1
|
Disis ML, Dang Y, Coveler AL, Childs JS, Higgins DM, Liu Y, Zhou J, Mackay S, Salazar LG. A Phase I/II Trial of HER2 Vaccine-Primed Autologous T-Cell Infusions in Patients with Treatment Refractory HER2-Overexpressing Breast Cancer. Clin Cancer Res 2023; 29:3362-3371. [PMID: 37093223 PMCID: PMC10754340 DOI: 10.1158/1078-0432.ccr-22-3578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/17/2023] [Accepted: 04/19/2023] [Indexed: 04/25/2023]
Abstract
PURPOSE High levels of type I T cells are needed for tumor eradication. We evaluated whether the HER2-specific vaccine-primed T cells are readily expanded ex vivo to achieve levels needed for therapeutic infusion. PATIENTS AND METHODS Phase I/II nonrandomized trial of escalating doses of ex vivo-expanded HER2-specific T cells after in vivo priming with a multiple peptide-based HER2 intracellular domain (ICD) vaccine. Vaccines were given weekly for a total of three immunizations. Two weeks after the third vaccine, patients underwent leukapheresis for T-cell expansion, then received three escalating cell doses over 7- to 10-day intervals. Booster vaccines were administered after the T-cell infusions. The primary objective was safety. The secondary objectives included extent and persistence of HER2-specific T cells, development of epitope spreading, and clinical response. Patients received a CT scan prior to enrollment and 1 month after the last T-cell infusion. RESULTS Nineteen patients received T-cell infusions. Treatment was well tolerated. One month after the last T-cell infusion, 82% of patients had significantly augmented T cells to at least one of the immunizing epitopes and 81% of patients demonstrated enhanced intramolecular epitope spreading compared with baseline (P < 0.05). There were no complete responses, one partial response (6%), and eight patients with stable disease (47%), for a disease control rate of 53%. The median survival for those with progressive disease was 20.5 months and for responders (PR+SD) was 45.0 months. CONCLUSIONS Adoptive transfer of HER2 vaccine-primed T cells was feasible, was associated with minimal toxicity, and resulted in an increased overall survival in responding patients. See related commentary by Crosby et al., p. 3256.
Collapse
Affiliation(s)
- Mary L. Disis
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, WA
| | - Yushe Dang
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, WA
| | - Andrew L. Coveler
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, WA
| | - Jennifer S Childs
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, WA
| | - Doreen M Higgins
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, WA
| | - Ying Liu
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, WA
| | | | | | - Lupe G. Salazar
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, WA
| |
Collapse
|
2
|
An Updated Review on Recent Advances in the Usage of Novel Therapeutic Peptides for Breast Cancer Treatment. Int J Pept Res Ther 2023. [DOI: 10.1007/s10989-023-10503-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
3
|
Gupta R, Jit BP, Kumar S, Mittan S, Tanwer P, Ray MD, Mathur S, Perumal V, Kumar L, Rath GK, Sharma A. Leveraging epigenetics to enhance the efficacy of cancer-testis antigen: a potential candidate for immunotherapy. Epigenomics 2022; 14:865-886. [DOI: 10.2217/epi-2021-0479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological malignancy in women. The phenotype is characterized by delayed diagnosis, recurrence and drug resistance. Inherent immunogenicity potential, oncogenic function and expression of cancer-testis/germline antigen (CTA) in ovarian cancer render them a potential candidate for immunotherapy. Revolutionary clinical findings indicate that tumor antigen-mediated T-cell and dendritic cell-based immunotherapeutic approaches provide an excellent strategy for targeting tumors. Currently, dendritic cell vaccination for the treatment of B-cell lymphoma and CTA-based T-cell receptor transduced T-cell therapy involving MAGE-A4 and NY-ESO-1 are well documented and shown to be effective. This review highlighted the mechanical aspects of epigenetic drugs that can elicit a CTA-based humoral and cellular immune response and implicate T-cell and dendritic cell-based immunotherapeutic approaches.
