after SOC therapies. While 4 patients progressed before completing the PVS, 12/31 (39%) demonstrated clinical benefit (2 complete responses, 4 partial responses, 6 stable disease). In the adjuvant setting, 46% of late stage patients remain disease free at a median of 22.5 months. CONCLUSIONS The TLPLDC vaccine is scalable, generates a personalized DC vaccine, and requires little autologous tumor tissue and few DC. The vaccine is safe, with primarily grade 0-2 toxicities, and nearly 40% clinical benefit rate in varied tumors, warranting further study. TRIAL REGISTRATION ISRCTN81339386, Registered 2/17/2016.


Archive | 2018

Therapeutic Cancer Vaccines for Melanoma

Kaitlin M. Peace; Garth S. Herbert; Tj Vreeland; G. Travis Clifton; George E. Peoples

Melanoma has long been a prominent focus of investigation for cancer immunotherapy due to its intrinsically immunogenic nature, typically a consequence of a high mutational load. Furthermore, tumor infiltration by lymphocytes provides evidence of endogenous immune activity, as does the spontaneous regression described in some tumors. Conversely, the melanoma microenvironment promotes upregulation of immunosuppressive cells (myeloid-derived suppressor cells and regulatory T cells) and cytokines (such as IL-6, IL-10, TNFα, TGFβ, and VEGF) facilitating evasion of the immune response. The tumor microenvironment contains several targets for immunomodulation that are under active investigation. In this chapter, we will explore the development of immunotherapy for the treatment of melanoma, beginning with early, nonspecific immunomodulation through more recent advances in melanoma vaccines with specific discussion of several phase III clinical trials for peptide, viral, dendritic cell, and whole tumor cell-based vaccines.


Cancer immunology research | 2016

Abstract B007: Improved disease-free survival in endometrial and ovarian cancer patients with low folate binding protein expression after treatment with the E39 peptide vaccine in a Phase I/IIa trial

Kaitlin M. Peace; Diane F. Hale; Tj Vreeland; Doreen O. Jackson; Julia M. Greene; John S. Berry; Alfred F. Trappey; Garth S. Herbert; Guy T. Clifton; Mark O. Hardin; Kathleen M. Darcy; Chad A. Hamilton; G. Larry Maxwell; George E. Peoples

