15 results on '"Marcia Meseck"'
Search Results
2. Supplementary Figure Legend from Combined Vaccination with NY-ESO-1 Protein, Poly-ICLC, and Montanide Improves Humoral and Cellular Immune Responses in Patients with High-Risk Melanoma
- Author
-
Nina Bhardwaj, Rachel Lubong Sabado, Sacha Gnjatic, Sreekumar Balan, Gustavo Gimenez, Christopher B. McClain, Andres M. Salazar, John Mandeli, Rose Marie Holman, Thomas U. Marron, Patrick A. Ott, Michael Lattanzi, Marcia Meseck, Ana B. Blazquez, and Anna Pavlick
- Abstract
Descriptions of Supplementary Figures 1, 2, and 3, and supplementary table 1
- Published
- 2023
3. Supplementary Fig 2 from Combined Vaccination with NY-ESO-1 Protein, Poly-ICLC, and Montanide Improves Humoral and Cellular Immune Responses in Patients with High-Risk Melanoma
- Author
-
Nina Bhardwaj, Rachel Lubong Sabado, Sacha Gnjatic, Sreekumar Balan, Gustavo Gimenez, Christopher B. McClain, Andres M. Salazar, John Mandeli, Rose Marie Holman, Thomas U. Marron, Patrick A. Ott, Michael Lattanzi, Marcia Meseck, Ana B. Blazquez, and Anna Pavlick
- Abstract
Supplementary Fig 2
- Published
- 2023
4. Supplementary Fig 1 from Combined Vaccination with NY-ESO-1 Protein, Poly-ICLC, and Montanide Improves Humoral and Cellular Immune Responses in Patients with High-Risk Melanoma
- Author
-
Nina Bhardwaj, Rachel Lubong Sabado, Sacha Gnjatic, Sreekumar Balan, Gustavo Gimenez, Christopher B. McClain, Andres M. Salazar, John Mandeli, Rose Marie Holman, Thomas U. Marron, Patrick A. Ott, Michael Lattanzi, Marcia Meseck, Ana B. Blazquez, and Anna Pavlick
- Abstract
Supplementary Fig 1
- Published
- 2023
5. Data from Combined Vaccination with NY-ESO-1 Protein, Poly-ICLC, and Montanide Improves Humoral and Cellular Immune Responses in Patients with High-Risk Melanoma
- Author
-
Nina Bhardwaj, Rachel Lubong Sabado, Sacha Gnjatic, Sreekumar Balan, Gustavo Gimenez, Christopher B. McClain, Andres M. Salazar, John Mandeli, Rose Marie Holman, Thomas U. Marron, Patrick A. Ott, Michael Lattanzi, Marcia Meseck, Ana B. Blazquez, and Anna Pavlick
- Abstract
Given its ability to induce both humoral and cellular immune responses, NY-ESO-1 has been considered a suitable antigen for a cancer vaccine. Despite promising results from early-phase clinical studies in patients with melanoma, NY-ESO-1 vaccine immunotherapy has not been widely investigated in larger trials; consequently, many questions remain as to the optimal vaccine formulation, predictive biomarkers, and sequencing and timing of vaccines in melanoma treatment. We conducted an adjuvant phase I/II clinical trial in high-risk resected melanoma to optimize the delivery of poly-ICLC, a TLR-3/MDA-5 agonist, as a component of vaccine formulation. A phase I dose-escalation part was undertaken to identify the MTD of poly-ICLC administered in combination with NY-ESO-1 and montanide. This was followed by a randomized phase II part investigating the MTD of poly-ICLC with NY-ESO-1 with or without montanide. The vaccine regimens were generally well tolerated, with no treatment-related grade 3/4 adverse events. Both regimens induced integrated NY-ESO-1–specific CD4+ T-cell and humoral responses. CD8+ T-cell responses were mainly detected in patients receiving montanide. T-cell avidity toward NY-ESO-1 peptides was higher in patients vaccinated with montanide. In conclusion, NY-ESO-1 protein in combination with poly-ICLC is safe, well tolerated, and capable of inducing integrated antibody and CD4+ T-cell responses in most patients. Combination with montanide enhances antigen-specific T-cell avidity and CD8+ T-cell cross-priming in a fraction of patients, indicating that montanide contributes to the induction of specific CD8+ T-cell responses to NY-ESO-1.
