51
|
Arigoni M, Barutello G, Lanzardo S, Longo D, Aime S, Curcio C, Iezzi M, Zheng Y, Barkefors I, Holmgren L, Cavallo F. A vaccine targeting angiomotin induces an antibody response which alters tumor vessel permeability and hampers the growth of established tumors. Angiogenesis 2012; 15:305-16. [PMID: 22426512 PMCID: PMC3338916 DOI: 10.1007/s10456-012-9263-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 02/21/2012] [Indexed: 12/21/2022]
Abstract
Angiomotin (Amot) is one of several identified angiostatin receptors expressed by the endothelia of angiogenic tissues. We have shown that a DNA vaccine targeting Amot overcome immune tolerance and induce an antibody response that hampers the progression of incipient tumors. Following our observation of increased Amot expression on tumor endothelia concomitant with the progression from pre-neoplastic lesions to full-fledged carcinoma, we evaluated the effect of anti-Amot vaccination on clinically evident tumors. Electroporation of plasmid coding for the human Amot (pAmot) significantly delayed the progression both of autochthonous tumors in cancer prone BALB-neuT and PyMT genetically engineered mice and transplantable TUBO tumor in wild-type BALB/c mice. The intensity of the inhibition directly correlated with the titer of anti-Amot antibodies induced by the vaccine. Tumor inhibition was associated with an increase of vessels diameter with the formation of lacunar spaces, increase in vessel permeability, massive tumor perivascular necrosis and an effective epitope spreading that induces an immune response against other tumor associated antigens. Greater tumor vessel permeability also markedly enhances the antitumor effect of doxorubicin. These data provide a rationale for the development of novel anticancer treatments based on anti-Amot vaccination in conjunction with chemotherapy regimens.
Collapse
|
52
|
The antitumor effects of adenoviral-mediated, intratumoral delivery of interleukin 23 require endogenous IL-12. Cancer Gene Ther 2011; 19:135-43. [PMID: 22158522 DOI: 10.1038/cgt.2011.78] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Interleukin (IL)-23 is a member of the IL-12 family of heterodimeric cytokines, comprised of p19 and p40 subunits, which exhibits immunostimulatory properties similar to IL-12. We have demonstrated previously that adenoviral-mediated, intratumoral delivery of IL-23 (Ad.IL-23) was able to induce systemic antitumor immunity. Here we demonstrate that Ad.IL-23 requires endogenous IL-12 for conferring an antitumor effect after adenoviral-mediated, intratumoral delivery. In contrast, Ad.IL-12 does not require IL-23 for its antitumor effects although endogenous IL-23 appears important for induction of systemic antitumor immunity by IL-12. However, despite the requirement for endogenous IL-12, co-delivery of IL-23 and IL-12 does not provide even an additive local or systemic antitumor effect, regardless of the dose. We further demonstrate that although the use of a single-chain IL-23 (scIL-23) results in higher level of expression and a more pronounced IL-23-mediated antitumor effect, there is still no synergy with IL-12. These results demonstrate that although significant antitumor effects are achieved by intratumoral injection of adenovirus expressing either scIL-23 or IL-12 alone and that IL-23 requires endogenous IL-12 for maximum antitumor benefit, the combined use of these cytokines provides no additive or synergistic effect.
Collapse
|
53
|
Ben Chaaben A, Busson M, Douik H, Boukouaci W, Mamoghli T, Chaouch L, Harzallah L, Dorra S, Fortier C, Ghanem A, Charron D, Krishnamoorthy R, Guemira F, Tamouza R. Association of IL-12p40 +1188 A/C polymorphism with nasopharyngeal cancer risk and tumor extension. ACTA ACUST UNITED AC 2011; 78:148-51. [PMID: 21623733 DOI: 10.1111/j.1399-0039.2011.01702.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The interleukin 12 (IL-12) cytokine, encoded by polymorphic genes, plays a central role in the T helper 1 cell-mediated immunity against tumors. We investigated whether the 3' untranslated region +1188 A/C polymorphism (rs 3212227) influences the nasopharyngeal carcinoma (NPC) risk in Tunisian patients. DNA analysis of 247 patients and 284 healthy individuals showed a higher frequency of the 1188 C allele and the CC genotype in patients than in controls (P = 0.00001 and P = 0.00005) suggesting that the C variant allele is associated with the susceptibility to NPC. Additional testing showed that the homozygous CC genotype is also associated with advanced stage of the tumor extension at presentation (P = 0.022). Our data suggest that the impaired production of IL-12 behaves as a risk factor for NPC occurrence and progression.
Collapse
Affiliation(s)
- A Ben Chaaben
- Clinical Biology Department, Salah Azaiz Institute of Cancer, Tunis, Tunisia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Landuzzi L, Antognoli A, Nicoletti G, Croci S, Palladini A, Ianzano ML, Murgo A, Stivani V, Grosso V, Nanni P, De Giovanni C, Lollini PL. HER-2/neu tolerant and non-tolerant mice for fine assessment of antimetastatic potency of dendritic cell-tumor cell hybrid vaccines. Vaccine 2011; 29:4690-7. [PMID: 21569812 DOI: 10.1016/j.vaccine.2011.04.096] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 03/24/2011] [Accepted: 04/26/2011] [Indexed: 01/20/2023]
Abstract
Main obstacles to cancer vaccine efficacy are pre-existing antigenic load and immunoescape mechanisms, including tolerance against self tumor-associated antigens. Here we explored the role of tolerance in an antimetastatic vaccine approach based on dendritic cell-tumor cell (DC-TC) hybrids, thanks to the comparison between BALB-neuT mice, transgenic for and tolerant to rat HER-2/neu, with their non-tolerant strain of origin BALB/c. Allogeneic DC-TC hybrid vaccine displayed a high antimetastatic activity in non-tolerant mice, but was far less effective in tolerant mice, even with intensified vaccine schedule. Tolerant BALB-neuT mice revealed a reduced ability to mount polarized Th1 responses. A further attempt to increase the antimetastatic activity by using LPS-matured DC hybrids failed. Allogeneic LPS-matured DC-TC hybrids induced high IFN-γ levels, but concomitantly also the highest production of IL-4 and IL-10 suggesting activation of mechanisms sustaining regulatory cells able to blunt vaccine efficacy. Our data in tolerant versus non-tolerant hosts suggest that clinical translation of effective DC-based strategies could benefit from more extensive investigations in tolerant transgenic models.
Collapse
Affiliation(s)
- Lorena Landuzzi
- Laboratory of Experimental Oncology, Rizzoli Orthopedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Abstract
Epidemiological and laboratory-based studies have identified infection with one of 15 high-risk human papillomavirus (HPV) types as a necessary but not sufficient cause of cervical cancer. The prevalence of genital HPV infections is high in young women, but most of the infections regress without interventions. Host genetic variations in genes involved in immune response pathways may be related to HPV clearance, and HPV E6/E7 oncoproteins interacting or downstream genes, both coding and non-coding, may contribute to the outcome of high risk HPV infection and cervical cancer. Of specific interest for this review has been the selection of genetic variants in genes involved in the above-referred pathways with a summary of their applications in association studies. Because the supportive and opposing data have been reported in different populations, well-designed international collaborative studies need to be conducted to define the consistency of the associations, paving the way to better define the patients at high risk of developing cervical cancer.
Collapse
Affiliation(s)
- Xiaojun Chen
- Department of Gynecology, Tumor Hospital of Nantong, Nantong, Jiangsu 226000, China ; Department of Epidemiology and Biostatistics, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | | | | | | |
Collapse
|
56
|
Matejuk A, Leng Q, Chou ST, Mixson AJ. Vaccines targeting the neovasculature of tumors. Vasc Cell 2011; 3:7. [PMID: 21385454 PMCID: PMC3061948 DOI: 10.1186/2045-824x-3-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 03/08/2011] [Indexed: 01/04/2023] Open
Abstract
Angiogenesis has a critical role in physiologic and disease processes. For the growth of tumors, angiogenesis must occur to carry sufficient nutrients to the tumor. In addition to growth, development of new blood vessels is necessary for invasion and metastases of the tumor. A number of strategies have been developed to inhibit tumor angiogenesis and further understanding of the interplay between tumors and angiogenesis should allow new approaches and advances in angiogenic therapy. One such promising angiogenic approach is to target and inhibit angiogenesis with vaccines. This review will discuss recent advances and future prospects in vaccines targeting aberrant angiogenesis of tumors. The strategies utilized by investigators have included whole endothelial cell vaccines as well as vaccines with defined targets on endothelial cells and pericytes of the developing tumor endothelium. To date, several promising anti-angiogenic vaccine strategies have demonstrated marked inhibition of tumor growth in pre-clinical trials with some showing no observed interference with physiologic angiogenic processes such as wound healing and fertility.
