1
|
Cao D, Song Q, Li J, Chard Dunmall LS, Jiang Y, Qin B, Wang J, Guo H, Cheng Z, Wang Z, Lemoine NR, Lu S, Wang Y. Redirecting anti-Vaccinia virus T cell immunity for cancer treatment by AAV-mediated delivery of the VV B8R gene. Mol Ther Oncolytics 2022; 25:264-275. [PMID: 35615262 PMCID: PMC9114156 DOI: 10.1016/j.omto.2022.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 04/21/2022] [Indexed: 11/28/2022] Open
Abstract
Immunotherapies, such as immune checkpoint inhibitors (ICIs) and chimeric antigen receptor-T (CAR-T) cells, are only efficient in a small proportion of tumor patients. One of the major reasons for this is the lack of immune cell infiltration and activation in the tumor microenvironment (TME). Recent research reported that abundant bystander CD8+ T cells targeting viral antigens exist in tumor infiltrates and that virus-specific memory T cells could be recalled to kill tumor cells. Therefore, virus-specific memory T cells may be effective candidates for tumor immunotherapy. In this study, we established subcutaneous tumor mice models that were pre-immunized with Vaccinia virus (VV) and confirmed that tumor cells with ectopic expression of the viral B8R protein could be recognized and killed by memory T cells. To create a therapeutic delivery system, we designed a recombinant adeno-associated virus (rAAV) with a modified tumor-specific promoter and used it to deliver VV B8R to tumor cells. We observed that rAAV gene therapy can retard tumor growth in VV pre-immunized mice. In summary, our study demonstrates that rAAV containing a tumor-specific promoter to restrict VV B8R gene expression to tumor cells is a potential therapeutic agent for cancer treatment in VV pre-immunized or VV-treated mice bearing tumors.
Collapse
Affiliation(s)
- Dujuan Cao
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qianqian Song
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Junqi Li
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Louisa S. Chard Dunmall
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Yuanyuan Jiang
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Bin Qin
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jianyao Wang
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Haoran Guo
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhenguo Cheng
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhimin Wang
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Nicholas R. Lemoine
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Shuangshuang Lu
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yaohe Wang
- National Center for International Research in Cell and Gene Therapy, Sino-British Research Centre for Molecular Oncology, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
2
|
Pires IS, Hammond PT, Irvine DJ. Engineering Strategies for Immunomodulatory Cytokine Therapies - Challenges and Clinical Progress. ADVANCED THERAPEUTICS 2021; 4:2100035. [PMID: 34734110 PMCID: PMC8562465 DOI: 10.1002/adtp.202100035] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Indexed: 12/15/2022]
Abstract
Cytokines are immunoregulatory proteins involved in many pathological states with promising potential as therapeutic agents. A diverse array of cytokines have been studied in preclinical disease models since the 1950s, some of which became successful biopharmaceutical products with the advancement of recombinant protein technology in the 1980s. However, following these early approvals, clinical translation of these natural immune signaling molecules has been limited due to their pleiotropic action in many cell types, and the fact that they have evolved to act primarily locally in tissues. These characteristics, combined with poor pharmacokinetics, have hindered the delivery of cytokines via systemic administration routes due to dose-limiting toxicities. However, given their clinical potential and recent clinical successes in cancer immunotherapy, cytokines continue to be extensively pursued in preclinical and clinical studies, and a range of molecular and formulation engineering strategies are being applied to reduce treatment toxicity while maintaining or enhancing therapeutic efficacy. This review provides a brief background on the characteristics of cytokines and their history as clinical therapeutics, followed by a deeper discussion on the engineering strategies developed for cytokine therapies with a focus on the translational relevance of these approaches.
Collapse
Affiliation(s)
- Ivan S Pires
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
3
|
Tornatore C, Rabin S, Baker-Cairns B, Keir S, Mocchetti I. Engraftment of C6-2B Cells into the Striatum of Aci Nude Rats as a Tool for Comparison of the in Vitro and in Vivo Phenotype of a Glioma Cell Line. Cell Transplant 2017; 6:317-26. [PMID: 9171164 DOI: 10.1177/096368979700600314] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The C6-2B is a well-characterized glioma cell line used extensively in the study of malignant glial biology. While the C6-2B cell line has traditionally been thought of as a homogenous cell line, the in vitro phenotype of the C6-2B cell line can vary considerably depending on the culture technique used and the stratum on which the cells are grown. Thus, we asked whether the in vitro phenotype of the C6-2B cell line was significantly different than the in vivo phenotype of the cell line once it was engrafted into the striatum of nude rats. Under culture conditions used in our laboratory, 100% of the C6 cells were found to express p75, the low-affinity nerve growth factor (NGF) receptor, and Major Histocompatability Class I (MHC Class I), while only 10-15% demonstrated vimentin reactivity. Immunohistochemistry was consistently negative for GFAP, trkA (the high-affinity receptor for NGF), CD4, CD8, and a macrophage specific marker (Ox-41). Once engrafted into the striatum of nude rats, the cells remained 100% p75 and MHC Class I positive, and again, only 15% of the cells demonstrated vimentin reactivity. The grafted cells retained this characteristic for 28 days in vivo. Although an immunoincompetent host was selected to minimize the effects an inflammatory response would have on the graft, a transient inflammatory response was detected. During the first week of engraftment, numerous MHC class II cells, some of which were macrophages, were seen infiltrating the graft. However, by 4 weeks postengraftment, no inflammatory cells were appreciated in the graft and surprisingly little reactive gliosis was seen in the penumbra of the tumor mass. Thus, the limited number of in vitro phe-notypic characteristics we examined in the C6-2B cell line remained constant once the cells were engrafted into the striatum of athymic nude rats.
Collapse
Affiliation(s)
- C Tornatore
- Department of Neurology, Georgetown University Medical Center, Washington, DC 20007, USA
| | | | | | | | | |
Collapse
|
4
|
Kim J, Kim W, Moon UJ, Kim HJ, Choi HJ, Sin JI, Park NH, Cho HR, Kwon B. Intratumorally Establishing Type 2 Innate Lymphoid Cells Blocks Tumor Growth. THE JOURNAL OF IMMUNOLOGY 2016; 196:2410-23. [PMID: 26829987 DOI: 10.4049/jimmunol.1501730] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/22/2015] [Indexed: 12/12/2022]
Abstract
A long-standing question in the field of tumor immunotherapy is how Th2 cytokines block tumor growth. Their antitumor effects are particularly prominent when they are secreted continuously in tumors, suggesting that Th2 cytokines may create a tumor microenvironment unfavorable for tumor growth independently of adaptive immunity. In this study, we show that local production of IL-33 establishes a high number of type 2 innate lymphoid cells (ILC2s) with potent antitumor activity. IL-33 promotes secretion of a massive amount of CXCR2 ligands from ILC2s but creates a tumor microenvironment where tumor cells express CXCR2 through a dysfunctional angiogenesis/hypoxia/reactive oxygen species axis. These two signaling events converge to reinforce tumor cell-specific apoptosis through CXCR2. Our results identify a previously unrecognized antitumor therapeutic pathway wherein ILC2s play a central role.
Collapse
Affiliation(s)
- Juyang Kim
- Biomedical Research Center, Ulsan University Hospital and School of Medicine, University of Ulsan, Ulsan 682-714, Republic of Korea
| | - Wonyoung Kim
- Biomedical Research Center, Ulsan University Hospital and School of Medicine, University of Ulsan, Ulsan 682-714, Republic of Korea
| | - U J Moon
- School of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Hyun J Kim
- School of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea
| | - Hye-Jeong Choi
- Department of Pathology, Ulsan University Hospital and School of Medicine, University of Ulsan, Ulsan 682-714, Republic of Korea
| | - Jeong-Im Sin
- Department of Microbiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, Republic of Korea
| | - Neung H Park
- Biomedical Research Center, Ulsan University Hospital and School of Medicine, University of Ulsan, Ulsan 682-714, Republic of Korea; Department of Internal Medicine, Ulsan University Hospital and School of Medicine, University of Ulsan, Ulsan 682-714, Republic of Korea; and
| | - Hong R Cho
- Biomedical Research Center, Ulsan University Hospital and School of Medicine, University of Ulsan, Ulsan 682-714, Republic of Korea; Department of Surgery, Ulsan University Hospital and School of Medicine, University of Ulsan, Ulsan 682-714, Republic of Korea
| | - Byungsuk Kwon
- Biomedical Research Center, Ulsan University Hospital and School of Medicine, University of Ulsan, Ulsan 682-714, Republic of Korea; School of Biological Sciences, University of Ulsan, Ulsan 680-749, Republic of Korea;
| |
Collapse
|
5
|
Schweighoffer T. Molecular cancer vaccines: Tumor therapy using antigen-specific immunizations. Pathol Oncol Res 2012; 3:164-76. [PMID: 18470726 DOI: 10.1007/bf02899917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/1997] [Accepted: 08/24/1997] [Indexed: 10/21/2022]
Abstract
Vaccination against tumors promises selective destruction of malignant cells by the host's immune system. Molecular cancer vaccines rely on recently identified tumor antigens as immunogens. Tumor antigens can be applied in many forms, as genes in recombinant vectors, as proteins or peptides representing T cell epitopes.Analysis of various aspects indicates some advantage for peptide-based vaccines over the other modalities. Further refinements and extensively monitored clinical trials are necessary to advance molecular cancer vaccines from concepts into powerful therapy.
Collapse
Affiliation(s)
- T Schweighoffer
- Department Cell Biology, Boehringer Ingelheim Research and Development, Dr. Boehringer-Gasse 5, A-l 120, Wien, Austria,
| |
Collapse
|
6
|
Shi YP, Guo Y, Li HP, Shen JJ, Yang JY. Establishment of a CBRH7919 cell line expressing rat IL-2 and B7-1. Shijie Huaren Xiaohua Zazhi 2011; 19:2660-2663. [DOI: 10.11569/wcjd.v19.i25.2660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [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 establish a CBRH7919 cell line CBRH7919 expressing rat IL-2 and B7-1 genes and to examine their ability to express the IL-2 and B7-1 genes in vitro.