- Published
- 2023
6. Supplementary Fig 3 from Combined Vaccination with NY-ESO-1 Protein, Poly-ICLC, and Montanide Improves Humoral and Cellular Immune Responses in Patients with High-Risk Melanoma
- Author
-
Nina Bhardwaj, Rachel Lubong Sabado, Sacha Gnjatic, Sreekumar Balan, Gustavo Gimenez, Christopher B. McClain, Andres M. Salazar, John Mandeli, Rose Marie Holman, Thomas U. Marron, Patrick A. Ott, Michael Lattanzi, Marcia Meseck, Ana B. Blazquez, and Anna Pavlick
- Abstract
Supplementary Fig 3
- Published
- 2023
7. Data from Myeloid-Derived Suppressor Cells as a Vehicle for Tumor-Specific Oncolytic Viral Therapy
- Author
-
Ping-Ying Pan, Shu-Hsia Chen, Savio Woo, Celia Divino, Stephen Ward, Marcia Meseck, Hui-ming Chen, Ge Ma, Karen Briley-Saebo, Brian A. Coakley, and Samuel Eisenstein
- Abstract
One of the several impediments to effective oncolytic virus therapy of cancer remains a lack of tumor-specific targeting. Myeloid-derived suppressor cells (MDSC) are immature myeloid cells induced by tumor factors in tumor-bearing hosts. The biodistribution kinetics of MDSC and other immune cell types in a murine hepatic colon cancer model was investigated through the use of tracking markers and MRI. MDSCs were superior to other immune cell types in preferential migration to tumors in comparison with other tissues. On the basis of this observation, we engineered a strain of vesicular stomatitis virus (VSV), an oncolytic rhabdovirus that bound MDSCs and used them as a delivery vehicle. Improving VSV-binding efficiency to MDSCs extended the long-term survival of mice bearing metastatic colon tumors compared with systemic administration of wild-type VSV alone. Survival was further extended by multiple injections of the engineered virus without significant toxicity. Notably, direct tumor killing was accentuated by promoting MDSC differentiation towards the classically activated M1-like phenotype. Our results offer a preclinical proof-of-concept for using MDSCs to facilitate and enhance the tumor-killing activity of tumor-targeted oncolytic therapeutics. Cancer Res; 73(16); 5003–15. ©2013 AACR.
- Published
- 2023
8. Supplementary Figure Legend from Myeloid-Derived Suppressor Cells as a Vehicle for Tumor-Specific Oncolytic Viral Therapy
- Author
-
Ping-Ying Pan, Shu-Hsia Chen, Savio Woo, Celia Divino, Stephen Ward, Marcia Meseck, Hui-ming Chen, Ge Ma, Karen Briley-Saebo, Brian A. Coakley, and Samuel Eisenstein
- Abstract
PDF file - 26K
- Published
- 2023
9. Supplementary Figures 1 - 7 from Myeloid-Derived Suppressor Cells as a Vehicle for Tumor-Specific Oncolytic Viral Therapy
- Author
-
Ping-Ying Pan, Shu-Hsia Chen, Savio Woo, Celia Divino, Stephen Ward, Marcia Meseck, Hui-ming Chen, Ge Ma, Karen Briley-Saebo, Brian A. Coakley, and Samuel Eisenstein
- Abstract
PDF file - 653K, MDSC purity (S1); Effective Feridex labeling of MDSCs (S2); VSV does not alter the ability of MDSCs to migrate to tumor sites (S3); Antibody conjugation improves viral delivery to tumor sites (S4); VSV-MDSCs exhibit more tumor specificity than free virus (S5); VSV-MDSC treatment does not result in any appreciable neuropathological toxicity (S6); The survival curves of LLC lung cancer-bearing mice after various treatments (S7).