Introduction: One of the most encouraging examples of targeted therapy for cancer is trastuzumab, but its success is dependent on levels of expression of its target, HER2. We have found that HER2 expression levels also have a significant impact on the efficacy of HER2-directed peptide vaccines. Analogous to HER2 in breast cancer, Folate Binding Protein (FBP) is over-expressed on ovarian and endometrial cancer cells (up to 80 - 90-fold higher) and increased FBP expression is associated with aggressive disease. As a result, multiple FBP-directed therapies are being developed. We are investigating E39 + GM-CSF, which is an HLA-A2-restricted FBP-derived peptide vaccine used to prevent recurrence in disease-free endometrial and ovarian cancer patients (pts) after standard of care (SOC), multi-modality therapy. We have shown that E39 is safe, effectively generates E39-specific immune responses, and may improve DFS when optimally dosed in a phase I/IIa trial.1 Little is known about the effects of FBP expression levels on FBP-directed therapies, including our E39 vaccine. Purpose: Here, we report clinical outcomes of patients based on FBP expression levels from a phase I/IIa trial of the E39+GM-CSF vaccine given for the prevention of recurrence in disease-free endometrial and ovarian cancer patients. Methods: Disease-free, HLA-A2+ pts were vaccinated (VG), while HLA-A2- pts were followed as untreated controls (CG). The VG received 6 monthly inoculations of E39+GM-CSF, including either 100, 500, or 1000mcg of peptide and 250mcg of GM-CSF. FBP expression testing was performed by immunohistochemistry and the results were graded 0-4+ based on the percentage of positively staining cells. Patient9s tumors were then categorized as low expression (FBPlo) if scored 0-1+ or high expression (FBPhi) if 2-4+. The pts were monitored for evidence of clinical recurrence through the SOC follow-up by their treating oncology team. Demographics, FBP expression and disease-free survival (DFS) were analyzed using appropriate statistical tests. Results: Thirty-eight enrolled pts underwent FBP expression testing (CG n = 20; VG n = 18). There were no clinicopathologic differences between groups (p≥0.1). Nineteen pts were found to be FBPlo (CG, n = 11; VG, n = 8) and 19 were FBPhi (CG, n = 9; VG, n = 10). Median follow up was 16.3 months. There was no significant difference in overall DFS between the CG and the VG (34.6% vs. 34.6%, p = 0.208). In FBPlo pts, there was improved DFS in the VG vs. CG (85.7% vs. 17.5%, p = 0.01) while there was no such difference in FBPhi pts (VG:13.9% vs. CG:44.4%, p = 0.83). Though groups were small, there was a dose-dependent effect on DFS; pts receiving 1000mcg (n = 4) had improved DFS compared to the Conclusion: This phase I/IIa trial has previously demonstrated that E39 is well-tolerated, elicits a strong, dose-dependent in vivo immune response and may improve DFS when properly dosed. This focused analysis based on FBP expression level revealed a DFS benefit in FBPlo, but not FBPhi, endometrial and ovarian cancer pts treated with E39. This may be due to immunotolerance from significantly higher endogenous exposure to the FBP antigen. This is concordant with findings in our trials of HER2-directed peptide vaccines in breast cancer pts. These findings warrant further study as they may have important implications regarding the target population for future E39 peptide vaccine trials. 1. Jackson DO, et al. Interim analysis of a phase I/IIa trial assessing E39+GM-CSF, a folate binding protein vaccine, to prevent recurrence in ovarian and endometrial cancer patients. In Press. Citation Format: Kaitlin M. Peace, Diane F. Hale, Timothy J. Vreeland, Doreen O. Jackson, Julia M. Greene, John S. Berry, IV, Alfred F. Trappey, Garth S. Herbert, Guy T. Clifton, Mark O. Hardin, Kathleen M. Darcy, Chad A. Hamilton, G. Larry Maxwell, George E. Peoples. Improved disease-free survival in endometrial and ovarian cancer patients with low folate binding protein expression after treatment with the E39 peptide vaccine in a Phase I/IIa trial [abstract]. In: Proceedings of the Second CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; 2016 Sept 25-28; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(11 Suppl):Abstract nr B007.


Cancer Research | 2013

Abstract P4-13-05: HLA-A2 is not a prognostic indicator in breast cancer: Implications for cancer vaccine trials

Alfred F. Trappey; John S. Berry; Tj Vreeland; Ct Guy; Hf Diane; Sk Alan; Sj Erika; L Ferrise; Nathan M. Shumway; Michael Papamichail; Sonia A. Perez; Sathibalan Ponniah; Elizabeth A. Mittendorf; Ge Peoples

Introduction Peptide-based cancer vaccines require presentation by a specific HLA molecule. HLA-A2 is the most common class I allele in the US (40-50% of the population) and, therefore, the most commonly targeted. Our group has been investigating HER2-derived peptide vaccines administered in the adjuvant setting to high risk breast cancer patients in order to prevent disease recurrence. This study was undertaken to compare known prognostic factors and disease-free survival (DFS) in control HLA-A2+ and HLA-A2- patients in order to better define these populations for future trial design. Methods Our group is currently enrolling patients in a phase II trial evaluating the HER2-derived peptide vaccines, AE37 (MHC Class II, HLA-non-restricted epitope) and GP2 (MHC Class I, HLA-A2+ restricted epitope). The studies are enrolling high-risk, disease-free breast cancer patients with any level of HER2 expression (IHC 1+, 2+ or 3+) after completion of standard of care therapy. Patients are HLA-typed. HLA-A2+ patients are randomized to GP2+GM-CSF or GM-CSF alone. HLA-A2- patients are randomized to AE37+GM-CSF or GM-CSF alone. Demographics between groups are compared using chi squared or fisher exact as appropriate. DFS is compared using log rank. Results Thus far, 407 patients have been enrolled to the study (181 HLA-A2+ and 226 HLA-A2-). Demographics are shown in Table 1. There are no differences between groups with respect to age, node positivity, grade, tumor size, ER/PR status, HER2 over-expression, or triple negative breast cancer. Of those enrolled, 83 HLA-A2+ patients and 109 HLA-A2- patients have been randomized to the control groups. Within the control group, there are no differences between the HLA-A2+ and HLA-A2- patients regarding age, node positivity, grade, tumor size, ER/PR status, HER2 over-expression, or triple negative breast cancer (Table 2). With a median follow-up of 30 months, DFS is similar between A2+ and A2- control patients (83% v. 80%, p = 0.93). Conclusions Baseline clinico-pathologic factors are similar between HLA-A2+ and HLA-A2- breast cancer patients with no correlations to known prognostic factors. Well-matched blinded control patients treated only with GM-CSF demonstrate no differences in DFS between HLA-A2+ and HLA-A2- patients. Therefore, it does not appear that HLA-A2 status is a prognostic factor in breast cancer, and HLA-A2+ and HLA-A2- patients should be comparable in peptide-based breast cancer vaccine trials. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr P4-13-05.


