1
|
Nishinakamura H, Shinya S, Irie T, Sakihama S, Naito T, Watanabe K, Sugiyama D, Tamiya M, Yoshida T, Hase T, Yoshida T, Karube K, Koyama S, Nishikawa H. Coactivation of innate immune suppressive cells induces acquired resistance against combined TLR agonism and PD-1 blockade. Sci Transl Med 2025; 17:eadk3160. [PMID: 39937883 DOI: 10.1126/scitranslmed.adk3160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/02/2024] [Accepted: 01/03/2025] [Indexed: 02/14/2025]
Abstract
Immune checkpoint blockade therapy has been successfully applied in clinical settings as a standard therapy for many cancer types, but its clinical efficacy is restricted to patients with immunologically hot tumors. Various strategies to modify the tumor microenvironment (TME), such as Toll-like receptor (TLR) agonists that can stimulate innate immunity, have been explored but have not been successful. Here, we show a mechanism of acquired resistance to combination treatment consisting of an agonist for multiple TLRs, OK-432 (Picibanil), and programmed cell death protein 1 (PD-1) blockade. Adding the TLR agonist failed to convert the TME from immunogenically cold to hot and did not augment antitumor immunity, particularly CD8+ T cell responses, in multiple animal models. The failure was attributed to the coactivation of innate suppressive cells, such as polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) expressing CXCR2, through high CXCL1 production by macrophages in the TME upon OK-432 treatment. A triple combination treatment with OK-432, PD-1 blockade, and a CXCR2 neutralizing antibody overcame the resistance induced by PMN-MDSCs, resulting in a stronger antitumor effect than that of any dual combinations or single treatments. The accumulation of PMN-MDSCs was similarly observed in the pleural effusions of patients with lung cancer after OK-432 administration. We propose that successful combination cancer immunotherapy intended to stimulate innate antitumor immunity requires modulation of unwanted activation of innate immune suppressive cells, including PMN-MDSCs.
Collapse
Affiliation(s)
- Hitomi Nishinakamura
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center Japan, Tokyo, 104-0045/Chiba 277-8577, Japan
| | - Sayoko Shinya
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center Japan, Tokyo, 104-0045/Chiba 277-8577, Japan
- Discovery and Research, Ono Pharmaceutical Co. Ltd., Osaka, 618-8585, Japan
| | - Takuma Irie
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center Japan, Tokyo, 104-0045/Chiba 277-8577, Japan
| | - Shugo Sakihama
- Laboratory of Hemato-Immunology, Graduate School of Health Sciences, University of the Ryukyus, Nishihara, 903-0125, Japan
| | - Takeo Naito
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center Japan, Tokyo, 104-0045/Chiba 277-8577, Japan
| | - Keisuke Watanabe
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center Japan, Tokyo, 104-0045/Chiba 277-8577, Japan
| | - Daisuke Sugiyama
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Motohiro Tamiya
- Respiratory Medicine, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Tatsuya Yoshida
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Tetsunari Hase
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Takao Yoshida
- Discovery and Research, Ono Pharmaceutical Co. Ltd., Osaka, 618-8585, Japan
| | - Kennosuke Karube
- Department of Pathology and Laboratory Medicine, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shohei Koyama
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center Japan, Tokyo, 104-0045/Chiba 277-8577, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center Japan, Tokyo, 104-0045/Chiba 277-8577, Japan
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Division of Cancer Immune Multicellular System Regulation, Center for Cancer Immunotherapy and Immunobiology (CCII), Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
2
|
Poschel DB, Kehinde-Ige M, Klement JD, Yang D, Merting AD, Savage NM, Shi H, Liu K. IRF8 Regulates Intrinsic Ferroptosis through Repressing p53 Expression to Maintain Tumor Cell Sensitivity to Cytotoxic T Lymphocytes. Cells 2023; 12:310. [PMID: 36672246 PMCID: PMC9856547 DOI: 10.3390/cells12020310] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/07/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
Ferroptosis has emerged as a cytotoxic T lymphocyte (CTL)-induced tumor cell death pathway. The regulation of tumor cell sensitivity to ferroptosis is incompletely understood. Here, we report that interferon regulatory factor 8 (IRF8) functions as a regulator of tumor cell intrinsic ferroptosis. Genome-wide gene expression profiling identified the ferroptosis pathway as an IRF8-regulated pathway in tumor cells. IRF8.KO tumor cells acquire resistance to intrinsic ferroptosis induction and IRF8-deficient tumor cells also exhibit decreased ferroptosis in response to tumor-specific CTLs. Irf8 deletion increased p53 expression in tumor cells and knocking out p53 in IRF8.KO tumor cells restored tumor cell sensitivity to intrinsic ferroptosis induction. Furthermore, IRF8.KO tumor cells grew significantly faster than WT tumor cells in immune-competent mice. To restore IRF8 expression in tumor cells, we designed and synthesized codon usage-optimized IRF8-encoding DNA to generate IRF8-encoding plasmid NTC9385R-mIRF8. Restoring IRF8 expression via a lipid nanoparticle-encapsulated NTC9385R-mIRF8 plasmid therapy suppressed established tumor growth in vivo. In human cancer patients, nivolumab responders have a significantly higher IRF8 expression level in their tumor cells as compared to the non-responders. Our data determine that IRF8 represses p53 expression to maintain tumor cell sensitivity to intrinsic ferroptosis.
Collapse
Affiliation(s)
- Dakota B. Poschel
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
- Georgia Cancer Center, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Mercy Kehinde-Ige
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
- Georgia Cancer Center, Augusta, GA 30912, USA
| | - John D. Klement
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
- Georgia Cancer Center, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Dafeng Yang
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
- Georgia Cancer Center, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Alyssa D. Merting
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
- Georgia Cancer Center, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Natasha M. Savage
- Department of Pathology, Medical College of Georgia, Augusta, GA 30912, USA
| | - Huidong Shi
- Georgia Cancer Center, Augusta, GA 30912, USA
| | - Kebin Liu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
- Georgia Cancer Center, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| |
Collapse
|
3
|
Al Subeh ZY, Poschel DB, Redd PS, Klement JD, Merting AD, Yang D, Mehta M, Shi H, Colson YL, Oberlies NH, Pearce CJ, Colby AH, Grinstaff MW, Liu K. Lipid Nanoparticle Delivery of Fas Plasmid Restores Fas Expression to Suppress Melanoma Growth In Vivo. ACS NANO 2022; 16:12695-12710. [PMID: 35939651 PMCID: PMC9721370 DOI: 10.1021/acsnano.2c04420] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Fas ligand (FasL), expressed on the surface of activated cytotoxic T lymphocytes (CTLs), is the physiological ligand for the cell surface death receptor, Fas. The Fas-FasL engagement initiates diverse signaling pathways, including the extrinsic cell death signaling pathway, which is one of the effector mechanisms that CTLs use to kill tumor cells. Emerging clinical and experimental data indicate that Fas is essential for the efficacy of CAR-T cell immunotherapy. Furthermore, loss of Fas expression is a hallmark of human melanoma. We hypothesize that restoring Fas expression in tumor cells reverses human melanoma resistance to T cell cytotoxicity. DNA hypermethylation, at the FAS promoter, down-regulates FAS expression and confers melanoma cell resistance to FasL-induced cell death. Forced expression of Fas in tumor cells overcomes melanoma resistance to FasL-induced cell death in vitro. Lipid nanoparticle-encapsulated mouse Fas-encoding plasmid therapy eliminates Fas+ tumor cells and suppresses established melanoma growth in immune-competent syngeneic mice. Similarly, lipid nanoparticle-encapsulated human FAS-encoding plasmid (hCOFAS01) therapy significantly increases Fas protein levels on tumor cells of human melanoma patient-derived xenograft (PDX) and suppresses the established human melanoma PDX growth in humanized NSG mice. In human melanoma patients, FasL is expressed in activated and exhausted T cells, Fas mRNA level positively correlates with melanoma patient survival, and nivolumab immunotherapy increases FAS expression in tumor cells. Our data demonstrate that hCOFAS01 is an effective immunotherapeutic agent for human melanoma therapy with dual efficacy in increasing tumor cell FAS expression and in enhancing CTL tumor infiltration.