Collapse
Affiliation(s)
- Agata Matejuk
- Department of Pathology, University of Maryland Baltimore, MSTF Building, 10 South Pine Street, Baltimore, MD 21201, USA.
| | | | | | | |
Collapse
|
57
|
Cavallo F, De Giovanni C, Nanni P, Forni G, Lollini PL. 2011: the immune hallmarks of cancer. Cancer Immunol Immunother 2011; 60:319-26. [PMID: 21267721 PMCID: PMC3042096 DOI: 10.1007/s00262-010-0968-0] [Citation(s) in RCA: 252] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Accepted: 12/27/2010] [Indexed: 12/26/2022]
Abstract
Ten years after the publication of the position paper “The hallmarks of cancer” (Hanahan and Weinberg Cell 100:57–70, 2000), it has become increasingly clear that mutated cells on their way to giving rise to a tumor have also to learn how to thrive in a chronically inflamed microenvironment, evade immune recognition, and suppress immune reactivity. Genetic and molecular definition of these three immune hallmarks of cancer offers the opportunity to learn how to deploy specific countermeasures to reverse the situation in favor of the immune system and, eventually, the patient. This new information could be channeled to address what seem to be the three major hallmarks for the immune control of cancer progression: effective procedures to activate immune reactivity; characterization of not-disposable oncoantigens; and counteraction of immune suppression.
Collapse
Affiliation(s)
- Federica Cavallo
- Department of Clinical and Biological Sciences, University of Turin, Molecular Biotechnology Center, Via Nizza 52, 10126 Turin, Italy.
| | | | | | | | | |
Collapse
|
58
|
Bolli E, Quaglino E, Arigoni M, Lollini PL, Calogero R, Forni G, Cavallo F. Oncoantigens for an immune prevention of cancer. Am J Cancer Res 2010; 1:255-264. [PMID: 21969087 PMCID: PMC3180047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 12/19/2010] [Indexed: 05/31/2023] Open
Abstract
Vaccines are one of the main arms of preventive medicine. Recently a large series of experiments with cancer-prone genetically engineered mice have shown that preventive vaccines are also extremely efficacious inhibitors of the progression of carcinogenesis. Early vaccination affords significant and persistent protection, whereas its efficacy fades when neoplastic lesions become more advanced. Our current attempts to use combination strategies and technological advances to make vaccines effective in cancer prevention able to cure more advanced stages of cancer lesions are based on the temporary and systemic T(reg) removal, the preparation of new bimodular plasmids for DNA vaccination, and the search for fresh target oncoantigens.
Collapse
Affiliation(s)
- Elisabetta Bolli
- Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of TurinItaly
| | - Elena Quaglino
- Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of TurinItaly
| | - Maddalena Arigoni
- Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of TurinItaly
| | - Pier-Luigi Lollini
- Department of Hematology and Oncological Sciences, University of BolognaItaly
| | - Raffaele Calogero
- Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of TurinItaly
| | - Guido Forni
- Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of TurinItaly
| | - Federica Cavallo
- Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of TurinItaly
| |
Collapse
|
59
|
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: 2.8] [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
|
60
|
Croci S, Recktenwald CV, Lichtenfels R, Nicoletti G, Dressler SP, De Giovanni C, Astolfi A, Palladini A, Shin-ya K, Landuzzi L, Nanni P, Lollini PL, Seliger B. Proteomic and PROTEOMEX profiling of mammary cancer progression in a HER-2/neu oncogene-driven animal model system. Proteomics 2010; 10:3835-53. [DOI: 10.1002/pmic.200900643] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
61
|
Pennisi M, Pappalardo F, Palladini A, Nicoletti G, Nanni P, Lollini PL, Motta S. Modeling the competition between lung metastases and the immune system using agents. BMC Bioinformatics 2010; 11 Suppl 7:S13. [PMID: 21106120 PMCID: PMC2957681 DOI: 10.1186/1471-2105-11-s7-s13] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The Triplex cell vaccine is a cancer cellular vaccine that can prevent almost completely the mammary tumor onset in HER-2/neu transgenic mice. In a translational perspective, the activity of the Triplex vaccine was also investigated against lung metastases showing that the vaccine is an effective treatment also for the cure of metastases. A future human application of the Triplex vaccine should take into account several aspects of biological behavior of the involved entities to improve the efficacy of therapeutic treatment and to try to predict, for example, the outcomes of longer experiments in order to move faster towards clinical phase I trials. To help to address this problem, MetastaSim, a hybrid Agent Based - ODE model for the simulation of the vaccine-elicited immune system response against lung metastases in mice is presented. The model is used as in silico wet-lab. As a first application MetastaSim is used to find protocols capable of maximizing the total number of prevented metastases, minimizing the number of vaccine administrations. RESULTS The model shows that it is possible to obtain "in silico" a 45% reduction in the number of vaccinations. The analysis of the results further suggests that any optimal protocol for preventing lung metastases formation should be composed by an initial massive vaccine dosage followed by few vaccine recalls. CONCLUSIONS Such a reduction may represent an important result from the point of view of translational medicine to humans, since a downsizing of the number of vaccinations is usually advisable in order to minimize undesirable effects. The suggested vaccination strategy also represents a notable outcome. Even if this strategy is commonly used for many infectious diseases such as tetanus and hepatitis-B, it can be in fact considered as a relevant result in the field of cancer-vaccines immunotherapy. These results can be then used and verified in future "in vivo" experiments, and their outcome can be used to further improve and refine the model.
Collapse
Affiliation(s)
- Marzio Pennisi
- Department of Mathematics and Computer Science, University of Catania, V.le A. Doria 6, Catania, Italy
| | - Francesco Pappalardo
- Department of Mathematics and Computer Science, University of Catania, V.le A. Doria 6, Catania, Italy
| | - Ariannna Palladini
- Laboratory of Immunology and Biology of Metastasis, Cancer Research Section, Department of Experimental Pathology, University of Bologna, Bologna, Italy
| | - Giordano Nicoletti
- Laboratory of Experimental Oncology, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Patrizia Nanni
- Laboratory of Immunology and Biology of Metastasis, Cancer Research Section, Department of Experimental Pathology, University of Bologna, Bologna, Italy
| | - Pier-Luigi Lollini
- Department of Hematology and Oncologic Sciences "L. e A. Seragnoli", University of Bologna, Bologna, Italy
| | - Santo Motta
- Department of Mathematics and Computer Science, University of Catania, V.le A. Doria 6, Catania, Italy
| |
Collapse
|
62
|
Palladini A, Nicoletti G, Pappalardo F, Murgo A, Grosso V, Stivani V, Ianzano ML, Antognoli A, Croci S, Landuzzi L, De Giovanni C, Nanni P, Motta S, Lollini PL. In silico modeling and in vivo efficacy of cancer-preventive vaccinations. Cancer Res 2010; 70:7755-63. [PMID: 20924100 DOI: 10.1158/0008-5472.can-10-0701] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer vaccine feasibility would benefit from reducing the number and duration of vaccinations without diminishing efficacy. However, the duration of in vivo studies and the huge number of possible variations in vaccination protocols have discouraged their optimization. In this study, we employed an established mouse model of preventive vaccination using HER-2/neu transgenic mice (BALB-neuT) to validate in silico-designed protocols that reduce the number of vaccinations and optimize efficacy. With biological training, the in silico model captured the overall in vivo behavior and highlighted certain critical issues. First, although vaccinations could be reduced in number without sacrificing efficacy, the intensity of early vaccinations was a key determinant of long-term tumor prevention needed for predictive utility in the model. Second, after vaccinations ended, older mice exhibited more rapid tumor onset and sharper decline in antibody levels than young mice, emphasizing immune aging as a key variable in models of vaccine protocols for elderly individuals. Long-term studies confirmed predictions of in silico modeling in which an immune plateau phase, once reached, could be maintained with a reduced number of vaccinations. Furthermore, that rapid priming in young mice is required for long-term antitumor protection, and that the accuracy of mathematical modeling of early immune responses is critical. Finally, that the design and modeling of cancer vaccines and vaccination protocols must take into account the progressive aging of the immune system, by striving to boost immune responses in elderly hosts. Our results show that an integrated in vivo-in silico approach could improve both mathematical and biological models of cancer immunoprevention.
Collapse
Affiliation(s)
- Arianna Palladini
- Cancer Research Section, Department of Experimental Pathology, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Watanabe A, Hara M, Chosa E, Nakamura K, Sekiya R, Shimizu T, Onitsuka T. Combination of adoptive cell transfer and antibody injection can eradicate established tumors in mice--an in vivo study using anti-OX40mAb, anti-CD25mAb and anti-CTLA4mAb-. Immunopharmacol Immunotoxicol 2010; 32:238-45. [PMID: 20001272 DOI: 10.3109/08923970903222355] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Boosting an effective immune response against established tumors remains a difficult challenge. This study shows the combination of 1) adoptive cell transfer using CD25 depleted splenocytes co-cultured with irradiated tumor cells, and 2) antibody injection therapy using CTLA4 blockade, the elimination of Treg and OX40, which together could eradicate an established MethA tumor in over 50% of the BALB/c mice. Each element of the protocol was shown to be necessary, as elimination of any factor except anti-CD25 antibody injection failed to eradicate the tumor.