METHODS: The IL-2 and B7-1 genes were amplified by RT-PCR and subcloned into retroviral vectors pBaBe-puro and pMSCV-neo, respectively, to obtain the recombinant retroviral vectors pBaBe-puro-IL-2 and pMSCV-neo-B7-1. The recombinant plasmids were then transfected into the 293FT packaging cells. The obtained infectious viruses were used to infect the CBRH7919 cell line, and puro/G418-resistant clones were acquired after puro/G418 selection. The expression of IL-2 and B7-1 was detected using Q-PCR, Western blot and ELISA.
RESULTS: The rat IL-2 and B7-1 genes were successfully amplified by RT-PCR, and the recombinant plasmids pBaBe-puro-IL-2 and pMSCV-neo-B7-1 were successfully constructed and verified by direct sequencing. A CBRH7919 cell line (CBRH7919/IL-2/B7-1) expressing rat IL-2 and B7-1 was established. Q-PCR analysis showed that the expression levels of IL-2 and B7-1 mRNAs in CBRH-7919/IL-2/B7-1 cells were 4.15 and 17.04 times higher than those in CBRH-7919-pmscv-neo cells. Western blot analysis showed that the expression level of B7-1 protein in CBRH-7919/IL-2/B7-1 cells was 3 times more than that in unmodified cells, while ELISA showed that expression level of IL-2 was 190 times more than that in unmodified cells.
CONCLUSION: A CBRH7919 cell line stably and effectively expressing rat IL-2 and B7-1 genes was obtained and provides a good basis for further research of immuno-gene therapy of liver cancer.
Collapse
|
7
|
Kim YS. Tumor Therapy Applying Membrane-bound Form of Cytokines. Immune Netw 2009; 9:158-68. [PMID: 20157604 PMCID: PMC2816950 DOI: 10.4110/in.2009.9.5.158] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Accepted: 10/11/2009] [Indexed: 12/18/2022] Open
Abstract
Tumor therapy using cytokines has been developed for last two decades. Several recombinant cytokines and tumor cell vaccines produced by cytokine gene transfer have been in clinical trials, but several side effects hamper routine clinical applications. Many cytokines are originally expressed as membrane-bound form and then processed to secretory form exerting paracrine effects. Though functional differences of these two types of cytokines are elusive yet, the membrane-bound form of cytokine may exert its effects on restricted target cells as a juxtacrine, which are in physical contacts. With the efforts to improve antitumor activities of cytokines in cancer patients, developing new strategies to alleviate life-threatening side effects became an inevitable goal of tumor immunologists. Among these, tumor cell vaccines expressing cytokines as membrane-bound form on tumor cell surface have been developed by genetic engineering techniques with the hope of selective stimulation of the target cells that are in cell-to-cell contacts. In this review, recent progress of tumor cell vaccines expressing membrane-bound form of cytokines will be discussed.
Collapse
Affiliation(s)
- Young Sang Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Korea
| |
Collapse
|
8
|
Gene therapy of benign gynecological diseases. Adv Drug Deliv Rev 2009; 61:822-35. [PMID: 19446586 DOI: 10.1016/j.addr.2009.04.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Accepted: 04/28/2009] [Indexed: 11/22/2022]
Abstract
Gene therapy is the introduction of genetic material into patient's cells to achieve therapeutic benefit. Advances in molecular biology techniques and better understanding of disease pathogenesis have validated the use of a variety of genes as potential molecular targets for gene therapy based approaches. Gene therapy strategies include: mutation compensation of dysregulated genes; replacement of defective tumor-suppressor genes; inactivation of oncogenes; introduction of suicide genes; immunogenic therapy and antiangiogenesis based approaches. Preclinical studies of gene therapy for various gynecological disorders have not only shown to be feasible, but also showed promising results in diseases such as uterine leiomyomas and endometriosis. In recent years, significant improvement in gene transfer technology has led to the development of targetable vectors, which have fewer side-effects without compromising their efficacy. This review provides an update on developing gene therapy approaches to treat common gynecological diseases such as uterine leiomyoma and endometriosis.
Collapse
|
9
|
Induction of Antitumor Response by In Vivo Allogeneic Major Histocompatibility Complex Gene Transfer Using Electroporation. J Surg Res 2009; 154:60-7. [DOI: 10.1016/j.jss.2008.05.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Revised: 05/12/2008] [Accepted: 05/27/2008] [Indexed: 11/21/2022]
|
10
|
Abstract
On the basis of experimental models and some human data, we can assume that tumor outgrowth results from the balance between immunosurveillance (the extrinsic tumor suppressor mechanisms) and immunosubversion dictated by transformed cells and/or the corrupted surrounding microenvironment. Cancer immunosurveillance relies mainly upon conventional lymphocytes exerting either lytic or secretory functions, whereas immunosubversion results from the activity of regulatory T or suppressor myeloid cells and soluble mediators. Although specific tools to target or ablate dendritic cells (DCs) became only recently available, accumulating evidence points to the critical role of the specialized DC system in dictating most of the conventional and regulatory functions of tumor-specific T lymphocytes. Although DC can be harnessed to silence tumor development, tumors in turn can exploit DC to evade immunity. Indeed, DCs harbor defects in their differentiation and stimulatory functions in cancer-bearing hosts and can actively promote T-cell tolerance to self-tumor antigens. In this review, we will focus on the dual role of DC during tumor progression and discuss pharmacoimmunological strategies to harness DC against cancer.
Collapse
|
11
|
Reszka R, Zhu JH, Weber F, Walther W, Greferath R, Dyballa S. Liposome Mediated Transfer of Marker and Cytokine Genes Into Rat and Human Glioblastoma Cells in Vitro and in Vivo. J Liposome Res 2008. [DOI: 10.3109/08982109509039915] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
12
|
Current status of experimental therapeutics for prostate cancer. Cancer Lett 2008; 266:116-34. [DOI: 10.1016/j.canlet.2008.02.065] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2007] [Revised: 02/22/2008] [Accepted: 02/22/2008] [Indexed: 11/17/2022]
|
13
|
Abstract
First, the testing of therapeutic gene vectors in vitro is described. This is followed by a discussion of the administration of therapeutic vectors in vivo. Two methods for assessing the development of anti-tumor immunity after cytokine gene therapy are provided. In addition, two methods for the generation of murine tumor models in syngeneic hostsone subcutaneous and one orthotopicare also included.
Collapse
Affiliation(s)
- Y L Kwong
- Baylor College of Medicine, Houston, Texas, USA
| | | | | |
Collapse
|
14
|
Scott-Ram NR. Review Biologicals & Immunologicals: Gene therapy: review of recent patenting activity. Expert Opin Ther Pat 2008. [DOI: 10.1517/13543776.5.3.219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
15
|
Choi JW, Lim HY, Chang M, Cheon J, Kim YS. Anti‐tumor immunity induced by tumor cells express a membrane‐bound form of IL‐2 and SDF‐1. Anim Cells Syst (Seoul) 2008. [DOI: 10.1080/19768354.2008.9647173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
16
|
Nguyen van Binh P, Duc HT. Analyses and perspectives in cancer immunotherapy. Biomed Pharmacother 2006; 60:621-8. [PMID: 16978826 DOI: 10.1016/j.biopha.2006.07.092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Accepted: 07/28/2006] [Indexed: 01/15/2023] Open
Abstract
Since the last two decades, rapid progress has been made in the field of cancer immunotherapy relevant to manipulation of adaptative cytotoxic T lymphocytes (CTLs) and innate immunity natural killer (NK) cells as well as antibodies. Many possibilities are now offered for therapeutic purposes contributing to better approaches in treatment of cancer.
Collapse
Affiliation(s)
- P Nguyen van Binh
- Inserm, U602, Micro-environnement et Physiopathologie de la Différenciation, Hôpital Paul-Brousse, 16, avenue Paul-Vaillant-Couturier, 94807 Villejuif cedex, France
| | | |
Collapse
|
17
|
Liu J, Xia X, Torrero M, Barrett R, Shillitoe EJ, Li S. The mechanism of exogenous B7.1-enhanced IL-12-mediated complete regression of tumors by a single electroporation delivery. Int J Cancer 2006; 119:2113-8. [PMID: 16823840 DOI: 10.1002/ijc.22100] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Electroporation-based mono-gene therapy has received great interest in recent years but coadministration of different therapeutic genes for treatment of tumors has not been well explored. We hypothesize that electroporation is capable of delivering multiple genes that induce an additive or synergistic antitumor effect. To test this hypothesis, we used mice that were bearing SCCVII or TRAMP tumors. Established tumors with a diameter of 4-5 mm were injected with control plasmid DNA or plasmid DNA encoding B7.1, IL-12 or both via electroporation. Tumor regression, CTL activity and the level of B7.1, IL-12 and Stat1 expression were determined in both wild-type mice and in mice with a knock-out of the Stat1 gene. Remarkably, a single coadministration of the plasmids that encoded IL-12 and B7.1 eradicated tumors in 80% of mice. The therapeutic effect was associated with high levels of endogenous B7.1 expression, activity of cytotoxic lymphocytes, and activation of Stat1. Both exogenous B7.1 and IL-12 were required for inducing a high level of Stat1 activation in tumors, which occurred through a mechanism that was independent of the host Stat1. Both stimulators were also required for inducing the strong cytotoxic lymphocyte activity and for increasing the level and extending the duration of endogenous B7.1 expression. We therefore propose a 2-signal stimulation model to explain the synergistic effect of the coadministration of IL-12 and B7.1 on the regression of the tumors.