Collapse
Affiliation(s)
- Zeinab Y. Al Subeh
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Dakota B. Poschel
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
- Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Priscilla S. Redd
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
- Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - John D. Klement
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
- Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Alyssa D. Merting
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
- Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Dafeng Yang
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
- Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Megh Mehta
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
| | - Huidong Shi
- Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912, USA
| | - Yolonda L. Colson
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114. USA
| | - Nicholas H. Oberlies
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC 27402, USA
| | | | - Aaron H. Colby
- Ionic Pharmaceuticals, Brookline, MA 02445, USA
- Department of Biomedical Engineering, Boston University, Boston, MA 02215. USA
| | - Mark W. Grinstaff
- Ionic Pharmaceuticals, Brookline, MA 02445, USA
- Department of Biomedical Engineering, Boston University, Boston, MA 02215. USA
| | - Kebin Liu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
- Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| |
Collapse
|
4
|
Aaes TL, Vandenabeele P. The intrinsic immunogenic properties of cancer cell lines, immunogenic cell death, and how these influence host antitumor immune responses. Cell Death Differ 2021; 28:843-860. [PMID: 33214663 PMCID: PMC7937679 DOI: 10.1038/s41418-020-00658-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 01/30/2023] Open
Abstract
Modern cancer therapies often involve the combination of tumor-directed cytotoxic strategies and generation of a host antitumor immune response. The latter is unleashed by immunotherapies that activate the immune system generating a more immunostimulatory tumor microenvironment and a stronger tumor antigen-specific immune response. Studying the interaction between antitumor cytotoxic therapies, dying cancer cells, and the innate and adaptive immune system requires appropriate experimental tumor models in mice. In this review, we discuss the immunostimulatory and immunosuppressive properties of cancer cell lines commonly used in immunogenic cell death (ICD) studies being apoptosis or necroptosis. We will especially focus on the antigenic component of immunogenicity. While in several cancer cell lines the epitopes of endogenously expressed tumor antigens are known, these intrinsic epitopes are rarely determined in experimental apoptotic or necroptotic ICD settings. Instead by far the most ICD research studies investigate the antigenic response against exogenously expressed model antigens such as ovalbumin or retroviral epitopes (e.g., AH1). In this review, we will argue that the immune response against endogenous tumor antigens and the immunopeptidome profile of cancer cell lines affect the eventual biological readouts in the typical prophylactic tumor vaccination type of experiments used in ICD research, and we will propose additional methods involving immunopeptidome profiling, major histocompatibility complex molecule expression, and identification of tumor-infiltrating immune cells to document intrinsic immunogenicity following different cell death modalities.
Collapse
Affiliation(s)
- Tania Løve Aaes
- grid.11486.3a0000000104788040Unit for Cell Clearance in Health and Disease, VIB Center for Inflammation Research, Ghent, Belgium ,grid.5342.00000 0001 2069 7798Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium ,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Peter Vandenabeele
- grid.5342.00000 0001 2069 7798Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium ,Cancer Research Institute Ghent (CRIG), Ghent, Belgium ,grid.11486.3a0000000104788040Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium
| |
Collapse
|
5
|
DeLeo AB, Appella E. The p53 Saga: Early Steps in the Development of Tumor Immunotherapy. THE JOURNAL OF IMMUNOLOGY 2021; 204:2321-2328. [PMID: 32312843 DOI: 10.4049/jimmunol.1901343] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/31/2019] [Indexed: 12/31/2022]
Abstract
This year marks the 40th anniversary of the initial identification of p53 as a transformation-related Ag, which was the result of our effort to identify an antigenically distinct tumor Ag of a chemically induced mouse tumor and develop a cancer vaccine. Many researchers at the time viewed this effort as folly. Since then, its characterization has progressed from being an attractive cancer vaccine candidate to recognition as a key player in regulating critical pathways controlling the cell cycle and oncogenesis. Advances in molecular immunology and oncology have enhanced the role of p53 in both fields. It is now apparent that p53 plays a critical role in controlling immune recognition and responses in normal tissues as well as the tumor microenvironment. Together with the advances in clinical implementation of p53-based cancer immunotherapy, they highlight the importance of p53 in many areas of basic and translational cancer research.
Collapse
Affiliation(s)
- Albert B DeLeo
- University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232; and
| | - Ettore Appella
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814
| |
Collapse
|
6
|
Aaes TL, Verschuere H, Kaczmarek A, Heyndrickx L, Wiernicki B, Delrue I, De Craene B, Taminau J, Delvaeye T, Bertrand MJM, Declercq W, Berx G, Krysko DV, Adjemian S, Vandenabeele P. Immunodominant AH1 Antigen-Deficient Necroptotic, but Not Apoptotic, Murine Cancer Cells Induce Antitumor Protection. THE JOURNAL OF IMMUNOLOGY 2020; 204:775-787. [DOI: 10.4049/jimmunol.1900072] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 12/11/2019] [Indexed: 11/19/2022]
|
7
|
Characterization of retroviral vector derived DNA-isoforms by PCR and sequencing. J Verbrauch Lebensm 2019. [DOI: 10.1007/s00003-019-01215-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
8
|
Muraoka D, Seo N, Hayashi T, Tahara Y, Fujii K, Tawara I, Miyahara Y, Okamori K, Yagita H, Imoto S, Yamaguchi R, Komura M, Miyano S, Goto M, Sawada SI, Asai A, Ikeda H, Akiyoshi K, Harada N, Shiku H. Antigen delivery targeted to tumor-associated macrophages overcomes tumor immune resistance. J Clin Invest 2019; 129:1278-1294. [PMID: 30628894 DOI: 10.1172/jci97642] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 01/03/2019] [Indexed: 12/13/2022] Open
Abstract
Immune checkpoint inhibitors and adoptive transfer of gene-engineered T cells have emerged as novel therapeutic modalities for hard-to-treat solid tumors; however, many patients are refractory to these immunotherapies, and the mechanisms underlying tumor immune resistance have not been fully elucidated. By comparing the tumor microenvironment of checkpoint inhibition-sensitive and -resistant murine solid tumors, we observed that the resistant tumors had low immunogenicity. We identified antigen presentation by CD11b+F4/80+ tumor-associated macrophages (TAMs) as a key factor correlated with immune resistance. In the resistant tumors, TAMs remained inactive and did not exert antigen-presenting activity. Targeted delivery of a long peptide antigen to TAMs by using a nano-sized hydrogel (nanogel) in the presence of a TLR agonist activated TAMs, induced their antigen-presenting activity, and thereby transformed the resistant tumors into tumors sensitive to adaptive immune responses such as adoptive transfer of tumor-specific T cell receptor-engineered T cells. These results indicate that the status and function of TAMs have a significant impact on tumor immune sensitivity and that manipulation of TAM functions would be an effective approach for improving the efficacy of immunotherapies.