Collapse
Affiliation(s)
- Akira Watanabe
- Department of Cardiovascular, Thoracic and General Surgery, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan.
| | | | | | | | | | | | | |
Collapse
|
64
|
Masuelli L, Marzocchella L, Focaccetti C, Lista F, Nardi A, Scardino A, Mattei M, Turriziani M, Modesti M, Forni G, Schlom J, Modesti A, Bei R. Local delivery of recombinant vaccinia virus encoding for neu counteracts growth of mammary tumors more efficiently than systemic delivery in neu transgenic mice. Cancer Immunol Immunother 2010; 59:1247-58. [PMID: 20364378 PMCID: PMC3561760 DOI: 10.1007/s00262-010-0850-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Accepted: 03/18/2010] [Indexed: 01/14/2023]
Abstract
Recombinant vaccinia virus has been widely employed as a cancer vaccine in several clinical trials. In this study we explored, employing BALB/c mice transgenic for the rat neu oncogene, the ability of the recombinant vaccinia virus neu (rV-neuT) vaccine to inhibit growth of neu+ mammary carcinomas and whether the efficacy of vaccination was dependent on: (a) carcinogenesis stage at which the vaccination was initiated; (b) number of vaccinations and (c) route of delivery (systemic vs. local). BALB-neuT mice were vaccinated one, two and three times by subcutaneous (s.c.) and intramammary gland (im.g.) injection with rV-neuT or V-wt (wild-type vaccinia virus) starting at the stage in which mouse mammary gland displays atypical hyperplasia, carcinoma in situ or invasive carcinoma. We demonstrated that vaccination using rV-neuT was more effective when started at an earlier stage of mammary carcinogenesis and after three vaccinations. The im.g. vaccination was more effective than the s.c. vaccination in inhibiting mammary carcinogenesis, eliciting anti-Neu antibodies, increasing anti-Neu IgG2a/G3 isotypes and inducing antibodies able to trigger mammary tumor cells apoptosis and antibody-dependent cellular cytotoxicity. The better protective ability of rV-neuT im.g. vaccination was associated with its capacity to induce a superior degree of in vivo mammary cancer cells apoptosis. Our research suggests that intratumoral vaccination using recombinant vaccinia virus could be employed to increase the activity of a genetic cancer vaccine. This study may have important implications for the design of cancer vaccine protocols for the treatment of breast cancer and of accessible tumors using recombinant vaccinia virus.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Cancer Vaccines
- Cell Proliferation
- Female
- Gene Transfer Techniques
- Male
- Mammary Glands, Animal/immunology
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- NIH 3T3 Cells
- Rats
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Receptor, ErbB-2/metabolism
- Vaccines, Synthetic
- Vaccinia virus/genetics
Collapse
Affiliation(s)
- Laura Masuelli
- Department of Experimental Medicine, University of Rome ‘Sapienza’, Rome, Italy
| | - Laura Marzocchella
- Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Chiara Focaccetti
- Department of Biology, STA, University of Rome “Tor Vergata”, Rome, Italy
| | - Florigio Lista
- Centro Studi e Ricerche Sanità e Veterinaria Esercito, Rome, Italy
| | - Alessandra Nardi
- Department of Mathematics, University of Rome “Tor Vergata”, Rome, Italy
| | - Antonio Scardino
- Department of Experimental Medicine, University of Rome ‘Sapienza’, Rome, Italy
| | - Maurizio Mattei
- Department of Biology, STA, University of Rome “Tor Vergata”, Rome, Italy
| | - Mario Turriziani
- Department of Internal Medicine, University ‘Tor Vergata’, Rome, Italy
| | - Mauro Modesti
- Department of Surgery, University of Rome ‘Sapienza’, Rome, Italy
| | - Guido Forni
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | - Andrea Modesti
- Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Roberto Bei
- Department of Experimental Medicine and Biochemical Sciences, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
65
|
Kurzik-Dumke U, Hörner M, Nicotra MR, Koslowski M, Natali PG. In vivo evidence of htid suppressive activity on ErbB-2 in breast cancers over expressing the receptor. J Transl Med 2010; 8:58. [PMID: 20565727 PMCID: PMC2909173 DOI: 10.1186/1479-5876-8-58] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Accepted: 06/17/2010] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Htid encoded proteins are physiological partners of a wide spectrum of molecules relevant to neoplastic transformation. One of the molecular ligands of the cytosolic hTid-L and hTid-I forms is the ErbB-2 receptor variably over expressed in diverse solid tumors. Altered ErbB-2 signalling is associated with an unfavourable prognosis in about 30% of human breast malignancies. METHODS We evaluated htid and HER-2 expression by quantitative real time PCR in tumors of different TNMG status and by immunohistochemistry in a cohort of breast tumors of the Luminal A, B, HER-2 and triple negative subtype. RESULTS The RT-PCR analysis revealed that aberrant expression of all three htid forms correlates with malignant transformation. Furthermore, elevated hTid-L expression can be associated with less aggressive tumors. The immunohistochemical testing revealed that tumors of the luminal A subtype are characterized by a high level of htid (81%). In contrast htid expression is significantly lower in tumors of the Luminal B (20%) and HER-2 (18%) subtype over expressing the receptor and in the triple negative (40%) more aggressive malignancies. A statistically significant inverse correlation between htid and ErbB-2 expression was found in human breast (p < 0,0001) and non-mammary tumors (p < 0,007), and in transgenic mice carrying the rat HER-2/neu oncogene. CONCLUSIONS Our findings provide in vivo evidence that htid is a tissue independent and evolutionarily conserved suppressor of ErbB-2.
Collapse
Affiliation(s)
- Ursula Kurzik-Dumke
- Institute of Medical Microbiology and Hygiene, Comparative Tumor Biology Group, University Medical Center, Johannes Gutenberg University, Obere Zahlbacher Str. 63, 55131 Mainz, Germany
| | - Manuela Hörner
- Institute of Medical Microbiology and Hygiene, Comparative Tumor Biology Group, University Medical Center, Johannes Gutenberg University, Obere Zahlbacher Str. 63, 55131 Mainz, Germany
| | | | - Michael Koslowski
- Experimental and Translational Oncology III, University Medical Center, Johannes Gutenberg University, Obere Zahlbacher Str. 63, 55131 Mainz, Germany
| | - Pier G Natali
- Immunology Laboratory, "Regina Elena" National Cancer Institute, Via delle Messi d'Oro 156, 0158 Rome and CIMBO Laboratories, "G.d'Annunzio" University, Chieti, Italy
| |
Collapse
|
66
|
Liu L, Xu Y, Liu Z, Chen J, Zhang Y, Zhu J, Liu J, Liu S, Ji G, Shi H, Shen H, Hu Z. IL12 polymorphisms, HBV infection and risk of hepatocellular carcinoma in a high-risk Chinese population. Int J Cancer 2010; 128:1692-6. [PMID: 20521253 DOI: 10.1002/ijc.25488] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Accepted: 05/27/2010] [Indexed: 12/13/2022]
Abstract
To investigate the association between the potentially functional polymorphisms in IL12A and IL12B, HBV infection and risk of hepatocellular carcinoma in a Chinese population, we genotyped three polymorphisms, rs568408 (3'UTR G>A), rs2243115 (5'UTR T>G) in IL12A and rs3212227 (3'UTR A>C) in IL12B in a case-control study of 869 hepatocellular carcinoma (HCC) cases and 891 cancer-free controls. We found that the IL12A rs568408 GA/AA variant genotypes were associated with a significantly increased risk of HCC (adjusted odds ratio (OR) = 1.53, 95% confidence interval (CI) = 1.17-2.00), compared with the wild-type GG homozygote. In the stratified analyses, the increased risk of HCC associated with rs568408 GA/AA was more evident in patients who were negative for HBsAg (adjusted OR = 1.71, 95% CI = 1.23-2.39). However, no significant associations between IL12A rs2243115 T/G, IL12B rs3212227 A/C and risk of HCC were observed. Our findings indicate that IL12A rs568408 may contribute to the risk of HCC and modify HCC risk associated with HBV infection.
