1
|
Fessler K, Zhang J, Sandhu AK, Hui Y, Kapoor AR, Ayeh SK, Karanika S, Karakousis PC, Markham RB, Gordy JT. Brief Communication: Combination of an MIP3α-Antigen Fusion Therapeutic DNA Vaccine With Treatments of IFNα and 5-Aza-2'Deoxycytidine Enhances Activated Effector CD8+ T Cells Expressing CD11c in the B16F10 Melanoma Model. J Immunother 2025; 48:1-5. [PMID: 39397434 PMCID: PMC11617275 DOI: 10.1097/cji.0000000000000542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 09/10/2024] [Indexed: 10/15/2024]
Abstract
Previous studies in the B16F10 mouse melanoma model have demonstrated that combining a DNA vaccine comprised of regions of gp100 and tyrosinase-related protein 2 fused to macrophage-inflammatory protein 3-alpha (MIP3α) with recombinant interferon alpha (IFN) and 5-Aza-2'-deoxycytidine (5Aza) treatments resulted in significantly greater antitumor activity and immunogenicity in the tumor microenvironment (TME). This brief report details that the combination of vaccine with treatments IFN and 5Aza results in an increase in the TME of a distinct CD11c+ CD8+ T-cell population. This cell population correlates with tumor size, is primarily comprised of effector or effector memory T cells, and has a more robust response to ex vivo stimulation as compared with CD11c- CD8+ T cells. In conclusion, this combination therapy results in a greater presence of highly active effector CD8+ T cells expressing CD11c in the TME, which are likely primary contributors to treatment efficacy.
Collapse
Affiliation(s)
- Kaitlyn Fessler
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Jiaqi Zhang
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Avinaash K. Sandhu
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Yinan Hui
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Aakanksha R. Kapoor
- Division of Infectious Diseases, Center for Tuberculosis Research, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
| | - Samuel K. Ayeh
- Division of Infectious Diseases, Center for Tuberculosis Research, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
| | - Styliani Karanika
- Division of Infectious Diseases, Center for Tuberculosis Research, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
| | - Petros C. Karakousis
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Division of Infectious Diseases, Center for Tuberculosis Research, Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD, United States
| | - Richard B. Markham
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - James T. Gordy
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
2
|
Fessler K, Zhang J, Sandhu AK, Hui Y, Kapoor AR, Ayeh SK, Karanika S, Karakousis PC, Markham RB, Gordy JT. Combination of a MIP3α-antigen fusion therapeutic DNA vaccine with treatments of IFNα and 5-Aza-2'Deoxycytidine enhances activated effector CD8+ T cells expressing CD11c in the B16F10 melanoma model. RESEARCH SQUARE 2024:rs.3.rs-3243336. [PMID: 37645859 PMCID: PMC10462250 DOI: 10.21203/rs.3.rs-3243336/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Previous studies in the B16F10 mouse melanoma model have demonstrated that combining a DNA vaccine comprised of regions of gp100 and tyrosinase-related protein 2 fused to Macrophage-inflammatory protein 3-alpha (MIP3α) with recombinant Interferon alpha (IFN) and 5-Aza-2'-Deoxycytidine (5Aza) treatments resulted in significantly greater anti-tumor activity and immunogenicity in the tumor microenvironment (TME). This brief report details that the combination of vaccine with treatments IFN and 5Aza results in both the upregulation of genes expressing CD11c-interacting proteins and an increase in the TME of a distinct CD11c+ CD8+ T cell population. This cell population correlates with tumor size, is primarily comprised of effector or effector memory T cells, and has a more robust response to ex vivo stimulation as compared to CD11c- CD8+ T cells as measured by surface activation markers 4-1BB (CD137) and KLRG1 (Killer cell lectin-like receptor G1) and intracellular IFNγ production. In conclusion, this combination therapy results in greater presence of highly active effector CD8+ T-cells expressing CD11c in the TME that correlate with and are likely primary contributors to treatment efficacy.
Collapse
Affiliation(s)
| | - Jiaqi Zhang
- Johns Hopkins Bloomberg School of Public Health
| | | | - Yinan Hui
- Johns Hopkins Bloomberg School of Public Health
| | | | | | | | | | | | | |
Collapse
|
3
|
Fessler K, Zhang J, Sandhu AK, Hui Y, Kapoor AR, Ayeh SK, Karanika S, Karakousis PC, Markham RB, Gordy JT. Combination of a MIP3α-antigen fusion therapeutic DNA vaccine with treatments of IFNα and 5-Aza-2'Deoxycytidine enhances activated effector CD8+ T cells expressing CD11c in the B16F10 melanoma model. RESEARCH SQUARE 2024:rs.3.rs-3243336. [PMID: 37645859 PMCID: PMC10462250 DOI: 10.21203/rs.3.rs-3243336/v2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Previous studies in the B16F10 mouse melanoma model have demonstrated that combining a DNA vaccine comprised of regions of gp100 and tyrosinase-related protein 2 fused to Macrophage-inflammatory protein 3-alpha (MIP3α) with recombinant Interferon alpha (IFN) and 5-Aza-2'-Deoxycytidine (5Aza) treatments resulted in significantly greater anti-tumor activity and immunogenicity in the tumor microenvironment (TME). This brief report details that the combination of vaccine with treatments IFN and 5Aza results in both the upregulation of genes expressing CD11c-interacting proteins and an increase in the TME of a distinct CD11c+ CD8+ T cell population. This cell population correlates with tumor size, is primarily comprised of effector or effector memory T cells, and has a more robust response to ex vivo stimulation as compared to CD11c- CD8+ T cells as measured by surface activation markers 4-1BB (CD137) and KLRG1 (Killer cell lectin-like receptor G1) and intracellular IFNγ production. In conclusion, this combination therapy results in greater presence of highly active effector CD8+ T-cells expressing CD11c in the TME that correlate with and are likely primary contributors to treatment efficacy.
Collapse
Affiliation(s)
| | - Jiaqi Zhang
- Johns Hopkins Bloomberg School of Public Health
| | | | - Yinan Hui
- Johns Hopkins Bloomberg School of Public Health
| | | | | | | | | | | | | |
Collapse
|
4
|
Pearson JRD, Puig-Saenz C, Thomas JE, Hardowar LD, Ahmad M, Wainwright LC, McVicar AM, Brentville VA, Tinsley CJ, Pockley AG, Durrant LG, McArdle SEB. TRP-2 / gp100 DNA vaccine and PD-1 checkpoint blockade combination for the treatment of intracranial tumors. Cancer Immunol Immunother 2024; 73:178. [PMID: 38954031 PMCID: PMC11219641 DOI: 10.1007/s00262-024-03770-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024]
Abstract
Intracranial tumors present a significant therapeutic challenge due to their physiological location. Immunotherapy presents an attractive method for targeting these intracranial tumors due to relatively low toxicity and tumor specificity. Here we show that SCIB1, a TRP-2 and gp100 directed ImmunoBody® DNA vaccine, generates a strong TRP-2 specific immune response, as demonstrated by the high number of TRP2-specific IFNγ spots produced and the detection of a significant number of pentamer positive T cells in the spleen of vaccinated mice. Furthermore, vaccine-induced T cells were able to recognize and kill B16HHDII/DR1 cells after a short in vitro culture. Having found that glioblastoma multiforme (GBM) expresses significant levels of PD-L1 and IDO1, with PD-L1 correlating with poorer survival in patients with the mesenchymal subtype of GBM, we decided to combine SCIB1 ImmunoBody® with PD-1 immune checkpoint blockade to treat mice harboring intracranial tumors expressing TRP-2 and gp100. Time-to-death was significantly prolonged, and this correlated with increased CD4+ and CD8+ T cell infiltration in the tissue microenvironment (TME). However, in addition to PD-L1 and IDO, the GBM TME was found to contain a significant number of immunoregulatory T (Treg) cell-associated transcripts, and the presence of such cells is likely to significantly affect clinical outcome unless also tackled.