Collapse
Affiliation(s)
- Jianguo Liu
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | | | | | | | | | | |
Collapse
|
18
|
Lasfar A, Lewis-Antes A, Smirnov SV, Anantha S, Abushahba W, Tian B, Reuhl K, Dickensheets H, Sheikh F, Donnelly RP, Raveche E, Kotenko SV. Characterization of the mouse IFN-lambda ligand-receptor system: IFN-lambdas exhibit antitumor activity against B16 melanoma. Cancer Res 2006; 66:4468-77. [PMID: 16618774 DOI: 10.1158/0008-5472.can-05-3653] [Citation(s) in RCA: 215] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recently discovered type III IFNs (IFN-lambda) exert their antiviral and immunomodulatory activities through a unique receptor complex composed of IFN-lambdaR1 and interleukin-10 receptor 2. To further study type III IFNs, we cloned and characterized mouse IFN-lambda ligand-receptor system. We showed that, similar to their human orthologues, mIFN-lambda2 and mIFN-lambda3 signal through the IFN-lambda receptor complex, activate IFN stimulated gene factor 3, and are capable of inducing antiviral protection and MHC class I antigen expression in several cell types including B16 melanoma cells. We then used the murine B16 melanoma model to investigate the potential antitumor activities of IFN-lambdas. We developed B16 cells constitutively expressing murine IFN-lambda2 (B16.IFN-lambda2 cells) and evaluated their tumorigenicity in syngeneic C57BL/6 mice. Although constitutive expression of mIFN-lambda2 in melanoma cells did not affect their proliferation in vitro, the growth of B16.IFN-lambda2 cells, when injected s.c. into mice, was either retarded or completely prevented. We found that rejection of the modified tumor cells correlated with their level of IFN-lambda2 expression. We then developed IFN-lambda-resistant B16.IFN-lambda2 cells (B16.IFN-lambda2Res cells) and showed that their tumorigenicity was also highly impaired or completely abolished similar to B16.IFN-lambda2 cells, suggesting that IFN-lambdas engage host mechanisms to inhibit melanoma growth. These in vivo experiments show the antitumor activities of IFN-lambdas and suggest their strong therapeutic potential.
Collapse
Affiliation(s)
- Ahmed Lasfar
- Department of Biochemistry and Molecular Biology, University of Medicine and Dentistry-New Jersey Medical School, Newark, New Jersey 07103, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Cormier SA, Taranova AG, Bedient C, Nguyen T, Protheroe C, Pero R, Dimina D, Ochkur SI, O’Neill K, Colbert D, Lombari TR, Constant S, McGarry MP, Lee JJ, Lee NA. Pivotal Advance: eosinophil infiltration of solid tumors is an early and persistent inflammatory host response. J Leukoc Biol 2006; 79:1131-9. [PMID: 16617160 PMCID: PMC3496422 DOI: 10.1189/jlb.0106027] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tumor-associated eosinophilia has been observed in numerous human cancers and several tumor models in animals; however, the details surrounding this eosinophilia remain largely undefined and anecdotal. We used a B16-F10 melanoma cell injection model to demonstrate that eosinophil infiltration of tumors occurred from the earliest palpable stages with significant accumulations only in the necrotic and capsule regions. Furthermore, the presence of diffuse extracellular matrix staining for eosinophil major basic protein was restricted to the necrotic areas of tumors, indicating that eosinophil degranulation was limited to this region. Antibody-mediated depletion of CD4+ T cells and adoptive transfer of eosinophils suggested, respectively, that the accumulation of eosinophils is not associated with T helper cell type 2-dependent immune responses and that recruitment is a dynamic, ongoing process, occurring throughout tumor growth. Ex vivo migration studies have identified what appears to be a novel chemotactic factor(s) released by stressed/dying melanoma cells, suggesting that the accumulation of eosinophils in tumors occurs, in part, through a unique mechanism dependent on a signal(s) released from areas of necrosis. Collectively, these studies demonstrate that the infiltration of tumors by eosinophils is an early and persistent response that is spatial-restricted. It is more important that these data also show that the mechanism(s) that elicit this host response occur, independent of immune surveillance, suggesting that eosinophils are part of an early inflammatory reaction at the site of tumorigenesis.
Collapse
MESH Headings
- Animals
- CD4 Antigens/immunology
- CD4-Positive T-Lymphocytes/immunology
- Cells, Cultured
- Chemotactic Factors/metabolism
- Chemotaxis/drug effects
- Chemotaxis/physiology
- Culture Media, Conditioned/chemistry
- Culture Media, Conditioned/pharmacology
- Eosinophilia/etiology
- Eosinophilia/physiopathology
- Eosinophils/immunology
- Eosinophils/transplantation
- Immunologic Surveillance
- Immunotherapy, Adoptive
- Inflammation/immunology
- Inflammation/pathology
- Injections, Subcutaneous
- Interleukin-5/genetics
- Lymphocyte Depletion
- Melanoma, Experimental/complications
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Microscopy, Confocal
- Necrosis
- Neoplasm Transplantation
- Th2 Cells/immunology
Collapse
Affiliation(s)
- Stephania A. Cormier
- Division of Hematology and Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard Scottsdale, AZ 85259
- Department of Biological Sciences, Louisiana State University, 202 Life Sciences Annex, Baton Rouge, LA 70803
| | - Anna G. Taranova
- Division of Pulmonary Medicine, Mayo Clinic Arizona, 13400 East Shea Boulevard Scottsdale, AZ 85259
| | - Carrie Bedient
- Division of Pulmonary Medicine, Mayo Clinic Arizona, 13400 East Shea Boulevard Scottsdale, AZ 85259
| | - Thanh Nguyen
- Division of Hematology and Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard Scottsdale, AZ 85259
| | - Cheryl Protheroe
- Division of Hematology and Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard Scottsdale, AZ 85259
| | - Ralph Pero
- Division of Hematology and Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard Scottsdale, AZ 85259
| | - Dawn Dimina
- Division of Pulmonary Medicine, Mayo Clinic Arizona, 13400 East Shea Boulevard Scottsdale, AZ 85259
| | - Sergei I. Ochkur
- Division of Pulmonary Medicine, Mayo Clinic Arizona, 13400 East Shea Boulevard Scottsdale, AZ 85259
| | - Katie O’Neill
- Division of Hematology and Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard Scottsdale, AZ 85259
| | - Dana Colbert
- Division of Hematology and Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard Scottsdale, AZ 85259
| | - Theresa R. Lombari
- Laboratory Animal Research Core (LARC) Facility, Mayo Clinic Arizona, 13400 East Shea Boulevard Scottsdale, AZ 85259
| | - Stephanie Constant
- Department of Microbiology and Tropical Medicine, George Washington University, 2300 Eye Street NW, Washington, DC 20037
| | - Michael P. McGarry
- Division of Pulmonary Medicine, Mayo Clinic Arizona, 13400 East Shea Boulevard Scottsdale, AZ 85259
| | - James J. Lee
- Division of Pulmonary Medicine, Mayo Clinic Arizona, 13400 East Shea Boulevard Scottsdale, AZ 85259
| | - Nancy A. Lee
- Division of Hematology and Oncology, Mayo Clinic Arizona, 13400 East Shea Boulevard Scottsdale, AZ 85259
| |
Collapse
|
20
|
Abstract
Leiomyomas (fibroids) are common estrogen-dependent uterine tumours that cause significant morbidity for women and a substantial economic impact on health delivery systems. Currently, there is no effective medical treatment option for this condition-hysterectomy is the mainstay of management. This is not an attractive choice for many women, especially patients desiring to preserve their fertility potential. Gene therapy is becoming a clinical reality, with more than 600 clinical trials worldwide. Researchers have recently attempted to develop a gene-therapy-based approach for the ablation of uterine fibroids. The localized nature of this condition and its accessibility using different imaging or endoscopic techniques make it an attractive target for direct delivery of gene-based vectors. Recent work from our laboratory suggests the potential use of a dominant-negative form of estrogen receptor (ER) to inactivate estrogen signalling in leiomyoma cells and induce apoptosis. Our in vivo data in a mouse model demonstrate the ability of an adenovirus-expressing dominant-negative ER to arrest leiomyoma growth. We and others also have described the utility of the herpes simplex virus-thymidine kinase (HSV-TK) plus ganciclovir (GCV) suicide gene-therapy system to effectively eradicate leiomyoma cells by utilizing the bystandard effect phenomena and the high expression of gap-junction protein in these tumours. Further work on rat models will pave the way for future leiomyoma gene-therapy clinical trials and allow the realization of gene therapy as a viable non-surgical option for this common problem in women's health.
Collapse
Affiliation(s)
- Ayman Al-Hendy
- Department of Obstetrics & Gynecology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.
| | | |
Collapse
|
21
|
Curigliano G, Spitaleri G, Pietri E, Rescigno M, de Braud F, Cardillo A, Munzone E, Rocca A, Bonizzi G, Brichard V, Orlando L, Goldhirsch A. Breast cancer vaccines: a clinical reality or fairy tale? Ann Oncol 2005; 17:750-62. [PMID: 16293674 DOI: 10.1093/annonc/mdj083] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The characterization of tumor antigens recognized by immune effector cells has opened the perspective of developing therapeutic vaccines in the field of breast cancer. The potential advantages of the vaccines are: (i) the induction of a robust immune response against tumors that are spontaneously weekly immunogenic; (ii) the tumor specificity for some antigens; (iii) the good tolerance and safety profile and (iv) the long-term immune memory, critical to prevent efficiently tumor recurrence. Most trials evaluating breast cancer vaccines have been carried out in patients with extended metastatic breast cancer, characterized by aggressive tumors, resistant to standard cytotoxic treatments, so that clinical efficacy was difficult to achieve. However, some significant immune responses against tumor antigens induced upon vaccinations were recorded. The aim of this review is to analyze the activity of vaccination strategies in current clinical trials. Data of clinical activity have been observed by using vaccines targeting HER2/neu protein, human telomerase reverse transcriptase, carcinoembryonic antigen and carbohydrate antigen given after stem cell rescue. The review discusses possible future directions for vaccine development and applications in the adjuvant setting.
