101
|
Wagner S, Jasinska J, Breiteneder H, Kundi M, Pehamberger H, Scheiner O, Zielinski CC, Wiedermann U. Delayed tumor onset and reduced tumor growth progression after immunization with a Her-2/neu multi-peptide vaccine and IL-12 in c-neu transgenic mice. Breast Cancer Res Treat 2007; 106:29-38. [PMID: 17203384 DOI: 10.1007/s10549-006-9469-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2006] [Accepted: 11/25/2006] [Indexed: 11/28/2022]
Abstract
Passive immunotherapy with monoclonal antibodies is a routinely performed but cost intensive treatment against certain cancers. Induction of humoral anti-tumor responses by active peptide immunization has therefore become a favorable treatment concept. We have recently identified three peptides representing B-cell epitopes of the extracellular domain of Her-2/neu each of them inducing Her-2/neu specific immune responses with anti-tumor activity in vitro. The present study was performed to evaluate the in vivo protective capacity of a combined vaccination with these three peptides in FVB/N transgenic mice spontaneously developing c-neu overexpressing breast cancers. The three Her-2/neu peptides coupled to tetanus toxoid were administered with or without addition of recombinant IL-12. At the time all untreated mice had developed tumors about 40% of peptide-immunized mice and nearly 60% of mice immunized with the peptide vaccine co-applied with IL-12 remained tumor free. Moreover, co-administration of IL-12 had a significant impact on the retardation of tumor progression. The enhanced anti-tumor efficacy of the vaccine by IL-12 was associated with a Th1 biased immune response as demonstrated by an increased IFN-gamma production in vitro and elevated Her-2-specific IgG levels. Our findings clearly demonstrate that this multi-peptide vaccine is effective in tumor prevention and support its use against minimal disease, drug-resistant tumors or even for prophylaxis against cancers overexpressing Her-2/neu.
Collapse
MESH Headings
- Adjuvants, Immunologic/pharmacology
- Adjuvants, Immunologic/therapeutic use
- Animals
- Antibodies/blood
- Antibodies/metabolism
- Antibody Formation
- Antibody Specificity
- Cancer Vaccines/pharmacology
- Cancer Vaccines/therapeutic use
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Epitopes, B-Lymphocyte/immunology
- Female
- Glycoproteins/genetics
- Glycoproteins/immunology
- Glycoproteins/metabolism
- Humans
- Immunoglobulin G/blood
- Interferon-gamma/blood
- Interleukin-12/pharmacology
- Interleukin-12/therapeutic use
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Rats
- Receptor, ErbB-2/immunology
- Recombinant Proteins/pharmacology
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Time Factors
- Vaccines, Subunit/pharmacology
- Vaccines, Subunit/therapeutic use
Collapse
Affiliation(s)
- Stefan Wagner
- Department of Pathophysiology, Center for Physiology and Pathophysiology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
102
|
Hamid O, Solomon JC, Scotland R, Garcia M, Sian S, Ye W, Groshen SL, Weber JS. Alum with Interleukin-12 Augments Immunity to a Melanoma Peptide Vaccine: Correlation with Time to Relapse in Patients with Resected High-Risk Disease. Clin Cancer Res 2007; 13:215-22. [PMID: 17200357 DOI: 10.1158/1078-0432.ccr-06-1450] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We attempted to augment immunity to melanoma antigens using interleukin-12 (IL-12) with aluminum hydroxide (alum) for sustained release or granulocyte macrophage colony-stimulating factor (GM-CSF) added to a multipeptide vaccine. EXPERIMENTAL DESIGN Sixty patients with high-risk resected melanoma were randomized to receive melanoma peptides gp100(209-217) (210M), MART-1(26-35) (27L), and tyrosinase(368-376) (370D) with adjuvant Montanide ISA 51 and either IL-12 at 30 ng/kg with alum (group A), IL-12 at 100 ng/kg with alum (group B), or IL-12 at 30 ng/kg with 250 mug GM-CSF (group C). RESULTS Three patients had stage IIC (5%), 50 had stage III (83%), and 7 had stage IV (12%) melanoma. Most toxicities were grade 1/2 and resolved rapidly. Significant toxicity included grade 3 colitis and visual changes and grade 3 headache resolving after stopping IL-12 but continuing peptide vaccine. A higher rate of post-vaccine 6-month immune response to gp100 and MART-1 was observed in group A (15 of 19) or B (19 of 20) that received IL-12 plus alum versus group C with IL-12/GM-CSF (4 of 21; P < 0.001). Post-vaccine enzyme-linked immunospot response rates to peptide analogues in group B were higher than group A (P = 0.031 for gp100 and P = 0.010 for MART-1); both were higher than group C (P < 0.001 for gp100 and P < 0.026 for MART-1). With a median of 24 months of follow-up, 23 patients have relapsed. Post-vaccine immune response to MART-1 was associated with relapse-free survival (P = 0.012). CONCLUSIONS IL-12 with alum augmented an immune response to melanoma antigens compared with IL-12 with GM-CSF. Immune response was associated with time to relapse.
Collapse
Affiliation(s)
- Omid Hamid
- The Angeles Clinic and Research Institute, Santa Monica, USA
| | | | | | | | | | | | | | | |
Collapse
|
103
|
Affiliation(s)
- B St Croix
- Tumor Angiogenesis Section, Mouse Cancer Genetics Program, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| |
Collapse
|
104
|
Pappalardo F, Motta S, Lollini PL, Mastriani E. Analysis of vaccine’s schedules using models. Cell Immunol 2006; 244:137-40. [PMID: 17442286 DOI: 10.1016/j.cellimm.2007.03.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Accepted: 02/11/2007] [Indexed: 11/23/2022]
Abstract
We present a further, yet important, step in applying Catania Mouse Model & simulator (SimTriplex) of immune system response to vaccination. In particular we show that, as immune response can induce toxicity, one can calibrate the vaccine administrations in such a way to avoid toxicity effects, keeping immunoprevention from cancer. This result increases the model's potential applications.
Collapse
Affiliation(s)
- F Pappalardo
- Faculty of Pharmacy, University of Catania, Italy.
| | | | | | | |
Collapse
|
105
|
Chiodoni C, Iezzi M, Guiducci C, Sangaletti S, Alessandrini I, Ratti C, Tiboni F, Musiani P, Granger DN, Colombo MP. Triggering CD40 on endothelial cells contributes to tumor growth. ACTA ACUST UNITED AC 2006; 203:2441-50. [PMID: 17043144 PMCID: PMC2118135 DOI: 10.1084/jem.20060844] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Inflammatory cells can either promote or inhibit tumor growth. Here we studied whether CD40, a key molecule for adaptive immune response, has any role in mammary carcinogenesis of BALB/NeuT transgenic tumor-prone mice. We transferred the HER2/neu oncogene into CD40-null background to obtain the CD40-KO/NeuT strain. CD40-KO/NeuT mice showed delayed tumor onset and reduced tumor multiplicity. BM (BM) transplantation experiments excluded a role of BM-derived cells in the reduced tumorigenicity associated with CD40 deficiency. Rather, CD40 expressed by endothelial cells (ECs) takes part to the angiogenic process. Accordingly, large vessels, well organized around the tumor lobular structures, characterize BALB/NeuT tumors, whereas tiny numerous vessels with scarce extracellular matrix are dispersed in the parenchyma of poorly organized CD40-KO/NeuT tumors. Activated platelets, which may interact with and activate ECs, are a possible source of CD40L. Their localization within tumor vessels prompted the idea of treating BALB/NeuT and CD40-KO/NeuT mice chronically with the anti-platelet drug clopidogrel, known to inhibit platelet CD40L expression. Treatment of BALB/NeuT mice reduced tumor growth to a level similar to CD40-deficient mice, whereas CD40-KO/NeuT mice treated or not showed the same attenuated tumor outgrowth, indicating that activated platelets are the likely source of CD40L in this model. Collectively, these data point to a participation of CD40/CD40L in the angiogenic processes associated with mammary carcinogenesis of BALB/NeuT mice.
Collapse
Affiliation(s)
- Claudia Chiodoni
- Immunotherapy and Gene Therapy Unit, Department of Experimental Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, 20133 Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
106
|
Abstract
This article discusses the current understanding of the interactions between tumors and cells of the immune system, particularly at the early stages of carcinogenesis. A growing body of data suggests that these interactions help shape the eventual development of tumors. Inflammation is a common feature of several cancers, and the immune system can serve as a two-edged sword against cancer, capable of supporting and suppressing cancer. Data from human studies show that the immune system is capable of detecting the smallest expansions of transformed cells, well before the development of clinical cancer. These advances suggest a need to change the current emphasis for harnessing antitumor immunity from therapy to prevention of cancers.
Collapse
Affiliation(s)
- Radek Spisek
- Laboratory of Tumor Immunology and Immunotherapy, The Rockefeller University, New York, NY 10021, USA
| | | |
Collapse
|
107
|
Ambrosino E, Spadaro M, Iezzi M, Curcio C, Forni G, Musiani P, Wei WZ, Cavallo F. Immunosurveillance of Erbb2 Carcinogenesis in Transgenic Mice Is Concealed by a Dominant Regulatory T-Cell Self-Tolerance. Cancer Res 2006; 66:7734-40. [PMID: 16885376 DOI: 10.1158/0008-5472.can-06-1432] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To assess the role of CD4(+)CD25(+)Foxp3(+) regulatory T (Treg) cells in overcoming immunosurveillance of Erbb2 (HER-2/neu) mammary lesions, we studied the effects of their sustained removal in BALB/c female mice made transgenic for the rat Erbb2 (r-Erbb2) oncogene (BALB-neuT mice), which develop multiple mammary carcinomas. During the progression of these lesions, Treg cells expand in the spleen, tumor draining lymph nodes, and tumors. Repeated administration of anti-CD25 antibodies extends tumor-free survival, reduces carcinoma multiplicity, and leads to the manifestation of a natural antibody and CTL-mediated reactivity against r-Erbb2. Loss of Foxp3(+) Treg cells during anti-CD25 treatment remarkably caused the disappearance of Gr1(+) immature myeloid cells, suggesting a cross-talk between these two inhibitory immune cell types. Treg cell expansion associated with r-Erbb2 overexpression may be seen as a physiologic response to dampen the immune reaction elicited by local anomalous overexpression of a self-antigen.
