1
|
Pierini S, Tanyi JL, Simpkins F, George E, Uribe-Herranz M, Drapkin R, Burger R, Morgan MA, Facciabene A. Ovarian granulosa cell tumor characterization identifies FOXL2 as an immunotherapeutic target. JCI Insight 2020; 5:136773. [PMID: 32814714 PMCID: PMC7455139 DOI: 10.1172/jci.insight.136773] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022] Open
Abstract
Granulosa cell tumors (GCT) are rare ovarian malignancies. Due to the lack of effective treatment in late relapse, there is a clear unmet need for novel therapies. Forkhead Box L2 (FOXL2) is a protein mainly expressed in granulosa cells (GC) and therefore is a rational therapeutic target. Since we identified tumor infiltrating lymphocytes (TILs) as the main immune population within GCT, TILs from 11 GCT patients were expanded, and their phenotypes were interrogated to determine that T cells acquired late antigen-experienced phenotypes and lower levels of PD1 expression. Importantly, TILs maintained their functionality after ex vivo expansion as they vigorously reacted against autologous tumors (100% of patients) and against FOXL2 peptides (57.1% of patients). To validate the relevance of FOXL2 as a target for immune therapy, we developed a plasmid DNA vaccine (FoxL2–tetanus toxin; FoxL2-TT) by fusing Foxl2 cDNA with the immune-enhancing domain of TT. Mice immunization with FoxL2-TT controlled growth of FOXL2-expressing ovarian (BR5) and breast (4T1) cancers in a T cell–mediated manner. Combination of anti–PD-L1 with FoxL2-TT vaccination further reduced tumor progression and improved mouse survival without affecting the female reproductive system and pregnancy. Together, our results suggest that FOXL2 immune targeting can produce substantial long-term clinical benefits. Our study can serve as a foundation for trials testing immunotherapeutic approaches in patients with ovarian GCT. FOXL2 may serve as a immunotherapeutic target for tumor infiltrating lymphocytes in ovarian granulosa cell tumors.
Collapse
Affiliation(s)
- Stefano Pierini
- Department of Radiation Oncology and.,Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Janos L Tanyi
- Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fiona Simpkins
- Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Erin George
- Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mireia Uribe-Herranz
- Department of Radiation Oncology and.,Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ronny Drapkin
- Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert Burger
- Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mark A Morgan
- Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andrea Facciabene
- Department of Radiation Oncology and.,Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2
|
Savelyeva N, Allen A, Chotprakaikiat W, Harden E, Jobsri J, Godeseth R, Wang Y, Stevenson F, Ottensmeier C. Linked CD4 T Cell Help: Broadening Immune Attack Against Cancer by Vaccination. Curr Top Microbiol Immunol 2019; 405:123-143. [PMID: 27704269 DOI: 10.1007/82_2016_500] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the last decade, immunotherapy with monoclonal antibodies targeting immunological check points has become a breakthrough therapeutic modality for solid cancers. However, only up to 50 % of patients benefit from this powerful approach. For others vaccination might provide a plausible addition or alternative. For induction of effective anticancer immunity CD4+ T cell help is required, which is often difficult to induce to self cancer targets because of tolerogenic mechanisms. Our approach for cancer vaccines has been to incorporate into the vaccine design sequences able to activate foreign T cell help, through genetically linking cancer targets to microbial sequences (King et al. in Nat Med 4(11):1281-1286, 1998; Savelyeva et al. in Nat Biotechnol 19(8):760-764, 2001). This harnesses the non-tolerized CD4 T cell repertoire available in patients to help induction of effective immunity against fused cancer antigens. Multiple immune effector mechanisms including antibody, CD8+ T cells as well as CD4 effector T cells can be activated using this strategy. Delivery via DNA vaccines has already indicated clinical efficacy. The same principle of linked T cell help has now been transferred to other novel vaccine modalities to further potentiate immunity against cancer targets.
Collapse
Affiliation(s)
- Natalia Savelyeva
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK.
| | - Alex Allen
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| | - Warayut Chotprakaikiat
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
- Oral Biology Department, Naresuan University, Phitsanulok, Thailand
| | - Elena Harden
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| | - Jantipa Jobsri
- Oral Biology Department, Naresuan University, Phitsanulok, Thailand
| | - Rosemary Godeseth
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| | - Yidao Wang
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| | - Freda Stevenson
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| | - Christian Ottensmeier
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Tremona Road, Southampton, SO16 6YD, UK
| |
Collapse
|
3
|
Nagaraj S, Reddy PN, Ramlal S, Paul S, Peddayelachagiri B, Parida DM. A novel tandem repeat cloning technique for creation of multiple short peptide repeats to differentiate closely related antigens. J Immunol Methods 2019; 469:11-17. [PMID: 30660621 DOI: 10.1016/j.jim.2019.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 12/11/2018] [Accepted: 01/16/2019] [Indexed: 11/17/2022]
Abstract
Antibody cross-reactivity is a problem often associated with closely related antigens. This study was aimed to develop a method enabling differentiation of closely related toxins based on antigen designing strategy. The method involves identification of disparate amino acids (AA) confined to target antigen in comparison with two or more closely related antigens, their assembly into a DNA oligomer and further cloning as six tandem repeats (TR) using restriction and ligation strategy into a desired vector and finally generation of antigen specific antibodies. The practical utility of this method was demonstrated by generating and testing the specificity of polyclonal antibodies against staphylococcal enterotoxin C (SEC). Cross-reactivity is a problem often associated with SEC in immunoassays due to its amino acid sequence identity with staphylococcal enterotoxin B (SEB) (40-60%). To circumvent the same, the above-mentioned strategy was applied. Unique AA of SEC (36 AA) in comparison to SEB were selected, reassembled and with deduced corresponding nucleotides, an oligomer of 117 bases was designed. Using primers with restriction overhangs, three constructs were created each with two repeats using a common restriction site. The resulting three constructs were sequentially cloned into alternating restriction sites of pRSET A vector in directional orientation, expressed in E. coli for rTR/SEC protein which was used to generate specific polyclonal antibodies against SEC. Specificity was compared with antibody raised against whole SEC recombinant protein using Western blot and dot blot assays. High specificity was achieved through the developed strategy signifying its possible application to address cross-reactivity problem associated with closely related antigens.
Collapse
Affiliation(s)
- Sowmya Nagaraj
- Microbiology Division, Defence Food Research Laboratory, Siddarthanagar, Mysore, Karnataka 570011, India
| | - Prakash Narayana Reddy
- Department of Biotechnology, Vignan's Foundation for Science, Technology and Research (Deemed to be University), Vadlamudi, Guntur District, Andhra Pradesh 522 213, India
| | - Shylaja Ramlal
- Microbiology Division, Defence Food Research Laboratory, Siddarthanagar, Mysore, Karnataka 570011, India.
| | - Soumya Paul
- Microbiology Division, Defence Food Research Laboratory, Siddarthanagar, Mysore, Karnataka 570011, India
| | - Bhavani Peddayelachagiri
- Microbiology Division, Defence Food Research Laboratory, Siddarthanagar, Mysore, Karnataka 570011, India
| | - D Manmohan Parida
- Microbiology Division, Defence Food Research Laboratory, Siddarthanagar, Mysore, Karnataka 570011, India
| |
Collapse
|
4
|
|
5
|
Meleshko AN, Petrovskaya NA, Savelyeva N, Vashkevich KP, Doronina SN, Sachivko NV. Phase I clinical trial of idiotypic DNA vaccine administered as a complex with polyethylenimine to patients with B-cell lymphoma. Hum Vaccin Immunother 2017; 13:1-6. [PMID: 28272989 DOI: 10.1080/21645515.2017.1285477] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
We report on the design of a phase I, non-randomized, open-label study of idiotypic DNA vaccination in patients with B-cell non-Hodgkin's lymphoma (ISRCTN31090206). The study uses DNA fusion gene vaccination encoding patient-specific single chain variable fragment, or idiotype, linked to an immunostimulatory sequence. Two types of immunostimulatory sequence are being explored: potato virus X coat protein and human chemokine MIP3α. Linear polyethylenimine with low molecular weight (8 kDa) is used as a synthetic vehicle for vaccine delivery. Humoral and T-cellular immune responses to vaccination will be measured by ELISA and ELISPOT, respectively. The primary study endpoints are safety, tolerability and immunogenicity of DNA-PEI vaccination.