Collapse
Affiliation(s)
- Daisuke Muraoka
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan.,Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Naohiro Seo
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan.,ERATO Akiyoshi Bio-Nanotransporter Project, Japan Science and Technology Agency (JST), Tokyo, Japan
| | - Tae Hayashi
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Yoshiro Tahara
- ERATO Akiyoshi Bio-Nanotransporter Project, Japan Science and Technology Agency (JST), Tokyo, Japan.,Department of Polymer Chemistry, Kyoto University Graduate School of Engineering, Kyoto, Japan.,Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, Fukuoka, Japan
| | - Keisuke Fujii
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Isao Tawara
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Mie, Japan
| | - Yoshihiro Miyahara
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Kana Okamori
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Seiya Imoto
- Division of Health Medical Data Science, Health Intelligence Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Rui Yamaguchi
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Mitsuhiro Komura
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Satoru Miyano
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Masahiro Goto
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, Fukuoka, Japan
| | - Shin-Ichi Sawada
- ERATO Akiyoshi Bio-Nanotransporter Project, Japan Science and Technology Agency (JST), Tokyo, Japan.,Department of Polymer Chemistry, Kyoto University Graduate School of Engineering, Kyoto, Japan
| | - Akira Asai
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Hiroaki Ikeda
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kazunari Akiyoshi
- ERATO Akiyoshi Bio-Nanotransporter Project, Japan Science and Technology Agency (JST), Tokyo, Japan.,Department of Polymer Chemistry, Kyoto University Graduate School of Engineering, Kyoto, Japan
| | - Naozumi Harada
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan.,ERATO Akiyoshi Bio-Nanotransporter Project, Japan Science and Technology Agency (JST), Tokyo, Japan.,United Immunity Co., Ltd., Mie, Japan
| | - Hiroshi Shiku
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan.,ERATO Akiyoshi Bio-Nanotransporter Project, Japan Science and Technology Agency (JST), Tokyo, Japan
| |
Collapse
|
9
|
Rodolfo M, Parmiani G. Growth Inhibition of Murine Colonic Adenocarcinoma by Tumor Immune but not by IL-2-Activated or Alloactivated Lymphocytes. TUMORI JOURNAL 2018; 73:1-9. [PMID: 3493573 DOI: 10.1177/030089168707300101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The antigenic profile of C-26 and C-51 BALB/c colonic adenocarcinomas was examined by in vivo and in vitro assays. Mice immunized with irradiated C-26 or C-51 tumor cells from freshly excised tumor nodules or from in vitro-growing cell lines were able to reject a challenge of both tumors. Spleen lymphocytes of immune but not of normal mice were effective in cross-inhibiting tumor growth in vivo in a Winn assay. Tissue-associated antigens common to C-26 and C-51 and to their metastases but not to other syngeneic neoplasms were detected in vitro by cytotoxic T lymphocytes obtained after 5 days of a secondary culture of immune lymphocytes and irradiated tumor cells. Activated lymphocytes were obtained by exposure of spleen cells to interleukin 2 or by allostimulation. Such lymphocytes, although cytotoxic in vitro on C-26 and C-51 carcinomas, were unable to significantly reduce in vivo tumor growth in the Winn assay.
Collapse
|
10
|
Uversky VN, El-Baky NA, El-Fakharany EM, Sabry A, Mattar EH, Uversky AV, Redwan EM. Functionality of intrinsic disorder in tumor necrosis factor-α and its receptors. FEBS J 2017; 284:3589-3618. [PMID: 28746777 DOI: 10.1111/febs.14182] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/15/2017] [Accepted: 07/20/2017] [Indexed: 01/02/2023]
Abstract
Tumor necrosis factor-α (TNF-α) is a pleiotropic inflammatory cytokine that exerts potent cytotoxic effects on solid tumor cells, while not affecting their normal counterparts. It is also known that TNF-α exerts many of its biological functions via interaction with specific receptors. To understand the potential roles of intrinsic disorder in the functioning of this important cytokine, we explored the peculiarities of intrinsic disorder distribution in human TNF-α and its homologs from various species, ranging from zebrafish to chimpanzee. We also studied the peculiarities of intrinsic disorder distribution in human TNF-α receptors, TNFR1 and TNFR2. Analysis revealed that cytoplasmic domains of TNF-α and its receptors are expected to be highly disordered. Furthermore, although the sequence identities of analyzed TNF-α homologs range from 99.57% (between human and chimpanzee proteins) to 22.33% (between frog and fish proteins), their intrinsic disorder profiles are characterized by a remarkable similarity. These observations indicate that the peculiarities of distribution of the intrinsic disorder propensity within the amino acid sequences are evolutionary conserved, and therefore could be of functional importance for this family of proteins. We also show that disordered and flexible regions of human TNF-α and its TNFR1 and TNFR2 receptors are crucial for some of their biological activities.
Collapse
Affiliation(s)
- Vladimir N Uversky
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Nawal Abd El-Baky
- Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria, Egypt
| | - Esmail M El-Fakharany
- Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria, Egypt
| | - Amira Sabry
- Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria, Egypt
| | - Ehab H Mattar
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Alexey V Uversky
- Center for Data Analytics and Biomedical Informatics, Department of Computer and Information Sciences, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria, Egypt
| |
Collapse
|
11
|
Fujii K, Miyahara Y, Harada N, Muraoka D, Komura M, Yamaguchi R, Yagita H, Nakamura J, Sugino S, Okumura S, Imoto S, Miyano S, Shiku H. Identification of an immunogenic neo-epitope encoded by mouse sarcoma using CXCR3 ligand mRNAs as sensors. Oncoimmunology 2017. [PMID: 28638727 PMCID: PMC5467990 DOI: 10.1080/2162402x.2017.1306617] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The CXCR3 ligands CXCL9, 10, and 11 play critical roles in the amplification of immune responses by recruiting CXCR3+ immune effector cells to the tumor site. Taking advantage of this property of CXCR3 ligands, we aimed to establish a novel approach to identify immunogenic mutated-antigens. We examined the feasibility of using CXCR3 ligand mRNAs as sensors for detection of specific immune responses in human and murine systems. We further investigated whether this approach is applicable for the identification of immunogenic mutated-antigens by using murine sarcoma lines. Rapid synthesis of CXCR3 ligand mRNAs occurred shortly after specific immune responses in both human and murine immune systems. Particularly, in CMS5 tumor-bearing mice, we detected specific immune responses to mutated mitogen-activated protein kinase 2 (ERK2), which has previously been identified as an immunogenic mutated-antigen. Furthermore, by combining this approach with whole-exome and transcriptome sequencing analyses, we identified an immunogenic neo-epitope derived from mutated staphylococcal nuclease domain-containing protein 1 (Snd1) in CMS7 tumor-bearing mice. Most importantly, we successfully detected the specific immune response to this neo-epitope even without co-administration of anti-cytotoxic T-lymphocyte protein-4 (CTLA-4), anti-programmed cell death-1 (PD-1) and anti-glucocorticoid-induced TNFR-related protein (GITR) antibodies, which vigorously augmented the immune response and consequently enabled us to detect the specific immune response to this neo-epitope by conventional IFNγ intracellular staining method. Our data indicate the potential usefulness of this strategy for the identification of immunogenic mutated-antigens. We propose that this approach would be of great help for the development of personalized cancer vaccine therapies in future.
