1
|
Medler TR, Blair TC, Crittenden MR, Gough MJ. Defining Immunogenic and Radioimmunogenic Tumors. Front Oncol 2021; 11:667075. [PMID: 33816320 PMCID: PMC8017281 DOI: 10.3389/fonc.2021.667075] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/02/2021] [Indexed: 12/21/2022] Open
Abstract
In the cancer literature tumors are inconsistently labeled as ‘immunogenic’, and experimental results are occasionally dismissed since they are only tested in known ‘responsive’ tumor models. The definition of immunogenicity has moved from its classical definition based on the rejection of secondary tumors to a more nebulous definition based on immune infiltrates and response to immunotherapy interventions. This review discusses the basis behind tumor immunogenicity and the variation between tumor models, then moves to discuss how these principles apply to the response to radiation therapy. In this way we can identify radioimmunogenic tumor models that are particularly responsive to immunotherapy only when combined with radiation, and identify the interventions that can convert unresponsive tumors so that they can also respond to these treatments.
Collapse
Affiliation(s)
- Terry R Medler
- Earle A. Chiles Research Institute, Providence Cancer Institute, Providence Portland Medical Center, Portland, OR, United States
| | - Tiffany C Blair
- Earle A. Chiles Research Institute, Providence Cancer Institute, Providence Portland Medical Center, Portland, OR, United States.,Molecular Microbiology and Immunology, OHSU, Portland, OR, United States
| | - Marka R Crittenden
- Earle A. Chiles Research Institute, Providence Cancer Institute, Providence Portland Medical Center, Portland, OR, United States.,Molecular Microbiology and Immunology, OHSU, Portland, OR, United States.,The Oregon Clinic, Portland, OR, United States
| | - Michael J Gough
- Earle A. Chiles Research Institute, Providence Cancer Institute, Providence Portland Medical Center, Portland, OR, United States.,Molecular Microbiology and Immunology, OHSU, Portland, OR, United States
| |
Collapse
|
2
|
|
3
|
CD8+ tumor-infiltrating T cells are trapped in the tumor-dendritic cell network. Neoplasia 2013; 15:85-94. [PMID: 23359264 DOI: 10.1593/neo.121572] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 11/26/2012] [Accepted: 11/29/2012] [Indexed: 12/22/2022] Open
Abstract
Chemotherapy enhances the antitumor adaptive immune T cell response, but the immunosuppressive tumor environment often dominates, resulting in cancer relapse. Antigen-presenting cells such as tumor-associated macrophages (TAMs) and tumor dendritic cells (TuDCs) are the main protagonists of tumor-infiltrating lymphocyte (TIL) immunosuppression. TAMs have been widely investigated and are associated with poor prognosis, but the immunosuppressive activity of TuDCs is less well understood. We performed two-photon imaging of the tumor tissue to examine the spatiotemporal interactions between TILs and TuDCs after chemotherapy. In a strongly immunosuppressive murine tumor model, cyclophosphamide-mediated chemotherapy transiently enhanced the antitumor activity of adoptively transferred ovalbumin-specific CD8(+) T cell receptor transgenic T cells (OTI) but barely affected TuDC compartment within the tumor. Time lapse imaging of living tumor tissue showed that TuDCs are organized as a mesh with dynamic interconnections. Once infiltrated into the tumor parenchyma, OTI T cells make antigen-specific and long-lasting contacts with TuDCs. Extensive analysis of TIL infiltration on histologic section revealed that after chemotherapy the majority of OTI T cells interact with TuDCs and that infiltration is restricted to TuDC-rich areas. We propose that the TuDC network exerts antigen-dependent unproductive retention that trap T cells and limit their antitumor effectiveness.
Collapse
|
4
|
Hebishima T, Tada S, Takeshima SN, Akaike T, Ito Y, Aida Y. Induction of antigen-specific immunity by pH-sensitive carbonate apatite as a potent vaccine carrier. Biochem Biophys Res Commun 2011; 415:597-601. [DOI: 10.1016/j.bbrc.2011.10.114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 10/25/2011] [Indexed: 11/29/2022]
|
5
|
Hebishima T, Matsumoto Y, Watanabe G, Soma GI, Kohchi C, Taya K, Hayashi Y, Hirota Y. Oral administration of immunopotentiator from Pantoea agglomerans 1 (IP-PA1) improves the survival of B16 melanoma-inoculated model mice. Exp Anim 2011; 60:101-9. [PMID: 21512265 DOI: 10.1538/expanim.60.101] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
To investigate the usefulness of the immunopotentiator from Pantoea agglomerans 1 (IP-PA1) as a supportive drug in melanoma therapy, we analyzed the immunological effects of IP-PA1 on melanoma-inoculated model mice. Oral administration of IP-PA1 increased the serum levels of tumor necrosis factor (TNF)-α at 2 h after the administration and interferon (IFN)-γ and IL-12 at 12 h after the administration in naïve BALB/cCrSlc mice as evaluated by ELISA. IP-PA1 did not affect the proliferation of melanoma cells directly determined by the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay. Combinatory treatment of IP-PA1 with doxorubicin for 9 days increased the serum levels of IFN-γ and IL-12 by 71.0 and 15.3%, respectively, compared to the treatment of doxorubicin alone in melanoma-bearing C57BL/6NCrSlc mice as evaluated by ELISA. It also increased the proportion of natural killer (NK) cells and the ratio of CD4(+) to CD8(+) T cells in the spleen from 6.1 ± 0.3 to 7.4 ± 0.5% and from 1.25 ± 0.03 to 1.38 ± 0.04, respectively, compared to the treatment of doxorubicin alone as analyzed by flow cytometry. The mean survival period of melanoma-bearing, doxorubicin treated mice was prolonged from 31.4 ± 7.1 to 35.3 ± 8.4, 51.1 ± 5.4, and 45.0 ± 8.4 days by combinatory treatment of IP-PA1 at the daily doses of 0.1, 0.5, and 1 mg/kg, respectively. In conclusion, the results of the present study suggest the usefulness of IP-PA1 as a supportive drug in melanoma therapy.
Collapse
Affiliation(s)
- Takehisa Hebishima
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Tokyo 183-8509, Japan
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Zeelenberg IS, van Maren WWC, Boissonnas A, Van Hout-Kuijer MA, Den Brok MHMGM, Wagenaars JAL, van der Schaaf A, Jansen EJR, Amigorena S, Théry C, Figdor CG, Adema GJ. Antigen localization controls T cell-mediated tumor immunity. THE JOURNAL OF IMMUNOLOGY 2011; 187:1281-8. [PMID: 21705625 DOI: 10.4049/jimmunol.1003905] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Effective antitumor immunotherapy requires the identification of suitable target Ags. Interestingly, many of the tumor Ags used in clinical trials are present in preparations of secreted tumor vesicles (exosomes). In this study, we compared T cell responses elicited by murine MCA101 fibrosarcoma tumors expressing a model Ag at different localizations within the tumor cell in association with secreted vesicles (exosomes), as a nonsecreted cell-associated protein, or as secreted soluble protein. Remarkably, we demonstrated that only the tumor-secreting vesicle-bound Ag elicited a strong Ag-specific CD8(+) T cell response, CD4(+) T cell help, Ag-specific Abs, and a decrease in the percentage of immunosuppressive regulatory T cells in the tumor. Moreover, in a therapeutic tumor model of cryoablation, only in tumors secreting vesicle-bound Ag could Ag-specific CD8(+) T cells still be detected up to 16 d after therapy. We concluded that the localization of an Ag within the tumor codetermines whether a robust immunostimulatory response is elicited. In vivo, vesicle-bound Ag clearly skews toward a more immunogenic phenotype, whereas soluble or cell-associated Ag expression cannot prevent or even delay outgrowth and results in tumor tolerance. This may explain why particular immunotherapies based on these vesicle-bound tumor Ags are potentially successful. Therefore, we conclude that this study may have significant implications in the discovery of new tumor Ags suitable for immunotherapy and that their location should be taken into account to ensure a strong antitumor immune response.
Collapse
Affiliation(s)
- Ingrid S Zeelenberg
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, 6525GA Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Zeelenberg IS, Ostrowski M, Krumeich S, Bobrie A, Jancic C, Boissonnas A, Delcayre A, Le Pecq JB, Combadière B, Amigorena S, Théry C. Targeting tumor antigens to secreted membrane vesicles in vivo induces efficient antitumor immune responses. Cancer Res 2008; 68:1228-35. [PMID: 18281500 DOI: 10.1158/0008-5472.can-07-3163] [Citation(s) in RCA: 235] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Expression of non-self antigens by tumors can induce activation of T cells in vivo, although this activation can lead to either immunity or tolerance. CD8+ T-cell activation can be direct (if the tumor expresses MHC class I molecules) or indirect (after the capture and cross-presentation of tumor antigens by dendritic cells). The modes of tumor antigen capture by dendritic cells in vivo remain unclear. Here we examine the immunogenicity of the same model antigen secreted by live tumors either in association with membrane vesicles (exosomes) or as a soluble protein. We have artificially addressed the antigen to secreted vesicles by coupling it to the factor VIII-like C1C2 domain of milk fat globule epidermal growth factor-factor VIII (MFG-E8)/lactadherin. We show that murine fibrosarcoma tumor cells that secrete vesicle-bound antigen grow slower than tumors that secrete soluble antigen in immunocompetent, but not in immunodeficient, host mice. This growth difference is due to the induction of a more potent antigen-specific antitumor immune response in vivo by the vesicle-bound than by the soluble antigen. Finally, in vivo secretion of the vesicle-bound antigen either by tumors or by vaccination with naked DNA protects against soluble antigen-secreting tumors. We conclude that the mode of secretion can determine the immunogenicity of tumor antigens and that manipulation of the mode of antigen secretion may be used to optimize antitumor vaccination protocols.