Collapse
Affiliation(s)
- Li Liu
- Department of Epidemiology and Biostatistics, Cancer Center, Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Rolla S, Ria F, Occhipinti S, Di Sante G, Iezzi M, Spadaro M, Nicolò C, Ambrosino E, Merighi IF, Musiani P, Forni G, Cavallo F. Erbb2 DNA vaccine combined with regulatory T cell deletion enhances antibody response and reveals latent low-avidity T cells: potential and limits of its therapeutic efficacy. THE JOURNAL OF IMMUNOLOGY 2010; 184:6124-32. [PMID: 20435927 DOI: 10.4049/jimmunol.0901215] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Rat (r)Erbb2 transgenic BALB-neuT mice genetically predestined to develop multiple invasive carcinomas allow an assessment of the potential of a vaccine against the stages of cancer progression. Because of rErbb2 expression in the thymus and its overexpression in the mammary gland, CD8(+) T cell clones reacting at high avidity with dominant rErbb2 epitopes are deleted in these mice. In BALB-neuT mice with diffuse and invasive in situ lesions and almost palpable carcinomas, a temporary regulatory T cells depletion combined with anti-rErbb2 vaccine markedly enhanced the anti-rErbb2 Ab response and allowed the expansion of latent pools of low-avidity CD8(+) T cells bearing TCRs repertoire reacting with the rErbb2 dominant peptide. This combination of a higher Ab response and activation of a low-avidity cytotoxic response persistently blocked tumor progression at stages in which the vaccine alone was ineffective. However, when diffuse and invasive microscopic cancers become almost palpable, this combination was no longer able to secure a significant extension of mice survival.
Collapse
Affiliation(s)
- Simona Rolla
- Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Farkas AM, Finn OJ. Vaccines based on abnormal self-antigens as tumor-associated antigens: immune regulation. Semin Immunol 2010; 22:125-31. [PMID: 20403708 DOI: 10.1016/j.smim.2010.03.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Accepted: 03/15/2010] [Indexed: 01/21/2023]
Abstract
Abnormal expression of "self" antigens on tumors compared with normal cells provides opportunities and challenges for development of cancer vaccines. We review recent work in pre-clinical transgenic mouse models and in clinical trials that has elucidated multiple regulatory mechanisms that interfere with the induction of effective immunity. We discuss these as being either part of the normal function of the immune system or being driven by the tumor microenvironment. Collectively this work shows that it is possible to design vaccines based on tumor-associated antigens and elicit effective immunity against abnormal expression of these antigens on tumors without causing autoimmunity.
Collapse
Affiliation(s)
- Adam M Farkas
- Department of Immunology, University of Pittsburgh School of Medicine, E1044, Biomedical Science Tower, 200 Lothrop St., Pittsburgh, PA 15261, USA
| | | |
Collapse
|
69
|
Zhu S, Lee DA, Li S. IL-12 and IL-27 sequential gene therapy via intramuscular electroporation delivery for eliminating distal aggressive tumors. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:2348-54. [PMID: 20139275 PMCID: PMC2824785 DOI: 10.4049/jimmunol.0902371] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Eradication of residual malignancies and metastatic tumors via a systemic approach is the key for successfully treating cancer and increasing cancer patient survival. Systemic administration of IL-12 protein in an acute large dose is effective but toxic. Systemic administration of IL-12 gene by persistently expressing a low level of IL-12 protein may reduce the systemic toxicity but only eradicates IL-12-sensitive tumors. In this study, we discovered that sequential administration of IL-12- and IL-27-encoding DNA, referred to as sequential IL-12-->IL-27 (IL-12 administration followed by IL-27 administration 10 d after) gene therapy, not only eradicated IL-12-sensitive CT26 tumors from 100% of mice but also eradicated the highly malignant 4T1 tumors from 33% of treated mice in multiple independent experiments. This IL-12-->IL-27 sequential gene therapy is not only superior to IL-12-encoding plasmid DNA given a total of two times at a 10-d interval sequential gene therapy for eliminating tumors but also for inducing CTL activity, increasing T cell infiltration into tumors, and yielding a large number of tumor-specific IFN-gamma-positive CD8 T cells. Notably, depletion of either T or NK cells during the IL-27 treatment phase reverses tumor eradication, suggesting an NK cell requirement for this sequential gene therapy-mediated tumor eradication. Both reversal of the administration sequence and coadministration of IL-12 and IL-27 impaired tumor eradication in 4T1 tumor-bearing mice. This IL-12-->IL-27 sequential gene therapy, via sequential administration of IL-12- and IL-27-encoding plasmid DNA into tumor-bearing mice through i.m. electroporation, provides a simple but effective approach for eliminating inaccessible residual tumors.
Collapse
Affiliation(s)
- Shiguo Zhu
- School of Medicine & Institute of Medical Science, Shanghai Jiao Tong University, 280 Chongqing South Road, Shanghai 200025, PR China
| | - Dean Anthony Lee
- The University of Texas MD Anderson Cancer Center, Division of Pediatrics, Houston, TX 77030
| | - Shulin Li
- Department of Comparative Biomedical Sciences, Louisiana State University, Skip Bertman Drive, Baton Rouge, LA 70803
| |
Collapse
|
70
|
Parajuli N, Müller-Holzner E, Böck G, Werner ER, Villunger A, Doppler W. Infiltrating CD11b+CD11c+ cells have the potential to mediate inducible nitric oxide synthase-dependent cell death in mammary carcinomas of HER-2/neu transgenic mice. Int J Cancer 2010; 126:896-908. [PMID: 19653277 DOI: 10.1002/ijc.24805] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The development of autochtonous mammary tumors in HER-2/neu transgenic mice is facilitated by immune tolerance to the neu-transgene. However, appropriate vaccination strategies can initiate immune system-mediated antitumor response by a process that requires IFN-gamma. We investigated the role of inducible nitric oxide synthase (iNOS) induction by IFN-gamma to promote tumor cell apoptosis. Tumors from FVBN202 mice expressing the normal neu gene under the control of the MMTV-LTR were treated in slice cultures with IFN-gamma for up to 24 hr. Apoptosis was induced, which depended on iNOS enzymatic activity. iNOS expression was predominantly found in infiltrating CD11b(+)CD11c(+) myeloid cells and at much lower levels in the tumor epithelium. By contrast, IFN-gamma treatment of explant cultures of tumor epithelial cells was not sufficient to efficiently induce iNOS, emphasizing an important role of the integrity of tumor tissue architecture, which was preserved in the slice cultures. This notion was further supported by the upregulation of iNOS costimulatory cytokines TNF-alpha and IL-1beta in slice cultures but not in explants and the capability of purified CD11b(+)CD11c(+) cells to enhance iNOS expression of tumor cells in cocultures. The findings suggest that tumor-infiltrating myeloid cells in immuno-tolerant HER-2/neu transgenic mice possess tumor killing ability via induction of iNOS and underline the capacity of antitumor strategies designed to stimulate infiltrating myeloid cells.
Collapse
Affiliation(s)
- Nirmala Parajuli
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | | | | | | | | | | |
Collapse
|
71
|
De Giovanni C, Nicoletti G, Palladini A, Croci S, Landuzzi L, Antognoli A, Murgo A, Astolfi A, Ferrini S, Fabbi M, Orengo AM, Amici A, Penichet ML, Aurisicchio L, Iezzi M, Musiani P, Nanni P, Lollini PL. A multi-DNA preventive vaccine for p53/Neu-driven cancer syndrome. Hum Gene Ther 2010; 20:453-64. [PMID: 19215191 DOI: 10.1089/hum.2008.172] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The highly aggressive cancer syndrome of female mice carrying a p53 knockout allele and a rat HER-2/neu (Neu) transgene (BALB-p53Neu) can be prevented by a cell vaccine presenting three components: Neu, interleukin (IL)-12 production, and allogeneic major histocompatibility complex (MHC) alleles (Triplex cell vaccine). Here we tested a second-generation Triplex DNA-based vaccine (Tri-DNA), consisting of the combination of three gene components (a transmembrane-extracellular domain fragment of the Neu gene, IL-12 genes, and the H-2D(q) allogeneic MHC gene), carried by separate plasmids. The Tri-DNA vaccine was at least as effective as the Triplex cell vaccine for cancer immunoprevention, giving a similar delay in the onset of mammary cancer and complete protection from salivary cancer. Both vaccines induced anti-Neu antibodies of the murine IgG2a isotype at similar levels. The Tri-DNA vaccine gave more restricted immunostimulation, consisting of a fully helper T cell type 1 (Th1)-polarized response, with effective production of interferon (IFN)-gamma in response to the vaccine but no spontaneous production, and no induction of anti-Neu IgG3 antibodies. On the other hand, the Triplex cell vaccine induced both Th1 and Th2 cytokines, a strong increase in spontaneous IFN-gamma production, and high levels of IgG3 antibodies recognizing Neu-positive syngeneic cells. In conclusion, the Tri-DNA vaccine is as effective as Triplex cell vaccine, exploiting a more restricted immune stimulation.
Collapse
Affiliation(s)
- C De Giovanni
- Cancer Research Section, Department of Experimental Pathology, Alma Mater Studiorum, University of Bologna, I-40126 Bologna, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Warren RL, Holt RA. A census of predicted mutational epitopes suitable for immunologic cancer control. Hum Immunol 2010; 71:245-54. [PMID: 20035814 DOI: 10.1016/j.humimm.2009.12.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Revised: 12/09/2009] [Accepted: 12/17/2009] [Indexed: 11/25/2022]
Abstract
The adaptive immune system can protect against spontaneously arising tumors, and the potential exists to reduce cancer incidence by priming adaptive immune responses with vaccines. Immunologic cancer control has been implemented for cancers caused by infectious agents, but not for spontaneous cancers caused by mutation. This is largely due to the high cost of preventative clinical trials and the lack of validated tumor epitopes. Here we evaluate, computationally, all known somatic mutations in human tumors for their antigenic potential. All possible human leukocyte antigen (HLA) class I presented peptides containing recurrent somatic cancer mutations with frequency > 5% were screened by three independent epitope prediction algorithms (SYFPEITHI, BIMAS, and IEDB). Using stringent filters, a total of 20 genes, 35 mutations, and 159 candidate epitopes were identified, each presented by up to four distinct HLA class I alleles. The top-ranking gene from our survey was KRAS, which figures prominently because there are frequent hotspot mutations in numerous, prevalent cancers, and mutant peptides are predicted to be presented by several common HLA alleles. From our data, we estimate that prophylactic vaccination could provide meaningful levels of prevention of tumors associated with common recurrent mutations.
Collapse
Affiliation(s)
- René L Warren
- British Columbia Cancer Agency, Genome Sciences Centre, Vancouver, British Columbia, Canada
| | | |
Collapse
|
73
|
Vaccine protocols optimization: In silico experiences. Biotechnol Adv 2010; 28:82-93. [DOI: 10.1016/j.biotechadv.2009.10.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2009] [Revised: 09/15/2009] [Accepted: 09/30/2009] [Indexed: 11/21/2022]
|
74
|
Reay J, Kim SH, Lockhart E, Kolls J, Robbins PD. Adenoviral-mediated, intratumor gene transfer of interleukin 23 induces a therapeutic antitumor response. Cancer Gene Ther 2009; 16:776-85. [PMID: 19390568 PMCID: PMC2745505 DOI: 10.1038/cgt.2009.27] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2008] [Revised: 01/19/2009] [Accepted: 03/20/2009] [Indexed: 11/30/2022]
Abstract
Interleukin 23 (IL-23) is a member of the IL-12 family of heterodimeric cytokines, composed of p19 and p40 subunits, which exhibits immunostimulatory properties similar to IL-12. IL-23 has been shown to possess potent antitumor activities in several establishment models of cancer and a few therapeutic models, but the efficacy of local, adenoviral-mediated expression of IL-23 in established tumors has yet to be investigated. Here we have examined the antitumor activity of adenovirally delivered IL-23 in a day-7 MCA205 murine fibrosarcoma tumor model. Three intratumoral injections of adenovirus expressing IL-23 (Ad.IL-23) significantly increased animal survival and resulted in complete rejection of 40% of tumors, with subsequent generation of protective immunity and MCA205-specific cytotoxic T lymphocytes. In addition, we have shown that the antitumor activity of IL-23 is independent of IL-17, perforin and Fas ligand, but dependent on interferon-gamma, CD4(+) and CD8(+) T cells. These results demonstrate that direct intratumoral injection of adenovirus expressing IL-23 results in enhanced survival, tumor eradication and generation of protective immunity by generation of a Th1-type immune response.
Collapse
Affiliation(s)
- Ja'Nean Reay
- Department of Microbiology and Molecular Genetics University of Pittsburgh School of Medicine Pittsburgh, PA
| | - Seon-Hee Kim
- Department of Microbiology and Molecular Genetics University of Pittsburgh School of Medicine Pittsburgh, PA
| | - Euan Lockhart
- Department of Microbiology and Molecular Genetics University of Pittsburgh School of Medicine Pittsburgh, PA
| | - Jay Kolls
- Department of Pediatrics Childrens' Hospital of Pittsburgh Pittsburgh, PA
| | - Paul D. Robbins
- Department of Microbiology and Molecular Genetics University of Pittsburgh School of Medicine Pittsburgh, PA
| |
Collapse
|
75
|
Abstract
Recent scientific advances have expanded our understanding of the immune system and its response to malignant cells. The clinical goal of tumour immunotherapy is to provide either passive or active immunity against malignancies by harnessing the immune system to target tumours. Monoclonal antibodies, cytokines, cellular immunotherapy, and vaccines have increasingly become successful therapeutic agents for the treatment of solid and haematological cancers in preclinical models, clinical trials, and practice. In this article, we review recent advances in the immunotherapy of cancer, focusing on new strategies and future perspectives as well as on clinical trials attempting to enhance the efficacy of immunotherapeutic modalities and translate this knowledge into effective cancer therapies.
Collapse
|
76
|
Pennisi M, Pappalardo F, Ping Zhang, Motta S. Searching of optimal vaccination schedules. ACTA ACUST UNITED AC 2009; 28:67-72. [DOI: 10.1109/memb.2009.932919] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
77
|
Tamoxifen combined to anti-HER-2/neu cell vaccine does not hamper cancer immunopreventive efficacy. Vaccine 2009; 27:2065-9. [DOI: 10.1016/j.vaccine.2009.01.113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Revised: 01/16/2009] [Accepted: 01/22/2009] [Indexed: 11/22/2022]
|
78
|
Koski GK, Cohen PA, Roses RE, Xu S, Czerniecki BJ. Reengineering dendritic cell-based anti-cancer vaccines. Immunol Rev 2009; 222:256-76. [PMID: 18364007 DOI: 10.1111/j.1600-065x.2008.00617.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite initial enthusiasm, dendritic cell (DC)-based anti-cancer vaccines have yet to live up to their promise as one of the best hopes for generating effective anti-tumor immunity. One of the principal reasons for the generally disappointing results achieved thus far could be that the full potential of DCs has not been effectively exploited. Here, we argue that dramatic improvements in vaccine efficacy will probably require a careful re-evaluation of current vaccine design. The formulation of new strategies must take into account the natural history of DCs, particularly their role in helping the immune system deal with infection. Equally critical is the emerging importance of soluble factors, notably interleukin-12, in modulating the quality of immune responses. Vaccines should also be designed to recruit helper T cells and antibody-producing B cells rather than simply cytotoxic T lymphocytes. Finally, the judicious selection of tumor, target antigen, and disease stage best suited for treatment should serve as the foundation of trial designs. Our discussion addresses a recent clinical vaccine trial to treat early breast cancer, where many elements of this new strategy were put into practice.
Collapse
Affiliation(s)
- Gary K Koski
- Department of Immunology, Center for Surgery Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | | | |
Collapse
|
79
|
Chen X, Han S, Wang S, Zhou X, Zhang M, Dong J, Shi X, Qian N, Wang X, Wei Q, Shen H, Hu Z. Interactions of IL-12A and IL-12B Polymorphisms on the Risk of Cervical Cancer in Chinese Women. Clin Cancer Res 2008; 15:400-5. [DOI: 10.1158/1078-0432.ccr-08-1829] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
80
|
Antibodies targeted to TRAIL receptor-2 and ErbB-2 synergize in vivo and induce an antitumor immune response. Proc Natl Acad Sci U S A 2008; 105:16254-9. [PMID: 18838682 DOI: 10.1073/pnas.0806849105] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Despite the development of human epidermal growth factor receptor-2 (ErbB-2/HER2)-targeted therapies, there remains an unmet medical need for breast cancer patients with ErbB-2 overexpression. We investigated the therapeutic activity of an agonist mAb to mouse tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor-2 (DR5) against ErbB2-driven breast cancer. Established tumors in BALB/c transgenic mice expressing a constitutively active ErbB-2/neuT were treated with anti-DR5 mAb and/or anti-ErbB-2 mAb and monitored for tumor progression. Treatment with anti-DR5 or anti-ErbB2 mAb as single agents significantly delayed tumor growth, although all tumors eventually progressed. Remarkably, treatment with a combination of anti-DR5 and anti-ErbB-2 mAbs induced complete response in a majority of mice. In vivo blockade of CD11b(+) cells, but not natural killer cell depletion, significantly abrogated the early antitumor response. Notably, depletion of CD8(+) T cells provoked primary and secondary tumor relapse, revealing the induction of antitumor immunity by the combination treatment. Combined therapy with anti-DR5 and anti-ErbB-2 mAbs further significantly suppressed the growth of advanced spontaneous tumors in ErbB-2/neuT transgenic mice, even when treatment was delayed until tumors were palpable. We thus demonstrated that the combination of anti-DR5 and anti-ErbB2 mAbs might be an effective form of treatment for ErbB-2-overexpressing breast cancer.