Collapse
Affiliation(s)
- G Curigliano
- Department of Medicine, Clinical Pharmacology and New Drugs Development Unit, Division of Experimental Oncology, Laboratory of Experimental Immunology, European Institute of Oncology, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Tseng SH, Chen Y, Chang CJ, Tai KF, Lin SM, Hwang LH. Induction of T-Cell Apoptosis in Rats by Genetically Engineered Glioma Cells Expressing Granulocyte-Macrophage Colony-Stimulating Factor and B7.1. Clin Cancer Res 2005; 11:1639-49. [PMID: 15746069 DOI: 10.1158/1078-0432.ccr-04-1366] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To evaluate antitumor effects on intracerebral gliomas of genetically engineered tumor vaccines expressing granulocyte-macrophage colony-timulating factor (GM-CSF), B7.1, or both (combination). EXPERIMENTAL DESIGN A rat glioma cell line, RT-2, was engineered with a retroviral vector to express GM-CSF, B7.1, or combination. Tumorigenicity of engineered cells and therapeutic effects of s.c. given irradiated or live tumor vaccines on parental intracerebral gliomas were studied. Immune cell infiltration induced at vaccine and tumor sites was examined by histologic and immunohistochemical staining. Apoptosis of T cells from vaccine sites was analyzed with fluorescence-activated cell sorting. RESULTS Engineered RT-2 cells exhibited reduced s.c. tumorigenicity in rats with reduced tumor growth and prolonged animal survival time compared with control rats. Rats with intracerebral gliomas s.c. treated with irradiated or live GM-CSF-expressing vaccines had 60% and 100% survival rates, respectively, significantly better than the control groups (P < 0.05). In contrast, rats treated with vaccines expressing B7.1 or the combination had no or mild therapeutic effects. Studies revealed less T-cell infiltration at both vaccine and tumor sites in rats treated with vaccines expressing B7.1 or the combination than in rats treated with a vaccine expressing GM-CSF. Cell sorting analyses revealed higher proportions of apoptotic T cells at vaccine sites of rats treated with the combination than those treated with vaccine expressing GM-CSF. CONCLUSIONS Combination of GM-CSF- and B7.1-expressing tumor vaccines exerted no synergistic, or even worse, therapeutic effects on gliomas compared with single GM-CSF-secreting tumor vaccine. The worse therapeutic effects of the GM-B7.1-expressing tumor vaccine than the GM-CSF-expressing tumor vaccine were related to the reduced T-cell amount and increased T-cell apoptosis in the former.
Collapse
Affiliation(s)
- Sheng-Hong Tseng
- Department of Surgery, National Taiwan University Hospital, College of Medicine, 7 Chung-Shan South Road, Taipei 100, Taiwan
| | | | | | | | | | | |
Collapse
|
23
|
Abstract
The prognosis of patients with advanced head and neck cancer has not changed significantly in the last twenty years, despite concerted efforts to optimize treatment using conventional modalities such as surgery, radiotherapy and chemotherapy. Novel therapeutic approaches based on our increasing understanding of the molecular changes that underlie the development of cancer have the potential to alter this situation. Gene therapy involves the delivery of genetic sequences in to tumour or normal cells for a therapeutic purpose. A number of viral and non-viral vectors have been developed that have the ability to deliver therapeutic genes specifically to tumours. These therapeutic genes can exert their effects by correcting existing genetic abnormalities, by killing cells directly or indirectly through recruitment of the immune system. In this review, the various gene therapy strategies that are under development are presented with particular reference to the treatment of head and neck cancer.
Collapse
Affiliation(s)
- Kevin J Harrington
- Targeted Therapy Laboratory, Cancer Research UK, Centre for Cell and Molecular Biology, Chester Beatty Laboratories, Institute of Cancer Research, London, SW3 6JB, UK.
| | | | | |
Collapse
|
24
|
Driessens G, Hamdane M, Cool V, Velu T, Bruyns C. Highly Successful Therapeutic Vaccinations Combining Dendritic Cells and Tumor Cells Secreting Granulocyte Macrophage Colony-stimulating Factor. Cancer Res 2004; 64:8435-42. [PMID: 15548715 DOI: 10.1158/0008-5472.can-04-0774] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In an attempt to induce potent immune antitumor activities, we investigated, within the rat 9L gliosarcoma model, distal therapeutic vaccinations associating three therapies: dendritic cell vaccination, intratumoral granulocyte macrophage colony-stimulating factor (GM-CSF) gene transfer, and tumor apoptosis induction. Vaccines of dendritic cells coinjected with processed GM-CSF secreting 9L cells induced systemic responses, resulting in the complete regression of distant preimplanted 9L tumor masses in, with the best strategy, 94% of male rats. All of the cured rats developed a long-term resistance to a rechallenge with parental cells. The curative responses were correlated with the detection of elevated specific cytotoxic activities and a CD4+, CD8+ T cell-, and natural killer (NK) cell-mediated IFN-gamma production. The survival rate of the rat seemed more directly linked to the amount of GM-CSF secreted by the transduced tumor cells, which in turn depended on the toxicity of the apoptosis-inducing treatment, than to the level of apoptosis induced. Unexpectedly, alive GM-CSF secreting 9L cells became apoptotic when injected in vivo. Thus we documented the positive role of apoptosis in the induction of therapeutic antitumor responses by comparing, at equal GM-CSF exogenous supply, the effects of dendritic cells coinjected with apoptotic or necrotic 9L cells. The data showed the superior therapeutic efficiency of combined vaccines containing apoptotic tumor cells. In conclusion, vaccinations with dendritic cells associated with apoptotic tumor cells secreting GM-CSF show a very high therapeutic potency that should show promise for the treatment of human cancer.
Collapse
Affiliation(s)
- Gregory Driessens
- Interdisciplinary Research Institute (IRIBHM), Faculty of Medicine, Université Libre de Bruxelles, Bruxelles, Belgium
| | | | | | | | | |
Collapse
|
25
|
Abstract
Parvoviruses comprise a group of single-stranded DNA viruses with greater potential for gene therapy applications. Unique characteristics of paroviruses, such as non-pathogenicity, antioncogenicity and methods of efficient recombinant vector production, have drawn more attention towards utilising parvovirus-based vectors in cancer gene therapy. Although > 30 different parvoviruses have been identified so far, recombinant vectors derived from adeno-associated virus (AAV), minute virus of mice (MVM), LuIII and parvovirus H1 have been successfully tested in many preclinical models of human diseases, including cancer. The present article will focus on the potential of non-replicating and autonomously replicating parvoviral vectors in cancer gene therapy, including strategies that target tumour cells directly or indirectly.
Collapse
Affiliation(s)
- Selvarangan Ponnazhagan
- Department of Pathology, LHRB 513, 701 19th Street South, University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA.
| |
Collapse
|
26
|
Huang H, Xiang J. Synergistic effect of lymphotactin and interferon gamma-inducible protein-10 transgene expression in T-cell localization and adoptive T-cell therapy of tumors. Int J Cancer 2004; 109:817-25. [PMID: 15027114 DOI: 10.1002/ijc.20043] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The lack of efficient T-cell infiltration of tumors is a major obstacle to successful adoptive T-cell therapy. We have previously demonstrated that adenovirus (AdV)-mediated transgene lymphotactin (Lptn) or IP-10 expression in tumors can significantly enhance T-cell tumor infiltration. In this study, active OVA-specific CD8+ T cells were prepared by coculturing naive OVA-specific CD8+ T cells from transgenic OT I mice with OVA-I peptide-pulsed dendritic cells in vitro. These XCR-1- and CXCR3-expressing T cells predominantly secreted IFN-gamma and displayed significant killing activity (84% at effector:target cell ratio of 1.5) against OVA-expressing EG7 tumor cells through perforin-mediated pathway. Our data also showed that chemokine Lptn and IP-10 not only can chemoattract, but also stimulate proliferation of CD8+ T cells in vitro, and that a mixture of Lptn and IP-10 can more efficiently chemoattract CD8+ T cells than either one of them. Furthermore, we demonstrated that the transferred CD8+ T cells detected in group of tumors treated with both AdVLptn and AdVIP-10 (group a) are around 4 and 2 times more than that in groups of tumors treated with control AdVpLpA (group b) and either AdVIP-10 (group c) or AdVLptn (group d), respectively. Around 87.5% of mice in group a were tumor-free compared to the aggressive tumor growth in all 8 mice of group b and 25% or 37.5% cured mice seen in groups c and d (p<0.05). Thus, our results indicate that enhancement of adoptive T-cell therapy can be obtained by double tranmsgene Lptn and IP-10 expression, which facilitates CD8+ T-cell tumor localization through proliferation and chemoattraction of the transferred CD8+ T cells by in situ chemokine transgene expressions in the tumors. Collectively, our data provide solid evidence of a potent synergy between adoptive T-cell therapy and adenovirus-mediated Lptn and IP-10 gene transfer into tumor tissues, which culminated in the T-cell tumor localization and eradication of well-established tumor masses.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Cell Division
- Chemokine CXCL10
- Chemokines, C
- Chemokines, CXC/genetics
- Chemotaxis, Leukocyte/immunology
- Cytotoxicity, Immunologic
- Dendritic Cells/immunology
- Drug Synergism
- Female
- Gene Expression Regulation, Neoplastic
- Gene Transfer Techniques
- Immunotherapy, Adoptive
- Lymphocyte Activation
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Lymphokines/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasms/immunology
- Neoplasms/pathology
- Neoplasms/therapy
- Ovalbumin/immunology
- Receptors, Antigen, T-Cell/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Sialoglycoproteins/genetics
- Transgenes
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Hui Huang
- Research Unit, Saskatchewan Cancer Agency, Department of Microbiology, University of Saskatchewan, Saskatoon, Canada
| | | |
Collapse
|
27
|
Wang YQ, Ugai SI, Shimozato O, Yu L, Kawamura K, Yamamoto H, Yamaguchi T, Saisho H, Tagawa M. Induction of systemic immunity by expression of interleukin-23 in murine colon carcinoma cells. Int J Cancer 2003; 105:820-4. [PMID: 12767068 DOI: 10.1002/ijc.11160] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Interleukin-23 (IL-23), a novel cytokine composed of a newly identified p19 molecule and the p40 subunit of IL-12, can stimulate the proliferation in vitro of memory T cells. We examined whether Colon 26 murine colon carcinoma cells that were retrovirally transduced with the p19-linked p40 gene (Colon 26/IL-23) could produce antitumor effects in inoculated mice. The growth of Colon 26/IL-23 tumors developed in immunocompetent mice was significantly retarded and the tumors disappeared thereafter. Spleen cells from the mice that received Colon 26/IL-23 cells produced significant amounts of interferon-gamma, when they were cultured with irradiated Colon 26 but not irrelevant cells. Depletion of CD8(+) T cells suppressed the production of interferon-gamma. The mice that had rejected Colon 26/IL-23 tumors were resistant to subsequent challenge of parent but not irrelevant tumor cells. Colon 26/IL-23 tumors were not rejected in nude mice but the growth was retarded compared to parent tumors. Treatment of nude mice with anti-asialo GM(1) antibody did not influence the growth of Colon 26/IL-23 tumors. These data suggest that expression of IL-23 in tumors produces T cell-dependent antitumor effects and induces systemic immunity.
