1
|
Malla R, Srilatha M, Muppala V, Farran B, Chauhan VS, Nagaraju GP. Neoantigens and cancer-testis antigens as promising vaccine candidates for triple-negative breast cancer: Delivery strategies and clinical trials. J Control Release 2024; 370:707-720. [PMID: 38744346 DOI: 10.1016/j.jconrel.2024.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/15/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
Immunotherapy is gaining prominence as a promising strategy for treating triple-negative breast cancer (TNBC). Neoantigens (neoAgs) and cancer-testis antigens (CTAs) are tumor-specific targets originating from somatic mutations and epigenetic changes in cancer cells. These antigens hold great promise for personalized cancer vaccines, as supported by preclinical and early clinical evidence in TNBC. This review delves into the potential of neoAgs and CTAs as vaccine candidates, emphasizing diverse strategies and delivery approaches. It also highlights the current status of vaccination modalities undergoing clinical trials in TNBC therapy. A comprehensive understanding of neoAgs, CTAs, vaccination strategies, and innovative delivery methods is crucial for optimizing neoAg-based immunotherapies in clinical practice.
Collapse
Affiliation(s)
- RamaRao Malla
- Cancer Biology Lab, Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati 517502, AP, India
| | - Veda Muppala
- Department of Neuroscience, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Batoul Farran
- Division of Hematology and Oncology, Department of Medicine, Henry Ford Health, Detroit, MI 48202, USA
| | - Virander Singh Chauhan
- Molecular Medicine Group, Molecular Medicines International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
2
|
C-type lectin receptor DCIR contributes to hippocampal injury in acute neurotropic virus infection. Sci Rep 2021; 11:23819. [PMID: 34893671 PMCID: PMC8664856 DOI: 10.1038/s41598-021-03201-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
Neurotropic viruses target the brain and contribute to neurologic diseases. C-type lectin receptors (CLRs) are pattern recognition receptors that recognize carbohydrate structures on endogenous molecules and pathogens. The myeloid CLR dendritic cell immunoreceptor (DCIR) is expressed by antigen presenting cells and mediates inhibitory intracellular signalling. To investigate the effect of DCIR on neurotropic virus infection, mice were infected experimentally with Theiler’s murine encephalomyelitis virus (TMEV). Brain tissue of TMEV-infected C57BL/6 mice and DCIR−/− mice were analysed by histology, immunohistochemistry and RT-qPCR, and spleen tissue by flow cytometry. To determine the impact of DCIR deficiency on T cell responses upon TMEV infection in vitro, antigen presentation assays were utilised. Genetic DCIR ablation in C57BL/6 mice was associated with an ameliorated hippocampal integrity together with reduced cerebral cytokine responses and reduced TMEV loads in the brain. Additionally, absence of DCIR favoured increased peripheral cytotoxic CD8+ T cell responses following TMEV infection. Co-culture experiments revealed that DCIR deficiency enhances the activation of antigen-specific CD8+ T cells by virus-exposed dendritic cells (DCs), indicated by increased release of interleukin-2 and interferon-γ. Results suggest that DCIR deficiency has a supportive influence on antiviral immune mechanisms, facilitating virus control in the brain and ameliorates neuropathology during acute neurotropic virus infection.