Collapse
Affiliation(s)
- A N Meleshko
- a Belarusian Research Center for Pediatric Oncology, Hematology and Immunology , Minsk , Belarus
| | - N A Petrovskaya
- b N.N. Alexandrov National Cancer Centre of Belarus , Minsk , Belarus
| | - N Savelyeva
- c Cancer Sciences Unit, Faculty of Medicine, University of Southampton , Southampton , UK
| | - K P Vashkevich
- a Belarusian Research Center for Pediatric Oncology, Hematology and Immunology , Minsk , Belarus
| | - S N Doronina
- a Belarusian Research Center for Pediatric Oncology, Hematology and Immunology , Minsk , Belarus
| | - N V Sachivko
- b N.N. Alexandrov National Cancer Centre of Belarus , Minsk , Belarus
| |
Collapse
|
6
|
McCann KJ, Mander A, Cazaly A, Chudley L, Stasakova J, Thirdborough S, King A, Lloyd-Evans P, Buxton E, Edwards C, Halford S, Bateman A, O'Callaghan A, Clive S, Anthoney A, Jodrell DI, Weinschenk T, Simon P, Sahin U, Thomas GJ, Stevenson FK, Ottensmeier CH. Targeting Carcinoembryonic Antigen with DNA Vaccination: On-Target Adverse Events Link with Immunologic and Clinical Outcomes. Clin Cancer Res 2016; 22:4827-4836. [PMID: 27091407 PMCID: PMC5330406 DOI: 10.1158/1078-0432.ccr-15-2507] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 03/29/2016] [Indexed: 12/22/2022]
Abstract
PURPOSE We have clinically evaluated a DNA fusion vaccine to target the HLA-A*0201-binding peptide CAP-1 from carcinoembryonic antigen (CEA605-613) linked to an immunostimulatory domain (DOM) from fragment C of tetanus toxin. EXPERIMENTAL DESIGN Twenty-seven patients with CEA-expressing carcinomas were recruited: 15 patients with measurable disease (arm-I) and 12 patients without radiological evidence of disease (arm-II). Six intramuscular vaccinations of naked DNA (1 mg/dose) were administered up to week 12. Clinical and immunologic follow-up was up to week 64 or clinical/radiological disease. RESULTS DOM-specific immune responses demonstrated successful vaccine delivery. All patients without measurable disease compared with 60% with advanced disease responded immunologically, while 58% and 20% expanded anti-CAP-1 CD8+ T cells, respectively. CAP-1-specific T cells were only detectable in the blood postvaccination but could also be identified in previously resected cancer tissue. The gastrointestinal adverse event diarrhea was reported by 48% of patients and linked to more frequent decreases in CEA (P < 0.001) and improved global immunologic responses [anti-DOM responses of greater magnitude (P < 0.001), frequency (P = 0.004), and duration] compared with patients without diarrhea. In advanced disease patients, decreases in CEA were associated with better overall survival (HR = 0.14, P = 0.017). CAP-1 peptide was detectable on MHC class I of normal bowel mucosa and primary colorectal cancer tissue by mass spectrometry, offering a mechanistic explanation for diarrhea through CD8+ T-cell attack. CONCLUSIONS Our data suggest that DNA vaccination is able to overcome peripheral tolerance in normal and tumor tissue and warrants testing in combination studies, for example, by vaccinating in parallel to treatment with an anti-PD1 antibody. Clin Cancer Res; 22(19); 4827-36. ©2016 AACR.
Collapse
Affiliation(s)
- Katy J McCann
- Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - Ann Mander
- Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - Angelica Cazaly
- Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - Lindsey Chudley
- Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - Jana Stasakova
- Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - Stephen Thirdborough
- Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - Andrew King
- University Hospital Southampton NHS Trust, Southampton, UK
| | - Paul Lloyd-Evans
- NHS Blood and Transplant, Clinical Biotechnology Centre, University of Bristol, Bristol, UK
| | - Emily Buxton
- Cancer Research UK Centre for Drug Development, London, UK
| | - Ceri Edwards
- Cancer Research UK Centre for Drug Development, London, UK
| | - Sarah Halford
- Cancer Research UK Centre for Drug Development, London, UK
| | - Andrew Bateman
- Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton, Southampton, UK
- University Hospital Southampton NHS Trust, Southampton, UK
| | | | | | | | - Duncan I Jodrell
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Petra Simon
- TRON gGmbH, Translational Oncology at the University Medical Center, Johannes Gutenberg-University, Mainz, Germany
- BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Ugur Sahin
- TRON gGmbH, Translational Oncology at the University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Gareth J Thomas
- Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton, Southampton, UK
- University Hospital Southampton NHS Trust, Southampton, UK
| | - Freda K Stevenson
- Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - Christian H Ottensmeier
- Southampton Experimental Cancer Medicine Centre, Cancer Sciences Unit, University of Southampton, Southampton, UK
- University Hospital Southampton NHS Trust, Southampton, UK
| |
Collapse
|
7
|
Strategies to enhance immunogenicity of cDNA vaccine encoded antigens by modulation of antigen processing. Vaccine 2016; 34:5132-5140. [PMID: 27593157 DOI: 10.1016/j.vaccine.2016.08.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/21/2016] [Accepted: 08/12/2016] [Indexed: 11/20/2022]
Abstract
Most vaccines are based on protective humoral responses while for intracellular pathogens CD8(+) T cells are regularly needed to provide protection. However, poor processing efficiency of antigens is often a limiting factor in CD8(+) T cell priming, hampering vaccine efficacy. The multistage cDNA vaccine H56, encoding three secreted Mycobacterium tuberculosis antigens, was used to test a complete strategy to enhance vaccine' immunogenicity. Potential CD8(+) T cell epitopes in H56 were predicted using the NetMHC3.4/ANN program. Mice were immunized with H56 cDNA using dermal DNA tattoo immunization and epitope candidates were tested for recognition by responding CD8(+) T cells in ex vivo assays. Seven novel CD8(+) T cell epitopes were identified. H56 immunogenicity could be substantially enhanced by two strategies: (i) fusion of the H56 sequence to cDNA of proteins that modify intracellular antigen processing or provide CD4(+) T cell help, (ii) by substitution of the epitope's hydrophobic C-terminal flanking residues for polar glutamic acid, which facilitated their proteasome-mediated generation. We conclude that this whole strategy of in silico prediction of potential CD8(+) T cell epitopes in novel antigens, followed by fusion to sequences with immunogenicity-enhancing properties or modification of epitope flanking sequences to improve proteasome-mediated processing, may be exploited to design novel vaccines against emerging or 'hard to treat' intracellular pathogens.
Collapse
|
8
|
Ugel S, Facciponte JG, De Sanctis F, Facciabene A. Targeting tumor vasculature: expanding the potential of DNA cancer vaccines. Cancer Immunol Immunother 2015; 64:1339-48. [PMID: 26267042 PMCID: PMC11028665 DOI: 10.1007/s00262-015-1747-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 07/28/2015] [Indexed: 01/16/2023]
Abstract
Targeting the tumor vasculature with anti-angiogenesis modalities is a bona fide validated approach that has complemented cancer treatment paradigms. Tumor vasculature antigens (TVA) can be immunologically targeted and offers multiple theoretical advantages that may enhance existing strategies against cancer. We focused on tumor endothelial marker 1 (TEM1/CD248) as a model TVA since it is broadly expressed on many different cancers. Our DNA-based vaccine approach demonstrated that CD248 can be effectively targeted immunologically; anti-tumor responses were generated in several mouse models; and CD8(+)/CD4(+) T cell responses were elicited against peptides derived from CD248 protein. Our work supports our contention that CD248 is a novel immunotherapeutic target for cancer treatment and highlights the efficient, safe and translatable use of DNA-based immunotherapy. We next briefly highlight ongoing investigations targeting CD248 with antibodies as a diagnostic imaging agent and as a therapeutic antibody in an early clinical trial. The optimal approach for generating effective DNA-based cancer vaccines for several tumor types may be a combinatorial approach that enhances immunogenicity such as combination with chemotherapy. Additional combination approaches are discussed and include those that alleviate the immunosuppressive tumor microenvironment induced by myeloid-derived suppressor cells and T regulatory cells. Targeting the tumor vasculature by CD248-based immunological modalities expands the armamentarium against cancer.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Cancer Vaccines/therapeutic use
- Combined Modality Therapy
- Disease Models, Animal
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Humans
- Immunotherapy/methods
- Neoplasms/immunology
- Neoplasms/therapy
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/therapy
- T-Lymphocytes/immunology
- Vaccines, DNA/therapeutic use
Collapse
Affiliation(s)
- Stefano Ugel
- Ovarian Cancer Research Center (OCRC), University of Pennsylvania School of Medicine, Biomedical Research Building II/III, 13th Floor, 421 Curie Blvd., Philadelphia, PA 19104 USA
- Immunology Section, Department of Pathology and Diagnostics, University of Verona, 37134 Verona, Italy
| | - John G. Facciponte
- Ovarian Cancer Research Center (OCRC), University of Pennsylvania School of Medicine, Biomedical Research Building II/III, 13th Floor, 421 Curie Blvd., Philadelphia, PA 19104 USA
| | - Francesco De Sanctis
- Ovarian Cancer Research Center (OCRC), University of Pennsylvania School of Medicine, Biomedical Research Building II/III, 13th Floor, 421 Curie Blvd., Philadelphia, PA 19104 USA
- Immunology Section, Department of Pathology and Diagnostics, University of Verona, 37134 Verona, Italy
| | - Andrea Facciabene
- Ovarian Cancer Research Center (OCRC), University of Pennsylvania School of Medicine, Biomedical Research Building II/III, 13th Floor, 421 Curie Blvd., Philadelphia, PA 19104 USA
| |
Collapse
|
9
|
Ragonnaud E, Holst P. The rationale of vectored gene-fusion vaccines against cancer: evolving strategies and latest evidence. THERAPEUTIC ADVANCES IN VACCINES 2014; 1:33-47. [PMID: 24757514 DOI: 10.1177/2051013613480446] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The development of vaccines that target tumor antigens in cancer has proven difficult. A major reason for this is that T cells specific for tumor self-antigens and neoantigens are eliminated or inactivated through mechanisms of tolerance. Antigen fusion strategies which increase the ability of vaccines to stimulate T cells that have escaped tolerance mechanisms, may have a particular potential as immunotherapies. This review highlights antigen fusion strategies that have been successful in stimulating the induction of T-cell immunity against cancer and counteracting tumor-associated tolerance. In preclinical studies, these strategies have shown to improve the potency of vectored vaccines through fusion of tumor antigen to proteins or protein domains that increase CD4+ T-cell help, CD8+ T-cell responses or both the CD4+ and CD8+ T-cell responses. However, in clinical trials such strategies seem to be less efficient when provided as a DNA vaccine. The first clinical trial using a viral vectored fusion-gene vaccine is expected to be tested as a partner in a heterologous prime-boost regimen directed against cervical cancer.