Collapse
Affiliation(s)
- Yan-Qing Wang
- Division of Pathology, Chiba Cancer Center Research Institute, Chiba, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Trioma cell vaccination is a potent new immunotherapeutic strategy for the treatment of B cell neoplasias. It is based on the specific redirection of tumor antigens against surface receptors on professional antigen-presenting cells (APC) that internalize antigens and present immunogenic peptides to T-lymphocytes. Tumor cells are converted to trioma cells by fusion with xenogeneic hybridomas expressing an anti-APC specificity. The trioma cell is a polyvalent vaccine that contains potentially all lymphoma-derived antigens. Apart from the expression of the APC-binding arm by the trioma cell, another requirement for successful tumor protection is the xenogeneic moiety of the trioma cells. We show that, despite their xenogenicity, trioma cells persist for extended periods in vaccinated animals. Trioma cells could be identified in spleens as long as about 12 weeks post vaccination. By using a suicide gene approach, trioma cells could partly be eliminated from immunized mice, whereby the antitumor effect was partly abrogated. We argue that not all trioma cells are immediately lysed in vivo and that the cell cycle of the remaining cells is arrested after having undergone few divisions. Trioma cells surviving in vivo may be instrumental for efficient induction of tumor immunity.
Collapse
Affiliation(s)
- Nicolas Graf
- GSF-Institut für Molekulare Immunologie, München, Germany
| | | | | |
Collapse
|
29
|
Zöller M. Immunotherapy of cancer by active vaccination: does allogeneic bone marrow transplantation after non-myeloablative conditioning provide a new option? Technol Cancer Res Treat 2003; 2:237-60. [PMID: 12779354 DOI: 10.1177/153303460300200307] [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: 11/17/2022] Open
Abstract
The critical role of antigen-specific T cells in cancer immunotherapy has been amply demonstrated in many model systems. Though success of clinical trials still remains far behind expectation, the continuous improvement in our understanding of the biology of the immune response will provide the basis of optimized cancer vaccines and allow for new modalities of cancer treatment. This review focuses on the current status of active therapeutic vaccination and future prospects. The latter will mainly be concerned with allogeneic bone marrow cell transplantation after non-myeloablative conditioning, because it is my belief that this approach could provide a major breakthrough in cancer immunotherapy. Concerning active vaccination protocols the following aspects will be addressed: i) the targets of immunotherapeutic approaches; ii) the response elements needed for raising a therapeutically successful immune reaction; iii) ways to achieve an optimal confrontation of the immune system with the tumor and iv) supportive regimen of immunomodulation. Hazards which one is most frequently confronted with in trials to attack tumors with the inherent weapon of immune defense will only be briefly mentioned. Many question remain to be answered in the field of allogeneic bone marrow transplantation after non-myeloablative conditioning to optimize the therapeutic setting for this likely very powerful tool of cancer therapy. Current considerations to improve engraftment and to reduce graft versus host disease while strengthening graft versus tumor reactivity will be briefly reviewed. Finally, I will discuss whether tumor-reactive T cells can be "naturally" maintained during the process of T cell maturation in the allogeneic host. Provided this hypothesis can be substantiated, a T cell vaccine will meet a pool of virgin T cells in the allogeneically reconstituted host, which are tolerant towards the host, but not anergised towards tumor antigens presented by MHC molecules of the host.
Collapse
Affiliation(s)
- Margot Zöller
- Dept. of Tumor Progression & Immune Defense, German Cancer Research Center, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany.
| |
Collapse
|
30
|
Abstract
The development of biochemical and genetic schemes to characterize cancer antigens led to the recognition that malignant melanoma frequently evokes a host response. While the generation of brisk T-cell infiltrates in early stage disease is correlated with prolonged survival, host reactions in most cases are insufficient to impede tumor progression. One variable that may limit the potency of the host response against nascent melanoma is the mixture of cytokines present in the tumor microenvironment. In a murine melanoma model, we identified granulocyte-macrophage colony stimulating factor (GM-CSF) as the most potent molecule for augmenting tumor immunity following gene transfer into melanoma cells. Vaccination with irradiated melanoma cells engineered to secrete GM-CSF enhances host responses through improved tumor antigen presentation by recruited dendritic cells and macrophages. Melanoma-specific CD4(+) and CD8(+) T-cells, CD1d-restricted NKT-cells, and antibodies mediate tumor rejection. Initial testing of this immunization strategy in patients with metastatic melanoma revealed the consistent induction of cellular and humoral antitumor responses that provoked the extensive necrosis of distant metastases without significant toxicity.
Collapse
Affiliation(s)
- Glenn Dranoff
- Department of Adult Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| |
Collapse
|
31
|
Rousseau RF, Haight AE, Hirschmann-Jax C, Yvon ES, Rill DR, Mei Z, Smith SC, Inman S, Cooper K, Alcoser P, Grilley B, Gee A, Popek E, Davidoff A, Bowman LC, Brenner MK, Strother D. Local and systemic effects of an allogeneic tumor cell vaccine combining transgenic human lymphotactin with interleukin-2 in patients with advanced or refractory neuroblastoma. Blood 2003; 101:1718-26. [PMID: 12406881 DOI: 10.1182/blood-2002-08-2493] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In murine models, transgenic chemokine-cytokine tumor vaccines overcome many of the limitations of single-agent immunotherapy by producing the sequence of T-cell attraction followed by proliferation. The safety and immunologic effects of this approach in humans were tested in 21 patients with relapsed or refractory neuroblastoma. They received up to 8 subcutaneous injections of a vaccine combining lymphotactin (Lptn)- and interleukin-2 (IL-2)-secreting allogeneic neuroblastoma cells in a dose-escalating scheme. Severe adverse reactions were limited to reversible panniculitis in 5 patients and bone pain in 1 patient. Injection-site biopsies revealed increased cellularity caused by infiltration of CD4+ and CD8+ lymphocytes, eosinophils, and Langerhans cells. Systemically, the vaccine produced a 2-fold (P =.035) expansion of CD4+ T cells, a 3.5-fold (P =.039) expansion of natural killer (NK) cells, a 2.1-fold (P =.014) expansion of eosinophils, and a 1.6-fold (P =.049) increase in serum IL-5. When restimulated in vitro by the immunizing cell line, T cells collected after vaccination showed a 2.3-fold increase (P =.02) of T-helper (TH2)-type CD3+IL-4+ cells. Supernatant collected from restimulated cells showed increased amounts of IL-4 (11.4-fold; P =.021) and IL-5 (8.7-fold; P =.002). Six patients had significant increases in NK cytolytic activity. Fifteen patients made immunoglobulin G (IgG) antibodies that bound to the immunizing cell line. Measurable tumor responses included complete remission in 2 patients and partial response in 1 patient. Hence, allogeneic tumor cell vaccines combining transgenic Lptn with IL-2 appear to have little toxicity in humans and can induce an antitumor immune response.
Collapse
MESH Headings
- Adolescent
- CD4 Lymphocyte Count
- CD4-Positive T-Lymphocytes/immunology
- Cancer Vaccines/adverse effects
- Cancer Vaccines/therapeutic use
- Chemokines, C
- Child
- Child, Preschool
- Cytokines/blood
- DNA, Complementary/genetics
- Female
- Humans
- Hypersensitivity, Delayed/etiology
- Immunization Schedule
- Immunoglobulin G/biosynthesis
- Immunoglobulin G/immunology
- Immunophenotyping
- Infant
- Injections, Subcutaneous
- Interleukin-2/administration & dosage
- Interleukin-2/genetics
- Interleukin-2/metabolism
- Interleukin-2/therapeutic use
- Killer Cells, Natural/immunology
- Lymphokines/administration & dosage
- Lymphokines/genetics
- Lymphokines/metabolism
- Lymphokines/therapeutic use
- Male
- Neuroblastoma/pathology
- Neuroblastoma/therapy
- Panniculitis/etiology
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/metabolism
- Recombinant Fusion Proteins/therapeutic use
- Remission Induction
- Salvage Therapy
- Sialoglycoproteins/administration & dosage
- Sialoglycoproteins/genetics
- Sialoglycoproteins/metabolism
- Sialoglycoproteins/therapeutic use
- Skin/pathology
- Th2 Cells/immunology
- Transduction, Genetic
- Treatment Outcome
- Tumor Cells, Cultured/drug effects
- Tumor Cells, Cultured/metabolism
- Tumor Cells, Cultured/radiation effects
- Tumor Cells, Cultured/transplantation
Collapse
Affiliation(s)
- Raphaël F Rousseau
- Center for Cell and Gene Therapy, Texas Children's Cancer Center, and the Department of Pathology, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
This is a brief discussion on the progress of gene therapy and the limitations of present-day gene therapy clinical trials based on a review of 464 human trial protocols from the U.S. National Institutes of Health (NIH) and the U.S. Federal Drug Administration (FDA). The paper also discusses an aspect of the gene therapy research of the author, who, together with colleagues, conducted the first gene therapy clinical trial in Korea in 1995.