Collapse
|
3
|
Pavasutthipaisit S, Stoff M, Ebbecke T, Ciurkiewicz M, Mayer-Lambertz S, Störk T, Pavelko KD, Lepenies B, Beineke A. CARD9 Deficiency Increases Hippocampal Injury Following Acute Neurotropic Picornavirus Infection but Does Not Affect Pathogen Elimination. Int J Mol Sci 2021; 22:ijms22136982. [PMID: 34209576 PMCID: PMC8268812 DOI: 10.3390/ijms22136982] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/20/2021] [Accepted: 06/25/2021] [Indexed: 12/11/2022] Open
Abstract
Neurotropic viruses target the brain and contribute to neurologic diseases. Caspase recruitment domain containing family member 9 (CARD9) controls protective immunity in a variety of infectious disorders. To investigate the effect of CARD9 in neurotropic virus infection, CARD9−/− and corresponding C57BL/6 wild-type control mice were infected with Theiler’s murine encephalomyelitis virus (TMEV). Brain tissue was analyzed by histology, immunohistochemistry and molecular analyses, and spleens by flow cytometry. To determine the impact of CARD9 deficiency on T cell responses in vitro, antigen presentation assays were utilized. Genetic ablation of CARD9 enhanced early pro-inflammatory cytokine responses and accelerated infiltration of T and B cells in the brain, together with a transient increase in TMEV-infected cells in the hippocampus. CARD9−/− mice showed an increased loss of neuronal nuclear protein+ mature neurons and doublecortin+ neuronal precursor cells and an increase in β-amyloid precursor protein+ damaged axons in the hippocampus. No effect of CARD9 deficiency was found on the initiation of CD8+ T cell responses by flow cytometry and co-culture experiments using virus-exposed dendritic cells or microglia-enriched glial cell mixtures, respectively. The present study indicates that CARD9 is dispensable for the initiation of early antiviral responses and TMEV elimination but may contribute to the modulation of neuroinflammation, thereby reducing hippocampal injury following neurotropic virus infection.
Collapse
Affiliation(s)
- Suvarin Pavasutthipaisit
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (S.P.); (M.S.); (M.C.); (T.S.)
- Center for Systems Neuroscience, 30559 Hannover, Germany; (T.E.); (B.L.)
- Department of Pathology, Faculty of Veterinary Medicine, Mahanakorn University of Technology, Bangkok 10530, Thailand
| | - Melanie Stoff
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (S.P.); (M.S.); (M.C.); (T.S.)
| | - Tim Ebbecke
- Center for Systems Neuroscience, 30559 Hannover, Germany; (T.E.); (B.L.)
- Institute for Immunology and Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hannover, Germany;
| | - Malgorzata Ciurkiewicz
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (S.P.); (M.S.); (M.C.); (T.S.)
| | - Sabine Mayer-Lambertz
- Institute for Immunology and Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hannover, Germany;
| | - Theresa Störk
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (S.P.); (M.S.); (M.C.); (T.S.)
| | - Kevin D. Pavelko
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA;
| | - Bernd Lepenies
- Center for Systems Neuroscience, 30559 Hannover, Germany; (T.E.); (B.L.)
- Institute for Immunology and Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hannover, Germany;
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (S.P.); (M.S.); (M.C.); (T.S.)
- Center for Systems Neuroscience, 30559 Hannover, Germany; (T.E.); (B.L.)
- Correspondence: ; Tel.: +49-51-195-38640
| |
Collapse
|
4
|
Targeting Glioblastoma: Advances in Drug Delivery and Novel Therapeutic Approaches. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000124] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
5
|
Neier SC, Ferrer A, Wilton KM, Smith SEP, Kelcher AMH, Pavelko KD, Canfield JM, Davis TR, Stiles RJ, Chen Z, McCluskey J, Burrows SR, Rossjohn J, Hebrink DM, Carmona EM, Limper AH, Kappes DJ, Wettstein PJ, Johnson AJ, Pease LR, Daniels MA, Neuhauser C, Gil D, Schrum AG. The early proximal αβ TCR signalosome specifies thymic selection outcome through a quantitative protein interaction network. Sci Immunol 2020; 4:4/32/eaal2201. [PMID: 30770409 DOI: 10.1126/sciimmunol.aal2201] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 01/17/2019] [Indexed: 12/18/2022]
Abstract
During αβ T cell development, T cell antigen receptor (TCR) engagement transduces biochemical signals through a protein-protein interaction (PPI) network that dictates dichotomous cell fate decisions. It remains unclear how signal specificity is communicated, instructing either positive selection to advance cell differentiation or death by negative selection. Early signal discrimination might occur by PPI signatures differing qualitatively (customized, unique PPI combinations for each signal), quantitatively (graded amounts of a single PPI series), or kinetically (speed of PPI pathway progression). Using a novel PPI network analysis, we found that early TCR-proximal signals distinguishing positive from negative selection appeared to be primarily quantitative in nature. Furthermore, the signal intensity of this PPI network was used to find an antigen dose that caused a classic negative selection ligand to induce positive selection of conventional αβ T cells, suggesting that the quantity of TCR triggering was sufficient to program selection outcome. Because previous work had suggested that positive selection might involve a qualitatively unique signal through CD3δ, we reexamined the block in positive selection observed in CD3δ0 mice. We found that CD3δ0 thymocytes were inhibited but capable of signaling positive selection, generating low numbers of MHC-dependent αβ T cells that expressed diverse TCR repertoires and participated in immune responses against infection. We conclude that the major role for CD3δ in positive selection is to quantitatively boost the signal for maximal generation of αβ T cells. Together, these data indicate that a quantitative network signaling mechanism through the early proximal TCR signalosome determines thymic selection outcome.