Collapse
Affiliation(s)
- Joshua R D Pearson
- John Van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - Carles Puig-Saenz
- John Van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - Jubini E Thomas
- John Van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - Lydia D Hardowar
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Murrium Ahmad
- John Van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - Louise C Wainwright
- Bioscience Support Facility, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Adam M McVicar
- Bioscience Support Facility, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Victoria A Brentville
- Scancell Ltd, Unit 202, Bellhouse Building, Oxford Science Park, Sanders Road, Oxford, OX4 4GA, UK
| | - Chris J Tinsley
- John Van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - A Graham Pockley
- John Van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, UK
| | - Lindy G Durrant
- Scancell Ltd, Unit 202, Bellhouse Building, Oxford Science Park, Sanders Road, Oxford, OX4 4GA, UK
| | | |
Collapse
|
5
|
Tu HF, Wong M, Tseng SH, Ingavat N, Olczak P, Notarte KI, Hung CF, Roden RBS. Virus-like particle vaccine displaying an external, membrane adjacent MUC16 epitope elicits ovarian cancer-reactive antibodies. J Ovarian Res 2024; 17:19. [PMID: 38225646 PMCID: PMC10790439 DOI: 10.1186/s13048-023-01325-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 12/12/2023] [Indexed: 01/17/2024] Open
Abstract
BACKGROUND MUC16 is a heavily glycosylated cell surface mucin cleaved in the tumor microenvironment to shed CA125. CA125 is a serum biomarker expressed by > 95% of non-mucinous advanced stage epithelial ovarian cancers. MUC16/CA125 contributes to the evasion of anti-tumor immunity, peritoneal spread and promotes carcinogenesis; consequently, it has been targeted with antibody-based passive and active immunotherapy. However, vaccination against this self-antigen likely requires breaking B cell tolerance and may trigger autoimmune disease. Display of self-antigens on virus-like particles (VLPs), including those produced with human papillomavirus (HPV) L1, can efficiently break B cell tolerance. RESULTS A 20 aa juxta-membrane peptide of the murine MUC16 (mMUC16) or human MUC16 (hMUC16) ectodomain was displayed either via genetic insertion into an immunodominant loop of HPV16 L1-VLPs between residues 136/137, or by chemical coupling using malemide to cysteine sulfhydryl groups on their surface. Female mice were vaccinated intramuscularly three times with either DNA expressing L1-MUC16 fusions via electroporation, or with alum-formulated VLP chemically-coupled to MUC16 peptides. Both regimens were well tolerated, and elicited MUC16-specific serum IgG, although titers were higher in mice vaccinated with MUC16-coupled VLP on alum as compared to L1-MUC16 DNA vaccination. Antibody responses to mMUC16-targeted vaccination cross-reacted with hMUC16 peptide, and vice versa; both were reactive with the surface of CA125+ OVCAR3 cells, but not SKOV3 that lack detectable CA125 expression. Interestingly, vaccination of mice with mMUC16 peptide mixed with VLP and alum elicited mMUC16-specific IgG, implying VLPs provide robust T help and that coupling may not be required to break tolerance to this epitope. CONCLUSION Vaccination with VLP displaying the 20 aa juxta-membrane MUC16 ectodomain, which includes the membrane proximal cleavage site, is likely to be well tolerated and induce IgG targeting ovarian cancer cells, even after CA125 is shed.
Collapse
Affiliation(s)
- Hsin-Fang Tu
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Margaret Wong
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Ssu-Hsueh Tseng
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Nattha Ingavat
- Downstream Processing (DSP), Bioprocessing Technology Institute (BTI), Agency for Science, Technology, and Research (A*STAR), Singapore, 138632, Singapore
| | - Pola Olczak
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Kin Israel Notarte
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Richard B S Roden
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21287, USA.
- Department of Oncology, Johns Hopkins University, Baltimore, MD, 21287, USA.
- Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD, 21287, USA.
| |
Collapse
|
6
|
Shah S, Al-Omari A, Cook KW, Paston SJ, Durrant LG, Brentville VA. What do cancer-specific T cells 'see'? DISCOVERY IMMUNOLOGY 2022; 2:kyac011. [PMID: 38567060 PMCID: PMC10917189 DOI: 10.1093/discim/kyac011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/18/2022] [Accepted: 12/02/2022] [Indexed: 04/04/2024]
Abstract
Complex cellular interactions between the immune system and cancer can impact tumour development, growth, and progression. T cells play a key role in these interactions; however, the challenge for T cells is to recognize tumour antigens whilst minimizing cross-reactivity with antigens associated with healthy tissue. Some tumour cells, including those associated with viral infections, have clear, tumour-specific antigens that can be targeted by T cells. A high mutational burden can lead to increased numbers of mutational neoantigens that allow very specific immune responses to be generated but also allow escape variants to develop. Other cancer indications and those with low mutational burden are less easily distinguished from normal tissue. Recent studies have suggested that cancer-associated alterations in tumour cell biology including changes in post-translational modification (PTM) patterns may also lead to novel antigens that can be directly recognized by T cells. The PTM-derived antigens provide tumour-specific T-cell responses that both escape central tolerance and avoid the necessity for individualized therapies. PTM-specific CD4 T-cell responses have shown tumour therapy in murine models and highlight the importance of CD4 T cells as well as CD8 T cells in reversing the immunosuppressive tumour microenvironment. Understanding which cancer-specific antigens can be recognized by T cells and the way that immune tolerance and the tumour microenvironment shape immune responses to cancer is vital for the future development of cancer therapies.
Collapse
Affiliation(s)
- Sabaria Shah
- Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Abdullah Al-Omari
- Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Katherine W Cook
- Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Samantha J Paston
- Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Lindy G Durrant
- Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Victoria A Brentville
- Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| |
Collapse
|
7
|
Xu H, Yue Z, Pang H, Elahi E, Li J, Wang L. Integrative model for discovering linked topics in science and technology. J Informetr 2022. [DOI: 10.1016/j.joi.2022.101265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
8
|
He X, Zhou S, Quinn B, Jahagirdar D, Ortega J, Long MD, Abrams SI, Lovell JF. An In Vivo Screen to Identify Short Peptide Mimotopes with Enhanced Antitumor Immunogenicity. Cancer Immunol Res 2022; 10:314-326. [PMID: 34992135 DOI: 10.1158/2326-6066.cir-21-0332] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/23/2021] [Accepted: 01/04/2022] [Indexed: 11/16/2022]
Abstract
Tumor-associated self-antigens are potential cancer vaccine targets but suffer from limited immunogenicity. There are examples of mutated, short self-peptides inducing epitope-specific CD8⁺ T cells more efficiently than the wild-type epitope, but current approaches cannot yet reliably identify such epitopes, which are referred to as enhanced mimotopes ("e-mimotopes"). Here, we present a generalized strategy to develop e-mimotopes, using the tyrosinase-related protein 2 (Trp2) peptide Trp2180-188, which is a murine major histocompatibility complex class I (MHC-I) epitope, as a test case. Using a vaccine adjuvant that induces peptide particle formation and strong cellular responses with nanogram antigen doses, a two-step method systematically identified e-mimotope candidates with murine immunization. First, position-scanning peptide micro libraries were generated in which each position of the wild-type epitope sequence was randomized. Randomization of only one specific residue of the Trp2 epitope increased antitumor immunogenicity. Second, all 20 amino acids were individually substituted and tested at that position, enabling the identification of two e-mimotopes with single amino-acid mutations. Despite similar MHC-I affinity compared to the wild-type epitope, e-mimotope immunization elicited improved Trp2-specific cytotoxic T-cell phenotypes and improved T-cell receptor affinity for both the e-mimotopes and the native epitope, resulting in better outcomes in multiple prophylactic and therapeutic tumor models. The screening method was also applied to other targets with other murine MHC-I restriction elements, including epitopes within glycoprotein 70 and Wilms' Tumor Gene 1, to identify additional e-mimotopes with enhanced potency.
