1
|
Nguyen-Hoai T, Hohn O, Pezzutto A, Westermann J. Gene Gun Her2/neu DNA Vaccination: Evaluation of Vaccine Efficacy in a Syngeneic Her2/neu Mouse Tumor Model. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2521:129-154. [PMID: 35732996 DOI: 10.1007/978-1-0716-2441-8_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Genetic vaccination using naked plasmid DNA is an immunization strategy both against infectious diseases and cancer.In order to improve efficacy of DNA vaccines, particularly in large animals and humans, different strategies have been pursued. These vaccination strategies are based on different application routes, schedules and coexpression of immunomodulatory molecules as adjuvants. Our mouse tumor model offers the possibility to investigate Her2/neu DNA vaccines in different settings, that is, intramuscular or intradermal application with or without coexpression of adjuvants. The immunogenicity of predicted peptides for Her2/neu specific memory T cells were screened and confirmed after intramuscular and intradermal application. Protection from tumor growth in tumor challenge experiments and both T cell and humoral immune responses against Her2/neu peptides are used as surrogate parameters for vaccine efficacy.
Collapse
Affiliation(s)
- Tam Nguyen-Hoai
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | | | - Antonio Pezzutto
- Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Jörg Westermann
- Department Hematology, Oncology and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
2
|
Argenziano M, Occhipinti S, Scomparin A, Angelini C, Novelli F, Soster M, Giovarelli M, Cavalli R. Exploring chitosan-shelled nanobubbles to improve HER2 + immunotherapy via dendritic cell targeting. Drug Deliv Transl Res 2022; 12:2007-2018. [PMID: 35672651 PMCID: PMC9172608 DOI: 10.1007/s13346-022-01185-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2022] [Indexed: 11/29/2022]
Abstract
Immunotherapy is a valuable approach to cancer treatment as it is able to activate the immune system. However, the curative methods currently in clinical practice, including immune checkpoint inhibitors, present some limitations. Dendritic cell vaccination has been investigated as an immunotherapeutic strategy, and nanotechnology-based delivery systems have emerged as powerful tools for improving immunotherapy and vaccine development. A number of nanodelivery systems have therefore been proposed to promote cancer immunotherapy. This work aims to design a novel immunotherapy nanoplatform for the treatment of HER2 + breast cancer, and specially tailored chitosan-shelled nanobubbles (NBs) have been developed for the delivery of a DNA vaccine. The NBs have been functionalized with anti-CD1a antibodies to target dendritic cells (DCs). The NB formulations possess dimensions of approximately 300 nm and positive surface charge, and also show good physical stability up to 6 months under storage at 4 °C. In vitro characterization has confirmed that these NBs are capable of loading DNA with good encapsulation efficiency (82%). The antiCD1a-functionalized NBs are designed to target DCs, and demonstrated the ability to induce DC activation in both human and mouse cell models, and also elicited a specific immune response that was capable of slowing tumor growth in mice in vivo. These findings are the proof of concept that loading a tumor vaccine into DC-targeted chitosan nanobubbles may become an attractive nanotechnology approach for the future immunotherapeutic treatment of cancer.
Collapse
Affiliation(s)
- Monica Argenziano
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125, Turin, Italy
| | - Sergio Occhipinti
- Department of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, 10126, Turin, Italy
| | - Anna Scomparin
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125, Turin, Italy
| | - Costanza Angelini
- Department of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, 10126, Turin, Italy
| | - Francesco Novelli
- Department of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, 10126, Turin, Italy
| | - Marco Soster
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125, Turin, Italy
| | - Mirella Giovarelli
- Department of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, 10126, Turin, Italy
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125, Turin, Italy.
| |
Collapse
|
3
|
Florczak A, Deptuch T, Lewandowska A, Penderecka K, Kramer E, Marszalek A, Mackiewicz A, Dams-Kozlowska H. Functionalized silk spheres selectively and effectively deliver a cytotoxic drug to targeted cancer cells in vivo. J Nanobiotechnology 2020; 18:177. [PMID: 33261651 PMCID: PMC7709326 DOI: 10.1186/s12951-020-00734-y] [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: 09/25/2020] [Accepted: 11/19/2020] [Indexed: 12/31/2022] Open
Abstract
Background Chemotherapy is often a first-line therapeutic approach for the treatment of a wide variety of cancers. Targeted drug delivery systems (DDSs) can potentially resolve the problem of chemotherapeutic drug off-targeting effects. Herein, we examined in vivo models to determine the efficacy of Her2-targeting silk spheres (H2.1MS1) as DDSs for delivering doxorubicin (Dox) to Her2-positive and Her2-negative primary and metastatic mouse breast cancers. Results The specific accumulation of H2.1MS1 spheres was demonstrated at the site of Her2-positive cancer. Dox delivered only by functionalized H2.1MS1 particles selectively inhibited Her2-positive cancer growth in primary and metastatic models. Moreover, the significant effect of the Dox dose and the frequency of treatment administration on the therapeutic efficacy was indicated. Although the control MS1 spheres accumulated in the lungs in Her2-positive metastatic breast cancer, the Dox-loaded MS1 particles did not treat cancer. Histopathological examination revealed no systemic toxicity after multiple administrations and at increased doses of Dox-loaded silk spheres. Although the studies were performed in immunocompetent mice, the H2.1MS1 silk spheres efficiently delivered the drug, which exerted a therapeutic effect. Conclusion Our results indicated that functionalized silk spheres that enable cell-specific recognition, cellular internalization, and drug release represent an efficient strategy for cancer treatment in vivo.![]()
Collapse
Affiliation(s)
- Anna Florczak
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland. .,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland.
| | - Tomasz Deptuch
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland.,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland
| | - Anna Lewandowska
- Department of Tumor Pathology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland.,Department of Tumor Pathology and Prophylaxis, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland
| | - Karolina Penderecka
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland.,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland
| | - Elzbieta Kramer
- Department of Tumor Pathology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland
| | - Andrzej Marszalek
- Department of Tumor Pathology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland.,Department of Tumor Pathology and Prophylaxis, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland
| | - Andrzej Mackiewicz
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland.,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland
| | - Hanna Dams-Kozlowska
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland. .,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland.
| |
Collapse
|
4
|
An HER2 DNA vaccine with evolution-selected amino acid substitutions reveals a fundamental principle for cancer vaccine formulation in HER2 transgenic mice. Cancer Immunol Immunother 2019; 68:1143-1155. [PMID: 31177328 DOI: 10.1007/s00262-019-02333-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 03/26/2019] [Indexed: 12/19/2022]
Abstract
Enhancement of endogenous immunity to tumor-associated self-antigens and neoantigens is the goal of preventive vaccination. Toward this goal, we compared the efficacy of the following HER2 DNA vaccine constructs: vaccines encoding wild-type HER2, hybrid HER2 vaccines consisting of human HER2 and rat Neu, HER2 vaccines with single residue substitutions and a novel human HER2 DNA vaccine, ph(es)E2TM. ph(es)E2TM was designed to contain five evolution-selected substitutions: M198V, Q398R, F425L, H473R and A622T that occur frequently in 12 primate HER2 sequences. These ph(es)E2TM substitutions score 0 to 1 in blocks substitutions matrix (BLOSUM), indicating minimal biochemical alterations. h(es)E2TM recombinant protein is recognized by a panel of anti-HER2 mAbs, demonstrating the preservation of HER2 protein structure. Compared to native human HER2, electrovaccination of HER2 transgenic mice with ph(es)E2TM induced a threefold increase in HER2-binding antibody (Ab) and elevated levels of IFNγ-producing T cells. ph(es)E2TM, but not pE2TM immune serum, recognized HER2 peptide p95 355LPESFDGDPASNTAP369, suggesting a broadening of epitope recognition induced by the minimally modified HER2 vaccine. ph(es)E2TM vaccination reduced tumor growth more effectively than wild-type HER2 or HER2 vaccines with more extensive modifications. The elevation of tumor immunity by ph(es)E2TM vaccination would create a favorable tumor microenvironment for neoantigen priming, further enhancing the protective immunity. The fundamental principle of exploiting evolution-selected amino acid substitutions is novel, effective and applicable to vaccine development in general.
Collapse
|
5
|
Nilofar Danishmalik S, Lee SH, Sin JI. Tumor regression is mediated via the induction of HER263-71- specific CD8+ CTL activity in a 4T1.2/HER2 tumor model: no involvement of CD80 in tumor control. Oncotarget 2018; 8:26771-26788. [PMID: 28460461 PMCID: PMC5432296 DOI: 10.18632/oncotarget.15816] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 02/20/2017] [Indexed: 02/01/2023] Open
Abstract
In the CT26/HER2 and 4T1.2/HER2 tumor models, CT26/HER2 cells form tumors that continue to grow, whereas 4T1.2/HER2 cells form tumors that eventually regress. Here, we investigated the differences in the behaviors of these two cell lines. When immune cells from 4T1.2/HER2 tumor-bearing animals were stimulated with HER2 class I peptides, they displayed a 2-fold increase in IFN-γ levels, in response to the peptides, HER263-71 and HER2342-350. In contrast, extremely high levels of antigen-non-specific IFN-γ production were observed with immune cells and sera from CT26/HER2 tumor-bearing mice. However, IFN-γ had no effect on tumor progression in the CT26/HER2 model, as determined by an IFN-γ knockout assay. 4T1.2/HER2 tumor-bearing mice displayed CTL activity in response to HER263-71 but not to HER2342-350, whereas no such induction was observed in CT26/HER2 tumor-bearing mice. When 4T1.2/HER2 cell-challenged mice were depleted of CD8+ T cells, they lost their tumor-regressing activity, suggesting an antitumor role of HER263-71-specific CD8+ CTLs in the control of this tumor type. CT26/HER2 cells also expressed CD80. However, CD80-transfected 4T1.2/HER2 and CD80-non-expressing CT26/HER2 cells failed to alter their tumorigenicity, suggesting no role of CD80 in tumor control. Despite increased levels of myeloid-derived suppressor cells in the tumor, they were not associated with tumor progression in the CT26/HER2 model, as determined by a cell depletion assay. Overall, these data show that, contrary to CT26/HER2 tumors, 4T1.2/HER2 tumors regress via the induction of HER263-71-specific CD8+ CTLs and that CD80 is not associated with the regression of these tumors.
Collapse
Affiliation(s)
- Sayyed Nilofar Danishmalik
- BK21 Plus Graduate Program and Department of Microbiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, Korea
| | - Si-Hyeong Lee
- BK21 Plus Graduate Program and Department of Microbiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, Korea
| | - Jeong-Im Sin
- BK21 Plus Graduate Program and Department of Microbiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, Korea
| |
Collapse
|
6
|
Lo Iacono M, Cavallo F, Quaglino E, Rolla S, Iezzi M, Pupa SM, De Giovanni C, Lollini PL, Musiani P, Forni G, Calogero RA. A Limited Autoimmunity to p185neu Elicited by DNA and Allogeneic Cell Vaccine Hampers the Progression of Preneoplastic Lesions in HER-2/NEU Transgenic Mice. Int J Immunopathol Pharmacol 2016; 18:351-63. [PMID: 15888257 DOI: 10.1177/039463200501800217] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Prevention of the progression of precancerous lesions by vaccines is a virtually uncharted territory. Their potential, however, is being assessed in transgenic mice which develop autochthonous tumors with defined stages of progression. In this paper we show that the DNA micro-array technology significantly helps assessment of the preventive efficacy of a combined DNA and cell vaccine. All female rat Her-2/neu transgenic BALB/c (BALB-neuT) mice develop an invasive carcinoma in each of their mammary glands within 25 weeks of age. This is elicited by the activated transforming rat Her-2/neu oncogene embedded in their genome. We have previously shown that vaccination of mice bearing multiple in situ carcinomas with DNA plasmids which code for the extracellular and transmembrane domain of rat p185neu, the product of the rat Her-2/neu oncogene, followed by a boost with rat p185neu+ allogeneic cells engineered to secrete interferon-γ, keeps 48% of mice tumor free until week 32. We have now extended our follow-up until mice reach one year of age and show that protection vanishes as time progresses. This observation suggests that the accuracy of the results studying immunotherapy against life-threatening tumors is a function of the length of the follow-up. The application of microarrays, and the concordance of morphologic and gene expression data led us to identify antibody as the main mechanism induced by vaccination. Protection is associated with a break of tolerance and a limited autoimmunity against the
Collapse
Affiliation(s)
- M Lo Iacono
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Chotprakaikiat W, Allen A, Bui-Minh D, Harden E, Jobsri J, Cavallo F, Gleba Y, Stevenson FK, Ottensmeier C, Klimyuk V, Savelyeva N. A plant-expressed conjugate vaccine breaks CD4(+) tolerance and induces potent immunity against metastatic Her2(+) breast cancer. Oncoimmunology 2016; 5:e1166323. [PMID: 27471642 PMCID: PMC4938312 DOI: 10.1080/2162402x.2016.1166323] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 02/29/2016] [Accepted: 03/11/2016] [Indexed: 02/06/2023] Open
Abstract
Passive antibody therapy for cancer is an effective but costly treatment modality. Induction of therapeutically potent anticancer antibodies by active vaccination is an attractive alternative but has proven challenging in cancer due to tolerogenic pressure in patients. Here, we used the clinically relevant cancer target Her2, known to be susceptible to targeting by antibody therapy, to demonstrate how potent antibody can be induced by vaccination. A novel 44kD Her2 protein fragment was generated and found to be highly effective at inducing anti-Her2 antibody including trastuzumab-like reactivities. In the tolerant and spontaneous BALB-neuT mouse model of metastatic breast cancer this Her2-targeting vaccine was only effective if the fragment was conjugated to a foreign immunogenic carrier; Fragment C of tetanus toxin. Only the conjugate vaccine induced high affinity anti-Her2 antibody of multiple isotypes and suppressed tumor development. The magnitude of CD4(+) T-cell help and breadth of cytokines secreted by the CD4(+) T helper (Th) cells induced to the foreign antigen was critical. We used a highly efficient plant-based bio-manufacturing process for protein antigens, magnICON, for vaccine expression, to underpin feasibility of future clinical testing. Hence, our novel Her2-targeting conjugate vaccine combines preclinical efficacy with clinical deliverability, thus setting the scene for therapeutic testing.
Collapse
Affiliation(s)
| | - Alex Allen
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Elena Harden
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Jantipa Jobsri
- Oral Biology Department, Naresuan University, Phitsanulok, Thailand
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | | | - Freda K. Stevenson
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Christian Ottensmeier
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Natalia Savelyeva
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
8
|
Wei WZ, Jones RF, Juhasz C, Gibson H, Veenstra J. Evolution of animal models in cancer vaccine development. Vaccine 2015; 33:7401-7407. [PMID: 26241945 DOI: 10.1016/j.vaccine.2015.07.075] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 07/02/2015] [Indexed: 12/29/2022]
Abstract
Advances in cancer vaccine development are facilitated by animal models reflecting key features of human cancer and its interface with host immunity. Several series of transplantable preneoplastic and neoplastic mouse mammary lesions have been used to delineate mechanisms of anti-tumor immunity. Mimicking immune tolerance to tumor-associated antigens (TAA) such as HER2/neu, transgenic mice developing spontaneous mammary tumors are strong model systems for pre-clinical vaccine testing. In these models, HER2 DNA vaccines are easily administered, well-tolerated, and induce both humoral and cellular immunity. Although engineered mouse strains have advanced cancer immunotherapy, basic shortcomings remain. For example, multiple mouse strains have to be tested to recapitulate genetic regulation of immune tolerance in humans. Outbred domestic felines more closely parallel humans in the natural development of HER2 positive breast cancer and their varying genetic background. Electrovaccination with heterologous HER2 DNA induces robust adaptive immune responses in cats. Importantly, homologous feline HER2 DNA with a single amino acid substitution elicits unique antibodies to feline mammary tumor cells, unlocking a new vaccine principle. As an alternative approach to targeted vaccination, non-surgical tumor ablation such as cryoablation induces anti-tumor immunity via in situ immunization, particularly when combined with toll-like receptor (TLR) agonist. As strategies for vaccination advance, non-invasive monitoring of host response becomes imperative. As an example, magnetic resonance imaging (MRI) and positron emission tomography (PET) scanning following administration of tryptophan metabolism tracer [11C]-alpha-methyl-tryptophan (AMT) provides non-invasive imaging of both tumor growth and metabolic activities. Because AMT is a substrate of indoleamine-pyrrole 2,3-dioxygenase (IDO), an enzyme that produces the immune regulatory molecule kynurenine, AMT imaging can provide novel insight of host response. In conclusion, new feline models improve the predictive power of cancer immunotherapy and real-time PET imaging enables mechanistic monitoring of host immunity. Strategic utilization of these new tools will expedite cancer vaccine development.
Collapse
Affiliation(s)
- Wei-Zen Wei
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States.
| | - Richard F Jones
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States
| | - Csaba Juhasz
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States
| | - Heather Gibson
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States
| | - Jesse Veenstra
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, United States
| |
Collapse
|
9
|
Patel JM, Vartabedian VF, Kim MC, He S, Kang SM, Selvaraj P. Influenza virus-like particles engineered by protein transfer with tumor-associated antigens induces protective antitumor immunity. Biotechnol Bioeng 2015; 112:1102-10. [PMID: 25689082 PMCID: PMC4621003 DOI: 10.1002/bit.25537] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/18/2014] [Accepted: 12/30/2014] [Indexed: 01/06/2023]
Abstract
Delivery of antigen in particulate form using either synthetic or natural particles induces stronger immunity than soluble forms of the antigen. Among naturally occurring particles, virus-like particles (VLPs) have been genetically engineered to express tumor-associated antigens (TAAs) and have shown to induce strong TAA-specific immune responses due to their nano-particulate size and ability to bind and activate antigen-presenting cells. In this report, we demonstrate that influenza VLPs can be modified by a protein transfer technology to express TAAs for induction of effective antitumor immune responses. We converted the breast cancer HER-2 antigen to a glycosylphosphatidylinositol (GPI)-anchored form and incorporated GPI-HER-2 onto VLPs by a rapid protein transfer process. Expression levels on VLPs depended on the GPI-HER-2 concentration added during protein transfer. Vaccination of mice with protein transferred GPI-HER-2-VLPs induced a strong Th1 and Th2-type anti-HER-2 antibody response and protected mice against a HER-2-expressing tumor challenge. The Soluble form of GPI-HER-2 induced only a weak Th2 response under similar conditions. These results suggest that influenza VLPs can be enriched with TAAs by protein transfer to develop effective VLP-based subunit vaccines against cancer without chemical or genetic modifications and thus preserve the immune stimulating properties of VLPs for easier production of antigen-specific therapeutic cancer vaccines.
Collapse
MESH Headings
- Animals
- Antibodies, Neoplasm/blood
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Disease Models, Animal
- Drug Carriers
- Humans
- Immunity
- Mice
- Neoplasms/immunology
- Neoplasms/prevention & control
- Orthomyxoviridae/genetics
- Orthomyxoviridae/metabolism
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Receptor, ErbB-2/metabolism
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
- Jaina M. Patel
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322. USA
| | - Vincent F. Vartabedian
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322. USA
| | - Min-Chul Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
- Animal and Plant Quarantine Agency, Anyang City, Gyeonggi-do, Korea
| | - Sara He
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322. USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Periasamy Selvaraj
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322. USA
| |
Collapse
|
10
|
Gene Gun Her2/neu DNA Vaccination: Evaluation of Vaccine Efficacy in a Syngeneic Her2/neu Mouse Tumor Model. Methods Mol Biol 2015; 1317:17-37. [PMID: 26072399 DOI: 10.1007/978-1-4939-2727-2_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Genetic vaccination using naked plasmid DNA is an immunization strategy both against infectious diseases and cancer. In order to improve the efficacy of DNA vaccines, particularly in large animals and humans, different strategies have been pursued. These vaccination strategies are based on different application routes, schedules, and coexpression of immunomodulatory molecules as adjuvants. Our mouse tumor model offers the possibility to investigate Her2/neu DNA vaccines in different settings, i.e., intramuscular or intradermal application with or without coexpression of adjuvants. Protection from tumor growth in tumor challenge experiments and both T cell and humoral immune responses against Her2/neu peptides are used as surrogate parameters for vaccine efficacy.
Collapse
|
11
|
Kaumaya PTP, Foy KC. Peptide vaccines and targeting HER and VEGF proteins may offer a potentially new paradigm in cancer immunotherapy. Future Oncol 2012; 8:961-87. [PMID: 22894670 DOI: 10.2217/fon.12.95] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The ErbB family (HER-1, HER-2, HER-3 and HER-4) of receptor tyrosine kinases has been the focus of cancer immunotherapeutic strategies while antiangiogenic therapies have focused on VEGF and its receptors VEGFR-1 and VEGFR-2. Agents targeting receptor tyrosine kinases in oncology include therapeutic antibodies to receptor tyrosine kinase ligands or the receptors themselves, and small-molecule inhibitors. Many of the US FDA-approved therapies targeting HER-2 and VEGF exhibit unacceptable toxicities, and show problems of efficacy, development of resistance and unacceptable safety profiles that continue to hamper their clinical progress. The combination of different peptide vaccines and peptidomimetics targeting specific molecular pathways that are dysregulated in tumors may potentiate anticancer immune responses, bypass immune tolerance and circumvent resistance mechanisms. The focus of this review is to discuss efforts in our laboratory spanning two decades of rationally developing peptide vaccines and therapeutics for breast cancer. This review highlights the prospective benefit of a new, untapped category of therapies biologically targeted to EGF receptor (HER-1), HER-2 and VEGF with potential peptide 'blockbusters' that could lay the foundation of a new paradigm in cancer immunotherapy by creating clinical breakthroughs for safe and efficacious cancer cures.
Collapse
Affiliation(s)
- Pravin T P Kaumaya
- Departments of Obstetrics & Gynecology, OSU Wexner Medical Center, James Cancer Hospital & Solove Research Institute & the Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| | | |
Collapse
|
12
|
Baxevanis CN, Voutsas IF, Gritzapis AD, Perez SA, Papamichail M. HER-2/neu as a target for cancer vaccines. Immunotherapy 2010; 2:213-26. [PMID: 20635929 DOI: 10.2217/imt.09.89] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
A novel modality toward the treatment of HER-2/neu-positive malignancies, mostly including breast and, more recently prostate carcinomas, has been the use of vaccines targeting HER-2/neu extracellular and intracellular domains. HER-2/neu-specific vaccines have been demonstrated to generate durable T-cell anti-HER-2/neu immunity when tested in Phase I and II clinical trials with no significant toxicity or autoimmunity directed against normal tissues. Targeting of HER-2/neu in active immunotherapy may involve peptide and DNA vaccines. Moreover, active anti-HER-2/neu immunization could facilitate the ex vivo expansion of HER-2/neu-specific T cells for use in adoptive immunotherapy for the treatment of established metastatic disease. In addition, early data from trials examining the potential use of HER-2/neu-based vaccines in the adjuvant setting to prevent the relapse of breast cancer in high-risk patients have shown promising results. Future approaches include multiepitope preventive vaccines and combinatorial treatments for generating the most efficient protective anti-tumor immunity.
Collapse
|
13
|
Pakravan N, Soudi S, Hassan ZM. N-terminally fusion of Her2/neu to HSP70 decreases efficiency of Her2/neu DNA vaccine. Cell Stress Chaperones 2010; 15:631-8. [PMID: 20224916 PMCID: PMC3006617 DOI: 10.1007/s12192-010-0175-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Revised: 01/27/2010] [Accepted: 01/29/2010] [Indexed: 01/23/2023] Open
Abstract
DNA vaccines consisted of tumor-associated antigen (TAA) are well suited for immunotherapy against tumor. The construct can contain TAA fused to an appropriate molecule (biologic adjuvant) to improve the efficacy of anti-tumor immune response. Heat shock protein 70 (HSP70) has been shown to be an excellent candidate, capable of cross-priming TAA by antigen presenting cells leading to a robust T-cell response. However, the relationship between strong T-cell responses and tumor rejection is not always mutually exclusive, for which TAA loss or activation of suppressive mechanisms may occur. HSP70 fused to downstream of Her2/neu as DNA vaccine has been shown to be efficient against Her2-expressing tumors. In this study, we examined if N-terminally fusion of Her2/neu to HSP70 could also improve efficiency of Her2/neu DNA vaccine. Therefore, mice with an established Her2/neu expressing tumor were immunized with DNA vaccine consisting of extracellular and trans-membrane domain (EC+TM) of rat Her2/neu alone or N-terminally fused to HSP70 and immune response was evaluated. Administration of rat Her2/neu led to partial control of tumor progression. Surprisingly, fusion of HSP70 to N-terminal of rat Her2/neu led to tumor progression. Our result proposes that fusion direction of biologic adjuvant is an important consideration when Her2/neu is used.
Collapse
Affiliation(s)
- Nafiseh Pakravan
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Ale-Ahmad Avenue, P.O. Box 14115-331, Tehran, Iran.
| | | | | |
Collapse
|
14
|
Pakravan N, Soleimanjahi H, Hassan ZM. GP96 C-terminal improves Her2/neu DNA vaccine. J Gene Med 2010; 12:345-53. [PMID: 20232284 DOI: 10.1002/jgm.1445] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND DNA vaccines ensure protective immunity against tumors in a variety of experimental models. The favorite target tumor-associated antigens have been those that are frequently expressed by human tumors, such as Her2. However, the efficacy of active vaccination is limited because Her2 is a self-tolerated antigen. Many strategies have been applied to increase the efficacy of DNA vaccination, such as fusion or co-administration of Her2 with cytokine and co-stimulatory molecules. GP96 is involved in innate and adaptive immune responses and evokes potent activation and maturation of dendritic cells along with increased secretion of pro-inflammatory cytokines. On the basis of previous studies, we expected the C-terminal of GP96 to act as a package and as a suitable substitute for both cytokine and co-stimulatory genes. METHODS In the present study, the C-terminal of GP96 fused or co-administered with Her2/neu-containing constructs was used and the resultant immune response was evaluated and compared. RESULTS The data obtained showed that the construct containing the C-terminal of GP96 fused with Her2/neu, but not the co-administration of the two separated constructs, decreased CD4(+)CD25(+)foxp3(+) regulatory T cells at the tumor site, enhanced cytotoxic T lymphocyte activity and increased interferon-gamma secretion. CONCLUSIONS The C-terminal of GP96 has potent adjuvant activity in eliciting a significant immune response when fused with Her2/neu. It may be used as molecular adjuvant along with other tumor or bacterial/viral antigens.
Collapse
Affiliation(s)
- Nafiseh Pakravan
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | |
Collapse
|
15
|
Ladjemi MZ, Jacot W, Chardès T, Pèlegrin A, Navarro-Teulon I. Anti-HER2 vaccines: new prospects for breast cancer therapy. Cancer Immunol Immunother 2010; 59:1295-312. [PMID: 20532501 DOI: 10.1007/s00262-010-0869-2] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Accepted: 05/11/2010] [Indexed: 12/24/2022]
Abstract
Each year, breast cancer accounts for more than 400,000 new cancer cases and more than 130,000 cancer deaths in Europe. Prognosis of nonmetastatic breast cancer patients is directly related to the extent of the disease, mainly nodal spreading and tumor size, and to the molecular profile, particularly HER2 over-expression. In patients with HER2-over-expressing tumors, different studies have shown cellular and/or humoral immune responses against HER2 associated with a lower tumor development at early stages of the disease. These findings have led to the hypothesis that the generation of an anti-HER2 immune response should protect patients from HER2-over-expressing tumor growth. Taken together with the clinical efficiency of trastuzumab-based anti-HER2 passive immunotherapy, these observations allowed to envisage various vaccine strategies against HER2. The induction of a stable and strong immunity by cancer vaccines is expected to lead to establishment of immune memory, thereby preventing tumor recurrence. However, an immunological tolerance against HER2 antigen exists representing a barrier to effective vaccination against this oncoprotein. As a consequence, the current challenge for vaccines is to find the best conditions to break this immunological tolerance. In this review, we will discuss the different anti-HER2 vaccine strategies currently developed; considering the strategies having reached the clinical phases as well as those still in preclinical development. The used antigen can be either composed of tumoral allogenic cells or autologous cells, or specific to HER2. It can be delivered by dendritic cells or in a DNA, peptidic or proteic form. Another area of research concerns the use of anti-idiotypic antibodies mimicking HER2.
Collapse
Affiliation(s)
- Maha Zohra Ladjemi
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U896, Université Montpellier1, Montpellier, France
| | | | | | | | | |
Collapse
|
16
|
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
|
17
|
Jacob JB, Quaglino E, Radkevich-Brown O, Jones RF, Piechocki MP, Reyes JD, Weise A, Amici A, Wei WZ. Combining human and rat sequences in her-2 DNA vaccines blunts immune tolerance and drives antitumor immunity. Cancer Res 2010; 70:119-28. [PMID: 20048073 DOI: 10.1158/0008-5472.can-09-2554] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Immune tolerance to tumor-associated self-antigens poses a major challenge in the ability to mount an effective cancer vaccine response. To overcome immune tolerance to HER-2, we formulated DNA vaccines that express both human HER-2 and heterologous rat Neu sequences in separate plasmids or as single hybrid constructs that encode HER-2/Neu fusion proteins. Candidate vaccines were tested in Her-2 transgenic (Tg) mice of BALB/c (BALB), BALB/cxC57BL/6 F1 (F1), or C57BL/6 (B6) background, which exhibit decreasing immune responsiveness to HER-2. Analysis of various cocktails or hybrid vaccines defined a requirement for particular combination of HER/2/Neu sequences to effectively prime immune effector cells in HER-2 Tg mice. In B6 HER-2 Tg mice, rejection of HER-2-positive tumors protected mice from HER-2-negative tumors, providing evidence of epitope spreading. Our findings show that a strategy of combining heterologous antigen with self-antigens could produce a potent DNA vaccine that may be applicable to other tumor-associated antigens.
Collapse
Affiliation(s)
- Jennifer B Jacob
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan 48201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Ladjemi MZ, Jacot W, Pèlegrin A, Navarro-Teulon I. [Anti-HER2 vaccines: The HER2 immunotargeting future?]. ACTA ACUST UNITED AC 2009; 59:173-82. [PMID: 19481373 DOI: 10.1016/j.patbio.2009.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Accepted: 04/03/2009] [Indexed: 11/17/2022]
Abstract
Breast cancer is a widely spread women's disease. In spite of progress in the field of surgery and adjuvant therapies, the risk of breast cancer metastatic relapses remains high especially in those overexpressing HER2. Different studies have shown cellular and/or humoral immune responses against HER2 in patients with HER2-overexpressing tumors. This immune response is associated with a lower tumor development at early stages of the disease. These observations, associated with the efficiency today demonstrated by a trastuzumab-based anti-HER2 immunotherapy, allowed to envisage various vaccinal strategies against HER2. These findings have so led to the hypothesis that the generation of an anti-HER2 immune response should protect patients from HER2-overexpressing tumor growth, and induction of a stable and strong immunity by cancer vaccines is expected to lead to establishment of immune memory, thereby preventing tumor recurrence. However, an immunological tolerance against HER2 antigen exists representing a barrier to effective vaccination against this oncoprotein. As a consequence, the current challenge for vaccines is to find the best conditions to break this immunological tolerance. In this review, we will discuss the different anti-HER2 vaccine strategies currently developed; considering the strategies having reached the clinical phases as well as those still in preclinical development. The used antigen can be composed of tumoral allogenic cells or autologous cells or be specific of HER2. It can be delivered by denditric cells or in a DNA, peptidic or proteic form. Another area of the research concerns the use of anti-idiotypic antibodies mimicking HER2.
Collapse
Affiliation(s)
- M Z Ladjemi
- Inserm U896, CRLC Val-d'Aurelle Paul-Lamarque, institut de recherche en cancérologie de Montpellier (IRCM), université Montpellier-1, 34298 Montpellier cedex 5, France.
| | | | | | | |
Collapse
|
19
|
Rolla S, Marchini C, Malinarich S, Quaglino E, Lanzardo S, Montani M, Iezzi M, Angeletti M, Ramadori G, Forni G, Cavallo F, Amici A. Protective immunity against neu-positive carcinomas elicited by electroporation of plasmids encoding decreasing fragments of rat neu extracellular domain. Hum Gene Ther 2008; 19:229-40. [PMID: 18269312 DOI: 10.1089/hum.2006.196] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have shown that electroporation of plasmid carrying extracellular and transmembrane domains (EC-TM plasmid) encoded by the rat neu oncogene triggers a protective immune response toward rat p185(neu)-positive tumors in both wild-type BALB/c mice and cancer-prone rat neu-transgenic BALB-neuT mice. To identify the critical fragments that confer this protective immunity, mice were electroporated with plasmids encoding the TM domain associated with decreasing fragments of the EC domain and the antitumor protection afforded, the titer of antibody, and cytotoxic T lymphocyte (CTL) activity elicited to Neu protein were evaluated. Plasmids encoding EC fragments shortened by 70 (EC1-TM plasmid), 150 (EC2-TM), 230 (EC3-TM), 310 (EC4-TM), and 390 (EC5-TM) NH(2)-terminal residues afforded effective protection. Plasmids encoding shorter truncated proteins were ineffective. When the immunogenic protein was retained in the cytoplasm (EC1-TM, EC2-TM, and EC5-TM), only a CTL response was elicited, whereas when it was also expressed on the membrane (EC4-TM) both CTLs and antibodies were induced. EC4-TM encoding a truncated protein with an EC portion of only 344 amino acids conferred protection on both BALB/c and BALB-neuT mice comparable to that of EC-TM.
Collapse
Affiliation(s)
- Simona Rolla
- Molecular Biotechnology Center, Dipartimento di Scienze Cliniche e Biologiche, University of Turin, 10123 Turin, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Felicetti P, Mennecozzi M, Barucca A, Montgomery S, Orlandi F, Manova K, Houghton AN, Gregor PD, Concetti A, Venanzi FM. Tumor endothelial marker 8 enhances tumor immunity in conjunction with immunization against differentiation Ag. Cytotherapy 2008; 9:23-34. [PMID: 18236207 DOI: 10.1080/14653240601048369] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND We have previously shown that xenogeneic DNA vaccines encoding rat neu and melanosomal differentiation Ag induce tumor immunity. Others have developed vaccines targeting tumor neovasculature. Tumor endothelial marker 8 (TEM8) is expressed in the neovasculature of human tumors, and in the mouse melanoma B16, but its expression is limited in normal adult tissues. We describe a DNA vaccine combining xenogeneic tumor Ag and TEM8. METHODS In-situ hybridization was used to detect TEM8 RNA in mouse tumors. Mice transgenic for the rat neu proto-oncogene were immunized with DNA vaccines encoding TEM8 and the extracellular domain of rat neu and challenged with the 233-VSGA1 breast cancer cell line. In parallel experiments, C57BL/6 mice were immunized with TEM8 and human tyrosinase-related protein 1 (hTYRP1/hgp75) and challenged with B16F10 melanoma. RESULTS TEM8 was expressed in the stroma of transplantable mouse breast and melanoma tumors. In both model systems, TEM8 DNA had no activity as a single agent but significantly enhanced the anti-tumor immunity of neu and hTYRP1/hgp75 DNA vaccines when given in concert. The observed synergy was dependent upon CD8+ T cells, as depletion of this cell population just prior to tumor challenge obviated the effect of the TEM8 vaccine in both tumor models. DISCUSSION A local immune response to TEM8 may increase inflammation or tumor necrosis within the tumor, resulting in improved Ag presentation of HER2/neu and hTYRP1/hgp75. Alternatively, TEM8 expression in host APC may alter T-cell interactions or homing. In this way, TEM8 may act more as an adjuvant than an immunologic target.
Collapse
Affiliation(s)
- P Felicetti
- Swim Across America Laboratory of Tumor Immunology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Wei WZ, Jacob J, Radkevich-Brown O, Whittington P, Kong YCM. The "A, B and C" of Her-2 DNA vaccine development. Cancer Immunol Immunother 2008; 57:1711-7. [PMID: 18273615 DOI: 10.1007/s00262-008-0464-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Accepted: 01/24/2008] [Indexed: 11/26/2022]
Abstract
INTRODUCTION The development of Her-2 DNA vaccine has progressed through three phases that can be categorized as phase "A": the pursuit of Her-2 as a tumor-associated "antigen", phase "B": tilting the "balance" between tumor immunity and autoimmunity and phase "C": the on-going "clinical trials". MATERIALS AND METHODS In phase "A", a panel of human ErbB-2 or Her-2 plasmids were constructed to encode non-transforming Her-2 derivatives. The immunogenicity and anti-tumor activity of Her-2 DNA vaccines were tested in human Her-2 transgenic mice with or without the depletion of regulatory T cells (Tregs). However, Treg depletion or other immune modulating regimens may increase the risk of autoimmunity. In phase "B", the balance between tumor immunity and autoimmunity was assessed by monitoring the development of experimental autoimmune thyroiditis (EAT). To test the efficacy of Her-2 DNA vaccines in cancer patients, clinical trials have been initiated in phase "C". RESULTS AND CONCLUSIONS Significant anti-Her-2 and anti-tumor activity was observed when Her-2 transgenic mice were electro-vaccinated after Treg depletion. Susceptibility to EAT was also enhanced by Treg depletion and there was mutual amplification between Her-2 immunity and EAT development. Although Tregs regulate both EAT and Her-2 immunity, their effector mechanisms may differ. It may be possible to amplify tumor immunity with improved strategies that can by-pass undue autoimmunity. Critical information will be revealed in the next decade to expedite the development of cancer vaccines.
Collapse
Affiliation(s)
- Wei-Zen Wei
- Karmanos Cancer Institute and Department of Immunology and Microbiology, Wayne State University, 110 E. Warren Ave, Detroit, MI 48201, USA.
| | | | | | | | | |
Collapse
|
22
|
Cho HJ, Shin HJ, Han IK, Jung WW, Kim YB, Sul D, Oh YK. Induction of mucosal and systemic immune responses following oral immunization of mice with Lactococcus lactis expressing human papillomavirus type 16 L1. Vaccine 2007; 25:8049-57. [PMID: 17936447 DOI: 10.1016/j.vaccine.2007.09.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2007] [Revised: 08/04/2007] [Accepted: 09/06/2007] [Indexed: 12/21/2022]
Abstract
Human papillomavirus type 16 L1 (HPV16 L1) has shown considerable promise as a parenteral vaccine for prevention of cervical cancers. Here, we report the possibility of oral vaccination for HPV16 L1 using Lactococcus lactis (L. lactis) as a live vector. L. lactis MG1363 was transformed with two types of HPV16 L1-encoding plasmids for intracellular expression or secretion. L. lactis transformed with HPV16 L1-encoding plasmids retained biochemical lactic acid production capability. The mucosal and systemic immune responses were affected by the cellular location of expressed HPV16 L1 proteins in L. lactis. Serum IgG responses were induced after oral immunizations of L. lactis secreting HPV16 L1. Vaginal IgA immune responses were observed following oral immunization with L. lactis expressing HPV16 L1 in an intracellular form, but not with L. lactis secreting HPV16 L1. Furthermore, induction of HPV16 L1-specific mucosal immune responses was affected by immunization frequency. Six immunizations over 5 weeks were required to induce vaginal immune responses. The levels of HPV16 L1-specific vaginal IgA were maintained until 12 weeks after the first vaccination. These results suggest the feasibility of L. lactis as an oral vaccine vehicle of HPV16 L1 and demonstrate the importance of cellular loci of expressed antigen for induction of vaginal and systemic immune responses.
Collapse
Affiliation(s)
- Hee-Jeong Cho
- School of Life Sciences and Biotechnology, Korea University, Anam-dong, Seongbuk-gu, Seoul 136-713, South Korea
| | | | | | | | | | | | | |
Collapse
|
23
|
Garrett JT, Rawale S, Allen SD, Phillips G, Forni G, Morris JC, Kaumaya PTP. Novel engineered trastuzumab conformational epitopes demonstrate in vitro and in vivo antitumor properties against HER-2/neu. THE JOURNAL OF IMMUNOLOGY 2007; 178:7120-31. [PMID: 17513761 DOI: 10.4049/jimmunol.178.11.7120] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Trastuzumab is a growth-inhibitory humanized Ab targeting the oncogenic protein HER-2/neu. Although trastuzumab is approved for treatment of advanced breast cancer, a number of concerns exist with passive immunotherapy. Treatment is expensive and has a limited duration of action, necessitating repeated administrations of the mAb. Active immunotherapy with conformational B cell epitopes affords the possibility of generating an enduring immune response, eliciting protein-reactive high-affinity anti-peptide Abs. The three-dimensional structure of human HER-2 in complex with trastuzumab reveals that the Ag-binding region of HER-2 spans residues 563-626 that comprises an extensive disulfide-bonding pattern. To delineate the binding region of HER-2, we have designed four synthetic peptides with different levels of conformational flexibility. Chimeric peptides incorporating the measles virus fusion "promiscuous" T cell epitope via a four-residue linker sequence were synthesized, purified, and characterized. All conformational peptides were recognized by trastuzumab and prevented the function of trastuzumab inhibiting tumor cell proliferation, with 563-598 and 597-626 showing greater reactivity. All epitopes were immunogenic in FVB/N mice with Abs against 597-626 and 613-626 recognizing HER-2. The 597-626 epitope was immunogenic in outbred rabbits eliciting Abs which recognized HER-2, competed with trastuzumab for the same epitope, inhibited proliferation of HER-2-expressing breast cancer cells in vitro and caused their Ab-dependent cell-mediated cytotoxicity. Moreover, immunization with the 597-626 epitope significantly reduced tumor burden in transgenic BALB-neuT mice. These results suggest the peptide B cell immunogen is appropriate as a vaccine for HER-2-overexpressing cancers because the resulting Abs show analogous biological properties to trastuzumab.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents/chemical synthesis
- Antineoplastic Agents/therapeutic use
- Cancer Vaccines/chemical synthesis
- Cancer Vaccines/therapeutic use
- Cell Line, Tumor
- Epitopes/chemistry
- Epitopes/therapeutic use
- Female
- Humans
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Molecular Sequence Data
- Peptide Fragments/chemical synthesis
- Peptide Fragments/therapeutic use
- Protein Conformation
- Protein Engineering
- Rabbits
- Rats
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/immunology
- Trastuzumab
Collapse
Affiliation(s)
- Joan T Garrett
- Departments of Obstetrics and Gynecology, Arthur G. James Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Lin CC, Tu CF, Yen MC, Chen MC, Hsieh WJ, Chang WC, Chang WT, Lai MD. Inhibitor of heat-shock protein 90 enhances the antitumor effect of DNA vaccine targeting clients of heat-shock protein. Mol Ther 2007; 15:404-10. [PMID: 17235320 DOI: 10.1038/sj.mt.6300014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Geldanamycin (GA), a heat-shock protein (HSP) 90 inhibitor, induces degradation of HSP90 client proteins, which may promote the presentation of degradation peptides with major histocompatibility complex class I on cancer cells. We hypothesized that GA may enhance the efficacy of DNA vaccination, and investigated the therapeutic effect of the combination of GA and a DNA vaccine against HSP90 clients p185(neu) and Met. The efficacy of various doses of GA combined with an N-terminal neu (N'-neu) DNA vaccine was investigated in a transplanted tumor constitutively overexpressing endogenous p185(neu). Low-dose (2.5 mug) but not high-dose (10 microg) GA enhanced the effect of N'-neu DNA vaccination on the inhibition of murine bladder tumor-2 tumors in syngeneic C3H mice. Anti-p185(neu) antibody titers were similar among all treated groups. Significantly increased infiltrations of CD8(+) T cells and NK cells were observed at tumor sites. GA sensitized tumor cells to the cytotoxic effects of lymphocytes. Depletion of CD8(+) T cells eliminated most of the therapeutic efficacy; in contrast, depletion of CD4(+) T cells enhanced the therapeutic efficacy. A similar enhancing effect was observed for the combination of GA and a DNA vaccine targeting the Met oncogene. Our results support the use of combination of GA and DNA vaccination against GA-targeted proteins.
Collapse
Affiliation(s)
- Chi-Chen Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Gallo P, Dharmapuri S, Nuzzo M, Maldini D, Cipriani B, Forni G, Monaci P. Adenovirus vaccination against neu oncogene exerts long-term protection from tumorigenesis in BALB/neuT transgenic mice. Int J Cancer 2007; 120:574-84. [PMID: 17096348 DOI: 10.1002/ijc.22274] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The transforming rat HER2/neu oncogene (neu), when embedded in the genome of transgenic BALB/c (neuT) mice, provokes the development of an invasive carcinoma in each of their 10 mammary glands. We used the neuT mice model system to evaluate the immunization efficiency and the protective effect of intramuscular injection of adenovirus (Ad) and/or of DNA with electrostimulation (DNA+ES), both expressing the rat p185(neu) protein. A neu cDNA sequence, which exclusively contains codons preferred by highly expressed mammalian genes, was used in this study. This "optimized" cDNA displayed higher expression in cultured cells and greater cell-mediated response than the original gene when injected as DNA+ES. Ad expressing the optimized sequence (Ad5-neu.opt) induced a higher immune response, as measured by the frequency of IFN-gamma-secreting spleen cells and antibody titers. Different Ad/DNA combinations and immunization schedules confirmed the superiority of Ad5-neu.opt in inducing a strong Th1-skewed humoral and CD8(+) cell-mediated response. Two Ad5-neu.opt injections of 10(9) viral particles at week 10 and 12 were sufficient to induce the highest response, which persisted at detectable levels up to 33 weeks of age. Anti-Ad5 antibodies elicited by previous injections neutralized the effect of an additional Ad5-neu.opt immunization at week 19. A group, which received 3 injections of DNA+ES at week 23, 27 and 31, in addition to the 3 Ad injections at week 10, 12 and 19 showed an increased frequency of IFN-gamma(+), CD8(+) PBMC at week 25, which persisted at detectable levels till week 38. Ad5-neu.opt administration at 10 and 12 weeks of age had a significant impact on tumor progression. At 44 weeks, 40% of the mice were completely protected from tumors with a mean tumor of 3.8. In contrast, control mice developed 10 tumors and died by week 27. Vaccination blocked the tumor development at the atypical hyperplasia stage present at the time of treatment. Tumors developing at later times express reduced levels of rat p185(neu) protein.
Collapse
MESH Headings
- Adenoviridae/genetics
- Amino Acid Sequence
- Animals
- Cell Line
- Cell Line, Tumor
- Codon/genetics
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Genetic Vectors/administration & dosage
- Genetic Vectors/genetics
- Humans
- Injections, Intramuscular
- Mammary Glands, Animal/chemistry
- Mammary Glands, Animal/immunology
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Molecular Sequence Data
- NIH 3T3 Cells
- Proliferating Cell Nuclear Antigen/analysis
- Rats
- Receptor, ErbB-2/analysis
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Time Factors
- Vaccination/methods
Collapse
Affiliation(s)
- Pasquale Gallo
- Molecular and Cell Biology Department, I.R.B.M.P. Angeletti, Pomezia, Roma, Italy
| | | | | | | | | | | | | |
Collapse
|
26
|
Rolla S, Nicoló C, Malinarich S, Orsini M, Forni G, Cavallo F, Ria F. Distinct and non-overlapping T cell receptor repertoires expanded by DNA vaccination in wild-type and HER-2 transgenic BALB/c mice. THE JOURNAL OF IMMUNOLOGY 2007; 177:7626-33. [PMID: 17114432 DOI: 10.4049/jimmunol.177.11.7626] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Central tolerance to tumor-associated Ags is an immune-escape mechanism that significantly limits the TCR repertoires available for tumor eradication. The repertoires expanded in wild-type BALB/c and rat-HER-2/neu (rHER-2) transgenic BALB-neuT mice following DNA immunization against rHER-2 were compared by spectratyping the variable (V)beta and the joining (J)beta CDR 3. Following immunization, BALB/c mice raised a strong response. Every mouse used one or more CD8+ T cell rearrangements of the Vbeta9-Jbeta1.2 segments characterized by distinct length of the CDR3 and specific for 63-71 or 1206-1214 rHER-2 peptides. In addition, two CD4+ T cell rearrangements recurred in >50% of mice. Instead, BALB-neuT mice displayed a limited response to rHER-2. Their repertoire is smaller and uses different rearrangements confined to CD4+ T cells. Thus, central tolerance in BALB-neuT mice acts by silencing the BALB/c mice self-reactive repertoire and reducing the size of the CD8+ T cell component. CD8+ and CD4+ T cells from both wild-type and transgenic mice home to tumors. This definition of the T cell repertoires available is critical to the designing of immunological maneuvers able to elicit an effective immune reaction against HER-2-driven carcinogenesis.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Neoplasm/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Female
- Gene Rearrangement, beta-Chain T-Cell Antigen Receptor
- Genes, T-Cell Receptor beta
- Immune Tolerance
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- RNA, Messenger/analysis
- Receptor, ErbB-2/immunology
- Receptors, Antigen, T-Cell/immunology
- Recombinant Proteins/immunology
- Reverse Transcriptase Polymerase Chain Reaction
- Transfection
- Tumor Escape
- Vaccines, DNA
Collapse
Affiliation(s)
- Simona Rolla
- Dipartimento di Scienze Cliniche e Biologiche, University of Turin, Turin, Italy
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Significant progress made in the field of tumor immunology by the characterization of a large number of tumor antigens, and the better understanding of the mechanisms preventing immune responses to malignancies has led to the extensive study of cancer immunization approaches such as DNA vaccines encoding tumor antigens. This article reviews major aspects of DNA immunization in cancer. It gives a brief history and then discusses the proposed mechanism of action, preclinical and clinical studies, and methods of enhancing the immune responses induced by DNA vaccines.
Collapse
Affiliation(s)
- Rodica Stan
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | |
Collapse
|
28
|
Jacob J, Radkevich O, Forni G, Zielinski J, Shim D, Jones RF, Wei WZ. Activity of DNA vaccines encoding self or heterologous Her-2/neu in Her-2 or neu transgenic mice. Cell Immunol 2006; 240:96-106. [PMID: 16930573 DOI: 10.1016/j.cellimm.2006.07.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2006] [Revised: 06/30/2006] [Accepted: 07/05/2006] [Indexed: 11/20/2022]
Abstract
To assess the efficacy of self versus heterologous ErbB-2 vaccines, the reactivity to human and rat ErbB-2 (Her-2 and neu, respectively) DNA vaccines were tested in normal, Her-2 or neu transgenic mice. When immunized with either Her-2 or neu DNA, normal BALB/c and C57BL/6 mice produced cross-reactive T cells, but only antigen specific antibodies. In Her-2 Tg mice, weak to no anti-Her-2 response was induced by either self Her-2 or heterologous neu DNA, demonstrating profound tolerance to Her-2 and the inability to induce anti-Her-2 immunity with either vaccine. In NeuT mice, vaccination with self neu but not heterologous Her-2 DNA induced anti-neu antibodies and delayed spontaneous tumorigenesis. Both neu and Her-2 DNA induced anti-neu T cell response, but depletion of CD8 T cells did not change the delay in tumorigenesis. Therefore, in NeuT mice, both self and heterologous DNA activated anti-neu T cells, although T cell response did not reach sufficient level to suppress spontaneous tumorigenesis. Rather, induction of anti-neu antibodies by self neu DNA is associated with the delay in spontaneous tumor growth. Overall, NeuT mice were more responsive to DNA vaccination than Her-2 Tg mice and this may be associated with the continuous production of neu by the 10 mammary glands undergoing tumor progression.
Collapse
MESH Headings
- Animals
- Antibody Formation/immunology
- Antigens/immunology
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Cross Reactions/immunology
- Female
- Humans
- Immune Tolerance/immunology
- Interferon-gamma/metabolism
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Rats
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Jennifer Jacob
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Chan T, Sami A, El-Gayed A, Guo X, Xiang J. HER-2/neu-gene engineered dendritic cell vaccine stimulates stronger HER-2/neu-specific immune responses compared to DNA vaccination. Gene Ther 2006; 13:1391-402. [PMID: 16724093 DOI: 10.1038/sj.gt.3302797] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
HER-2/neu is a candidate for developing breast cancer-targeted immunotherapeutics. Although DNA-based and HER-2/neu transgene-modified dendritic cell (DC)-based vaccines are potent at eliciting HER-2/neu-specific antitumor immunity, there has been no side-by-side study comparing them directly. The present study utilizes an in vivo murine tumor model expressing HER-2/neu antigen to compare the efficacy between adenovirus (AdVneu)-transfected dendritic cells (DC(neu)) and plasmid DNA (pcDNAneu) vaccine. Our data showed that DC(neu) upregulated the expression of immunologically important molecules and inflammatory cytokines and partially converted regulatory T (Tr)-cell suppression through interleukin-6 (IL-6) secretion. Vaccination of DC(neu) induced stronger HER-2/neu-specific humoral and cellular immune responses than DNA vaccination, which downregulated HER-2/neu expression and lysed HER-2/neu-positive tumor cells in vitro, respectively. In two HER-2/neu-expressing tumor models, DC(neu) completely protected mice from tumor cell challenge compared to partial or no protection observed in DNA-immunized mice. In addition, DC(neu) significantly delayed breast cancer development in transgenic mice in comparison to DNA vaccine (P<0.05). Taken together, we have demonstrated that HER-2/neu-gene-modified DC vaccine is more potent than DNA vaccine in both protective and preventive animal tumor models. Therefore, DCs genetically engineered to express tumor antigens such as HER-2/neu represent a new direction in DC vaccine of breast cancer.
Collapse
Affiliation(s)
- T Chan
- Cancer Research Unit, Saskatchewan Cancer Agency, University of Saskatchewan, 20 Campus Drive, Saskatoon, Saskatchewan, Canada S7N 4H4
| | | | | | | | | |
Collapse
|
30
|
Zhang H, Knutson KL, Hellstrom KE, Disis ML, Hellstrom I. Antitumor efficacy of CD137 ligation is maximized by the use of a CD137 single-chain Fv-expressing whole-cell tumor vaccine compared with CD137-specific monoclonal antibody infusion. Mol Cancer Ther 2006; 5:149-55. [PMID: 16432173 DOI: 10.1158/1535-7163.mct-05-0206] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tumor-destructive immune responses can be generated by engaging CD137 (4-1BB) via infusing a monoclonal antibody specific for CD137 or vaccinating with a single-chain Fv (scFv) CD137-expressing whole-cell tumor vaccine. We assessed whether such a vaccine can induce tumor rejection in the neu-transgenic (neu-Tg) mouse breast cancer model and compared the antitumor efficacy of vaccination with the infusion of a CD137-specific antibody. Mammary carcinoma cells (MMC) from a neu-Tg mouse were transfected to stably express surface scFv derived from the anti-CD137 rat hybridoma 1D8 or 3H3. The anti-CD137 scFv-expressing cells were rejected when transplanted into neu-Tg mice by a mechanism that involved both CD4(+) and CD8(+) T cells, and vaccination with such cells delayed the outgrowth of MMC cells transplanted 3 days previously. T cells from neu-Tg mice that had been vaccinated proliferated and produced IFN-gamma when stimulated by MMC but not by antigen-negative variant breast cancer cells that did not express the neu tumor antigen. In addition, antibodies binding to the MMC but not to antigen-negative variant cells were detected in sera from some but not all of the immunized mice. Complete regression of s.c. transplanted MMC tumors was observed in mice repeatedly immunized against MMC-1D8 starting on the day the MMC cells were transplanted. In contrast, repeated administration of either of two different anti-CD137 monoclonal antibodies did not induce complete tumor regression, although tumor growth was delayed.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antigens, CD/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/immunology
- Cancer Vaccines/pharmacology
- Cell Transplantation
- Female
- Graft Rejection/immunology
- Immunoglobulin Fragments/immunology
- Infusions, Parenteral
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/therapy
- Mice
- Mice, Inbred Strains
- Mice, Transgenic
- Receptors, Nerve Growth Factor/immunology
- Receptors, Tumor Necrosis Factor/immunology
- T-Lymphocytes/immunology
- Tumor Necrosis Factor Receptor Superfamily, Member 9
Collapse
Affiliation(s)
- Hongtao Zhang
- Department of Pathology, Center for Translational Medicine in Women's Health, University of Washington, Seattle, USA
| | | | | | | | | |
Collapse
|
31
|
Smorlesi A, Papalini F, Amici A, Orlando F, Pierpaoli S, Mancini C, Provinciali M. Evaluation of different plasmid DNA delivery systems for immunization against HER2/neu in a transgenic murine model of mammary carcinoma. Vaccine 2006; 24:1766-75. [PMID: 16288939 DOI: 10.1016/j.vaccine.2005.10.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2005] [Revised: 09/15/2005] [Accepted: 10/10/2005] [Indexed: 11/18/2022]
Abstract
Studies of DNA vaccination against HER2/neu showed the effectiveness of immunization protocols in models of transplantable or spontaneous tumors; scarce information, however, has been provided to identify the procedure of DNA administration that more effectively contributes to the activation of immune system against spontaneously arising HER2/neu-positive tumors. We compared the effectiveness of different procedures of DNA vaccine delivery (intradermic injection (ID), gene gun (GG) delivery and intramuscular injection (IM) alone or with electroporation) in a murine transgenic model of mammary carcinoma overexpressing HER2/neu. We highlighted the role of DNA delivery system in the success of DNA vaccination showing that, among the analysed methods, intramuscular injection of the vaccine, particularly when associated to electroporation, elicits a better protection against HER2/neu spontaneous tumor development inducing antibody and cell-mediated immune responsiveness against HER2/neu and a Th1 polarization of the immune response.
Collapse
MESH Headings
- Animals
- Antibodies, Neoplasm/blood
- Biolistics
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cytokines/analysis
- Cytotoxicity Tests, Immunologic
- Disease Models, Animal
- Electroporation
- Female
- Genes, erbB-2
- Immunization, Passive
- Injections, Intradermal
- Injections, Intramuscular
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Mice, Transgenic
- Plasmids
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Arianna Smorlesi
- Laboratory of Tumor Immunology, Immunology Center, INRCA Gerontology Research Department, Via Birarelli 8, 60121 Ancona, Italy
| | | | | | | | | | | | | |
Collapse
|
32
|
Williams JA, Luke J, Johnson L, Hodgson C. pDNAVACCultra vector family: high throughput intracellular targeting DNA vaccine plasmids. Vaccine 2005; 24:4671-6. [PMID: 16448726 DOI: 10.1016/j.vaccine.2005.08.033] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
DNA vaccines have the potential to provide a safe route for protective immunity to neoplasms and infectious agents. However, current DNA vaccine plasmids are not optimal with additional non-essential DNA, nor do they facilitate controlled or flexible targeting of antigens to various intracellular destinations. A family of DNA vaccine vectors, optimized and minimized to comply with FDA guidelines regarding content and elimination of extraneous materials, was constructed. The resulting vectors are much smaller than existing vectors, drive higher levels of target gene expression, facilitate high throughput cloning applications, and allow simultaneous cloning into multiple vectors that feature various intracellular targeting destinations for the protein product. The ability to control expression and trafficking is intended to provide a rapid, rational approach to cancer therapy and emerging infectious diseases.
Collapse
Affiliation(s)
- James A Williams
- Nature Technology Corporation, 4701 Innovation Drive, Lincoln, NE 68521, USA.
| | | | | | | |
Collapse
|
33
|
Astolfi A, Landuzzi L, Nicoletti G, De Giovanni C, Croci S, Palladini A, Ferrini S, Iezzi M, Musiani P, Cavallo F, Forni G, Nanni P, Lollini PL. Gene expression analysis of immune-mediated arrest of tumorigenesis in a transgenic mouse model of HER-2/neu-positive basal-like mammary carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:1205-16. [PMID: 15793299 PMCID: PMC1602398 DOI: 10.1016/s0002-9440(10)62339-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We previously showed that a vaccine combining interleukin 12 and allogeneic p185(neu)-positive mammary carcinoma cells completely prevented multifocal mammary carcinogenesis in HER-2/neu transgenic mice. To identify the molecular events responsible for effective tumor prevention and to define the tumor gene expression signature, we used microarrays to analyze the expression profile of mammary tissue of untreated transgenic mice and of vaccine-treated, tumor-free mice at different time points. Mammary tissue from vaccinated mice displayed a gene expression profile different from that of untreated, tumor-bearing mice but similar to that of normal/hyperplastic mammary gland. Comparison of treated and untreated mice at 15 weeks of age revealed up-regulation of genes encoding antibodies, chemokines, gamma-interferon-induced genes and inflammatory molecules, and down-regulation of early genes induced by tumor development. The gene expression signature of HER-2/neu-transformed tumor cells showed modulation of genes promoting proliferation, angiogenesis, migration, invasion, and metastasis and inhibiting apoptosis and immune response. Meta-analysis of microarray data on human breast cancer showed that the signature of tumors arising in murine HER-2/neu transgenic model correctly classified human HER-2/neu-expressing tumors and normal breast tissue. Moreover murine and human HER-2/neu-positive tumors share the signature of basal-like breast cancers. This gene expression analysis reveals the immune events associated with prevention of tumor development and shows that HER-2/neu transgenic mice represent a good model of a poor-prognosis group of human breast tumors.
Collapse
Affiliation(s)
- Annalisa Astolfi
- Department of Experimental Pathology, Cancer Research Section, University of Bologna, viale Filopanti 22, I-40126 Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Viehl CT, Becker-Hapak M, Lewis JS, Tanaka Y, Liyanage UK, Linehan DC, Eberlein TJ, Goedegebuure PS. A tat fusion protein-based tumor vaccine for breast cancer. Ann Surg Oncol 2005; 12:517-25. [PMID: 15889213 DOI: 10.1245/aso.2005.06.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2004] [Accepted: 02/03/2005] [Indexed: 11/18/2022]
Abstract
BACKGROUND We recently reported that dendritic cells (DCs) transduced with a fusion protein between Her2/neu and the protein transduction domain Tat (DC-Tat-extracellular domain [ECD]) induced Her2/neu-specific CD8(+) T cells in vitro. This study tested the in vivo efficacy of DC-Tat-ECD in a murine breast cancer model. METHODS FVB/N mice received one or two weekly intraperitoneal immunizations with syngeneic DC-Tat-ECD followed by a tumor challenge with syngeneic neu(+) breast cancer cells, and tumor development was monitored. To test for Her2/neu specificity, CD4(+) and CD8(+) cells were isolated through magnetic bead separation and analyzed for specific interferon gamma release. RESULTS Intraperitoneally injected DCs migrated to secondary lymphoid organs, as evidenced by small-animal positron emission tomography studies. Immunized mice developed palpable tumors significantly later than control mice injected with DC-Tat-empty (P = .001 and P < .05 for two immunizations and for one immunization, respectively) or mice that received no DCs (P = .001 and P < .05). Similarly, immunized mice had smaller resulting tumors than mice injected with DC-Tat-empty (P < .05 and P < .01) or untreated mice (P < .001 and P < .001). Significantly more tumor-specific CD8(+) splenocytes were found in twice-immunized mice than in untreated animals (P < .001). Similarly, a T-helper type 1 CD4(+) T-cell response was observed. CONCLUSIONS Protein-transduced DCs may be effective vaccines for the treatment of cancer.
Collapse
Affiliation(s)
- Carsten T Viehl
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Quaglino E, Mastini C, Iezzi M, Forni G, Musiani P, Klapper LN, Hardy B, Cavallo F. The adjuvant activity of BAT antibody enables DNA vaccination to inhibit the progression of established autochthonous Her-2/neu carcinomas in BALB/c mice. Vaccine 2005; 23:3280-7. [PMID: 15837233 DOI: 10.1016/j.vaccine.2005.01.086] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2004] [Revised: 01/03/2005] [Accepted: 01/05/2005] [Indexed: 11/29/2022]
Abstract
Over the course of 33 weeks from birth, the mammary glands of virgin female BALB/c mice transgenic for the transforming rat Her-2/neu oncogene progress from atypical hyperplasia to invasive carcinoma. By week 12, all their mammary glands display many foci of in situ carcinoma. DNA vaccination at weeks 12 and 14 through in vivo electroporation of a plasmid encoding for the extracellular and transmembrane domain of the protein product of rat Her-2/neu oncogene kept 33% of mice tumor-free until week 35, when the experiment ended. To improve its efficacy the vaccine was combined with a T cell stimulatory monoclonal antibody (BAT). When each plasmid electroporation was followed by intravenous administration of 10 microg of BAT monoclonal antibody at weeks 13 and 15, 55% of mice remained tumor free (p < 0.0001) and stronger T cell and antibody-mediated immune responses were elicited. These data suggest that costimulation by BAT monoclonal antibody enables DNA vaccination to establish an effective protection against incipient carcinomas.
Collapse
Affiliation(s)
- Elena Quaglino
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Jia ZC, Zou LY, Ni B, Wan Y, Zhou W, Lv YB, Geng M, Wu YZ. Effective induction of antitumor immunity by immunization with plasmid DNA encoding TRP-2 plus neutralization of TGF-beta. Cancer Immunol Immunother 2005; 54:446-52. [PMID: 15750831 PMCID: PMC11032863 DOI: 10.1007/s00262-004-0619-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2004] [Accepted: 09/08/2004] [Indexed: 11/29/2022]
Abstract
Plasmid DNA vaccine is an appealing cancer immunotherapy. However, it is a weak immunogen and immunization with plasmid DNA encoding self-antigens, such as melanoma-associated antigens, could not induce antitumor immunity because of tolerance. In this study, we investigated the feasibility of using a plasmid DNA encoding Xenopus laevis transforming growth factor-beta 5 (aTGF-beta5) as an immunogen to induce neutralizing antibodies against murine TGF-beta1 (mTGF-beta1) and thus enhance the efficacy of plasmid DNA vaccine encoding murine tyrosinase-related protein 2 (mTRP-2) through neutralization of TGF-beta. The results showed that immunization with aTGF-beta5 resulted in the generation of mTGF-beta1-neutralizing antibodies, and immunization with a combination of aTGF-beta5 and mTRP-2 induced specific cytotoxic T lymphocytes (CTLs). On the contrary, immunization with mTRP-2 alone could not elicit the CTL response. Moreover, immunization of C57BL/6 wild-type mice with a combination of aTGF-beta5 and mTRP-2 induced the protective and therapeutic antitumor immunity to B16F10 melanoma, whereas the antitumor activity was abrogated in both CD4-deficient mice and CD8-deficient mice on the C57BL/6 background. Our results indicate that immunization with aTGF-beta5 is capable of breaking immune tolerance and induces mTGF-beta1-neutralizing antibodies. Neutralization of TGF-beta can enhance the efficacy of DNA vaccine encoding mTRP-2 and the induction of antitumor immunity by this immunization strategy is associated with CD4+ and CD8+ T cells.
Collapse
Affiliation(s)
- Zheng-Cai Jia
- Institute of Immunology, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 China
| | - Li-Yun Zou
- Institute of Immunology, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 China
| | - Bing Ni
- Institute of Immunology, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 China
| | - Ying Wan
- Institute of Immunology, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 China
| | - Wei Zhou
- Institute of Immunology, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 China
| | - Yan-Bo Lv
- Institute of Immunology, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 China
| | - Miao Geng
- Institute of Immunology, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 China
| | - Yu-Zhang Wu
- Institute of Immunology, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 China
| |
Collapse
|
37
|
|
38
|
Montgomery RB, Makary E, Schiffman K, Goodell V, Disis ML. Endogenous Anti-HER2 Antibodies Block HER2 Phosphorylation and Signaling through Extracellular Signal-Regulated Kinase. Cancer Res 2005. [DOI: 10.1158/0008-5472.650.65.2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Abstract
Immunologic targeting of the oncoprotein HER2/neu with monoclonal antibodies is an important component of current therapeutic strategies for patients with locally and systemically advanced breast cancer. Engineered antibodies targeting HER2 may have agonist or antagonist effects on HER2, but little is known about whether endogenous antibodies modulate HER2 activity. Vaccination of patients with HER2 peptides successfully induced antibodies in a minority of patients with HER2-expressing malignancy. A subset of antibodies specifically suppressed phosphorylation of HER2 on tyrosine Y1248, a residue critical for HER2 signaling through extracellular signal-regulated kinase. These antibodies also suppressed extracellular signal-regulated kinase phosphorylation and inhibited colony formation in soft agar. The majority of the antibodies that suppressed HER2 phosphorylation displayed specificity for amino acids 328 to 345 and 369 to 384. The isotype of anti-HER2 antibodies was predominantly IgG3 of low avidity, suggesting a Th1 response to peptide vaccine. Endogenous anti-HER2 antibodies can effectively suppress HER2 kinase activity and downstream signaling to inhibit the transformed phenotype of HER2-expressing tumor cells.
Collapse
Affiliation(s)
- R. Bruce Montgomery
- 1Division of Oncology, Department of Medicine, VA Puget Sound HCS and
- 2University of Washington, Seattle, Washington
| | - Ekram Makary
- 1Division of Oncology, Department of Medicine, VA Puget Sound HCS and
| | | | | | | |
Collapse
|
39
|
Pupa SM, Tagliabue E, Ménard S, Anichini A. HER-2: A biomarker at the crossroads of breast cancer immunotherapy and molecular medicine. J Cell Physiol 2005; 205:10-8. [PMID: 15887236 DOI: 10.1002/jcp.20387] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The oncoprotein encoded by the HER-2 oncogene is a member of the HER family of receptor tyrosine kinases and is actually the first successfully exploited target molecule in new biomolecular therapies of solid tumors. The association of HER-2 overexpression with human tumors, its extracellular accessibility, as well as its involvement in tumor aggressiveness are all factors that make this receptor an appropriate target for tumor-specific therapy. In addition, HER-2 overexpression fosters its immunogenicity, as shown by the frequency of B and T cell-mediated responses against this oncoprotein in cancer patients, and it is being investigated as a promising molecule for either passive and active immunotherapy strategies. This review summarizes a number of immune intervention approaches that target HER-2 in breast cancer.
Collapse
Affiliation(s)
- Serenella M Pupa
- Molecular Targeting Unit, Department of Experimental Oncology, Istituto Nazionale Tumori, Milan, Italy
| | | | | | | |
Collapse
|
40
|
de Gruijl TD, Pinedo HM, Scheper RJ. Immunotherapy of Cancer by Dendritic Cell-Targeted Gene Transfer. Cancer Gene Ther 2005. [DOI: 10.1007/978-1-59259-785-7_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
41
|
Abstract
DNA vaccines have been used to generate protective immunity against tumors in a variety of experimental models. The favorite target antigens have been those that are frequently expressed by human tumors, such as carcinoembryonic antigen (CEA), ErbB2/neu, and melanoma-associated antigens. DNA vaccines have the advantage of being simple to construct, produce and deliver. They can activate all arms of the immune system, and allow substantial flexibility in modifying the type of immune response generated through codelivery of cytokine genes. DNA vaccines can be applied by intramuscular, dermal/epidermal, oral, respiratory and other routes, and pose relatively few safety concerns. Compared to other nucleic acid vectors, they are usually devoid of viral or bacterial antigens and can be designed to deliver only the target tumor antigen(s). This is likely to be important when priming a response against weak tumor antigens. DNA vaccines have been more effective in rodents than in larger mammals or humans. However, a large number of methods that might be applied clinically have been shown to ameliorate these vaccines. This includes in vivo electroporation, and/or inclusion of various immunostimulatory molecules, xenoantigens (or their epitopes), antigen-cytokine fusion genes, agents that improve antigen uptake or presentation, and molecules that activate innate immunity mechanisms. In addition, CpG motifs carried by plasmids can overcome the negative effects of regulatory T cells. There have been few studies in humans, but recent clinical trials suggest that plasmid/virus, or plasmid/antigen-adjuvant, prime-boost strategies generate strong immune responses, and confirm the usefulness of plasmid-based vaccination.
Collapse
Affiliation(s)
- Gérald J Prud'homme
- Department of Laboratory Medicine and Pathobiology, St. Michael's Hospital and University of Toronto, Ontario M5B 1W8, Canada.
| |
Collapse
|
42
|
Ni J, Ni Y, Wang X, Xu W, Wang Y, Xiong S. Application of a Gene Vaccine Targeting HER-2/Neu in Immunocontraception. DNA Cell Biol 2004; 23:807-14. [PMID: 15684707 DOI: 10.1089/dna.2004.23.807] [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] [Indexed: 11/12/2022] Open
Abstract
HER-2/neu was widely used as a target for tumor prevention and therapy because of its overexpression in many tumors. However, it also plays an important role in proliferation of endometrium, embryo implantation, and development. Here, HER-2/neu was used in immunocontraception. A gene vaccine encoding the extracellular domain of human HER-2/neu was constructed. After immunization, it especially elicited both humoral and cellular responses in mice. Embryo implantation was interfered by intravenous and intraluminal injection of anti-HER-2/neu serum or lymphocytes. Lower fertility was induced after vaccination when compared with the control groups, while injuries to the uterus and ovary were not observed. Our results suggested a new and impactful target for contraceptive vaccines development.
Collapse
Affiliation(s)
- Jing Ni
- Department of Immunology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | |
Collapse
|
43
|
Wang X, Wang JP, Maughan MF, Lachman LB. Alphavirus replicon particles containing the gene for HER2/neu inhibit breast cancer growth and tumorigenesis. Breast Cancer Res 2004; 7:R145-55. [PMID: 15642163 PMCID: PMC1064108 DOI: 10.1186/bcr962] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2004] [Revised: 09/09/2004] [Accepted: 10/13/2004] [Indexed: 11/24/2022] Open
Abstract
Introduction Overexpression of the HER2/neu gene in breast cancer is associated with an increased incidence of metastatic disease and with a poor prognosis. Although passive immunotherapy with the humanized monoclonal antibody trastuzumab (Herceptin) has shown some effect, a vaccine capable of inducing T-cell and humoral immunity could be more effective. Methods Virus-like replicon particles (VRP) of Venezuelan equine encephalitis virus containing the gene for HER2/neu (VRP-neu) were tested by an active immunotherapeutic approach in tumor prevention models and in a metastasis prevention model. Results VRP-neu prevented or significantly inhibited the growth of HER2/neu-expressing murine breast cancer cells injected either into mammary tissue or intravenously. Vaccination with VRP-neu completely prevented tumor formation in and death of MMTV-c-neu transgenic mice, and resulted in high levels of neu-specific CD8+ T lymphocytes and serum IgG. Conclusion On the basis of these findings, clinical testing of this vaccine in patients with HER2/neu+ breast cancer is warranted.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Bioimmunotherapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The Graduate School of Biomedical Sciences, The University of Texas Health Sciences Center, Houston, Texas, USA
| | - Jian-Ping Wang
- Department of Bioimmunotherapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Lawrence B Lachman
- Department of Bioimmunotherapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The Graduate School of Biomedical Sciences, The University of Texas Health Sciences Center, Houston, Texas, USA
| |
Collapse
|
44
|
Lin CC, Chou CW, Shiau AL, Tu CF, Ko TM, Chen YL, Yang BC, Tao MH, Lai MD. Therapeutic HER2/Neu DNA Vaccine Inhibits Mouse Tumor Naturally Overexpressing Endogenous Neu. Mol Ther 2004; 10:290-301. [PMID: 15294176 DOI: 10.1016/j.ymthe.2004.05.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2004] [Accepted: 05/10/2004] [Indexed: 10/26/2022] Open
Abstract
The therapeutic efficacy of HER2/c-erbB-2/neu DNA immunization on mouse tumor cells expressing exogenous human or rat p185neu but not on mouse tumor cells naturally expressing mouse p185neu has been demonstrated. We investigated the feasibility of using N-terminal rat neu DNA immunization on mouse tumor overexpressing endogenous p185neu and enhancing the therapeutic efficacy of this vaccine by fusion to various cytokine genes, including interleukin-2 (IL-2), interleukin-4 (IL-4), or granulocyte-macrophage colony-stimulating factor. In a therapeutic model, N'-neu-IL-2 DNA vaccine was significantly better than N'-neu DNA vaccine, and N'-neu DNA vaccine was significantly better than control DNA or N'-neu-IL-4 DNA vaccine. The therapeutic efficacy of DNA vaccines was correlated with tumor infiltration of CD8+ T cells. Depletion of CD8+ T cells completely abolished the therapeutic effects of N'-neu-IL-2 DNA vaccine and N'-neu DNA vaccine. Depletion of CD4+ T cells after tumor implantation had no influence on N'-neu-IL-2 DNA vaccine, but enhanced the therapeutic efficacy of N'-neu DNA vaccine. Our results demonstrate that rat N'-neu DNA vaccine has a therapeutic effect on established tumor through the CD8+ T-cell-dependent pathway. Depletion of CD4+ T cells or fusion to the IL-2 gene can thus further enhance the therapeutic effects of N'-neu DNA immunization on mouse tumor expressing endogenous p185neu.
Collapse
Affiliation(s)
- Chi-Chen Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Chang SY, Lee KC, Ko SY, Ko HJ, Kang CY. Enhanced efficacy of DNA vaccination against Her-2/neu tumor antigen by genetic adjuvants. Int J Cancer 2004; 111:86-95. [PMID: 15185348 DOI: 10.1002/ijc.20232] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Certain types of malignant tumors overexpress Her-2/neu, a transmembrane glycoprotein of the class I receptor tyrosine kinase erbB family. To develop an effective Her-2/neu vaccine for selective immunotherapy of these malignancies, we prepared Her-2/neu DNA plasmid encoding the transmembrane and extracellular domain (pHM) and tested the ability of this construct to induce antitumor immunity in animal models. In addition, we investigated the effects of cytokine used as a genetic adjuvant. Modulation by factors that affect T-cell function or hematopoiesis, including interleukin-12, interleukin-15, interleukin-18, interleukin-23, Eta-1, Flt3L and GM-CSF, was studied in the forms of monocistronic and bicistronic plasmid. Our results demonstrated that vaccination of pHM could induce successful antitumor immunity against Her-2/neu-expressing murine tumor cells in BALB/c mice. We also showed that the antitumor activity of pHM was augmented by coadministration and coexpression of different cytokines. Despite the similar levels of gene expression, the antitumor effects of bicistronic plasmids coexpressing Her-2/neu antigen and cytokine were improved in comparison with coadministration of separate monocistronic plasmid. In particular, coexpression of interleukin-18 or GM-CSF with Her-2/neu increased antitumor activity in both preventive and therapeutic experiments. These findings can help in the decision concerning which of the various cytokine adjuvants should be used for the development of a Her-2/neu DNA vaccine. In addition, our results from a large panel of cytokine adjuvants in the various tumor models may provide an insight into the important immune components of antitumor immunity.
Collapse
Affiliation(s)
- Sun Young Chang
- Laboratory of Immunology, College of Pharmacy, Seoul National University, Seoul, Korea
| | | | | | | | | |
Collapse
|
46
|
De Giovanni C, Nicoletti G, Landuzzi L, Astolfi A, Croci S, Comes A, Ferrini S, Meazza R, Iezzi M, Di Carlo E, Musiani P, Cavallo F, Nanni P, Lollini PL. Immunoprevention of HER-2/neu Transgenic Mammary Carcinoma through an Interleukin 12-Engineered Allogeneic Cell Vaccine. Cancer Res 2004; 64:4001-9. [PMID: 15173014 DOI: 10.1158/0008-5472.can-03-2984] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study evaluated the ability of cytokine-engineered allogeneic (H-2(q)) HER-2/neu-positive cells to prevent tumor development in mammary cancer-prone virgin female BALB/c (H-2(d)) mice transgenic for the transforming rat HER-2/neu oncogene (BALB-neuT mice). Repeated vaccinations with cells engineered to release interleukin (IL)-2, IL-12, IL-15, or IFN-gamma showed that IL-12-engineered cell vaccines had the most powerful immunopreventive activity, with >80% of 1-year-old BALB-neuT mice free of tumors. On the contrary all of the untreated mice and all of the mice vaccinated with IL-12-engineered cells lacking either HER-2/neu or allogeneic antigens developed mammary carcinomas within 22 or 33 weeks, respectively. Whole mount, histology, immunohistochemistry, and gene expression profile analysis showed that vaccination with IL-12-engineered cells maintained 26-week mammary glands free of neoplastic growth, with a gene expression profile that clustered with that of untreated preneoplastic glands. The IL-12-engineered cell vaccine elicited a high production of IFN-gamma and IL-4 and a strong anti-HER-2/neu antibody response. Immune protection was lost or markedly impaired in BALB-neuT mice lacking IFN-gamma or antibody production, respectively. The protection afforded by the IL-12-engineered cell vaccine was equal to that provided by the systemic administration of recombinant IL-12 in combination with HER-2/neu H-2(q) cell vaccine. However, IL-12-engineered cell vaccine induced much lower circulating IL-12 and IFN-gamma, and therefore lower potential side effects and systemic toxicity.