Collapse
Affiliation(s)
- Rashmi Gupta
- Department of Biochemistry, National Cancer Institute – India, Jhajjar Campus, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Bimal Prasad Jit
- Department of Biochemistry, National Cancer Institute – India, Jhajjar Campus, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Santosh Kumar
- Department of Biochemistry, National Cancer Institute – India, Jhajjar Campus, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Sandeep Mittan
- Montefiore Medical Center, Albert Einstein College of Medicine, NY 10467, USA
| | - Pranay Tanwer
- Laboratory Oncology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - M D Ray
- Department of Surgical Oncology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sandeep Mathur
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Vanamail Perumal
- Department of Obstetrics & Gynecology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Lalit Kumar
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - G K Rath
- Department of Radiotherapy, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Ashok Sharma
- Department of Biochemistry, National Cancer Institute – India, Jhajjar Campus, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| |
Collapse
|
4
|
Inflammation and immunity in ovarian cancer. EJC Suppl 2020; 15:56-66. [PMID: 33240443 PMCID: PMC7569134 DOI: 10.1016/j.ejcsup.2019.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/15/2019] [Accepted: 12/27/2019] [Indexed: 12/30/2022] Open
Abstract
The standard first-line therapy for ovarian cancer is a combination of surgery and carboplatin/paclitaxel-based chemotherapy. Patients with longer survival and improved response to chemotherapy usually present T-cell inflamed tumours. The presence of tumour-infiltrating T cells (TILs) notably varies among the different subtypes of ovarian tumours, being highest in high-grade serous ovarian carcinoma, intermediate in endometrioid tumours, and lowest in low-grade serous, mucinous and clear cell tumours. Interestingly, the presence of TILs is often accompanied by a strong immunosuppressive tumour environment. A better understanding of the immune response against ovarian cancer and the tumour immune evasion mechanisms will enable improved prognostication, response prediction and immunotherapy of this disease. This article provides an overview of some ovarian cancer cell features relevant for antitumour response, such as tumour-associated antigens, including neoantigens, expression of inhibitory molecules, and other mechanisms of immune evasion. Moreover, we describe relevant immune cell types found in epithelial ovarian tumours, including T and B lymphocytes, regulatory T cells, natural killer cells, tumour-associated macrophages, myeloid-derived suppressor cells and neutrophils. We focus on how these components influence the burden of the tumour and the clinical outcome. The presence of spontaneous tumour-specific T lymphocytes and the existence of multiple immune evasion mechanisms in epithelial ovarian cancer (EOC) support the immunogenicity of this tumour. Tumour-infiltrating T lymphocytes (TILs) have been associated with disease outcome in EOC, indicating their clinical significance. The subtypes of EOC, mutations in TP53 and breast and ovarian cancer susceptibility protein 1/2 and the immune expression signature are factors associated to TIL density in EOC. The tumour microenvironment in EOC consists of a dynamic and complex network of soluble factors, inhibitory receptors and immunosuppressive cells.
Collapse
|
5
|
Knutson KL, Block MS, Norton N, Erskine CL, Hobday TJ, Dietz AB, Padley D, Gustafson MP, Puglisi-Knutson D, Mangskau TK, Chumsri S, Dueck AC, Karyampudi L, Wilson G, Degnim AC. Rapid Generation of Sustainable HER2-specific T-cell Immunity in Patients with HER2 Breast Cancer using a Degenerate HLA Class II Epitope Vaccine. Clin Cancer Res 2019; 26:1045-1053. [PMID: 31757875 DOI: 10.1158/1078-0432.ccr-19-2123] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/14/2019] [Accepted: 11/18/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE Patients with HER2+ breast cancer benefit from trastuzumab-containing regimens with improved survival. Adaptive immunity, including cytotoxic T-cell and antibody immunity, is critical to clinical efficacy of trastuzumab. Because Th cells are central to the activation of these antitumor effectors, we reason that HER2 patients treated with trastuzumab may benefit by administering vaccines that are designed to stimulate Th-cell immunity. PATIENTS AND METHODS We developed a degenerate HER2 epitope-based vaccine consisting of four HLA class II-restricted epitopes mixed with GM-CSF that should immunize most (≥84%) patients. The vaccine was tested in a phase I trial. Eligible women had resectable HER2+ breast cancer and had completed standard treatment prior to enrollment and were disease free. Patients were vaccinated monthly for six doses and monitored for safety and immunogenicity. RESULTS Twenty-two subjects were enrolled and 20 completed all six vaccines. The vaccine was well tolerated. All patients were alive at analysis with a median follow-up of 2.3 years and only two experienced disease recurrence. The percent of patients that responded with augmented T-cell immunity was high for each peptide ranging from 68% to 88%, which led to 90% of the patients generating T cells that recognized naturally processed HER2 antigen. The vaccine also augmented HER2-specific antibody. Immunity was sustained in patients with little sign of diminishing at 2 years following the vaccination. CONCLUSIONS Degenerate HLA-DR-based HER2 vaccines induce sustainable HER2-specific T cells and antibodies. Future studies, could evaluate whether vaccination during adjuvant treatment with trastuzumab-containing regimens improves patient outcomes.