Collapse
Affiliation(s)
- Xuedan He
- Biomedical Engineering, University at Buffalo, State University of New York
| | - Shiqi Zhou
- Biomedical Engineering, University at Buffalo, State University of New York
| | - Breandan Quinn
- Biomedical Engineering, University at Buffalo, State University of New York
| | | | | | - Mark D Long
- Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center
| | | | - Jonathan F Lovell
- Biomedical Engineering, University at Buffalo, State University of New York
| |
Collapse
|
9
|
Rezaei T, Davoudian E, Khalili S, Amini M, Hejazi M, de la Guardia M, Mokhtarzadeh A. Strategies in DNA vaccine for melanoma cancer. Pigment Cell Melanoma Res 2021; 34:869-891. [PMID: 33089665 DOI: 10.1111/pcmr.12933] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/05/2020] [Accepted: 09/22/2020] [Indexed: 11/29/2022]
Abstract
According to reports of the international agency for cancer on research, although malignant melanoma shows less prevalence than nonmelanoma skin cancers, it is the major cause of skin cancer mortality. Given that, the production of effective vaccines to control melanoma is eminently required. In this regard, DNA-based vaccines have been extensively investigated for melanoma therapy. DNA vaccines are capable of inducing both cellular and humoral branches of immune responses. These vaccines possess some valuable advantages such as lack of severe side effects and high stability compared to conventional vaccination methods. The ongoing studies are focused on novel strategies in the development of DNA vaccines encoding artificial polyepitope immunogens based on the multiple melanoma antigens, the inclusion of molecular adjuvants to increase the level of immune responses, and the improvement of delivery approaches. In this review, we have outlined the recent advances in the field of melanoma DNA vaccines and described their implications in clinical trials as a strong strategy in the prevention and control of melanoma.
Collapse
Affiliation(s)
- Tayebeh Rezaei
- Department of Molecular Medicine and Biotechnology, Faculty of Medicine, Arak University of Medical Science, Arak, Iran
| | - Elham Davoudian
- Department of Microbiology, School of Paramedical Sciences, Ilam University of Medical Sciences, Ilam, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hejazi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
10
|
Cucu CI, Giurcăneanu C, Popa LG, Orzan OA, Beiu C, Holban AM, Grumezescu AM, Matei BM, Popescu MN, Căruntu C, Mihai MM. Electrochemotherapy and Other Clinical Applications of Electroporation for the Targeted Therapy of Metastatic Melanoma. MATERIALS 2021; 14:ma14143985. [PMID: 34300902 PMCID: PMC8305146 DOI: 10.3390/ma14143985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/10/2021] [Accepted: 07/11/2021] [Indexed: 12/21/2022]
Abstract
Electrochemotherapy (ECT) is an effective bioelectrochemical procedure that uses controlled electrical pulses to facilitate the increase of intracellular concentration of certain substances (electropermeabilization/ reversible electroporation). ECT using antitumor drugs such as bleomycin and cisplatin is a minimally invasive targeted therapy that can be used as an alternative for oncologic patients not eligible for surgery or other standard therapies. Even though ECT is mainly applied as palliative care for metastases, it may also be used for primary tumors that are unresectable due to size and location. Skin neoplasms are the main clinical indication of ECT, the procedure reporting good curative results and high efficiency across all tumor types, including melanoma. In daily practice, there are many cases in which the patient’s quality of life can be significantly improved by a safe procedure such as ECT. Its popularity must be increased because it has a safe profile and minor local adverse reactions. The method can be used by dermatologists, oncologists, and surgeons. The aim of this paper is to review recent literature concerning electrochemotherapy and other clinical applications of electroporation for the targeted therapy of metastatic melanoma.
Collapse
Affiliation(s)
- Corina Ioana Cucu
- Department of Oncologic Dermatology-“Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.I.C.); (C.G.); (O.A.O.); (C.B.); (M.M.M.)
| | - Călin Giurcăneanu
- Department of Oncologic Dermatology-“Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.I.C.); (C.G.); (O.A.O.); (C.B.); (M.M.M.)
| | - Liliana Gabriela Popa
- Department of Oncologic Dermatology-“Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.I.C.); (C.G.); (O.A.O.); (C.B.); (M.M.M.)
- Correspondence: ; Tel.: +40-727-173-767
| | - Olguța Anca Orzan
- Department of Oncologic Dermatology-“Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.I.C.); (C.G.); (O.A.O.); (C.B.); (M.M.M.)
| | - Cristina Beiu
- Department of Oncologic Dermatology-“Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.I.C.); (C.G.); (O.A.O.); (C.B.); (M.M.M.)
| | - Alina Maria Holban
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania;
- Research Institute of the University of Bucharest, 050657 Bucharest, Romania
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 1-7 Polizu Street, 011061 Bucharest, Romania;
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 1-7 Polizu Street, 011061 Bucharest, Romania;
| | - Bogdan Mircea Matei
- Department of Biophysics and Cellular Biotechnology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Marius Nicolae Popescu
- Department of Physical and Rehabilitation Medicine, “Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Constantin Căruntu
- Faculty of Medicine, “Titu Maiorescu” University, 22 Dambrovnicului, 031593 Bucharest, Romania;
| | - Mara Mădălina Mihai
- Department of Oncologic Dermatology-“Elias” Emergency University Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (C.I.C.); (C.G.); (O.A.O.); (C.B.); (M.M.M.)
- Research Institute of the University of Bucharest, 050657 Bucharest, Romania
| |
Collapse
|
11
|
Almshayakhchi R, Nagarajan D, Vadakekolathu J, Guinn BA, Reeder S, Brentville V, Metheringham R, Pockley AG, Durrant L, McArdle S. A Novel HAGE/WT1-ImmunoBody ® Vaccine Combination Enhances Anti-Tumour Responses When Compared to Either Vaccine Alone. Front Oncol 2021; 11:636977. [PMID: 34262856 PMCID: PMC8273701 DOI: 10.3389/fonc.2021.636977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 05/14/2021] [Indexed: 02/02/2023] Open
Abstract
Many cancers, including myeloid leukaemia express the cancer testis antigen (CTA) DDX43 (HAGE) and/or the oncogene Wilms’ tumour (WT1). Here we demonstrate that HAGE/WT1-ImmunoBody® vaccines derived T-cells can kill ex-vivo human CML cell lines expressing these antigens and significantly delay B16/HHDII+/DR1+/HAGE+/WT1+ tumour growth in the HHDII/DR1 mice and prolonged mouse survival in the prophylactic setting in comparison to non-immunised control mice. We show that immunisation of HHDII/DR1 mice with HAGE- and WT1-ImmunoBody® DNA vaccines in a prime-boost regime in two different flanks induce significant IFN-γ release by splenocytes from treated mice, and a significant level of cytotoxicity against tumour targets expressing HAGE/WT1 in vitro. More importantly, the combined HAGE/WT1 ImmunoBody® vaccine significantly delayed tumour growth in the B16/HHDII+/DR1+/HAGE+/WT1+ tumour model and prolonged mouse survival in the prophylactic setting in comparison to non-immunised control mice. Overall, this work demonstrates that combining both HAGE- and WT1-ImmunoBody® into a single vaccine is better than either vaccine alone. This combination vaccine could be given to patients whose cancer expresses HAGE and WT1 in parallel with existing therapies in order to decrease the chance of disease progression and relapse.