Collapse
Affiliation(s)
- Ingrid S Zeelenberg
- Institut National de la Santé et de la Recherche Médicale U653, Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Teramoto K, Kontani K, Fujita T, Ozaki Y, Sawai S, Tezuka N, Fujino S, Itoh Y, Taguchi O, Kannagi R, Ogasawara K. Successful tumor eradication was achieved by collaboration of augmented cytotoxic activity and anti-angiogenic effects following therapeutic vaccines containing helper-activating analog-loaded dendritic cells and tumor antigen DNA. Cancer Immunol Immunother 2007; 56:331-42. [PMID: 16896967 PMCID: PMC11031089 DOI: 10.1007/s00262-006-0192-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2006] [Accepted: 06/07/2006] [Indexed: 11/30/2022]
Abstract
We reported previously that pigeon cytochrome c-derived peptides (Pan-IA), which bind broad ranges of MHC class II molecules efficiently, activate T helper (Th) function in mice. In an experimental model, Pan-IA DNA vaccines augmented antitumor immunity in tumor antigen-immunized mice. To elicit more potent antitumor immunity and to eradicate tumors in a therapeutic setting, Pan-IA-loaded dendritic cells (DCs) were inoculated in combination with vaccines including ovalbumin (OVA) antigen DNA in tumor-bearing mice. Seventy percent of the immunized mice survived tumor-free for at least 4 months after treatment. In contrast, mice vaccinated with OVA DNA, either with or without naïve DCs, did not eliminate the tumors and died within 5 weeks. Only in mice vaccinated with OVA DNA and Pan-IA-loaded DCs were both cytotoxic and helper responses specific for OVA induced at the spleen and tumor sites as well as at the vaccination sites. Furthermore, accumulation of OVA-specific CD4(+) and CD8(+) T lymphocytes and interferon-gamma-mediated anti-angiogenesis were observed in the tumors of these mice. Thus, the combined vaccination primed both tumor-specific cytotoxicity and helper immunity resulting in augmented tumor lysis ability and anti-angiogenic effects. This is the first report to show that most established tumors were successfully eradicated by collaboration of potent antitumor immunity and anti-angiogenic effects by vaccination with tumor antigens and helper-activating analogs. This novel vaccination strategy is broadly applicable, regardless of identifying helper epitopes in target molecules, and contributes to the development of therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Koji Teramoto
- Department of Surgery, Shiga University of Medical Science, Seta-tsukinowa, Otsu, 520-2192 Japan
| | - Keiichi Kontani
- Second Department of Surgery, Kagawa University Faculty of Medicine, 1750-1 Miki-cho, Kita-gun, 761-0793 Japan
| | - Takuya Fujita
- Department of Surgery, Shiga University of Medical Science, Seta-tsukinowa, Otsu, 520-2192 Japan
| | - Yoshitomo Ozaki
- Department of Surgery, Shiga University of Medical Science, Seta-tsukinowa, Otsu, 520-2192 Japan
| | - Satoru Sawai
- Department of Surgery, Shiga University of Medical Science, Seta-tsukinowa, Otsu, 520-2192 Japan
| | - Noriaki Tezuka
- Department of Surgery, Shiga University of Medical Science, Seta-tsukinowa, Otsu, 520-2192 Japan
| | - Shozo Fujino
- Department of Surgery, Shiga University of Medical Science, Seta-tsukinowa, Otsu, 520-2192 Japan
| | - Yasushi Itoh
- Department of Pathology, Shiga University of Medical Science, Seta-tsukinowa, Otsu, 520-2192 Japan
| | - Osamu Taguchi
- Division of Molecular Pathology, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa, Nagoya, 464-8681 Japan
| | - Reiji Kannagi
- Division of Molecular Pathology, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa, Nagoya, 464-8681 Japan
| | - Kazumasa Ogasawara
- Department of Pathology, Shiga University of Medical Science, Seta-tsukinowa, Otsu, 520-2192 Japan
| |
Collapse
|
9
|
Engelhorn ME, Guevara-Patiño JA, Noffz G, Hooper AT, Lou O, Gold JS, Kappel BJ, Houghton AN. Autoimmunity and tumor immunity induced by immune responses to mutations in self. Nat Med 2006; 12:198-206. [PMID: 16444264 DOI: 10.1038/nm1363] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2005] [Accepted: 01/03/2006] [Indexed: 11/08/2022]
Abstract
Little is known about the consequences of immune recognition of mutated gene products, despite their potential relevance to autoimmunity and tumor immunity. To identify mutations that induce immunity, here we have developed a systematic approach in which combinatorial DNA libraries encoding large numbers of random mutations in two syngeneic tyrosinase-related proteins are used to immunize black mice. We show that the libraries of mutated DNA induce autoimmune hypopigmentation and tumor immunity through cross-recognition of nonmutated gene products. Truncations are present in all immunogenic clones and are sufficient to elicit immunity to self, triggering recognition of normally silent epitopes. Immunity is further enhanced by specific amino acid substitutions that promote T helper cell responses. Thus, presentation of a vast repertoire of antigen variants to the immune system can enhance the generation of adaptive immune responses to self.
Collapse
Affiliation(s)
- Manuel E Engelhorn
- Swim Across America Laboratory, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10021, USA.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
|
11
|
Abstract
The idea of enhancing or establishing effective immune response against endogenously developed tumor cells is not novel. More than a hundred years ago, bacterial components were used to develop antitumor immune response. Later, when a number of immune system-effecting cytokines had been discovered, they were used for systemic treatment of cancer patients. However, systemic treatment often resulted in even negative outcome. Recent developments of genetic approaches of cell modifications allowed developing of modern techniques of targeted tumor cell elimination. In the present paper, we review modern trends of the antitumor response enhancement based on immunoregulatory gene transfer into different cell types both in vivo and in vitro. Almost all these approaches are based on the activation of the adaptive arm of the immune system in response to tumor cells. However, recent studies indicate that the innate arm of the immune system, as well as adaptive arm, is involved in tumor suppression. The innate immune system uses nonrearranging germline receptors, which could trigger cellular effector responses that are conditional (or instructive) to the subsequent adaptive immune response. Last years' viewpoints on 'self' and 'non-self' recognition and primary induction of the immune response have changed. The key role of lymphocytes is pathogen recognition and, following immune response induction, switched on the central role of dendritic cells in 'non-self' recognition and induction of both innate and adaptive responses. Moreover, innate response is supposed to be an essential starting point in induction of successful and effective acquired response. Most cancer vaccines do not have 'non-self' marks presentation due to their endogenous origin, thus lacking their effectiveness in the induction of the specific long-lasting immune response. Taking this point into consideration, we can conclude that to make cancer vaccine more effective we have to present tumor antigens, together with the molecules that can potentially activate downstream 'non-self' recognition events not in parallel, but as a consequence of tumor antigen processing and presentation.
