1
|
Suryo Rahmanto Y, Bal S, Loh KH, Yu Y, Richardson DR. Melanotransferrin: search for a function. Biochim Biophys Acta Gen Subj 2011; 1820:237-43. [PMID: 21933697 DOI: 10.1016/j.bbagen.2011.09.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 08/29/2011] [Accepted: 09/05/2011] [Indexed: 10/17/2022]
Abstract
BACKGROUND Melanotransferrin was discovered in the 1980s as one of the first melanoma tumour antigens. The molecule is a transferrin homologue that is found predominantly bound to the cell membrane by a glycosyl-phosphatidylinositol anchor. MTf was described as an oncofoetal antigen expressed in only small quantities in normal tissues, but in much larger amounts in neoplastic cells. Several diseases are associated with expression of melanotransferrin, including melanoma and Alzheimer's disease, although the significance of the protein to the pathogenesis of these conditions remains unclear. SCOPE OF REVIEW In this review, we discuss the roles of melanotransferrin in physiological and pathological processes and its potential use as an immunotherapy. MAJOR CONCLUSIONS Although the exact biological functions of melanotransferrin remain elusive, a growing number of roles have been attributed to the protein, including iron transport/metabolism, angiogenesis, proliferation, cellular migration and tumourigenesis. GENERAL SIGNIFICANCE The high expression of melanotransferrin in several disease states, particularly malignant melanoma, remains intriguing and may have clinical significance. Further studies on the biology of this protein may provide new insights as well as potential therapeutic avenues for cancer treatment. This article is part of a Special Issue entitled Transferrins: Molecular mechanisms of iron transport and disorders.
Collapse
Affiliation(s)
- Yohan Suryo Rahmanto
- Department of Pathology and Bosch Institute, Blackburn Building (D06), University of Sydney, Sydney, New South Wales, 2006 Australia
| | | | | | | | | |
Collapse
|
2
|
Röhn TA, Reitz A, Paschen A, Nguyen XD, Schadendorf D, Vogt AB, Kropshofer H. A Novel Strategy for the Discovery of MHC Class II–Restricted Tumor Antigens: Identification of a Melanotransferrin Helper T-Cell Epitope. Cancer Res 2005; 65:10068-78. [PMID: 16267033 DOI: 10.1158/0008-5472.can-05-1973] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CD4+ helper T cells play a critical role in orchestrating host immune responses, including antitumor immunity. The limited availability of MHC class II-associated tumor antigens is still viewed as a major obstacle in the use of CD4+ T cells in cancer vaccines. Here, we describe a novel approach for the identification of MHC class II tumor-associated antigens (TAAs). By combining two-dimensional liquid chromatography and nanoelectrospray ionization tandem mass spectrometry, we developed a highly sensitive method for the detection of human leukocyte antigen (HLA)-DR-associated peptides of dendritic cells upon exposure to necrotic tumor cells. This approach led to the identification of a novel MHC class II-restricted TAA epitope derived from melanotransferrin. The epitope stimulated T cells derived from melanoma patients and healthy individuals and displayed promiscuity in HLA-DR restriction. Moreover, the same peptide was also presented by MHC class II-positive melanoma cells. This strategy may contribute to increase the number of tumor epitopes presented by MHC class II molecules and may support the development of more efficacious vaccines against cancer.
Collapse
Affiliation(s)
- Till A Röhn
- Pharmaceutical Research and Roche Center for Medical Genomics, F. Hoffmann La Roche Ltd., Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
3
|
Kaufman HL, Divgi CR. Optimizing Prostate Cancer Treatment by Combining Local Radiation Therapy with Systemic Vaccination. Clin Cancer Res 2005; 11:6757-62. [PMID: 16203760 DOI: 10.1158/1078-0432.ccr-05-0644] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Howard L Kaufman
- Department of Surgery, Columbia University College of Physicians & Surgeons, New York, New York 10032, USA.
| | | |
Collapse
|
4
|
Abstract
The development of vaccines for melanoma has been accelerated by the identification of melanoma-associated antigens, a better understanding of basic immunologic principles, and the ability to construct complex vectors for immunization. The location and context in which T-cell priming occurs significantly influences the type and magnitude of immune response. Furthermore, there is a delicate balance between the generation of tumor-specific immunity and the emergence of tumor escape variants. We have focused on the direct intra-tumoral delivery of poxvirus vaccines expressing costimulatory molecules as a strategy for overcoming local immunosuppression in the treatment of established melanoma. Poxviruses provide potent danger signals and, in the presence of costimulation, local administration provides a mechanism to prime tumor-specific T-cell responses. The clinical application of this approach will likely depend on the ability to induce systemic anti-tumor immunity following local injection and we are evaluating this in current clinical trials. These studies may have important implications for the design of vaccine strategies for melanoma and other tumors.
Collapse
Affiliation(s)
- Heidi Hörig
- Department of Surgery, Columbia University Medical Center, 177 Fort Washington Avenue, MHB 7SK, New York, NY 10032, USA
| | | |
Collapse
|
5
|
Zaloudik J, Li W, Jacob L, Kieny MP, Somasundaram R, Acres B, Song H, Zhang T, Li J, Herlyn D. Inhibition of tumor growth by recombinant vaccinia virus expressing GA733/CO17-1A/EpCAM/KSA/KS1-4 antigen in mice. Cancer Gene Ther 2002; 9:382-9. [PMID: 11960289 DOI: 10.1038/sj.cgt.7700452] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The human colorectal carcinoma (CRC)-associated GA733 antigen (Ag), also named CO17-1A/EpCAM/KSA/KS1-4, has been a useful target in passive immunotherapy of CRC patients with monoclonal antibody (mAb) and in active immunotherapy with anti-idiotypic antibodies or with recombinant protein. These approaches have targeted single epitopes (monoclonal anti-GA733 antibodies and anti-idiotypic antibodies) or extracellular domain epitopes (recombinant protein), primarily by B cells. To determine whether a reagent that induces immunity to a larger number of both B- and T-cell epitopes might represent a superior vaccine, we analyzed the capacity of full-length GA733 Ag expressing multiple potentially immunogenic epitopes and encoded by recombinant vaccinia virus (VV GA733-2) to induce humoral, cellular, and/or protective immunity in mice. VV GA733-2 induced Ag-specific antibodies that reacted predominantly to unknown epitopes on the Ag and lysed Ag-positive CRC targets in conjunction with murine peritoneal macrophages as effector cells. Immunized mice developed Ag-specific, proliferative and delayed-type hypersensitive lymphocytes. VV GA733-2 inhibited growth of ras-transformed syngeneic tumor cells expressing the human GA733 Ag in mice. These results suggest the potential of VV GA733-2 as a candidate vaccine for patients with CRC, possibly in combination with recombinant GA733-2-expressing adenovirus, which has been shown to induce cytolytic antibodies and T cells as well as tumor protective effects in mice. The combined vaccine approach may be superior to the use of either vaccine alone in patients who are pre-immune to both viruses.