Collapse
|
81
|
Her-2 DNA versus cell vaccine: immunogenicity and anti-tumor activity. Cancer Immunol Immunother 2008; 58:759-67. [PMID: 18836716 DOI: 10.1007/s00262-008-0599-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2008] [Accepted: 09/18/2008] [Indexed: 01/22/2023]
Abstract
Direct comparison and ranking of vaccine formulations in pre-clinical studies will expedite the identification of cancer vaccines for clinical trials. Two human ErbB-2 (Her-2) vaccines, naked DNA and whole cell vaccine, were tested side-by-side in wild type and Her-2 transgenic mice. Both vaccines can induce humoral and cellular immunity to the entire repertoire of Her-2 epitopes. Mice were electro-vaccinated i.m. with a mixture of pGM-CSF and pE2TM, the latter encodes Her-2 extracellular and transmembrane domains. Alternatively, mice were injected i.p. with human ovarian cancer SKOV3 cells that have amplified Her-2. In wild type mice, comparable levels of Her-2 antibodies (Ab) were induced by these two vaccines. However, T cell immunity and protection against Her-2(+) tumors were superior in DNA vaccinated mice. In BALB Her-2 transgenic (Tg) mice, which were tolerant to Her-2, DNA and cell vaccines were administered after regulatory T cells (Treg) were removed by anti-CD25 mAb. Again, comparable levels of Her-2 Ab were induced, but DNA vaccines rendered greater anti-tumor activity. In B6xDR3 Her-2 Tg mice that expressed the autoimmune prone HLA-DR3 allele, higher levels of Her-2 Ab were induced by SKOV3 cell than by Her-2 DNA. But anti-tumor activity was still more profound in DNA vaccinated mice. Therefore, Her-2 DNA vaccine induced greater anti-tumor immunity than cell vaccine, whether mice were tolerant to Her-2 or susceptible to autoimmunity. Through such side-by-side comparisons in appropriate pre-clinical test systems, the more effective vaccine formulations will emerge as candidates for clinical trials.
Collapse
|
82
|
Quaglino E, Mastini C, Forni G, Cavallo F. ErbB2 transgenic mice: a tool for investigation of the immune prevention and treatment of mammary carcinomas. CURRENT PROTOCOLS IN IMMUNOLOGY 2008; Chapter 20:Unit 20.9.1-20.9-10. [PMID: 18729063 DOI: 10.1002/0471142735.im2009s82] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
The epidermal growth factor receptor belongs to a superfamily of receptor tyrosine kinases (RTK) that includes ErbB2. ErbB2 is involved in normal physiological processes, such as embryogenesis, cell proliferation, differentiation, adhesion motility, and apoptosis, while its malfunction or overexpression is responsible for development defects, diabetes, and cancer. The human ortholog of ErbB2 is referred as Her-2 (human ErbB2) while the rat ortholog is referred as neu (rat ErbB2). As ErbB2 is directly involved in carcinogenesis, mice transgenic for the rat neu oncogene allow straightforward assessment of the ability of drugs and vaccines to inhibit the progression of neu-driven cancer. Information from this model may provide indications on the efficacy of similar treatments in patients. This commentary provides key information regarding the use of these transgenic mouse models for evaluation of the efficacy of anti-tumor strategies.
Collapse
Affiliation(s)
- Elena Quaglino
- Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | | | | | | |
Collapse
|
83
|
Müller-Hermelink N, Braumüller H, Pichler B, Wieder T, Mailhammer R, Schaak K, Ghoreschi K, Yazdi A, Haubner R, Sander CA, Mocikat R, Schwaiger M, Förster I, Huss R, Weber WA, Kneilling M, Röcken M. TNFR1 signaling and IFN-gamma signaling determine whether T cells induce tumor dormancy or promote multistage carcinogenesis. Cancer Cell 2008; 13:507-18. [PMID: 18538734 DOI: 10.1016/j.ccr.2008.04.001] [Citation(s) in RCA: 240] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2004] [Revised: 05/23/2007] [Accepted: 04/08/2008] [Indexed: 12/12/2022]
Abstract
Immune responses may arrest tumor growth by inducing tumor dormancy. The mechanisms leading to either tumor dormancy or promotion of multistage carcinogenesis by adaptive immunity are poorly characterized. Analyzing T antigen (Tag)-induced multistage carcinogenesis in pancreatic islets, we show that Tag-specific CD4+ T cells home selectively into the tumor microenvironment around the islets, where they either arrest or promote transition of dysplastic islets into islet carcinomas. Through combined TNFR1 signaling and IFN-gamma signaling, Tag-specific CD4+ T cells induce antiangiogenic chemokines and prevent alpha(v)beta(3) integrin expression, tumor angiogenesis, tumor cell proliferation, and multistage carcinogenesis, without destroying Tag-expressing islet cells. In the absence of either TNFR1 signaling or IFN-gamma signaling, the same T cells paradoxically promote angiogenesis and multistage carcinogenesis. Thus, tumor-specific T cells can directly survey multistage carcinogenesis through cytokine signaling.
Collapse
MESH Headings
- Animals
- Antigens, Viral, Tumor/genetics
- Antigens, Viral, Tumor/metabolism
- Blood Glucose/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/pathology
- CD4-Positive T-Lymphocytes/transplantation
- Cell Movement
- Cell Proliferation
- Cell Survival
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Cells, Cultured
- GTPase-Activating Proteins/genetics
- GTPase-Activating Proteins/metabolism
- Immunotherapy/methods
- Insulinoma/blood supply
- Insulinoma/genetics
- Insulinoma/immunology
- Insulinoma/metabolism
- Insulinoma/pathology
- Insulinoma/therapy
- Integrin alphaVbeta3/metabolism
- Interferon-gamma/metabolism
- Mice
- Mice, Inbred C3H
- Mice, Knockout
- Mice, Transgenic
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/pathology
- Pancreatic Neoplasms/blood supply
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/therapy
- Receptors, Tumor Necrosis Factor, Type I/deficiency
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Signal Transduction
- Th1 Cells/immunology
- Th1 Cells/pathology
- Time Factors
- Whole-Body Irradiation
Collapse
Affiliation(s)
- Nele Müller-Hermelink
- Department of Dermatology, Eberhard Karls University, Liebermeisterstrasse 25, 72076 Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Using Genetically Modified Microvascular Free Flaps to Deliver Local Cancer Immunotherapy with Minimal Systemic Toxicity. Plast Reconstr Surg 2008; 121:1541-1553. [PMID: 18453976 DOI: 10.1097/prs.0b013e31816ff6aa] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
85
|
Park JM, Terabe M, Steel JC, Forni G, Sakai Y, Morris JC, Berzofsky JA. Therapy of advanced established murine breast cancer with a recombinant adenoviral ErbB-2/neu vaccine. Cancer Res 2008; 68:1979-87. [PMID: 18339880 DOI: 10.1158/0008-5472.can-07-5688] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
ErbB-2 (HER-2/neu) is a transforming oncogene expressed by a substantial fraction of breast cancers, and monoclonal antibody therapy directed toward this antigen is an established treatment modality. However, not all tumors respond, and with a monoclonal antibody directed to a single epitope, there is always the risk of tumor escape. Furthermore, passive antibody therapy requires continual treatment. Whereas cancer vaccines have prevented the growth of tumors, it has been far more difficult to treat large established tumors. Here, we show that vaccination with a recombinant adenovirus expressing a truncated ErbB-2 antigen can cure large established subcutaneous ErbB-2-expressing breast cancers in mice, and can also cure extensive established lung metastatic disease. We also show that the mechanism of protection involves antibody-mediated blockade of ErbB-2 function, independent of Fc receptors. We conclude that a vaccine inducing antibodies to a functional oncogenic receptor could have tremendous therapeutic potential against cancers overexpressing such molecules.
Collapse
Affiliation(s)
- Jong Myun Park
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland 20892-1578, USA
| | | | | | | | | | | | | |
Collapse
|
86
|
|
87
|
Systemic IL-12 gene therapy for treating malignancy via intramuscular electroporation. Methods Mol Biol 2008. [PMID: 18370211 DOI: 10.1007/978-1-59745-194-9_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Interleukin 12 (IL-12) is effective in treating systemic microscopic malignancies by inducing T helper 1 (T(H)1) response, inhibiting angiogenesis, and triggering secondary cytokine production. Unfortunately, daily systemic administration of an acute dose of IL-12 protein is very costly and severely toxic. Here, a simple, economic, and less toxic approach, intramuscular administration of IL-12 gene, is provided for treating tumors in three tumor models. The results indicate that intramuscular administration of IL-12 encoding plasmid DNA via electroporation is a promising technology for treating systemic residual malignancies (less than 3-5 mm in diameter), as illustrated by the inhibition of tumor growth and lung metastases as well as the extension of survival rate. This approach is not effective in treating tumors larger than 3-5 mm in diameter.