Collapse
Affiliation(s)
- Keisuke Fujii
- Department of Immuno-Gene Therapy, Graduate School of Medicine, Mie University, Mie, Japan
| | - Yoshihiro Miyahara
- Department of Immuno-Gene Therapy, Graduate School of Medicine, Mie University, Mie, Japan
| | - Naozumi Harada
- Department of Immuno-Gene Therapy, Graduate School of Medicine, Mie University, Mie, Japan
| | - Daisuke Muraoka
- Department of Immuno-Gene Therapy, Graduate School of Medicine, Mie University, Mie, Japan.,Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Mitsuhiro Komura
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Rui Yamaguchi
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Junko Nakamura
- Department of Immuno-Gene Therapy, Graduate School of Medicine, Mie University, Mie, Japan
| | - Sahoko Sugino
- Department of Immuno-Gene Therapy, Graduate School of Medicine, Mie University, Mie, Japan
| | - Satoshi Okumura
- Department of Immuno-Gene Therapy, Graduate School of Medicine, Mie University, Mie, Japan
| | - Seiya Imoto
- Division of Health Medical Data Science, Health Intelligence Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Satoru Miyano
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hiroshi Shiku
- Department of Immuno-Gene Therapy, Graduate School of Medicine, Mie University, Mie, Japan
| |
Collapse
|
12
|
Galanos C, Gumenscheimer M, Mühlradt P, Jirillo E, Freudenberg M. MALP-2, a Mycoplasma lipopeptide with classical endotoxic properties: end of an era of LPS monopoly? ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519000060061001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although some activities of LPS are shared by other bacterial components, for half a century LPS has been regarded as unique in displaying many pathophysiological activities. Here we report on a synthetic lipopeptide, MALP-2 from Mycoplasma fermentans , which expresses potent endotoxin-like activity and whose lethal toxicity is comparable to that of LPS. With the exception of the Limulus lysate gelation test, in which MALP-2 was approximately 1000-fold less active than LPS, the synthetic lipopeptide induced all activities tested for, and in most cases to an extent comparable to that of LPS. Unlike LPS, the biological activities of MALP-2 were expressed both in LPSresponder and in LPS-non-responder mice (BALB/c/l, C57BL10/ScCr), indicating that MALP-2 signaling, unlike that of LPS, is not transduced via the Toll-like receptor (Tlr) 4 protein. MALP-2 expressed no toxicity in normal or sensitized Tlr2 knockout (Tlr2— /—) mice indicating that its toxic activity is induced via Tlr2 signaling. The phenomenology of the lethal shock induced by MALP-2 in normal or sensitized mice, i.e. the kinetics of its development and symptoms of illness exhibited by the treated animals, was very reminiscent of the lethal shock induced by LPS.
Collapse
Affiliation(s)
- C. Galanos
- Max-Planck Institut für Immunbiologie, Freiburg, Germany
| | | | - P.F. Mühlradt
- Immunobiology Research Group, GBF, Braunschweig, Germany
| | - E. Jirillo
- Department of Immunology, Faculty of Medicina, Policlinico University, Bari, Italy
| | | |
Collapse
|
13
|
Kashima H, Momose F, Umehara H, Miyoshi N, Ogo N, Muraoka D, Shiku H, Harada N, Asai A. Epirubicin, Identified Using a Novel Luciferase Reporter Assay for Foxp3 Inhibitors, Inhibits Regulatory T Cell Activity. PLoS One 2016; 11:e0156643. [PMID: 27284967 PMCID: PMC4902191 DOI: 10.1371/journal.pone.0156643] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 05/17/2016] [Indexed: 12/21/2022] Open
Abstract
Forkhead box protein p3 (Foxp3) is crucial to the development and suppressor function of regulatory T cells (Tregs) that have a significant role in tumor-associated immune suppression. Development of small molecule inhibitors of Foxp3 function is therefore considered a promising strategy to enhance anti-tumor immunity. In this study, we developed a novel cell-based assay system in which the NF-κB luciferase reporter signal is suppressed by the co-expressed Foxp3 protein. Using this system, we screened our chemical library consisting of approximately 2,100 compounds and discovered that a cancer chemotherapeutic drug epirubicin restored the Foxp3-inhibited NF-κB activity in a concentration-dependent manner without influencing cell viability. Using immunoprecipitation assay in a Treg-like cell line Karpas-299, we found that epirubicin inhibited the interaction between Foxp3 and p65. In addition, epirubicin inhibited the suppressor function of murine Tregs and thereby improved effector T cell stimulation in vitro. Administration of low dose epirubicin into tumor-bearing mice modulated the function of immune cells at the tumor site and promoted their IFN-γ production without direct cytotoxicity. In summary, we identified the novel action of epirubicin as a Foxp3 inhibitor using a newly established luciferase-based cellular screen. Our work also demonstrated our screen system is useful in accelerating discovery of Foxp3 inhibitors.
Collapse
Affiliation(s)
- Hajime Kashima
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Fumiyasu Momose
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie 514-8507, Japan
| | - Hiroshi Umehara
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Nao Miyoshi
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Naohisa Ogo
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Daisuke Muraoka
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Hiroshi Shiku
- Mie University Center for Comprehensive Cancer Immunotherapy, Mie 514-8507, Japan
| | - Naozumi Harada
- Mie University Center for Comprehensive Cancer Immunotherapy, Mie 514-8507, Japan
- * E-mail: (AA); (NH)
| | - Akira Asai
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
- * E-mail: (AA); (NH)
| |
Collapse
|
14
|
Humar M, Azemar M, Maurer M, Groner B. Adaptive Resistance to Immunotherapy Directed Against p53 Can be Overcome by Global Expression of Tumor-Antigens in Dendritic Cells. Front Oncol 2014; 4:270. [PMID: 25340039 PMCID: PMC4186483 DOI: 10.3389/fonc.2014.00270] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 09/17/2014] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy of cancer utilizes dendritic cells (DCs) for antigen presentation and the induction of tumor-specific immune responses. However, the therapeutic induction of anti-tumor immunity is limited by tumor escape mechanisms. In this study, immortalized dendritic D2SC/1 cells were transduced with a mutated version of the p53 tumor suppressor gene, p53M234I, or p53C132F/E168G, which are overexpressed in MethA fibrosarcoma tumor cells. In addition, D2SC/1 cells were fused with MethA tumor cells to generate a vaccine that potentially expresses a large repertoire of tumor-antigens. Cellular vaccines were transplanted onto Balb/c mice and MethA tumor growth and anti-tumor immune responses were examined in vaccinated animals. D2SC/1–p53M234I and D2SC/1–p53C132F/E168G cells induced strong therapeutic and protective MethA tumor immunity upon transplantation in Balb/c mice. However, in a fraction of immunized mice MethA tumor growth resumed after an extended latency period. Analysis of these tumors indicated loss of p53 expression. Mice, pre-treated with fusion hybrids generated from D2SC/1 and MethA tumor cells, suppressed MethA tumor growth and averted adaptive immune escape. Polyclonal B-cell responses directed against various MethA tumor proteins could be detected in the sera of D2SC/1–MethA inoculated mice. Athymic nude mice and Balb/c mice depleted of CD4+ or CD8+ T-cells were not protected against MethA tumor cell growth after immunization with D2SC/1–MethA hybrids. Our results highlight a potential drawback of cancer immunotherapy by demonstrating that the induction of a specific anti-tumor response favors the acquisition of tumor phenotypes promoting immune evasion. In contrast, the application of DC/tumor cell fusion hybrids prevents adaptive immune escape by a T-cell dependent mechanism and provides a simple strategy for personalized anti-cancer treatment without the need of selectively priming the host immune system.