Collapse
MESH Headings
- 3T3 Cells
- Animals
- Antibodies, Monoclonal/immunology
- Antibody-Dependent Cell Cytotoxicity/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/immunology
- Cell Division
- Epithelial Cell Adhesion Molecule
- Female
- Genetic Therapy/methods
- Humans
- Hypersensitivity, Delayed/immunology
- Immunity, Cellular
- Lymphocyte Activation
- Mice
- Mice, Inbred BALB C
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/therapy
- T-Lymphocytes/immunology
- Transfection
- Vaccinia virus/genetics
Collapse
Affiliation(s)
- Jan Zaloudik
- The Wistar Institute, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Ye Z, Hellström I, Hayden-Ledbetter M, Dahlin A, Ledbetter JA, Hellström KE. Gene therapy for cancer using single-chain Fv fragments specific for 4-1BB. Nat Med 2002; 8:343-8. [PMID: 11927939 DOI: 10.1038/nm0402-343] [Citation(s) in RCA: 118] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Monoclonal antibodies against the T-cell activation molecule 4-1BB have been effective in the treatment of established mouse tumors. To create a vaccine that stimulates the immune system similarly to the efficacious monoclonal anti-4-1BB antibody, 1D8, we constructed a vector encoding cell-bound single-chain Fv fragments from 1D8. We transfected the vector into cells from the K1735 melanoma, selected because of its low immunogenicity and very low expression of major histocompatibility complex class I. The transfected cells induced a strong type 1 T-helper cell response, for which CD4+ but not CD8+ T lymphocytes were necessary and that involved natural killer cells. Vaccinated mice rejected established wild-type K1735 tumors growing as subcutaneous nodules or in the lung. An analogous approach may be effective against micrometastases in human patients, including tumors whose expression of major histocompatibility complex class I is very low.
Collapse
Affiliation(s)
- Zhengmao Ye
- Pacific Northwest Research Institute, Seattle, Washington, USA
| | | | | | | | | | | |
Collapse
|
7
|
Chang JS, Choi MJ, Cheong HS, Kim K. Development of Th1-mediated CD8+ effector T cells by vaccination with epitope peptides encapsulated in pH-sensitive liposomes. Vaccine 2001; 19:3608-14. [PMID: 11395193 DOI: 10.1016/s0264-410x(01)00104-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
There have been many studies for tumor therapy mediated by cytotoxic T lymphocytes (CTL) that recognize tumor-associated antigen. It is generally accepted that CTL responses are induced when antigen is delivered into the cytosol. The pH-sensitive liposomes as vehicles are well known for their capacity to deliver the antigen into the cytosol. In this work, immunization of mice with CTL epitope peptides from Hantaan nucleocapsid protein (M6) or human papilloma virus E7 encapsulated in pH-sensitive liposomes induced effective antigen-specific CTL responses. The CTL responses induced by M6 peptide encapsulated in pH-sensitive liposomes blocked the formation of tumor mass from Hantaan NP transfected B16 melanoma cells in C57BL/6 mice and delayed the growth of preinoculated melanoma cells. During the blockade of the tumor growth, the CTL response was maintained for at least approximately 6 weeks, and the mice secreted Th1 type cytokines such as IL-2 and IFN-gamma. These results suggested that the pH-sensitive liposomes might provide an effective peptide delivery system for CTL-mediated tumor therapy.
Collapse
Affiliation(s)
- J S Chang
- Drug Delivery Research Lab., Mogam Biotechnology Research Institute, 341 Pojung-ri, Koosung-Myon, Yongin City, Kyonggi-do 449-910, South Korea
| | | | | | | |
Collapse
|
8
|
Pandey M, Mathew A, Nair MK. Cancer vaccines: a step towards prevention and treatment of cancer. Eur J Surg Oncol 1999; 25:209-14. [PMID: 10218468 DOI: 10.1053/ejso.1998.0629] [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: 02/02/2023] Open
Abstract
Breakthroughs in basic science and applied biology over the last quarter of a century have had a great deal of influence on our understanding of disease processes. The structure, function and importance of various lipid and protein molecules within cells is now well known and is central in future developments of diagnostic and therapeutic modalities. Since the discovery of the double helix DNA structure by Watson and Crick, molecular biology has come a long way enabling inroads to be made in manipulating DNA and evolving into the discipline of molecular oncology. For tumours having a viral carcinogenesis, preventive vaccines directed against viruses reduce the chance of tumour formation. Therapeutic vaccination, on the other hand, is less successful. To overcome the latter, various methods involving the use of tumour-specific antibodies, anti-idiotypic antibodies, peptides, proteins and carbohydrate products of human tumours, etc. have been tried. However, as the majority of human tumours are antigenic and not immunogenic, the problem remains. Recently, inoculation with DNA plasmids, encoding a variety of proteins, has been able to generate T-cell specific responses in vivo. This novel concept may help scientist to overcome the immunological tolerance and anti-tumour ineffectiveness induced by many human cancer cells and may lead to generation of tumour-specific vaccines.