Collapse
Affiliation(s)
- Keith L Knutson
- Department of Immunology, Mayo Clinic, Jacksonville, Florida.
| | | | - Nadine Norton
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | | | | | - Allan B Dietz
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Douglas Padley
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Michael P Gustafson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | | | - Toni Kay Mangskau
- Mayo Clinic Cancer Education Program, Mayo Clinic, Rochester, Minnesota
| | | | - Amylou C Dueck
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, Arizona
| | | | | | - Amy C Degnim
- Department of Surgery, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
6
|
Zhou L, Li Y, Li X, Chen G, Liang H, Wu Y, Tong J, Ouyang W. Propranolol Attenuates Surgical Stress-Induced Elevation of the Regulatory T Cell Response in Patients Undergoing Radical Mastectomy. THE JOURNAL OF IMMUNOLOGY 2016; 196:3460-9. [PMID: 26969754 DOI: 10.4049/jimmunol.1501677] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 02/11/2016] [Indexed: 12/27/2022]
Abstract
Surgical stress and inflammatory response induce the release of catecholamines and PGs, which may be key factors in facilitating cancer recurrence through immunosuppression. Animal studies have suggested the efficacy of perioperative blockades of catecholamines and PGs in reducing immunosuppression. In this study, to our knowledge, we present the first report of the effects of perioperative propranolol and/or parecoxib on peripheral regulatory T cells (Tregs) in breast cancer patients. Patients were randomly assigned to control, propranolol, parecoxib, and propranolol plus parecoxib groups. We demonstrated that levels of circulating epinephrine, norepinephrine, and PGE2increased in response to surgery. Meanwhile, peripheral FOXP3 mRNA level and Treg frequencies were elevated on postoperative day 7. Propranolol administration, rather than parecoxib, attenuated such elevation of Tregs, indicating the critical roles for catecholamines in surgery-induced promotion of Tregs. Besides, propranolol plus parecoxib treatment demonstrated no additive or synergistic effects. Furthermore, a study of Treg activity on CD4(+)T cell responses to specific tumor Ags was performed in the control and propranolol groups. Propranolol abrogated the increased Treg activity and accompanying suppression of CD4(+)T cell responses after surgery. Finally, we conducted ex vivo experiments on the effects of varying concentrations of epinephrine and/or propranolol on Treg proliferation over PBMCs from breast cancer patients, to provide further direct evidence strengthening our clinical observations. Epinephrine markedly promoted Treg proliferation, whereas propranolol prevented such enhancement effect. In conclusion, our study highlights beneficial roles for propranolol in inhibiting Treg responses in vivo and in vitro, and demonstrates that propranolol could alleviate surgical stress-induced elevation of Tregs in breast cancer patients.