Collapse
Affiliation(s)
- Rukaia Almshayakhchi
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.,Centre for Health, Ageing and Understanding Disease, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Divya Nagarajan
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.,Centre for Health, Ageing and Understanding Disease, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Jayakumar Vadakekolathu
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.,Centre for Health, Ageing and Understanding Disease, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Barbara-Ann Guinn
- Department of Biomedical Sciences, University of Hull, Hull, United Kingdom
| | - Stephen Reeder
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.,Centre for Health, Ageing and Understanding Disease, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Victoria Brentville
- Scancell Ltd, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Rachael Metheringham
- Scancell Ltd, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - A Graham Pockley
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.,Centre for Health, Ageing and Understanding Disease, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Lindy Durrant
- Scancell Ltd, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Stephanie McArdle
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.,Centre for Health, Ageing and Understanding Disease, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| |
Collapse
|
12
|
Nagarajan D, Pearson J, Brentville V, Metheringham R, Pockley AG, Durrant L, McArdle SE. ImmunoBody®-HAGE derived vaccine induces immunity to HAGE and delays the growth and metastasis of HAGE-expressing tumours in vivo. Immunol Cell Biol 2021; 99:972-989. [PMID: 34105800 DOI: 10.1111/imcb.12485] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/28/2021] [Accepted: 06/07/2021] [Indexed: 12/01/2022]
Abstract
The management of patients with triple-negative breast cancer (TNBC) continues to pose a significant clinical challenge. Less than 30% of women with metastatic TNBC survive 5 years, despite adjuvant chemotherapy and the initial higher rates of clinical response that can be achieved with neoadjuvant chemotherapy. ImmunoBody® is a plasmid DNA designed to encode a human antibody molecule with complementary determining regions (CDRs) engineered to express cytotoxic and helper T cell epitopes derived from the cancer antigen of interest. HAGE is a Cancer Testis Antigen, which is expressed in TNBC. Herein, we have identified a 30-amino-acid-long HAGE-derived sequence containing HLA-A2 and HLA-DR1 restricted epitopes and demonstrated that the use of this sequence as peptide (with CpG/IFA) or incorporated into an ImmunoBody® vaccine can generate specific IFNγ secreting splenocytes in HHDII/DR1 mice. T-cell responses elicited by the ImmunoBody®-HAGE vaccine were superior to peptide immunisation. Moreover, splenocytes from ImmunoBody®-HAGE vaccinated mice stimulated in vitro could recognise HAGE+ tumour cells and the human TNBC cell line MDA-MB-231. More importantly, the growth of implanted B16/HHDII/DR1/HAGE+ cells was significantly delayed by the ImmunoBody®-HAGE vaccine in both prophylactic and experimental metastasis settings. Overall, we demonstrate the potential of HAGE-derived vaccines for treating HAGE-expressing cancers and that such vaccines could be considered as therapeutic options for patients with HAGE+ TNBC after conventional treatment to prevent disease recurrence.
Collapse
Affiliation(s)
- Divya Nagarajan
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Centre for Health, Ageing and Understanding Disease, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Joshua Pearson
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK.,Centre for Health, Ageing and Understanding Disease, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Victoria Brentville
- Scancell Ltd, Biodiscovery Institute, University of Nottingham, University Park, NG7 2RD, UK
| | - Rachael Metheringham
- Scancell Ltd, Biodiscovery Institute, University of Nottingham, University Park, NG7 2RD, UK
| | - A Graham Pockley
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Lindy Durrant
- Scancell Ltd, Biodiscovery Institute, University of Nottingham, University Park, NG7 2RD, UK
| | - Stephanie E McArdle
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| |
Collapse
|
13
|
Nagy NA, de Haas AM, Geijtenbeek TBH, van Ree R, Tas SW, van Kooyk Y, de Jong EC. Therapeutic Liposomal Vaccines for Dendritic Cell Activation or Tolerance. Front Immunol 2021; 12:674048. [PMID: 34054859 PMCID: PMC8155586 DOI: 10.3389/fimmu.2021.674048] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells (DCs) are paramount in initiating and guiding immunity towards a state of activation or tolerance. This bidirectional capacity of DCs sets them at the center stage for treatment of cancer and autoimmune or allergic conditions. Accordingly, many clinical studies use ex vivo DC vaccination as a strategy to boost anti-tumor immunity or to suppress immunity by including vitamin D3, NF-κB inhibitors or retinoic acid to create tolerogenic DCs. As harvesting DCs from patients and differentiating these cells in vitro is a costly and cumbersome process, in vivo targeting of DCs has huge potential as nanoparticulate platforms equipped with activating or tolerogenic adjuvants can modulate DCs in their natural environment. There is a rapid expansion of the choices of nanoparticles and activation- or tolerance-promoting adjuvants for a therapeutic vaccine platform. In this review we highlight the most recent nanomedical approaches aimed at inducing immune activation or tolerance via targeting DCs, together with novel fundamental insights into the mechanisms inherent to fostering anti-tumor or tolerogenic immunity.
Collapse
Affiliation(s)
- Noémi Anna Nagy
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Aram M. de Haas
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Teunis B. H. Geijtenbeek
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Ronald van Ree
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
- Department of Otorhinolaryngology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Sander W. Tas
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, Amsterdam Rheumatology and Immunology Center, University of Amsterdam, Amsterdam, Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Esther C. de Jong
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
14
|
Dendritic Cell Tumor Vaccination via Fc Gamma Receptor Targeting: Lessons Learned from Pre-Clinical and Translational Studies. Vaccines (Basel) 2021; 9:vaccines9040409. [PMID: 33924183 PMCID: PMC8074394 DOI: 10.3390/vaccines9040409] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023] Open
Abstract
Despite significant recent improvements in the field of immunotherapy, cancer remains a heavy burden on patients and healthcare systems. In recent years, immunotherapies have led to remarkable strides in treating certain cancers. However, despite the success of checkpoint inhibitors and the advent of cellular therapies, novel strategies need to be explored to (1) improve treatment in patients where these approaches fail and (2) make such treatments widely and financially accessible. Vaccines based on tumor antigens (Ag) have emerged as an innovative strategy with the potential to address these areas. Here, we review the fundamental aspects relevant for the development of cancer vaccines and the critical role of dendritic cells (DCs) in this process. We first offer a general overview of DC biology and routes of Ag presentation eliciting effective T cell-mediated immune responses. We then present new therapeutic avenues specifically targeting Fc gamma receptors (FcγR) as a means to deliver antigen selectively to DCs and its effects on T-cell activation. We present an overview of the mechanistic aspects of FcγR-mediated DC targeting, as well as potential tumor vaccination strategies based on preclinical and translational studies. In particular, we highlight recent developments in the field of recombinant immune complex-like large molecules and their potential for DC-mediated tumor vaccination in the clinic. These findings go beyond cancer research and may be of relevance for other disease areas that could benefit from FcγR-targeted antigen delivery, such as autoimmunity and infectious diseases.
Collapse
|
15
|
Paston SJ, Brentville VA, Symonds P, Durrant LG. Cancer Vaccines, Adjuvants, and Delivery Systems. Front Immunol 2021; 12:627932. [PMID: 33859638 PMCID: PMC8042385 DOI: 10.3389/fimmu.2021.627932] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/12/2021] [Indexed: 12/11/2022] Open
Abstract
Vaccination was first pioneered in the 18th century by Edward Jenner and eventually led to the development of the smallpox vaccine and subsequently the eradication of smallpox. The impact of vaccination to prevent infectious diseases has been outstanding with many infections being prevented and a significant decrease in mortality worldwide. Cancer vaccines aim to clear active disease instead of aiming to prevent disease, the only exception being the recently approved vaccine that prevents cancers caused by the Human Papillomavirus. The development of therapeutic cancer vaccines has been disappointing with many early cancer vaccines that showed promise in preclinical models often failing to translate into efficacy in the clinic. In this review we provide an overview of the current vaccine platforms, adjuvants and delivery systems that are currently being investigated or have been approved. With the advent of immune checkpoint inhibitors, we also review the potential of these to be used with cancer vaccines to improve efficacy and help to overcome the immune suppressive tumor microenvironment.