Collapse
Affiliation(s)
- M Pandey
- Department of Surgical Oncology, Regional Cancer Centre, Medical College P.O., Trivandrum, Kerala, India.
| | | | | |
Collapse
|
9
|
Zbar AP, Lemoine NR, Wadhwa M, Thomas H, Snary D, Kmiot WA. Biological therapy: approaches in colorectal cancer. Strategies to enhance carcinoembryonic antigen (CEA) as an immunogenic target. Br J Cancer 1998; 77:683-93. [PMID: 9514045 PMCID: PMC2149974 DOI: 10.1038/bjc.1998.114] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- A P Zbar
- Academic Department of Colorectal Surgery, Hammersmith Hospital, London, UK
| | | | | | | | | | | |
Collapse
|
10
|
Anti-metastatic effect of oncolysates from murine melanoma cells transfected with recombinant vaccinia virus encoding human IL-2. Chin J Cancer Res 1997. [DOI: 10.1007/bf02974970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
11
|
Irvine KR, Chamberlain RS, Shulman EP, Surman DR, Rosenberg SA, Restifo NP. Enhancing efficacy of recombinant anticancer vaccines with prime/boost regimens that use two different vectors. J Natl Cancer Inst 1997; 89:1595-601. [PMID: 9362157 DOI: 10.1093/jnci/89.21.1595] [Citation(s) in RCA: 122] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The identification of tumor-associated antigens and the cloning of DNA sequences encoding them have enabled the development of anticancer vaccines. Such vaccines target tumors by stimulating an immune response against the antigens. One method of vaccination involves the delivery of antigen-encoding DNA sequences, and a number of recombinant vectors have been used for this purpose. To optimize the efficacy of recombinant vaccines, we compared primary and booster treatment regimens that used a single vector (i.e., homologous boosting) with regimens that used two different vectors (i.e., heterologous boosting). METHODS Pulmonary tumors (experimental metastases) were induced in BALB/c mice inoculated with CT26.CL25 murine colon carcinoma cells, which express recombinant bacterial beta-galactosidase (the model antigen). Protocols for subsequent vaccination used three vectors that encoded beta-galactosidase--vaccinia (cowpox) virus, fowlpox virus, naked bacterial plasmid DNA. Mouse survival was evaluated in conjunction with antibody and cytotoxic T-lymphocyte responses to beta-galactosidase. RESULTS Heterologous boosting resulted in significantly longer mouse survival than homologous boosting (all P<.0001, two-sided). Potent antigen-specific cytotoxic T lymphocytes were generated following heterologous boosting with poxvirus vectors. This response was not observed with any of the homologous boosting regimens. Mice primed with recombinant poxvirus vectors generated highly specific antibodies against viral proteins. CONCLUSIONS The poor efficacy of homologous boosting regimens with viral vectors was probably a consequence of the induction of a strong antiviral antibody response. Heterologous boosting augmented antitumor immunity by generating a strong antigen-specific cytotoxic T-lymphocyte response. These data suggest that heterologous boosting strategies may be useful in increasing the efficacy of recombinant DNA anticancer vaccines that have now entered clinical trials.
Collapse
Affiliation(s)
- K R Irvine
- Surgery Branch, Division of Clinical Sciences, National Cancer Institute, Bethesda, MD 20892-1502, USA
| | | | | | | | | | | |
Collapse
|
12
|
Abstract
Most of these therapies, although still in the infant stages of their development, offer the potential for major advances in colorectal cancer therapy. Gene therapy is an entirely new medicinal paradigm for the treatment of cancer. Currently, the clinical application of these methods is limited by the need for a more through understanding of cancer immunology and the availability of better vector systems for efficient and selective tumor gene transfer. As increasing numbers of scientists and clinicians address these issues, better therapies will likely emerge.
Collapse
Affiliation(s)
- S L Eck
- Department of Medicine, University of Pennsylvania Medical Center, Philadelphia, USA
| |
Collapse
|
13
|
Effects of granulocyte-macrophage colony stimulating factor gene encoded vaccinia virus vector on murine pulmonary metastatic melanoma. Chin J Cancer Res 1997. [DOI: 10.1007/bf02974714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
14
|
Parmiani G, Colombo MP, Melani C, Arienti F. Cytokine gene transduction in the immunotherapy of cancer. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 1997; 40:259-307. [PMID: 9217928 DOI: 10.1016/s1054-3589(08)60142-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- G Parmiani
- Gene Therapy Program, Istituto Nazionale Tumori, Milan, Italy
| | | | | | | |
Collapse
|
15
|
Boursnell ME, Rutherford E, Hickling JK, Rollinson EA, Munro AJ, Rolley N, McLean CS, Borysiewicz LK, Vousden K, Inglis SC. Construction and characterisation of a recombinant vaccinia virus expressing human papillomavirus proteins for immunotherapy of cervical cancer. Vaccine 1996; 14:1485-94. [PMID: 9014288 PMCID: PMC7130629 DOI: 10.1016/s0264-410x(96)00117-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The presence and consistent expression of the genes encoding the human papillomavirus (HPV) E6 and E7 proteins in the great majority of cervical tumours presents the opportunity for an immunotherapeutic approach for control of the disease. This report describes the construction and characterisation of a recombinant vaccinia virus designed to express modified forms of the E6 and E7 proteins from HPV16 and HPV18, the viruses most commonly associated with cervical cancer. The recombinant virus (designated TA-HPV) was based on the Wyeth vaccine strain of vaccinia, and was shown to express the desired gene products. Studies in mice indicated that the recombinant virus was less neurovirulent than the parental virus and was capable of inducing an HPV-specific CTL response. This pre-clinical evaluation has provided a basis for the initiation of human trials in cervical cancer patients.
Collapse
|
16
|
Moss B. Genetically engineered poxviruses for recombinant gene expression, vaccination, and safety. Proc Natl Acad Sci U S A 1996; 93:11341-8. [PMID: 8876137 PMCID: PMC38059 DOI: 10.1073/pnas.93.21.11341] [Citation(s) in RCA: 349] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Vaccinia virus, no longer required for immunization against smallpox, now serves as a unique vector for expressing genes within the cytoplasm of mammalian cells. As a research tool, recombinant vaccinia viruses are used to synthesize and analyze the structure-function relationships of proteins, determine the targets of humoral and cell-mediated immunity, and investigate the types of immune response needed for protection against specific infectious diseases and cancer. The vaccine potential of recombinant vaccinia virus has been realized in the form of an effective oral wild-life rabies vaccine, although no product for humans has been licensed. A genetically altered vaccinia virus that is unable to replicate in mammalian cells and produces diminished cytopathic effects retains the capacity for high-level gene expression and immunogenicity while promising exceptional safety for laboratory workers and potential vaccine recipients.