Collapse
Affiliation(s)
- Matjaz Humar
- Pharmaceutical Biology and Biotechnology, Albert-Ludwigs-University of Freiburg , Freiburg , Germany
| | - Marc Azemar
- Internistische Onkologie, Tumor Biology Center , Freiburg , Germany
| | - Martina Maurer
- Basilea Pharmaceutica International Ltd. , Basel , Switzerland
| | - Bernd Groner
- Institute for Biomedical Research, Georg Speyer Haus , Frankfurt am Main , Germany
| |
Collapse
|
15
|
Muraoka D, Harada N, Hayashi T, Tahara Y, Momose F, Sawada SI, Mukai SA, Akiyoshi K, Shiku H. Nanogel-based immunologically stealth vaccine targets macrophages in the medulla of lymph node and induces potent antitumor immunity. ACS NANO 2014; 8:9209-9218. [PMID: 25180962 DOI: 10.1021/nn502975r] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Because existing therapeutic cancer vaccines provide only a limited clinical benefit, a different vaccination strategy is necessary to improve vaccine efficacy. We developed a nanoparticulate cancer vaccine by encapsulating a synthetic long peptide antigen within an immunologically inert nanoparticulate hydrogel (nanogel) of cholesteryl pullulan (CHP). After subcutaneous injection to mice, the nanogel-based vaccine was efficiently transported to the draining lymph node, and was preferentially engulfed by medullary macrophages but was not sensed by other macrophages and dendritic cells (so-called "immunologically stealth mode"). Although the function of medullary macrophages in T cell immunity has been unexplored so far, these macrophages effectively cross-primed the vaccine-specific CD8(+) T cells in the presence of a Toll-like receptor (TLR) agonist as an adjuvant. The nanogel-based vaccine significantly inhibited in vivo tumor growth in the prophylactic and therapeutic settings, compared to another vaccine formulation using a conventional delivery system, incomplete Freund's adjuvant. We also revealed that lymph node macrophages were highly responsive to TLR stimulation, which may underlie the potency of the macrophage-oriented, nanogel-based vaccine. These results indicate that targeting medullary macrophages using the immunologically stealth nanoparticulate delivery system is an effective vaccine strategy.
Collapse
Affiliation(s)
- Daisuke Muraoka
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine , Mie 514-8507, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Castle JC, Loewer M, Boegel S, de Graaf J, Bender C, Tadmor AD, Boisguerin V, Bukur T, Sorn P, Paret C, Diken M, Kreiter S, Türeci Ö, Sahin U. Immunomic, genomic and transcriptomic characterization of CT26 colorectal carcinoma. BMC Genomics 2014; 15:190. [PMID: 24621249 PMCID: PMC4007559 DOI: 10.1186/1471-2164-15-190] [Citation(s) in RCA: 288] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 02/21/2014] [Indexed: 12/15/2022] Open
Abstract
Background Tumor models are critical for our understanding of cancer and the development of cancer therapeutics. Here, we present an integrated map of the genome, transcriptome and immunome of an epithelial mouse tumor, the CT26 colon carcinoma cell line. Results We found that Kras is homozygously mutated at p.G12D, Apc and Tp53 are not mutated, and Cdkn2a is homozygously deleted. Proliferation and stem-cell markers, including Top2a, Birc5 (Survivin), Cldn6 and Mki67, are highly expressed while differentiation and top-crypt markers Muc2, Ms4a8a (MS4A8B) and Epcam are not. Myc, Trp53 (tp53), Mdm2, Hif1a, and Nras are highly expressed while Egfr and Flt1 are not. MHC class I but not MHC class II is expressed. Several known cancer-testis antigens are expressed, including Atad2, Cep55, and Pbk. The highest expressed gene is a mutated form of the mouse tumor antigen gp70. Of the 1,688 non-synonymous point variations, 154 are both in expressed genes and in peptides predicted to bind MHC and thus potential targets for immunotherapy development. Based on its molecular signature, we predicted that CT26 is refractory to anti-EGFR mAbs and sensitive to MEK and MET inhibitors, as have been previously reported. Conclusions CT26 cells share molecular features with aggressive, undifferentiated, refractory human colorectal carcinoma cells. As CT26 is one of the most extensively used syngeneic mouse tumor models, our data provide a map for the rationale design of mode-of-action studies for pre-clinical evaluation of targeted- and immunotherapies. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-190) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- John C Castle
- TRON gGmbH - Translational Oncology, Johannes Gutenberg-University Medical Center gGmbH, Langenbeckstr, 1, Building 708, 55131 Mainz, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Reis BS, Jungbluth AA, Frosina D, Holz M, Ritter E, Nakayama E, Ishida T, Obata Y, Carver B, Scher H, Scardino PT, Slovin S, Subudhi SK, Reuter VE, Savage C, Allison JP, Melamed J, Jäger E, Ritter G, Old LJ, Gnjatic S. Prostate cancer progression correlates with increased humoral immune response to a human endogenous retrovirus GAG protein. Clin Cancer Res 2013; 19:6112-25. [PMID: 24081977 DOI: 10.1158/1078-0432.ccr-12-3580] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Human endogenous retroviruses (HERV) encode 8% of the human genome. While HERVs may play a role in autoimmune and neoplastic disease, no mechanistic association has yet been established. We studied the expression and immunogenicity of a HERV-K GAG protein encoded on chromosome 22q11.23 in relation to the clinical course of prostate cancer. EXPERIMENTAL DESIGN In vitro expression of GAG-HERV-K was analyzed in panels of normal and malignant tissues, microarrays, and cell lines, and effects of demethylation and androgen stimulation were evaluated. Patient sera were analyzed for seroreactivity to GAG-HERV-K and other self-antigens by ELISA and seromics (protein array profiling). RESULTS GAG-HERV-K expression was most frequent in prostate tissues and regulated both by demethylation of the promoter region and by androgen stimulation. Serum screening revealed that antibodies to GAG-HERV-K are found in a subset of patients with prostate cancer (33 of 483, 6.8%) but rarely in male healthy donors (1 of 55, 1.8%). Autoantibodies to GAG-HERV-K occurred more frequently in patients with advanced prostate cancer (29 of 191 in stage III-IV, 21.0%) than in early prostate cancer (4 of 292 in stages I-II, 1.4%). Presence of GAG-HERV-K serum antibody was correlated with worse survival of patients with prostate cancer, with a trend for faster biochemical recurrence in patients with antibodies to GAG-HERV-K. CONCLUSIONS Preferential expression of GAG-HERV-K ch22q11.23 in prostate cancer tissue and increased frequency of autoantibodies observed in patients with advanced prostate cancer make this protein one of the first bona fide retroviral cancer antigens in humans, with potential as a biomarker for progression and biochemical recurrence rate of prostate cancer. Clin Cancer Res; 19(22); 6112-25. ©2013 AACR.