Collapse
Affiliation(s)
| | | | - Peter Symonds
- Biodiscovery Institute, Scancell Limited, Nottingham, United Kingdom
| | - Lindy G. Durrant
- Biodiscovery Institute, University of Nottingham, Faculty of Medicine and Health Sciences, Nottingham, United Kingdom
| |
Collapse
|
16
|
Dry Formulation of Virus-Like Particles in Electrospun Nanofibers. Vaccines (Basel) 2021; 9:vaccines9030213. [PMID: 33802376 PMCID: PMC8000389 DOI: 10.3390/vaccines9030213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 11/16/2022] Open
Abstract
Biologics can be combined with liquid polymer materials and electrospun to produce a dry nanofibrous scaffold. Unlike spray-drying and freeze-drying, electrospinning minimizes the physiological stress on sensitive materials, and nanofiber mat properties such as hydrophobicity, solubility, and melting temperature can be tuned based on the polymer composition. In this study, we explored the dry formulation of a virus-like particle (VLP) vaccine by electrospinning VLP derived from rabbit hemorrhagic disease virus modified to carry the MHC-I gp100 tumor-associated antigen epitope. VLP were added to a polyvinylpyrrolidone (PVP) solution (15% w/v) followed by electrospinning at 24 kV. Formation of a nanofibrous mat was confirmed by scanning electron microscopy, and the presence of VLP was confirmed by transmission electron microscopy and Western blot. VLP from the nanofibers induced T-cell activation and interferon- (IFN-) γ production in vitro. To confirm in vivo cytotoxicity, Pmel mice treated by injection with gp100 VLP from nanofibers induced a gp100 specific immune response, lysing approximately 65% of gp100-pulsed target cells, comparable to mice vaccinated with gp100 VLP in PBS. VLP from nanofibers also induced an antibody response. This work shows that electrospinning can be used to dry-formulate VLP, preserving both humoral and cell-mediated immunity.
Collapse
|
17
|
Cuzzubbo S, Mangsbo S, Nagarajan D, Habra K, Pockley AG, McArdle SEB. Cancer Vaccines: Adjuvant Potency, Importance of Age, Lifestyle, and Treatments. Front Immunol 2021; 11:615240. [PMID: 33679703 PMCID: PMC7927599 DOI: 10.3389/fimmu.2020.615240] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022] Open
Abstract
Although the discovery and characterization of multiple tumor antigens have sparked the development of many antigen/derived cancer vaccines, many are poorly immunogenic and thus, lack clinical efficacy. Adjuvants are therefore incorporated into vaccine formulations to trigger strong and long-lasting immune responses. Adjuvants have generally been classified into two categories: those that ‘depot’ antigens (e.g. mineral salts such as aluminum hydroxide, emulsions, liposomes) and those that act as immunostimulants (Toll Like Receptor agonists, saponins, cytokines). In addition, several novel technologies using vector-based delivery of antigens have been used. Unfortunately, the immune system declines with age, a phenomenon known as immunosenescence, and this is characterized by functional changes in both innate and adaptive cellular immunity systems as well as in lymph node architecture. While many of the immune functions decline over time, others paradoxically increase. Indeed, aging is known to be associated with a low level of chronic inflammation—inflamm-aging. Given that the median age of cancer diagnosis is 66 years and that immunotherapeutic interventions such as cancer vaccines are currently given in combination with or after other forms of treatments which themselves have immune-modulating potential such as surgery, chemotherapy and radiotherapy, the choice of adjuvants requires careful consideration in order to achieve the maximum immune response in a compromised environment. In addition, more clinical trials need to be performed to carefully assess how less conventional form of immune adjuvants, such as exercise, diet and psychological care which have all be shown to influence immune responses can be incorporated to improve the efficacy of cancer vaccines. In this review, adjuvants will be discussed with respect to the above-mentioned important elements.
Collapse
Affiliation(s)
- Stefania Cuzzubbo
- Université de Paris, PARCC, INSERM U970, 75015, Paris, France.,Laboratoire de Recherches Biochirurgicales (Fondation Carpentier), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges Pompidou, Paris, France
| | - Sara Mangsbo
- Ultimovacs AB, Uppsala, Sweden.,Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Divya Nagarajan
- Department of Immunology, Genetics and Clinical pathology Rudbeck laboratories, Uppsala University, Uppsala, Sweden
| | - Kinana Habra
- The School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.,The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Alan Graham Pockley
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.,Centre for Health, Ageing and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Stephanie E B McArdle
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom.,Centre for Health, Ageing and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| |
Collapse
|
18
|
Fomsgaard A, Liu MA. The Key Role of Nucleic Acid Vaccines for One Health. Viruses 2021; 13:258. [PMID: 33567520 PMCID: PMC7916035 DOI: 10.3390/v13020258] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 01/07/2023] Open
Abstract
The ongoing SARS-CoV-2 pandemic has highlighted both the importance of One Health, i.e., the interactions and transmission of pathogens between animals and humans, and the potential power of gene-based vaccines, specifically nucleic acid vaccines. This review will highlight key aspects of the development of plasmid DNA Nucleic Acid (NA) vaccines, which have been licensed for several veterinary uses, and tested for a number of human diseases, and will explain how an understanding of their immunological and real-world attributes are important for their efficacy, and how they helped pave the way for mRNA vaccines. The review highlights how combining efforts for vaccine development for both animals and humans is crucial for advancing new technologies and for combatting emerging diseases.
Collapse
Affiliation(s)
- Anders Fomsgaard
- Department of Virology and Microbiological Special Diagnostic, Statens Serum Institut, 5 Artillerivej, DK-2300 Copenhagen, Denmark
| | - Margaret A. Liu
- ProTherImmune, 3656 Happy Valley Road, Lafayette, CA 94549, USA
| |
Collapse
|
19
|
Odales J, Guzman Valle J, Martínez-Cortés F, Manoutcharian K. Immunogenic properties of immunoglobulin superfamily members within complex biological networks. Cell Immunol 2020; 358:104235. [PMID: 33137645 PMCID: PMC7548077 DOI: 10.1016/j.cellimm.2020.104235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/04/2020] [Accepted: 10/04/2020] [Indexed: 12/01/2022]
Abstract
Antibody-based therapies induce CDR-specific T and B cell responses. Idiotype-anti-idiotype network alters immune system memory compartment. Antigenized antibodies are efficient vaccine immunogen.
Antibodies, T cell receptors and major histocompatibility complex molecules are members of the immunoglobulin superfamily and have pivotal roles in the immune system. The fine interrelation between them regulates several immune functions. Here, we describe lesser-known functions ascribed to these molecules in generating and maintaining immune response. Particularly, we outline the contribution of antibody- and T cell receptor-derived complementarity-determining region neoantigens, antigenized antibodies, as well as major histocompatibility complex class I molecules-derived epitopes to the induction of protective/therapeutic immune responses against pathogens and cancer. We discuss findings of our own and other studies describing protective mechanisms, based on immunogenic properties of immunoglobulin superfamily members, and evaluate the perspectives of application of this class of immunogens in molecular vaccines design.