Collapse
Affiliation(s)
| | - Peter Holst
- ISIM - Center for Medical Parasitology, Copenhagen, Denmark
| |
Collapse
|
10
|
Facciponte JG, Ugel S, De Sanctis F, Li C, Wang L, Nair G, Sehgal S, Raj A, Matthaiou E, Coukos G, Facciabene A. Tumor endothelial marker 1-specific DNA vaccination targets tumor vasculature. J Clin Invest 2014; 124:1497-511. [PMID: 24642465 DOI: 10.1172/jci67382] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 01/16/2014] [Indexed: 12/11/2022] Open
Abstract
Tumor endothelial marker 1 (TEM1; also known as endosialin or CD248) is a protein found on tumor vasculature and in tumor stroma. Here, we tested whether TEM1 has potential as a therapeutic target for cancer immunotherapy by immunizing immunocompetent mice with Tem1 cDNA fused to the minimal domain of the C fragment of tetanus toxoid (referred to herein as Tem1-TT vaccine). Tem1-TT vaccination elicited CD8+ and/or CD4+ T cell responses against immunodominant TEM1 protein sequences. Prophylactic immunization of animals with Tem1-TT prevented or delayed tumor formation in several murine tumor models. Therapeutic vaccination of tumor-bearing mice reduced tumor vascularity, increased infiltration of CD3+ T cells into the tumor, and controlled progression of established tumors. Tem1-TT vaccination also elicited CD8+ cytotoxic T cell responses against murine tumor-specific antigens. Effective Tem1-TT vaccination did not affect angiogenesis-dependent physiological processes, including wound healing and reproduction. Based on these data and the widespread expression of TEM1 on the vasculature of different tumor types, we conclude that targeting TEM1 has therapeutic potential in cancer immunotherapy.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Cell Line, Tumor
- Female
- Humans
- Immune Tolerance
- Immunodominant Epitopes
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Microvessels/immunology
- Microvessels/pathology
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/genetics
- Neoplasm Proteins/immunology
- Neoplasms, Experimental/blood supply
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/therapy
- Pregnancy
- Tetanus Toxoid/genetics
- Tetanus Toxoid/immunology
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Vaccines, DNA/therapeutic use
Collapse
|
11
|
Abstract
The goal of active vaccination is to induce all the immune effector pathways and to establish immunological memory allowing prolonged surveillance against pathogens or cancer cells. DNA vaccination platform is an intriguing strategy owing to its ability to mobilize both branches of the immune system (i.e., innate immunity as well as adaptive immunity). Since plasmids offer several advantages for biotechnological applications due to their modular structure and easy manipulation, a wide range of strategies can be applied to improve DNA vaccine performance. This chapter discusses this topic in detail taking into account antigen/epitope selection and optimization, inclusion of intracellular targeting sequences and genetic adjuvants, and provision of T cell help.
Collapse
|
12
|
Aldrich JF, Shearer MH, Lowe DB, Winn RE, Jumper CA, Kennedy RC, Bright RK. The role of gamma interferon in DNA vaccine-induced tumor immunity targeting simian virus 40 large tumor antigen. Cancer Immunol Immunother 2013; 62:371-82. [PMID: 22926061 PMCID: PMC11028630 DOI: 10.1007/s00262-012-1338-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: 05/02/2012] [Accepted: 08/09/2012] [Indexed: 11/28/2022]
Abstract
The central role of CD4+ T lymphocytes in mediating DNA vaccine-induced tumor immunity against the viral oncoprotein simian virus 40 (SV40) large tumor antigen (Tag) has previously been described by our laboratory. In the present study, we extend our previous findings by examining the roles of IFN-γ and Th1-associated effector cells within the context of DNA immunization in a murine model of pulmonary metastasis. Immunization of BALB/c mice with plasmid DNA encoding SV40 Tag (pCMV-Tag) generated IFN-γ-secreting T lymphocytes that produced this cytokine upon in vitro stimulation with mKSA tumor cells. The role of IFN-γ as a mediator of protection against mKSA tumor development was assessed via in vivo IFN-γ neutralization, and these experiments demonstrated a requirement for this cytokine in the induction immune phase. Neutralization of IFN-γ was associated with a reduction in Th1 cytokine-producing CD4+ and CD8+ splenocytes, as assessed by flow cytometry analysis, and provided further evidence for the role of CD4+ T lymphocytes as drivers of the cellular immune response. Depletion of NK cells and CD8+ T lymphocytes demonstrated the expendability of these cell types individually, but showed a requirement for a resident cytotoxic cell population within the immune effector phase. Our findings demonstrate the importance of IFN-γ in the induction of protective immunity stimulated by pCMV-Tag DNA-based vaccine and help to clarify the general mechanisms by which DNA vaccines trigger immunity to tumor cells.
Collapse
Affiliation(s)
- Joel F. Aldrich
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 6591, Lubbock, TX 79430 USA
| | - Michael H. Shearer
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 6591, Lubbock, TX 79430 USA
| | - Devin B. Lowe
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 6591, Lubbock, TX 79430 USA
- Present Address: Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
| | - Richard E. Winn
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 6591, Lubbock, TX 79430 USA
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430 USA
| | - Cynthia A. Jumper
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 6591, Lubbock, TX 79430 USA
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430 USA
| | - Ronald C. Kennedy
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 6591, Lubbock, TX 79430 USA
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430 USA
| | - Robert K. Bright
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 6591, Lubbock, TX 79430 USA
| |
Collapse
|
13
|
Suppression of breast tumor growth by DNA vaccination against phosphatase of regenerating liver 3. Gene Ther 2013; 20:834-45. [PMID: 23364316 DOI: 10.1038/gt.2013.5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 11/14/2012] [Accepted: 12/31/2012] [Indexed: 12/18/2022]
Abstract
Phosphatase of regenerating liver (PRL)-3 is highly expressed in multiple cancers and has important roles in cancer development. Some small-molecule inhibitors and antibodies targeting PRL-3 have been recently reported to inhibit tumor growth effectively. To determine whether PRL-3-targeted DNA vaccination can induce immune response to prevent or inhibit the tumor growth, we established mouse D2F2 breast cancer cells expressing PRL-3 (D2F2/PRL-3) and control cells (D2F2/NC) with lentivirus, and constructed pVAX1-Igκ-PRL-3 plasmid (named as K-P3) as DNA vaccine to immunize BALB/c mice. We found that the K-P3 vaccine delivered by gene gun significantly prevented the growth of D2F2/PRL-3 compared with pVAX1-vector (P<0.01), but not of D2F2/NC, and improved the survival of D2F2/PRL-3-innoculated mice. Both PRL-3-targeted cytotoxic T lymphocytes (CTLs) and T-helper type 1 cell immune response (production of high levels of interferon-γ and tumor necrosis factor-α) were found to be involved in the preventive effect. Furthermore, PRL-3-targeted DNA immunization inhibited tumor growth of D2F2/PRL-3 cells in mice. We also evaluated the potential of immunization with PRL-3 protein, but no significant therapeutic or preventive effect was obtained on tumor growth. To enhance the immunity of PRL-3, we incorporated different molecular adjuvants, such as Mycobacterium tuberculosis heat-shock protein, CTL antigen 4 and M. tuberculosis T-cell stimulatory epitope (MT), into K-P3 vaccine for expressing the fusion proteins. We found that these adjuvant molecules did not significantly improve the antitumor activity of PRL-3 vaccine, but enhanced the production of PRL-3 antibodies in immunized mice. Summarily, our findings demonstrate that PRL-3-targeted DNA vaccine can generate significantly preventive and therapeutic effects on the growth of breast cancer expressing PRL-3 through the induction of cellular immune responses to PRL-3.
Collapse
|
14
|
The development of gene-based vectors for immunization. Vaccines (Basel) 2013. [PMCID: PMC7151937 DOI: 10.1016/b978-1-4557-0090-5.00064-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
15
|
Oosterhuis K, Aleyd E, Vrijland K, Schumacher TN, Haanen JB. Rational design of DNA vaccines for the induction of human papillomavirus type 16 E6- and E7-specific cytotoxic T-cell responses. Hum Gene Ther 2012; 23:1301-12. [PMID: 22971245 DOI: 10.1089/hum.2012.101] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Many DNA vaccine candidates have been developed for the treatment of human papillomavirus type 16 (HPV16)-induced malignancies. Most of these vaccines consist of a fusion of E7 with a "carrier-protein" that functions to increase the potency of the vaccine. The nature of these carrier-proteins varies widely, and the mechanisms proposed to explain the enhanced immunogenicity of such fusions are often linked to the biological function of the carrier-protein. However, the potentiating effect of these carrier-proteins might also be explained by more general mechanisms, such as the provision of CD4+ T-cell help, increased antigen stability, or altered subcellular localization of the antigen. To assess whether these more generic mechanisms could suffice to generate highly immunogenic DNA vaccines, we evaluated a series of modular HPV16 E7 DNA vaccines in which the presence of CD4+ T-cell help, the presence of an endogenous carrier-protein, and the subcellular localization of the antigen could be systematically altered. Using this approach, we demonstrate that the addition of an element that provides CD4+ T-cell help, elements that enforce endoplasmic reticulum (ER) localization/retention are both necessary and sufficient to create markedly effective HPV16 E7-directed DNA vaccines. Importantly, the resulting design rules also apply to an HPV16 E6-directed DNA vaccine. The developed "HELP(ER)" HPV DNA vaccines encode only very limited additional sequences besides the antigen, thereby reducing the risk of antigenic competition and/or autoimmunity.