Collapse
Affiliation(s)
- S S Larin
- Institute of Gene Biology Russian Academy of Sciences, Moscow, Russia
| | | | | |
Collapse
|
12
|
Kurte M, López M, Aguirre A, Escobar A, Aguillón JC, Charo J, Larsen CG, Kiessling R, Salazar-Onfray F. A Synthetic Peptide Homologous to Functional Domain of Human IL-10 Down-Regulates Expression of MHC Class I and Transporter Associated with Antigen Processing 1/2 in Human Melanoma Cells. THE JOURNAL OF IMMUNOLOGY 2004; 173:1731-7. [PMID: 15265902 DOI: 10.4049/jimmunol.173.3.1731] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Tumor cells treated with IL-10 were shown to have decreased, but peptide-inducible expression of MHC class I, decreased sensitivity to MHC class I-restricted CTL, and increased NK sensitivity. These findings could be explained, at least partially, by a down-regulation of TAP1/TAP2 expression. In this study, IT9302, a nanomeric peptide (AYMTMKIRN), homologous to the C-terminal of the human IL-10 sequence, was demonstrated to mimic these previously described IL-10 effects on MHC class I-related molecules and functions. We observed a dose-dependent down-regulation of MHC class I at the cell surface of melanoma cells after 24-h treatment with IT9302. The IL-10 homologue peptide also caused a dose-dependent inhibition of the IFN-gamma-mediated surface induction of MHC class I in a melanoma cell line. We demonstrated, using Western blot and flow cytometry, that IT9302 inhibits the expression of TAP1 and TAP2 proteins, but not MHC class I H chain or low molecular protein molecules. Finally, peptide-treated melanoma cells were shown to be more sensitive to lysis by NK cells in a dose-dependent way. Taken together, these results demonstrate that a small synthetic peptide derived from IL-10 can mimic the Ag presentation-related effects mediated by this cytokine in human melanomas and increase tumor sensitivity to NK cells, which can be relevant in the designing of future strategies for cancer immune therapy.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 2
- ATP Binding Cassette Transporter, Subfamily B, Member 3
- ATP-Binding Cassette Transporters/biosynthesis
- ATP-Binding Cassette Transporters/genetics
- Cell Line, Tumor/drug effects
- Cell Line, Tumor/metabolism
- Cysteine Endopeptidases/biosynthesis
- Cysteine Endopeptidases/genetics
- Cytotoxicity, Immunologic
- Dose-Response Relationship, Drug
- Eye Neoplasms/metabolism
- Eye Neoplasms/pathology
- Gene Expression Regulation, Neoplastic/drug effects
- Genes, MHC Class I
- Histocompatibility Antigens Class I/biosynthesis
- Humans
- Interferon-gamma/antagonists & inhibitors
- Interferon-gamma/pharmacology
- Interleukin-10/agonists
- Interleukin-10/chemistry
- Killer Cells, Lymphokine-Activated/immunology
- Melanoma/metabolism
- Melanoma/pathology
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Oligopeptides/pharmacology
- Protein Structure, Tertiary
- Recombinant Proteins
Collapse
Affiliation(s)
- Mónica Kurte
- Disciplinary Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
Adoptive cellular therapy remains a potentially powerful method of eradicating established tumors. T-cells have been particularly potent effector cells, as demonstrated in animal models and clinical studies, and it is apparent that the stimulation of certain subpopulations of T-cells that are reactive to tumor antigens can lead to more therapeutic T-cells. The use of gene transfer techniques has resulted in more effective and specific methods to generate these tumor-specific T-cells. Another area of tremendous interest is in the adoptive transfer of DCs manipulated to present tumor antigen to resting, naive T-cells. Gene transfer techniques may offer more optimal ways to generate therapeutic DCs. Adoptive immunotherapy may ultimately [figure: see text] have its greatest use in patients undergoing cellular rescue after ablative chemotherapy; the infusion of immunocompetent T-cells, genetically modified stem cells, or programmed DCs may offer the opportunity to direct a patient's immune response to eliminate residual microscopic disease.
Collapse
Affiliation(s)
- Alicia Terando
- Division of Surgical Oncology, University of Michigan Medical Center, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
14
|
Abstract
Over the past decade, the unprecedented growth in science and technology has fueled the development of novel treatment strategies to combat disease. The creative and innovative efforts of scientists and clinicians to overcome the multitude of unforeseen obstacles to success is no better exemplified than in the field of cancer gene therapy. Since its inception, developers of cancer gene therapy have been charged with the challenge of altering basic tumor biology or, alternatively, the host responses for the purpose of tumor eradication and prevention. Several major therapeutic strategies have emerged from preclinical studies, and results from these early studies hold promise for altering the clinical outcome in a variety of malignancies. These strategies may be broadly subcategorized and range in intent from alteration of the tumor cell phenotype by replacement of defective cellular response genes (e.g., mutated or deleted tumor suppressor genes) to the enhancement of the immunological response to cancer (e.g., amplification of the cell surface antigen signature or modulation of the host response). Not surprisingly, the increasingly intricate nature of tumor biology revealed over the past several years has effectively raised the bar of success for those involved in the development of effective molecular and cancer gene therapy strategies. This, in turn, has led to the development of more complex therapies that frequently draw upon multiple disciplines in an effort to optimize treatment response.
Collapse
Affiliation(s)
- James C Cusack
- Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Cox Building, Room 626, 100 Blossom Street, Boston, MA 02114, USA.
| | | |
Collapse
|
15
|
Tanaka H, Tanaka J, Kjaergaard J, Shu S. Depletion of CD4+ CD25+ regulatory cells augments the generation of specific immune T cells in tumor-draining lymph nodes. J Immunother 2002; 25:207-17. [PMID: 12000862 DOI: 10.1097/00002371-200205000-00003] [Citation(s) in RCA: 155] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Recent studies have identified a unique population of CD4+CD25+ regulatory T cells that is crucial for the prevention of spontaneous autoimmune diseases. Further studies demonstrated that depletion of CD4+CD25+ T cells enhances immune responses to nonself antigens. Because immune responses to malignant tumors are weak and ineffective, depletion of regulatory T cells has been reported to result in tumor regression. In the current study, using the weakly immunogenic MCA205 sarcoma and the poorly immunogenic B16/BL6/D5 (D5) melanoma, depletion of CD4+CD25+ T cells by the administration of anti-CD25 monoclonal antibodies (mAb), PC61 induced some tumor growth retardation, but all mice eventually succumbed to tumors. In our laboratory, immunotherapy by the transfer of tumor-immune T cells has demonstrated potent antitumor effects. A reliable source of tumor-reactive T cells has been lymph nodes (LN) draining progressive tumors. Therapeutic effector T cells can be generated by in vitro activation of draining LN cells with anti-CD3 mAb followed by culture in interleukin-2. In this system, PC61 mAb depletion of CD4+CD25+ T cells before or on day 8 of tumor growth resulted in increased sensitization in the draining LN. The therapeutic efficacy of activated tumor-draining LN cells from mAb depleted mice increased approximately three fold while maintaining specificity when tested in adoptive immunotherapy of established pulmonary metastases. Specific interferon-gamma secretion by LN T cells from mice treated with PC61 mAb 1 day before tumor inoculation increased significantly. However, this increase was not demonstrated with LN T cells from mice treated on day 8 despite their enhanced therapeutic reactivities. Our results indicate that although the antitumor immunity enhanced by the depletion of CD4+CD25+ T cells is insufficient to eradicate tumors, it augments the sensitization of immune T cells in the draining LN, thus, facilitating adoptive immunotherapy.
Collapse
Affiliation(s)
- Hiroshi Tanaka
- Center for Surgery Research/FF50, The Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, U.S.A
| | | | | | | |
Collapse
|
16
|
McKeever U, Barman S, Hao T, Chambers P, Song S, Lunsford L, Hsu YY, Roy K, Hedley ML. Protective immune responses elicited in mice by immunization with formulations of poly(lactide-co-glycolide) microparticles. Vaccine 2002; 20:1524-31. [PMID: 11858858 DOI: 10.1016/s0264-410x(01)00509-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Parenteral administration of microparticle encapsulated DNA elicits immune responses to the encoded antigens. Experiments were performed to test whether the addition of certain lipophilic agents to such formulations enhanced the activity of a beta-galactosidase (beta-gal) DNA vaccine. Addition of either taurocholic acid (TA) or monomethoxy polyethylene-glycol-distearoylphosphatidylehanolamine (PEG-DSPE) increased the efficiency of DNA encapsulation. Immunization of mice with encapsulated DNA formulations containing either compound significantly increased the number of antibody positive responders over that achieved with non-lipid containing particles. Moreover, responding animals demonstrated trends towards higher antibody titers and increased T cell responses. Tumor protection against the CT26.CL25 tumor cell line was demonstrated with lipid and non-lipid containing formulations. These results are the first demonstration of protection obtained by parenteral administration of PLG encapsulated DNA vaccines.
Collapse
Affiliation(s)
- U McKeever
- Zycos Inc., 44 Hartwell Avenue, Lexington, MA 02421, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Yu D, Thomas-Tikhonenko A. Intratumoral delivery of an interferon gamma retrovirus-producing cells inhibits growth of a murine melanoma by a non-immune mechanism. Cancer Lett 2001; 173:145-54. [PMID: 11597789 DOI: 10.1016/s0304-3835(01)00612-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Interferon gamma (IFNgamma) is a potent inhibitor of cell growth effective against a wide range of tumor-derived cell lines. We cloned murine IFNgamma cDNA into a retroviral vector and created a packaging cell line (Am-gamma) producing this IFNgamma-encoding retrovirus. In a pilot experiment, admixing and co-injection of equal numbers of retrovirus-producing and target B16 melanoma cells led to high rates of infection and strong suppression of neoplastic growth. This effect was observed in the absence of measurable systemic production of IFNgamma and could be reproduced in animals lacking cytotoxic immune responses. Tumor angiogenesis was unaffected and no increase in apoptosis was apparent; however, mitotic indices were greatly reduced in Am-gamma-containing abortive tumors. We thus concluded that IFNgamma directly affects proliferation of B16 cells. Indeed, exposure of B16 cells to IFNgamma in vitro inhibits cell division, as measured by a thymidine incorporation assay. Most importantly, repeated intratumoral injections of Am-gamma stunted growth of established B16 melanomas in 75% of treated animals. Thus, this approach can serve as a prototype for new anti-cancer modalities.