Collapse
Affiliation(s)
- B Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-0445, USA
| |
Collapse
|
17
|
Cole DJ, Wilson MC, Baron PL, O'Brien P, Reed C, Tsang KY, Schlom J. Phase I study of recombinant CEA vaccinia virus vaccine with post vaccination CEA peptide challenge. Hum Gene Ther 1996; 7:1381-94. [PMID: 8818725 DOI: 10.1089/hum.1996.7.11-1381] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- D J Cole
- MUSC Dept. of Surgery, Charleston, South Carolina, 29425, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Rao JB, Chamberlain RS, Bronte V, Carroll MW, Irvine KR, Moss B, Rosenberg SA, Restifo NP. IL-12 is an effective adjuvant to recombinant vaccinia virus-based tumor vaccines: enhancement by simultaneous B7-1 expression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 1996; 156:3357-65. [PMID: 8617961 PMCID: PMC1952187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
A number of cytokines and costimulatory molecules involved in immune activation have recently been identified including IL-12, a heterodimeric cytokine that supports the development of cell-mediated immunity, and B7-1, a costimulatory molecule involved in the activation of T lymphocytes. We explored the use of these immunomodulants as molecularly defined adjuvants in the function of recombinant anticancer vaccines using a murine model adenocarcinoma, CT26, transduced with a model Ag, beta-galactosidase (beta-gal). Although IL-12 given alone to mice bearing tumors established for 3 days did not have consistent antitumor activity, a profound therapeutic effect was observed when IL-12 administration was combined with a recombinant vaccinia virus (rVV) encoding beta-gal called VJS6. On the basis of the reported synergistic effects of IL-12 and the costimulatory molecule B7-1 (CD80) in vitro, we immunized mice with a double recombinant vaccinia encoding both the model tumor Ag and the costimulatory molecule B7-1, designated B7-1 beta-gal rVV. The adjuvant administration of IL-12 after immunization with this virus significantly enhanced survival in tumor-bearing animals. T cell subset depletions demonstrated that the in vivo activity of IL-12 was largely independent of CD4+ T lymphocytes, whereas the in vivo activity of a B7-1 rVV required both CD4+ and CD8+ T cells to elicit maximal therapeutic effect. To our knowledge, this is the first description of B7-1 and IL-12 cooperation in vivo and represents a novel strategy to enhance the efficacy of recombinant anticancer vaccines.
Collapse
Affiliation(s)
- J B Rao
- Howard Hughes Medical Institute-National Institutes of Health Research Scholars Program, Surgery Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Irvine KR, Rao JB, Rosenberg SA, Restifo NP. Cytokine enhancement of DNA immunization leads to effective treatment of established pulmonary metastases. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 1996; 156:238-45. [PMID: 8598468 PMCID: PMC2040336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
DNA immunization can result in the induction of Ag-specific cellular and humoral immune responses and in protective immunity in several Ag systems. To evaluate the utility of DNA-based immunization as a potential cancer treatment strategy, we employed an experimental murine tumor, CT26, expressing the model tumor-associated Ag, beta-galactosidase (beta-gal), designated CT26.CL25. A plasmid expressing beta-gal (pCMV/beta-gal) administered by particle-mediated gene delivery to the epidermis using a hand-held, helium-driven "gene gun" induced beta-gal-specific Ab and lytic responses. Immunization with this construct prevented the growth of pulmonary metastatic tumor, and the adoptive transfer of splenocytes generated by pCMV/beta-gal in vivo immunization and cultured in vitro with the beta-gal876-884 immunodominant peptide reduced the number of established pulmonary nodules. DNA immunization alone had little or no impact on the growth of established lung metastases. To enhance the function of DNA immunization for active immunotherapy, a panel of cytokines was added as adjuvants following DNA administration. Significant reduction in the number of established metastases was observed when human rIL-2, mouse rIL-6, human rIL-7, or mouse rIL-12 were given after DNA inoculation; mouse rIL-12 as an adjuvant had the most profound effect. These findings suggest that the cytokines involved in the activation and expansion of lymphocyte populations may improve the therapeutic effects of DNA immunization. Given the ease with which plasmid DNA can be prepared to high purity for safe use in humans with infectious diseases and cancers, DNA immunization administered together with cytokine adjuvant may be an attractive alternative to recombinant viral vaccines.
Collapse
Affiliation(s)
- K R Irvine
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
20
|
Abstract
The identification of tumor-associated antigens (TAA) recognized by T lymphocytes makes the development of antigen-specific synthetic and recombinant vaccines possible. The expression of TAA within a recombinant vector increases control over the kinetics and quantity, the molecular form, and the subcellular location of the immunogen delivered. The next generation of antitumor vaccines employs cytokines and costimulatory molecules expressed in concert with TAA that are capable of augmenting the activation and proliferation of antitumor immune responses. The ultimate goal of these new strategies, the treatment of established cancer, is now being realized in animal models.
Collapse
Affiliation(s)
- K R Irvine
- Surgery Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | | |
Collapse
|
21
|
Colombo MP, Rodolfo M. Tumor cells engineered to produce cytokines or cofactors as cellular vaccines: do animal studies really support clinical trials? Cancer Immunol Immunother 1995; 41:265-70. [PMID: 8536271 PMCID: PMC11037825 DOI: 10.1007/bf01517213] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/1995] [Accepted: 10/09/1995] [Indexed: 01/31/2023]
Affiliation(s)
- M P Colombo
- Division of Experimental Oncology D, Istituto Nazionale Tumori, Milano, Italy
| | | |
Collapse
|
22
|
Darrow TL, Abdel-Wahab Z, Seigler HF. Immunotherapy of Human Melanoma With Gene-Modified Tumor Cell Vaccines. Cancer Control 1995; 2:415-423. [PMID: 10862182 DOI: 10.1177/107327489500200505] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The incidence of melanoma in the United States is increasing at a faster rate than that of any other cancer. The prognosis for metastatic disease is poor, and more effective treatments for disseminated disease are needed. Since melanoma is one of the more immunogenic tumors, strategies have focussed on immune recognition. In vitro studies suggest that potent tumor-specific cytotoxic T cells can be induced against human melanoma. Melanoma specific T-cell activation depends on appropriate presentation to the immune system of recently defined melanoma-associated antigens presented in the context of self-HLA gene products. Full T-cell activation requires the co- stimulation by B7-CD28 interactions at the T-cell surface and the elaboration of immune cytokines to promote T-cell growth. Data from animal models of tumor-specific immunization with tumor cells engineered to express immune cytokines or the B7 co-stimulatory molecule suggest that gene therapy for human melanoma may be an effective means to treat disseminated disease.