Collapse
MESH Headings
- Animals
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/immunology
- Female
- Immunoglobulin G/immunology
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mice
- Mice, Inbred BALB C
- Mice, SCID
- Mice, Transgenic
- NIH 3T3 Cells
- Rats
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Receptors, Interleukin-2/immunology
- Self Tolerance/immunology
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Elena Ambrosino
- Department of Clinical and Biological Sciences, University of Turin, Ospedale San Luigi Gonsaga, I-10043 Orbassano, Italy
| | | | | | | | | | | | | | | |
Collapse
|
108
|
Cywinski AL, Dunnion DJ, Teobald I, Tucker VC, Browning MJ. Hybrid cells formed by fusion of Epstein - Barr virus-associated B-lymphoblastoid cells and either marrow-derived or solid tumour-derived cell lines display different co-stimulatory phenotypes and abilities to activate allogeneic T-cell responses in vitro. ACTA ACUST UNITED AC 2006; 68:115-26. [PMID: 16866881 DOI: 10.1111/j.1399-0039.2006.00623.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A panel of stable cell hybrids was generated by fusing a range of marrow-derived and solid tumour-derived human cell lines with the B-lymphoblastoid cell lines, HMy2 or KR4, and expression of immunologically relevant accessory and co-stimulatory molecules, and ability to stimulate allogeneic T-cell responses in vitro was investigated. Hybrid cell lines generated from three marrow-derived tumour cells consistently expressed both MHC class I and class II molecules, a range of accessory and T-cell co-stimulatory ligand molecules, including CD80 and CD86, and directly stimulated markedly enhanced T-cell proliferative responses in vitro, as compared with the parent tumour cell lines. The responses were blocked by addition of CTLA4-Ig fusion protein to the cultures, indicating a role of CD28/B7 interaction in induction of T-cell activation. By contrast, hybrid cells derived from three solid tumours only expressed MHC class II when the parent tumour cell line expressed MHC class II and consistently failed to express CD80 or CD86. These hybrid cells also stimulated greater T-cell proliferative responses in vitro than the parent tumour cell lines, although effective co-stimulation depended on the presence of responder non-T cells in the cultures. The expression of co-stimulatory ligand molecules and ability to directly stimulate strong allogeneic T-cell responses correlated with the EBV latency type of the hybrid cells. These data suggest that phenotypic and functional differences in fusion cells of professional antigen- presenting cells and tumour cells arise as a result of the parent tumour cell type.
Collapse
Affiliation(s)
- A L Cywinski
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | | | | | | | | |
Collapse
|
109
|
Abstract
Despite tremendous progress in basic and epidemiological research, effective prevention of most types of cancer is still lacking. Vaccine use in cancer therapy remains a promising but difficult prospect. However, new mouse models that recapitulate significant features of human cancer progression show that vaccines can keep precancerous lesions under control and might eventually be the spearhead of effective and reliable ways to prevent cancer.
Collapse
Affiliation(s)
- Pier-Luigi Lollini
- Section of Cancer Research, Department of Experimental Pathology, University of Bologna, Italy
| | | | | | | |
Collapse
|
110
|
Cavallo F, Offringa R, van der Burg SH, Forni G, Melief CJM. Vaccination for Treatment and Prevention of Cancer in Animal Models. Adv Immunol 2006; 90:175-213. [PMID: 16730264 DOI: 10.1016/s0065-2776(06)90005-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Two approaches to immunological intervention in tumor-host interactions in mouse models are discussed in this review. The first is described with reference to experiments in which CD8(+) T lymphocytes are used to kill established transplantable tumors. Peptides and their optimal presentation by dendritic cells and intervention in immune regulatory mechanisms are the key issues for efficient induction of T-killer cell-mediated tumor eradication. The time frame of tumor therapy and the threat imposed by tumor growth in transplantable models and cancer patients require the induction of a robust T-cell reaction. Prevention of the progression of small preneoplastic lesions, on the other hand, requires the significant and prolonged immune protection sought in the second approach. This is based on antibody production and the coordinated activation of multiple low-avidity cell-mediated mechanisms elicited by DNA vaccination in genetically modified cancer-prone mice, transgenic for a mutant Her-2/neu growth factor receptor expressed at the plasma membrane surface of preneoplastic mammary gland epithelial cells. Vaccination with appropriate DNA formulations results in prolonged immune inhibition of the progression of preneoplastic mammary lesions but is ineffective against established tumors. The use of molecularly defined adjuvants and intervention in immune regulatory mechanisms are critical in both the elicitation of an effective T-cell mediated reaction required for tumor debulking in the first set of models and the induction by vaccination of a sustained immune memory able to prevent the expansion of preneoplastic lesions in genetically cancer-prone mice.
Collapse
Affiliation(s)
- Federica Cavallo
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | | | | | | | | |
Collapse
|
111
|
Tegerstedt K, Lindencrona JA, Curcio C, Andreasson K, Tullus C, Forni G, Dalianis T, Kiessling R, Ramqvist T. A single vaccination with polyomavirus VP1/VP2Her2 virus-like particles prevents outgrowth of HER-2/neu-expressing tumors. Cancer Res 2005; 65:5953-7. [PMID: 15994974 DOI: 10.1158/0008-5472.can-05-0335] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Murine polyomavirus (MPyV) VP1 virus-like particles (VLPs), containing a fusion protein between MPyV VP2 and the extracellular and transmembrane domain of HER-2/neu (Her2), Her2(1-683)PyVLPs, were tested for their ability to vaccinate against Her2-expressing tumors in two different in vivo models. Protection was assessed both against a lethal challenge with a BALB/c mammary carcinoma transfected with human Her2 (D2F2/E2) and against the outgrowth of autochthonous mammary carcinomas in BALB-neuT mice, transgenic for the activated rat Her2 oncogene. A single injection of Her2(1-683)PyVLPs before tumor inoculation induced a complete rejection of D2F2/E2 tumor cells in BALB/c mice. Similarly, a single injection of Her2(1-683)PyVLPs at 6 weeks of age protected BALB-neuT mice with atypical hyperplasia from a later outgrowth of mammary carcinomas, whereas all controls developed palpable tumors in all mammary glands. VLPs containing only VP1 and VP2 did not induce protection. The protection elicited by Her2(1-683)PyVLPs vaccination was most likely due to a cellular immune response because a Her2-specific response was shown in an ELISPOT assay, whereas antibodies against Her2 were not detected in any of the two models. The results show the feasibility of using MPyV-VLPs carrying Her2 fusion proteins as safe and efficient vaccines against Her2-expressing tumors.
Collapse
Affiliation(s)
- Karin Tegerstedt
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Centrum Karolinska, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Dela Cruz JS, Morrison SL, Penichet ML. Insights into the mechanism of anti-tumor immunity in mice vaccinated with the human HER2/neu extracellular domain plus anti-HER2/neu IgG3-(IL-2) or anti-HER2/neu IgG3-(GM-CSF) fusion protein. Vaccine 2005; 23:4793-803. [PMID: 15967544 DOI: 10.1016/j.vaccine.2005.04.041] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2004] [Revised: 03/31/2005] [Accepted: 04/04/2005] [Indexed: 11/29/2022]
Abstract
In the present study, we demonstrate that a physical association between the extracellular domain of human HER2/neu receptor (ECDHER2) plus anti-HER2/neu IgG3-(IL-2) or anti-HER2/neu IgG3-(GM-CSF) was required to elicit the most effective anti-tumor immune response against a syngeneic tumor expressing rat HER2/neu. Immune effectors including CD4+, CD8+, and NK cells contributed to protection against tumor growth. Vaccinated B-cell deficient mice did not elicit tumor protection, suggesting a critical role for B-cells in a protective immune response. These results provide insights into the mechanisms responsible for the protective tumor immunity elicited when antibody-(IL-2 or GM-CSF) are used as enhancers of vaccines targeting tumor antigens.
Collapse
Affiliation(s)
- Jay S Dela Cruz
- Department of Microbiology, Immunology and Molecular Genetics and Molecular Biology Institute, University of California, Box 148906, Los Angeles, CA 90095-1489, USA
| | | | | |
Collapse
|
113
|
Park JM, Terabe M, Sakai Y, Munasinghe J, Forni G, Morris JC, Berzofsky JA. Early role of CD4+ Th1 cells and antibodies in HER-2 adenovirus vaccine protection against autochthonous mammary carcinomas. THE JOURNAL OF IMMUNOLOGY 2005; 174:4228-36. [PMID: 15778385 DOI: 10.4049/jimmunol.174.7.4228] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
HER-2 is an oncogenic tumor-associated Ag that is overexpressed in several human tumors including breast and ovarian cancer. The efficacy and mechanism of a HER-2-expressing recombinant adenoviral vaccine to protect against tumorigenesis was examined using HER-2 transgenic (BALB-neuT) mice, which develop spontaneous breast tumors in all 10 mammary glands, and also using a transplantable mouse tumor model. Vaccination beginning at 6-8 wk of age (through 19 wk of age) prevented development of spontaneous mammary tumors even after 50 wk, whereas the animals in the control groups had tumors in all mammary glands by 25 wk. Such long-term protection after the last boost has not been achieved previously in this transgenic mouse in which the oncogene is continuously spawning tumorigenesis. Using beta(2)-microglobulin-knockout, IFN-gamma-knockout, and B cell-deficient mice, CD4(+) and CD8(+) cell depletion, and Ab transfer studies, we show that induction of anti-HER-2/neu Abs are both necessary and sufficient for protection, and the IgG2a isotype is most effective. In contrast, CD8(+) T cells are not necessary at all, and CD4(+) T cells are necessary for only 36-48 h after immunization to provide help for B cells but not as effector cells. Equal protection in immunized mice deficient in FcgammaRI/III excluded an FcR-mediated mechanism. Anti-HER-2 serum not only inhibited growth of mammary tumor cell lines expressing HER-2 in vitro but also protected mice from tumors in vivo, suggesting a direct action of Ab on the tumor cells. Such a vaccine may provide Ab-mediated protection against HER-2-expressing breast cancers in humans.