Collapse
Affiliation(s)
- Carla De Giovanni
- Department of Experimental Pathology, University of Bologna, Bologna.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Eralp Y, Wang X, Wang JP, Maughan MF, Polo JM, Lachman LB. Doxorubicin and paclitaxel enhance the antitumor efficacy of vaccines directed against HER 2/neu in a murine mammary carcinoma model. Breast Cancer Res 2004; 6:R275-83. [PMID: 15217493 PMCID: PMC468620 DOI: 10.1186/bcr787] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2004] [Revised: 02/26/2004] [Accepted: 03/11/2004] [Indexed: 11/16/2022] Open
Abstract
Introduction The purpose of the present study was to determine whether cytotoxic chemotherapeutic agents administered prior to immunotherapy with gene vaccines could augment the efficacy of the vaccines. Methods Mice were injected in the mammary fat pad with an aggressive breast tumor cell line that expresses HER2/neu. The mice were treated 3 days later with a noncurative dose of either doxorubicin or paclitaxel, and the following day with a gene vaccine to HER2/neu. Two more doses of vaccine were given 14 days apart. Two types of gene vaccines were tested: a plasmid vaccine encoding a self-replicating RNA (replicon) of Sindbis virus (SINCP), in which the viral structural proteins were replaced by the gene for neu; and a viral replicon particle derived from an attenuated strain of Venezuelan equine encephalitis virus, containing a replicon RNA in which the Venezuelan equine encephalitis virus structural proteins were replaced by the gene for neu. Results Neither vaccination alone nor chemotherapy alone significantly reduced the growth of the mammary carcinoma. In contrast, chemotherapy followed by vaccination reduced tumor growth by a small, but significant amount. Antigen-specific CD8+ T lymphocytes were induced by the combined treatment, indicating that the control of tumor growth was most probably due to an immunological mechanism. The results demonstrated that doxorubicin and paclitaxel, commonly used chemotherapeutic agents for the treatment of breast cancer, when used at immunomodulating doses augmented the antitumor efficacy of gene vaccines directed against HER2/neu. Conclusions The combination of chemotherapeutic agents plus vaccine immunotherapy may induce a tumor-specific immune response that could be beneficial for the adjuvant treatment of patients with minimal residual disease. The regimen warrants further evaluation in a clinical setting.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/therapeutic use
- Antineoplastic Agents/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/immunology
- Breast Neoplasms/therapy
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/therapeutic use
- Carcinoma/drug therapy
- Carcinoma/immunology
- Carcinoma/therapy
- Cell Line
- Combined Modality Therapy/methods
- Combined Modality Therapy/trends
- Disease Models, Animal
- Doxorubicin/administration & dosage
- Doxorubicin/therapeutic use
- Drug Administration Schedule
- Epitopes/physiology
- Female
- Humans
- Interferon-gamma/metabolism
- Lymphocyte Activation/physiology
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Paclitaxel/administration & dosage
- Paclitaxel/therapeutic use
- Receptor, ErbB-2/therapeutic use
- T-Lymphocyte Subsets/physiology
- T-Lymphocytes, Cytotoxic/physiology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/therapeutic use
Collapse
Affiliation(s)
- Yesim Eralp
- Department of Bioimmunotherapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Current address: University of Istanbul, Institute of Oncology, Istanbul, Turkey
| | - Xiaoyan Wang
- Department of Bioimmunotherapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The Graduate School of Biomedical Sciences, The University of Texas Health Sciences Center, Houston, Texas, USA
| | - Jian-Ping Wang
- Department of Bioimmunotherapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - John M Polo
- Chiron Corporation, Emeryville, California, USA
| | - Lawrence B Lachman
- Department of Bioimmunotherapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The Graduate School of Biomedical Sciences, The University of Texas Health Sciences Center, Houston, Texas, USA
| |
Collapse
|
48
|
Piechocki MP, Ho YS, Pilon S, Wei WZ. Human ErbB-2 (Her-2) transgenic mice: a model system for testing Her-2 based vaccines. THE JOURNAL OF IMMUNOLOGY 2004; 171:5787-94. [PMID: 14634087 DOI: 10.4049/jimmunol.171.11.5787] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Her-2 transgenic (Tg) mice were generated with wild-type human c-ErbB-2 (Her-2) under the whey acidic protein promoter. They are tolerant to Her-2 and appropriate for testing Her-2 vaccines. The expression of transmembrane ErbB-2 from the whey acidic protein-Her-2 cassette and its up-regulation by insulin and hydrocortisone was verified by in vitro transfection. The transgene cassette was microinjected into fertilized eggs from B6C3 (C3H x C57BL/6) females mated with B6C3 males. Transgene-positive mice were backcrossed onto C57BL/6 mice. Human ErbB-2 was expressed in the secretory mammary epithelia during pregnancy and lactation and expressed constitutively in the Bergman glia cells within the molecular layer of the cerebellum. Overt, neoplastic transformation was not detected in any tissue examined. Tolerance to Her-2 was demonstrated by inoculating mice with a syngenic tumor expressing high levels of human ErbB-2. Tumors grew exclusively in Her-2 Tg mice without inducing an Ab response, while the nontransgenic littermates remained tumor free for 10 mo and mounted a robust anti-ErbB-2 Ab response. When immunized five times with plasmid DNA encoding secErbB-2 and GM-CSF, respectively, approximately 33% of the Her-2 Tg mice rejected a lethal challenge of EL-4/E2 tumor cells, whereas all immunized littermates rejected the tumor. Therefore, Her-2 Tg mice express human ErbB-2 in the brain and mammary gland and demonstrated tolerance to ErbB-2 which was partially overcome by DNA vaccination. The breakable tolerance of Her-2 Tg mice resembles that in human and these mice are particularly suited for testing human ErbB-2 based vaccines.
Collapse
MESH Headings
- Animals
- Antibodies, Neoplasm/biosynthesis
- Cell Line, Tumor
- Female
- Genetic Vectors
- Humans
- Immune Tolerance/genetics
- Male
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic/immunology
- Milk Proteins/biosynthesis
- Milk Proteins/genetics
- Milk Proteins/immunology
- Models, Animal
- Models, Immunological
- Neoplasm Transplantation
- Organ Specificity/genetics
- Organ Specificity/immunology
- Promoter Regions, Genetic/immunology
- Receptor, ErbB-2/biosynthesis
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Transfection
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Marie P Piechocki
- Karmanos Cancer Institute, Department of Otolaryngology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | | | | | | |
Collapse
|
49
|
Nelson EL, Prieto D, Alexander TG, Pushko P, Lofts LA, Rayner JO, Kamrud KI, Fralish B, Smith JF. Venezuelan Equine Encephalitis Replicon Immunization Overcomes Intrinsic Tolerance and Elicits Effective Anti-tumor Immunity to the ‘Self’ tumor-associated antigen, neu in a Rat Mammary Tumor Model. Breast Cancer Res Treat 2003; 82:169-83. [PMID: 14703064 DOI: 10.1023/b:brea.0000004373.09678.bb] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Many tumor-associated antigens (TAAs) represent 'self' antigens and as such, are subject to the constraints of immunologic tolerance. There are significant barriers to eliciting anti-tumor immune responses of sufficient magnitude. We have taken advantage of a Venezuelan equine encephalitis-derived alphavirus replicon vector system with documented in vivo tropism for immune system dendritic cells. We have overcome the intrinsic tolerance to the 'self' TAA rat neu and elicited an effective anti-tumor immune response using this alphavirus replicon vector system and a designed target antigen in a rigorous rat mammary tumor model. We have demonstrated the capacity to generate 50% protection in tumor challenge experiments (p = 0.004) and we have confirmed the establishment of immunologic memory by both second tumor challenge and Winn Assay (p = 0.009). Minor antibody responses were identified and supported the establishment of T helper type 1 (Th1) anti-tumor immune responses by isotype. Animals surviving in excess of 300 days with established effective anti-tumor immunity showed no signs of autoimmune phenomena. Together these experiments support the establishment of T lymphocyte dependent, Th1-biased anti-tumor immune responses to a non-mutated 'self' TAA in an aggressive tumor model. Importantly, this tumor model is subject to the constraints of immunologic tolerance present in animals with normal developmental, temporal, and anatomical expression of a non-mutated TAA. These data support the continued development and potential clinical application of this alphaviral replicon vector system and the use of appropriately designed target antigen sequences for anti-tumor immunotherapy.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Neoplasm/immunology
- Disease Models, Animal
- Encephalitis Virus, Venezuelan Equine/genetics
- Female
- Genetic Vectors/immunology
- Genetic Vectors/therapeutic use
- Humans
- Immunization
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/therapy
- Molecular Sequence Data
- Neoplasm Proteins/immunology
- Rats
- Rats, Inbred F344
- Receptor, ErbB-2/chemistry
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Replicon/immunology
- Self Tolerance
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Edward L Nelson
- Department of Medicine, Division of Hematology/Oncology, School of Medicine and Molecular Biology and Biochemistry, University of California, Irvine, CA 97697-4060, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Sudowe S, Ludwig-Portugall I, Montermann E, Ross R, Reske-Kunz AB. Transcriptional targeting of dendritic cells in gene gun-mediated DNA immunization favors the induction of type 1 immune responses. Mol Ther 2003; 8:567-75. [PMID: 14529829 DOI: 10.1016/s1525-0016(03)00242-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cutaneous dendritic cells (DC) are pivotal for the elicitation of antigen-specific immune responses following gene gun-mediated biolistic transfection of the skin. We transcriptionally targeted transgene expression to DC using vectors containing the murine fascin promoter (pFascin) to control antigen production and compared the immune response elicited with conventional DNA immunization using plasmid constructs with the ubiquitously active CMV promoter (pCMV). Biolistic transfection with pFascin initiated a marked type 1 immune response characterized by the occurrence of a large population of IFN-gamma-producing T helper (Th) cells in spleen and draining lymph nodes. Consistently, immunoglobulin production was dominated by IgG2a antibodies. In contrast, the humoral response after repeated administration of pCMV was strongly enhanced and characterized by a type 2-like isotype pattern (IgG1 > IgG2a). Cytokine production analysis in vitro indicated compartmentalization of the immune response, revealing large numbers of IL-4-producing Th cells in the lymph nodes and dominant presence of IFN-gamma-producing Th cells in the spleen. Biolistic transfection with pFascin, like immunization with pCMV, led to potent induction of cytotoxic T cells as was assessed by JAM test. Thus gene gun immunization with plasmids that focus transgene expression and antigen production specifically to DC propagates type 1-biased cellular immune responses.
Collapse
Affiliation(s)
- Stephan Sudowe
- Clinical Research Unit Allergology, Department of Dermatology, Johannes Gutenberg-University, Obere Zahlbacher Strasse 63, D-55101 Mainz, Germany.
| | | | | | | | | |
Collapse
|