Collapse
Affiliation(s)
- Koen Oosterhuis
- Division of Immunology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
16
|
Hou H, Zhang Z, Zhao W, Hou J. Generating DNA sequences encoding tandem peptide repeats suitable for expression and immunological application. World J Microbiol Biotechnol 2012; 28:2175-80. [PMID: 22806040 DOI: 10.1007/s11274-012-1023-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 02/03/2012] [Indexed: 11/24/2022]
Abstract
Tandem repeats of single short peptide sequences are useful for many purposes. Here we describe a method called ligation-PCR to construct DNA sequences encoding numerous tandem peptide repeats that can stably produce such repeats in both prokaryotic and eukaryotic cells. The method employs double-strand target monomers consisting of a short peptide coding sequences. These sequences contain 3-bp cohesive overhangs to ensure correct repeat orientation and reading frame during ligation. The ligation products are PCR amplified and directly cloned into a new TA-cloning vector, pZeroT. Constructs containing tandem 10-amino-acid myc-tag peptide coding sequence repeats that ranged from approximately 0.45-1.2 kb, representing 15-40 copies of the corresponding peptide, were successfully obtained by this method. When one of the constructs was subcloned into prokaryotic vector pET-28 c (+) and eukaryotic vector rGHpcDNA3.0, and introduced into E. Coli and COS-7 cells, respectively, proteins containing tandem myc-tag peptide repeats were expressed with expected molecular weights. Purified proteins from E. Coli could successfully stimulate a peptide specific immune response. This method provides a means to manipulate peptides at the nucleic acid level, and can serve as the basis for biological peptide synthesis, epitope-specific antibody production, and epitope-based DNA vaccine construction.
Collapse
Affiliation(s)
- Hongwei Hou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cell Biology, Peking University Cancer Hospital and Institute, 52 Fucheng Road, Beijing 100142, People's Republic of China
| | | | | | | |
Collapse
|
17
|
Wei Y, Sun Y, Song C, Li H, Li Y, Zhang K, Gong J, Liu F, Liu Z, August JT, Jin B, Yang K. Enhancement of DNA vaccine efficacy by targeting the xenogeneic human chorionic gonadotropin, survivin and vascular endothelial growth factor receptor 2 combined tumor antigen to the major histocompatibility complex class II pathway. J Gene Med 2012; 14:353-62. [PMID: 22438278 DOI: 10.1002/jgm.2624] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND A number of strategies have been used to improve the efficacy of the DNA vaccine for the treatment of tumors. These strategies, ranging from activating CD4+ T cell, manipulating antigen presentation and/or processing to anti-angiogenesis, focus on one certain aspect in the functioning of the vaccine. Therefore, their combination is necessary for rational DNA vaccines design by synergizing different regimens and overcoming the limitations of each strategy. METHODS A DNA fragment (HSV) encoding the C terminal 37 amino acids of human chorionic gonadotropin β chain (hCGβ), 5 different HLA-restricted cytotoxic T lymphocyte epitopes from human survivin and the third and fourth extracellular domains of vascular endothelial growth factor receptor 2 (VEGFR2) was inserted into the sequence between the luminal and transmembrane domain of human lysosome-associated membrane protein-1 cDNA for the construction of a novel DNA vaccine. RESULTS This novel vaccine, named p-L/HSV, has a potent antitumor effect on the LL/2 lung carcinoma model in syngeneic C57BL/6 mice. The immunologic mechanism involved in the antitumor effect referred to the activation of both cellular and humoral immune response. In addition, the tumor vasculature was abrogated as observed by immunohistochemistry in p-L/HSV immunized mice. Furthermore, the immunized mice received an additional boost with p-L/HSV 6 months later and showed a strong immune recall response. CONCLUSIONS The present study indicates that the strategies of combining antitumor with antiangiogenesis and targeting the tumor antigen to the major histocompatibility complex class II pathway cooperate well. Such a study may shed new light on designing vaccine for cancer in the future.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- CD8-Positive T-Lymphocytes
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/therapy
- Chorionic Gonadotropin, beta Subunit, Human/genetics
- Epitopes/genetics
- Female
- Genetic Vectors/administration & dosage
- HEK293 Cells
- Humans
- Immunity, Active/genetics
- Inhibitor of Apoptosis Proteins/genetics
- Inhibitor of Apoptosis Proteins/immunology
- Lysosomal Membrane Proteins/genetics
- Lysosomal Membrane Proteins/immunology
- Major Histocompatibility Complex/genetics
- Major Histocompatibility Complex/immunology
- Mice
- Mice, Inbred C57BL
- Survivin
- T-Lymphocytes, Cytotoxic/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vascular Endothelial Growth Factor Receptor-2/genetics
- Vascular Endothelial Growth Factor Receptor-2/immunology
Collapse
Affiliation(s)
- Yuying Wei
- Department of Immunology, The Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Iurescia S, Fioretti D, Fazio VM, Rinaldi M. Epitope-driven DNA vaccine design employing immunoinformatics against B-cell lymphoma: A biotech's challenge. Biotechnol Adv 2012; 30:372-83. [DOI: 10.1016/j.biotechadv.2011.06.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 06/16/2011] [Accepted: 06/23/2011] [Indexed: 12/16/2022]
|
19
|
Waheed MT, Thönes N, Müller M, Hassan SW, Gottschamel J, Lössl E, Kaul HP, Lössl AG. Plastid expression of a double-pentameric vaccine candidate containing human papillomavirus-16 L1 antigen fused with LTB as adjuvant: transplastomic plants show pleiotropic phenotypes. PLANT BIOTECHNOLOGY JOURNAL 2011; 9:651-60. [PMID: 21447051 DOI: 10.1111/j.1467-7652.2011.00612.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Human papillomavirus (HPV) causes cervical cancer in women worldwide, which is currently prevented by vaccines based on virus-like particles (VLPs). However, these vaccines have certain limitations in their availability to developing countries, largely due to elevated costs. Concerning the highest burden of disease in resource-poor countries, development of an improved mucosal and cost-effective vaccine is a necessity. As an alternative to VLPs, capsomeres have been shown to be highly immunogenic and can be used as vaccine candidate. Furthermore, coupling of an adjuvant like Escherichia coli heat-labile enterotoxin subunit B (LTB) to an antigen can increase its immunogenicity and reduce the costs related to separate co-administration of adjuvants. Our study demonstrates the expression of two pentameric proteins: the modified HPV-16 L1 (L1_2xCysM) and LTB as a fusion protein in tobacco chloroplasts. Homoplasmy of the transplastomic plants was confirmed by Southern blotting. Western blot analysis showed that the LTB-L1 fusion protein was properly expressed in the plastids and the recombinant protein was estimated to accumulate up to 2% of total soluble protein. Proper folding and display of conformational epitopes for both LTB and L1 in the fusion protein was confirmed by GM1-ganglioside binding assay and antigen capture ELISA, respectively. However, all transplastomic lines showed chlorosis, male sterility and growth retardation, which persisted in the ensuing four generations studied. Nevertheless, plants reached maturity and produced seeds by pollination with wild-type plants. Taken together, these results pave the way for the possible development of a low-cost adjuvant-coupled vaccine with potentially improved immunogenicity against cervical cancer.
Collapse
Affiliation(s)
- Mohammad T Waheed
- Department of Applied Plant Sciences and Plant Biotechnology (DAPP), University of Natural Resources and Applied Life Sciences (BOKU), Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Oosterhuis K, Öhlschläger P, van den Berg JH, Toebes M, Gomez R, Schumacher TN, Haanen JB. Preclinical development of highly effective and safe DNA vaccines directed against HPV 16 E6 and E7. Int J Cancer 2011; 129:397-406. [DOI: 10.1002/ijc.25894] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Accepted: 11/30/2010] [Indexed: 01/08/2023]
|
21
|
Orlandi F, Guevara-Patiño JA, Merghoub T, Wolchok JD, Houghton AN, Gregor PD. Combination of epitope-optimized DNA vaccination and passive infusion of monoclonal antibody against HER2/neu leads to breast tumor regression in mice. Vaccine 2011; 29:3646-54. [PMID: 21435405 DOI: 10.1016/j.vaccine.2011.03.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 02/27/2011] [Accepted: 03/05/2011] [Indexed: 12/31/2022]
Abstract
HER2/neu is an oncogene amplified and over-expressed in 20-30% of breast adenocarcinomas. Treatment with the humanized monoclonal antibody trastuzumab has shown efficacy in combination with cytotoxic agents, although resistance occurs over time. Novel approaches are needed to further increase antibody efficacy. In this study, we provide evidence in a mouse breast cancer therapeutic tumor model that the combination of active immunization with a modified HER2/neu DNA vaccine and passive infusion of an anti-HER2/neu monoclonal antibody leads to significant regression of established tumors. Our data indicate that combination therapy with a HER2/neu DNA vaccine and trastuzumab may have clinical activity in breast cancer patients.
Collapse
Affiliation(s)
- Francesca Orlandi
- The Swim Across America Laboratory, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, United States
| | | | | | | | | | | |
Collapse
|
22
|
Induction of TLR4-dependent CD8+ T cell immunity by murine β-defensin2 fusion protein vaccines. Vaccine 2011; 29:3476-82. [PMID: 21382485 DOI: 10.1016/j.vaccine.2011.02.061] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Revised: 01/13/2011] [Accepted: 02/16/2011] [Indexed: 02/02/2023]
Abstract
Our laboratory previously described the strategy of fusing chemokine receptor ligands to antigens in order to generate immunogenic DNA vaccines. In the present study, we produced mouse β-2 defensin (mBD2) fusion proteins using both ovalbumin (OVA) and gp100 as model antigens. Superior cross-presentation by dendritic cells (DC) was observed for mBD2 fused antigens over unfused antigens in vitro. In vivo, we observed significant increases in the expansion of adoptively transferred antigen-specific MHC class I, but not class II-restricted T cells after immunization with mBD2 fused antigen over antigen alone. This enhanced expansion of class I restricted T cells was Toll-like receptor 4 (TLR4) dependent, but CC chemokine receptor 6 (CCR6) independent. Superior tumor resistance was observed for mBD2-fusion protein vaccines, compared to unfused antigen, in both B16-OVA and B16 tumor models. These data suggest that production of mBD2 fusion proteins is feasible and that the vaccines facilitate in vivo expansion of adoptively transferred T cells through a TLR4-dependent mechanism.