Collapse
Affiliation(s)
- Steven C Neier
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA.,Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Alejandro Ferrer
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Katelynn M Wilton
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA.,Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA.,Medical Scientist Training Program, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Stephen E P Smith
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - April M H Kelcher
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA.,Mayo Graduate School, Mayo Clinic College of Medicine, Rochester, MN, USA.,Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Kevin D Pavelko
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Jenna M Canfield
- Molecular Pathogenesis and Therapeutics PhD Graduate Program, University of Missouri, Columbia, MO, USA
| | - Tessa R Davis
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Robert J Stiles
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Zhenjun Chen
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia
| | - James McCluskey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Scott R Burrows
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia.,School of Medicine, University of Queensland, Brisbane, Queensland 4006, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia.,Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
| | - Deanne M Hebrink
- Thoracic Diseases Research Unit, Division of Pulmonary Critical Care and Internal Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Eva M Carmona
- Thoracic Diseases Research Unit, Division of Pulmonary Critical Care and Internal Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Andrew H Limper
- Thoracic Diseases Research Unit, Division of Pulmonary Critical Care and Internal Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Dietmar J Kappes
- Blood Cell Development and Cancer Keystone, Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Peter J Wettstein
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA.,Department of Surgery, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA.,Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Larry R Pease
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Mark A Daniels
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA.,Department of Surgery, School of Medicine, University of Missouri, Columbia, MO, USA
| | | | - Diana Gil
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA. .,Department of Surgery, School of Medicine, University of Missouri, Columbia, MO, USA.,Department of Bioengineering, College of Engineering, University of Missouri, Columbia, MO, USA
| | - Adam G Schrum
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA. .,Department of Surgery, School of Medicine, University of Missouri, Columbia, MO, USA.,Department of Bioengineering, College of Engineering, University of Missouri, Columbia, MO, USA
| |
Collapse
|
6
|
McCarthy C, Jayawardena N, Burga LN, Bostina M. Developing Picornaviruses for Cancer Therapy. Cancers (Basel) 2019; 11:E685. [PMID: 31100962 PMCID: PMC6562951 DOI: 10.3390/cancers11050685] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/02/2019] [Accepted: 05/08/2019] [Indexed: 12/24/2022] Open
Abstract
Oncolytic viruses (OVs) form a group of novel anticancer therapeutic agents which selectively infect and lyse cancer cells. Members of several viral families, including Picornaviridae, have been shown to have anticancer activity. Picornaviruses are small icosahedral non-enveloped, positive-sense, single-stranded RNA viruses infecting a wide range of hosts. They possess several advantages for development for cancer therapy: Their genomes do not integrate into host chromosomes, do not encode oncogenes, and are easily manipulated as cDNA. This review focuses on the picornaviruses investigated for anticancer potential and the mechanisms that underpin this specificity.
Collapse
Affiliation(s)
- Cormac McCarthy
- Department of Microbiology and Immunology, University of Otago, Dunedin 9016, New Zealand.
| | - Nadishka Jayawardena
- Department of Microbiology and Immunology, University of Otago, Dunedin 9016, New Zealand.
| | - Laura N Burga
- Department of Microbiology and Immunology, University of Otago, Dunedin 9016, New Zealand.
| | - Mihnea Bostina
- Department of Microbiology and Immunology, University of Otago, Dunedin 9016, New Zealand.
- Otago Micro and Nano Imaging, University of Otago, Dunedin 9016, New Zealand.
| |
Collapse
|
7
|
Breaking tolerance with engineered class I antigen-presenting molecules. Proc Natl Acad Sci U S A 2019; 116:3136-3145. [PMID: 30728302 DOI: 10.1073/pnas.1807465116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Successful efforts to activate T cells capable of recognizing weak cancer-associated self-antigens have employed altered peptide antigens to activate T cell responses capable of cross-reacting on native tumor-associated self. A limitation of this approach is the requirement for detailed knowledge about the altered self-peptide ligands used in these vaccines. In the current study we considered allorecognition as an approach for activating CTL capable of recognizing weak or self-antigens in the context of self-MHC. Nonself antigen-presenting molecules typically contain polymorphisms that influence interactions with the bound peptide and TCR interface. Recognition of these nonself structures results in peptide-dependent alloimmunity. Alloreactive T cells target their inducing alloantigens as well as third-party alloantigens but generally fail to target self-antigens. Certain residues located on the alpha-1/2 domains of class I antigen-presenting molecules primarily interface with TCR. These residues are more conserved within and across species than are residues that determine peptide antigen binding properties. Class I variants designed with amino acid substitutions at key positions within the conserved helical structures are shown to provide strong activating signals to alloreactive CD8 T cells while avoiding changes in naturally bound peptide ligands. Importantly, CTL activated in this manner can break self-tolerance by reacting to self-peptides presented by native MHC. The ability to activate self-tolerant T cells capable of cross-reacting on self-peptide-MHC in vivo represents an approach for inducing autoimmunity, with possible application in cancer vaccines.
Collapse
|
8
|
Malo CS, Huggins MA, Goddery EN, Tolcher HMA, Renner DN, Jin F, Hansen MJ, Pease LR, Pavelko KD, Johnson AJ. Non-equivalent antigen presenting capabilities of dendritic cells and macrophages in generating brain-infiltrating CD8 + T cell responses. Nat Commun 2018; 9:633. [PMID: 29434238 PMCID: PMC5809416 DOI: 10.1038/s41467-018-03037-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 01/15/2018] [Indexed: 01/05/2023] Open
Abstract
The contribution of antigen-presenting cell (APC) types in generating CD8+ T cell responses in the central nervous system (CNS) is not fully defined, limiting the development of vaccines and understanding of immune-mediated neuropathology. Here, we generate a transgenic mouse that enables cell-specific deletion of the H-2Kb MHC class I molecule. By deleting H-2Kb on dendritic cells and macrophages, we compare the effect of each APC in three distinct models of neuroinflammation: picornavirus infection, experimental cerebral malaria, and a syngeneic glioma. Dendritic cells and macrophages both activate CD8+ T cell responses in response to these CNS immunological challenges. However, the extent to which each of these APCs contributes to CD8+ T cell priming varies. These findings reveal distinct functions for dendritic cells and macrophages in generating CD8+ T cell responses to neurological disease.
Collapse
Affiliation(s)
- Courtney S Malo
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Matthew A Huggins
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Emma N Goddery
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Heather M A Tolcher
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Danielle N Renner
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
- Neurobiology of Disease Graduate Program, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Fang Jin
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Michael J Hansen
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Larry R Pease
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Kevin D Pavelko
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA.
- Department of Neurology, Mayo Clinic, 200 First St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
9
|
Pavelko KD, Bell MP, Harrington SM, Dong H. B7-H1 Influences the Accumulation of Virus-Specific Tissue Resident Memory T Cells in the Central Nervous System. Front Immunol 2017; 8:1532. [PMID: 29170671 PMCID: PMC5684101 DOI: 10.3389/fimmu.2017.01532] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 10/27/2017] [Indexed: 01/01/2023] Open
Abstract
Therapies that target the PD-1/B7-H1 axis have revolutionized cancer treatment, yet precise knowledge of how this pathway provides benefit continues to evolve. Here, we report a novel role for the immune checkpoint ligand B7-H1 in the accumulation of tissue-resident memory CD8+ T-cells (TRM). After intracranial infection, Theiler's murine encephalomyelitis virus (TMEV) generates TRM that are maintained in the central nervous system (CNS) tissues of B7-H1WT animals. Although no differences in acute T-cell responses between B7-H1WT and B7-H1KO are observed, at long-term periods post-infection the maintenance of CD8+ TRM is diminished in B7-H1KO animals. This is accompanied by redistribution of the resident CD8+ population from primarily CD103+ TRM to a diminished population of TRM and a preponderance of non-specified PD-1+ CD103- CD8+ T-cells. T-cell transfer studies demonstrate that host B7-H1 is necessary for maintaining TRM and limiting accumulation of PD-1+ CD103- CD8+ T-cells. The lack of host B7-H1 results in compromised control of a heterologous virus re-challenge demonstrating a functional defect in TRM mediated virus control. This study reveals a new role for B7-H1 in TRM and pro-inflammatory PD-1+ CD103- CD8+ T-cell accumulation in the CNS and gives insight for using B7-H1/PD-1 blockade in modulating long-term T-cell protection.
Collapse
Affiliation(s)
- Kevin D. Pavelko
- Department of Immunology, College of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Michael P. Bell
- Department of Immunology, College of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Susan M. Harrington
- Department of Immunology, College of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Haidong Dong
- Department of Immunology, College of Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Urology, College of Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
10
|
The Effect of Vector Silencing during Picornavirus Vaccination against Experimental Melanoma and Glioma. PLoS One 2016; 11:e0162064. [PMID: 27560502 PMCID: PMC4999064 DOI: 10.1371/journal.pone.0162064] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/16/2016] [Indexed: 01/06/2023] Open
Abstract
Virus vector-based vaccination against tumor-specific antigens remains a promising therapeutic approach to overcome the immune suppressive tumor microenvironment. However, the extent that the desired CD8 T cell response against the targeted tumor antigen is impacted by the CD8 T cell response against the virus vector is unclear. To address this question, we used picornavirus vaccination with Theiler’s murine encephalomyelitis virus (TMEV) as our vector against tumor-expressed ovalbumin (OVA257-264) antigen in both the B16-OVA murine melanoma and GL261-quad cassette murine glioma models. Prior to vaccination, we employed vector silencing to inhibit the CD8 T cell response against the immunodominant TMEV antigen, VP2121-130. We then monitored the resulting effect on the CD8 T cell response against the targeted tumor-specific antigen, ovalbumin. We demonstrate that employing vector silencing in the context of B16-OVA melanoma does not reduce tumor burden or improve survival, while TMEV-OVA vaccination without vector silencing controls tumor burden. Meanwhile, employing vector silencing during picornavirus vaccination against the GL261-quad cassette glioma resulted in a lower frequency of tumor antigen-specific CD8 T cells. The results of this study are relevant to antigen-specific immunotherapy, in that the virus vector-specific CD8 T cell response is not competing with tumor antigen-specific CD8 T cells. Furthermore, vector silencing may have the adverse consequence of reducing the tumor antigen-specific CD8 T cell response, as demonstrated by our findings in the GL261-quad cassette model.
Collapse
|
11
|
Bell MP, Pavelko KD. Enhancing the Tumor Selectivity of a Picornavirus Virotherapy Promotes Tumor Regression and the Accumulation of Infiltrating CD8+ T Cells. Mol Cancer Ther 2016; 15:523-30. [PMID: 26823492 DOI: 10.1158/1535-7163.mct-15-0459] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 11/29/2015] [Indexed: 01/09/2023]
Abstract
Picornaviruses have emerged as promising cancer therapies due to their ability to drive cytotoxic cellular immune responses and for promoting oncolysis. These properties include preferential replication in tumor cells, the induction of strong innate and adaptive immune responses, and the ease with which their genomes can be manipulated. We have developed Theiler's murine encephalomyelitis virus (TMEV) as an immunotherapy vector that promotes strong adaptive immune responses to tumor antigens embedded within its genome. To further explore its usefulness as cancer therapy, we investigated whether direct intratumoral delivery of TMEV could promote tumor regression. We generated several picornavirus hybrids using substrains of TMEV that have unique immunopathologic characteristics, despite their extensive sequence homology. These hybrids exhibit a unique propensity to infect and replicate in melanoma. We have identified GD7-KS1, a virus that is particularly effective at replicating and infecting B16 melanoma in vitro and provides benefit as an oncolytic therapy in vivo after intratumoral injection. In addition, this virus promotes the mobilization and accumulation of CD8(+) T cells within treated tumors. Altogether, these findings demonstrate that picornavirus substrains can be used to rationally design virus hybrids that promote antitumor responses and add to the known strategies identified by us and others to further enhance the therapeutic potential of vectors used to treat cancer.
Collapse
Affiliation(s)
- Michael P Bell
- Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | - Kevin D Pavelko
- Department of Immunology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
12
|
Renner DN, Malo CS, Jin F, Parney IF, Pavelko KD, Johnson AJ. Improved Treatment Efficacy of Antiangiogenic Therapy when Combined with Picornavirus Vaccination in the GL261 Glioma Model. Neurotherapeutics 2016; 13:226-36. [PMID: 26620211 PMCID: PMC4720676 DOI: 10.1007/s13311-015-0407-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The addition of antiangiogenic therapy to the standard-of-care treatment regimen for recurring glioblastoma has provided some clinical benefits while also delineating numerous caveats, prompting evaluation of the elicited alterations to the tumor microenvironment. Of critical importance, given the steadily increasing incorporation of immunotherapeutic approaches clinically, is an enhanced understanding of the interplay between angiogenic and immune response pathways within tumors. In the present study, the GL261 glioma mouse model was used to determine the effects of antiangiogenic treatment in an immune-competent host. Following weekly systemic administration of aflibercept, an inhibitor of vascular endothelial growth factor, tumor volume was assessed by magnetic resonance imaging and changes to the tumor microenvironment were determined. Treatment with aflibercept resulted in reduced tumor burden and increased survival compared with controls. Additionally, decreased vascular permeability and preservation of the integrity of tight junction proteins were observed. Treated tumors also displayed hallmarks of anti-angiogenic evasion, including marked upregulation of vascular endothelial growth factor expression and increased tumor invasiveness. Aflibercept was then administered in combination with a picornavirus-based antitumor vaccine and tumor progression was evaluated. This combination therapy significantly delayed tumor progression and extended survival beyond that observed for either therapy alone. As such, this work demonstrates the efficacy of combined antiangiogenic and immunotherapy approaches for treating established gliomas and provides a foundation for further evaluation of the effects of antiangiogenic therapy in the context of endogenous or vaccine-induced inflammatory responses.
Collapse
Affiliation(s)
- Danielle N Renner
- Neurobiology of Disease Graduate Program, Mayo Clinic, Rochester, MN, USA
| | | | - Fang Jin
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Ian F Parney
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, USA
| | | | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
13
|
Chan SL, Lindquist LD, Hansen MJ, Girtman MA, Pease LR, Bram RJ. Calcium-Modulating Cyclophilin Ligand Is Essential for the Survival of Activated T Cells and for Adaptive Immunity. THE JOURNAL OF IMMUNOLOGY 2015; 195:5648-56. [PMID: 26561552 DOI: 10.4049/jimmunol.1500308] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 10/18/2015] [Indexed: 01/26/2023]
Abstract
Calcium-modulating cyclophilin ligand (CAML) is an endoplasmic reticulum resident protein that is widely expressed. Although it has been demonstrated to participate in the tail-anchored protein insertion pathway, its physiological role in the mature immune system is unknown. In this work, we show that mature, peripheral T cells require CAML for survival specifically following TCR-induced activation. In this study, we examined mature T cells from spleen and lymph nodes of tamoxifen-inducible CAML knockout mice (tCAML(-/-)). Whereas CAML-deficient T cells were able to express the early activation markers CD25 and CD69, and produce IL-2 normally upon stimulation, deficient cells proliferated less and died. Cells did not require CAML for entry into the S phase of the cell cycle, thus implicating its survival function at a relatively late step in the T cell activation sequence. In addition, CAML was required for homeostatic proliferation and for Ag-dependent cell killing in vivo. These results demonstrate that CAML critically supports T cell survival and cell division downstream of T cell activation.