- Published
- 2023
10. Abstract CT270: Immunogenicity of PGV_001 neoantigen vaccine in a Phase-I clinical trial, across various types of cancers in adjuvant setting
- Author
-
Mansi Saxena, Thomas Marron, Julia Kodysh, Alex Rubinsteyn, John Finnigan, Ana Blasquez, Marcia Meseck, Tim O'Donnell, Daniela Delbeau, Mathew Galsky, Deborah Doroshow, Brett Miles, Krzysztof Misiukiewicz, Hanna Irie, Amy Tiersten, Samir Parekh, Marshall Posner, Andrea Wolf, John Mandeli, Rachel Brody, Sacha Gnjatic, Eric Schadt, Philip Friedlander, and Nina Bhardwaj
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: Immunotherapies such as checkpoint blockade, have demonstrated remarkable clinical efficacy yet a large percentage of patients do not respond, potentially due to a paucity of pre-existing immune priming against neoantigens. We developed a personalized genome vaccine (PGV_001) platform to generate neoantigen vaccines targeting each patient’s unique mutanome. Primary objectives of the study were to determine 1) the safety and tolerability; 2) the feasibility of PGV_001 production and administration; and 3) the immunogenicity of PGV_001. Secondary objectives included immunophenotyping vaccine driven cellular and soluble immune milieu in peripheral blood. We previously reported on the clinical efficacy, and here we report, analysis of vaccine-driven immune responses in all treated patients. Methods: The study (Trial Registration NCT02721043) enrolled patients with resected malignancies, including Head and neck squamous cell carcinomas, breast cancer and bladder cancer, or, in the case of multiple myeloma treated with autologous stem cell transplant; all patients determined to have a high risk of disease recurrence (>30% over 5 years). Tumor-derived and germline RNA and DNA was sequenced to predict neoantigens utilizing our custom computation pipeline, OpenVax. Approximately 10 peptides were synthesized per patient, and a mixture of these peptides was administered as 10 subcutaneous and intradermal vaccines over 27 weeks in combination with poly-ICLC and helper Tetanus peptide as adjuvants. Immune responses were analyzed utilizing assays including IFN-gamma ELISPOT, antigen specific T cell expansion followed by flow cytometry, etc. Results: In total 148 neoantigen peptides were manufactured for 15 patients. Overall, 136 PGV_001 doses were administered to 13 patients. Vaccine-specific T cell immunity was observed against multiple vaccine neoepitopes in all 13 subjects. Of the peptides administered, 45% of vaccine antigens (57/126) induced de novo immunity, starting as early as Week8 and often sustaining past last vaccination. Notably, while the vaccine driven T cell immunity was CD4 T cell dominant, most evaluated subjects also displayed vaccine induced polyfunctional CD8-T cell responses. Additional studies are ongoing to define qualities of reactive T cells, evaluate vaccine-induced humoral responses and probe the circulating inflammatory immune milieu. These will be presented. Conclusions: We have established a platform for generating personalized neoantigen vaccines. 100% of the vaccinated patients developed an immune response specific to the vaccine neoantigens predicted by OpenVax. Subjects who received treatment experienced mild Grade 1 or 2 adverse reactions as per the CTEP v 4.0 NCI CTCAE. This vaccine trial reached the primary endpoint of safety, tolerability, feasibility and immunogenicity. Based on the PGV_001 platform two clinical trials, one in patients with glioblastoma multiforme (NCT03223103) in combination with TT fields and second in patients with urothelial carcinoma (NCT03359239) in combination with Atezolizumab have been performed. Data from these trials is under evaluation. Citation Format: Mansi Saxena, Thomas Marron, Julia Kodysh, Alex Rubinsteyn, John Finnigan, Ana Blasquez, Marcia Meseck, Tim O'Donnell, Daniela Delbeau, Mathew Galsky, Deborah Doroshow, Brett Miles, Krzysztof Misiukiewicz, Hanna Irie, Amy Tiersten, Samir Parekh, Marshall Posner, Andrea Wolf, John Mandeli, Rachel Brody, Sacha Gnjatic, Eric Schadt, Philip Friedlander, Nina Bhardwaj. Immunogenicity of PGV_001 neoantigen vaccine in a Phase-I clinical trial, across various types of cancers in adjuvant setting [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr CT270.