Collapse
Affiliation(s)
- Bernardo Sgarbi Reis
- Authors' Affiliations: Ludwig Institute for Cancer Research, New York Branch at Memorial Sloan-Kettering Cancer Center; Departments of Surgery, Medicine, Pathology, Biostatistics, and Immunology, Memorial Sloan-Kettering Cancer Center; NYU Langone Medical Center, New York; Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama; RIKEN Bioresource Center, Tsukuba, Ibaraki, Japan; and Klinik für Onkologie und Hämatologie, Krankenhaus Nordwest, Frankfurt, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Zimmerman MA, Rahman NT, Yang D, Lahat G, Lazar AJ, Pollock RE, Lev D, Liu K. Unphosphorylated STAT1 promotes sarcoma development through repressing expression of Fas and bad and conferring apoptotic resistance. Cancer Res 2012; 72:4724-32. [PMID: 22805310 DOI: 10.1158/0008-5472.can-12-1347] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
STAT1 exists in phosphorylated (pSTAT1) and unphosphorylated (uSTAT1) forms each regulated by IFN-γ. Although STAT1 is a key mediator of the IFN-γ signaling pathway, an essential component of the host cancer immunosurveillance system, STAT1 is also overexpressed in certain human cancers where the functions of pSTAT1 and uSTAT1 are ill defined. Using a murine model of soft tissue sarcoma (STS), we show that disruption of the IFN effector molecule IRF8 decreases pSTAT1 and increases uSTAT1 in STS cells, thereby increasing their metastatic potential. We determined that the IRF8 gene promoter was hypermethylated frequently in human STS. An analysis of 123 human STS specimens revealed that high uSTAT1 levels in tumor cells was correlated with a reduction in disease-specific survival (DSS), whereas high pSTAT1 levels in tumor cells were correlated with an increase in DSS. In addition, uSTAT1 levels were negatively correlated with pSTAT1 levels in these STS specimens. Mechanistic investigations revealed that IRF8 suppressed STAT1 transcription by binding the STAT1 promoter. RNAi-mediated silencing of STAT1 in STS cells was sufficient to increase expression of the apoptotic mediators Fas and Bad and to elevate the sensitivity of STS cells to Fas-mediated apoptosis. Together, our findings show how the phosphorylation status of pSTAT1 determines its function as a tumor suppressor, with uSTAT1 acting as a tumor promoter that acts by elevating resistance to Fas-mediated apoptosis to promote immune escape.
Collapse
Affiliation(s)
- Mary A Zimmerman
- Department of Biochemistry and Molecular Biology, Georgia Health Sciences University, Augusta, Georgia 30912, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Takahashi T, Shiku H. Cell surface antigens: invaluable landmarks reflecting the nature of cells. CANCER IMMUNITY 2012; 12:2. [PMID: 22896747 PMCID: PMC3380351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Affiliation(s)
- Toshitada Takahashi
- Comprehensive Health Science Center, Aichi Health Promotion Foundation, Higashiura Town, Aichi, Japan.
| | | |
Collapse
|
20
|
Kemmler CB, Clambey ET, Kedl RM, Slansky JE. Elevated tumor-associated antigen expression suppresses variant peptide vaccine responses. THE JOURNAL OF IMMUNOLOGY 2011; 187:4431-9. [PMID: 21940675 DOI: 10.4049/jimmunol.1101555] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Variant peptide vaccines are used clinically to expand T cells that cross-react with tumor-associated Ags (TAA). To investigate the effects of elevated endogenous TAA expression on variant peptide-induced responses, we used the GP70 TAA model. Although young BALB/c mice display T cell tolerance to the TAA GP70(423-431) (AH1), expression of GP70 and suppression of AH1-specific responses increases with age. We hypothesized that as TAA expression increases, the AH1 cross-reactivity of variant peptide-elicited T cell responses diminishes. Controlling for immunosenescence, we showed that elevated GP70 expression suppressed AH1 cross-reactive responses elicited by two AH1 peptide variants. A variant that elicited almost exclusively AH1 cross-reactive T cells in young mice elicited few or no T cells in aging mice with Ab-detectable GP70 expression. In contrast, a variant that elicited a less AH1 cross-reactive T cell response in young mice successfully expanded AH1 cross-reactive T cells in all aging mice tested. However, these T cells bound the AH1/MHC complex with a relatively short half-life and responded poorly to ex vivo stimulation with the AH1 peptide. Variant peptide vaccine responses were also suppressed when AH1 peptide is administered tolerogenically to young mice before vaccination. Analyses of variant-specific precursor T cells from naive mice with Ab-detectable GP70 expression determined that these T cells expressed PD-1 and had downregulated IL-7Rα expression, suggesting they were anergic or undergoing deletion. Although variant peptide vaccines were less effective as TAA expression increases, data presented in this article also suggest that complementary immunotherapies may induce the expansion of T cells with functional TAA recognition.
Collapse
Affiliation(s)
- Charles B Kemmler
- Integrated Department of Immunology, School of Medicine, University of Colorado Denver and National Jewish Health, Denver, CO 80206, USA
| | | | | | | |
Collapse
|
21
|
Hiraoka N, Yamazaki-Itoh R, Ino Y, Mizuguchi Y, Yamada T, Hirohashi S, Kanai Y. CXCL17 and ICAM2 are associated with a potential anti-tumor immune response in early intraepithelial stages of human pancreatic carcinogenesis. Gastroenterology 2011; 140:310-21. [PMID: 20955708 DOI: 10.1053/j.gastro.2010.10.009] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Revised: 09/29/2010] [Accepted: 10/08/2010] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Anti-tumor immunity changes over the course of tumor progression; it is not clear how or when the developing tumor overcomes immune surveillance. Intraductal papillary mucinous neoplasm (IPMN) is an intraepithelial precursor lesion of pancreatic cancer that progresses from adenoma to carcinoma. We investigated when and how the human anti-tumor immune reaction changes during pancreatic tumor development. METHODS Using immunohistochemical analysis of cells isolated from patients with IPMN, the numbers of tumor-infiltrating lymphocytes and dendritic cells and the maturation state of dendritic cells in the regional lymph nodes were investigated during tumor progression. Gene expression profiles were compared among epithelial neoplastic cells at each stage of tumor development. Biological functions of the selected gene products were analyzed using syngeneic mouse models. RESULTS The anti-tumor immune reaction changed from an immune response to immune tolerance between the stages of intraductal papillary mucinous adenoma (IPMA) and intraductal papillary mucinous carcinoma (IPMC). Chemokine (C-X-C motif) ligand 17 (CXCL17) and intercellular adhesion molecule 2 (ICAM2) were involved in immune surveillance during tumor development-their expression levels were up-regulated exclusively in IPMA and disappeared from IPMC. CXCL17 and ICAM2 induced infiltration and accumulation of the tumor epithelial layer by immature myeloid dendritic cells. This was followed by a cellular immune reaction and ICAM2 simultaneously promoted the susceptibility of the tumor cells to cytotoxic T-cell-mediated cytolysis. These processes had a synergistic effect to increase the anti-tumor immune response. CONCLUSIONS Immune surveillance occurs during the early intraepithelial stages of human pancreatic carcinogenesis and is mediated by expression of CXCL17 and ICAM2.