Collapse
MESH Headings
- Adenoviridae
- Animals
- Antibody Formation/drug effects
- Antigens, Neoplasm/administration & dosage
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/therapeutic use
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cancer Vaccines/standards
- Female
- Immunoglobulin G
- Mammary Neoplasms, Animal/immunology
- Mammary Neoplasms, Animal/prevention & control
- Mammary Neoplasms, Animal/therapy
- Mice
- Mice, Knockout
- Mice, Transgenic
- Neoplasm Transplantation/immunology
- Receptor, ErbB-2/administration & dosage
- Receptor, ErbB-2/immunology
- Receptor, ErbB-2/therapeutic use
- T-Lymphocytes/immunology
- Th1 Cells/immunology
- Time Factors
Collapse
Affiliation(s)
- Jong Myun Park
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
114
|
Preiss S, Kammertoens T, Lampert C, Willimsky G, Blankenstein T. Tumor-induced antibodies resemble the response to tissue damage. Int J Cancer 2005; 115:456-62. [PMID: 15700321 DOI: 10.1002/ijc.20914] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Tumor-associated antibodies are frequently detected in cancer patients. To ask whether the recognized antigens are rejection antigens, we screened a cDNA expression library of the mouse TS/A tumor with TS/A-immune serum and isolated 8 IgG-reactive clones, representing self-antigens that were expressed in normal tissues and other tumor lines. Three of the antigens had previously been identified in the human system by this cloning strategy. None of the antigens revealed to be a rejection antigen in normal mice demonstrated by an otherwise effective plasmid immunization. For one of the identified antigens, alpha-catenin, it is shown that the induction of IgG antibodies by protein immunization does not correlate with tumor rejection. For another antigen, vimentin, it is shown that vimentin-deficient but not vimentin-competent mice reject vimentin-expressing tumors indicating T -cell tolerance despite the fact that tumor cell immunization induces antivimentin IgG antibodies. Tissue damage induced by adenovirus infection induced an antibody response similar to tumor cell immunization, exemplified with 2 of the antigens. We conclude that the tumor-induced antibodies mirror tissue damage and that the antibody-inducing antigens can serve as rejection antigens if they are recognized as foreign.
Collapse
Affiliation(s)
- Susanne Preiss
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | | | | | | |
Collapse
|
115
|
Astolfi A, Rolla S, Nanni P, Quaglino E, De Giovanni C, Iezzi M, Musiani P, Forni G, Lollini PL, Cavallo F, Calogero RA. Immune prevention of mammary carcinogenesis in HER-2/neu transgenic mice: a microarray scenario. Cancer Immunol Immunother 2005; 54:599-610. [PMID: 15690207 PMCID: PMC11034326 DOI: 10.1007/s00262-004-0635-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2004] [Accepted: 10/01/2004] [Indexed: 12/22/2022]
Abstract
Neoplastic transformation is a multistep process in which gene products of specific regulatory pathways are involved at each stage. Identification of these overexpressed or mutated gene products provides an unprecedented opportunity to address the immune system against defined antigens and eliminate transformed cells. Mice transgenic for these oncogenes (e.g. HER-2/neu, a prototype of deregulated oncogenic protein kinase membrane receptors) are ideal experimental models for assessing the potential of active immunization. The demonstration that vaccines can cure HER-2/neu transplantable tumors, prevent their onset and delay the progression of preneoplastic lesions in mice at risk suggests that efficient immunological inhibition of HER-2/neu carcinogenesis can be achieved by specific vaccination. To further explore this issue, halting of tumor progression in the mammary glands of BALB-neuT mice with two immunization protocols in two laboratories has been studied independently by DNA microarray analysis. Combination of the two sets of results revealed a clear correlation between them when the tumor mass was titrated by transcription profiling. It was also clear that both protocols induced a strong, polyclonal antibody response and halted tumor growth at a condition very similar to that at which the vaccination began. Differences in the expression profiles were mainly related to the expression levels of a few chemokines and T-cell-specific genes that may be in some way correlated with the efficacy of the vaccination. Last, combination of the expression data with the protection results indicated that chronic vaccination is needed to maintain an active IFN-gamma-mediated response in the mammary gland.
Collapse
Affiliation(s)
- Annalisa Astolfi
- Cancer Research Section, Department of Experimental Pathology, University of Bologna, Bologna, 40126 Italy
| | - Simona Rolla
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Italy
| | - Patrizia Nanni
- Cancer Research Section, Department of Experimental Pathology, University of Bologna, Bologna, 40126 Italy
| | - Elena Quaglino
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Italy
| | - Carla De Giovanni
- Cancer Research Section, Department of Experimental Pathology, University of Bologna, Bologna, 40126 Italy
| | - Manuela Iezzi
- Aging Research Center, CeSi, G. D’Annunzio University Foundation, Chieti, 66013 Italy
| | - Piero Musiani
- Aging Research Center, CeSi, G. D’Annunzio University Foundation, Chieti, 66013 Italy
| | - Guido Forni
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Italy
| | - Pier-Luigi Lollini
- Cancer Research Section, Department of Experimental Pathology, University of Bologna, Bologna, 40126 Italy
| | - Federica Cavallo
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Italy
| | - Raffaele A. Calogero
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, 10043 Italy
| |
Collapse
|
116
|
O'Mara LA, Norian LA, Kreamalmeyer D, White JM, Allen PM. T cell-mediated delay of spontaneous mammary tumor onset: increased efficacy with in vivo versus in vitro activation. THE JOURNAL OF IMMUNOLOGY 2005; 174:4662-9. [PMID: 15814690 DOI: 10.4049/jimmunol.174.8.4662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Peripheral tolerance to shared Ags expressed on both tumors and normal self-tissues presents a major barrier to T cell-based immunotherapy as a treatment for cancer. To assess the activity of tumor-specific T cells against spontaneously arising carcinomas in the context of shared Ag expression, we developed a model system whereby an identified tumor Ag, tumor ERK (tERK), is expressed transgenically on both normal mammary tissue and spontaneous mammary carcinomas. Transfer of in vitro-activated, tERK-specific DUC18 T cells delayed spontaneous tumor development in tERK-expressing mice when T cells were given before the development of palpable carcinomas. However, antitumor activity mediated by in vitro-activated DUC18 T cells, as measured by responsiveness against a transplanted tERK-expressing fibrosarcoma challenge, was lost within days of transfer. This loss was due to expression of tERK as a self-Ag on normal tissues and was independent of the presence of mammary tumors. In contrast, transferred naive DUC18 T cells maintained a long-term protective function in tERK-expressing mice. Ten-fold fewer naive T cells activated in vivo were able to replicate the delay in spontaneous tumor development achieved by in vitro-activated T cells. These results are in contrast to our earlier studies using transplanted tumors alone, in which in vitro-activated DUC18 T cells were more efficacious than naive DUC18 T cells and highlight the need to perform tumor studies in the presence of tumor Ag expression on normal self-tissue.
Collapse
Affiliation(s)
- Leigh A O'Mara
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
117
|
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: 38] [Impact Index Per Article: 1.9] [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
|
118
|
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
|
119
|
Abstract
The concept that the immune system recognizes and controls cancer was first postulated over a century ago, and cancer immunity has continued to be vigorously debated and experimentally tested. Mounting evidence in humans and mice supports the involvement of cytokines in tumor initiation, growth, and metastasis. The idea that the immune system detects stressed, transformed, and frankly malignant cells underpins much of the excitement currently surrounding new cytokine therapies in cancer treatment. In this review, we define the contrasting roles that cytokines play in promoting tumor immunity, inflammation, and carcinogenesis. We also discuss the more promising aspects of clinical cytokine use in cancer patients.
Collapse
Affiliation(s)
- Mark J Smyth
- Cancer Immunology Program, Trescowthick Laboratories, Peter MacCallum Cancer Center (Peter Mac), Victoria, Australia.
| | | | | | | |
Collapse
|
120
|
Abstract
Sophisticated genetic technologies have led to the development of mouse models of human cancers that recapitulate important features of human oncogenesis. Many of these genetically engineered mouse models promise to be very relevant and relatively rapid systems for determining the efficacy of chemopreventive agents and their mechanisms of action. The validation of such models for chemoprevention will help the selection of appropriate agents for large-scale clinical trials and allow the testing of combination therapies.