Collapse
Affiliation(s)
- Lei Zhou
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, China
| | - Yunli Li
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, China
| | - Xiaoxiao Li
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, China
| | - Gong Chen
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, China
| | - Huiying Liang
- Department of Endocrinology, Second Xiangya Hospital of Central South University, Changsha 410008, Hunan, China
| | - Yuhui Wu
- Department of Breast Surgery, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China;
| | - Jianbin Tong
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, China; Medical Central Laboratory, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, China;
| | - Wen Ouyang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, China; Seniors Anesthesia and Perioperative Management Research Center, Central South University, Changsha 410013, Hunan, China; and State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410013, Hunan, China
| |
Collapse
|
7
|
Escalante P, Peikert T, Van Keulen VP, Erskine CL, Bornhorst CL, Andrist BR, McCoy K, Pease LR, Abraham RS, Knutson KL, Kita H, Schrum AG, Limper AH. Combinatorial Immunoprofiling in Latent Tuberculosis Infection. Toward Better Risk Stratification. Am J Respir Crit Care Med 2015; 192:605-17. [PMID: 26030344 DOI: 10.1164/rccm.201412-2141oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Most immunocompetent patients diagnosed with latent tuberculosis infection (LTBI) will not progress to tuberculosis (TB) reactivation. However, current diagnostic tools cannot reliably distinguish nonprogressing from progressing patients a priori, and thus LTBI therapy must be prescribed with suboptimal patient specificity. We hypothesized that LTBI diagnostics could be improved by generating immunomarker profiles capable of categorizing distinct patient subsets by a combinatorial immunoassay approach. OBJECTIVES A combinatorial immunoassay analysis was applied to identify potential immunomarker combinations that distinguish among unexposed subjects, untreated patients with LTBI, and treated patients with LTBI and to differentiate risk of reactivation. METHODS IFN-γ release assay (IGRA) was combined with a flow cytometric assay that detects induction of CD25(+)CD134(+) coexpression on TB antigen-stimulated T cells from peripheral blood. The combinatorial immunoassay analysis was based on receiver operating characteristic curves, technical cut-offs, 95% bivariate normal density ellipse prediction, and statistical analysis. Risk of reactivation was estimated with a prediction formula. MEASUREMENTS AND MAIN RESULTS Sixty-five out of 150 subjects were included. The combinatorial immunoassay approach identified at least four different T-cell subsets. The representation of these immune phenotypes was more heterogeneous in untreated patients with LTBI than in treated patients with LTBI or unexposed groups. Patients with IGRA(+) CD4(+)CD25(+)CD134(+) T-cell phenotypes had the highest estimated reactivation risk (4.11 ± 2.11%). CONCLUSIONS These findings suggest that immune phenotypes defined by combinatorial assays may potentially have a role in identifying those at risk of developing TB; this potential role is supported by risk of reactivation modeling. Prospective studies will be needed to test this novel approach.
Collapse
Affiliation(s)
- Patricio Escalante
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine.,2 Public Health Department, Olmsted County Tuberculosis Clinic, Rochester, Minnesota; and.,3 Mayo Clinic Center for Tuberculosis, Rochester, Minnesota
| | - Tobias Peikert
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine.,4 Department of Immunology, and
| | | | | | - Cathy L Bornhorst
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Boleyn R Andrist
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Kevin McCoy
- 2 Public Health Department, Olmsted County Tuberculosis Clinic, Rochester, Minnesota; and.,3 Mayo Clinic Center for Tuberculosis, Rochester, Minnesota
| | | | - Roshini S Abraham
- 5 Department of Laboratory Medicine, Mayo Clinic, Rochester, Minnesota
| | | | | | | | - Andrew H Limper
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine
| |
Collapse
|
8
|
Peres LDP, da Luz FAC, Pultz BDA, Brígido PC, de Araújo RA, Goulart LR, Silva MJB. Peptide vaccines in breast cancer: The immunological basis for clinical response. Biotechnol Adv 2015; 33:1868-77. [PMID: 26523780 DOI: 10.1016/j.biotechadv.2015.10.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 10/15/2015] [Accepted: 10/29/2015] [Indexed: 02/07/2023]
Abstract
This review discusses peptide-based vaccines in breast cancer, immune responses and clinical outcomes, which include studies on animal models and phase I, phase I/II, phase II and phase III clinical trials. Peptide-based vaccines are powerful neoadjuvant immunotherapies that can directly target proteins expressed in tumor cells, mainly tumor-associated antigens (TAAs). The most common breast cancer TAA epitopes are derived from MUC1, HER2/neu and CEA proteins. Peptides derived from TAAs could be successfully used to elicit CD8 and CD4 T cell-specific responses. Thus, choosing peptides that adapt to natural variations of human leukocyte antigen (HLA) genes is critical. The most attractive advantage is that the target response is more specific and less toxic than for other therapies and vaccines. Prominent studies on NeuVax - E75 (epitope for HER2/neu and GM-CSF) in breast cancer and DPX-0907 (HLA-A2-TAAs) expressed in breast cancer, ovarian and prostate cancer have shown the efficacy of peptide-based vaccines as neoadjuvant immunotherapy against cancer. Future peptide vaccine strategies, although a challenge to be applied in a broad range of breast cancers, point to the development of degenerate multi-epitope immunogens against multiple targets.