Collapse
Affiliation(s)
- Siaw-Li Chan
- Department of Immunology, Mayo Clinic, Rochester, MN 55905; and
| | - Lonn D Lindquist
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905
| | | | - Megan A Girtman
- Department of Immunology, Mayo Clinic, Rochester, MN 55905; and
| | - Larry R Pease
- Department of Immunology, Mayo Clinic, Rochester, MN 55905; and
| | - Richard J Bram
- Department of Immunology, Mayo Clinic, Rochester, MN 55905; and Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
14
|
Renner DN, Jin F, Litterman AJ, Balgeman AJ, Hanson LM, Gamez JD, Chae M, Carlson BL, Sarkaria JN, Parney IF, Ohlfest JR, Pirko I, Pavelko KD, Johnson AJ. Effective Treatment of Established GL261 Murine Gliomas through Picornavirus Vaccination-Enhanced Tumor Antigen-Specific CD8+ T Cell Responses. PLoS One 2015; 10:e0125565. [PMID: 25933216 PMCID: PMC4416934 DOI: 10.1371/journal.pone.0125565] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 03/25/2015] [Indexed: 11/18/2022] Open
Abstract
Glioblastoma (GBM) is among the most invasive and lethal of cancers, frequently infiltrating surrounding healthy tissue and giving rise to rapid recurrence. It is therefore critical to establish experimental model systems and develop therapeutic approaches that enhance anti-tumor immunity. In the current study, we have employed a newly developed murine glioma model to assess the efficacy of a novel picornavirus vaccination approach for the treatment of established tumors. The GL261-Quad system is a variation of the GL261 syngeneic glioma that has been engineered to expresses model T cell epitopes including OVA257-264. MRI revealed that both GL261 and GL261-Quad tumors display characteristic features of human gliomas such as heterogeneous gadolinium leakage and larger T2 weighted volumes. Analysis of brain-infiltrating immune cells demonstrated that GL261-Quad gliomas generate detectable CD8+ T cell responses toward the tumor-specific Kb:OVA257-264 antigen. Enhancing this response via a single intracranial or peripheral vaccination with picornavirus expressing the OVA257-264 antigen increased anti-tumor CD8+ T cells infiltrating the brain, attenuated progression of established tumors, and extended survival of treated mice. Importantly, the efficacy of the picornavirus vaccination is dependent on functional cytotoxic activity of CD8+ T cells, as the beneficial response was completely abrogated in mice lacking perforin expression. Therefore, we have developed a novel system for evaluating mechanisms of anti-tumor immunity in vivo, incorporating the GL261-Quad model, 3D volumetric MRI, and picornavirus vaccination to enhance tumor-specific cytotoxic CD8+ T cell responses and track their effectiveness at eradicating established gliomas in vivo.
Collapse
Affiliation(s)
- Danielle N. Renner
- Neurobiology of Disease Graduate Program, Mayo Clinic, Rochester, MN, United States of America
- Department of Immunology, Mayo Clinic, Rochester, MN, United States of America
| | - Fang Jin
- Department of Immunology, Mayo Clinic, Rochester, MN, United States of America
| | - Adam J. Litterman
- Department of Neurosurgery, University of Minnesota, Minneapolis MN, United States of America
| | - Alexis J. Balgeman
- Summer Undergraduate Research Fellowship, Mayo Clinic, Rochester, MN, United States of America
| | - Lisa M. Hanson
- Department of Immunology, Mayo Clinic, Rochester, MN, United States of America
| | - Jeffrey D. Gamez
- Department of Neurology, Mayo Clinic, Rochester, MN, United States of America
| | - Michael Chae
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, United States of America
| | - Brett L. Carlson
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States of America
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States of America
| | - Ian F. Parney
- Department of Immunology, Mayo Clinic, Rochester, MN, United States of America
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, United States of America
| | - John R. Ohlfest
- Department of Neurosurgery, University of Minnesota, Minneapolis MN, United States of America
| | - Istvan Pirko
- Department of Neurology, Mayo Clinic, Rochester, MN, United States of America
| | - Kevin D. Pavelko
- Department of Immunology, Mayo Clinic, Rochester, MN, United States of America
- * E-mail: (AJJ); (KDP)
| | - Aaron J. Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN, United States of America
- Department of Neurology, Mayo Clinic, Rochester, MN, United States of America
- * E-mail: (AJJ); (KDP)
| |
Collapse
|
15
|
Bell MP, Renner DN, Johnson AJ, Pavelko KD. A CD8 T-cell epitope variant enhances immune targeting to a recombinant picornavirus vaccine antigen. Viral Immunol 2014; 27:361-6. [PMID: 25025983 DOI: 10.1089/vim.2014.0045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Recombinant virus vaccines are often less effective due to immunodominant responses against endogenous vector antigens. However, the use of small RNA virus vectors provides an opportunity to limit host exposure to endogenous virus antigens and focus immune responses on the desired vaccine antigen. Using the Daniel's strain of Theiler's murine encephalomyelitis virus, we have identified strategies to modulate responses to endogenous viral proteins by manipulating the host CD8+ T-cell repertoire prior to infection or through the use of mutations introduced into the virus genome. Both of these approaches enhance responses to vaccine antigens introduced into the picornavirus. However, the use of mutant immunodominant epitopes provides an opportunity for enhancing vaccine responses without further manipulation of the host. Using this strategy, we demonstrate that modification of the consensus MHC class I anchor residue within the virus genome can promote enhanced immunity to foreign antigens and self-antigens embedded in the virus genome.