Collapse
Affiliation(s)
- Jeffrey E Green
- Transgenic Oncogenesis Group, Laboratory of Cell Regulation and Carcinogenesis, National Cancer Institute, Besthesda, MD 20892, USA.
| | | |
Collapse
|
121
|
Spadaro M, Ambrosino E, Iezzi M, Di Carlo E, Sacchetti P, Curcio C, Amici A, Wei WZ, Musiani P, Lollini PL, Cavallo F, Forni G. Cure of Mammary Carcinomas in Her-2 Transgenic Mice through Sequential Stimulation of Innate (Neoadjuvant Interleukin-12) and Adaptive (DNA Vaccine Electroporation) Immunity. Clin Cancer Res 2005; 11:1941-52. [PMID: 15756020 DOI: 10.1158/1078-0432.ccr-04-1873] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Purpose: Whereas neoadjuvant therapy is emerging as a treatment option in early primary breast cancer, no data are available on the use of antiangiogenic and immunomodulatory agents in a neoadjuvant setting. In a model of Her-2 spontaneous mammary cancer, we investigated the efficacy of neoadjuvant interleukin 12 (IL-12) followed by “immune-surgery” of the residual tumor.
Experimental Design: Female BALB/c mice transgenic for the rat Her-2 oncogene inexorably develop invasive carcinomas in all their mammary glands by the 23rd week of age. Mice with multifocal in situ carcinomas received four weekly i.p. injections of 100 ng IL-12 followed by a 3-week rest. This course was given four times. A few mice additionally received DNA plasmids encoding portions of the Her-2 receptor electroporated through transcutaneous electric pulses.
Results: The protection elicited by IL-12 in combination with two DNA vaccine electroporations kept 63% of mice tumor-free. Complete protection of all 1-year-old mice was achieved when IL-12-treated mice received four vaccine electroporations. Pathologic findings, in vitro tests, and the results from immunization of both IFN-γ and immunoglobulin gene knockout transgenic mice and of adoptive transfer experiments all show that IL-12 augments the B- and T-cell response elicited by vaccination and slightly decreases the number of regulatory T cells. In addition, IL-12 strongly inhibits tumor angiogenesis.
Conclusions: In Her-2 transgenic mice, IL-12 impairs tumor progression and triggers innate immunity so markedly that DNA vaccination becomes effective at late points in time when it is ineffective on its own.
Collapse
Affiliation(s)
- Michela Spadaro
- Department of Clinical and Biological Sciences, University of Turin, Ospedale San Luigi Gonzaga, Regione Gonzole 10, I-10043 Orbassano, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
122
|
Lollini PL, De Giovanni C, Pannellini T, Cavallo F, Forni G, Nanni P. Cancer immunoprevention. Future Oncol 2005; 1:57-66. [PMID: 16555976 DOI: 10.1517/14796694.1.1.57] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Immunoprevention is a fresh approach to cancer prevention based on the stimulation of the immune system before tumor onset. Immunoprevention was effective in various models of carcinogen-induced or autochthonous tumor progression. Vaccines made of cells or DNA plasmids combined with appropriate adjuvants completely blocked mammary carcinogenesis in HER-2/neu transgenic mice. At variance with cancer immunotherapy, the mediators of immunoprevention are antibodies and T-cell-derived cytokines, rather than cytotoxic T-cells. Immunopreventive approaches and chemoprevention with tamoxifen or cyclooxygenase-2 inhibitors can be combined advantageously. The success obtained in preclinical studies suggests that cancer immunoprevention should progress to clinical testing.
Collapse
Affiliation(s)
- Pier-Luigi Lollini
- Cancer Research Section, Department of Experimental Pathology, University of Bologna, Viale Filopanti 22, I-40126 Bologna, Italy.
| | | | | | | | | | | |
Collapse
|
123
|
Abstract
The interferon-gamma (IFN-gamma)/interleukin-12 (IL-12) pathway is a pivotal player in the immune system and is central to controlling mycobacterial infections. We highlight the most recent and relevant advances in understanding this pathway and their repercussions on basic and clinical science. Human mutations in IFN-gamma receptor-1 (IFN-gammaR1), IFN-gammaR2, IL-12p40, IL-12 receptor-beta1, signal transducer and activator of transcription-1, and nuclear factor-kappaB essential modulator are analyzed in the context of genetic susceptibility to mycobacterial diseases. A diagnostic and therapeutic approach is described. The IFN-gamma/IL-12 pathway is central in immune control of both environmental and autochthonous challenges, as reflected in human mutations and animal models. Besides being crucial for mycobacterial control, the IFN-gamma/IL-12 pathway is also involved in the pathogenesis of autoimmune disease as well as tumor development and control. Genotype-phenotype correlations have been established for certain genes in this pathway, some of which have therapeutic implications.
Collapse
Affiliation(s)
- Sergio D Rosenzweig
- Division of Immunology, Department of Pediatrics, Hospital Nacional de Pediatría Juan P. Garrahan, Buenos Aires, Argentina
| | | |
Collapse
|
124
|
van Mierlo GJD, Boonman ZFHM, Dumortier HMH, den Boer AT, Fransen MF, Nouta J, van der Voort EIH, Offringa R, Toes REM, Melief CJM. Activation of dendritic cells that cross-present tumor-derived antigen licenses CD8+ CTL to cause tumor eradication. THE JOURNAL OF IMMUNOLOGY 2005; 173:6753-9. [PMID: 15557168 DOI: 10.4049/jimmunol.173.11.6753] [Citation(s) in RCA: 133] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The fate of naive CD8(+) T cells is determined by the environment in which they encounter MHC class I presented peptide Ags. The manner in which tumor Ags are presented is a longstanding matter of debate. Ag presentation might be mediated by tumor cells in tumor draining lymph nodes or via cross-presentation by professional APC. Either pathway is insufficient to elicit protective antitumor immunity. We now demonstrate using a syngeneic mouse tumor model, expressing an Ag derived from the early region 1A of human adenovirus type 5, that the inadequate nature of the antitumor CTL response is not due to direct Ag presentation by the tumor cells, but results from presentation of tumor-derived Ag by nonactivated CD11c(+) APC. Although this event results in division of naive CTL in tumor draining lymph nodes, it does not establish a productive immune response. Treatment of tumor-bearing mice with dendritic cell-stimulating agonistic anti-CD40 mAb resulted in systemic efflux of CTL with robust effector function capable to eradicate established tumors. For efficacy of anti-CD40 treatment, CD40 ligation of host APC is required because adoptive transfer of CD40-proficient tumor-specific TCR transgenic CTL into CD40-deficient tumor-bearing mice did not lead to productive antitumor immunity after CD40 triggering in vivo. CpG and detoxified LPS (MPL) acted similarly as agonistic anti-CD40 mAb with respect to CD8(+) CTL efflux and tumor eradication. Together these results indicate that dendritic cells, depending on their activation state, orchestrate the outcome of CTL-mediated immunity against tumors, leading either to an ineffective immune response or potent antitumor immunity.
Collapse
MESH Headings
- Adenovirus E1A Proteins/immunology
- Adenovirus E1A Proteins/metabolism
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Antigens, Viral, Tumor/immunology
- Antigens, Viral, Tumor/metabolism
- CD11c Antigen/biosynthesis
- CD40 Antigens/biosynthesis
- CD40 Antigens/immunology
- Cross-Priming/immunology
- Cytotoxicity, Immunologic
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Injections, Intralesional
- Injections, Intravenous
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/prevention & control
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Geertje J D van Mierlo
- Department of Immunohematology and Bloodtransfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
125
|
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
|
126
|
Croci S, Nicoletti G, Landuzzi L, De Giovanni C, Astolfi A, Marini C, Di Carlo E, Musiani P, Forni G, Nanni P, Lollini PL. Immunological Prevention of a Multigene Cancer Syndrome. Cancer Res 2004; 64:8428-34. [PMID: 15548714 DOI: 10.1158/0008-5472.can-04-2341] [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] [Indexed: 11/16/2022]
Abstract
Vaccines effectively prevent the onset of tumors in transgenic mice carrying activated oncogenes; however, human tumors are caused by combined alterations in oncogenes and oncosuppressor genes. We evaluated the impact of prophylactic vaccines in HER-2/neu transgenic, p53 wild-type/null mice that succumb to an aggressive cancer syndrome comprising mammary and salivary gland carcinomas and rhabdomyosarcoma. A vaccine made of allogeneic mammary carcinoma cells expressing HER-2/neu and interleukin 12 afforded long-term protection from tumor onset. Tumor prevention was mediated by T cell-derived cytokines, in particular gamma-interferon, and by anti-HER-2/neu antibodies. HER-2/neu expression was inhibited in target tissues of vaccinated mice, and somatic loss of the wild-type p53 allele did not occur. A highly effective vaccine against a single oncoprotein induced a powerful immune response that arrested multistep carcinogenesis in distinct target tissues.
Collapse
Affiliation(s)
- Stefania Croci
- Cancer Research Section, Department of Experimental Pathology, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Abstract
Reliable animal models are critical for evaluating immunotherapies and for defining tumor immunology paradigms. Tumor immunologists are moving away from traditional transplantation tumor systems because they do not adequately model human malignancies. Transgenic mouse models in which tumors arise spontaneously have been developed for most cancers. The models use one of three technologies: tissue-specific promoters to drive expression of SV40 large T antigen or tissue-specific oncogenes; deletion of tumor suppressor genes by gene targeting; or, conditional deletion of tumor suppressor genes or activation of oncogenes via Cre-lox technology. Knockin mice expressing human tumor antigens and gene-targeted mice with deletions for immunologically relevant molecules have been integral to advancing knowledge of the tumor-host relationship. Although animal models are becoming more sophisticated, additional improvements are needed so that more realistic models can be developed.