Collapse
Affiliation(s)
- Dae Seog Heo
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University Medical College, Korea
| |
Collapse
|
33
|
Kawai K, Tani K, Asano S, Akaza H. Ex vivo gene therapy using granulocyte-macrophage colony-stimulating factor-transduced tumor vaccines. MOLECULAR UROLOGY 2002; 4:43-6. [PMID: 12006240 DOI: 10.1089/10915360050138567] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
There is no standard effective therapy for metastatic renal-cell carcinoma (RCC) or prostate cancer. Both of these cancers may be immunogenic, so therapy targeted to a tumor-associated antigen may be effective. Transduction of the gene encoding granulocyte-macrophage colony-stimulating factor has shown promise in preclinical studies, and clinical trials are in their early stages. Both autologous cancer cells and partially HLA-matched allogenic cells are being studied. No dose-limiting side effects have been observed, and a few patients have had transient objective tumor regressions. Further trials with more frequent and, probably, longer immunization schedules are needed to define efficacy.
Collapse
Affiliation(s)
- K Kawai
- Department of Urology, Institute of Clinical Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| | | | | | | |
Collapse
|
34
|
Abstract
The concept of immunotherapy of cancer is more than a century old, but only recently have molecularly defined therapeutic approaches been developed. In this review, we focus on the most promising approach, active therapeutic vaccination. The identification of tumour antigens can now be accelerated by methods allowing the amplification of gene products selectively or preferentially transcribed in the tumour. However, determining the potential immunogenicity of such gene products remains a demanding task, since major histocompatibility complex (MHC) restriction of T cells implies that for any newly defined antigen, immunogenicity will have to be defined for any individual MHC haplotype. Tumour-derived peptides eluted from MHC molecules of tumour tissue are also a promising source of antigen. Tumour antigens are mostly of weak immunogenicity, because the vast majority are tumour-associated differentiation antigens already 'seen' by the patient's immune system. Effective therapeutic vaccination will thus require adjuvant support, possibly by new approaches to immunomodulation such as bispecific antibodies or antibody-cytokine fusion proteins. Tumour-specific antigens, which could be a more potent target for immunotherapy, mostly arise by point mutations and have the disadvantage of being not only tumour-specific, but also individual-specific. Therapeutic vaccination will probably focus on defined antigens offered as protein, peptide or nucleic acid. Irrespective of the form in which the antigen is applied, emphasis will be given to the activation of dendritic cells as professional antigen presenters. Dendritic cells may be loaded in vitro with antigen, or, alternatively, initiation of an immune response may be approached in vivo by vaccination with RNA or DNA, given as such or packed into attenuated bacteria. The importance of activation of T helper cells has only recently been taken into account in cancer vaccination. Activation of cytotoxic T cells is facilitated by the provision of T helper cell-derived cytokines. T helper cell-dependent recruitment of elements of non-adaptive defence, such as leucocytes, natural killer cells and monocytes, is of particular importance when the tumour has lost MHC class I expression. Barriers to successful therapeutic vaccination include: (i) the escape mechanisms developed by tumour cells in response to immune attack; (ii) tolerance or anergy of the evoked immune response; (iii) the theoretical possibility of provoking an autoimmune reaction by vaccination against tumour-associated antigens; and (iv) the advanced age of many patients, implying reduced responsiveness of the senescent immune system.
Collapse
Affiliation(s)
- S Matzku
- Department of Oncology, Biomedical Research, Merck KGaA, Darmstadt, Germany
| | | |
Collapse
|
35
|
Harrington KJ, Melcher AA, Bateman AR, Ahmed A, Vile RG. Cancer gene therapy: Part 2. Candidate transgenes and their clinical development. Clin Oncol (R Coll Radiol) 2002; 14:148-69. [PMID: 12069125 DOI: 10.1053/clon.2001.0004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Kevin J Harrington
- CRC Centre for Cell and Molecular Biology, Institute for Cancer Research, London, UK.
| | | | | | | | | |
Collapse
|
36
|
Kronenberger K, Dieckmann A, Selmayr M, Strehl J, Wahl U, Lindhofer H, Kraal G, Mocikat R. Impact of the lymphoma idiotype on in vivo tumor protection in a vaccination model based on targeting antigens to antigen-presenting cells. Blood 2002; 99:1327-31. [PMID: 11830483 DOI: 10.1182/blood.v99.4.1327] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Trioma cell vaccination is a potent new immunologic approach for the therapy of malignant B-cell lymphoma. It is based on targeting tumor antigens to internalizing receptors on antigen-presenting cells (APCs). Tumor cells are fused to an APC-specific hybridoma, where they are converted to trioma cells that include potentially all lymphoma-derived antigens and that express the APC-binding arm. In this study, the mechanisms of trioma-mediated tumor immunity in immunocompetent mice were dissected, and it was shown in this model system that humoral anti-idiotypic immunity is indeed detectable after idiotype-specific immunization but that it does not reflect the degree of tumor protection obtained in vivo. Immunization against the idiotype alone was not sufficient for efficient tumor rejection in vivo. Targeting tumor antigens to APCs is only successful in terms of inducing tumor protection when designed as a polyvalent vaccination protocol.
Collapse
MESH Headings
- Animals
- Antibodies, Anti-Idiotypic/immunology
- Antibody Formation/immunology
- Antigen-Presenting Cells/cytology
- Antigen-Presenting Cells/immunology
- Antigens, Neoplasm/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/standards
- Female
- Hybridomas/cytology
- Hybridomas/immunology
- Hybridomas/transplantation
- Lymphoma, B-Cell/pathology
- Lymphoma, B-Cell/prevention & control
- Lymphoma, B-Cell/therapy
- Mice
- Mice, Inbred BALB C
- Rats
- Receptors, Fc/immunology
- Survival Rate
- Tumor Cells, Cultured/cytology
- Tumor Cells, Cultured/immunology
- Tumor Cells, Cultured/transplantation
Collapse
|
37
|
Hicks C, Keoshkerian E, French R, Lindeman R. Adenoviral mediated interleukin-2 gene transfer to human long-term bone marrow stromal cultures. Cancer Biother Radiopharm 2001; 16:537-43. [PMID: 11789030 DOI: 10.1089/10849780152752137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Human long-term bone marrow stromal cultures (LTBMC) have been transduced to express the immunostimulatory cytokine interleukin-2 (IL-2). LTBMC were established from normal bone marrow and characterized by gross morphology, immunohistochemistry, immunophenotyping, and immunofluorescent staining. A stromal marrow cell (SMC) population expressing the markers CD68, CD29, CD13, and CD54 was identified. These cells were successfully transduced with an adenoviral vector containing the human IL-2 gene. Production of IL-2 was demonstrated for at least 13 days post-transduction and was maximal on day 10 at 19,350 pg (or 38 IU) of IL-2/10(6) cells/24 h. IL-2 expressed by the transduced SMC was demonstrated to be biologically active, resulting in the proliferation of autologous T lymphocytes in a mixed lymphocyte reaction. Autologous human LTBMC can be obtained and manipulated with relative ease and may be a useful vehicle for sustained local cytokine delivery.
Collapse
Affiliation(s)
- C Hicks
- Department of Haematology, Prince of Wales Hospital, High Street, Randwick, NSW, 2031, Sydney, Australia.
| | | | | | | |
Collapse
|
38
|
Kianmanesh A, Hackett NR, Lee JM, Kikuchi T, Korst RJ, Crystal RG. Intratumoral administration of low doses of an adenovirus vector encoding tumor necrosis factor alpha together with naive dendritic cells elicits significant suppression of tumor growth without toxicity. Hum Gene Ther 2001; 12:2035-49. [PMID: 11747595 DOI: 10.1089/10430340152677395] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Although tumor necrosis factor alpha (TNF-alpha) is a potent cytokine with a myriad of innate immune antitumor properties, systemic administration of TNF-alpha is associated with significant toxicity, limiting the use of the TNF-alpha protein as an antitumor therapeutic. On the basis of the knowledge that dendritic cells (DCs) play a central role in initiating antitumor adaptive immune responses, we hypothesized that intratumoral administration of low doses of an adenovirus encoding TNF-alpha (AdTNF-alpha) together with syngeneic DCs would act synergistically to suppress preexisting tumors. As a model, four different tumor cell lines, all resistant in vitro to the TNF-alpha protein, were implanted in syngeneic mice, and established tumors received intratumor AdTNF-alpha alone or in combination with DCs. At high doses (10(9) PFU), AdTNF-alpha alone suppressed tumor growth, but was associated with systemic toxicity. A 100-fold lower AdTNF-alpha concentration (10(7) PFU) or high doses of the control vector AdNull had no systemic toxicity, but also minimal suppression of tumor growth. In contrast, local administration of the low dose (10(7) PFU) of AdTNF-alpha in combination with syngeneic DCs (AdTNF-alpha + DCs) elicited marked tumor suppression without toxicity. Administration of AdTNF-alpha + DCs into tumors elicited tumor-specific cytotoxic T cells and protected animals against subsequent challenge with the same tumor, suggesting that AdTNF-alpha + DC therapy induced tumor-specific adaptive immune host responses. Consistent with this concept, studies with syngeneic knockout mice showed that MHC class I molecules on DCs as well as CD8(+) T cells were necessary for the antitumor effect of intratumor AdTNF-alpha + DCs. These data demonstrate that the combination of intratumoral administration of the TNF-alpha cDNA together with naive DCs can evoke tumor suppression without systemic toxicity, providing a new paradigm for the use of TNF-alpha as antitumor therapy.