Cancer Research | 2013

Abstract P2-14-01: Breast cancer patients with HER2 low-expression: An under-recognized group at significant risk for recurrence

Tj Vreeland; Bs John; Alfred F. Trappey; Erika J Schneble; Diane F. Hale; Gt Clifton; Nathan M. Shumway; Sonia A. Perez; Michael Papamichail; Sathibalan Ponniah; Ge Peoples; Elizabeth A. Mittendorf

Background: HER2 over-expression is associated with more aggressive malignant disease. The introduction of trastuzumab and other HER2-directed therapies, however, has led to improved prognosis for patients (pts) with HER2 over-expressing (OE) tumors. Currently, no HER2-targeted therapies are available for patients with HER2 low-expressing (LE) (1+, 2+ by IHC) tumors. We are conducting a randomized, controlled Phase II trial of multiple peptide vaccines enrolling patients with any level of HER2 expression (1+, 2+ and 3+). Here, we report survival data based on levels of HER2 expression in our unvaccinated, control pts. Methods: After standard of care therapy, disease-free, high-risk BCa pts were randomized to receive either peptide+GM-CSF (Vaccine Group, VG) or GM-CSF alone (Control Group, CG) in six, monthly doses followed by four boosters every six months. Pts were prospectively followed for recurrence. Demographic information was available for all pts and was compared between groups using chi square or fisher exact tests. Disease-Free Survival (DFS) was compared using log rank. Results: To date, we have enrolled 196 pts in the CG. 96 pts had HER2 OE tumors, 100 had LE tumors. The only significant demographic difference between the CG OE and LE groups was more ER/PR positive patients in LE (LE 72% vs OE 51%, p = 0.008). 83% of CG OE pts received trastuzumab, 3% of CG LE pts received trastuzumab. At a median f/u of 30 mo, DFS was significantly higher for CG OE vs CG LE (92.5% v 65.5%, p = 0.001). Conclusions: In the cohort of control pts from our ongoing vaccine trial, conducted in an era when Tz has been standard of care therapy for patients with HER2 OE tumors, we have shown that HER2 LE pts are at higher risk of recurrence than OE pts, despite having more ER/PR positive. This calls for increased efforts to develop novel therapies for patients with HER2 LE disease. We have previously shown a trend towards increased DFS with the HER2 vaccines, AE37 (p = 0.13, median f/u 22 mo) and E75 (p = 0.16, median f/u 60mo) in HER2 LE pts, suggesting that these vaccines may represent one such novel therapeutic approach. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr P2-14-01.


Cancer Research | 2013

Abstract P4-13-02: Preliminary results for the phase 1 trial of a dual HER2 peptide cancer vaccine in breast and ovarian cancer patients

John S. Berry; Alfred F. Trappey; Tj Vreeland; Erika J Schneble; Gt Clifton; Diane F. Hale; Alan K. Sears; Sathibalan Ponniah; Nathan M. Shumway; Elizabeth A. Mittendorf; Ge Peoples