- Published
- 2023
11. Abstract CT108: Immunogenicity of Poly-ICLC matured dendritic cells as an adjuvant for NY-ESO-1 and Melan-A-MART-1 peptide vaccination compared to Montanide® ISA-51 VG, in study subjects with melanoma in complete clinical remission but at high risk of disease recurrence
- Author
-
Mansi Saxena, Ashleih Burke, Anna Pavlick, Ana Blazquez, Gustavo Gimenez, Marcia Meseck, Michael Donovan, Denise Rodriguez, Mireia Castillo-Martin, Tin Htwe Thin, Rachel Sabado, John Mandeli, Sacha Gnjatic, Philip Friedlander, and Nina Bhardwaj
- Subjects
Cancer Research ,Oncology - Abstract
Background: Dendritic cells (DCs) play a critical role in anti-tumor immune response. However, tumor-induced immunosuppression promotes DC dysfunction and T cell exhaustion resulting in evasion of tumor immunity. In this study, we aimed to compare two approaches for engaging DCs and inducing an immune response to tumor antigens in the absence of tumor (melanoma) Methods: This is a Phase II open-label, randomized, two-arm study to compare Arm A: Poly-ICLC-matured DC+PolyICLC to Arm B: Montanide ISA-51-VG+Poly-ICLC, as adjuvants for NY-ESO-1 and Melan-A/MART-1 long peptides in patients with melanoma in complete clinical remission but at high risk of disease recurrence (NCT02334735). Each arm also received helper peptide, keyhole limpet hemocyanin (KLH) with the first vaccine and Poly-ICLC on day 2 of each vaccine. 36 patients were consented and randomized. Of these, 31 subjects received treatment, 16 in Arm A and 15 in Arm B. Immunohistochemistry (IHC) was used to determine expression of NY-ESO-1 and Melan-A/MART-1 and to determine the immune infiltrate landscape in the primary tumors. Humoral responses, TCR clonality, and inflammatory pathways were assessed by ELISA, bulk TCR sequencing, and Olink, respectively. Functional T-cell responses were investigated ex-vivo by interferon (IFN)-g enzyme-linked immunospot assay (ELISPOT) and after expansion by intracellular cytokine staining. Results: Arm B induced a stronger humoral response vs Arm A against both Melan-A/MART-1 and NY-ESO-1. A stronger ex vivo IFN-g response as well as expanded CD4+ T cell response against NY-ESO-1 was also induced in Arm B. However, the response to Melan-A/MART-1, as measured by ex vivo ELISPOT and expanded CD4+T cell assay, was comparable in both arms. Interestingly, while similar proportions of patients in each arm displayed a CD8+ T cell response to NYESO-1, more patients in Arm A vs Arm B responded with a CD8+ T cell response to Melan-A/MART-1 (9/16 responders in Arm A vs 4/14 in Arm B). Melan-A expression was observed in 81% of the patients but did not correlate with antigen specific immune response. TCR sequencing, Olink analysis and evaluation of NY-ESO-1 expression and immune infiltrates in the primary tumors is ongoing. Conclusion: This trial reached the primary endpoint of safety and tolerability. Arm B induced a stronger antibody and CD4+ T cell response, especially to NY-ESO-1, whereas the Arm A vaccine appears to be more efficient at eliciting CD8+ T cell responses against MelanA/MART1. The seemingly enhanced CD8 T cell response in Arm A versus Arm B, could be attributed to frequency of specific HLA-I alleles in either arm or pre-existing immune response in select patients. A deeper investigation into correlation between HLA type and cellular immune response to vaccine is ongoing. Citation Format: Mansi Saxena, Ashleih Burke, Anna Pavlick, Ana Blazquez, Gustavo Gimenez, Marcia Meseck, Michael Donovan, Denise Rodriguez, Mireia Castillo-Martin, Tin Htwe Thin, Rachel Sabado, John Mandeli, Sacha Gnjatic, Philip Friedlander, Nina Bhardwaj. Immunogenicity of Poly-ICLC matured dendritic cells as an adjuvant for NY-ESO-1 and Melan-A-MART-1 peptide vaccination compared to Montanide® ISA-51 VG, in study subjects with melanoma in complete clinical remission but at high risk of disease recurrence [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr CT108.
- Published
- 2022
12. Abstract LB048: An adjuvant personalized neoantigen peptide vaccine for the treatment of malignancies (PGV-001)
- Author
-
Deborah B. Doroshow, Tim O'Donnel, Krysztof Misiukiewicz, Julia Kodysh, Mathew Galsky, Jeff Hammerbacher, Ana Belen Blazquez, John P. Finnigan, Thomas U. Marron, Sacha Gnjatic, Rachel Brody, Marshall R. Posner, Philip Friedlander, Brett A. Miles, Hanna Irie, Nina Bhardwaj, Alex Rubinsteyn, Eric E. Schadt, Marcia Meseck, Mansi Saxena, Andrea S. Wolf, John Mandeli, Samir Parekh, and Amy Tiersten
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Immunogenicity ,Cancer ,medicine.disease ,Neoantigen Peptide ,Vaccination ,Tolerability ,Internal medicine ,medicine ,Adverse effect ,business ,Adjuvant ,Multiple myeloma - Abstract
Background: The majority of novel cancer immunotherapies rely on adequate priming of T cells to tumor-specific neoantigens, which is believed to be lacking in patients who do not respond to therapy. We developed a personalized genomic vaccine (PGV-001) in which patient-specific synthetic neoantigen peptides (25 mer),are formulated and administered to patients with multiple cancer types in the adjuvant setting (NCT02721043). Methods: This trial enrolled patients whom had undergone curative-intent surgery (solid tumor patients) or autologous stem cell transplant (multiple myeloma patients), and for whom there was >30% chance of recurrence. Sequencing of tumor and germline DNA and RNA was performed and the OpenVax custom computation pipeline was used to identify candidate neoantigens; this platform ranks transcribed mutations using predicted MHC-I binding affinity and neoantigen abundance. A maximum of 10 peptides were synthesized per patient. Peptides were administered over the course of 27 weeks with poly-ICLC and a tetanus helper peptide. Primary objectives were to determine the safety and tolerability of vaccination, feasibility of vaccine production and administration, and immunogenicity. Results: Within 15 patients enrolled, the OpenVax pipeline identified an average of 67.1 neoantigens/patient (range 8-193), only two patients did not have adequate number of neoantigens identified to synthesize 10 peptides. 13 of the 15 patients received PGV-001, including 10 patients with solid tumor diagnoses and 3 patients with multiple myeloma, 11 of whom received all 10 doses, while 1 experienced progression of disease while on treatment. The vaccine was well tolerated, with grade 1 injection site reactions in 31% of patients, and grade 1 fever in one patient; there were no other significant adverse events. While one patient was lost to follow-up, of the remaining 12 patients the median progression-free survival from the time of their surgery or transplant of 618 days. With a mean follow-up of 925 days, 4 patients remain without evidence of disease, 4 patients are receiving subsequent lines of therapy, and 4 patients have died, though notably only two with documented recurrence of their malignancy. Initial analysis of the patient samples analyzed confirms immunogenicity. T cell responses were measured using ex vivo ELISpot and intracellular cytokine staining following expansion with neoantigen peptide libraries, both demonstrating induction of IFN-gamma, TNF-alpha and IL-2. Notably, robust T cell reactivity was only seen at the completion of all 10 vaccines, supporting the need for a prolonged schedule. Conclusions: PGV-001 was successfully synthesized for 15 patients and administered successfully to 13 patients without significant adverse events. Immune monitoring of immunogenicity is ongoing, with initial analysis demonstrating induction of neoantigen-specific CD4 and CD8 T cell expansion. Citation Format: Thomas Urban Marron, Mansi Saxena, Nina Bhardwaj, Marcia Meseck, Alex Rubinsteyn, John Finnigan, Julia Kodysh, Ana Blazquez, Tim O'Donnel, Mathew Galsky, Deborah Doroshow, Brett Miles, Krysztof Misiukiewicz, Hanna Irie, Amy Tiersten, Samir Parekh, Marshall Posner, Andrea Wolf, John Mandeli, Rachel Brody, Sacha Gnjatic, Eric Schadt, Philip Friedlander, Jeffrey Hammerbacher. An adjuvant personalized neoantigen peptide vaccine for the treatment of malignancies (PGV-001) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr LB048.
- Published
- 2021
13. Abstract CT173: PGV-001: A phase I trial of a multipeptide personalized neoantigen vaccine in the adjuvant setting
- Author
-
Julia Kodysh, John P. Finnigan, Timothy O'Donnell, Thomas U. Marron, Marcia Meseck, Alex Rubinsteyn, Philip Friedlander, Ana Belen Blazquez, Nina Bhardwaj, and Mansi Saxena
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,biology ,business.industry ,medicine.medical_treatment ,Immunogenicity ,Cancer ,Human leukocyte antigen ,medicine.disease ,Vaccination ,Antigen ,Internal medicine ,MHC class I ,biology.protein ,Medicine ,business ,Adjuvant ,CD8 - Abstract
Background: Mutation-derived tumor antigens (MTAs) arise as a result of somatic mutations, such as nucleotide substitutions and small insertions/deletions. MTAs can serve as specific targets for antitumor therapy, including neoantigen vaccines. The goal of neoantigen vaccination is to help prime T cells to recognize such tumor-specific mutations. Here we describe a phase I trial testing a personalized genomic vaccine (PGV-001) in multiple histologies in the adjuvant setting (NCT02721043). Methods: This trial included patients with histologic diagnosis of solid malignancies or multiple myeloma with a >30% risk of recurrence but no measurable disease at the time of first vaccination. For each patient, HLA typing was performed, and the patient's tumor and germline DNA and tumor RNA were sequenced. Mutated peptides containing predicted neoantigens were selected using the OpenVax computational pipeline, which prioritizes somatic mutations by expression of the mutant allele in the tumor RNA and predicted MHC class I epitope binding for the patient's HLA type. A maximum of 10 peptides was included in each patient's personalized vaccine. The vaccine was administered over the course of 6 months, given in combination with poly-ICLC as the adjuvant. The primary objectives were to determine safety and tolerability, feasibility, and immunogenicity of the PGV-001 neoantigen vaccine. Results: The neoantigen vaccine was successfully administered to 13 patients, spanning 5 different tumor types. For each patient, an average of 1730 somatic mutations were identified (range 521-5106), of which 349 were coding variants (range 68-1493), 88 were coding and expressed in the tumor RNA (range 9-233), and 71 were coding, expressed and resulted in predicted MHC class I ligands (range 8-193). The urothelial, head & neck, and lung tumors resulted in larger numbers of predicted neoantigens than in the cases of multiple myeloma or breast tumors. Despite this difference, each of the 13 patient tumor samples showed enough of a neoantigen load to enable vaccination. We have also initiated analysis of immunogenicity, and early reports demonstrate neoantigen-specific CD4 and CD8 T-cell responses. Conclusions: The PGV-001 personalized neoantigen vaccine was successfully administered to 13 patients, where each patient's neoantigen load was adequate for vaccine synthesis. While we have demonstrated the computational feasibility of identification of neoantigens for inclusion in the PGV-001 genomic vaccine, clinical outcomes will be reported separately. Citation Format: Julia Kodysh, Thomas Marron, Alex Rubinsteyn, Tim O'Donnell, John Finnigan, Ana Blazquez, Mansi Saxena, Marcia Meseck, Philip Friedlander, Nina Bhardwaj. PGV-001: A phase I trial of a multipeptide personalized neoantigen vaccine in the adjuvant setting [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr CT173.