Collapse
Affiliation(s)
- TL Darrow
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | |
Collapse
|
23
|
Herlyn D, Somasundaram R, Li W, Jacob L. Animal models of human-derived cancer vaccines. CELL BIOPHYSICS 1995; 27:15-30. [PMID: 7493396 DOI: 10.1007/bf02822524] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Preclinical cancer vaccine studies must address vaccine safety, immunogenicity, and efficacy, as well as mechanism of vaccine action. Animal models of vaccines employing human tumor-associated antigen or epitopes (TAA, TAE) differ fundamentally from those employing tumor-specific antigens or epitopes (TSA, TSE). TSA and TSE vaccines will most likely demonstrate similar toxicity, immunogenicity, and efficacy in both tumor-bearing animals and patients. In contrast, TAA/TAE immunizations may have to overcome a host's immunological tolerance to TAA/TAE expressed not only on tumor, but also on normal tissues; immunity to TAA/TAE will potentially target normal tissues and thus may induce autoimmunity. Various experimental models for human-derived TAA/TAE vaccines have been developed. These models include transgenic mice, mice with severe combined immunodeficiency (SCID), and non-human primates. Recently, unique animal models of TAA/TAE cancer vaccines have been developed, taking advantage of the discovery of animal tissue antigens with significant sequence homologies to human TAA/TAE. These models mimic perhaps most closely the situation in cancer patients.
Collapse
Affiliation(s)
- D Herlyn
- Wistar Institute, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
24
|
|
25
|
Bronte V, Tsung K, Rao JB, Chen PW, Wang M, Rosenberg SA, Restifo NP. IL-2 enhances the function of recombinant poxvirus-based vaccines in the treatment of established pulmonary metastases. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 1995; 154:5282-92. [PMID: 7730632 PMCID: PMC2041892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Neoplastic cells are generally poor immunogens. Transfection of the murine tumor CT-26 with beta-galactosidase (beta-gal), a protein from Escherichia coli, did not alter its growth rate in vivo, or its lethality, and did not elicit a measurable anti-beta-gal immune response. Immunization with beta-gal-expressing recombinant vaccinia viruses (rVV) elicited specific anti-beta-gal cytolytic T lymphocytes, but rVV-beta-gal was only marginally therapeutic when given to tumor-bearing mice. With the aim of expanding the immune response against beta-gal, used here as a model tumor Ag, we gave mice exogenous IL-2 starting 12 h after the poxvirus. The therapeutic effectiveness of the combination of poxvirus and IL-2 was far greater than either of these treatments alone. When the cDNA for IL-2 was inserted into the viral genome of the rVV construct to make a double recombinant (drVV), antitumor activity was further augmented. One mechanism of action may be the enhanced activation or expansion of cytotoxic T cells, because a marked increase in primary cytotoxic responses against vaccinia determinants was observed. Interestingly, other cytokines (mGM-CSF, mTNF-alpha, and mIFN-gamma) inserted into the rVV genome did not modify the efficacy of the rVV constructs. The increase in specific CTL responses against beta-gal by drVV expressing the tumor-associated Ags (TAA) and IL-2 was more pronounced in mice bearing the lacZ-transduced tumor than in those bearing the parental cell line, suggesting that the TAA presented by growing tumor cells can either pre-activate or otherwise amplify the immune response induced by the rVV. Unfortunately, in several long-term surviving mice, tumor recurred that no longer expressed beta-gal. These results indicate that treatment of disseminated tumors by using recombinant viruses expressing TAA can be enhanced by IL-2 provided exogenously, or encoded within the recombinant virus.
Collapse
Affiliation(s)
- V Bronte
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Irvine KR, McCabe BJ, Rosenberg SA, Restifo NP. Synthetic oligonucleotide expressed by a recombinant vaccinia virus elicits therapeutic CTL. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 1995; 154:4651-7. [PMID: 7722317 PMCID: PMC1976247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
P815A is a naturally occurring tumor rejection Ag of the methylcholanthrene-induced murine mastocytoma P815. The gene encoding the Ag P815A, designated P1A, is identical to that encoded in the normal genome of the DBA/2 mouse. A recombinant vaccinia virus (rVV) was constructed that expressed a synthetic oligonucleotide encoding the minimal determinant peptide of the tumor-associated Ag. Although the rVV recombinant expressing this mini-gene was recognized efficiently in vitro, it was an ineffective immunogen in vivo. The addition of an endoplasmic reticulum insertion signal sequence to the NH2 terminus of the minimal determinant resulted in a rVV that elicited CD8+ T cells that could lyse P815 mastocytoma cells in vitro and that were therapeutic in vivo. Recombinant viruses expressing synthetic oligonucleotide sequences preceded by the insertion signal sequences allow the expression of Ag directly into the endoplasmic reticulum, where binding to MHC class I molecules is most efficient. Vaccines based on synthetic oligonucleotides could be constructed with ease and rapidity but, most importantly, such constructs avoid the dangers associated with the expression of full length genes encoding TAA that are potentially oncogenic.
Collapse
Affiliation(s)
- K R Irvine
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
27
|
McCabe BJ, Irvine KR, Nishimura MI, Yang JC, Spiess PJ, Shulman EP, Rosenberg SA, Restifo NP. Minimal determinant expressed by a recombinant vaccinia virus elicits therapeutic antitumor cytolytic T lymphocyte responses. Cancer Res 1995; 55:1741-7. [PMID: 7536130 PMCID: PMC2248453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Anticancer vaccine strategies can now target intracellular antigens that are involved in the process of malignant transformation, such as oncogene products or mutated tumor suppressor genes. Fragments of these antigens, generally 8-10 amino acids in length and complexed with MHC class I molecules, can be recognized by CD8+ T lymphocytes (TCD8+). To explore the possibility of using a genetically encoded, minimally sized fragment of an intracellular antigen as an immunogen, we constructed a recombinant vaccinia virus encoding an 8-residue peptide derived from chicken ovalbumin that is known to associate with the mouse H-2Kb molecule. Compared to standard methods of immunization, recombinant molecule. Compared to standard methods of immunization, recombinant vaccinia virus expressing the minimal determinant as well as full length ovalbumin were the only approaches that elicited specific primary lytic responses in C57BL/6 mice against E.G7OVA, a transfectant of the murine thymoma EL4 containing the ovalbumin gene. Stimulating these effectors in vitro with OVA257-264 peptide induced H-2Kb-restricted TCD8+ that not only lysed but also specifically secreted IFN-gamma in response to an antigen. Furthermore, when transferred adoptively, these anti-OVA257-264 TCD8+ cells significantly reduced the growth of established ovalbumin-transfected tumors in a pulmonary metastasis model system. Synthetic transfected tumors in a pulmonary metastasis model system. Synthetic oligonucleotides encoding minimal antigenic determinants within expression constructs may be a useful approach for treatment of neoplastic disease, thus avoiding the potential hazards of immunizing with full-length cDNAs that are potentially oncogenic.