BACKGROUND: HER2 is a commonly expressed tumor-associated antigen in breast (BrCa) and ovarian cancer (OvCa) and, therefore, an attractive target for immunotherapy. We have investigated HER2-derived peptides as vaccines mixed with GM-CSF to include GP2 (a HLA-A2 and HLA-A3 restricted, CD8+ eliciting epitope) and AE37 (a HLA unrestricted, MHC class II, CD4+ eliciting epitope). Both peptide vaccines (PV) have shown clinical promise individually. There is clear rationale for combining GP2 and AE37 to elicit a more robust immune response (IR) of both CD4+ and CD8+ T cells. Here, we summarize initial toxicity (tox) and in vivo IR data from a phase 1 trial of the combined PV. METHODS: The trial is being performed as a five cohort, 3+3 dose-escalation, safety trial. Clinically disease-free, HLA-A2+ and A3+, BrCa and OvCa patients with tumors expressing any level of HER2 (IHC 1-3+) and who have completed standard-of-care therapy are accrued. In the first cohort, three patients received six, monthly intradermal inoculations (R1-R6) of 100mcg of AE37, 100mcg of GP2, and 125mcg of GM-CSF or 100:100:125. The second cohort received 250mcg of AE37, 100mcg of GP2, and 125mcg of GM-CSF or 250:100:125. Three additional cohorts were vaccinated: 250:250:125, 500:250:125, and 500:500:125. Toxicity was graded 48-72 hours post vaccination using NCI Toxicity Criteria v4.0. After each inoculation, local reactions (LR) are measured via the sensitive ballpoint pen method and reported as the orthogonal mean (OM). IR is assessed in vivo by delayed type hypersensitivity (DTH) reactions with separate intradermal inoculations of AE37, AE36, and GP2 antigens, measured both pre-vaccination (R0) and after the vaccine series (R6) via the sensitive ballpoint pen method, and reported as the OM. Means were compared using paired t-tests. RESULTS: 28 patients enrolled; 8 withdrew consent, 1 recurred prior to completing R6, 3 had an intercurrent illness, 14 patients completed R1-R6, and the vaccine series is ongoing in 2 patients. Six patients did not receive any inoculations and, therefore, are not included in this safety analysis. In 22 patients, the vaccine was well tolerated (max local tox: 23% Grade (Gr) 1, 73% Gr 2, 4% Gr 3; max systemic tox: 14% Gr 0, 50% Gr 1, 36% Gr 2). No dose-limiting toxicity was observed. For the 14 patients who completed the VS, the median age was 51(35-83). Breast tumor size was 3.3±1.1cm and ovarian tumor size was 10.0±2.3cm. Compared to GP2 LR at R1 (15.5±4.1mm), LR increased at R2 (31.7±5.9mm), R3 (42.9±7.4mm), R4 (35.3±7.3mm), R5 (45.0±9.9mm), and R6 (25.9±6.7mm, p = 0.17). Compared to the AE37 LR at R1 (18.5±3.8mm), LR increased at R2 (37.3±6.7mm), R3 (36.4±4.6mm), R4 (42.2±5.9mm), R5 (46.0±8.9mm), and R6 (36.2±6.6mm). Unless stated, all LR p-values < 0.05. After the VS, AE37 DTH increased from 0.0±0.0mm to 19.6±6.7mm (p<0.01), AE36 DTH increased from 0.0±0.0mm to 10.3±3.9mm (p<0.01), and GP2 DTH reactions increased from 0.3±0.2mm to 4.1±2.0mm (p = 0.056). CONCLUSIONS: Initial results from a phase I trial of a vaccine combining GP2 and AE37 peptides show that dual administration of the peptides is well tolerated at all tested dosing levels. Additionally, the combination is capable of stimulating strong peptide-specific in vivo immune responses. Continued testing of this vaccination strategy is underway. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr P4-13-02.

Collaboration


Dive into the Tj Vreeland's collaboration.

Top Co-Authors

Avatar

Diane F. Hale

San Antonio Military Medical Center

View shared research outputs
Top Co-Authors

Avatar

Gt Clifton

University of Texas MD Anderson Cancer Center

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

Alfred F. Trappey

San Antonio Military Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ge Peoples

San Antonio Military Medical Center

View shared research outputs
Top Co-Authors

Avatar

George E. Peoples

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

John S. Berry

San Antonio Military Medical Center

View shared research outputs
Top Co-Authors

Avatar

Doreen O. Jackson

San Antonio Military Medical Center

View shared research outputs
Top Co-Authors

Avatar

Garth S. Herbert

San Antonio Military Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sathibalan Ponniah

Uniformed Services University of the Health Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge