1
|
Sánchez-Beltrán J, Soler Díaz J, Herraiz C, Olivares C, Cerdido S, Cerezuela-Fuentes P, García-Borrón JC, Jiménez-Cervantes C. An MGRN1-Based Biomarker Combination Accurately Predicts Melanoma Patient Survival. Int J Mol Sci 2025; 26:1739. [PMID: 40004203 PMCID: PMC11855888 DOI: 10.3390/ijms26041739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025] Open
Abstract
With ever-increasing incidence and high metastatic potential, cutaneous melanoma is the deadliest skin cancer. Risk prediction based on the Tumor-Node-Metastasis (TNM) staging system has medium accuracy with intermediate IIB-IIIB stages, as roughly 25% of patients with low-medium-grade TNM, and hence a favorable prognostic, undergo an aggressive disease with short survival and around 15% of deaths arise from metastases of thin, low-risk lesions. Therefore, reliable prognostic biomarkers are required. We used genomic and clinical information of melanoma patients from the TCGA-SKCM cohort and two GEO studies for discovery and validation of potential biomarkers, respectively. Neither mutation nor overexpression of major melanoma driver genes provided significant prognostic information. Conversely, expression of MGRN1 and the melanocyte-specific genes MLANA, PMEL, and TYRP1 provided a simple 4-gene signature identifying with high-sensitivity (>80%), low-medium TNM patients with adverse outcomes. Transcriptomic analysis of tumors with this signature, or from low-medium-grade TNM patients with poor outcomes, revealed comparable dysregulation of an inflammatory response, cell cycle progression, and DNA damage/repair programs. A functional analysis of MGRN1-knockout cells confirmed these molecular features. Therefore, the simple MGRN1-MLANA-PMEL-TYRP1 combination of biomarkers complemented TNM staging prognostic accuracy and pointed to the dysregulation of immunological responses and genomic stability as determinants of a melanoma outcome.
Collapse
Affiliation(s)
- José Sánchez-Beltrán
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, Campus de Excelencia Internacional Regional (CEIR), Campus Mare Nostrum (CMN), University of Murcia, 30120 Murcia, Spain; (J.S.-B.); (J.S.D.); (C.H.); (C.O.); (S.C.); (J.C.G.-B.)
- Biomedical Research Institute of Murcia (Instituto Murciano de Investigación Biosanitaria, IMIB), 30120 Murcia, Spain;
| | - Javier Soler Díaz
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, Campus de Excelencia Internacional Regional (CEIR), Campus Mare Nostrum (CMN), University of Murcia, 30120 Murcia, Spain; (J.S.-B.); (J.S.D.); (C.H.); (C.O.); (S.C.); (J.C.G.-B.)
| | - Cecilia Herraiz
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, Campus de Excelencia Internacional Regional (CEIR), Campus Mare Nostrum (CMN), University of Murcia, 30120 Murcia, Spain; (J.S.-B.); (J.S.D.); (C.H.); (C.O.); (S.C.); (J.C.G.-B.)
- Biomedical Research Institute of Murcia (Instituto Murciano de Investigación Biosanitaria, IMIB), 30120 Murcia, Spain;
| | - Conchi Olivares
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, Campus de Excelencia Internacional Regional (CEIR), Campus Mare Nostrum (CMN), University of Murcia, 30120 Murcia, Spain; (J.S.-B.); (J.S.D.); (C.H.); (C.O.); (S.C.); (J.C.G.-B.)
- Biomedical Research Institute of Murcia (Instituto Murciano de Investigación Biosanitaria, IMIB), 30120 Murcia, Spain;
| | - Sonia Cerdido
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, Campus de Excelencia Internacional Regional (CEIR), Campus Mare Nostrum (CMN), University of Murcia, 30120 Murcia, Spain; (J.S.-B.); (J.S.D.); (C.H.); (C.O.); (S.C.); (J.C.G.-B.)
- Biomedical Research Institute of Murcia (Instituto Murciano de Investigación Biosanitaria, IMIB), 30120 Murcia, Spain;
| | - Pablo Cerezuela-Fuentes
- Biomedical Research Institute of Murcia (Instituto Murciano de Investigación Biosanitaria, IMIB), 30120 Murcia, Spain;
- Medical Oncology Department, Hospital Clínico Universitario Virgen de La Arrixaca, 30120 Murcia, Spain
| | - José Carlos García-Borrón
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, Campus de Excelencia Internacional Regional (CEIR), Campus Mare Nostrum (CMN), University of Murcia, 30120 Murcia, Spain; (J.S.-B.); (J.S.D.); (C.H.); (C.O.); (S.C.); (J.C.G.-B.)
- Biomedical Research Institute of Murcia (Instituto Murciano de Investigación Biosanitaria, IMIB), 30120 Murcia, Spain;
| | - Celia Jiménez-Cervantes
- Department of Biochemistry, Molecular Biology and Immunology, School of Medicine, Campus de Excelencia Internacional Regional (CEIR), Campus Mare Nostrum (CMN), University of Murcia, 30120 Murcia, Spain; (J.S.-B.); (J.S.D.); (C.H.); (C.O.); (S.C.); (J.C.G.-B.)
- Biomedical Research Institute of Murcia (Instituto Murciano de Investigación Biosanitaria, IMIB), 30120 Murcia, Spain;
| |
Collapse
|
2
|
Abstract
PURPOSE OF REVIEW Checkpoint inhibitors block co-inhibitory signals which serves to promote T cell activation/reinvigoration in the periphery and tumor microenvironment. A brief historical background as well as a summary of key observations related to the composition and prognostic value of tumor-infiltrating lymphocytes (TILs) is discussed. RECENT FINDINGS Solid tumor patients that respond to checkpoint inhibitors have greater CD8+ T cell densities (at the tumor margin) associated with a gene inflammation signature and high tumor mutational burden. The precise specificity of effector (CD8+ T cell) TIL remains poorly defined and this deficiency represents a major challenge for the field of cancer immunology. High mutational burden cancers such as melanoma provides compelling evidence that missense mutations create neoantigens which can serve as target antigens for the immune system. Emerging evidence suggests that neoantigen-specific TILs are the major effector cells that mediate tumor regression due to checkpoint inhibition.
Collapse
Affiliation(s)
- Gerald P Linette
- Center for Cellular Immunotherapies and the Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA.
- Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, Philadelphia, PA, USA.
| | - Beatriz M Carreno
- Center for Cellular Immunotherapies and the Parker Institute for Cancer Immunotherapy, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
3
|
Lischer C, Eberhardt M, Jaitly T, Schinzel C, Schaft N, Dörrie J, Schuler G, Vera J. Curatopes Melanoma: A Database of Predicted T-cell Epitopes from Overly Expressed Proteins in Metastatic Cutaneous Melanoma. Cancer Res 2019; 79:5452-5456. [PMID: 31416842 DOI: 10.1158/0008-5472.can-19-0296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/25/2019] [Accepted: 08/08/2019] [Indexed: 11/16/2022]
Abstract
Therapeutic anticancer vaccination has been adapted as an immunotherapy in several solid tumors. However, the selection of promising candidates from the total quantity of possible epitopes poses a challenge to clinicians and bioinformaticians alike, and very few epitopes have been tested in experimental or clinical settings to validate their efficacy. Here, we present a comprehensive database of predicted nonmutated peptide epitopes derived from genes that are overly expressed in a group of 32 melanoma biopsies compared with healthy tissues and that were filtered against expression in a curated list of survival-critical tissues. We hypothesize that these "self-tolerant" epitopes have two desirable properties: they do not depend on mutations, being immediately applicable to a large patient collective, and they potentially cause fewer autoimmune reactions. To support epitope selection, we provide an aggregated score of expected therapeutic efficiency as a shortlist mechanism. The database has applications in facilitating epitope selection and trial design and is freely accessible at https://www.curatopes.com. SIGNIFICANCE: A database is presented that predicts and scores antitumor T-cell epitopes, with a focus on tolerability and avoidance of severe autoimmunity, offering a supplementary epitope set for further investigation in immunotherapy.
Collapse
Affiliation(s)
- Christopher Lischer
- Department of Dermatology, Universitätsklinikum Erlangen and Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Eberhardt
- Department of Dermatology, Universitätsklinikum Erlangen and Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tanushree Jaitly
- Department of Dermatology, Universitätsklinikum Erlangen and Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Cornelia Schinzel
- Department of Dermatology, Universitätsklinikum Erlangen and Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Niels Schaft
- Department of Dermatology, Universitätsklinikum Erlangen and Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jan Dörrie
- Department of Dermatology, Universitätsklinikum Erlangen and Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Gerold Schuler
- Department of Dermatology, Universitätsklinikum Erlangen and Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Julio Vera
- Department of Dermatology, Universitätsklinikum Erlangen and Faculty of Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
4
|
Zhao F, Zhang R, Wang J, Wu D, Pan M, Li M, Guo M, Dou J. Effective tumor immunity to melanoma mediated by B16F10 cancer stem cell vaccine. Int Immunopharmacol 2017; 52:238-244. [PMID: 28950176 DOI: 10.1016/j.intimp.2017.09.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/18/2017] [Accepted: 09/19/2017] [Indexed: 12/29/2022]
Abstract
Although tumor vaccines have been considered a promising immunotherapy approach, therapeutic tumor vaccines are mostly disappointing in the clinic due to vaccine weak immunogenicity. Cancer stem cells (CSCs) may broaden the antigenic breadth and effectively induce the immune responses against autologous cancer cells. Here we report on the development of the B16F10 CD133+CD44+CSCs (B16F10 CSCs) vaccine to induce tumor immunity to melanoma in mice. Efficacy of against melanoma was evaluated by analysis of tumor growth and mouse survival. Immunogenicity was assessed by ELISA and flow cytometric assays, including serum cytokines, cytotoxic activity of NK cells and splenocytes in the immunized mice. The results showed that the B16F10 CSC vaccine resulted in tumor shrinkage and mouse lifespan extension. The cytotoxic activity and IFN-γ level were significantly increased in mice immunized with B16F10 CSC vaccine compared with the mice immunized with control vaccines. Additionally, New York esophageal squamous cell carcinoma-1, an efficient tumor associated antigen over-expressed by B16F10 CSCs, was markedly reduced in expression in melanoma tissue, suggesting decrease of CSC subpopulation due to B16F10 CSC vaccination. Collectively, the findings may represent a new powerful approach for treatment of melanoma by B16F10 CSC vaccination.
Collapse
Affiliation(s)
- Fengshu Zhao
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Rong Zhang
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jing Wang
- Department of Gynecology & Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Di Wu
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Meng Pan
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Miao Li
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Mei Guo
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jun Dou
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
5
|
Allison KE, Coomber BL, Bridle BW. Metabolic reprogramming in the tumour microenvironment: a hallmark shared by cancer cells and T lymphocytes. Immunology 2017. [PMID: 28621843 DOI: 10.1111/imm.12777] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Altered metabolism is a hallmark of cancers, including shifting oxidative phosphorylation to glycolysis and up-regulating glutaminolysis to divert carbon sources into biosynthetic pathways that promote proliferation and survival. Therefore, metabolic inhibitors represent promising anti-cancer drugs. However, T cells must rapidly divide and survive in harsh microenvironments to mediate anti-cancer effects. Metabolic profiles of cancer cells and activated T lymphocytes are similar, raising the risk of metabolic inhibitors impairing the immune system. Immune checkpoint blockade provides an example of how metabolism can be differentially impacted to impair cancer cells but support T cells. Implications for research with metabolic inhibitors are discussed.
Collapse
Affiliation(s)
- Katrina E Allison
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Brenda L Coomber
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Byram W Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
6
|
Bentzen AK, Marquard AM, Lyngaa R, Saini SK, Ramskov S, Donia M, Such L, Furness AJS, McGranahan N, Rosenthal R, Straten PT, Szallasi Z, Svane IM, Swanton C, Quezada SA, Jakobsen SN, Eklund AC, Hadrup SR. Large-scale detection of antigen-specific T cells using peptide-MHC-I multimers labeled with DNA barcodes. Nat Biotechnol 2016; 34:1037-1045. [PMID: 27571370 DOI: 10.1038/nbt.3662] [Citation(s) in RCA: 237] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 08/04/2016] [Indexed: 01/17/2023]
Abstract
Identification of the peptides recognized by individual T cells is important for understanding and treating immune-related diseases. Current cytometry-based approaches are limited to the simultaneous screening of 10-100 distinct T-cell specificities in one sample. Here we use peptide-major histocompatibility complex (MHC) multimers labeled with individual DNA barcodes to screen >1,000 peptide specificities in a single sample, and detect low-frequency CD8 T cells specific for virus- or cancer-restricted antigens. When analyzing T-cell recognition of shared melanoma antigens before and after adoptive cell therapy in melanoma patients, we observe a greater number of melanoma-specific T-cell populations compared with cytometry-based approaches. Furthermore, we detect neoepitope-specific T cells in tumor-infiltrating lymphocytes and peripheral blood from patients with non-small cell lung cancer. Barcode-labeled pMHC multimers enable the combination of functional T-cell analysis with large-scale epitope recognition profiling for the characterization of T-cell recognition in various diseases, including in small clinical samples.
Collapse
Affiliation(s)
- Amalie Kai Bentzen
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Andrea Marion Marquard
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | - Rikke Lyngaa
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Sunil Kumar Saini
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Sofie Ramskov
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Marco Donia
- Center for Cancer Immune Therapy, Department of Hematology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Oncology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Lina Such
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| | - Andrew J S Furness
- CRUK Lung Cancer Center of Excellence, UCL Cancer Institute, London, UK
- Cancer Immunology Unit, UCL Cancer Institute, University College London, London, UK
| | - Nicholas McGranahan
- CRUK Lung Cancer Center of Excellence, UCL Cancer Institute, London, UK
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Rachel Rosenthal
- CRUK Lung Cancer Center of Excellence, UCL Cancer Institute, London, UK
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Per Thor Straten
- Center for Cancer Immune Therapy, Department of Hematology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Zoltan Szallasi
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy, Department of Hematology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Oncology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Charles Swanton
- CRUK Lung Cancer Center of Excellence, UCL Cancer Institute, London, UK
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Sergio A Quezada
- CRUK Lung Cancer Center of Excellence, UCL Cancer Institute, London, UK
- Cancer Immunology Unit, UCL Cancer Institute, University College London, London, UK
| | - Søren Nyboe Jakobsen
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
- Immudex, Copenhagen, Denmark
| | - Aron Charles Eklund
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | - Sine Reker Hadrup
- Section for Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Copenhagen, Denmark
| |
Collapse
|
7
|
Gfeller D, Bassani-Sternberg M, Schmidt J, Luescher IF. Current tools for predicting cancer-specific T cell immunity. Oncoimmunology 2016; 5:e1177691. [PMID: 27622028 DOI: 10.1080/2162402x.2016.1177691] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/06/2016] [Accepted: 04/06/2016] [Indexed: 12/20/2022] Open
Abstract
Tumor exome and RNA sequencing data provide a systematic and unbiased view on cancer-specific expression, over-expression, and mutations of genes, which can be mined for personalized cancer vaccines and other immunotherapies. Of key interest are tumor-specific mutations, because T cells recognizing neoepitopes have the potential to be highly tumoricidal. Here, we review recent developments and technical advances in identifying MHC class I and class II-restricted tumor antigens, especially neoantigen derived MHC ligands, including in silico predictions, immune-peptidome analysis by mass spectrometry, and MHC ligand validation by biochemical methods on T cells.
Collapse
Affiliation(s)
- David Gfeller
- Ludwig Center for Cancer Research, University of Lausanne, Epalinges, Switzerland; Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Julien Schmidt
- Ludwig Center for Cancer Research, University of Lausanne , Epalinges, Switzerland
| | - Immanuel F Luescher
- Ludwig Center for Cancer Research, University of Lausanne , Epalinges, Switzerland
| |
Collapse
|
8
|
Overgaard NH, Frøsig TM, Welner S, Rasmussen M, Ilsøe M, Sørensen MR, Andersen MH, Buus S, Jungersen G. Establishing the pig as a large animal model for vaccine development against human cancer. Front Genet 2015; 6:286. [PMID: 26442104 PMCID: PMC4584933 DOI: 10.3389/fgene.2015.00286] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 08/28/2015] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy has increased overall survival of metastatic cancer patients, and cancer antigens are promising vaccine targets. To fulfill the promise, appropriate tailoring of the vaccine formulations to mount in vivo cytotoxic T cell (CTL) responses toward co-delivered cancer antigens is essential. Previous development of therapeutic cancer vaccines has largely been based on studies in mice, and the majority of these candidate vaccines failed to induce therapeutic responses in the subsequent human clinical trials. Given that antigen dose and vaccine volume in pigs are translatable to humans and the porcine immunome is closer related to the human counterpart, we here introduce pigs as a supplementary large animal model for human cancer vaccine development. IDO and RhoC, both important in human cancer development and progression, were used as vaccine targets and 12 pigs were immunized with overlapping 20mer peptides spanning the entire porcine IDO and RhoC sequences formulated in CTL-inducing adjuvants: CAF09, CASAC, Montanide ISA 51 VG, or PBS. Taking advantage of recombinant swine MHC class I molecules (SLAs), the peptide-SLA complex stability was measured for 198 IDO- or RhoC-derived 9-11mer peptides predicted to bind to SLA-1*04:01, −1*07:02, −2*04:01, −2*05:02, and/or −3*04:01. This identified 89 stable (t½ ≥ 0.5 h) peptide-SLA complexes. By IFN-γ release in PBMC cultures we monitored the vaccine-induced peptide-specific CTL responses, and found responses to both IDO- and RhoC-derived peptides across all groups with no adjuvant being superior. These findings support the further use of pigs as a large animal model for vaccine development against human cancer.
Collapse
Affiliation(s)
- Nana H Overgaard
- Department of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark Copenhagen, Denmark
| | - Thomas M Frøsig
- Department of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark Copenhagen, Denmark
| | - Simon Welner
- Department of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark Copenhagen, Denmark
| | - Michael Rasmussen
- Department of International Health, Immunology and Microbiology, University of Copenhagen Copenhagen, Denmark
| | - Mette Ilsøe
- Department of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark Copenhagen, Denmark
| | - Maria R Sørensen
- Department of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark Copenhagen, Denmark
| | - Mads H Andersen
- Center for Cancer Immune Therapy, Department of Hematology, Copenhagen University Hospital Herlev, Denmark
| | - Søren Buus
- Department of International Health, Immunology and Microbiology, University of Copenhagen Copenhagen, Denmark
| | - Gregers Jungersen
- Department of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark Copenhagen, Denmark
| |
Collapse
|
9
|
Plant Viruses as Nanoparticle-Based Vaccines and Adjuvants. Vaccines (Basel) 2015; 3:620-37. [PMID: 26350598 PMCID: PMC4586470 DOI: 10.3390/vaccines3030620] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 07/22/2015] [Accepted: 07/29/2015] [Indexed: 12/11/2022] Open
Abstract
Vaccines are considered one of the greatest medical achievements in the battle against infectious diseases. However, the intractability of various diseases such as hepatitis C, HIV/AIDS, malaria, tuberculosis, and cancer poses persistent hurdles given that traditional vaccine-development methods have proven to be ineffective; as such, these challenges have driven the emergence of novel vaccine design approaches. In this regard, much effort has been put into the development of new safe adjuvants and vaccine platforms. Of particular interest, the utilization of plant virus-like nanoparticles and recombinant plant viruses has gained increasing significance as an effective tool in the development of novel vaccines against infectious diseases and cancer. The present review summarizes recent advances in the use of plant viruses as nanoparticle-based vaccines and adjuvants and their mechanism of action. Harnessing plant-virus immunogenic properties will enable the design of novel, safe, and efficacious prophylactic and therapeutic vaccines against disease.
Collapse
|