Collapse
Affiliation(s)
- D Yu
- Department of Pathobiology, University of Pennsylvania, 3800 Spruce Street, 368E, Philadelphia, PA 19104-6051, USA
| | | |
Collapse
|
18
|
Tanabe KK, Cusack JC. Gene Therapy. Surgery 2001. [DOI: 10.1007/978-3-642-57282-1_86] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
19
|
Peng L, Krauss JC, Plautz GE, Mukai S, Shu S, Cohen PA. T cell-mediated tumor rejection displays diverse dependence upon perforin and IFN-gamma mechanisms that cannot be predicted from in vitro T cell characteristics. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:7116-24. [PMID: 11120842 DOI: 10.4049/jimmunol.165.12.7116] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Experimental pulmonary metastases have been successfully treated by adoptive transfer of tumor-sensitized T cells from perforin knockout (KO) or Fas/APO-1 ligand(KO) mice, suggesting a prominent role for secretion of cytokines such as IFN-gamma. In the present study we confirmed that rejection of established methylcholanthrene-205 (MCA-205) pulmonary metastases displayed a requirement for T cell IFN-gamma expression. However, this requirement could be obviated by transferring larger numbers of tumor-sensitized IFN-gamma (KO) T cells or by immunosensitizing sublethal irradiation (500 rad) of the host before adoptive therapy. Extrapulmonary tumors (MCA-205 s.c. and intracranial) that required adjunct sublethal irradiation for treatment efficacy also displayed no requirement for host or T cell expression of IFN-gamma. Nonetheless, rejection of MCA-205 s.c. tumors and i.p. EL-4 tumors, but not MCA-205 pulmonary or intracranial tumors, displayed a significant requirement for T cell perforin expression (i.e., CTL participation). The capacity of T cells to lyse tumor targets and secrete IFN-gamma in vitro before adoptive transfer was nonpredictive of the roles of these activities in subsequent tumor rejection. Adoptive therapy studies employing KO mice are therefore indispensable for revealing a diversity of tumor rejection mechanisms that may lack in vitro correlation due to delays in their induction. Seemingly contradictory KO data from different studies are reconciled by the capacity of anti-tumor T cells to rely on alternative mechanisms when treated in larger numbers, the variable participation of CTL at different anatomic locations of tumor, and the apparent capacity of sublethal irradiation to provide a therapeutic alternative to host or T cell IFN-gamma production.
Collapse
MESH Headings
- Animals
- Brain Neoplasms/immunology
- Brain Neoplasms/therapy
- Cells, Cultured
- Cytotoxicity Tests, Immunologic
- Dose-Response Relationship, Immunologic
- Female
- Graft Rejection/genetics
- Graft Rejection/immunology
- Graft Rejection/radiotherapy
- Immunotherapy, Adoptive
- Injections, Intraperitoneal
- Interferon-gamma/metabolism
- Interferon-gamma/physiology
- Interferon-gamma/radiation effects
- Lung Neoplasms/immunology
- Lung Neoplasms/radiotherapy
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Lymph Nodes/immunology
- Lymph Nodes/metabolism
- Lymph Nodes/pathology
- Lymphocyte Activation
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/transplantation
- Lymphoma/genetics
- Lymphoma/immunology
- Lymphoma/radiotherapy
- Lymphoma/therapy
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasm Transplantation
- Perforin
- Pore Forming Cytotoxic Proteins
- Sarcoma, Experimental/genetics
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/radiotherapy
- Sarcoma, Experimental/therapy
- Tumor Cells, Cultured/transplantation
- Whole-Body Irradiation
Collapse
Affiliation(s)
- L Peng
- Center for Surgery Research, Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Hu HM, Winter H, Urba WJ, Fox BA. Divergent roles for CD4+ T cells in the priming and effector/memory phases of adoptive immunotherapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:4246-53. [PMID: 11035058 DOI: 10.4049/jimmunol.165.8.4246] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The requirement for CD4(+) Th cells in the cross-priming of antitumor CTL is well accepted in tumor immunology. Here we report that the requirement for T cell help can be replaced by local production of GM-CSF at the vaccine site. Experiments using mice in which CD4(+) T cells were eliminated, either by Ab depletion or by gene knockout of the MHC class II beta-chain (MHC II KO), revealed that priming of therapeutic CD8(+) effector T cells following vaccination with a GM-CSF-transduced B16BL6-D5 tumor cell line occurred independently of CD4(+) T cell help. The adoptive transfer of CD8(+) effector T cells, but not CD4(+) effector T cells, led to complete regression of pulmonary metastases. Regression of pulmonary metastases did not require either host T cells or NK cells. Transfer of CD8(+) effector T cells alone could cure wild-type animals of systemic tumor; the majority of tumor-bearing mice survived long term after treatment (>100 days). In contrast, adoptive transfer of CD8(+) T cells to tumor-bearing MHC II KO mice improved survival, but eventually all MHC II KO mice succumbed to metastatic disease. WT mice cured by adoptive transfer of CD8(+) T cells were resistant to tumor challenge. Resistance was mediated by CD8(+) T cells in mice at 50 days, while both CD4(+) and CD8(+) T cells were important for protection in mice challenged 150 days following adoptive transfer. Thus, in this tumor model CD4(+) Th cells are not required for the priming phase of CD8(+) effector T cells; however, they are critical for both the complete elimination of tumor and the maintenance of a long term protective antitumor memory response in vivo.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, T-Independent/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/transplantation
- Cancer Vaccines/immunology
- Cell Count
- Cytotoxicity, Immunologic/genetics
- Dendritic Cells/immunology
- Dendritic Cells/pathology
- Female
- Histocompatibility Antigens Class II/genetics
- Immunologic Memory/genetics
- Immunophenotyping
- Killer Cells, Natural/immunology
- Lung Neoplasms/genetics
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Lymph Nodes/immunology
- Lymph Nodes/metabolism
- Lymph Nodes/pathology
- Lymphocyte Activation/genetics
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Skin Pigmentation/genetics
- Skin Pigmentation/immunology
- Survival Analysis
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/pathology
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Cells, Cultured/immunology
- Tumor Cells, Cultured/transplantation
Collapse
Affiliation(s)
- H M Hu
- Laboratory of Molecular and Tumor Immunology, Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR 97213, USA
| | | | | | | |
Collapse
|
21
|
Ferrantini M, Belardelli F. Gene therapy of cancer with interferon: lessons from tumor models and perspectives for clinical applications. Semin Cancer Biol 2000; 10:145-57. [PMID: 10936064 DOI: 10.1006/scbi.2000.0333] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cytokine gene transfer is a current approach in studies of gene therapy of cancer IFNs represent valuable cytokines for these studies, since they exert multiple biological effects, including anti-tumor activities. Early studies have been focused on IFN-gamma. Recently, several reports have shown that the transfer of type I IFN (especially IFN-alpha) genes represents a powerful approach for inducing tumor suppression. Recent studies have underscored new IFN-induced activities on immune cells. This knowledge adds a further rationale for the use of IFN-alpha in strategies of gene therapy of cancer and can be exploited for the design of more selective and effective anticancer treatments.