Collapse
Affiliation(s)
- A Kianmanesh
- Division of Pulmonary and Critical Care Medicine, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
39
|
Abstract
Cancer is one of the leading causes of death in Western society. Despite improvements in screening, diagnosis and treatment of cancer, many patients ultimately succumb to their disease. Advances in molecular biology and our increased understanding of how the immune system functions have led to an intense interest in the development of cancer vaccines.
Collapse
Affiliation(s)
- A C Armstrong
- CRC Department of Medical Oncology, University of Manchester & Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Manchester M20 4BX, UK
| | | |
Collapse
|
40
|
Harrington KJ, Spitzweg C, Bateman AR, Morris JC, Vile RG. Gene therapy for prostate cancer: current status and future prospects. J Urol 2001; 166:1220-33. [PMID: 11547047 DOI: 10.1016/s0022-5347(05)65742-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE Locally advanced, relapsed and metastatic prostate cancer has a dismal prognosis with conventional therapies offering no more than palliation. In recent years advances achieved in understanding the molecular biology of cancer have afforded clinicians and scientists the opportunity to develop a range of novel genetic therapies for this disease. MATERIALS AND METHODS We performed a detailed review of published reports of gene therapy for prostate cancer. Particular emphasis was placed on recent developments in the arena of nonviral (plasmid DNA, DNA coated gold particles, liposomes and polymer DNA complexes) and viral (adenovirus, retrovirus, adeno-associated virus, herpes virus and pox virus) vectors. Therapeutic strategies were categorized as corrective, cytoreductive and immunomodulatory gene therapy for the purpose of data analysis and comparison. RESULTS Locoregional administration of nonviral and viral vectors can yield impressive local gene expression and therapeutic effects but to our knowledge no efficient systemically delivered vector is available to date. Corrective gene therapy to restore normal patterns of tumor suppressor gene (p53, Rb, p21 and p16) expression or negate the effect of mutated tumor promoting oncogenes (ras, myc, erbB2 and bcl-2) have efficacy in animal models but this approach suffers from the fact that each cancer cell must be targeted. A wide variety of cytoreductive strategies are under development, including suicide, anti-angiogenic, radioisotopic and pro-apoptotic gene therapies. Each approach has strengths and weaknesses, and may best be suited for use in combination. Immunomodulatory gene therapy seeks to generate an effective local immune response that translates to systemic antitumor activity. Currently most studies involve immunostimulatory cytokine genes, such as granulocyte-macrophage colony-stimulating factor, or interleukin-2 or 12. CONCLUSIONS Various therapeutic genes have proved activity against prostate cancer in vitro and in vivo. However, the chief challenge facing clinical gene therapy strategies is the lack of efficient gene delivery by local and systemic routes. For the foreseeable future vector development may remain a major focus of ongoing research. Despite this caveat it is anticipated that gene therapy approaches may significantly contribute to the management of prostate cancer in the future.
Collapse
Affiliation(s)
- K J Harrington
- Molecular Medicine Program and Department of Endocrinology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | | | |
Collapse
|
41
|
|
42
|
de Wilt JH, Bout A, Eggermont AM, van Tiel ST, de Vries MW, ten Hagen TL, de Roos WK, Valerio D, van der Kaaden ME. Adenovirus-mediated interleukin 3 beta gene transfer by isolated limb perfusion inhibits growth of limb sarcoma in rats. Hum Gene Ther 2001; 12:489-502. [PMID: 11268282 DOI: 10.1089/104303401300042384] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cytokine gene transfer using (multiple) intratumoral injections can induce tumor regression in several animal models, but this administration technique limits the use for human gene therapy. In the present studies we describe tumor growth inhibition of established limb sarcomas after a single isolated limb perfusion (ILP) with recombinant adenoviral vectors harboring the rat IL-3 beta gene (IG.Ad.CMV.rIL-3 beta). In contrast, a single intratumoral injection or intravenous administration did not affect tumor growth. Dose-finding studies demonstrated a dose-dependent response with a loss of antitumor effect below 1 x 10(9) IU of IG.Ad.CMV.rIL-3 beta. Perfusions with adenoviral vectors bearing a weaker promoter (MLP promoter) driving the rIL-3 beta gene did not result in antitumor responses, suggesting that the rIL-3 beta-mediated antitumor effect depends on the amount of rIL-3 beta protein expressed by the infected cells. Furthermore, it was shown by direct comparison that ILP with IG.Ad.CMV.rIL-3 beta in the ROS-1 osteosarcoma model is at least as efficient as the established therapy with the combination of TNF-alpha and melphalan. Treatment with IG.Ad.CMV.rIL-3 beta induced a transient dose-dependent leukocytosis accompanied by an increase in peripheral blood levels of histamine. Leukocyte infiltrations were also histopathologically demonstrated in tumors after perfusion. These results demonstrate that ILP with recombinant adenoviral vectors carrying the IL-3 beta transgene inhibits tumor growth in rats and suggest that cytokine gene therapy using this administration technique might be beneficial for clinical cancer treatment.
Collapse
Affiliation(s)
- J H de Wilt
- Department of Surgical Oncology, University Hospital Rotterdam Dijkzigt/Daniel de Hoed Cancer Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ozbek S, Peters M, Breuhahn K, Mann A, Blessing M, Fischer M, Schirmacher P, Mackiewicz A, Rose-John S. The designer cytokine hyper-IL-6 mediates growth inhibition and GM-CSF-dependent rejection of B16 melanoma cells. Oncogene 2001; 20:972-9. [PMID: 11314032 DOI: 10.1038/sj.onc.1204180] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2000] [Revised: 12/05/2000] [Accepted: 12/14/2000] [Indexed: 11/09/2022]
Abstract
The low immunogenic B16 melanoma cell line was transfected with a mammalian expression vector containing the complementary DNA for a sIL-6R/IL-6 fusion protein, termed Hyper-IL-6 (H-IL-6), which was shown to have biological activities at 100-1000-fold lower concentrations than IL-6 in combination with sIL-6R. The secreted p84 glycoprotein was detected in the supernatant of transfected cells and was fully active on BAF3/gp130 cells, which respond to IL-6/sIL-6R but not to IL-6 alone. Administration of recombinant H-IL-6 to C57BL/6 mice resulted in a prolonged acute phase protein gene expression indicating long systemic persistence of the fusion protein. Transfected B16 cells (B16/H-IL6 cells) showed morphological alterations in combination with a dramatic growth inhibition in vitro. Subcutaneous injection in C57BL/6 mice resulted in an almost complete rejection of B16/H-IL6 cells. This effect was partially abolished in FVB/BL/6 mice transgenic for a GM-CSF receptor antagonist, indicating a GM-CSF-dependent rejection of H-IL-6 transfected B16 cells. These results demonstrate that the anti-tumor effect of cytokines like IL-6 which are secreted by transfected melanoma cells at least in part depends on GM-CSF activity.
Collapse
Affiliation(s)
- S Ozbek
- Department of Medicine, Section of Pathophysiology, University of Mainz, Obere Zahlbacher Strasse 63, D-55101 Mainz, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Nagai H, Hara I, Horikawa T, Oka M, Kamidono S, Ichihashi M. Elimination of CD4(+) T cells enhances anti-tumor effect of locally secreted interleukin-12 on B16 mouse melanoma and induces vitiligo-like coat color alteration. J Invest Dermatol 2000; 115:1059-64. [PMID: 11121142 DOI: 10.1046/j.1523-1747.2000.00156.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
CD4(+) T cells have been reported to suppress immunity against cancer in certain animal models. In this study, we investigated the role of CD4(+) T cells in the anti-tumor immune response when interleukin-12-producing melanoma cells are inoculated in mice. We found that interleukin-12-transfected B16 melanoma showed retarded tumor growth in syngeneic mice; however, all the mice developed tumors eventually. In vivo depletion of CD4(+) T cells led to complete regression of B16/interleukin-12 tumors in 12 of 20 mice (60%). Immunohistochemical analyses revealed that a number of CD8(+) T cells accumulated in close proximity to the B16/interleukin-12 tumors in the CD4(+) T cell-depleted mice, whereas CD8(+) T cells were only scarcely observed at the periphery of the tumors in control immunocompetent mice. Furthermore, 10 of 20 mice treated with both B16/interleukin-12 inoculation and CD4(+) T cell depletion exhibited vitiligo-like coat color alteration. B16/interleukin-12 tumors completely regressed in all the mice with vitiligo. Histologic examination showed that CD8(+) lymphocytes accumulated around the hair bulbs of mice with vitiligo, but not in those without vitiligo. These results suggest that CD4(+) T cells have an inhibitory effect on tumor rejection by suppressing cytotoxic CD8(+) T cells in this melanoma loading model with local interleukin-12 secretion. To investigate the mechanism of enhanced anti-tumor effects by CD4(+) T cell depletion, we examined the T helper type 1/2 cytokine profile in the tumor draining lymph nodes of B16/interleukin-12-bearing mice with or without CD4(+) T cell depletion using the reverse transcription-polymerase chain reaction method. We found that CD4(+) T cell depletion eliminated T helper type 2 cells and resulted in a T helper type 1-dominant cytokine profile in tumor draining lymph nodes. We emphasize that this T helper type 1-dominant cytokine profile may generate further activated CD8(+) T cells against B16 melanoma cells, lead B16/interleukin-12 to regress, and result in the destruction of the melanocytes in hair bulbs due to cross-antigenicity between both cell types. This mouse model not only demonstrates the depletion of CD4(+) T cells as a useful strategy for cancer gene therapy with interleukin-12 but also provides a model for human melanoma-associated vitiligo.J Invest Dermatol 115:1059-1064 2000
Collapse
Affiliation(s)
- H Nagai
- Department of Dermatology, Kobe University School of Medicine, Kobe, Japan.