Collapse
Affiliation(s)
- Nobuyoshi Hiraoka
- Pathology Division, National Cancer Center Research Institute, Tokyo, Japan.
| | | | | | | | | | | | | |
Collapse
|
22
|
James E, Yeh A, King C, Korangy F, Bailey I, Boulanger DS, Van den Eynde BJ, Murray N, Elliott TJ. Differential suppression of tumor-specific CD8+ T cells by regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2010; 185:5048-55. [PMID: 20921533 DOI: 10.4049/jimmunol.1000134] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In the CT26 BALB/c murine model of colorectal carcinoma, depletion of regulatory T cells (Tregs) prior to tumor inoculation results in protective immunity to both CT26 and other BALB/c-derived tumors of diverse histological origin. In this paper, we show that cross-protection can be conferred by adoptively transferred CD8(+) CTLs. Other schedules for inducing immunity to CT26 have been described, but they do not lead to cross-protection. We show that Treg ablation facilitates the development of new CTL specificities that are normally cryptic, and have mapped the root epitope of one of these responses. This work has allowed us to demonstrate how the specificity of CTL responses to tumor Ags can be controlled via differential suppression of CTL specificities by Tregs, and how this can result in very different physiological outcomes.
Collapse
Affiliation(s)
- Edward James
- Cancer Sciences Division, School of Medicine, University of Southampton, Southampton, Hampshire, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Watanabe A, Hara M, Chosa E, Nakamura K, Sekiya R, Shimizu T, Onitsuka T. Combination of adoptive cell transfer and antibody injection can eradicate established tumors in mice--an in vivo study using anti-OX40mAb, anti-CD25mAb and anti-CTLA4mAb-. Immunopharmacol Immunotoxicol 2010; 32:238-45. [PMID: 20001272 DOI: 10.3109/08923970903222355] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Boosting an effective immune response against established tumors remains a difficult challenge. This study shows the combination of 1) adoptive cell transfer using CD25 depleted splenocytes co-cultured with irradiated tumor cells, and 2) antibody injection therapy using CTLA4 blockade, the elimination of Treg and OX40, which together could eradicate an established MethA tumor in over 50% of the BALB/c mice. Each element of the protocol was shown to be necessary, as elimination of any factor except anti-CD25 antibody injection failed to eradicate the tumor.
Collapse
Affiliation(s)
- Akira Watanabe
- Department of Cardiovascular, Thoracic and General Surgery, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan.
| | | | | | | | | | | | | |
Collapse
|
24
|
Wild-type p53 enhances efficiency of simian virus 40 large-T-antigen-induced cellular transformation. J Virol 2009; 83:10106-18. [PMID: 19625393 DOI: 10.1128/jvi.00174-09] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Abortive infection of BALB/c mouse embryo fibroblasts differing in p53 gene status (p53(+/+) versus p53(-/)(-)) with simian virus 40 (SV40) revealed a quantitatively and qualitatively decreased transformation efficiency in p53(-/-) cells compared to p53(+/+) cells, suggesting a supportive effect of wild-type (wt) p53 in the SV40 transformation process. SV40 transformation efficiency also was low in immortalized p53(-/-) BALB/c 10-1 cells but could be restored to approximately the level in immortalized p53(+/+) BALB/c 3T3 cells by reconstituting wt p53, but not mutant p53 (mutp53), expression. Stable expression of large T antigen (LT) in p53(+/+) 3T3 cells resulted in full transformation, while LT expression in p53(-/-) 10-1 cells could not promote growth in suspension or in soft agar to a significant extent. The helper effect of wt p53 is mediated by its cooperation with LT and resides in the p53 N terminus, as an N-terminally truncated p53 (DeltaNp53) could not rescue the p53-null phenotype. The p53 N terminus serves as a scaffold for recruiting transcriptional regulators like p300/CBP and Mdm2 into the LT-p53 complex. Consequently, LT affected global and specific gene expression in p53(+/+) cells significantly more than in p53(-/-) cells. Our data suggest that recruitment of transcriptional regulators into the LT-p53 complex may help to modify cellular gene expression in response to the needs of cellular transformation.
Collapse
|
25
|
Hendrickson RC, Cicinnati VR, Albers A, Dworacki G, Gambotto A, Pagliano O, Tüting T, Mayordomo JI, Visus C, Appella E, Shabanowitz J, Hunt DF, DeLeo AB. Identification of a 17beta-hydroxysteroid dehydrogenase type 12 pseudogene as the source of a highly restricted BALB/c Meth A tumor rejection peptide. Cancer Immunol Immunother 2009; 59:113-24. [PMID: 19562340 DOI: 10.1007/s00262-009-0730-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 06/03/2009] [Indexed: 12/23/2022]
Abstract
Mass spectrometric analysis identified the peptide recognized by a cytotoxic T lymphocyte (CTL) specific for the chemically induced BALB/c Meth A sarcoma as derived from a 17beta-hydroxysteroid dehydrogenase type 12 (Hsd17b12) pseudogene present in the BALB/c genome, but only expressed in Meth A sarcoma. The sequence of the peptide is TYDKIKTGL and corresponds to Hsd17b12(114-122) with threonine instead of isoleucine at codon 114 and is designated Hsd17b12(114T). Immunization of mice with an Hsd17b12(114T) peptide-pulsed dendritic cell-based vaccine or a non-viral plasmid construct expressing the Hsd17b12(114T) peptide protected the mice from lethal Meth A tumor challenge in tumor rejection assays. A Hsd17b12(114-122) peptide-pulsed vaccine was ineffective in inducing resistance in mice to Meth A sarcoma. These results confirm the immunogenicity of the identified tumor peptide, as well as demonstrate the efficacies of these vaccine vehicles. These findings suggest that the role of the human homolog of Hsd17b12, HSD17B12, as a potential human tumor antigen be explored.
Collapse
|
26
|
Norian LA, Rodriguez PC, O'Mara LA, Zabaleta J, Ochoa AC, Cella M, Allen PM. Tumor-infiltrating regulatory dendritic cells inhibit CD8+ T cell function via L-arginine metabolism. Cancer Res 2009; 69:3086-94. [PMID: 19293186 DOI: 10.1158/0008-5472.can-08-2826] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DC) have a critical effect on the outcome of adaptive immune responses against growing tumors. Whereas it is generally assumed that the presence of phenotypically mature DCs should promote protective antitumor immunity, evidence to the contrary does exist. We describe here a novel mechanism by which tumor-infiltrating dendritic cells (TIDC) actively contribute to the suppression of protective CD8(+) T-cell-based antitumor immunity. Using the BALB/NeuT model of spontaneously arising mammary carcinoma, we found that canonical MHC II(+)/CD11b(+)/CD11c(high) TIDCs act as regulatory DCs to suppress CD8(+) T-cell function, resulting in diminished T-cell-based antitumor immunity in vivo. Stimulation of naive T cells with regulatory TIDCs resulted in an altered cell fate program characterized by minimal T-cell expansion, impaired IFNgamma production, and anergy. Suppression by regulatory TIDCs overcame stimulatory signals provided by standard DCs, occurred in the absence of cognate interactions with T cells, and was mediated primarily by arginase metabolism of l-arginine. Immunosuppressive TIDCs were found in every murine tumor type examined and were phenotypically distinct from tumor-infiltrating CD11c(int-low)/CD11b(+)/Gr-1(+) myeloid-derived suppressor cells. Thus, within the tumor microenvironment, MHC II(+) TIDCs can function as potent suppressors of CD8(+) T-cell immunity.