Collapse
Affiliation(s)
- Suzanne Ostrand-Rosenberg
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle Baltimore, MD 21250, USA.
| |
Collapse
|
128
|
Takeuchi N, Hiraoka SI, Zhou XY, Nagafuku M, Ono S, Tsujimura T, Nakazawa M, Yura Y, Hamaoka T, Fujiwara H. Anti–HER-2/neu Immune Responses Are Induced before the Development of Clinical Tumors but Declined following Tumorigenesis in HER-2/neu Transgenic Mice. Cancer Res 2004; 64:7588-95. [PMID: 15492286 DOI: 10.1158/0008-5472.can-04-1081] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
HER-2/neu oncogene products have been implicated as a potential target of T cell-mediated immune responses to HER-2/neu-induced tumors. Using HER-2/neu transgenic mice (oncomice), we investigated whether, and if so how, anti-HER-2/neu immune responses are induced and modulated in these oncomice from birth to tumor initiation. Female oncomice carrying the activated HER-2/neu oncogene displayed apparent hyperplasia in mammary glands at 10 weeks of age and developed mammary carcinomas around an average age of 26 weeks. Unfractionated spleen cells from 10- to 15-week-old oncomice that were cultured without any exogenous stimuli exhibited cytotoxicity against the F31 tumor cell line established from an HER-2/neu-induced mammary carcinoma mass. The final antitumor effectors were a macrophage lineage of cells. However, this effector population was activated, depending on the stimulation of oncomouse CD4(+) T cells with oncomouse-derived antigen-presenting cell (APC) alone or with wild-type mouse APC in the presence of F31 membrane fractions, suggesting the presence of HER-2/neu-primed CD4(+) T cells and HER-2/neu-presenting APC in 10- to 15-week-old oncomice. These antitumor cytotoxic responses were detected at approximately 5 weeks of age and peaked at age 10 to 15 weeks. However, the responses then declined at tumor-bearing stages in which the expression of target proteins could progressively increase. This resulted from the dysfunction of CD4(+) T cells but not of APC or effector macrophages. These results indicate that an anti-HER-2/neu CD4(+) T cell-mediated immune response was generated at the pretumorigenic stage but did not prevent tumorigenesis and declined after the development of clinical tumors.
Collapse
Affiliation(s)
- Noritami Takeuchi
- Department of Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Mocellin S, Rossi CR, Nitti D. Cancer vaccine development: on the way to break immune tolerance to malignant cells. Exp Cell Res 2004; 299:267-78. [PMID: 15350526 DOI: 10.1016/j.yexcr.2004.06.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2004] [Revised: 06/15/2004] [Indexed: 10/26/2022]
Abstract
Exploiting a naturally occurring defense system, the immunotherapeutic approach embodies an ideal nontoxic treatment for cancer. Despite the evidence that immune effectors can play a significant role in controlling tumor growth either in natural conditions or in response to therapeutic manipulation, the cascade of molecular events leading to tumor rejection by the immune system remains to be fully elucidated. Nevertheless, some recent tumor immunology advancements might drastically change the way to design the next generation of cancer vaccines, hopefully improving the effectiveness of this therapeutic approach. In the present work, we will focus on three main areas of particular interest for the development of novel vaccination strategies: (a) cellular or molecular mechanisms of immune tolerance to malignant cells; (b) synergism between innate and adaptive immune response; (c) tumor-immune system interactions within the tumor microenvironment.
Collapse
Affiliation(s)
- Simone Mocellin
- Department of Oncological and Surgical Sciences, University of Padova, Italy.
| | | | | |
Collapse
|
130
|
Nanni P, Landuzzi L, Nicoletti G, De Giovanni C, Rossi I, Croci S, Astolfi A, Iezzi M, Di Carlo E, Musiani P, Forni G, Lollini PL. Immunoprevention of Mammary Carcinoma in HER-2/neu Transgenic Mice Is IFN-γ and B Cell Dependent. THE JOURNAL OF IMMUNOLOGY 2004; 173:2288-96. [PMID: 15294941 DOI: 10.4049/jimmunol.173.4.2288] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A vaccine combining IL-12 and allogeneic mammary carcinoma cells expressing p185(neu) completely prevents tumor onset in HER-2/neu transgenic BALB/c mice (NeuT mice). The immune protection elicited was independent from CTL activity. We now formally prove that tumor prevention is mainly based on the production of anti-p185(neu) Abs. In the present studies, NeuT mice were crossed with knockout mice lacking IFN-gamma production (IFN-gamma(-/-)) or with B cell-deficient mice (microMT). Vaccination did not protect NeuT-IFN-gamma(-/-) mice, thus confirming a central role of IFN-gamma. The block of Ab production in NeuT-microMT mice was incomplete. About one third of NeuT-microMT mice failed to produce Abs and displayed a rapid tumor onset. By contrast, those NeuT-microMT mice that responded to the vaccine with a robust production of anti-p185(neu) Ab displayed a markedly delayed tumor onset. In these NeuT-microMT mice, the vaccine induced a lower level of IgG2a and IgG3 and a higher level of IgG2b than in NeuT mice. Moreover, NeuT-microMT mice failed to produce anti-MHC class I Abs in response to allogeneic H-2(q) molecules present in the cell vaccine. These findings show that inhibition of HER-2/neu carcinogenesis depends on cytokines and specific Abs, and that a highly effective vaccine can rescue Ab production even in B cell-deficient mice.
Collapse
Affiliation(s)
- Patrizia Nanni
- Cancer Research Section, Department of Experimental Pathology, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Airoldi I, Di Carlo E, Banelli B, Moserle L, Cocco C, Pezzolo A, Sorrentino C, Rossi E, Romani M, Amadori A, Pistoia V. The IL-12Rbeta2 gene functions as a tumor suppressor in human B cell malignancies. J Clin Invest 2004; 113:1651-9. [PMID: 15173892 PMCID: PMC419484 DOI: 10.1172/jci20303] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2003] [Accepted: 04/06/2004] [Indexed: 12/25/2022] Open
Abstract
The IL-12Rbeta2 gene is expressed in human mature B cell subsets but not in transformed B cell lines. Silencing of this gene may be advantageous to neoplastic B cells. Our objective was to investigate the mechanism(s) and the functional consequence(s) of IL-12Rbeta2 gene silencing in primary B cell tumors and transformed B cell lines. Purified tumor cells from 41 patients with different chronic B cell lymphoproliferative disorders, representing the counterparts of the major mature human B cell subsets, tested negative for IL-12Rbeta2 gene expression. Hypermethylation of a CpG island in the noncoding exon 1 was associated with silencing of this gene in malignant B cells. Treatment with the DNA methyltransferase inhibitor 5-Aza-2'-deoxycytidine restored IL-12Rbeta2 mRNA expression in primary neoplastic B cells that underwent apoptosis following exposure to human recombinant IL-12 (hrIL-12). hrIL-12 inhibited proliferation and increased the apoptotic rate of IL-12Rbeta2-transfected B cell lines in vitro. Finally, hrIL-12 strongly reduced the tumorigenicity of IL-12Rbeta2-transfected Burkitt lymphoma RAJI cells in SCID-NOD mice through antiproliferative and proapoptotic effects, coupled with neoangiogenesis inhibition related to human IFN-gamma-independent induction of hMig/CXCL9. The IL-12Rbeta2 gene acts as tumor suppressor in chronic B cell malignancies, and IL-12 exerts direct antitumor effects on IL-12Rbeta2-expressing neoplastic B cells.
Collapse
Affiliation(s)
- Irma Airoldi
- Laboratory of Oncology, G. Gaslini Institute, Genoa, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Airoldi I, Di Carlo E, Banelli B, Moserle L, Cocco C, Pezzolo A, Sorrentino C, Rossi E, Romani M, Amadori A, Pistoia V. The IL-12Rβ2 gene functions as a tumor suppressor in human B cell malignancies. J Clin Invest 2004. [DOI: 10.1172/jci200420303] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
133
|
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: 59] [Impact Index Per Article: 2.8] [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
|
134
|
Iinuma T, Homma S, Noda T, Kufe D, Ohno T, Toda G. Prevention of gastrointestinal tumors based on adenomatous polyposis coli gene mutation by dendritic cell vaccine. J Clin Invest 2004; 113:1307-17. [PMID: 15124022 PMCID: PMC398422 DOI: 10.1172/jci17323] [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] [Received: 11/06/2002] [Accepted: 02/25/2004] [Indexed: 11/17/2022] Open
Abstract
Here we describe the effect of immunization with dendritic cells loaded with syngeneic tumor cells (DC/Ts) by polyethylene glycol treatment, on tumor development in adenomatous polyposis coli (APC) gene mutant mouse models, APC1309 and APC(Min-/+), in which adenomatous polyps of the gastrointestinal tracts develop with a high incidence. Treatment with DC/Ts prevented the development of gastrointestinal tumors, and coadministration of DC/Ts and IL-12 caused a further reduction in tumor incidence. Splenocytes from APC1309 mice treated with DC/Ts and IL-12 showed no cytotoxic activity toward the tumor cells, but serum antibody specific to them was detected. IgG from the treated mice exhibited cytotoxic activity against the tumor cells in vitro. Predominance of Th2 cell response over Th1 response was also suggested by ELISPOT assays in the treated mice. Depletion in vivo of CD4(+) T cells, not CD8(+) T cells, by the intraperitoneal administration of corresponding mAb's decreased the antitumor effect of DC/T inoculation. Immunofluorescence microscopic studies showed that Ig was attached to tumor cells in mice treated with DC/Ts and IL-12. These findings indicate that DC/T vaccination prevents tumor development through APC gene mutation and that its preventive effects are mediated by humoral antitumor immunity.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/metabolism
- CD4-Positive T-Lymphocytes/immunology
- Cancer Vaccines/immunology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Cell Line, Tumor
- Dendritic Cells/immunology
- Gastrointestinal Neoplasms/pathology
- Gastrointestinal Neoplasms/prevention & control
- Genes, APC
- Interleukin-12/therapeutic use
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Fluorescence
- Mutation
- T-Lymphocytes, Cytotoxic/immunology
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th2 Cells/immunology
- Th2 Cells/metabolism
Collapse
Affiliation(s)
- Toshio Iinuma
- Department of Internal Medicine, Jikei University School of Medicine, Minato-ku, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
135
|
Mistiaen WP, Van Cauwelaert P, Muylaert P, Wuyts F, Harrisson F, Bortier H. Effect of prior malignancy on survival after cardiac surgery. Ann Thorac Surg 2004; 77:1593-7; discussion 1597. [PMID: 15111148 DOI: 10.1016/j.athoracsur.2003.08.049] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/28/2003] [Indexed: 11/24/2022]
Abstract
BACKGROUND The number of patients with a previously treated tumor, needing cardiac surgery is increasing. Whether this operation in these patients is justified is determined by the long-term outcome. METHODS Of 8620 patients referred for cardiac surgery, 205 had a documented malignant tumor. The time interval between the occurrence of the tumor and the cardiac surgery was recorded. These patients were matched with 205 patients without a tumor according to age, gender, comorbidity and type of cardiac surgery. The patients were followed retrospectively. A chi(2) Kaplan Meier and Cox' regression analysis were performed. RESULTS During follow-up, 95.8% of the patients were traced (2794 patient years). Univariate analysis showed that 5- and 10-year survival was better in patients without a malignant tumor in the history (0.91 +/- 0.02 versus 0.72 +/- 0.03 and 0.73 +/- 0.04 versus 0.40 +/- 0.05; p < 0.0001). For shorter time intervals, mortality for all causes and mortality due to the tumor increase significantly (p < 0.0001). Multivariate analysis identified 4 independent variables: a malignant tumor in the history (p < 0.001), chronic obstructive pulmonary disease (p = 0.003), age (p = 0.001), and impaired left ventricular function (p = 0.035) CONCLUSIONS A malignant tumor in the history is the most prognostic factor after cardiac surgery, but the operation is still rewarding. Fatal progression of the tumor is seen if the time interval between the occurrence of the malignant tumor and cardiac surgery is short. Other unfavorable factors are decreased left ventricular function, chronic obstructive pulmonary disease and high age.