Collapse
Affiliation(s)
- B J McCabe
- Surgery Branch, National Cancer Institute, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Herlyn D, Somasundaram R, Zaloudik J, Li W, Jacob L, Harris D, Kieny MP, Ricciardi R, Gonczol E, Sears H. Cloned antigens and antiidiotypes. Hybridoma (Larchmt) 1995; 14:159-66. [PMID: 7590774 DOI: 10.1089/hyb.1995.14.159] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Both monoclonal and polyclonal antiidiotypic antibodies mimicking the human colorectal carcinoma (CRC) associated antigen CO17-1A/GA733 have induced antigen-specific humoral and cellular immunity in CRC patients. The immune responses may underlie the clinical responses observed in some of the treated patients. Recently, the CO17-1A/GA733 antigen has been molecularly cloned and expressed in baculo-, adeno-, and vaccinia viruses. In preclinical studies, these recombinant antigen preparations elicited specific humoral immunity (cytotoxic antibodies) and cellular immunity (DTH-reactive and proliferative T cells). Antibody titers elicited in animals by recombinant antigen were significantly higher than those elicited by antiidiotypes. The recombinant antigen has a potential as a vaccine for CRC patients.
Collapse
Affiliation(s)
- D Herlyn
- Wistar Institute, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Sivanandham M, Scoggin SD, Tanaka N, Wallack MK. Therapeutic effect of a vaccinia colon oncolysate prepared with interleukin-2-gene encoded vaccinia virus studied in a syngeneic CC-36 murine colon hepatic metastasis model. Cancer Immunol Immunother 1994; 38:259-64. [PMID: 8168121 PMCID: PMC11038170 DOI: 10.1007/bf01533517] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/1993] [Accepted: 11/02/1993] [Indexed: 01/29/2023]
Abstract
Vaccinia CC-36 murine colon oncolysate (VCO) prepared with interleukin-2-gene encoded recombinant vaccinia virus (IL-2VCO) was used in the treatment of a syngeneic murine colon adenocarcinoma (CC-36) hepatic metastasis to test the beneficial effect of the interleukin-2-gene encoded vaccinia virus over a control recombinant vaccinia virus in producing a vaccinia oncolysate tumor cell vaccine. Results suggest that the IL-2VCO treatment significantly reduced the hepatic tumor burden in comparison with the controls that received either IL-2-gene-encoded recombinant vaccinia virus or a plain recombinant vaccinia virus or vaccinia oncolysate prepared with the plain recombinant virus. The survival of mice treated with IL-2VCO was also improved in comparison with mice treated with other preparations. The induction of a cytolytic T lymphocyte response was examined to elucidate the mechanism of the induction of antitumor responses in IL-2VCO-treated mice. Fresh peripheral blood lymphocytes (PBL) isolated from IL-2VCO-treated mice showed a higher cytolytic activity against CC-36 tumor cell target when compared to PBL from the mice of other treatment groups, suggesting that the IL-2VCO induced an antitumor cytolytic T lymphocyte response. These results suggest that a vaccinia oncolysate, prepared with recombinant vaccinia virus encoding an immunomodulating cytokine gene will enhance antitumor responses in the host.
Collapse
Affiliation(s)
- M Sivanandham
- Department of Surgery Research Laboratory, St. Vincent's Hospital and Medical Center, New York, NY 10011
| | | | | | | |
Collapse
|
30
|
|
31
|
Affiliation(s)
- C Roth
- Unité de Biologie Moléculaire du Gène, U.277 Inserm, Institut Pasteur, Paris, France
| | | | | |
Collapse
|
32
|
Paoletti E, Tartaglia J, Cox WI. Immunotherapeutic strategies for cancer using poxvirus vectors. Ann N Y Acad Sci 1993; 690:292-300. [PMID: 8368744 DOI: 10.1111/j.1749-6632.1993.tb44017.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- E Paoletti
- Virogenetics Corporation, Troy, New York 12180
| | | | | |
Collapse
|
33
|
Magarian-Blander J, Domenech N, Finn OJ. Specific and effective T-cell recognition of cells transfected with a truncated human mucin cDNA. Ann N Y Acad Sci 1993; 690:231-43. [PMID: 7690213 DOI: 10.1111/j.1749-6632.1993.tb44012.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Epithelial cell mucin has been characterized as a tumor-specific antigen in patients with pancreatic and breast cancer. Mucins are high molecular weight glycoproteins consisting of a heavily glycosylated tandemly repeating 20-amino acid sequence. Aberrant glycosylation of mucins on carcinomatous epithelial cells leads to the exposure of novel core epitopes that are recognized by cytotoxic T lymphocytes (CTLs). We previously reported the establishment of mucin-specific CTL clones that recognize mucin expressed on the surface of EBV-immortalized B cells transfected with the mucin cDNA (MUC1). This recognition was characterized as major histocompatibility complex (MHC)-unrestricted, because of the multivalent nature of mucin. The transfectants had to be incubated with an inhibitor of O-linked glycosylation, phenyl-N-acetyl-alpha-galactosaminide (phenyl-GalNAc) in order to unmask the tandem repeat core epitope recognized by CTLs. In the present study, we examined whether mucin molecules with fewer tandem repeats are capable of MHC-unrestricted recognition by mucin-specific CTL clones. A mucin cDNA expression vector expressing a "truncated" mucin molecule that contains only two tandem repeats was constructed. We found that mucin-specific CTL clones recognize the "truncated" mucin on allogeneic target cells, showing that recognition in this case was MHC-unrestricted as well. In addition, CTL clones lysed "truncated" mucin transfectants significantly better than full-length mucin transfectants treated with phenyl-GalNAc, and controls. The "truncated" construct may represent an effective means of immunizing patients with breast and pancreatic cancer, enabling them to mount a strong and efficient immune response against mucin-bearing tumor cells.