Collapse
Affiliation(s)
- Lívia de Paula Peres
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Instituto de Ciências Biomédicas (ICBIM) - Universidade Federal de Uberlândia - UFU, Uberlândia, MG, Brazil.
| | - Felipe Andrés Cordero da Luz
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Instituto de Ciências Biomédicas (ICBIM) - Universidade Federal de Uberlândia - UFU, Uberlândia, MG, Brazil
| | - Brunna dos Anjos Pultz
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Instituto de Ciências Biomédicas (ICBIM) - Universidade Federal de Uberlândia - UFU, Uberlândia, MG, Brazil
| | - Paula Cristina Brígido
- Laboratório de Tripanossomatídeos, Instituto de Ciências Biomédicas (ICBIM) - Universidade Federal de Uberlândia - UFU, Uberlândia, MG, Brazil
| | | | - Luiz Ricardo Goulart
- Laboratório de Nanobiotecnologia - Universidade Federal de Uberlândia - UFU, (INGEB), Uberlândia, MG, Brazil
| | - Marcelo José Barbosa Silva
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Instituto de Ciências Biomédicas (ICBIM) - Universidade Federal de Uberlândia - UFU, Uberlândia, MG, Brazil.
| |
Collapse
|
9
|
Mason PH, Domínguez D JF, Winter B, Grignolio A. Hidden in plain view: degeneracy in complex systems. Biosystems 2014; 128:1-8. [PMID: 25543071 DOI: 10.1016/j.biosystems.2014.12.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 12/20/2014] [Accepted: 12/23/2014] [Indexed: 12/27/2022]
Abstract
Degeneracy is a word with two meanings. The popular usage of the word denotes deviance and decay. In scientific discourse, degeneracy refers to the idea that different pathways can lead to the same output. In the biological sciences, the concept of degeneracy has been ignored for a few key reasons. Firstly, the word "degenerate" in popular culture has negative, emotionally powerful associations that do not inspire scientists to consider its technical meaning. Secondly, the tendency of searching for single causes of natural and social phenomena means that scientists can overlook the multi-stranded relationships between cause and effect. Thirdly, degeneracy and redundancy are often confused with each other. Degeneracy refers to dissimilar structures that are functionally similar while redundancy refers to identical structures. Degeneracy can give rise to novelty in ways that redundancy cannot. From genetic codes to immunology, vaccinology and brain development, degeneracy is a crucial part of how complex systems maintain their functional integrity. This review article discusses how the scientific concept of degeneracy was imported into genetics from physics and was later introduced to immunology and neuroscience. Using examples of degeneracy in immunology, neuroscience and linguistics, we demonstrate that degeneracy is a useful way of understanding how complex systems function. Reviewing the history and theoretical scope of degeneracy allows its usefulness to be better appreciated, its coherency to be further developed, and its application to be more quickly realized.
Collapse
Affiliation(s)
- P H Mason
- Woolcock Institute of Medical Research, University of Sydney, 431 Glebe Point Road, Glebe, 2037 NSW, Australia.