Collapse
Affiliation(s)
- Josué Odales
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México, DF 04510, Mexico
| | - Jesus Guzman Valle
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México, DF 04510, Mexico
| | - Fernando Martínez-Cortés
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México, DF 04510, Mexico
| | - Karen Manoutcharian
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), AP 70228, Ciudad Universitaria, México, DF 04510, Mexico.
| |
Collapse
|
20
|
Charles J, Chaperot L, Hannani D, Bruder Costa J, Templier I, Trabelsi S, Gil H, Moisan A, Persoons V, Hegelhofer H, Schir E, Quesada JL, Mendoza C, Aspord C, Manches O, Coulie PG, Khammari A, Dreno B, Leccia MT, Plumas J. An innovative plasmacytoid dendritic cell line-based cancer vaccine primes and expands antitumor T-cells in melanoma patients in a first-in-human trial. Oncoimmunology 2020; 9:1738812. [PMID: 32313721 PMCID: PMC7153838 DOI: 10.1080/2162402x.2020.1738812] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 01/28/2020] [Accepted: 01/28/2020] [Indexed: 12/11/2022] Open
Abstract
The efficacy of immune checkpoint inhibitors has been shown to depend on preexisting antitumor immunity; thus, their combination with cancer vaccines is an attractive therapeutic approach. Plasmacytoid dendritic cells (PDC) are strong inducers of antitumor responses and represent promising vaccine candidates. We developed a cancer vaccine approach based on an allogeneic PDC line that functioned as a very potent antigen-presenting cell in pre-clinical studies. In this phase Ib clinical trial, nine patients with metastatic stage IV melanoma received up to 60 million irradiated PDC line cells loaded with 4 melanoma antigens, injected subcutaneously at weekly intervals. The primary endpoints were safety and tolerability. The vaccine was well tolerated and no serious vaccine-induced side effects were recorded. Strikingly, there was no allogeneic response toward the vaccine, but a significant increase in the frequency of circulating anti-tumor specific T lymphocytes was observed in two patients, accompanied by a switch from a naïve to memory phenotype, thus demonstrating priming of antigen-specific T-cells. Signs of clinical activity were observed, including four stable diseases according to IrRC and vitiligoïd lesions. Four patients were still alive at week 48. We also demonstrate the in vitro enhancement of specific T cell expansion induced by the synergistic combination of peptide-loaded PDC line with anti-PD-1, as compared to peptide-loaded PDC line alone. Taken together, these clinical observations demonstrate the ability of the PDC line based-vaccine to prime and expand antitumor CD8+ responses in cancer patients. Further trials should test the combination of this vaccine with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Julie Charles
- Immunobiology and Immunotherapy of Chronic Diseases, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France.,Dermatology Department, Pôle Pluridisciplinaire de Médecine, CHU Grenoble Alpes, Grenoble, France
| | - Laurence Chaperot
- Immunobiology and Immunotherapy of Chronic Diseases, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France.,R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Dalil Hannani
- Immune checkpoint inhibitors, PDCline Pharma, Grenoble
| | - Juliana Bruder Costa
- Immunobiology and Immunotherapy of Chronic Diseases, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France.,Dermatology Department, Pôle Pluridisciplinaire de Médecine, CHU Grenoble Alpes, Grenoble, France.,R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Isabelle Templier
- Dermatology Department, Pôle Pluridisciplinaire de Médecine, CHU Grenoble Alpes, Grenoble, France
| | - Sabiha Trabelsi
- Dermatology Department, Pôle Pluridisciplinaire de Médecine, CHU Grenoble Alpes, Grenoble, France
| | - Hugo Gil
- Pathology Department, Institut de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble, France
| | - Anaick Moisan
- Immunobiology and Immunotherapy of Chronic Diseases, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France.,Cell Therapy and Engineering Unit, Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint Ismier, France
| | - Virginie Persoons
- Cell Therapy and Engineering Unit, Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint Ismier, France
| | - Harald Hegelhofer
- Cell Therapy and Engineering Unit, Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint Ismier, France
| | - Edith Schir
- Délégation à la Recherche Clinique et à l'Innovation, CHU Grenoble Alpes, Grenoble, France
| | | | | | - Caroline Aspord
- Immunobiology and Immunotherapy of Chronic Diseases, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France.,R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Olivier Manches
- Immunobiology and Immunotherapy of Chronic Diseases, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France.,R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Pierre G Coulie
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Amir Khammari
- Onco-dermatology Department, CHU Nantes, CIC 1413, CRCINA, Nantes University, Nantes, France
| | - Brigitte Dreno
- Onco-dermatology Department, CHU Nantes, CIC 1413, CRCINA, Nantes University, Nantes, France
| | - Marie-Thérèse Leccia
- Immunobiology and Immunotherapy of Chronic Diseases, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France.,Dermatology Department, Pôle Pluridisciplinaire de Médecine, CHU Grenoble Alpes, Grenoble, France
| | - Joel Plumas
- Immunobiology and Immunotherapy of Chronic Diseases, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France.,R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France.,Immune checkpoint inhibitors, PDCline Pharma, Grenoble
| |
Collapse
|
21
|
Xu Z, Wise MC, Chokkalingam N, Walker S, Tello‐Ruiz E, Elliott STC, Perales‐Puchalt A, Xiao P, Zhu X, Pumroy RA, Fisher PD, Schultheis K, Schade E, Menis S, Guzman S, Andersen H, Broderick KE, Humeau LM, Muthumani K, Moiseenkova‐Bell V, Schief WR, Weiner DB, Kulp DW. In Vivo Assembly of Nanoparticles Achieved through Synergy of Structure-Based Protein Engineering and Synthetic DNA Generates Enhanced Adaptive Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902802. [PMID: 32328416 PMCID: PMC7175333 DOI: 10.1002/advs.201902802] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/28/2019] [Indexed: 05/25/2023]
Abstract
Nanotechnologies are considered to be of growing importance to the vaccine field. Through decoration of immunogens on multivalent nanoparticles, designed nanovaccines can elicit improved humoral immunity. However, significant practical and monetary challenges in large-scale production of nanovaccines have impeded their widespread clinical translation. Here, an alternative approach is illustrated integrating computational protein modeling and adaptive electroporation-mediated synthetic DNA delivery, thus enabling direct in vivo production of nanovaccines. DNA-launched nanoparticles are demonstrated displaying an HIV immunogen spontaneously self-assembled in vivo. DNA-launched nanovaccines induce stronger humoral responses than their monomeric counterparts in both mice and guinea pigs, and uniquely elicit CD8+ effector T-cell immunity as compared to recombinant protein nanovaccines. Improvements in vaccine responses recapitulate when DNA-launched nanovaccines with alternative scaffolds and decorated antigen are designed and evaluated. Finally, evaluation of functional immune responses induced by DLnanovaccines demonstrates that, in comparison to control mice or mice immunized with DNA-encoded hemagglutinin monomer, mice immunized with a DNA-launched hemagglutinin nanoparticle vaccine fully survive a lethal influenza challenge, and have substantially lower viral load, weight loss, and influenza-induced lung pathology. Additional study of these next-generation in vivo-produced nanovaccines may offer advantages for immunization against multiple disease targets.