Collapse
Affiliation(s)
- J Magarian-Blander
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, Pennsylvania 15261
| | | | | |
Collapse
|
34
|
Affiliation(s)
- K O Lloyd
- Memorial Sloan-Kettering Cancer Center, New York, New York 10021
| |
Collapse
|
35
|
Affiliation(s)
- K E Hellström
- Bristol-myers Squibb Pharmaceutical Research Institute, Seattle, Washington 98121
| | | | | | | |
Collapse
|
36
|
Irvine K, Schlom J. Induction of delayed-type hypersensitivity responses by monoclonal anti-idiotypic antibodies to tumor cells expressing carcinoembryonic antigen and tumor-associated glycoprotein-72. Cancer Immunol Immunother 1993; 36:281-92. [PMID: 7682893 PMCID: PMC11038909 DOI: 10.1007/bf01741166] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/1992] [Accepted: 10/28/1992] [Indexed: 01/26/2023]
Abstract
The use of anti-idiotypic antibodies as immunogens represents one potential approach to active specific immunotherapy of cancer. Two panels of syngeneic monoclonal anti-idiotypic antibodies were generated. One panel was directed against mAb CC49 and the other to mAb COL-1. mAb CC49 recognizes the pancarcinoma antigen (Ag), tumor-associated glycoprotein-72 (TAG-72), and mAb COL-1 recognizes carcinoembryonic antigen (CEA). Seven anti-idiotypic (AI) antibodies (Ab2) designated AI49-1-7 were generated that recognize the variable region of mAb CC49. These mAb were shown to inhibit the interaction of mAb CC49 (Ab1) with TAG-72 (Ag). Five anti-idiotypic antibodies designated CAI-1-5 were also generated to the anti-CEA mAb, COL-1 (Ab1). These Ab2 were shown to inhibit the interaction between COL-1 (Ab1) and CEA (Ag). Immunization of mice, rats, and rabbits with Ab2 directed against CC49 or COL-1 could not elicit specific Ab3 humoral immune responses, i.e., antibody selectively reactive with their respective target antigens. However, immunization of mice with the CC49 anti-idiotypic antibody (Ab2), designated AI49-3, could induce a delayed-type hypersensitivity response (DTH) specific for tumor cells that express TAG-72. Similarly, immunization of mice with an anti-idiotypic antibody directed against COL-1, designated CAI-1, could induce specific DTH cell-mediated immune responses to murine tumor cells that express human CEA on their surface. These results thus demonstrate that while some anti-idiotype mAb may not be potent immunogens in eliciting Ab3 humoral responses, they are capable of eliciting specific cellular immune responses against human carcinoma-associated antigens. This type of mAb may ultimately be useful in active immunotherapy protocols for human carcinoma.
Collapse
Affiliation(s)
- K Irvine
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, Md 20892
| | | |
Collapse
|
37
|
Hand PH, Robbins PF, Salgaller ML, Poole DJ, Schlom J. Evaluation of human carcinoembryonic-antigen (CEA)-transduced and non-transduced murine tumors as potential targets for anti-CEA therapies. Cancer Immunol Immunother 1993; 36:65-75. [PMID: 8425211 PMCID: PMC11037972 DOI: 10.1007/bf01754404] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/1992] [Accepted: 08/19/1992] [Indexed: 01/30/2023]
Abstract
The MC-38 C57BL/6 mouse colon adenocarcinoma cell line has been transduced with a retroviral construct containing cDNA encoding the human carcinoembryonic antigen (CEA) gene [Robbins PF, Kantor JA, Salgaller M, Horan Hand P, Fernsten PD, Schlom J (1991) Cancer Res 51: 3657]. Two clones, MC-38-ceal and MC-38-cea2, expressed high levels of CEA on their cell surface. A third CEA-expressing cell line, MCA-102-cea3, was similarly derived by transduction of the MCA-102 C57BL/6 mouse fibrosarcoma cell line and is described here. In this study, the three CEA-transduced murine tumor cell lines (MC-38-ceal, MC-38-cea2, MCA-102-cea3) were evaluated for their tumorigenic potential, as well as their ability to serve as in vivo model systems for active and passive immunotherapy studies. Parameters that were investigated include tumor growth rate, the antibody response of the host to CEA, and the CEA content of the tumors. The MC-38-cea2 model appeared to be the most appropriate for immunotherapy studies. Biodistribution studies, using an 125I-labeled anti-CEA mAb, demonstrated efficient tumor targeting of MC-38-cea2 tumors in C57BL/6 and athymic mice.
Collapse
Affiliation(s)
- P H Hand
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | | | | | | | | |
Collapse
|
38
|
Hayashibe K. Melanoma-associated antigen synthesized in vitro for active specific immunotherapy. J Dermatol 1992; 19:823-6. [PMID: 1293170 DOI: 10.1111/j.1346-8138.1992.tb03790.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The immunogenicity of the antigen molecule is a prerequisite for active specific immunotherapy for melanoma. Since most of the melanoma-associated antigens recognized by the murine immune system are known to be not immunogenic in man, a detection and analysis system for melanoma-associated antigens is required to reflect in vivo immune responses in patients with melanoma. One of the promising approaches, an attempt to develop human monoclonal antibodies from B lymphocytes of patients with melanoma, has met with limited success due to the difficulties of producing large amounts of antibodies and using them in immunochemical assays, because most of them belong to the IgM class and have low affinity. Our approach is to utilize the screening of a cDNA expression library constructed from mRNA extracted from cultured melanoma cells with antibodies from patients with melanoma. The cloned cDNA, designated as D-1, had 1029 bp and showed no significant homology with viral and mammalian sequences stored in GENETYX. cDNA D-1 hybridized to a 2.0 kb mRNA species from 3 different cell lines of human melanoma, neuroblastoma, erythroleukemia, B lymphoid, and T lymphoid cells, but not from a renal carcinoma cell line, normal peripheral lymphocytes, or normal fibroblasts. The in vivo expression and distribution of mRNA related to cDNA D-1 has been examined in tissue specimens by in situ hybridization and shown to be rather restricted on melanoma cells. The polypeptide antigen encoded by cDNA D-1 may be a valuable immunogen for implementing active specific immunotherapy in patients with melanoma.
Collapse
Affiliation(s)
- K Hayashibe
- Department of Dermatology, Kobe University School of Medicine, Japan
| |
Collapse
|
39
|
Abstract
Melanoma cells have surface markers that are expressed differently than in normal melanocytes and nevus cells. Monoclonal antibodies may define a phenotypic map of the various melanocytic lesions and can be used in immunohistopathology and immunoscintigraphy. Monoclonal antibodies directed against melanoma-associated glycoproteins and glycolipids are being tested for therapy. Rearrangements or deletions on chromosome 1, 6, and 7 are the most frequently observed cytogenetic abnormalities. Molecular studies have not given a clear picture. A subset of HRAS alleles has been reported to be associated with melanoma. NRAS activation by point mutation has been found in one fourth of the cases. Allele losses at different loci have been reported. Genetic linkage studies have given conflicting results on the presence of a gene for the melanoma-dysplastic nevus syndrome on the short arm of chromosome 1.