Collapse
|
136
|
Quaglino E, Iezzi M, Mastini C, Amici A, Pericle F, Di Carlo E, Pupa SM, De Giovanni C, Spadaro M, Curcio C, Lollini PL, Musiani P, Forni G, Cavallo F. Electroporated DNA Vaccine Clears Away Multifocal Mammary Carcinomas in Her-2/neu Transgenic Mice. Cancer Res 2004; 64:2858-64. [PMID: 15087404 DOI: 10.1158/0008-5472.can-03-2962] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The transforming rat Her-2/neu oncogene embedded into the genome of virgin transgenic BALB/c mice (BALB-neuT) provokes the development of an invasive carcinoma in each of their 10 mammary glands. i.m. vaccination with DNA plasmids coding for the extracellular and transmembrane domains of the protein product of the Her-2/neu oncogene started when mice already display multifocal in situ carcinomas temporarily halts neoplastic progression, but all mice develop a tumor by week 43. By contrast, progressive clearance of neoplastic lesions and complete protection of all 1-year-old mice are achieved when the same plasmids are electroporated at 10-week intervals. Pathological findings, in vitro tests, and the results from the immunization of both IFN-gamma and immunoglobulin gene knockout BALB-neuT mice, and of adoptive transfer experiments, all suggest that tumor clearance rests on the combination of antibodies and IFN-gamma-releasing T cells. These findings show that an appropriate vaccine effectively inhibits the progression of multifocal preneoplastic lesions.
Collapse
MESH Headings
- Animals
- Antibody-Dependent Cell Cytotoxicity
- Carcinoma in Situ/genetics
- Carcinoma in Situ/immunology
- Carcinoma in Situ/therapy
- Electroporation/methods
- Female
- Genes, erbB-2/genetics
- Immunotherapy, Active/methods
- Immunotherapy, Adoptive/methods
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mice, Transgenic
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Elena Quaglino
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Chattergoon MA, Saulino V, Shames JP, Stein J, Montaner LJ, Weiner DB. Co-immunization with plasmid IL-12 generates a strong T-cell memory response in mice. Vaccine 2004; 22:1744-50. [PMID: 15068858 DOI: 10.1016/j.vaccine.2004.01.036] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Plasmid encoded exogenous IL-12 delivered as a DNA vaccine adjuvant has been shown to improve vaccine-induced immunity. In particular, pIL-12 greatly improves antigen (Ag)-specific cytotoxic tlymphocyte (CTL) activity in immunized mice. The longevity of this response has not previously been studied in detail. We have studied the effect of co-immunization with pIL-12 on HIV gp160 and Influenza A Hemeagglutinnin-specific memory immune responses. Mice co-immunized with pIL-12 and plasmid encoded antigens maintained a greater memory response than those immunized with the plasmid antigen alone which could be measured at least 6 months after vaccination. Further, this translated to an improved outcome after challenge of long term rested mice that were previously immunized. The strength of the immune response as well as the number of Ag-specific T-cells is proportional to the number of Ag-specific cells primed by the vaccination regimen.
Collapse
Affiliation(s)
- Michael A Chattergoon
- Department of Pathology, University of Pennsylvania School of Medicine, 422 Curie Blvd., 505 Stellar-Chance Laboratories, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
138
|
Lindencrona JA, Preiss S, Kammertoens T, Schüler T, Piechocki M, Wei WZ, Seliger B, Blankenstein T, Kiessling R. CD4+ T cell-mediated HER-2/neu-specific tumor rejection in the absence of B cells. Int J Cancer 2004; 109:259-64. [PMID: 14750178 DOI: 10.1002/ijc.11654] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
HER-2/neu (HER-2) is a cell surface proto-oncogene that is often overexpressed in carcinomas. Passive administration of anti-HER-2 antibodies in breast cancer patients has achieved promising results, but less is known about the role of antibodies in active immunization. We asked whether B cells/antibodies are needed for tumor immunity induced by plasmid (HER-2 and GM-CSF) immunization. HER-2 specific tumor immunity relied completely on both CD4+ and CD8+ T cells. IFN-gamma, and to a lesser extent IL-4, seemed to be crucial cytokines during tumor rejection. Protection was associated with production of anti-HER-2 IgG antibodies in B cell competent mice. After immunization, however, B cell-deficient mice rejected HER-2-expressing tumors as efficiently as control littermates. We conclude that T cells are the main effector cells in DNA vaccine induced immunity against HER-2 and that anti HER-2 antibodies are not necessary to elicit a protective anti tumor immune response in this model.
Collapse
Affiliation(s)
- Jan Alvar Lindencrona
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Quaglino E, Rolla S, Iezzi M, Spadaro M, Musiani P, De Giovanni C, Lollini PL, Lanzardo S, Forni G, Sanges R, Crispi S, De Luca P, Calogero R, Cavallo F. Concordant morphologic and gene expression data show that a vaccine halts HER-2/neu preneoplastic lesions. J Clin Invest 2004; 113:709-17. [PMID: 14991069 PMCID: PMC351320 DOI: 10.1172/jci19850] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2003] [Accepted: 12/23/2003] [Indexed: 11/17/2022] Open
Abstract
While much experimental data shows that vaccination efficiently inhibits a subsequent challenge by a transplantable tumor, its ability to inhibit the progress of autochthonous preneoplastic lesions is virtually unknown. In this article, we show that a combined DNA and cell vaccine persistently inhibits such lesions in a murine HER-2/neu mammary carcinogenesis model. At 10 weeks of age, all of the ten mammary gland samples from HER-2/neu-transgenic mice displayed foci of hyperplasia that progressed to invasive tumors. Vaccination with plasmids coding for the transmembrane and extracellular domain of rat p185neu followed by a boost with rp185neu+ allogeneic cells secreting IFN-gamma kept 48% of mice tumor free. At 22 weeks, their mammary glands were indistinguishable from those of 10-week-old untreated mice. Furthermore, the transcription patterns of the two sets of glands coincided. Of the 12,000 genes analyzed, 17 were differentially expressed and related to the antibody response. The use of B cell knockout mice as well as the concordance of morphologic and gene expression data demonstrated that the Ab response is the main mechanism facilitating tumor growth arrest. This finding suggests that a new way can be found to secure the immunologic control of the progression of HER-2/neu preneoplastic lesions.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/metabolism
- Cancer Vaccines
- Cell Membrane/metabolism
- Cluster Analysis
- Female
- Gene Expression Regulation
- Hyperplasia
- Immunohistochemistry
- Interferon-gamma/metabolism
- Mammary Neoplasms, Animal/prevention & control
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mice, Transgenic
- Microscopy, Confocal
- Models, Biological
- Neoplasm Transplantation
- Neoplasms/prevention & control
- Oligonucleotide Array Sequence Analysis
- Plasmids/metabolism
- Precancerous Conditions
- Protein Structure, Tertiary
- Rats
- Receptor, ErbB-2/biosynthesis
- Receptor, ErbB-2/genetics
- Time Factors
Collapse
Affiliation(s)
- Elena Quaglino
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Spadaro M, Lanzardo S, Curcio C, Forni G, Cavallo F. Immunological inhibition of carcinogenesis. Cancer Immunol Immunother 2004; 53:204-16. [PMID: 14722672 PMCID: PMC11032997 DOI: 10.1007/s00262-003-0483-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2003] [Accepted: 10/24/2003] [Indexed: 10/26/2022]
Abstract
The combination of new information provided by fundamental immunology, along with the refinement of genetic engineering techniques has given scientists the capacity to produce vaccines able to inhibit the growth of most if not every transplantable tumor. However, when faced with already established tumors, vaccines fail to afford any significant protection. Many studies are underway which seek to overcome this gloomy situation. However, another possibility is to follow the indications provided by a large quantity of experimental data and to evaluate the possibility of using immunotherapy to prevent the initial stages of tumor growth. Is it possible to prevent an autologous tumor by means of a vaccination performed before tumor onset? Could antitumor vaccines be a new form of preventive medicine in the wake of Jenner, Pasteur, and other pioneers? In this paper it is our intention to review the results obtained by our laboratory in the attempt to use natural and adaptive immunity in the control of carcinogenesis. Natural immunity boosted by IL-12 and IL-2 significantly hampers the progression of mammary lesions occurring in HER-2/neu transgenic mice genetically predestined to develop lethal mammary carcinomas. Specific immunity elicited by DNA vaccination provides a much stronger inhibition of the development of mammary lesions, and a significant number of transgenic mice are tumor free at 1 year of age. These experimental data suggest the possibility of using immunity as a means of controlling preneoplastic lesions and protecting healthy persons at risk of developing cancer.