Collapse
Affiliation(s)
- Michael P Bell
- 1 Department of Immunology, Mayo Graduate School, Mayo Clinic, Rochester, Minnesota
| | | | | | | |
Collapse
|
16
|
Bell MP, Renner DN, Johnson AJ, Pavelko KD. An elite controller of picornavirus infection targets an epitope that is resistant to immune escape. PLoS One 2014; 9:e94332. [PMID: 24710606 PMCID: PMC3978045 DOI: 10.1371/journal.pone.0094332] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 03/13/2014] [Indexed: 11/22/2022] Open
Abstract
The emergence of novel viral pathogens can lead to devastating consequences in the infected population. However, on occasion, rare hyper-responsive elite controllers are able to mount a protective primary response to infection and clear the new pathogen. Factors distinguishing elite controllers from other members of the population are not completely understood. We have been using Theiler's murine encephalomyelitis as a model of primary infection in mice and clearance of the virus is limited to one MHC genotype capable of generating a protective response to a single viral peptide VP2121-130. The genetics of host susceptibility to TMEV, a natural mouse pathogen, has been studied extensively and non-protective CD8 responses to other peptides have been documented, however, little is known why the protective response to infection focuses on the VP2121-130 peptide. To study this question, we have generated TMEV mutants that encode for mutations within the VP2121-130 peptide. We find that very few of mutants are able to assemble and infect in vitro. These mutations are not related to virus RNA structure since non-coding mutations do not interfere with assembly. In the rare event when functional VP2121-130 mutant viruses did emerge, they were attenuated to some level or retained the ability to develop an immune response to the wild-type VP2121-130 sequence, demonstrating that the virus is incapable of escaping the protective response. These findings advance our understanding of how characteristics of the host immune response and an infectious agent can interact to lead to the appearance of rare super controllers in a population. Furthermore, the immutable nature of the viral antigen highlights the importance of choosing appropriate vaccine antigens and has implications for the development of agents that are able to generate protective CD8 T-cell responses.
Collapse
Affiliation(s)
- Michael P. Bell
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Danielle N. Renner
- Neurobiology of Disease Program, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Aaron J. Johnson
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Neurology, Mayo Graduate School, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kevin D. Pavelko
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
17
|
Doddamane I, Butler R, Jhaveri A, Chung GG, Cheng D. Where does radioimmunotherapy fit in the management of breast cancer? Immunotherapy 2014; 5:895-904. [PMID: 23902558 DOI: 10.2217/imt.13.78] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Breast cancer is one of the most commonly diagnosed malignancies and is the main cause of death in women aged 40-49 years. Metastatic breast cancer is a heterogeneous disease that has a variety of different clinical presentations, ranging from solitary metastatic lesion to diffuse and multiple organ involvement. The biological heterogeneity of metastatic breast cancer has led to its unpredictable clinical behavior. One of the major challenges, therefore, is to identify predictive and prognostic models facilitating the selection of patients who can benefit from more aggressive and potentially curative options. This article provides an overview of the current management of metastatic breast cancer with focused emphasis on radioimmunotherapy.
Collapse
Affiliation(s)
- Indu Doddamane
- Department of Diagnostic Radiology, Yale University School of Medicine, 333 Cedar Street, PO Box 208042, New Haven, CT 06520-8042, USA.
| | | | | | | | | |
Collapse
|