Collapse
Affiliation(s)
- Ziyang Xu
- The Vaccine and Immunotherapy CenterThe Wistar InstitutePhiladelphiaPA19104USA
- Department of PharmacologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Megan C. Wise
- Inovio PharmaceuticalsPlymouth MeetingPhiladelphiaPA19422USA
| | - Neethu Chokkalingam
- The Vaccine and Immunotherapy CenterThe Wistar InstitutePhiladelphiaPA19104USA
| | - Susanne Walker
- The Vaccine and Immunotherapy CenterThe Wistar InstitutePhiladelphiaPA19104USA
| | - Edgar Tello‐Ruiz
- The Vaccine and Immunotherapy CenterThe Wistar InstitutePhiladelphiaPA19104USA
| | - Sarah T. C. Elliott
- The Vaccine and Immunotherapy CenterThe Wistar InstitutePhiladelphiaPA19104USA
| | | | - Peng Xiao
- The Vaccine and Immunotherapy CenterThe Wistar InstitutePhiladelphiaPA19104USA
| | - Xizhou Zhu
- The Vaccine and Immunotherapy CenterThe Wistar InstitutePhiladelphiaPA19104USA
| | - Ruth A. Pumroy
- Department of PharmacologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Paul D. Fisher
- Inovio PharmaceuticalsPlymouth MeetingPhiladelphiaPA19422USA
| | | | - Eric Schade
- Inovio PharmaceuticalsPlymouth MeetingPhiladelphiaPA19422USA
| | - Sergey Menis
- Department of Immunology and MicrobiologyThe Scripps Research InstituteLa JollaCA92037USA
- IAVI Neutralizing Antibody CenterThe Scripps Research InstituteLa JollaCA92037USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen DiscoveryThe Scripps Research InstituteLa JollaCA92037USA
| | - Stacy Guzman
- The Vaccine and Immunotherapy CenterThe Wistar InstitutePhiladelphiaPA19104USA
| | | | | | | | - Kar Muthumani
- The Vaccine and Immunotherapy CenterThe Wistar InstitutePhiladelphiaPA19104USA
| | - Vera Moiseenkova‐Bell
- Department of PharmacologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - William R. Schief
- Department of Immunology and MicrobiologyThe Scripps Research InstituteLa JollaCA92037USA
- IAVI Neutralizing Antibody CenterThe Scripps Research InstituteLa JollaCA92037USA
- Center for HIV/AIDS Vaccine Immunology and Immunogen DiscoveryThe Scripps Research InstituteLa JollaCA92037USA
- Ragon Institute of MGHMIT and HarvardCambridgeMA02139USA
| | - David B. Weiner
- The Vaccine and Immunotherapy CenterThe Wistar InstitutePhiladelphiaPA19104USA
| | - Daniel W. Kulp
- The Vaccine and Immunotherapy CenterThe Wistar InstitutePhiladelphiaPA19104USA
- Department of MicrobiologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| |
Collapse
|
22
|
Jacobs L, De Smidt E, Geukens N, Declerck P, Hollevoet K. DNA-Based Delivery of Checkpoint Inhibitors in Muscle and Tumor Enables Long-Term Responses with Distinct Exposure. Mol Ther 2020; 28:1068-1077. [PMID: 32101701 DOI: 10.1016/j.ymthe.2020.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 02/08/2020] [Indexed: 12/17/2022] Open
Abstract
Checkpoint-inhibiting antibodies elicit impressive clinical responses, but still face several issues. The current study evaluated whether DNA-based delivery can broaden the application of checkpoint inhibitors, specifically by pursuing cost-efficient in vivo production, facilitating combination therapies, and exploring administration routes that lower immune-related toxicity risks. We therefore optimized plasmid-encoded anti-CTLA-4 and anti-PD-1 antibodies, and studied their pharmacokinetics and pharmacodynamics when delivered alone and in combination via intramuscular or intratumoral electroporation in mice. Intramuscular electrotransfer of these DNA-based antibodies induced complete regressions in a subcutaneous MC38 tumor model, with plasma concentrations up to 4 and 14 μg/mL for anti-CTLA-4 and anti-PD-1 antibodies, respectively, and antibody detection for at least 6 months. Intratumoral antibody gene electrotransfer gave similar anti-tumor responses as the intramuscular approach. Antibody plasma levels, however, were up to 70-fold lower and substantially more transient, potentially improving biosafety of the expressed checkpoint inhibitors. Intratumoral delivery also generated a systemic anti-tumor response, illustrated by moderate abscopal effects and prolonged protection of cured mice against a tumor rechallenge. In conclusion, intramuscular and intratumoral DNA-based delivery of checkpoint inhibitors both enabled long-term anti-tumor responses despite distinct systemic antibody exposure, highlighting the potential of the tumor as delivery site for DNA-based therapeutics.
Collapse
Affiliation(s)
- Liesl Jacobs
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, Leuven, Belgium
| | - Elien De Smidt
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, Leuven, Belgium; PharmAbs - The KU Leuven Antibody Center, KU Leuven - University of Leuven, Leuven, Belgium
| | - Nick Geukens
- PharmAbs - The KU Leuven Antibody Center, KU Leuven - University of Leuven, Leuven, Belgium
| | - Paul Declerck
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, Leuven, Belgium; PharmAbs - The KU Leuven Antibody Center, KU Leuven - University of Leuven, Leuven, Belgium.
| | - Kevin Hollevoet
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, Leuven, Belgium.
| |
Collapse
|
23
|
Tan Z, Chiu MS, Yan CW, Wong YC, Huang H, Man K, Chen Z. Antimesothelioma Immunotherapy by CTLA-4 Blockade Depends on Active PD1-Based TWIST1 Vaccination. MOLECULAR THERAPY-ONCOLYTICS 2020; 16:302-317. [PMID: 32195318 PMCID: PMC7068049 DOI: 10.1016/j.omto.2020.01.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023]
Abstract
Checkpoint immunotherapy is a major breakthrough for cancer treatment, yet its efficacy is often limited against many types of malignancies, including malignant mesothelioma. Considering that the immunotherapeutic efficacy depends on immunosurveillance, we sought to develop an active immunization method to break immune tolerance to tumor self-antigen. Here, we demonstrated that TWIST1, the basic helix-loop-helix transcription factor, was associated with human mesothelioma tumorigenesis and required for the invasion and metastasis of mesothelioma in the immune-competent murine AB1 model. When conventional TWIST1 vaccines were not effective in vivo, programmed cell death protein 1 (PD1)-based vaccination provided prophylactic control by inducing long-lasting TWIST1-specific T cell responses against both subcutaneous and metastatic mesothelioma lethal challenges. Furthermore, while CTLA-4 blockade alone did not show any immunotherapeutic efficacy against established mesothelioma, its combination with PD1-based vaccination resulted in 60% complete remission. Mechanistically, these functional T cells recognized a novel highly conserved immunodominant TWIST1 epitope, exhibited cytotoxic activity and long-term memory, and led to durable tumor regression and survival benefit against established AB1 mesothelioma and 4T1 breast cancer. We concluded that PD1-based vaccination controls mesothelioma by breaking immune tolerance to the tumor self-antigen TWIST1. Our results warrant clinical development of the PD1-based vaccination to enhance immunotherapy against a wide range of TWIST1-expressing tumors.
Collapse
Affiliation(s)
- Zhiwu Tan
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China
| | - Mei Sum Chiu
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China
| | - Chi Wing Yan
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China
| | - Yik Chun Wong
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China
| | - Haode Huang
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China
| | - Kwan Man
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China
| | - Zhiwei Chen
- AIDS Institute and Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, PR China.,State Key Laboratory for Emerging Infectious Diseases, The University of Hong Kong, Hong Kong SAR, PR China
| |
Collapse
|
24
|
Wages NA, Slingluff CL, Bullock TN, Petroni GR. Tailoring early-phase clinical trial design to address multiple research objectives. Cancer Immunol Immunother 2020; 69:95-102. [PMID: 31807879 PMCID: PMC6952569 DOI: 10.1007/s00262-019-02442-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 11/27/2019] [Indexed: 12/22/2022]
Abstract
INTRODUCTION In contemporary oncology drug development, implementation of novel early-phase designs with the ability to address multiple research objectives is needed to better refine regimens. This paper describes an adaptive design strategy for identifying a range of optimal regimens based on two endpoints within multiple cohorts. The proposed design was developed to address objectives in an early-phase trial of cancer vaccines in combination with agonistic antibodies to CD40 and CD27. MATERIALS AND METHODS We describe a model-based design strategy that was developed for a trial evaluating the safety and immunogenicity of vaccination with (1) peptides plus CD40 antibody and TLR3 ligand, (2) systemic administration of an agonistic CD27 antibody, and (3) to assess immune response from (1) and (2) compared to optimal controls in participants with stage IIB-IV melanoma. RESULTS AND CONCLUSIONS The proposed design is a practical adaptive method for use with combined immunotherapy regimens with multiple objectives within multiple cohorts of interest. Further advances in the effectiveness of cancer immunotherapies will require new approaches that include redefining optimal strategies to take multiple regimens forward into later phases, incorporating additional endpoints in the dose selection process and testing drug combination therapies to improve efficacy and reduce toxicity. Our goal is to facilitate the acceptance and application of more novel designs in contemporary early development trials.