Collapse
Affiliation(s)
- Michela Spadaro
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Italy
| | - Stefania Lanzardo
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Italy
| | - Claudia Curcio
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Italy
| | - Guido Forni
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Italy
| | - Federica Cavallo
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Italy
| |
Collapse
|
141
|
Jasinska J, Wagner S, Radauer C, Sedivy R, Brodowicz T, Wiltschke C, Breiteneder H, Pehamberger H, Scheiner O, Wiedermann U, Zielinski CC. Inhibition of tumor cell growth by antibodies induced after vaccination with peptides derived from the extracellular domain of Her-2/neu. Int J Cancer 2004; 107:976-83. [PMID: 14601058 DOI: 10.1002/ijc.11485] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The anti Her-2/neu monoclonal antibody Trastuzumab has strong inhibiting effects on tumor growth in vitro and in vivo and is therefore used for immunotherapy in breast cancer patients. Due to necessity of frequent applications, however, cost intensiveness of Trastuzumab treatment and its limited duration of affectivity, an active immunization inducing a perhaps preventive and long-term immunity to Her-2/neu remains a desirable goal. We attempted to induce anti Her-2/neu antibodies by peptide vaccination and to test their efficacy in inhibiting tumor cell growth in vitro. By computer aided analyses, 7 putative B cell epitopes of Her-2/neu were defined and synthesized. These peptide epitopes were coupled to tetanus toxoid and used for immunization in BALB/c mice. Among these peptides, immunizations with 2 single peptides or a combination of 2 peptides induced anti-peptide antibody levels, primarily of the IgG1 isotype. These antibodies were also directed against the native Her-2/neu antigen, as shown in precipitation assays and ELISA with cell lysates of the Her-2/neu overexpressing breast cancer cell line SK-BR-3. Isolated IgG fractions from immune sera incubated with SK-BR-3 cells led to a moderate inhibition of the tumor cell growth in vitro, as well as to complement dependent cell lyses comparable to that achieved by incubation with Trastuzumab. Moreover, peptide immunization in rabbits generated anti-Her 2-neu IgG that, in contrast to mouse sera, were able to mediate a 31-46% lysis of SK-BR-3 cells in ADCC experiments. We conclude from our data that immunization with Her-2/neu peptides successfully induced humoral immune response with anti-tumor activity in an animal model.
Collapse
|
142
|
Mitola S, Strasly M, Prato M, Ghia P, Bussolino F. IL-12 regulates an endothelial cell-lymphocyte network: effect on metalloproteinase-9 production. THE JOURNAL OF IMMUNOLOGY 2004; 171:3725-33. [PMID: 14500672 DOI: 10.4049/jimmunol.171.7.3725] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
IL-12 is key cytokine in innate immunity and participates in tumor rejection by stimulating an IFN-gamma-mediated response characterized by CD8(+) mediated-cytotoxicity, inhibition of angiogenesis, and vascular injury. We previously demonstrated that activated lymphocytes stimulated with IL-12 induced an angiostatic program in cocultured vascular endothelial cells. In this study, we have extended this observation showing that a reciprocal modulation of cellular responses occurs. Actually, the presence of endothelial cells enhanced the inhibitory effect of IL-12 on metalloproteinase-9 expression in activated PBMC as well as their ability to transmigrate across an extracellular matrix. IL-12 triggered intracellular signaling, as indicated by STAT-1 activation, appeared to mainly operative in activated CD4 (+) cells challenged with IL-12, but it was also initiated in CD8(+) lymphocytes in the presence of endothelial cells. On the other hand, stimulated PBMC reduced the expression and the activity of metalloproteinase-9, up-regulated that of tissue inhibitor metalloproteinase-1, and stimulated the STAT-1 pathway in cocultured endothelial cells. We used neutralizing Abs to show that the IFN-inducible protein 10 (CXCL10) and monokine-induced by IFN-gamma (CXCL9) chemokines produced by both PBMC and endothelial cells are pivotal in inducing these effects. Altogether these results suggest the existence of an IL-12-regulated circuit between endothelium and lymphocytes resulting in a shift of proteolytic homeostasis at site of tissue injury.
Collapse
MESH Headings
- Basement Membrane/enzymology
- Basement Membrane/immunology
- CD4-Positive T-Lymphocytes/immunology
- Cell Communication/immunology
- Cell Migration Inhibition
- Cell Movement/immunology
- Cells, Cultured
- Chemokine CXCL10
- Chemokine CXCL9
- Chemokines, CXC/biosynthesis
- Chemokines, CXC/physiology
- Chemotaxis, Leukocyte/immunology
- Coculture Techniques
- DNA-Binding Proteins/biosynthesis
- Endothelium, Vascular/cytology
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/immunology
- Extracellular Matrix/enzymology
- Extracellular Matrix/immunology
- Humans
- Intercellular Signaling Peptides and Proteins/biosynthesis
- Intercellular Signaling Peptides and Proteins/physiology
- Interleukin-12/pharmacology
- Interleukin-12/physiology
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/enzymology
- Leukocytes, Mononuclear/immunology
- Lymphocyte Subsets/cytology
- Lymphocyte Subsets/enzymology
- Lymphocyte Subsets/immunology
- Matrix Metalloproteinase 9/biosynthesis
- Matrix Metalloproteinase Inhibitors
- STAT1 Transcription Factor
- Signal Transduction/immunology
- Trans-Activators/biosynthesis
Collapse
Affiliation(s)
- Stefania Mitola
- Institute for Cancer Research and Treatment and. Department of Oncological Sciences, University of Torino, Candiolo, Italy.
| | | | | | | | | |
Collapse
|
143
|
Manjili MH, Wang XY, Chen X, Martin T, Repasky EA, Henderson R, Subjeck JR. HSP110-HER2/neu chaperone complex vaccine induces protective immunity against spontaneous mammary tumors in HER-2/neu transgenic mice. THE JOURNAL OF IMMUNOLOGY 2004; 171:4054-61. [PMID: 14530326 DOI: 10.4049/jimmunol.171.8.4054] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Heat shock proteins (HSPs) are shown to be strong immunoadjuvants, eliciting both innate and adaptive immune responses against cancers. HSP110 is related in sequence to HSP70 and is approximately 4-fold more efficient in binding to and stabilizing denatured protein substrates compared with HSP70. In the present study we evaluated the ability of a heat shock complex of HSP110 with the intracellular domain (ICD) of human HER-2/neu to elicit effective antitumor immune responses and to inhibit spontaneous mammary tumors in FVB-neu (FVBN202) transgenic mice. The HSP110-ICD complex was capable of breaking tolerance against the rat neu protein and inhibiting spontaneous mammary tumor development. This vaccine induced ICD-specific IFN-gamma and IL-4 production. Depletion studies revealed that CD8(+) T cells were involved in protection against challenge with mouse mammary tumors, whereas CD4(+) T cells revealed partial protection. Increased IgG2a Ab titer in the sera of tumor-free animals after vaccination and elevated CD4(+) CD25(+) regulatory T cells in the PBL of tumor-bearing animals suggested that IFN-gamma-producing Th1 cells may be responsible for partial protection of CD4(+) T cells against the mammary tumor challenge, whereas CD4(+)CD25(+) regulatory T cells (Th2 cells) may suppress the antitumor immune responses. Together, these results suggest that HSP110-ICD complex can elicit effective IFN-gamma-producing T cells against spontaneous mammary tumors and that up-regulation of CD4(+) CD25(+) regulatory T cells may prevent complete eradication of the tumor following immunotherapy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Epitopes, T-Lymphocyte/immunology
- Female
- HSP110 Heat-Shock Proteins
- HSP70 Heat-Shock Proteins/administration & dosage
- HSP70 Heat-Shock Proteins/immunology
- Humans
- Immune Tolerance/genetics
- Immunoglobulin G/biosynthesis
- Intracellular Fluid/immunology
- Lymphocyte Activation/genetics
- Lymphocyte Count
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Mice, Transgenic
- Protein Structure, Tertiary/genetics
- Rats
- Receptor, ErbB-2/administration & dosage
- Receptor, ErbB-2/biosynthesis
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Receptors, Interleukin-2/biosynthesis
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Th1 Cells/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Masoud H Manjili
- Department of Molecular and Cellular Biophysics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.
| | | | | | | | | | | | | |
Collapse
|
144
|
Ruiz de Almodóvar C, López-Rivas A, Ruiz-Ruiz C. Interferon-Gamma and TRAIL in Human Breast Tumor Cells. TRAIL (TNF-RELATED APOPTOSIS-INDUCING LIGAND) 2004; 67:291-318. [PMID: 15110183 DOI: 10.1016/s0083-6729(04)67016-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Induction of apoptosis in tumor cells by death receptor activation is a novel therapeutic strategy. However, in systemic antitumor treatments, severe toxic effects have been observed with tumor necrosis factor-alpha (TNF-alpha) and CD95 ligand. TNF-alpha causes a lethal inflammatory response and CD95L produces lethal liver damage. Preclinical studies in mice and nonhuman primates showed no systemic cytotoxicity upon injection of recombinant TNF-related apoptosis-inducing ligand (TRAIL) at doses that effectively suppressed solid tumors such as colon and mammary carcinomas. Although unwanted effects of some TRAIL preparations have been reported in normal cells, these data suggest that TRAIL could be a suitable approach in cancer therapy. However, several mechanisms of resistance to TRAIL-mediated apoptosis have been described in tumor cells such as lack of TRAIL apoptotic receptors, enhanced expression of TRAIL-decoy receptors, and expression of apoptosis inhibitors. In combination regimes, interferon-gamma (IFN-gamma) could provide a promising antitumor therapeutic approach as it has been described to enhance cellular susceptibility to apoptosis in a variety of tumor cells. The mechanism by which IFN-gamma promotes cell death seems to be via the regulation of the expression of different proteins involved in apoptosis. Altogether, these data suggest a combination strategy to selectively kill tumor cells that need to be further explored.