Collapse
|
88
|
Piechocki MP, Dibbley SK, Lonardo F, Yoo GH. Gefitinib prevents cancer progression in mice expressing the activated rat HER2/neu. Int J Cancer 2008; 122:1722-9. [PMID: 18076070 DOI: 10.1002/ijc.23231] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We tested the efficacy of gefitinib in the prevention of HER2/neu-mediated breast cancer development in BALB-NeuT transgenic mice. Oral administration of gefitinib to female transgenic mice from 5 to 14 weeks of age reduced tumor multiplicity from 9.6 +/- 0.82 to 0.58 +/- 1.1 (83%). We observed a decrease in the number and size of lobules and lobular nodules in treated mice with a reduction in the overall disease burden per gland. Normal duct development in the mammary glands was not affected by gefitinib. The development of acinic cell carcinoma in the parotid glands of these animals was also reduced coincident with decreased stromal involvement during progression. Gefitinib eliminated phosphorylation of HER2 and HER3 and signaling through MAPK and Akt in lobular hyperplasias and carcinomas. At the same time MAPK activity and cytokine production in splenocytes and lymph nodes was increased in gefitinib-treated animals coincident with an increase in lymph node size. Delaying gefitinib treatment until mammary glands exhibited atypical lobular hyperplasias reduced efficacy. These studies demonstrate the critical role of HER2 signal transduction in the onset and progression of HER2/neu-dependent breast cancer and suggest a role for specific inhibitors to prevent the outgrowth of early hyperplastic disease.
Collapse
Affiliation(s)
- Marie P Piechocki
- Department of Otolaryngology-Head and Neck Surgery, Wayne State University and Karmanos Cancer Center, Detroit, MI 48201, USA.
| | | | | | | |
Collapse
|
89
|
Ostrand-Rosenberg S. Immune surveillance: a balance between protumor and antitumor immunity. Curr Opin Genet Dev 2008; 18:11-8. [PMID: 18308558 PMCID: PMC2699403 DOI: 10.1016/j.gde.2007.12.007] [Citation(s) in RCA: 323] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 12/19/2007] [Accepted: 12/20/2007] [Indexed: 01/11/2023]
Abstract
Precancerous and malignant cells can induce an immune response which results in the destruction of transformed and/or malignant cells, a process known as immune surveillance. However, immune surveillance is not always successful, resulting in 'edited' tumors that have escaped immune surveillance. Immunoediting is not simply because of the absence of antitumor immunity, but is because of protumor immunity that blocks antitumor adaptive and innate responses, and promotes conditions that favor tumor progression. Several immune protumor effector mechanisms are upregulated by chronic inflammation, leading to the hypothesis that inflammation promotes carcinogenesis and tumor growth by altering the balance between protumor and antitumor immunity, thereby preventing the immune system from rejecting malignant cells, and providing a tumor-friendly environment for progressive disease.
Collapse
Affiliation(s)
- Suzanne Ostrand-Rosenberg
- University of Maryland, Baltimore County, Department of Biological Sciences, 1000 Hilltop Circle, Baltimore, MD 21250, United States.
| |
Collapse
|
90
|
Nanni P, Nicoletti G, Palladini A, Croci S, Murgo A, Antognoli A, Landuzzi L, Fabbi M, Ferrini S, Musiani P, Iezzi M, De Giovanni C, Lollini PL. Antimetastatic activity of a preventive cancer vaccine. Cancer Res 2007; 67:11037-44. [PMID: 18006850 DOI: 10.1158/0008-5472.can-07-2499] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The development of prophylactic cancer vaccines that protect healthy hosts from tumor development leaves open the question whether such vaccines are also effective against established tumors and metastases. We tested the therapeutic activity of a proven prophylactic anti-HER-2/neu vaccine against successive stages of mammary carcinoma progression in HER-2/neu transgenic mice. The vaccine consisted of transgenic mammary carcinoma cells expressing HER-2/neu and two adjuvants: allogeneic class I histocompatibility antigens and interleukin (IL)-12. Vaccination of mice bearing lung micrometastases resulted in a 90% inhibition of metastasis development, whereas vaccination of mice with incipient local tumors was ineffective. The antimetastatic response was hampered by immune tolerance, as the protection of transgenic mice was lower than that of wild-type congenics not tolerant to HER-2/neu. A significant gain in immunotherapeutic activity in transgenic mice was obtained through the coadministration of anti-CD25 monoclonal antibody targeting regulatory T cells, which resulted in a >99% inhibition of metastasis. The immune responses elicited in transgenic mice comprised the activation of lung granulocytes and macrophages and of systemic adaptive responses based on helper T cells and their cytokines (IFN-gamma and IL-4) and anti-HER-2/neu antibodies. Dissection of relevant antimetastatic mechanisms by means of knockout mice and of depleting antibodies revealed a major difference between tumor prevention, which was completely dependent on anti-HER-2/neu antibodies, and metastasis therapy, which was antibody independent. In conclusion, a vaccine successfully developed for cancer immunoprevention showed a strong therapeutic activity against lung metastases mediated by protective immune mechanisms distinct from those preventing the onset of primary mammary carcinoma.
Collapse
Affiliation(s)
- Patrizia Nanni
- Section of Cancer Research, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Stewart TJ, Abrams SI. Altered immune function during long-term host-tumor interactions can be modulated to retard autochthonous neoplastic growth. THE JOURNAL OF IMMUNOLOGY 2007; 179:2851-9. [PMID: 17709499 DOI: 10.4049/jimmunol.179.5.2851] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Ag-specific and generalized forms of immunosuppression have been documented in animal tumor models. However, much of our knowledge on tumor-induced immunosuppression was acquired using tumor implant models, which do not reiterate the protracted nature of host-tumor interactions. Therefore, a transgenic mouse model of autochthonous mammary tumor development and progression was chosen to investigate the long-term consequences of neoplastic growth on the immune system. In vitro proliferation of unfractionated splenocytes from tumor-bearing mice, as assessed by [(3)H]thymidine uptake, was inhibited by the presence of suppressor cells within these splenocyte preparations, because purifying the T cells restored their biological activity. However, the level of inhibition did not correlate with either tumor load or the percentage of myeloid-derived CD11b+Gr1+ cells. To evaluate tumor-specific immune dysfunction, transgenic mice were challenged with autologous tumor cells. Mice with extensive, but not minimal autochthonous tumor burdens demonstrated a significantly enhanced rate of autologous tumor growth compared with age-matched controls. In contrast, an allogeneic tumor challenge was efficiently rejected from both groups of transgenic mice. It was also noted that allogeneic tumor challenge of mice with minimal disease significantly inhibited autochthonous primary tumor growth. We therefore demonstrated that 1) a generalized form of immunosuppression occurred, but not as a result of permanent alterations to T cell function, because purified T cell subsets retained normal biological activity following polyclonal or allostimulation; and 2) tumor-specific immunosuppression emerged as a consequence of tumor progression, but could be modulated to enhance antitumor responses against autochthonous primary neoplastic growth.
Collapse
Affiliation(s)
- Trina J Stewart
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, 10 Center Drive, Bethesda, MD 20892, USA
| | | |
Collapse
|
92
|
Abstract
When a vaccine-elicited immune response is directed against oncoantigens--proteins required for the neoplastic process--the chance that the tumour will evade the vaccine should be reduced. But how can these causal oncoantigens be identified? One approach is to find tumour-associated and microenvironment-associated oncoantigens required for progression from one tumour stage to the next by comparing gene signatures isolated from the different stages of tumour progression in cancer-prone transgenic mice. Mouse oncoantigens subsequently shown to be involved in human cancer can then be validated in mouse vaccination experiments. This provides the groundwork for the rational design of cancer vaccines for clinical trials.
Collapse
Affiliation(s)
- Federica Cavallo
- Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of Torino, Italy.
| | | | | |
Collapse
|
93
|
Wall EM, Milne K, Martin ML, Watson PH, Theiss P, Nelson BH. Spontaneous mammary tumors differ widely in their inherent sensitivity to adoptively transferred T cells. Cancer Res 2007; 67:6442-50. [PMID: 17616705 DOI: 10.1158/0008-5472.can-07-0622] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Immunotherapy of cancer can lead to the selection of antigen loss variants, which provides strong rationale to target oncogenes that are essential for tumor growth or viability. To investigate this concept, we tagged the HER2/neu oncogene with epitopes from ovalbumin to confer recognition by T-cell receptor transgenic CD8(+) (OT-I) and CD4(+) (OT-II) T cells. Transgenic mice expressing neu(OT-I/OT-II) developed mammary adenocarcinomas at 6 to 10 months of age. Adoptively transferred naive OT-I cells (with or without OT-II cells) proliferated vigorously on encountering neu(OT-I/OT-II)-expressing tumors. This was followed by the complete regression of 37% of tumors, whereas others showed partial/stable responses (40%) or progressive disease (23%). Those tumors undergoing complete regression never recurred. In mice with multiple primary tumors, simultaneous regressions and nonregressions were often seen, indicating that immune evasion occurred at a local rather than systemic level. The majority of nonregressing tumors expressed Neu(OT-I/OT-II) and MHC class I, and many avoided rejection through a profound block to T-cell infiltration. Thus, T cells directed against an essential oncogene can permanently eradicate a subset of spontaneous, established mammary tumors. However, in other tumors, local barriers severely limit the therapeutic response. To maximize the efficacy of immunotherapy against spontaneous cancers, predictive strategies that take into account the heterogeneity of the tumor microenvironment will be required.