Collapse
Affiliation(s)
- G Della Porta
- Division of Experimental Oncology A, Istituto Nazionale Tumori, Milan, Italy
| |
Collapse
|
40
|
|
41
|
Garratt RC, Jhotí H. A molecular model for the tumour-associated antigen, p97, suggests a Zn-binding function. FEBS Lett 1992; 305:55-61. [PMID: 1633859 DOI: 10.1016/0014-5793(92)80654-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The primary structure of p97 (melanotransferrin) has been compared with other members of the transferrin superfamily. A molecular structure of p97 has been modelled based on the crystal structure of diferric rabbit serum transferrin. The most significant amino acid substitutions in p97 are almost exclusively limited to only two regions; the C-lobe iron-binding cleft and the interlobe contact region. The latter includes within the N-terminal lobe a Zn-binding consensus sequence found in metallopeptidases, and in the C-terminal lobe a glutamic acid residue (Glu-394) capable of completing a potential thermolysin-like Zn-binding site. Thus, p97 may have a Zn-binding potential, unique amongst the transferrin superfamily.
Collapse
Affiliation(s)
- R C Garratt
- Departamento de Física y Ciência dos Materials, Universidade de São Paulo, Brazil
| | | |
Collapse
|
42
|
Stevenson FK. Update on tumor vaccines. INTERNATIONAL JOURNAL OF CLINICAL & LABORATORY RESEARCH 1992; 22:84-9. [PMID: 1504310 DOI: 10.1007/bf02591402] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Vaccination against tumor has always been an attractive idea for the treatment of patients bearing tumor. By harnessing the host's own immune response the attack on tumor cells would act on a continuing basis, with emerging tumor cells stimulating their own destruction. However, the approach has been hampered by our poor understanding of the nature of tumor antigens and of the pathways by which immune cells might operate against tumor growth. Recent developments in molecular biology and immunology are remedying this deficiency and bringing vaccination to the forefront of new approaches to treatment of a range of tumors. Results obtained in B-cell tumors, where the idiotypic immunoglobulin at the cell surface provides a well-defined tumor antigen, are already indicating exciting possibilities as well as delineating problems. There is considerable clinical evidence that patients have some intrinsic ability to control tumor growth and that certain tumors remain dormant for long periods. Attempts to understand and perhaps stimulate the mechanisms involved are being made through the use of biological modifiers and by manipulating potential effector cells in vitro. Ideally this approach, which may include non-specific and specific elements, could be combined with specific vaccination in order to combat the apparent ability of many tumor cells to evade host defences.
Collapse
Affiliation(s)
- F K Stevenson
- Molecular Immunology Group, Southampton University Hospitals, UK
| |
Collapse
|
43
|
Affiliation(s)
- K E Hellström
- Bristol-Myers Squibb Pharmaceutical Research Institute, Seattle, Washington 98121
| | | |
Collapse
|
44
|
Hu SL, Hellström I, Hellström KE. Recent advances in antitumor vaccines. BIOTECHNOLOGY (READING, MASS.) 1992; 20:327-43. [PMID: 1318138 DOI: 10.1016/b978-0-7506-9265-6.50020-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Immunization with anti-idiotypic antibodies can induce cell-mediated and humoral antitumor immunity in animal models. This immunity can sometimes cause tumor destruction. However, more needs to be learned about how best to induce the type of immune response that is responsible for tumor destruction, since the presence of anti-idiotypic antibodies has been shown occasionally to enhance, rather than to inhibit, tumor growth. There is evidence suggesting that immunization of human cancer patients with Ab2 can have therapeutic benefit, and also that patients who mount a vigorous Ab2 response following treatment with an Ab1 may do clinically better than those who do not make any Ab2. Although the generation of Ab2 related to infused antitumor Ab1 does not cause tumor rejection in the majority of patients, and although the clinical data from patients given Ab2 are scarce, the suggestion that Ab2 may cause destruction of human cancers indicates that further work in this area may become rewarding.
Collapse
|
45
|
Affiliation(s)
- E Kedar
- Lautenberg Center for General and Tumor Immunology, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | |
Collapse
|
46
|
|
47
|
Kaufman H, Schlom J, Kantor J. A recombinant vaccinia virus expressing human carcinoembryonic antigen (CEA). Int J Cancer 1991; 48:900-7. [PMID: 1860736 DOI: 10.1002/ijc.2910480618] [Citation(s) in RCA: 59] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Carcinoembryonic antigen (CEA) is a 180-kDa glycoprotein expressed on most gastrointestinal carcinomas. A 2.4-kb cDNA clone, containing the complete coding sequence, was isolated from a human colon tumor cell library and inserted into a vaccinia virus genome. This newly developed construct was characterized by Southern blotting, DNA hybridization studies, and polymerase chain reaction analysis. The CEA gene was stably integrated into the vaccinia virus thymidine kinase gene. The recombinant was efficiently replicated upon serial passages in cell cultures and in animals. The recombinant virus expresses on the surface of infected cells a protein product recognized by a monoclonal antibody (COL-I) directed against CEA. Immunization of mice with the vaccinia construct elicited a humoral immune response against CEA. Pilot studies also showed that administration of the recombinant CEA vaccinia construct was able to greatly reduce the growth in mice of a syngeneic murine colon adenocarcinoma which had been transduced with the human CEA gene. The use of this new recombinant CEA vaccinia construct may thus provide an approach in the specific active immunotherapy of human GI cancer and other CEA expressing carcinoma types.
Collapse
Affiliation(s)
- H Kaufman
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | | | | |
Collapse
|
48
|
Abstract
Vaccinia virus is no longer needed for smallpox immunization, but now serves as a useful vector for expressing genes within the cytoplasm of eukaryotic cells. As a research tool, recombinant vaccinia viruses are used to synthesize biologically active proteins and analyze structure-function relations, determine the targets of humoral- and cell-mediated immunity, and investigate the immune responses needed for protection against specific infectious diseases. When more data on safety and efficacy are available, recombinant vaccinia and related poxviruses may be candidates for live vaccines and for cancer immunotherapy.
Collapse
Affiliation(s)
- B Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
49
|
Livingston P. Active Specific Immunotherapy in the Treatment of Patients with Cancer. Immunol Allergy Clin North Am 1991. [DOI: 10.1016/s0889-8561(22)00329-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
50
|
Mackett M. The live vector approach—viruses. World J Microbiol Biotechnol 1991; 7:137-49. [DOI: 10.1007/bf00328983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|