Collapse
|
23
|
Signori E, Iurescia S, Massi E, Fioretti D, Chiarella P, De Robertis M, Rinaldi M, Tonon G, Fazio VM. DNA vaccination strategies for anti-tumour effective gene therapy protocols. Cancer Immunol Immunother 2010; 59:1583-91. [PMID: 20390416 PMCID: PMC11030090 DOI: 10.1007/s00262-010-0853-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Accepted: 03/26/2010] [Indexed: 10/19/2022]
Abstract
After more than 15 years of experimentation, DNA vaccines have become a promising perspective for tumour diseases, and animal models are widely used to study the biological features of human cancer progression and to test the efficacy of vaccination protocols. In recent years, immunisation with naked plasmid DNA encoding tumour-associated antigens or tumour-specific antigens has revealed a number of advantages: antigen-specific DNA vaccination stimulates both cellular and humoral immune responses; multiple or multi-gene vectors encoding several antigens/determinants and immune-modulatory molecules can be delivered as single administration; DNA vaccination does not induce autoimmune disease in normal animals; DNA vaccines based on plasmid vectors can be produced and tested rapidly and economically. However, DNA vaccines have shown low immunogenicity when tested in human clinical trials, and compared with traditional vaccines, they induce weak immune responses. Therefore, the improvement of vaccine efficacy has become a critical goal in the development of effective DNA vaccination protocols for anti-tumour therapy. Several strategies are taken into account for improving the DNA vaccination efficacy, such as antigen optimisation, use of adjuvants and delivery systems like electroporation, co-expression of cytokines and co-stimulatory molecules in the same vector, different vaccination protocols. In this review we discuss how the combination of these approaches may contribute to the development of more effective DNA vaccination protocols for the therapy of lymphoma in a mouse model.
Collapse
Affiliation(s)
- Emanuela Signori
- CNR-Institute of Neurobiology and Molecular Medicine, Via Fosso del Cavaliere100, 00133 Rome, Italy
- Section of Molecular Medicine and Biotechnology, PRABB, Centre of Integrated Research, Università Campus Bio-Medico di Roma, Via A. del Portillo 21, 00128 Rome, Italy
| | - Sandra Iurescia
- CNR-Institute of Neurobiology and Molecular Medicine, Via Fosso del Cavaliere100, 00133 Rome, Italy
| | - Emanuela Massi
- CNR-Institute of Neurobiology and Molecular Medicine, Via Fosso del Cavaliere100, 00133 Rome, Italy
- Section of Molecular Medicine and Biotechnology, PRABB, Centre of Integrated Research, Università Campus Bio-Medico di Roma, Via A. del Portillo 21, 00128 Rome, Italy
| | - Daniela Fioretti
- CNR-Institute of Neurobiology and Molecular Medicine, Via Fosso del Cavaliere100, 00133 Rome, Italy
| | - Pieranna Chiarella
- CNR-Institute of Neurobiology and Molecular Medicine, Via Fosso del Cavaliere100, 00133 Rome, Italy
- Section of Molecular Medicine and Biotechnology, PRABB, Centre of Integrated Research, Università Campus Bio-Medico di Roma, Via A. del Portillo 21, 00128 Rome, Italy
| | - Mariangela De Robertis
- CNR-Institute of Neurobiology and Molecular Medicine, Via Fosso del Cavaliere100, 00133 Rome, Italy
- Section of Molecular Medicine and Biotechnology, PRABB, Centre of Integrated Research, Università Campus Bio-Medico di Roma, Via A. del Portillo 21, 00128 Rome, Italy
| | - Monica Rinaldi
- CNR-Institute of Neurobiology and Molecular Medicine, Via Fosso del Cavaliere100, 00133 Rome, Italy
| | - Giancarlo Tonon
- Bio-ker S.r.l., POLARIS, Località Piscinamanna, 09010 Pula, Cagliari Italy
| | - Vito Michele Fazio
- Section of Molecular Medicine and Biotechnology, PRABB, Centre of Integrated Research, Università Campus Bio-Medico di Roma, Via A. del Portillo 21, 00128 Rome, Italy
| |
Collapse
|
24
|
Genetic immunization with CDR3-based fusion vaccine confers protection and long-term tumor-free survival in a mouse model of lymphoma. J Biomed Biotechnol 2010; 2010:316069. [PMID: 20445751 PMCID: PMC2860581 DOI: 10.1155/2010/316069] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 02/04/2010] [Indexed: 12/26/2022] Open
Abstract
Therapeutic vaccination against idiotype is a promising strategy for immunotherapy of B-cell malignancies. We have previously shown that CDR3-based DNA immunization can induce immune response against lymphoma and explored this strategy to provide protection in a murine B-cell lymphoma model. Here we performed vaccination employing as immunogen a naked DNA fusion product. The DNA vaccine was generated following fusion of a sequence derived from tetanus toxin fragment C to the VHCDR3109−116 epitope. Induction of tumor-specific immunity as well as ability to inhibit growth of the aggressive 38C13 lymphoma and to prolong survival of vaccinated mice has been tested. We determined that DNA fusion vaccine induced immune response, elicited a strong protective antitumor immunity, and ensured almost complete long-term tumor-free survival of vaccinated mice.
Our results show that CDR3-based DNA fusion vaccines hold promise for vaccination against lymphoma.
Collapse
|
25
|
DNA vaccines: developing new strategies against cancer. J Biomed Biotechnol 2010; 2010:174378. [PMID: 20368780 PMCID: PMC2846346 DOI: 10.1155/2010/174378] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Accepted: 02/05/2010] [Indexed: 12/14/2022] Open
Abstract
Due to their rapid and widespread development, DNA vaccines have entered into a variety of human clinical trials for vaccines against various diseases including cancer. Evidence that DNA vaccines are well tolerated and have an excellent safety profile proved to be of advantage as many clinical trials combines the first phase with the second, saving both time and money. It is clear from the results obtained in clinical trials that such DNA vaccines require much improvement in antigen expression and delivery methods to make them sufficiently effective in the clinic. Similarly, it is clear that additional strategies are required to activate effective immunity against poorly immunogenic tumor antigens. Engineering vaccine design for manipulating antigen presentation and processing pathways is one of the most important aspects that can be easily handled in the DNA vaccine technology. Several approaches have been investigated including DNA vaccine engineering, co-delivery of immunomodulatory molecules, safe routes of administration, prime-boost regimen and strategies to break the immunosuppressive networks mechanisms adopted by malignant cells to prevent immune cell function. Combined or single strategies to enhance the efficacy and immunogenicity of DNA vaccines are applied in completed and ongoing clinical trials, where the safety and tolerability of the DNA platform are substantiated.
In this review on DNA vaccines, salient aspects on this topic going from basic research to the clinic are evaluated. Some representative DNA cancer vaccine studies are also discussed.
Collapse
|
26
|
Intratumoral DNA electroporation induces anti-tumor immunity and tumor regression. Cancer Immunol Immunother 2010; 59:409-17. [PMID: 19730859 DOI: 10.1007/s00262-009-0760-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 08/21/2009] [Indexed: 12/27/2022]
Abstract
In situ expression of a foreign antigen and an immune-modulating cytokine by intratumoral DNA electroporation was tested as a cancer therapy regimen. Transgene expression in the tumors was sustained for 2-3 weeks after intratumoral electroporation with mammalian expression plasmid containing firefly luciferase cDNA. Electroporation with cDNA encoding tetanus toxin fragment C (TetC) induced tetanus toxin-binding antibody, demonstrating immune recognition of the transgene product. Intratumoral electroporation with TetC and IL-12 cDNA after mice were treated with CD25 mAb to remove regulatory T cells induced IFN-gamma producing T-cell response to tumor-associated antigen, heavy inflammatory infiltration, regression of established tumors and immune memory to protect mice from repeated tumor challenge. Intratumoral expression of immune-modulating molecules may be most suitable in the neoadjuvant setting to enhance the therapeutic efficacy and provide long-term protection.
Collapse
|
27
|
Oosterhuis K, van den Berg JH, Schumacher TN, Haanen JBAG. DNA vaccines and intradermal vaccination by DNA tattooing. Curr Top Microbiol Immunol 2010; 351:221-50. [PMID: 21107792 DOI: 10.1007/82_2010_117] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over the past two decades, DNA vaccination has been developed as a method for the induction of immune responses. However, in spite of high expectations based on their efficacy in preclinical models, immunogenicity of first generation DNA vaccines in clinical trials was shown to be poor, and no DNA vaccines have yet been licensed for human use. In recent years significant progress has been made in the development of second generation DNA vaccines and DNA vaccine delivery methods. Here we review the key characteristics of DNA vaccines as compared to other vaccine platforms, and recent insights into the prerequisites for induction of immune responses by DNA vaccines will be discussed. We illustrate the development of second generation DNA vaccines with the description of DNA tattooing as a novel DNA delivery method. This technique has shown great promise both in a small animal model and in non-human primates and is currently under clinical evaluation.
Collapse
Affiliation(s)
- K Oosterhuis
- Division of Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
28
|
Abstract
Standard therapies for many common cancers remain toxic and are often ineffective. Cellular immunotherapy has the potential to be a highly targeted alternative, with low toxicity to normal tissues but a high capacity to eradicate tumor. In this chapter we describe approaches that generate cellular therapies using active immunization with cells, proteins, peptides, or nucleic acids, as well as efforts that use adoptive transfer of effector cells that directly target antigens on malignant cells. Many of these approaches are proving successful in hematologic malignancy and in melanoma. In this chapter we discuss the advantages and limitations of each and how over the next decade investigators will attempt to broaden their reach, increase their efficacy, and simplify their application.
Collapse
Affiliation(s)
- Fatma V Okur
- Baylor College of Medicine, Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | | |
Collapse
|
29
|
Alvarez E, Moga E, Barquinero J, Sierra J, Briones J. Dendritic and tumor cell fusions transduced with adenovirus encoding CD40L eradicate B-cell lymphoma and induce a Th17-type response. Gene Ther 2009; 17:469-77. [PMID: 20010627 DOI: 10.1038/gt.2009.150] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Fusion of dendritic cells and tumor cells (FCs) constitutes a promising tool for generating an antitumor response because of their capacity to present tumor antigens and provide appropriate costimulatory signals. CD40-CD40L interaction has an important role in the maturation and survival of dendritic cells and provides critical help for T-cell priming. In this study, we sought to improve the effectiveness of FC vaccines in a murine model of B-cell lymphoma by engineering FCs to express CD40L by means of an adenovirus encoding CD40L (Adv-CD40L). Before transduction with Adv-CD40L, no CD40L expression was detected in FCs, DCs or tumor cells. The surface expression of CD40L in FC transduced with Adv-CD40L (FC-CD40L) ranged between 50 and 60%. FC-CD40L showed an enhanced expression of CD80, CD86, CD54 and MHC class II molecules and elicited a strong in vitro immune response in a syngeneic mixed lymphocyte reaction. Furthermore, FC-CD40L showed enhanced migration to secondary lymphoid organs. Splenocytes from mice treated with FC-CD40L had a dramatic increase in the production of IL-17, IL-6 and IFN-gamma, compared with controls. Treatment with the FC-CD40L vaccine induced regression of established tumors and increased survival. Our data demonstrate that FC transduced with Adv-CD40L enhances the antitumor effect of FC vaccines in a murine lymphoma model and this is associated with an increased Th17-type immune response.
Collapse
Affiliation(s)
- E Alvarez
- Department of Hematology, Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | | | | | | | | |
Collapse
|
30
|
Brandsma JL, Shlyankevich M, Su Y, Zelterman D, Rose JK, Buonocore L. Reversal of papilloma growth in rabbits therapeutically vaccinated against E6 with naked DNA and/or vesicular stomatitis virus vectors. Vaccine 2009; 28:8345-51. [PMID: 19615481 DOI: 10.1016/j.vaccine.2009.04.082] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Revised: 03/27/2009] [Accepted: 04/03/2009] [Indexed: 02/02/2023]
Abstract
Persistent infection with high-risk human papillomaviruses (HPVs) is the greatest risk factor for the development of HPV-associated cancers. In this study rabbits bearing persistent and potentially malignant papillomas were used to test the efficacy of vaccination with a recombinant DNA and/or vesicular stomatitis virus (VSV) targeting the cottontail rabbit papillomavirus (CRPV) E6 protein. Immune responses were primed with either vector and boosted twice with the homologous or heterologous E6 vector. Over the course of 18 weeks, E6 vaccination reduced papilloma volumes to one third the volume in the controls, and the rabbits boosted with an heterologous vector tended to mount stronger responses. Small and medium-sized papillomas responded significantly but only slightly better than large papillomas. Finally the initial papilloma burden per rabbit, ranging from <100 mm(3) to >1000 mm(3), was not prognostic of antitumor efficacy. In summary both E6 vaccines elicited significant therapeutic immunity, and their sequential use tended to be advantageous.
Collapse
Affiliation(s)
- Janet L Brandsma
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
Since the discovery, over a decade and a half ago, that genetically engineered DNA can be delivered in vaccine form and elicit an immune response, there has been much progress in understanding the basic biology of this platform. A large amount of data has been generated in preclinical model systems, and more sustained cellular responses and more consistent antibody responses are being observed in the clinic. Four DNA vaccine products have recently been approved, all in the area of veterinary medicine. These results suggest a productive future for this technology as more optimized constructs, better trial designs and improved platforms are being brought into the clinic.
Collapse
Affiliation(s)
- Michele A Kutzler
- Division of Infectious Diseases and HIV Medicine, The Department of Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, USA
| | | |
Collapse
|
32
|
DNA fusion gene vaccination mobilizes effective anti-leukemic cytotoxic T lymphocytes from a tolerized repertoire. Eur J Immunol 2008; 38:2118-30. [PMID: 18624299 DOI: 10.1002/eji.200838213] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The majority of known human tumor-associated antigens derive from non-mutated self proteins. T cell tolerance, essential to prevent autoimmunity, must therefore be cautiously circumvented to generate cytotoxic T cell responses against these targets. Our strategy uses DNA fusion vaccines to activate high levels of peptide-specific CTL. Key foreign sequences from tetanus toxin activate tolerance-breaking CD4(+) T cell help. Candidate MHC class I-binding tumor peptide sequences are fused to the C terminus for optimal processing and presentation. To model performance against a leukemia-associated antigen in a tolerized setting, we constructed a fusion vaccine encoding an immunodominant CTL epitope derived from Friend murine leukemia virus gag protein (FMuLV(gag)) and vaccinated tolerant FMuLV(gag)-transgenic (gag-Tg) mice. Vaccination with the construct induced epitope-specific IFN-gamma-producing CD8(+) T cells in normal and gag-Tg mice. The frequency and avidity of activated cells were reduced in gag-Tg mice, and no autoimmune injury resulted. However, these CD8(+) T cells did exhibit gag-specific cytotoxicity in vitro and in vivo. Also, epitope-specific CTL killed FBL-3 leukemia cells expressing endogenous FMuLV(gag) antigen and protected against leukemia challenge in vivo. These results demonstrate a simple strategy to engage anti-microbial T cell help to activate epitope-specific polyclonal CD8(+) T cell responses from a residual tolerized repertoire.
Collapse
|
33
|
Huebener N, Fest S, Strandsby A, Michalsky E, Preissner R, Zeng Y, Gaedicke G, Lode HN. A rationally designed tyrosine hydroxylase DNA vaccine induces specific antineuroblastoma immunity. Mol Cancer Ther 2008; 7:2241-51. [DOI: 10.1158/1535-7163.mct-08-0109] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
34
|
Tenbusch M, Kuate S, Tippler B, Gerlach N, Schimmer S, Dittmer U, Uberla K. Coexpression of GM-CSF and antigen in DNA prime-adenoviral vector boost immunization enhances polyfunctional CD8+ T cell responses, whereas expression of GM-CSF antigen fusion protein induces autoimmunity. BMC Immunol 2008; 9:13. [PMID: 18405363 PMCID: PMC2324072 DOI: 10.1186/1471-2172-9-13] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2007] [Accepted: 04/11/2008] [Indexed: 12/29/2022] Open
Abstract
Background Granulocyte-macrophage colony-stimulating factor (GM-CSF) has shown promising results as a cytokine adjuvant for antiviral vaccines and in various models of tumor gene therapy. To explore whether the targeting of antigens to GM-CSF receptors on antigen-presenting cells enhances antigen-specific CD8 T-cell responses, fusion proteins of GM-CSF and ovalbumin (OVA) were expressed by DNA and adenoviral vector vaccines. In addition, bicistronic vectors allowing independent expression of the antigen and the cytokine were tested in parallel. Results In vitro, the GM-CSF ovalbumin fusion protein (GM-OVA) led to the better stimulation of OVA-specific CD8+ T cells by antigen-presenting cells than OVA and GM-CSF given as two separate proteins. However, prime-boost immunizations of mice with DNA and adenoviral vector vaccines encoding GM-OVA suppressed CD8+ T-cell responses to OVA. OVA-specific IgG2a antibody levels were also reduced, while the IgG1 antibody response was enhanced. Suppression of CD8+ T cell responses by GM-OVA vaccines was associated with the induction of neutralizing antibodies to GM-CSF. In contrast, the coexpression of GM-CSF and antigens in DNA prime adenoviral boost immunizations led to a striking expansion of polyfunctional OVA-specific CD8+ T cells without the induction of autoantibodies. Conclusion The induction of autoantibodies suggests a general note of caution regarding the use of highly immunogenic viral vector vaccines encoding fusion proteins between antigens and host proteins. In contrast, the expansion of polyfunctional OVA-specific CD8+ T cells after immunizations with bicistronic vectors further support a potential application of GM-CSF as an adjuvant for heterologous prime-boost regimens with genetic vaccines. Since DNA prime adenoviral vector boost regimenes are presently considered as one of the most efficient ways to induce CD8+ T cell responses in mice, non-human primates and humans, further enhancement of this response by GM-CSF is a striking observation.
Collapse
Affiliation(s)
- Matthias Tenbusch
- Department of Molecular and Medical Virology, Ruhr-University Bochum, 44801 Bochum, Germany.
| | | | | | | | | | | | | |
Collapse
|
35
|
Engelhorn ME, Guevara-Patiño JA, Merghoub T, Liu C, Ferrone CR, Rizzuto GA, Cymerman DH, Posnett DN, Houghton AN, Wolchok JD. Mechanisms of immunization against cancer using chimeric antigens. Mol Ther 2008; 16:773-81. [PMID: 18301399 PMCID: PMC4399381 DOI: 10.1038/mt.2008.8] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Successful approaches to tumor immunotherapy must overcome the physiological state of tolerance of the immune system to self-tumor antigens. Immunization with appropriate variants of syngeneic antigens can achieve this. However, improvements in vaccine design are needed for efficient cancer immunotherapy. Here we explore nine different chimeric vaccine designs, in which the antigen of interest is expressed as an in-frame fusion with polypeptides that impact antigen processing or presentation. In DNA immunization experiments in mice, three of nine fusions elevated relevant CD8(+) T-cell responses and tumor protection relative to an unfused melanoma antigen. These fusions were: Escherichia coli outer membrane protein A (OmpA), Pseudomonas aeruginosa exotoxin A, and VP22 protein of herpes simplex virus-1. The gains of immunogenicity conferred by the latter two are independent of epitope presentation by major histocompatibility complex class II (MHC II). This finding has positive implications for immunotherapy in individuals with CD4(+) T-cell deficiencies. We present evidence that antigen instability is not a sine qua non condition for immunogenicity. Experiments using two additional melanoma antigens identified different optimal fusion partners, thereby indicating that the benefits of fusion vectors remain antigen specific. Therefore large fusion vector panels such as those presented here can provide information to promote the successful advancement of gene-based vaccines.