- Published
- 2020
14. Abstract A005: A phase I study of the safety and immunogenicity of a multipeptide personalized genomic vaccine in the adjuvant treatment of solid cancers
- Author
-
Eric M. Genden, Sacha Gnjatic, Brett A. Milles, Marcia Meseck, Julia Kodysh, Samir Parekh, Nina Bhardwaj, John Mandeli, Elisa Port, Krysztof Misiukiewicz, Amy Tiersten, Ashutosh K. Tewari, Hern J. Cho, Hooman Khorasani, Timothy O'Donnell, Jeff Hammerbacher, Milind Mahajan, Matthew D. Galsky, Hanna Irie, Ana Belen Blazquez, Eric E. Schadt, Andrea S. Wolf, John Holt, Thomas U. Marron, Sujit S. Nair, Michael J. Donovan, Rachel Lubong Sabado, William Oh, John P. Finnigan, Alex Rubinsteyn, Peter Dottino, and Phillip A. Friedlander
- Subjects
Cancer Research ,medicine.medical_treatment ,ELISPOT ,Immunogenicity ,Immunology ,Biology ,Virology ,Epitope ,Vaccination ,Cancer immunotherapy ,Antigen ,medicine ,Adjuvant ,Ex vivo - Abstract
Introduction: Mutation-derived tumor antigens (MTAs) arise as a direct result of somatic variations, including nucleotide substitutions, insertions, and deletions that occur during carcinogenesis. These somatic variations can be characterized via genetic sequencing and used to identify MTAs. We developed a platform for a fully-personalized MTA-based vaccine in the adjuvant treatment of solid and hematologic malignanicies. Methods: This is a single-arm, open label, proof-of-concept phase I study designed to test the safety and immunogenicity of Personalized Genomic Vaccine 001 (PGV001) that targets up to 10 predicted personal tumor neoantigens. The single-center study will enroll 20 eligible subjects with histologic diagnosis of solid and hematologic malignancies. Subjects must have no measurable disease at time of first vaccine administration, and 5-year disease recurrence risk of > 30%. Toxicity will be defined by CTCAE v5.0. Blood samples will be collected at various time points for immune response monitoring. Each patient’s vaccine peptides are selected by identifying somatic mutations from comparison of tumor and normal exome sequencing data, phasing somatic variants with co-occurring germline variants using tumor RNA sequencing data, and ranking mutated peptide sequences ”Openvax pipeline.” The process for determining somatic variants hews closely to the Broad Institute’s “Best Practices” for cancer SNVs and indels. The phasing of somatic and germline variants is implemented in a custom bioinformatics tool called Isovar. Mutated protein sequences containing phased variants are ranked according to two criteria: expression of the mutant allele in tumor RNA and aggregated predicted affinity to the patient’s Class I MHCs. Both quantities are normalized and multiplied together to create single ranked ordering of the candidate mutant sequences. Results: PGV001_002 (head and neck squamous cell cancer), who has completed vaccination, received 10 doses of vaccine comprising 10 long peptides (25 amino acid length) combined with poly-ICLC (toll-like receptor-3 agonist) intradermally. Vaccine-induced blood T-cell responses were determined, at weeks 0 (before-treatment) and 27 (after-treatment), ex vivo by interferon (IFN)-g enzyme-linked immunospot (ELISPOT) assay and after in vitro expansion by intracellular cytokine staining (ICS). Overlapping 15-16-mer assays peptides (OLPs) spanning the entirety of each ILP and 9-10-mer peptides corresponding to each predicted class I epitope (Min) were pooled and used to monitor immunogenicity. Ex vivo responses to these peptide pools were undetectable at week 0 but were evident at week 27 against 2 OLPs out of 10 (20%) and in 5 Min out of 10 (50%). After in vitro expansion, neoantigen-specific CD4+ and CD8+ T-cell responses were found in 5 out of 10 pooled peptides (50%). 7 out of 10 (70%) epitopes elicited polyfunctional T-cell responses (secretion of INF-α, TNF-α, and/or IL-2) from either CD4+ or CD8+ T-cells. Conclusion: To identify which predicted epitopes within the peptides pools stimulated the T-cell responses, we deconvoluted all the pools by either ex vivo and in vitro expansion. Ex vivo IFN-α production was detected in 1 (15-mer) peptide out of 15 (6.7%) and in 4 (9-10-mer) peptides out of 22 (18.2%). After expansion with single peptides, of 22 (9-10-mer) peptides tested, CD8+ T-cells were reactive against 13 peptides (59%), while CD4+ responses were seen in response to 11 of 15 (15-16-mer) peptides tested. Both CD4+ and CD8+ T-cell responses were polyfunctional. The PGV001 vaccine in our first patient showed both safety and immunogenicity, eliciting both CD4+ and CD8+ responses to the vaccine peptides. As we are enrolling additional patients, the information learned from this clinical trial will instruct the next generation of MTA-based vaccines, future development of immunotherapeutic approaches and rational combinations. Citation Format: Ana B. Blazquez, Alex Rubinsteyn, Julia Kodysh, John P. Finnigan, Thomas Marron, Rachel L. Sabado, Marcia Meseck, Timothy J. O'Donnell, Jeffrey Hammerbacher, Michael Donovan, John Holt, Milind Mahajan, John Mandeli, Krysztof Misiukiewicz, Eric M. Genden, Brett A. Milles, Hooman Khorasani, Peter R. Dottino, Hanna Irie, Amy B. Tiersten, Elisa R. Port, Andrea S. Wolf, Hern J. Cho, Ashutosh Tewari, Samir S. Parekh, Sujit Nair, Matthew D. Galsky, William K. Oh, Sacha Gnjatic, Eric E. Schadt, Phillip A. Friedlander, Nina Bhardwaj. A phase I study of the safety and immunogenicity of a multipeptide personalized genomic vaccine in the adjuvant treatment of solid cancers [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A005.