Collapse
Affiliation(s)
- M Ferrantini
- Laboratory of Virology, Istituto Superiore di Sanità, Rome, Italy
| | | |
Collapse
|
22
|
Affiliation(s)
- D M Pardoll
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
23
|
Murray JL, Hudson JM, Ross MI, Zhang HZ, Ioannides CG. Reduced recognition of metastatic melanoma cells by autologous MART-1 specific CTL: relationship to TAP expression. J Immunother 2000; 23:28-35. [PMID: 10687135 DOI: 10.1097/00002371-200001000-00005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Class I expression in context with T-cell receptor expression is crucial for peptide presentation and induction of CD8+ cytotoxic T lymphocytes (CTL). Presentation of class I bound peptides is dependent on transporter-associated proteins (TAP) expression and function. Tumor infiltrating lymphocytes from a patient with melanoma were isolated, expanded in vitro in the presence of interleukin-2, and tested for cytotoxicity against HLA-A2 positive, MART-1 positive autologous tumor cells, an HLA-A2-positive, MART-1 positive melanoma cell line (Mel-501), and HLA-A2-negative melanoma cells. Significant killing occurred against both A2-positive cell lines (63% and 65%, respectively), but not against the A2-negative line (18%) or A2-positive autologous tumor (1.5%). These CTL preferentially recognized the MART-1 peptide F119, 27-35, and gp100 peptide F125, 280-288, resulting in a 30% to 60% enhancement of lysis when autologous tumor or major histocompatibility complex class I "empty" T2 cells were pulsed with either peptide. To address whether the deficiency in autologous tumor recognition might be related to a deficiency in Ag presentation, we screened for the presence of TAP1 and TAP2 transcripts by polymerase chain reaction, Southern blotting, and scanning densitometry using sequence-specific primers and probes. Both TAP1 and TAP2 expression levels in the autologous tumor were minimal, yet were upregulated 7- to 18-fold, respectively, by interferon-gamma. Despite this increase, a similar increase in cytotoxicity did not occur. In short, deficiencies in TAP presentation may have functional significance for tumor escape from immunosurveillance and with respect to impending vaccine trials.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 2
- ATP Binding Cassette Transporter, Subfamily B, Member 3
- ATP-Binding Cassette Transporters/genetics
- Abdominal Neoplasms/immunology
- Abdominal Neoplasms/secondary
- Antigens, Neoplasm/immunology
- Cytotoxicity, Immunologic
- Female
- Gene Expression
- HLA-A2 Antigen/immunology
- Humans
- Interferon-gamma/immunology
- Interferon-gamma/pharmacology
- Lymphocytes, Tumor-Infiltrating/immunology
- MART-1 Antigen
- Major Histocompatibility Complex
- Melanoma/immunology
- Melanoma/pathology
- Melanoma/secondary
- Middle Aged
- Neoplasm Metastasis
- Neoplasm Proteins/immunology
- Peptides/immunology
- Phenotype
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- J L Murray
- Department of Bioimmunotherapy, MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
24
|
Kaufman HL, Rao JB, Irvine KR, Bronte V, Rosenberg SA, Restifo NP, Irivine KR. Interleukin-10 enhances the therapeutic effectiveness of a recombinant poxvirus-based vaccine in an experimental murine tumor model. J Immunother 1999; 22:489-96. [PMID: 10570747 PMCID: PMC2562555 DOI: 10.1097/00002371-199911000-00003] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Interleukin-10 (IL-10) has a wide range of in vivo biological activities and is a key regulatory cytokine of immune-mediated inflammation. The authors found that murine IL-10 given 12 hours after a recombinant vaccinia virus (rVV) containing the LacZ gene significantly enhanced the treatment of mice bearing 3-day-old pulmonary metastases expressing beta-galactosidase. Because IL-10 has been shown to inhibit the functions of key elements of both innate and acquired immune responses, the authors hypothesized that IL-10 might act by inhibiting clearance of the rVV, thus prolonging exposure to the experimental antigen. However, evidence that IL-10 was not acting primarily through such negative regulatory mechanisms included the following: (a) IL-10 also enhanced the therapeutic effectiveness of a recombinant fowlpox virus, which cannot replicate in mammalian cells; (b) Titers of rVV in immunized mice were lower, not higher; and (c) Although IL-10 did not alter levels of anti-vaccinia anti-bodies or natural killer cell activity, rVV-primed mice treated with IL-10 had enhanced vaccinia-specific cytotoxic T-lymphocyte activity. Thus, IL-10 enhanced the function of a recombinant poxvirus-based anti-cancer vaccine and may represent a potential adjuvant in the vaccination against human cancers using recombinant poxvirus-based vaccines.
Collapse
Affiliation(s)
- H L Kaufman
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Jung D, Hilmes C, Knuth A, Jaeger E, Huber C, Seliger B. Gene transfer of the Co-stimulatory molecules B7-1 and B7-2 enhances the immunogenicity of human renal cell carcinoma to a different extent. Scand J Immunol 1999; 50:242-9. [PMID: 10447932 DOI: 10.1046/j.1365-3083.1999.00588.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Stimulation of a specific antitumour immune response with recruitment and induction of T-cell effector functions represents an attractive concept in human cancer therapy. Different cytokines and the B7 co-stimulatory molecules are both able to provide proliferation and activation signals for T cells. In the present study, we first demonstrated the absence of both B7-1 and B7-2 expression in human renal cell carcinoma (RCC) cell lines. The lack of B7 expression was associated with a low or absent proliferative response of allogeneic and autologous T cells upon stimulation with tumour cells. In order to investigate the role of B7-1 and B7-2, the human RCC cell line, MZ1257RC, which expresses normal levels of adhesion molecules and major histocompatibility complex (MHC) class I surface antigens, was transfected with B7-1 and B7-2 expression vectors, respectively. The B7-1- and B7-2-transduced MZ1257RC cells were potent stimulators of allogeneic and autologous T-cell proliferation. B7-2 transfectants were approximately two- to threefold more effective in the induction of primary T-cell activation than B7-1-transduced cells. Interleukin (IL)-12 synergized with the B7/CD28 interaction to enhance allogeneic T-cell proliferation, independently of the B7 molecule transduced. In contrast, IL-2 only co-operatively increased T-cell activation in the presence of B7-2. Our results suggest the following: first, that co-stimulatory molecules are required for efficient T-cell responses directed against RCC; second, that B7-2 appears to be a more potent stimulator of tumour immunity as compared to B7-1; and third, that B7 molecules selectively co-operate with different T-cell stimulatory cytokines. The different activity of B7-1 and B7-2 molecules on the immunogenicity of RCC will have implications for the development and optimization of RCC-specific cancer vaccines.
Collapse
Affiliation(s)
- D Jung
- Johannes Gutenberg-Universität, III. Medizinische Klinik, Langenbeckstr. 1, 55101 Mainz, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Affiliation(s)
- D C LaTemple
- Department of Medicine, University of Pennsylvania, Philadelphia, USA
| | | |
Collapse
|
27
|
Salazar-Onfray F. Interleukin-10: a cytokine used by tumors to escape immunosurveillance. Cancer Immunol Immunother 1999; 16:86-94. [PMID: 10456656 DOI: 10.1007/bf02785841] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The mechanisms whereby malignant cells can elude the recognition of the immune system, by what is termed 'immunological escape', have attracted major attention of tumor immunologists during the past decade. In this review, the role of the immunosuppressive cytokine interleukin-10 (IL-10) will be discussed as a strategy used by tumors to avoid recognition by cytotoxic T lymphocytes (CTL).
Collapse
|
28
|
Nemunaitis J, Fong T, Burrows F, Bruce J, Peters G, Ognoskie N, Meyer W, Wynne D, Kerr R, Pippen J, Oldham F, Ando D. Phase I trial of interferon gamma retroviral vector administered intratumorally with multiple courses in patients with metastatic melanoma. Hum Gene Ther 1999; 10:1289-98. [PMID: 10365660 DOI: 10.1089/10430349950017978] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The purpose of this study was to determine the safety and antitumor activity of IFN-gamma retroviral vector in patients with advanced melanoma. Seventeen patients (9 single courses, 8 multiple courses) received a total of 363 intratumor injections of IFN-gamma retroviral vector (1 x 10(7) PFU/ml administered at 0.3, 0.5, and 1.0 ml per cohort). No grade III/IV adverse events were attributed to study medication. Replication-competent retrovirus was not detected in any of the 17 patients by polymerase chain reaction studies. Eight patients showed elevated anti-tumor antibody responses in comparison with baseline by ELISA. One of nine patients treated with a single course had an optimal response of stable disease, compared with eight of eight multiple-injected patients. Median survival of single-injected patients was 150 days, and patients who received multiple injections have still not achieved median survival duration, with four of eight still living (p = 0.0462, Wilcoxon; p = 0.0273, log rank). We conclude that intratumor injection of IFN-gamma is safe and well tolerated. Evidence of antitumor activity is suggested in patients with advanced malignancy that received multiple injections.
Collapse
|
29
|
Arosarena OA, Baranwal S, Strome S, Wolf GT, Krauss JC, Bradford CR, Carey TE. Expression of major histocompatibility complex antigens in squamous cell carcinomas of the head and neck: effects of interferon gene transfer. Otolaryngol Head Neck Surg 1999; 120:665-71. [PMID: 10229590 DOI: 10.1053/hn.1999.v120.a91770] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The effect of retroviral-mediated interferon-gamma (IFN-gamma) gene transfer on major histocompatibility complex (MHC) class I and II antigen expression was investigated in 13 head and neck squamous carcinoma cell lines. Six cell lines exhibited increased MHC class I expression, and 10 exhibited increased MHC class II expression after IFN-gamma gene transfer. Differences in MHC antigen expression between parental and transduced cell lines were significant (P = 0. 002) only for cell lines that upregulated MHC class II expression. After incubation in medium containing 100 U/mL recombinant IFN-gamma, or in medium from IFN-gamma retrovirus-transduced NIH 3T3 cells, 12 cell lines significantly upregulated MHC class I expression, and 9 significantly upregulated MHC class II expression. Only cell lines that exhibited increased MHC class II expression after retroviral transduction also upregulated class II expression with exogenous IFN-gamma treatment. Thus some head and neck squamous carcinoma cell lines can upregulate MHC class I and II expression after exogenous application of either IFN-gamma or IFN-gamma retroviral transduction. These are promising findings for head and neck cancer immunotherapy and gene therapy.