Collapse
Affiliation(s)
- Manuel E Engelhorn
- The Swim Across America Laboratory, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Rice J, Ottensmeier CH, Stevenson FK. DNA vaccines: precision tools for activating effective immunity against cancer. Nat Rev Cancer 2008; 8:108-20. [PMID: 18219306 DOI: 10.1038/nrc2326] [Citation(s) in RCA: 295] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
DNA vaccination has suddenly become a favoured strategy for inducing immunity. The molecular precision offered by gene-based vaccines, together with the facility to include additional genes to direct and amplify immunity, has always been attractive. However, the apparent failure to translate operational success in preclinical models to the clinic, for reasons that are now rather obvious, reduced initial enthusiasm. Recently, novel delivery systems, especially electroporation, have overcome this translational block. Here, we assess the development, current performance and potential of DNA vaccines for the treatment of cancer.
Collapse
Affiliation(s)
- Jason Rice
- Genetic Vaccine Group, Cancer Sciences Division, University of Southampton School of Medicine, Southampton General Hospital, Southampton,SO16 6YD, UK
| | | | | |
Collapse
|
37
|
Radcliffe JN, Roddick JS, Stevenson FK, Thirdborough SM. Prolonged Antigen Expression following DNA Vaccination Impairs Effector CD8+ T Cell Function and Memory Development. THE JOURNAL OF IMMUNOLOGY 2007; 179:8313-21. [DOI: 10.4049/jimmunol.179.12.8313] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
38
|
Facciabene A, Aurisicchio L, Elia L, Palombo F, Mennuni C, Ciliberto G, La Monica N. Vectors encoding carcinoembryonic antigen fused to the B subunit of heat-labile enterotoxin elicit antigen-specific immune responses and antitumor effects. Vaccine 2007; 26:47-58. [DOI: 10.1016/j.vaccine.2007.10.060] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Revised: 10/14/2007] [Accepted: 10/21/2007] [Indexed: 11/26/2022]
|
39
|
Wang YS, Wang GQ, Wen YJ, Wang L, Chen XC, Chen P, Kan B, Li J, Huang C, Lu Y, Zhou Q, Xu N, Li D, Fan LY, Yi T, Wu HB, Wei YQ. Immunity against Tumor Angiogenesis Induced by a Fusion Vaccine with Murine β-Defensin 2 and mFlk-1. Clin Cancer Res 2007; 13:6779-87. [PMID: 18006780 DOI: 10.1158/1078-0432.ccr-07-1587] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Yong-sheng Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Guo Xue Xiang No. 37, Chengdu, Sichuan, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Chiarella P, Massi E, De Robertis M, Signori E, Fazio VM. Adjuvants in vaccines and for immunisation: current trends. Expert Opin Biol Ther 2007; 7:1551-62. [DOI: 10.1517/14712598.7.10.1551] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
41
|
Mittal S, Marshall NA, Barker RN, Vickers MA. Immunomodulation against leukemias and lymphomas: a realistic future treatment? Crit Rev Oncol Hematol 2007; 65:101-8. [PMID: 17719232 DOI: 10.1016/j.critrevonc.2007.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Revised: 05/09/2007] [Accepted: 05/18/2007] [Indexed: 11/26/2022] Open
Abstract
Immunotherapy offers the potential for cure of malignancy without the side effects too commonly seen with conventional chemotherapy. The efficacy of allogenic transplantation and monoclonal antibodies in hematological malignancies illustrate this principle and are now part of routine care. Newer cell based and molecular approaches aimed at stimulating cytotoxic activity against host derived tumor associated antigens are able to 'boost' anti-tumor immunity as judged by immunological assays in vitro. Although clinically meaningful responses were originally less evident, more promising results are now being reported. Our growing understanding of tumor immunology provide rationales for further improvements in the field.
Collapse
Affiliation(s)
- S Mittal
- Department of Clinical Haematology, Aberdeen Royal Infirmary, Aberdeen AB25 2ZN, United Kingdom
| | | | | | | |
Collapse
|
42
|
Guinn BA, Kasahara N, Farzaneh F, Habib NA, Norris JS, Deisseroth AB. Recent Advances and Current Challenges in Tumor Immunology and Immunotherapy. Mol Ther 2007; 15:1065-71. [PMID: 17375068 DOI: 10.1038/sj.mt.6300138] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Despite advances in animal studies, where the cure of the majority of mice with pre-established (albeit early-stage) tumors has become almost standard, human clinical trials have been much less successful. Here we describe some of the most recent advances in the specialist field of tumor immunology and immunotherapy, highlighting salient work to identify key problem areas and potential solutions. We make particular note of recent developments in adoptive therapy; whole-cell, DNA, and peptide vaccines; and antibody therapy. We also describe the revival of interest in regulatory T cells and conclude by detailing the need for clinical trial read-out autonomy and methods to predict which patients will respond to a particular treatment.
Collapse
Affiliation(s)
- Barbara-ann Guinn
- Department of Haematological Medicine, King's College London School of Medicine, The Rayne, Institute, London, UK.
| | | | | | | | | | | |
Collapse
|
43
|
Nitschke C, Flechsig E, van den Brandt J, Lindner N, Lührs T, Dittmer U, Klein MA. Immunisation strategies against prion diseases: prime-boost immunisation with a PrP DNA vaccine containing foreign helper T-cell epitopes does not prevent mouse scrapie. Vet Microbiol 2007; 123:367-76. [PMID: 17499458 DOI: 10.1016/j.vetmic.2007.03.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Vaccination against prion diseases constitutes a promising approach for the treatment and prevention of the disease. Passive immunisation with antibodies binding to the cellular prion protein (PrP(C)) can protect against prion disease. However, immunotherapeutic strategies with active immunisation are limited due to the immune tolerance against the self-antigen. In order to develop an anti-prion vaccine, we designed a novel DNA fusion vaccine composed of mouse PrP and immune stimulatory helper T-cell epitopes of the tetanus toxin that have previously been reported to break tolerance to other self-antigens. This approach provoked a strong PrP(C)-specific humoral and cellular immune response in PrP null mice, but only low antibody titres were found in vaccinated wild-type mice. Furthermore, prime-boost immunisation with the DNA vaccine and recombinant PrP protein increased antibody titres in PrP null mice, but failed to protect wild-type mice from mouse scrapie.
Collapse
Affiliation(s)
- Cindy Nitschke
- Institute of Virology and Immunobiology, University of Wuerzburg, Versbacherstr. 7, D-97078 Wuerzburg, Germany
| | | | | | | | | | | | | |
Collapse
|
44
|
Radcliffe JN, Roddick JS, Friedmann PS, Stevenson FK, Thirdborough SM. Prime-Boost with Alternating DNA Vaccines Designed to Engage Different Antigen Presentation Pathways Generates High Frequencies of Peptide-Specific CD8+ T Cells. THE JOURNAL OF IMMUNOLOGY 2006; 177:6626-33. [PMID: 17082574 DOI: 10.4049/jimmunol.177.10.6626] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The route for presentation of Ag to CD8+ or CD4+ T cells following DNA vaccination is critical for determining outcome, but the pathways involved are unclear. In this study, we compare two different DNA vaccine designs aimed to elicit CD8+ T cell responses against a specific peptide-epitope either by direct- or cross-presentation. Each carries sequences from tetanus toxin (TT) to provide essential CD4+ T cell help. In the first already proven design, the peptide-epitope is fused to the N-terminal domain of fragment C from TT. This appears to act mainly by cross-presentation. In the second design, the peptide-epitope is encoded by a minigene, with induction of Th responses mediated by coexpression of a hybrid invariant chain molecule, incorporating a single determinant from TT (p30) in exchange for class II-associated invariant chain peptide. This design appears to act mainly via direct presentation from transfected APCs. Both vaccines mediated Th-dependent priming of CD8+ T cells in mice, but the kinetics and level of the responses differed markedly, consistent with engagement of distinct pathways of Ag presentation. Importantly, the vaccines could be combined in an alternating prime-boost regime, in either order, generating substantially expanded memory CD8+ T cells, with potent effector function. Taken together, these results demonstrate that vaccination protocols involving different modes of Ag presentation at prime and boost can significantly improve the effectiveness of immunization.
Collapse
MESH Headings
- Animals
- Antigen Presentation/genetics
- Antigen Presentation/immunology
- Antigens, Differentiation, B-Lymphocyte/administration & dosage
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigens, Differentiation, B-Lymphocyte/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cytotoxicity, Immunologic/genetics
- Egg Proteins/administration & dosage
- Egg Proteins/genetics
- Egg Proteins/immunology
- H-2 Antigens/immunology
- H-2 Antigens/metabolism
- Histocompatibility Antigens Class II/administration & dosage
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/immunology
- Immunization, Secondary/methods
- Interferon-gamma/biosynthesis
- Lymphocyte Count
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Ovalbumin/administration & dosage
- Ovalbumin/genetics
- Ovalbumin/immunology
- Peptide Fragments/administration & dosage
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes, Helper-Inducer/immunology
- Tetanus Toxin/administration & dosage
- Tetanus Toxin/genetics
- Tetanus Toxin/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/chemical synthesis
- Vaccines, DNA/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Joanna N Radcliffe
- Cancer Sciences, University of Southampton School of Medicine, Southampton General Hospital, Southampton, United Kingdom
| | | | | | | | | |
Collapse
|
45
|
Choudhury A, Mosolits S, Kokhaei P, Hansson L, Palma M, Mellstedt H. Clinical results of vaccine therapy for cancer: learning from history for improving the future. Adv Cancer Res 2006; 95:147-202. [PMID: 16860658 DOI: 10.1016/s0065-230x(06)95005-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Active, specific immunotherapy for cancer holds the potential of providing an approach for treating cancers, which have not been controlled by conventional therapy, with very little or no associated toxicity. Despite advances in the understanding of the immunological basis of cancer vaccine therapy as well as technological progress, clinical effectiveness of this therapy has often been frustratingly unpredictable. Hundreds of preclinical and clinical studies have been performed addressing issues related to the generation of a therapeutic immune response against tumors and exploring a diverse array of antigens, immunological adjuvants, and delivery systems for vaccinating patients against cancer. In this chapter, we have summarized a number of clinical trials performed in various cancers with focus on the clinical outcome of vaccination therapy. We have also attempted to draw objective inferences from the published data that may influence the clinical effectiveness of vaccination approaches against cancer. Collectively the data indicate that vaccine therapy is safe, and no significant autoimmune reactions are observed even on long term follow-up. The design of clinical trials have not yet been optimized, but meaningful clinical effects have been seen in B-cell malignancies, lung, prostate, colorectal cancer, and melanoma. It is also obvious that patients with limited disease or in the adjuvant settings have benefited most from this targeted therapy approach. It is imperative that future studies focus on exploring the relationship between immune and clinical responses to establish whether immune monitoring could be a reliable surrogate marker for evaluating the clinical efficacy of cancer vaccines.