| | - J F Domínguez D
- Experimental Neuropsychology Research Unit, School of Psychological Sciences, Monash University, Australia
| | - B Winter
- Cognitive and Information Sciences, University of California, Merced 5200 North Lake Rd., Merced, CA 95343, USA
| | - A Grignolio
- Section and Museum of History of Medicine, University of Rome "La Sapienza", viale dell'Università, 34a 00185 Rome, Italy
| |
Collapse
|
10
|
Galanis E, Atherton PJ, Maurer MJ, Knutson KL, Dowdy SC, Cliby WA, Haluska P, Long HJ, Oberg A, Aderca I, Block MS, Bakkum-Gamez J, Federspiel MJ, Russell SJ, Kalli KR, Keeney G, Peng KW, Hartmann LC. Oncolytic measles virus expressing the sodium iodide symporter to treat drug-resistant ovarian cancer. Cancer Res 2014; 75:22-30. [PMID: 25398436 DOI: 10.1158/0008-5472.can-14-2533] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Edmonston vaccine strains of measles virus (MV) have significant antitumor activity in mouse xenograft models of ovarian cancer. MV engineered to express the sodium iodide symporter gene (MV-NIS) facilitates localization of viral gene expression and offers a tool for tumor radiovirotherapy. Here, we report results from a clinical evaluation of MV-NIS in patients with taxol- and platinum-resistant ovarian cancer. MV-NIS was given intraperitoneally every 4 weeks for up to 6 cycles. Treatment was well tolerated and associated with promising median overall survival in these patients with heavily pretreated ovarian cancer; no dose-limiting toxicity was observed in 16 patients treated at high-dose levels (10(8)-10(9) TCID50), and their median overall survival of 26.5 months compared favorably with other contemporary series. MV receptor CD46 and nectin-4 expression was confirmed by immunohistochemistry in patient tumors. Sodium iodide symporter expression in patient tumors after treatment was confirmed in three patients by (123)I uptake on SPECT/CTs and was associated with long progression-free survival. Immune monitoring posttreatment showed an increase in effector T cells recognizing the tumor antigens IGFBP2 and FRα, indicating that MV-NIS treatment triggered cellular immunity against the patients' tumor and suggesting that an immune mechanism mediating the observed antitumor effect. Our findings support further clinical evaluation of MV-NIS as an effective immunovirotherapy.
Collapse
Affiliation(s)
- Evanthia Galanis
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota. Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota.
| | | | | | - Keith L Knutson
- Department of Immunology, Mayo Clinic, Rochester, Minnesota. Vaccine and Gene Therapy Institute of Florida, Port Saint Lucie, Florida
| | - Sean C Dowdy
- Division of Gynecological Surgery, Mayo Clinic, Rochester, Minnesota
| | - William A Cliby
- Division of Gynecological Surgery, Mayo Clinic, Rochester, Minnesota
| | - Paul Haluska
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Harry J Long
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Ann Oberg
- Department of Statistics, Mayo Clinic, Rochester, Minnesota
| | - Ileana Aderca
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Matthew S Block
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | - Gary Keeney
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Kah Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Lynn C Hartmann
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
11
|
Mittendorf EA, Alatrash G, Xiao H, Clifton GT, Murray JL, Peoples GE. Breast cancer vaccines: ongoing National Cancer Institute-registered clinical trials. Expert Rev Vaccines 2014; 10:755-74. [DOI: 10.1586/erv.11.59] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
12
|
Johnson LE, McNeel DG. Identification of prostatic acid phosphatase (PAP) specific HLA-DR1-restricted T-cell epitopes. Prostate 2012; 72:730-40. [PMID: 22529020 DOI: 10.1002/pros.21477] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 07/18/2011] [Indexed: 11/08/2022]
Abstract
BACKGROUND Prostatic acid phosphatase (PAP) is a prostate cancer tumor antigen and is an immunological target in several active immunotherapy clinical trials for the treatment of prostate cancer. We and others have demonstrated that PAP-specific T-cell responses can be elicited and augmented following antigen-specific immunization in both humans and animal models. We have previously reported that prostate cancer patients immunized with a DNA vaccine encoding PAP (pTVG-HP) developed both CD4+ and CD8+ T-cell responses. PAP-specific, CD4+ T-cell proliferative responses were generated in three out of four HLA-DRB1*0101 patients suggesting the possibility that DR1-restricted epitopes exist. METHODS To identify PAP-specific HLA-DRB1*0101 restricted epitopes, we immunized HLA-A2.01/HLA-DRB1*0101 (A2/DR1) transgenic mice with the pTVG-HP DNA vaccine. To map DRB1*0101-restricted epitopes, splenocytes from immunized mice were screened against a library of overlapping 15-residue, PAP-derived peptides using an IFNγ ELISPOT assay. RESULTS We identified four HLA-DRB1*0101 epitopes for PAP in A2/DR1 mice (PAP(161-175) , PAP(181-195) , PAP(191-205) , and PAP (351-365) ). T cells specific for one epitope (PAP(181-195) ) were found to be augmented after immunization in a HLA-DRB1*0101+ prostate cancer patient. CONCLUSIONS The identification of MHC class II epitopes may provide tools to directly monitor immune responses after vaccination and may be important for the design of future prostate cancer vaccines.