Collapse
Affiliation(s)
- O A Arosarena
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Kimura K, Nishimura H, Hirose K, Matsuguchi T, Nimura Y, Yoshikai Y. Immunogene therapy of murine fibrosarcoma using IL-15 gene with high translation efficiency. Eur J Immunol 1999; 29:1532-42. [PMID: 10359107 DOI: 10.1002/(sici)1521-4141(199905)29:05<1532::aid-immu1532>3.0.co;2-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We have recently found that translational efficiency is up-regulated by an alternative exon in IL-15 mRNA in mice. In a malignancy model using BALB/c mice and syngeneic Meth A fibrosarcoma (Meth A), we successfully applied immunological gene therapy with IL-15 protein using alternative IL-15 cDNA with high translational efficiency. Two expression vectors carrying the murine IL-15 gene were constructed for use in tumor immunotherapy, one utilizing IL-15 cDNA with alternative exon 5 and the second utilizing IL-15 cDNA with normal exon 5. The first vector induced the production of a large amount of IL-15 protein in Meth A cells, whereas tumor cells transfected by the second vector produced only a marginal level of IL-15 protein. Although cell growth of both transfectants in vitro remained unchanged, inoculation of clones transfected with normal IL-15 cDNA resulted in progressive tumor growth, while clones transfected with alternative IL-15 cDNA led to the rejection of the tumor. The clone producing high levels of IL-15 grew progressively in nude mice and mice treated with anti-CD4 monoclonal antibodies (mAb), whereas the growth of the transfectants was retarded in anti-CD8 mAb- or anti-asialo GM1 antibody-treated mice. Cured mice were shown to have generated immunity against a subsequent challenge with the wild type of Meth A but not against Meth 1 tumor cells, another type of fibrosarcoma derived from BALB/c mice. Thus, tumor therapy based on IL-15 gene transfection was effective against Meth A tumor cells, suggesting a possible application to human neoplasms.
Collapse
Affiliation(s)
- K Kimura
- Research Institute for Disease Mechanism and Control, First Department of Surgery, Nagoya University School of Medicine, Japan
| | | | | | | | | | | |
Collapse
|
31
|
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
|
32
|
Tanaka H, Yoshizawa H, Yamaguchi Y, Ito K, Kagamu H, Suzuki E, Gejyo F, Hamada H, Arakawa M. Successful Adoptive Immunotherapy of Murine Poorly Immunogenic Tumor with Specific Effector Cells Generated from Gene-Modified Tumor-Primed Lymph Node Cells. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.6.3574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
We previously reported that cytokine gene transfer into weakly immunogenic tumor cells could enhance the generation of precursor cells of tumor-reactive T cells and subsequently augment antitumor efficacy of adoptive immunotherapy. We investigated whether such potent antitumor effector T cells could be generated from mice bearing poorly immunogenic tumors. In contrast to similarly modified weakly immunogenic tumors, MCA102 cells, which are chemically induced poorly immunogenic fibrosarcoma cells transfected with cDNA for IL-2, IL-4, IL-6, IFN-γ, failed to augment the host immune reaction. Because priming of antitumor effector T cells in vivo requires two important signals provided by tumor-associated Ags and costimulatory molecules, these tumor cells were cotransfected with a B7-1 cDNA. Transfection of both IFN-γ and B7-1 (MCA102/B7-1/IFN-γ) resulted in regression of s.c. tumors, while tumor transfected with other combinations of cytokine and B7-1 showed progressive growth. Cotransfection of IFN-γ and B7-1 into other poorly immunogenic tumor B16 and LLC cells also resulted in the regression of s.c. tumors. Cells derived from lymph nodes draining MCA102/B7-1/IFN-γ tumors showed potent antitumor efficacy, eradicating established pulmonary metastases, but this effect was not seen with parental tumors. This mechanism of enhanced antitumor efficacy was further investigated, and T cells with down-regulated L-selectin expression, which constituted all the in vivo antitumor reactivity, were significantly increased in lymph nodes draining MCA102/B7-1/IFN-γ tumors. These T cells developed into potent antitumor effector cells after in vitro activation with anti-CD3/IL-2. The strategy presented here may provide a basis for developing potent immunotherapy for human cancers.
Collapse
Affiliation(s)
- Hiroshi Tanaka
- *Department of Medicine (II), Niigata University Medical School, Niigata, Japan; and
| | - Hirohisa Yoshizawa
- *Department of Medicine (II), Niigata University Medical School, Niigata, Japan; and
| | - Yoshifumi Yamaguchi
- *Department of Medicine (II), Niigata University Medical School, Niigata, Japan; and
| | - Kazuhisa Ito
- *Department of Medicine (II), Niigata University Medical School, Niigata, Japan; and
| | - Hiroshi Kagamu
- *Department of Medicine (II), Niigata University Medical School, Niigata, Japan; and
| | - Eiichi Suzuki
- *Department of Medicine (II), Niigata University Medical School, Niigata, Japan; and
| | - Fumitake Gejyo
- *Department of Medicine (II), Niigata University Medical School, Niigata, Japan; and
| | - Hirofumi Hamada
- †Department of Molecular Biotherapy Research, Cancer Chemotherapy Center, Cancer Institute, Tokyo, Japan
| | - Masaaki Arakawa
- *Department of Medicine (II), Niigata University Medical School, Niigata, Japan; and
| |
Collapse
|
33
|
Abstract
Transduction of a cytokine gene into neoplastic cells elicits a strong inflammatory host reaction that impairs tumor growth, and a long-lasting immune memory is established following their rejection. These findings have aroused great enthusiasm and expectations. Despite their enhanced immunogenicity, however, the immune reaction provoked by repeated injections of these engineered cells can do little more than inhibit the growth of initial tumors and metastases and is only minimally effective against established forms. Better therapeutic activity is thus being sought by combining such cells with tumor cells engineered with other genes.
Collapse
Affiliation(s)
- P Nanni
- Institute of Cancer Research, University of Bologna, Italy
| | | | | |
Collapse
|
34
|
Karavodin LM, Robbins J, Chong K, Hsu D, Ibanez C, Mento S, Jolly D, Fong TC. Generation of a systemic antitumor response with regional intratumoral injections of interferon gamma retroviral vector. Hum Gene Ther 1998; 9:2231-41. [PMID: 9794207 DOI: 10.1089/hum.1998.9.15-2231] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The generation of a lasting systemic immune response is a primary goal for cancer immunotherapy. Here we examine the ability of high-titer IFN-gamma retroviral vector injected into an accessible tumor to generate significant antitumor responses at a distal untreated site. CT26 or B16F10 murine tumors were inoculated subcutaneously to form solid tumors in BALB/c or C57BL/6 mice. Seven to 10 days postinoculation, high-titer IFN-gamma retroviral vector was directly injected into the subcutaneous tumor nodule, and optimal dose and course of therapy were determined. As a model for disseminated disease, mice were inoculated intravenously with CT26 cells to form pulmonary lesions, at the same time as the subcutaneous injections. Regression of subcutaneous tumor correlated with a systemic response at the distal lung metastases in the IFN-gamma-treated group (p < 0.0005). Splenocytes from mice with completely regressed tumors had a twofold increase in percent specific cytotoxicity in a standard CTL assay as compared with nonresponding mice. CD8+ T cells were shown to be essential for the regional and systemic antitumor response, as determined by in vivo cell depletion experiments. These data demonstrate that IFN-gamma retroviral vector gene therapy delivered intralesionally can generate significant inhibition of pulmonary tumor formation distal to the treatment site. The data from these preclinical studies suggest the potential clinical value of retroviral vector-mediated cytokine gene therapy for systemic cancer.
Collapse
|
35
|
Petersson M, Charo J, Salazar-Onfray F, Noffz G, Mohaupt M, Qin Z, Klein G, Blankenstein T, Kiessling R. Constitutive IL-10 Production Accounts for the High NK Sensitivity, Low MHC Class I Expression, and Poor Transporter Associated with Antigen Processing (TAP)-1/2 Function in the Prototype NK Target YAC-1. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.5.2099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Tumor cells that are treated with rIL-10 or transfected with the IL-10 gene show phenotypic changes. These include low but peptide-inducible expression of MHC class I, low sensitivity to specific CTL-mediated lysis, and increased NK sensitivity. In vitro-established mouse tumor lines were screened for IL-10 expression and production, and a large proportion of plasmocytomas or T cell lymphomas were found to produce IL-10. Since one of these lines was the prototype NK target cell YAC-1, we investigated whether the high IL-10 production of this cell line was related to its high NK sensitivity and its defects in MHC class I expression. The decrease in H-2 expression following the in vitro culture of in vivo-passaged YAC-1 cells was accompanied by a gradual increase in IL-10 production, whereas the reverse was found when passing in vitro-grown YAC-1 in vivo as an ascites tumor in syngenic mice. In addition, differences in YAC-1 MHC class I expression correlated with alterations in the functional activity of TAP-1/2 proteins. YAC-1 cells that were transduced with a retroviral IL-10 antisense construct (Y-IL-10 AS) only produced about half of the IL-10 that was produced by YAC-1 transduced with the control construct (Y-IL-10 Mock). Relative to Y-IL-10 Mock cells, the expression of H-2 on Y-IL-10 AS cells was markedly increased, and NK sensitivity was decreased. These data argue for a mechanism wherein IL-10 production is causally related to the low H-2 expression, decreased TAP function, and high NK sensitivity of YAC-1 cells.