Collapse
Affiliation(s)
- Carmen Ruiz de Almodóvar
- Department of Cellular Biology and Immunology Instituto de Parasitología y Biomedicina Consejo Superior de Investigaciones Científicas Granada E-18001, Spain
| | | | | |
Collapse
|
145
|
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: 1.3] [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
|
146
|
Abstract
Overexpression of the human epidermal growth factor-2 (HER2) oncogene in human breast carcinomas has been associated with a more aggressive course of disease. The reason for this association is still unclear, although it has been suggested to rest in increased proliferation, vessel formation, and/or invasiveness. Alternatively, prognosis may not be directly related to the presence of the oncoprotein on the cell membrane, but instead to the breast carcinoma subset identified by HER2 overexpression and characterized by a peculiar gene expression profile. HER2 has also been associated with sensitivity to anthracyclins and resistance to endocrine therapy, suggesting that tyrosine kinase receptor and hormone receptor pathways represent two major proliferation pathways exclusively active in breast carcinomas, one sensitive to chemotherapeutic drugs and the other to antiestrogens. HER2 currently represents one of the most appropriate targets for specific therapy. Indeed, trastuzumab, a monoclonal antibody directed against the extracellular domain of HER2, is therapeutically active in HER2-positive breast carcinomas. However, a consistent number of HER2-positive tumors is not responsive to HER2-driven therapy, indicating the need for a better understanding of the mechanism of action of this new biological drug in vivo. While preclinical studies suggest antibody-dependent cell cytotoxicity as the major mechanism, determination of NK activity at the time of treatment remains mandatory, especially in patients treated with immunosuppressive drugs. The efficacy of prophylactic vaccination has been fully demonstrated in preclinical models, whereas ongoing studies of active immunotherapy using a variety of vaccination regimens against HER2 in tumor-bearing mice and patients have met with only moderate success.
Collapse
Affiliation(s)
- Sylvie Ménard
- Molecular Targeting Unit, Department of Experimental Oncology, Istituto Nazionale Tumori, 20133 Milan, Italy.
| | | | | | | |
Collapse
|
147
|
Melani C, Chiodoni C, Forni G, Colombo MP. Myeloid cell expansion elicited by the progression of spontaneous mammary carcinomas in c-erbB-2 transgenic BALB/c mice suppresses immune reactivity. Blood 2003; 102:2138-45. [PMID: 12750171 DOI: 10.1182/blood-2003-01-0190] [Citation(s) in RCA: 220] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Transgenic female mice expressing the transforming rat oncogene c-erbB-2 (HER-2/neu) under the mouse mammary tumor virus (MMTV) promoter (BALB-neuT) spontaneously develop mammary carcinomas with a progression resembling that of human breast cancer. In these mice, activating antitumor immunotherapy fails to induce T cell-mediated cytotoxicity, suggesting a suppression of the immune response. We found a direct correlation between tumor multiplicity and an increased proportion of Gr-1+ (Ly6G)/Mac-1+(CD11b)/ER-MP12+(CD31) immature myeloid cells in the peripheral blood (PB) and spleen, suggesting that tumor load profoundly affects overall BALB-neuT hematopoiesis. In fact, myeloid colony formation was increased in bone marrow (BM) and spleen. The immature myeloid cells displayed suppressive activity on host T lymphocytes, which progressively failed to respond to alloantigens and CD3 triggering, while maintaining the ability to proliferate in response to nonspecific mitogens. Transplantation of normal BM into BALB-neuT mice readily resulted in hypertrophic hematopoiesis with myeloid cell expansion. This persistent influence of the tumor was mediated through the release of vascular endothelial growth factor (VEGF) but not granulocyte-macrophage colony-stimulating factor (GM-CSF), and was down-modulated when tumor load was reduced but not when BM was transplanted. Together, the data obtained in the BALB-neuT model of naturally occurring carcinogenesis show that tumor-associated immune suppression is secondary to a more general alteration of host hematopoiesis, conditioned by tumor-secreted soluble factors.
Collapse
Affiliation(s)
- Cecilia Melani
- Immunotherapy and Gene Therapy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, via G. Venezian 1, 20133 Milan, Italy.
| | | | | | | |
Collapse
|
148
|
Wolpoe ME, Lutz ER, Ercolini AM, Murata S, Ivie SE, Garrett ES, Emens LA, Jaffee EM, Reilly RT. HER-2/neu-specific monoclonal antibodies collaborate with HER-2/neu-targeted granulocyte macrophage colony-stimulating factor secreting whole cell vaccination to augment CD8+ T cell effector function and tumor-free survival in Her-2/neu-transgenic mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:2161-9. [PMID: 12902523 DOI: 10.4049/jimmunol.171.4.2161] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
HER-2/neu is overexpressed in several cancers including 30% of breast carcinomas, and correlates with a poor outcome. HER-2/neu-transgenic (neu-N) mice that overexpress the non-transforming rat neu develop spontaneous mammary carcinomas and demonstrate immunotolerance to the neu protein similar to that observed in patients with neu-expressing cancers. In neu-N mice, neu-targeted vaccination induces weak T cell and negligible Ab responses sufficient to delay but not eradicate transplanted neu-expressing tumor. Here we demonstrate that passive infusion of neu-specific mAbs in sequence with whole cell vaccination significantly improves tumor-free survival over either modality alone. Importantly, treatment of neu-N mice with vaccine in combination with two distinct neu-specific Abs is particularly efficacious, preventing tumor in 70% and eradicating established tumor in 30% of neu-N mice. In vivo lymphocyte subpopulation depletion experiments demonstrate that the efficacy of Ab, alone or combined with vaccine, is dependent on both CD4(+) and CD8(+) T cells. Furthermore, the in vivo antitumor effects of vaccine and Ab are associated with a significant increase in the number and function of neu-specific CD8(+) T cells. Collectively, these observations suggest that similarly increased efficacy could be obtained by combining neu-targeted vaccination and neu-specific Abs such as trastuzumab (Herceptin) in patients with neu-expressing cancers.
Collapse
MESH Headings
- 3T3 Cells
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/therapeutic use
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cell Line
- Cytotoxicity, Immunologic/genetics
- Cytotoxicity, Immunologic/immunology
- Epitopes, T-Lymphocyte/biosynthesis
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Female
- Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
- Immunization Schedule
- Injections, Subcutaneous
- Lymphocyte Count
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/mortality
- Mammary Neoplasms, Experimental/prevention & control
- Mice
- Mice, Transgenic
- Rats
- Receptor, ErbB-2/biosynthesis
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Receptor, ErbB-2/metabolism
- Survival Analysis
- Ubiquitins/metabolism
- Up-Regulation/genetics
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Matthew E Wolpoe
- Department of Otolaryngology-Head and Neck Surgery, Graduate Program in Immunology, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Abstract
Whether vaccines are designed to prepare the immune system for the encounter with a pathogen or with cancer, certain common challenges need to be faced, such as what antigen and what adjuvant to use, what type of immune response to generate and how to make it long lasting. Cancer, additionally, presents several unique hurdles. Cancer vaccines must overcome immune suppression exerted by the tumour, by previous therapy or by the effects of advanced age of the patient. If used for cancer prevention, vaccines must elicit effective long-term memory without the potential of causing autoimmunity. This article addresses the common and the unique challenges to cancer vaccines and the progress that has been made in meeting them. Considering how refractory cancer has been to standard therapy, efforts to achieve immune control of this disease are well justified.
Collapse
Affiliation(s)
- Olivera J Finn
- Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute, E1040 Biomedical Science Tower, Pittsburgh, Pennsylvania 15261, USA.
| |
Collapse
|
150
|
Forni G, Curcio C, Spadaro M, Iliffe J, Quaglino E, Di Carlo E, Musiani P, Lollini PL. Immunization in tumor prevention. Int Immunopharmacol 2003; 3:1151-8. [PMID: 12860170 DOI: 10.1016/s1567-5769(03)00026-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Recent experimental data suggest that immunity can be activated to prevent tumors. The rationale for prevention is strong because the setting is one endowed with an immune system that is neither impaired by tumor and treatment-induced suppression, nor tolerant to tumor-associated antigens. Oncogenic growth factor receptors are tumor antigens and rational targets for specific immunoprevention. Successful prevention of mammary carcinomas in Her-2/neu transgenic mice is cited as an evidence of the validity of this approach. The specific properties of the Her-2/neu gene product as an antigen and the nature of the immune responses induced by effective preventive treatments are summarized.
Collapse
Affiliation(s)
- Guido Forni
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Italy.
| | | | | | | | | | | | | | | |
Collapse
|