Collapse
Affiliation(s)
- Nolan A Wages
- Division of Translational Research and Applied Statistics, Department of Public Health Sciences, University of Virginia, P.O. Box 800717, Charlottesville, VA, USA.
| | - Craig L Slingluff
- Division of Surgical Oncology, Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Timothy N Bullock
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Gina R Petroni
- Division of Translational Research and Applied Statistics, Department of Public Health Sciences, University of Virginia, P.O. Box 800717, Charlottesville, VA, USA
| |
Collapse
|
25
|
Abstract
The incidence of melanoma continues to increase even as advances in immunotherapy have led to survival benefits in advanced stages. Vaccines are capable of inducing strong, antitumor immune responses with limited toxicity. Some vaccines have demonstrated clinical benefit in clinical trials alone; however, others have not despite inducing strong immune responses. Recent advancements have improved vaccine design, and combining vaccines with other immunotherapies offers promise. This review highlights the underlying principles of vaccine development, common components of vaccines, and the remaining challenges and future directions of vaccine therapy in melanoma.
Collapse
Affiliation(s)
- Minyoung Kwak
- Division of Surgical Oncology, Department of Surgery, University of Virginia, PO Box 800709, Charlottesville, VA 22908-0709, USA; Department of Surgery, SUNY Downstate, Brooklyn, NY, USA; Carter Immunology Center, University of Virginia, Charlottesville, VA, USA
| | - Katie M Leick
- Division of Surgical Oncology, Department of Surgery, University of Virginia, PO Box 800709, Charlottesville, VA 22908-0709, USA; Carter Immunology Center, University of Virginia, Charlottesville, VA, USA; Department of Surgery, University of Iowa, Iowa City, IA, USA
| | - Marit M Melssen
- Division of Surgical Oncology, Department of Surgery, University of Virginia, PO Box 800709, Charlottesville, VA 22908-0709, USA; Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA; Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Craig L Slingluff
- Division of Surgical Oncology, Department of Surgery, University of Virginia, PO Box 800709, Charlottesville, VA 22908-0709, USA; Carter Immunology Center, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
26
|
Sun X, Zeng L, Huang Y. Transcutaneous delivery of DNA/mRNA for cancer therapeutic vaccination. J Gene Med 2019; 21:e3089. [PMID: 30958606 DOI: 10.1002/jgm.3089] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/17/2019] [Accepted: 03/22/2019] [Indexed: 12/11/2022] Open
Abstract
Therapeutic vaccination is a promising strategy for the immunotherapy of cancers. It eradicates cancer cells by evoking and strengthening the patient's own immune system. Because of the easy access and sophisticated immune networks, the skin becomes an ideal target organ for vaccination. Genetic vaccines have been widely investigated, with the advantages of the delivery of multiple antigens and a lower cost for production compared to protein/peptide vaccines. This review summarizes the advances made with respect to the transcutaneous delivery of DNA/mRNA for cancer therapeutic vaccination and also gives a brief description of the immunological milieu of the skin and the importance of dendritic cell-targeting in vaccine delivery, as well as the technologies that aim to facilitate antigen delivery and modulate antigen-presenting cells, thus improving cellular responses. The applications of genetic vaccines encoding tumor antigens delivered through the skin route, both in preclinical and clinical trials, are outlined.
Collapse
Affiliation(s)
- Xiaoyi Sun
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Linghui Zeng
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
27
|
Liu MA. A Comparison of Plasmid DNA and mRNA as Vaccine Technologies. Vaccines (Basel) 2019; 7:E37. [PMID: 31022829 PMCID: PMC6631684 DOI: 10.3390/vaccines7020037] [Citation(s) in RCA: 268] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/19/2019] [Accepted: 04/20/2019] [Indexed: 12/13/2022] Open
Abstract
This review provides a comparison of the theoretical issues and experimental findings for plasmid DNA and mRNA vaccine technologies. While both have been under development since the 1990s, in recent years, significant excitement has turned to mRNA despite the licensure of several veterinary DNA vaccines. Both have required efforts to increase their potency either via manipulating the plasmid DNA and the mRNA directly or through the addition of adjuvants or immunomodulators as well as delivery systems and formulations. The greater inherent inflammatory nature of the mRNA vaccines is discussed for both its potential immunological utility for vaccines and for the potential toxicity. The status of the clinical trials of mRNA vaccines is described along with a comparison to DNA vaccines, specifically the immunogenicity of both licensed veterinary DNA vaccines and select DNA vaccine candidates in human clinical trials.
Collapse
Affiliation(s)
- Margaret A Liu
- ProTherImmune, 3656 Happy Valley Road, Lafayette, CA 94549, USA.
| |
Collapse
|
28
|
Affiliation(s)
- Xuedan He
- University at Buffalo; State University of New York; Buffalo NY 14260 USA
| | - Scott I. Abrams
- Roswell Park Comprehensive Cancer Center; Department of Immunology; Buffalo NY 14263 USA
| | - Jonathan F. Lovell
- University at Buffalo; State University of New York; Buffalo NY 14260 USA
| |
Collapse
|
29
|
Neutralizing Anti-Hemagglutinin Monoclonal Antibodies Induced by Gene-Based Transfer Have Prophylactic and Therapeutic Effects on Influenza Virus Infection. Vaccines (Basel) 2018; 6:vaccines6030035. [PMID: 29949942 PMCID: PMC6161145 DOI: 10.3390/vaccines6030035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 06/23/2018] [Accepted: 06/25/2018] [Indexed: 12/19/2022] Open
Abstract
Hemagglutinin (HA) of influenza virus is a major target for vaccines. HA initiates the internalization of the virus into the host cell by binding to host sialic acid receptors; therefore, inhibition of HA can significantly prevent influenza virus infection. However, the high diversity of HA permits the influenza virus to escape from host immunity. Moreover, the vaccine efficacy is poor in some high-risk populations (e.g., elderly or immunocompromised patients). Passive immunization with anti-HA monoclonal antibodies (mAbs) is an attractive therapy; however, this method has high production costs and requires repeated inoculations. To address these issues, several methods for long-term expression of mAb against influenza virus have been developed. Here, we provide an overview of methods using plasmid and viral adeno-associated virus (AAV) vectors that have been modified for higher expression of neutralizing antibodies in the host. We also examine two methods of injection, electro-transfer and hydrodynamic injection. Our results show that antibody gene transfer is effective against influenza virus infection even in immunocompromised mice, and antibody expression was detected in the serum and upper respiratory tract. We also demonstrate this method to be effective following influenza virus infection. Finally, we discuss the perspective of passive immunization with antibody gene transfer for future clinical trials.
Collapse
|
30
|
Cook KW, Durrant LG, Brentville VA. Current Strategies to Enhance Anti-Tumour Immunity. Biomedicines 2018; 6:E37. [PMID: 29570634 PMCID: PMC6027499 DOI: 10.3390/biomedicines6020037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 03/19/2018] [Accepted: 03/21/2018] [Indexed: 12/15/2022] Open
Abstract
The interaction of the immune system with cancer is complex, but new approaches are resulting in exciting therapeutic benefits. In order to enhance the immune response to cancer, immune therapies seek to either induce high avidity immune responses to tumour specific antigens or to convert the tumour to a more pro-inflammatory microenvironment. Strategies, including vaccination, oncolytic viruses, and adoptive cell transfer all seek to induce anti-tumour immunity. To overcome the suppressive tumour microenvironment checkpoint inhibitors and modulators of regulatory cell populations have been investigated. This review summarizes the recent advances in immune therapies and discusses the importance of combination therapies in the treatment of cancers.
Collapse
Affiliation(s)
- Katherine W Cook
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottinghamshire NG5 1PB, UK.
| | - Lindy G Durrant
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottinghamshire NG5 1PB, UK.
- Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, City Hospital Campus, Nottinghamshire NG5 1PB, UK.
| | - Victoria A Brentville
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottinghamshire NG5 1PB, UK.
| |
Collapse
|