Collapse
Affiliation(s)
- Max Petersson
- *Microbiology and Tumor Biology Center (MTC), Karolinska Institute, Stockholm, Sweden
| | - Jehad Charo
- *Microbiology and Tumor Biology Center (MTC), Karolinska Institute, Stockholm, Sweden
| | - Flavio Salazar-Onfray
- *Microbiology and Tumor Biology Center (MTC), Karolinska Institute, Stockholm, Sweden
| | - Gabriele Noffz
- †Max-Delbruck Center for Molecular Medicine (MDC), Berlin-Buch, Germany; and
| | - Mariette Mohaupt
- †Max-Delbruck Center for Molecular Medicine (MDC), Berlin-Buch, Germany; and
| | - Zhihai Qin
- †Max-Delbruck Center for Molecular Medicine (MDC), Berlin-Buch, Germany; and
| | - George Klein
- *Microbiology and Tumor Biology Center (MTC), Karolinska Institute, Stockholm, Sweden
| | - Thomas Blankenstein
- †Max-Delbruck Center for Molecular Medicine (MDC), Berlin-Buch, Germany; and
| | - Rolf Kiessling
- *Microbiology and Tumor Biology Center (MTC), Karolinska Institute, Stockholm, Sweden
- ‡Department of Experimental Oncology, Radiumhemmet, Karolinska Hospital, Stockholm, Sweden
| |
Collapse
|
36
|
Suzuki S, Umezu Y, Saijo Y, Satoh G, Abe Y, Satoh K, Nukiwa T. Exogenous recombinant human IL-12 augments MHC class I antigen expression on human cancer cells in vitro. TOHOKU J EXP MED 1998; 185:223-6. [PMID: 9823783 DOI: 10.1620/tjem.185.223] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We investigated whether expressions of MHC class I and class II antigens relevant to tumor antigen presentation were changed on human tumor cells cultured with or without recombinant human IL-12(rhIL-12). We showed that the expression of MHC class I antigen on UTC-8, 28-1Cl and SBC-3 cells was augmented when these cancer cells were cultured with rhIL-12. The expression of class II antigen was slightly raised on UTC-8 and 28-1Cl cells by rhIL-12, but not enhanced on SBC-3 cells. These results suggest that rhIL-12 may provide possible enhancement of immunologic tumor recognition, and cytotoxic activity of lymphocytes against tumors through the enhanced expression of MHC class I antigen.
Collapse
Affiliation(s)
- S Suzuki
- Department of Respiratory Oncology and Molecular Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | | | | | | | | | | | | |
Collapse
|
37
|
Genetic Approaches To Adoptive Cellular Immunotheraphy. Surg Oncol Clin N Am 1998. [DOI: 10.1016/s1055-3207(18)30257-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
38
|
|
39
|
Qian SB, Chen SS. Transduction of human hepatocellular carcinoma cells with human γ-interferon gene via retroviral vector. World J Gastroenterol 1998; 4:210-213. [PMID: 11819277 PMCID: PMC4723458 DOI: 10.3748/wjg.v4.i3.210] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the therapeutic potential of gamma interferon (IFN-γ) genemodified human hepatocellular carcinoma (HCC) cells.
METHODS: The IFN-γ gene was introduced retrovirally into four HCC cell lines. Secreted IFN-γ activity was assessed using bioassay. The expression of MHC molecules was detected by FACS. Tumorigenicity was analysed by tumor formation in nude mice.
RESULTS: Four IFN-γ gene transduced HCC cell lines secreted different amounts of IFN-γ, as in the same case of five clones derived from one HCC cell line. Transduction with IFN-γ caused significant increase in the expression of major histocompatibility complex (MHC) antigens on HCC cells. The expression of HLA class I was increased by 2-3 times in terms of mean fluorescence intensities, while for class II expression, the percentage of positive cells augmented from < 10% to > 50%. When equal amount of tumor cells were injected into nude mice, the tumor igenicity some transduced cells decreased dramantically.
CONCLUSION: IFN-γ gene transduction can convert weakly imunogenic HCC cells to activate antitumor immune response, and further pave the way for the future use of such gene modified tumor cells as a modality for the cancer immunotherapy.
Collapse
|
40
|
Emtage PCR, Wan Y, Bramson JL, Graham FL, Gauldie J. A Double Recombinant Adenovirus Expressing the Costimulatory Molecule B7-1 (Murine) and Human IL-2 Induces Complete Tumor Regression in a Murine Breast Adenocarcinoma Model. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.5.2531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Tumors that express tumor-specific antigens can maintain growth in an immunocompetent organism. Current hypotheses tend toward T cell anergy as a key component for the inhibition of immunoreactivity against such tumors. Anergy is thought to occur from hyperactive stimulation of the TCR in the absence of costimulation (costimulation leads to proliferation via IL 2 production). Subcutaneous injection of transgenic polyoma middle T transformed breast adenocarcinoma tumor cells (PyMT) in the hind flank of FVB/n mice results in the formation of tumor nodules at this site. We determined the MHC class I and class II, B7-1, and B7-2 expression in the tumor cells by flow cytometry and showed positive staining for only MHC class I. We show that a single E1-deleted adenovirus constructed to express both the costimulatory molecule B7-1 (murine) and human IL-2 genes (Ad5E1 mB7-1/human IL-2) elicits a very potent antitumor response when administered intratumorally. Ad5E1 mB7-1/human IL-2 induced rapid and complete regression (100%) of all tumors compared with Ad5 E1 mB7-1 (38%), Ad CAIL-2 (42%), and Ad5E1 dl70-3 (control vector) (0%). All mice that exhibited complete tumor regression were fully protected in tumor cell challenge experiments. The systemic immunity generated by intratumoral administration of the Ad vectors was associated with a strong anti-PyMT CTL response. These observations indicate that augmenting the immunogenicity of the tumor with coincident expression of B7-1 in combination with IL-2 may prove beneficial in direct tumor immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Frank L. Graham
- *Pathology and
- †Biology, McMaster University, Hamilton, Ontario, Canada
| | | |
Collapse
|
41
|
Tartour E, Gey A, Sastre-Garau X, Lombard Surin I, Mosseri V, Fridman WH. Prognostic value of intratumoral interferon gamma messenger RNA expression in invasive cervical carcinomas. J Natl Cancer Inst 1998; 90:287-94. [PMID: 9486814 DOI: 10.1093/jnci/90.4.287] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The production of the cytokine interferon gamma (IFN gamma) by activated peripheral blood mononuclear cells may be reduced in patients with invasive cervical carcinoma. This study was designed to assess the prognostic value of intratumoral IFN gamma messenger RNA (mRNA) levels in such patients. METHODS Biopsy specimens of primary cervical lesions were obtained from 27 patients with invasive squamous cell carcinoma before they received any therapy. Two prognostic groups were considered: 1) a group of 14 patients who had no apparent disease recurrence and who were alive 2 years after diagnosis (good-prognosis group) and 2) a group of 13 patients who had disease recurrence or died during the 2-year follow-up (poor-prognosis group). A competitive reverse transcription-polymerase chain reaction assay was used to measure levels of IFN gamma and beta actin mRNA. The expression of human leukocyte antigen (HLA) class II proteins (which is stimulated by IFN gamma) in tumor cells was studied by immunostaining. RESULTS Tumor specimens from all 14 patients in the good-prognosis group contained more than 10(3) IFN gamma mRNA copies per 5 x 10(5) beta actin mRNA copies, whereas tumor specimens from only six of the 13 patients in the poor-prognosis group contained this level of IFN gamma mRNA (two-sided P = .006). No clear relationship was observed between levels of IFN gamma mRNA and T-cell or natural killer cell infiltration in tumors; however, a statistically significant association was observed between HLA class II expression on tumor cells and IFN gamma mRNA levels (two-sided P = .01). CONCLUSIONS A subgroup of poor-prognosis cervical carcinoma patients who have low levels of intratumoral IFN gamma mRNA was identified.
Collapse
Affiliation(s)
- E Tartour
- Laboratoire d'Immunologie Clinique, INSERM U255, Institut Curie, Paris, France.
| | | | | | | | | | | |
Collapse
|
42
|
Zhang R, Minemura K, De Groot LJ. Immunotherapy for medullary thyroid carcinoma by a replication-defective adenovirus transducing murine interleukin-2. Endocrinology 1998; 139:601-8. [PMID: 9449631 DOI: 10.1210/endo.139.2.5764] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We have evaluated the feasibility of gene transduction using replication-defective adenovirus vector as a novel therapy for medullary thyroid carcinoma (MTC), a thyroid C cell neoplasm. Replication-defective adenoviruses were constructed to express murine interleukin-2 (mIL-2) gene and Escherichia coli beta-galactosidase (beta-gal; lacZ) gene under the control of the human cytomegalovirus (CMV) promoter (AdCMVmIL2, AdCMVbeta-gal) by homologous recombination. The efficiency of transduction was evaluated using AdCMVbeta-gal at different conditions. The gene transduction efficiency was dependent on multiplicity of infection, duration of exposure to the virus, and viral concentration. The expression of functional mIL-2 in transduced tumor cells was verified both in vitro and in vivo. Two cell lines (rat MTC and mMTC) secreted large amounts of functional mIL-2 after transduction, as tested in cytotoxic T lymphocyte (CTL) L-2 cells. When AdCMVmIL2-infected mMTC cells were injected s.c. into their host animals, tumors developed in 2 of 10 animals, in contrast to 9 of 10 animals injected with AdCMVbeta-gal-infected mMTC cells and all 10 animals injected with parental mMTC cells. Moreover protected animals developed a long lasting immunity against mMTC tumor cells and their splenocytes, showing cytotoxicity to parental tumor cells, and active natural killer (NK) cell activity. BALB/c-SCID (severe combined immune deficiency) mice were also used to evaluate the function of NK cells in antitumor activities. No tumor developed in SCID mice injected with AdCMVmIL2-infected cells, whereas all animals injected with either AdCMVbeta-gal-infected or parental mMTC cells developed tumors. Our data indicate that IL-2 production by MTC cells leads to rejection in syngeneic animals and suggest that both cytotoxic T cells and NK cells may play an important role. In addition, transduction of adenoviral vectors into tumor cells produces some nonspecific antitumor effects.