| | | | | | | | | | | |
Collapse
|
45
|
Thorsen F, Read TA, Lund-Johansen M, Tysnes BB, Bjerkvig R. Alginate-encapsulated producer cells: a potential new approach for the treatment of malignant brain tumors. Cell Transplant 2000; 9:773-83. [PMID: 11202564 DOI: 10.1177/096368970000900604] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
In recent years gene therapy has evolved as a new treatment for brain tumors, where genetically engineered cells can be used to deliver specific substances to target cells. However, clinical success has been limited due to insufficient gene transfer, lack of prolonged gene expression, and immunorejection of producer cells. These obstacles may be overcome by encapsulating producer cells into immunoisolating substances such as alginate. This may provide a stable in situ delivery system of specific proteins, which can interfere with tumor growth and differentiation. This article represents a fundamental study describing the in vitro and the in vivo behavior of alginate-encapsulated producer cells. The viability and cell cycle distribution of encapsulated NIH 3T3 cells was studied by confocal laser scanning microscopy (CLSM) and by flow cytometry. The CLSM study showed a high viability of the encapsulated NIH 3T3 cells during 9 weeks in culture. The flow cytometric analysis revealed a change in cellular ploidy after 1 week in culture, with normalization in ploidy after 3 and 9 weeks. The production of the bacterial E. coli beta-galactosidase in alginate-encapsulated BT4CnVlacZ cells was studied by x-gal staining, and the cells expressed prolonged beta-galactosidase activity. H528 hybridoma cells producing monoclonal antibodies (mAbs) against the human epidermal growth factor receptor (EGFR) were encapsulated in alginate, and the mAb release was determined. The release of mAbs stabilized around 400 ng/ml/h after 12 days in vitro. To actually demonstrate that alginate-encapsulated H528 cells potentially inhibit a heterogeneous glioma cell population, cell migration from human GaMg glioma spheroids was studied during stimulation with EGF in the presence of encapsulated H528 cells. The migration in vitro was totally inhibited in the presence of H528 encapsulated cells. Alginate beads with H528 cells were also implanted into rat brains, and after 9 weeks the distribution of mAbs within the brain was studied by immunohistochemistry. It is shown that the alginate entrapped H528 cells produce mAbs inside the brain for prolonged periods and that the mAbs are distributed within all CSF compartments. Encapsulated producer cells represent a potential delivery system for specific proteins to brain tumors. Different producer cells may be encapsulated in alginate to target phenotypic features and microenvironmental factors, which may influence the progressive growth of brain tumors.
Collapse
Affiliation(s)
- F Thorsen
- Department of Anatomy and Cell Biology, University of Bergen, Norway.
| | | | | | | | | |
Collapse
|
46
|
Cao L, Kulmburg P, Veelken H, Mackensen A, Mézes B, Lindemann A, Mertelsmann R, Rosenthal FM. Cytokine gene transfer in cancer therapy. Stem Cells 2000; 16 Suppl 1:251-60. [PMID: 11012169 DOI: 10.1002/stem.5530160831] [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: 11/06/2022]
Abstract
New strategies based on gene transfer technology are employed in cancer therapy. Cytokines are polypeptides involved in immunity and inflammation, and essentially control the magnitude of the immune response. Genetically modified tumor cells releasing various cytokines have been shown to enhance tumor immunogenicity and to induce the regression of preexisting tumors. In some instances, immunological memory has been generated to resist the subsequent challenge with unmodified, parental tumor cells. Cytokine gene transfer into antitumor effector cells, as well as antigen presenting cells, is also being investigated to augment antitumor immune responses.
Collapse
Affiliation(s)
- L Cao
- Department of Internal Medicine I (Hematology/Oncology), University Medical Center Freiburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Jiang XP, Yang DC, Elliott RL, Head JF. Vaccination with a mixed vaccine of autogenous and allogeneic breast cancer cells and tumor associated antigens CA15-3, CEA and CA125--results in immune and clinical responses in breast cancer patients. Cancer Biother Radiopharm 2000; 15:495-505. [PMID: 11155821 DOI: 10.1089/cbr.2000.15.495] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In breast cancer there is often overexpression of the breast cancer antigen CA15-3, the carcinoembryonic antigen (CEA) and the ovarian cancer antigen CA125, which makes them potential target antigens for immunotherapy. In this study, we used a multi-antigen vaccine, which included the following antigens: autologous breast cancer cells (AUTOC), allogeneic breast cancer MCF-7 cells (ALLOC), and the tumor associated antigens CA15-3, CEA and CA125, plus low doses of granulocyte/macrophage-colony-stimulating factor (GM-CSF) and interleukin 2 (IL-2). Forty-two breast cancer patients received weekly subcutaneous vaccination at the 1st, 2nd, 3rd, 7th, 11th and 15th weeks. Their lymphocyte proliferative responses to AUTOC, ALLOC, CA15-3, CEA and CA125 were tested in lymphocyte blastogenesis assays (LBA) before and after vaccination. The disease stage and serum CA15-3, CEA and CA125 concentrations were also determined pre- and post-vaccination. We found that the vaccine was safe, and the only major side effects were swelling at the site of injection, muscle pain, and weakness or fatigue. The vaccine induced a significant increase in post-vaccination lymphocyte proliferative responses to AUTOC, CA15-3, CEA and CA125 but not ALLOC, compared to pre-vaccination (p < 0.05, p < 0.01, p < 0.05, p < 0.01 and p > 0.05, respectively, a paired t Test). Computed tomography (CT), ultrasound or bone scan showed evidence of disease improvement in 2 (12%) patients after vaccination. Hepatic metastases were reduced in size and number and some actually disappeared one patient. Metastatic disease in the L5 vertebra and the skull decreased in size and some osteolytic sites completely healed in a second patient. In addition, 7 patients (44%) had stable disease and 7 patients (44%) had disease progression. We did not find vaccination significantly reduced serum tumor markers CA15-3, CEA and CA125 of these breast cancer patients. These results suggest that the vaccine mixture of autologous and allogeneic breast cancer cells and tumor associated antigens plus GM-CSF and IL-2 can be administered safely to breast cancer patients and there is evidence for improved immunity and clinical efficacy.
Collapse
Affiliation(s)
- X P Jiang
- Mastology Research Institute, Elliott Mastology Center, Baton Rouge, LA, USA
| | | | | | | |
Collapse
|
48
|
Trentin L, Perin A, Siviero M, Piazza F, Facco M, Gurrieri C, Galvan S, Adami F, Agostini C, Pizzolo G, Zambello R, Semenzato G. B7 costimulatory molecules from malignant cells in patients with B-cell chronic lymphoproliferative disorders trigger T-cell proliferation. Cancer 2000. [DOI: 10.1002/1097-0142(20000915)89:6<1259::aid-cncr10>3.0.co;2-g] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
49
|
Nawrocki S, Murawa P, Malicki J, Kapcinska M, Gryska K, Izycki D, Kaczmarek A, Laciak M, Czapczyk A, Karczewska A, Rose-John S, Mackiewicz A. Genetically modified tumour vaccines (GMTV) in melanoma clinical trials. Immunol Lett 2000; 74:81-6. [PMID: 10996632 DOI: 10.1016/s0165-2478(00)00253-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Since melanoma is a model immunogenic malignancy incurable in the disseminated phase of its natural course different immunotherapeutic approaches are tested in clinical trials. A number of tumour vaccines genetically modified (GMTV), with various immunostimulatory factors, are tested in phase I/II clinical trials. These factors include cytokines, tumour antigens (TA), costimulatory molecules or HLA antigens. We have designed a novel, mixed auto/allogeneic cellular melanoma vaccine modified with the IL-6 and the sIL-6R genes. Preclinical studies in a mouse model demonstrated that the IL-6/sIL-6R based vaccine is able to elicit efficient anti-tumour responses, mediated by CD8+ and NK cells, which resulted in inhibition of the tumour growth, metastases formation and prolonged survival of the animals treated. Irradiation of vaccine cells does not only lead to their sterilisation but also causes increased secretion of exogenous IL-6 and sIL-6R. Since January 1996 we have vaccinated more than one hundred metastatic melanoma patients. Promising clinical results (22% CR+PR, 32% SD) and the evidence of immune responses in the vaccinated patients have prompted us to design a phase III clinical trial which is to be open in 2000.
Collapse
Affiliation(s)
- S Nawrocki
- Department of Radiation Oncology, Great Poland Cancer Centre, ul. Garbary 15, 61-866, Poznań, Poland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Curiel-Lewandrowski C, Demierre MF. Advances in specific immunotherapy of malignant melanoma. J Am Acad Dermatol 2000; 43:167-85; quiz 186-8. [PMID: 10906637 DOI: 10.1067/mjd.2000.104513] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
UNLABELLED Management of malignant melanoma continues to present a challenge to dermatologists, particularly in advanced cases. In light of the steady increase in the worldwide incidence and mortality rates for melanoma, better understanding of the immune mechanisms regulating melanoma progression and interaction with the host's immune system seems eminently important. New studies on the role of immune mechanisms in the pathogenesis and clinical course of melanoma have recently been published. We review the immune mechanisms involved in tumor progression and ways in which these mechanisms may be applied toward immunotherapeutic management of malignant melanoma. LEARNING OBJECTIVE After the completion of this learning activity, participants should be familiar with (1) the immune mechanisms involved in host-tumor interaction and tumor rejection, (2) factors allowing the escape of melanoma cells from immune recognition, and (3) the current rationale for the different types of specific immunotherapy in melanoma. Better understanding of basic mechanisms in tumor immunology should raise awareness of future immunotherapeutic approaches in patients with melanoma, particularly in those who are at high risk of recurrence or who present with advanced disease.
Collapse
|