Collapse
Affiliation(s)
- Laura E Johnson
- Department of Medicine, University of Wisconsin, Madison, Wisconsin 53705, USA
| | | |
Collapse
|
13
|
Preston CC, Goode EL, Hartmann LC, Kalli KR, Knutson KL. Immunity and immune suppression in human ovarian cancer. Immunotherapy 2011; 3:539-56. [PMID: 21463194 DOI: 10.2217/imt.11.20] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Clinical outcomes in ovarian cancer are heterogeneous, independent of common features such as stage, response to therapy and grade. This disparity in outcomes warrants further exploration into tumor and host characteristics. One compelling issue is the response of the patient's immune system to her ovarian cancer. Several studies have confirmed a prominent role for the immune system in modifying disease course. This has led to the identification and evaluation of novel immune-modulating therapeutic approaches such as vaccination and antibody therapy. Antitumor immunity, however, is often negated by immune suppression mechanisms present in the tumor microenvironment. Thus, in the future, research into immunotherapy targeting ovarian cancer will probably become increasingly focused on combination approaches that simultaneously augment immunity while preventing local immune suppression. In this article, we summarize important immunological issues that could influence ovarian cancer outcome, including tumor antigens, endogenous immune responses, immune escape and new and developing immunotherapeutic strategies.
Collapse
|
14
|
Norell H, Poschke I, Charo J, Wei WZ, Erskine C, Piechocki MP, Knutson KL, Bergh J, Lidbrink E, Kiessling R. Vaccination with a plasmid DNA encoding HER-2/neu together with low doses of GM-CSF and IL-2 in patients with metastatic breast carcinoma: a pilot clinical trial. J Transl Med 2010; 8:53. [PMID: 20529245 PMCID: PMC2903523 DOI: 10.1186/1479-5876-8-53] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Accepted: 06/07/2010] [Indexed: 11/24/2022] Open
Abstract
Background Adjuvant trastuzumab (Herceptin) treatment of breast cancer patients significantly improves their clinical outcome. Vaccination is an attractive alternative approach to provide HER-2/neu (Her2)-specific antibodies and may in addition concomitantly stimulate Her2-reactive T-cells. Here we report the first administration of a Her2-plasmid DNA (pDNA) vaccine in humans. Patients and Methods The vaccine, encoding a full-length signaling-deficient version of the oncogene Her2, was administered together with low doses of GM-CSF and IL-2 to patients with metastatic Her2-expressing breast carcinoma who were also treated with trastuzumab. Six of eight enrolled patients completed all three vaccine cycles. In the remaining two patients treatment was discontinued after one vaccine cycle due to rapid tumor progression or disease-related complications. The primary objective was the evaluation of safety and tolerability of the vaccine regimen. As a secondary objective, treatment-induced Her2-specific immunity was monitored by measuring antibody production as well as T-cell proliferation and cytokine production in response to Her2-derived antigens. Results No clinical manifestations of acute toxicity, autoimmunity or cardiotoxicity were observed after administration of Her2-pDNA in combination with GM-CSF, IL-2 and trastuzumab. No specific T-cell proliferation following in vitro stimulation of freshly isolated PBMC with recombinant human Her2 protein was induced by the vaccination. Immediately after all three cycles of vaccination no or even decreased CD4+ T-cell responses towards Her2-derived peptide epitopes were observed, but a significant increase of MHC class II restricted T-cell responses to Her2 was detected at long term follow-up. Since concurrent trastuzumab therapy was permitted, λ-subclass specific ELISAs were performed to specifically measure endogenous antibody production without interference by trastuzumab. Her2-pDNA vaccination induced and boosted Her2-specific antibodies that could be detected for several years after the last vaccine administration in a subgroup of patients. Conclusion This pilot clinical trial demonstrates that Her2-pDNA vaccination in conjunction with GM-CSF and IL-2 administration is safe, well tolerated and can induce long-lasting cellular and humoral immune responses against Her2 in patients with advanced breast cancer. Trial registration The trial registration number at the Swedish Medical Products Agency for this trial is Dnr151:785/2001.
Collapse
Affiliation(s)
- Håkan Norell
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|