Collapse
Affiliation(s)
- R Zhang
- Department of Medicine, University of Chicago, Illinois 60637, USA
| | | | | |
Collapse
|
43
|
Abstract
PROBLEM The question of how trophoblastic and malignant cells evade immunologic recognition and rejection by their host was studied. METHOD OF STUDY A literature review was conducted. RESULTS Trophoblastic and malignant cells share a number of similarities. These include a lack of major histocompatibility complex antigen expression, resistance to lysis by natural killer cells, T-helper cell-2 (TH2)-biased response, prostaglandin E production, and response to transforming growth factor beta. In addition, the analogies between trophoblastic and malignant cells extend into immunotherapy in which anti-idiotype therapy has a viable role in the prevention of pregnancy loss and the treatment of cancer. CONCLUSIONS Trophoblastic and malignant cells use a number of similar mechanisms to resist rejection by their host. By using similar strategies these cells are able to successfully co-exist in an immunologically hostile environment.
Collapse
Affiliation(s)
- C A Mullen
- Department of Biology, Indiana University-Purdue University Indianapolis 46250-0457, USA
| |
Collapse
|
44
|
Affiliation(s)
- J S Lebkowski
- Applied Immune Sciences, Inc., Santa Clara, California 95054-1114, USA
| | | | | |
Collapse
|
45
|
Sjögren HO. Therapeutic immunization against cancer antigens using genetically engineered cells. IMMUNOTECHNOLOGY : AN INTERNATIONAL JOURNAL OF IMMUNOLOGICAL ENGINEERING 1997; 3:161-72. [PMID: 9358269 DOI: 10.1016/s1380-2933(97)00015-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- H O Sjögren
- Department of Cell and Molecular Biology, Wallengerg Laboratory, University of Lund, Sweden
| |
Collapse
|
46
|
Hellström KE, Gladstone P, Hellström I. Cancer vaccines: challenges and potential solutions. MOLECULAR MEDICINE TODAY 1997; 3:286-90. [PMID: 9257295 DOI: 10.1016/s1357-4310(97)01048-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Almost a century has passed since immunotherapy of cancer was first attempted using cancer immunogens (vaccines); however, its clinical impact remains modest. Although initial concerns about a lack of human tumor antigens have decreased, prevailing issues include inefficient procedures for immunization and downregulated expression of major histocompatibility complex (MHC) class I molecules in tumor cells. While immunization can be improved, deficient MHC class I expression remains a problem, because it hampers the ability of tumor cells to present antigens for killing by CD8+ T cells. These are the major mediators of tumor destruction, and they have little or no activity against antigen-negative bystander cells. However, there are reasons to be optimistic that therapeutic vaccination against cancer antigens might become a reality at last.
Collapse
Affiliation(s)
- K E Hellström
- Bristol-Myers Squibb Pharmacetuical Research Institute, Seattle, WA 98121, USA
| | | | | |
Collapse
|
47
|
Mackensen A, Lindemann A, Mertelsmann R. Immunostimulatory cytokines in somatic cells and gene therapy of cancer. Cytokine Growth Factor Rev 1997; 8:119-28. [PMID: 9244407 DOI: 10.1016/s1359-6101(96)00052-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The use of immunostimulatory cytokines has become an increasingly promising approach in cancer immunotherapy. The major goal is the activation of tumour-specific T lymphocytes capable of rejecting tumour cells from patients with low tumour burden or to protect patients from a recurrence of the disease. Strategies that provide high levels of immunostimulatory cytokines locally at the site of antigen have demonstrated pre-clinical and occasional clinical efficacy. Animal models using poorly immunogenic tumours revealed that tumour cells genetically engineered to produce cytokines like IL-2, IL-4, IL-7, IL-12, IFNs, GM-CSF or TNF-alpha were found to be effective in eradicating disseminated tumours. Experimental data obtained from these different animal models are reviewed here to provide an overview of this rapidly evolving field. The data obtained so far from clinical trials involving cytokine gene-modified cells have provided important information regarding the feasibility, safety, immunological effects and occasional clinical responses.
Collapse
Affiliation(s)
- A Mackensen
- Department of Hematology/Oncology, Freiburg Medical University Center, Germany.
| | | | | |
Collapse
|
48
|
Viret C, Lindemann A. Tumor immunotherapy by vaccination with cytokine gene transfected cells. Int Rev Immunol 1997; 14:193-212. [PMID: 9131387 DOI: 10.3109/08830189709116852] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- C Viret
- Yale University School of Medicine, Section of Immunobiology, New Haven, CT 06520-8011, USA
| | | |
Collapse
|
49
|
Roth JA, Cristiano RJ. Gene therapy for cancer: what have we done and where are we going? J Natl Cancer Inst 1997; 89:21-39. [PMID: 8978404 DOI: 10.1093/jnci/89.1.21] [Citation(s) in RCA: 404] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Gene-based therapies for cancer in clinical trials include strategies that involve augmentation of immunotherapeutic and chemotherapeutic approaches. These strategies include ex vivo and in vivo cytokine gene transfer, drug sensitization with genes for prodrug delivery, and the use of drug-resistance genes for bone marrow protection from high-dose chemotherapy. Inactivation of oncogene expression and gene replacement for tumor suppressor genes are among the strategies for targeting the underlying genetic lesions in the cancer cell. A review of clinical trial results to date, primarily in patients with very advanced cancers refractory to conventional treatments, indicates that these treatments can mediate tumor regression with acceptably low toxicity. Vector development remains a critical area for future research. Important areas for future research include modifying viral vectors to reduce toxicity and immunogenicity, increasing the transduction efficiency of nonviral vectors, enhancing vector targeting and specificity, regulating gene expression, and identifying synergies between gene-based agents and other cancer therapeutics.
Collapse
Affiliation(s)
- J A Roth
- Department of Thoracic and Cardiovascular Surgery, University of Texas M.D. Anderson Cancer Center, Houston 77030, USA
| | | |
Collapse
|
50
|
Toloza EM, Hunt K, Miller AR, McBride W, Lau R, Swisher S, Rhoades K, Arthur J, Choi J, Chen L, Chang P, Chen A, Glaspy J, Economou JS. Transduction of murine and human tumors using recombinant adenovirus vectors. Ann Surg Oncol 1997; 4:70-9. [PMID: 8985520 DOI: 10.1007/bf02316813] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Most cytokine-based cancer gene therapy clinical trials have used labor-intensive, retrovirus-mediated strategies resulting in unpredictable gene expression. Recombinant AdV vectors were evaluated for easier, more reproducible gene transfer into 12 human melanoma, 2 murine fibrosarcomas, and 8 other tumor cell lines. METHODS AdV vectors contained a reporter (Escherichia coli beta-galactosidase or firefly luciferase) or cytokine gene (human interleukin-2 [IL-2] or IL-7). Transduction efficiencies and expression levels were assessed by histochemical staining, flow cytometry, polymerase chain reaction, fluorometry, and enzyme-linked immunosorbent assay. Tumorigenicity was determined by subcutaneous injection of cells into syngeneic mice. RESULTS All cell lines studied were transduced with AdV. Most cell lines exhibited 100% transduction efficiencies (by flow cytometry) at multiplicities of infection (MOI) epsilon 10. Gene expression correlated linearly with MOI, but a cytopathic effect was observed at MOI > 100 with all vectors. Nanogram gene expression levels were routinely achieved. Irradiation (30 Gy) minimally affected expression levels. Tumorigenicity of AdV-IL-2-transduced fibrosarcoma cells in mice was inversely related to IL-2 production. A majority of mice that rejected their tumor challenge were immune to tumor rechallenge. CONCLUSIONS E1-deleted AdV vectors may prove useful in generating tumor vaccines ex vivo with high, transient cytokine expression levels.
Collapse
Affiliation(s)
- E M Toloza
- Division of Surgical Oncology, University of California at Los Angeles School of Medicine 90024, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|