- Published
- 2019
15. The Novel Role of Tyrosine Kinase Inhibitor in the Reversal of Immune Suppression and Modulation of Tumor Microenvironment for Immune-Based Cancer Therapies
- Author
-
Ping-Ying Pan, Celia M. Divino, Ge Ma, Junko Ozao-Choy, Shu Hsia Chen, Max Sung, Johnny Kao, Marcia Meseck, George X. Wang, and Myron Schwartz
- Subjects
Cancer Research ,Indoles ,medicine.drug_class ,Molecular Sequence Data ,Mice, Transgenic ,chemical and pharmacologic phenomena ,Biology ,Lymphocyte Activation ,T-Lymphocytes, Regulatory ,Article ,Tyrosine-kinase inhibitor ,Carcinoma, Lewis Lung ,Mice ,Immune system ,Sunitinib ,medicine ,Animals ,Cytotoxic T cell ,Myeloid Cells ,Pyrroles ,Amino Acid Sequence ,Protein Kinase Inhibitors ,Mice, Inbred BALB C ,Tumor microenvironment ,FOXP3 ,Interleukin ,Sunitinib malate ,Mice, Inbred C57BL ,Oncology ,Colonic Neoplasms ,Immunology ,Cancer research ,medicine.drug - Abstract
In tumor-bearing hosts, myeloid-derived suppressor cells (MDSC) and T regulatory cells (Treg) play important roles in immune suppression, the reversal of which is vitally important for the success of immune therapy. We have shown that ckit ligand is required for MDSC accumulation and Treg development. We hypothesized that sunitinib malate, a receptor tyrosine kinase inhibitor, could reverse MDSC-mediated immune suppression and modulate the tumor microenvironment, thereby improving the efficacy of immune-based therapies. Treatment with sunitinib decreased the number of MDSC and Treg in advanced tumor-bearing animals. Furthermore, it not only reduced the suppressive function of MDSCs but also prevented tumor-specific T-cell anergy and Treg development. Interestingly, sunitinib treatment resulted in reduced expression of interleukin (IL)-10, transforming growth factor-β, and Foxp3 but enhanced expression of Th1 cytokine IFN-γ and increased CTL responses in isolated tumor-infiltrating leukocytes. A significantly higher percentage and infiltration of CD8 and CD4 cells was detected in tumors of sunitinib-treated mice when compared with control-treated mice. More importantly, the expression of negative costimulatory molecules CTLA4 and PD-1 in both CD4 and CD8 T cells, and PDL-1 expression on MDSC and plasmacytoid dendritic cells, was also significantly decreased by sunitinib treatment. Finally, sunitinib in combination with our immune therapy protocol (IL-12 and 4-1BB activation) significantly improves the long-term survival rate of large tumor-bearing mice. These data suggest that sunitinib can be used to reverse immune suppression and as a potentially useful adjunct for enhancing the efficacy of immune-based cancer therapy for advanced malignancies. [Cancer Res 2009;69(6):2514–22]
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.