Collapse
Affiliation(s)
- Aniruddha Choudhury
- Department of Oncology, Cancer Centre Karolinska, Karolinska University, Hospital Solna, SE-171 76 Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
46
|
Munker R, Reibke R, Kolb HJ. Graft-versus-host and graft-versus-leukemia reactions: a summary of the Seventh International Symposium held in Garmisch-Partenkirchen, Germany, February 22nd–25th, 2006, Tolerance and Immunity, an update on lymphoid malignancies. Bone Marrow Transplant 2006; 38:593-607. [PMID: 16980992 DOI: 10.1038/sj.bmt.1705499] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The Seventh International Symposium on graft-versus-host and graft-versus-leukemia reactions was held in Garmisch Partenkirchen (Germany, near Lake Riessersee) between January 22nd and 25th, 2006. A total of more than 100 invited participants (scientists and clinicians working in the area of allogeneic stem cell transplantation) discussed research in the area of lymphoid malignancies. Major topics of the 2006 meeting were lymphocyte biology, experimental systems, lymphoma pathogenesis, cellular therapy in vivo and vitro, idiotype-specific responses and graft-versus-malignancy reactions for lymphomas and multiple myeloma. Further highlights were immune responses to blasts of ALL, haploidentical transplantation, role of natural killer cells, clinical guidelines for allogeneic transplantation and adoptive immunotherapy in chronic lymphocytic leukemia and multiple myeloma, new antibody-mediated strategies. As can be seen in the summaries of the individual presentations, progress was made in the understanding of lymphoma biology and in the clinical application of graft-versus-lymphoma or graft-versus-myeloma effects. Each day was followed by round-table discussions, which summarized new data and challenged established concepts. The discussions resulted in new insights and projects for basic research and clinical transplantation.
Collapse
Affiliation(s)
- R Munker
- Division of Hematology/Oncology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA.
| | | | | |
Collapse
|
47
|
Rice J, Dunn S, Piper K, Buchan SL, Moss PA, Stevenson FK. DNA fusion vaccines induce epitope-specific cytotoxic CD8(+) T cells against human leukemia-associated minor histocompatibility antigens. Cancer Res 2006; 66:5436-42. [PMID: 16707472 DOI: 10.1158/0008-5472.can-05-3130] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The graft-versus-leukemia effect of allogeneic stem-cell transplantation is believed to be mediated by T-cell recognition of minor histocompatibility antigens on recipient cells. For minor histocompatibility antigens HA-1 and HA-2, normal cell expression is restricted to hemopoietic cells, and boosting the immune response to these antigens may potentiate graft-versus-leukemia effect without accompanying graft-versus-host disease. To increase efficacy, expansion of HA-1- or HA-2-specific CTL before transplantation is desirable. However, primary HA-1- or HA-2-specific CTL expanded in vitro are often of low avidity. An alternative approach is to prime specific CTL responses in vivo by vaccination. Clearly, donor vaccination must be safe and specific. We have developed DNA fusion vaccines able to induce high levels of epitope-specific CTL using linked CD4(+) T-cell help. The vaccines incorporate a domain of tetanus toxin (DOM) fused to a sequence encoding a candidate MHC class I binding peptide. This design generates antitumor CD8(+) T-cell responses and protective immunity in preclinical models. For clinical application, we constructed vaccines encoding HLA-A*0201-restricted peptides from human HA-1 and HA-2, which were fused to DOM, and tested their performance in HLA-A*0201-transgenic mice. Priming induced epitope-specific, IFNgamma-producing CD8(+) T cells with cytotoxic function boosted to high levels with electroporation. Strikingly, these mouse T cells efficiently killed human lymphoblastoid cell lines expressing endogenous HA-1 or HA-2. High avidity is indicated by the independence of cytolysis from CD8/MHC class I interaction. These safe epitope-specific vaccines offer a potential strategy to prime HA-1- or HA-2-specific CTL in transplant donors before adoptive transfer.
Collapse
Affiliation(s)
- Jason Rice
- Molecular Immunology Group, Southampton University Hospitals Trust, Southampton, Hampshire, United Kingdom.
| | | | | | | | | | | |
Collapse
|
48
|
Naota H, Miyahara Y, Okumura S, Kuzushima K, Akatsuka Y, Hiasa A, Kitano S, Takahashi T, Yuta A, Majima Y, Shiku H. Generation of peptide-specific CD8+ T cells by phytohemagglutinin-stimulated antigen-mRNA-transduced CD4+ T cells. J Immunol Methods 2006; 314:54-66. [PMID: 16828790 DOI: 10.1016/j.jim.2006.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2005] [Revised: 05/10/2006] [Accepted: 05/15/2006] [Indexed: 12/29/2022]
Abstract
Functional analysis of antigen-specific CD8(+) T cells is important for understanding the immune response in various immunological disorders. To analyze CD8(+) T cell responses to a variety of antigens with no readily defined peptides available, we developed a system using CD4(+) phytohemagglutinin (PHA) blasts transduced with mRNA for antigen molecules. CD4(+) PHA blasts express MHC class I and II, and also CD80 and CD86 and are thus expected to serve as potent antigen presenting cells. EGFP mRNA could be transduced into and the protein expressed by more than 90% of either LCL or CD4(+) PHA blasts. Its expression stably persisted for more than 2 weeks after transduction. In experiments with HLA-A*2402 restricted CD8(+) CTL clones for either EBNA3A or a cancer-testis antigen, SAGE, mRNA-transduced lymphoid cells were appropriate target cells in ELISPOT assays or (51)Cr releasing assays. Finally, using CD4(+) PHA blasts transduced with mRNA of a cancer-testis antigen MAGE-A4, we successfully generated specific CTL clones that recognized a novel HLA-B*4002 restricted epitope, MAGE-A4(223-231). Messenger RNA-transduced CD4(+) PHA blasts are thus useful antigen presenting cells for analysis of CD8(+) T cell responses and induction of specific T cells for potential immunotherapy.
Collapse
Affiliation(s)
- Hiroaki Naota
- Second Department of Internal Medicine, Mie University School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Therapeutic vaccines targeting B cell lymphoma idiotype have reached an advanced stage of clinical development, with three multicenter randomized clinical trials ongoing. This review describes the rationale and development of this immunotherapeutic approach, the design of current phase III trials, and other active vaccination approaches likely to move forward into clinical testing for lymphomas. RECENT FINDINGS Several groups have achieved promising results in phase II trials of patient-specific idiotype vaccines, with very few side effects noted. Anti-idiotype antibodies, in addition to cytotoxic T cells, are now believed to be important effectors of antitumor immunity after idiotype vaccination. The manufacturing of autologous tumor idiotype proteins is being rapidly refined by the use of molecular technologies. Two trials involving more than 1000 patients are now under way, which use idiotype vaccination after induction chemotherapy; one trial completed accrual in early 2004. A third trial opened in 2004, using rituximab followed by idiotype vaccine with maintenance booster vaccines continuing throughout the period of normal B cell recovery. In accordance with the United States Food and Drug Administration, progression-free survival serves as the accepted primary efficacy endpoint in these studies. SUMMARY Lymphoma idiotype vaccination represents a promising immunotherapeutic approach targeting a patient-specific tumor antigen. The results of pivotal phase III trials for three first-generation idiotype vaccines will become available in the next several years. Advanced manufacturing techniques should permit application of this tailor-made treatment to large numbers of non-Hodgkin's lymphoma patients.
Collapse
Affiliation(s)
- Sara A Hurvitz
- Division of Hematology/Oncology, Center for Health Sciences, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | | |
Collapse
|
50
|
Steitz J, Tormo D, Schweichel D, Tüting T. Comparison of recombinant adenovirus and synthetic peptide for DC-based melanoma vaccination. Cancer Gene Ther 2006; 13:318-25. [PMID: 16151477 DOI: 10.1038/sj.cgt.7700894] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Optimal strategies for antigen-specific melanoma vaccination are currently being defined in experimental mouse models. Using a single H2-K(b)-binding peptide epitope derived from the melanosomal enzyme tyrosinase-related protein 2 (TRP2) in C57BL/6 mice, we show that adenovirus-transduced dendritic cells (DC) are clearly superior to peptide-pulsed DC for the induction of CD8+ T cells and antimelanoma immunity. Vaccine efficacy strictly depended on the presence of linked CD4+ T-cell help during the priming but not the effector phase of the immune response. These results provide important information for the translation of melanoma vaccine strategy in future clinical applications.
Collapse
Affiliation(s)
- J Steitz
- Department of Dermatology, Laboratory of Experimental Dermatology, University of Bonn, Bonn, Germany
| | | | | | | |
Collapse
|