Collapse
Affiliation(s)
- Erika M Wall
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada
| | | | | | | | | | | |
Collapse
|
94
|
Wang H, Yang G, Timme TL, Fujita T, Naruishi K, Frolov A, Brenner MK, Kadmon D, Thompson TC. IL-12 gene-modified bone marrow cell therapy suppresses the development of experimental metastatic prostate cancer. Cancer Gene Ther 2007; 14:819-27. [PMID: 17627292 DOI: 10.1038/sj.cgt.7701069] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
To investigate the immunomodulatory effects of interleukin-12 (IL-12) for treatment of metastatic prostate cancer, we administered adult bone marrow cells (BMC) that were genetically modified by retroviral vector-mediated IL-12 gene transduction in an experimental mouse model of prostate cancer metastasis. This therapy produced significant anti-metastatic effects in bone and lung and prolonged animal survival. Flow cytometric analysis indicated donor BMC could effectively home to bone and lung after treatment. Intensive infiltration of CD4 and CD8T cells in lung metastases and increased systemic natural killer and cytotoxic T lymphocyte activities indicated induction of a significant anti-metastatic immune response after treatment with IL-12 transduced BMC. Our results demonstrate the therapeutic potential of gene-modified BMC gene therapy.
Collapse
Affiliation(s)
- H Wang
- Scott Department of Urology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
95
|
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: 47] [Impact Index Per Article: 2.6] [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
|
96
|
De Giovanni C, Croci S, Nicoletti G, Landuzzi L, Palladini A, Pannellini T, Borgia L, Iezzi M, Di Carlo E, Orengo AM, Kennedy RC, Lollini PL, Nanni P, Musiani P. Inhibition of prostate carcinogenesis by combined active immunoprophylaxis. Int J Cancer 2007; 121:88-94. [PMID: 17290395 DOI: 10.1002/ijc.22586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The aim of this study is to investigate whether an active immunoprophylactic approach combining specific antigens and adjuvant stimuli would be able to inhibit prostate carcinogenesis in transgenic TRAMP mice. A vaccine consisting of allogeneic large T antigen (TAg)-positive SV40-transformed cells combined with systemic recombinant IL-12 was administered to TRAMP mice, starting from when they were still tumor-free at 5-6 weeks of age. The combined vaccine significantly inhibited prostate carcinogenesis, giving a more than doubled median latency time of prostatic tumors (53 weeks in comparison to 26 weeks in control mice). Vaccination with cells alone or IL-12 treatment alone was poorly effective (median latency of 30 and 39 weeks, respectively). The combined vaccine induced a very high CD4 response biased toward the Th1 pathway, with the induction of a humoral response that included TAg-specific antibodies. Therefore, such active immunoprophylactic approach based on the combination of allogeneic SV40 TAg-positive cells and systemic administration of recombinant IL-12 significantly delayed autochthonous urogenital carcinogenesis driven by SV40 TAg in TRAMP mice.
Collapse
Affiliation(s)
- Carla De Giovanni
- Cancer Research Section, Department of Experimental Pathology, University of Bologna, Bologna, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Singh R, Paterson Y. In the FVB/N HER-2/neu transgenic mouse both peripheral and central tolerance limit the immune response targeting HER-2/neu induced by Listeria monocytogenes-based vaccines. Cancer Immunol Immunother 2007; 56:927-38. [PMID: 17131121 PMCID: PMC11030683 DOI: 10.1007/s00262-006-0237-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2006] [Accepted: 10/12/2006] [Indexed: 10/23/2022]
Abstract
Listeria monocytogenes-based vaccines for HER-2/neu are capable of breaking tolerance in FVB/N rat HER-2/neu transgenic mice. The growth of implanted NT-2 tumors, derived from a spontaneously occurring tumor in the FVB/N HER-2/neu transgenic mouse, was significantly slower in these mice following vaccination with a series of L. monocytogenes-based vaccines for HER-2/neu. Mechanisms of T cell tolerance that exist in these transgenic mice include the absence of functional high avidity anti-HER-2/neu CD8(+) T cells and the presence of CD4(+)CD25(+) regulatory T cells. The in vivo depletion of these regulatory T cells resulted in the slowing in growth of tumors even without the treatment of mice with an anti-HER-2/neu vaccine. The average avidities of responsive CD8(+) T cells to six of the nine epitopes in HER-2/neu we examined, four of which were identified in this study, are shown here to be of a lower average avidity in the transgenic mice versus wild type FVB/N mice. In contrast, the average avidity of CD8(+) T cells to three epitopes that showed the lowest avidity in the wild-type mice did not differ between wild type and transgenic mice. This study demonstrates the ability of L. monocytogenes-based vaccines to impact upon tolerance to HER-2/neu in FVB/N HER-2/neu transgenic mice and further defines some of the aspects of tolerance in these mice.
Collapse
Affiliation(s)
- Reshma Singh
- Department of Microbiology, School of Medicine, University of Pennsylvania, 323 Johnson Pavilion, 36th St. and Hamilton Walk, Philadelphia, PA 19104-6076 USA
| | - Yvonne Paterson
- Department of Microbiology, School of Medicine, University of Pennsylvania, 323 Johnson Pavilion, 36th St. and Hamilton Walk, Philadelphia, PA 19104-6076 USA
| |
Collapse
|
98
|
Walewska R, Teobald I, Dunnion D, Abdulmajed H, Aldred M, Sadler J, Chapman C, Browning M. Preclinical development of hybrid cell vaccines for multiple myeloma. Eur J Haematol 2007; 78:11-20. [PMID: 17302859 DOI: 10.1111/j.1600-0609.2006.00769.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Immunotherapy may provide alternative or supplementary treatment of multiple myeloma (MM). We propose that hybrid cells, formed by fusing professional antigen-presenting cells with malignant plasma cells, would induce immune responses capable of mediating tumour regression. The human B-lymphoblastoid cell line, HMy2, was fused in vitro with CD138+ bead-separated myeloma plasma cells from five patients with MM. The hybrid cell lines generated in these studies grew stably in tissue culture, and maintained their phenotypic and functional characteristics, providing self-renewing cell lines with potential for therapeutic vaccination. The hybrid cells stimulated allogeneic and autologous T-cell proliferative responses in vitro to a considerably greater degree than their respective parent myeloma plasma cells, and directly activated both CD4+ and CD8+ T-cell responses. The enhanced T-cell stimulation correlated with expression of CD80 on the hybrid cells, and was inhibited by CTLA4-Ig fusion protein. The hybrid cell lines expressed several tumour-associated antigens known to be expressed in myeloma. These data show that self-replicating cell lines with enhanced immunostimulatory properties and potential for therapeutic vaccination can be generated by in vitro fusion of ex vivo myeloma cells and B-lymphoblastoid cell lines.
Collapse
Affiliation(s)
- Renata Walewska
- Department of Infection, Immunity & Inflammation, University of Leicester, Leicester, UK
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Airoldi I, Di Carlo E, Cocco C, Taverniti G, D'Antuono T, Ognio E, Watanabe M, Ribatti D, Pistoia V. Endogenous IL-12 triggers an antiangiogenic program in melanoma cells. Proc Natl Acad Sci U S A 2007; 104:3996-4001. [PMID: 17360466 PMCID: PMC1820697 DOI: 10.1073/pnas.0609028104] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The IL12RB2 gene acts as a tumor suppressor in human B cell malignancies. Indeed, Il12rb2 knockout (KO) mice develop spontaneously B cell tumors, but also lung epithelial tumors. This latter phenotype may be related to (i) impairment of host IL-12-mediated immunosurveillance and/or (ii) IL-12 inability to inhibit directly the growth of IL-12 unresponsive malignant cells. To address this issue, we transplanted IL-12R(+) B16 melanoma cells into syngeneic Il12rb2 KO mice with the following rationale: (i) these mice have severe defects in IFN-gamma production, as well as in cytotoxic T lymphocyte and natural killer cell cytotoxicity, and (ii) they produce but do not use IL-12 that can potentially bind to and target tumor cells only. Il12rb2 KO mice displayed higher endogenous serum levels of IL-12 and developed smaller B16 tumors than WT animals. These tumors showed reduced proliferation, increased apoptosis, and defective microvessel formation related to down-regulated expression of a set of proangiogenic genes previously unrelated to IL-12. Such effects depended on direct activity of endogenous IL-12 on tumor cells in KO mice, and hydrodynamic delivered IL-12 caused further reduced tumorigenicity of B16 cells in these mice. A previously undescribed mechanism of the IL-12 antitumor activity has been here identified and characterized.
Collapse
Affiliation(s)
- Irma Airoldi
- Laboratory of Oncology, G. Gaslini Institute, Largo G. Gaslini 5, 16148 Genoa, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
100
|
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: 63] [Impact Index Per Article: 3.5] [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
|