Collapse
Affiliation(s)
- Lyse A Norian
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Haridas S, Bowers M, Tusano J, Mehojah J, Kirkpatrick M, Burnham DK. The impact of Meth A fibrosarcoma derived EMAP II on dendritic cell migration. Cytokine 2008; 44:304-9. [PMID: 18951814 DOI: 10.1016/j.cyto.2008.09.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2008] [Revised: 08/19/2008] [Accepted: 09/12/2008] [Indexed: 10/21/2022]
Abstract
Studies have suggested that tumors are capable of modulating dendritic cell (DC) phenotype. A soluble protein produced by certain tumors, endothelial monocyte-activating polypeptide II (EMAP II) has been suggested as an anti-tumor agent based on its anti-angiogenic activity. However, this factor has not been evaluated for effects on DC. In this study, we analyzed the effect of Meth A fibrosarcoma supernatant and recombinant human EMAP II on DC migration. This included the migration of Langerhans cells from mouse ear skin sections and the migration of cells of a dendritic cell line (JAWS II) in a transwell culture system. The results of these studies indicated that EMAP II stimulates the migration of DC. Additional studies showed that the presence of the ascites form of the Meth A tumor led to a decrease in Langerhans cell (LC) numbers in the skin, and this decrease could be partially blocked by neutralizing antibody specific for EMAP II. Subcutaneous injection at the base of the ear of recombinant human EMAP II also led to a decrease in epidermal LC similar to that observed in tumor bearing mice. Together, these results suggest novel roles for EMAP II in modulating the migration of DC and suggest that these effects may modify Meth A tumor/host interactions.
Collapse
Affiliation(s)
- Seema Haridas
- Department of Microbiology and Molecular Genetics, 307 LSE, Stillwater, OK 74078, USA
| | | | | | | | | | | |
Collapse
|
28
|
Lennox ES. How are tumour antigens related to normal antigens? CIBA FOUNDATION SYMPOSIUM 2008; 96:89-103. [PMID: 6343008 DOI: 10.1002/9780470720776.ch6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Tumour cells share with normal cells antigens characteristic of defined states of differentiation. Is there anything else? An example of tumour antigens not expressed anywhere in normal tissue is the set of tumour-specific transplantation antigens (TSTA) of murine chemically induced sarcomas. There is evidence that at least one TSTA specificity is retrovirus-derived, is carried on the envelope protein gp70, and probably arises by the recombination events that yield the diverse gp70s of the MCF strains of murine leukaemia viruses. Whether a similar mechanism can generate human tumour antigens depends on the yet unanswered question of whether human cells have retroviruses in their genomes capable of recombination. Aside from this, the only other mechanism known for antigen expression on tumours is via their oncogenes, which seem to make normal cell products. Such products, or secondary consequences of their production, were they normally expressed only at an early stage of development, would be candidates for 'fetal antigens'. While only the two mechanisms mentioned above seem the ready sources of 'tumour-associated antigens', it would be too early--in the face of ever more startling information about gene mobility and rearrangements--to think we have exhausted possible mechanisms for generating tumour antigens.
Collapse
|
29
|
Norian LA, Allen PM. Rapid maturation of effector T cells in tumors, but not lymphoid organs, during tumor regression. PLoS One 2007; 2:e821. [PMID: 17786193 PMCID: PMC1950566 DOI: 10.1371/journal.pone.0000821] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2007] [Accepted: 08/09/2007] [Indexed: 12/18/2022] Open
Abstract
Increasing the efficacy of adoptively transferred, tumor antigen specific T cells is a major goal of immunotherapy. Clearly, a more thorough understanding of the effector phase of T cell responses, within the tumor site itself, would be beneficial. To examine this issue, we adoptively transferred tumor antigen-specific effector T cells into tumor-bearing mice, then performed kinetic evaluations of their phenotype, function, and survival in tumors, draining lymph nodes (dLNs), and spleens during regression of murine fibrosarcomas. Effector function in tumors was quantitated through the use of a novel intratumoral cytolytic assay. This approach revealed dynamic changes in the phenotype, cytolytic capacity, and viability of tumor infiltrating effector T cells during the course of tumor regression. Over a period of days, T cells within tumors rapidly transitioned from a CD25(hi)/CD27(hi) to a CD25(low)/CD27(low) phenotype and displayed an increase in cytolytic capacity, indicative of effector maturation. Simultaneously, however, the viability of maturing T cells within tumors diminished. In contrast, transferred T cells trafficking through lymphoid organs were much more static, as they maintained a stable phenotype, robust cytolytic activity, and high viability. Therefore, there exists a marked phenotypic and functional divergence between tumor-infiltrating effector T cells and their counterparts in lymphoid organs. Our results indicate that the population of tumor-infiltrating T cells is unique in experiencing rapid effector maturation post-transfer, and suggest that strategies aimed at prolonging the survival of CD25(low)/CD27(low) full effectors, which displayed the highest levels of intratumoral cytolytic activity, should enhance the efficacy of T cell based tumor immunotherapies.
Collapse
Affiliation(s)
- Lyse A Norian
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America.
| | | |
Collapse
|
30
|
di Palma A, Matarese G, Leone V, Di Matola T, Acquaviva F, Acquaviva AM, Ricchi P. Aspirin reduces the outcome of anticancer therapy in Meth A-bearing mice through activation of AKT-glycogen synthase kinase signaling. Mol Cancer Ther 2006; 5:1318-24. [PMID: 16731765 DOI: 10.1158/1535-7163.mct-05-0473] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aspirin displays, at millimolar concentrations, several mechanisms independent from its ability to inhibit cyclooxygenases. Occasionally, the mechanisms displayed in vitro have been clearly related to an effect of clinical relevance in vivo. An expanding literature has been focusing on the cytoprotective effect of aspirin in neurodegenerative disorders and the activation of AKT pathway in neuroprotection and induction of resistance to anticancer drugs. In this work, we tested the ability of aspirin to activate the AKT survival pathway in methylcholanthrene-induced fibrosarcoma cells (Meth A) transplanted into BALB/c nude mice and the clinical effect of aspirin cotreatment during etoposide (VP-16)-based anticancer therapy. We found that cotreatment with aspirin reduced VP-16-induced apoptosis and activated AKT in vitro and in vivo. In Meth A-bearing mice, aspirin administration also activated glycogen synthase kinase-3 and reduced the activity and the efficacy of anticancer therapy in VP-16 cotreated animals. Our data suggest that the antiapoptotic effect of aspirin operates in vivo through the activation of AKT-glycogen synthase kinase pathway causing a decrease in the outcome of VP-16-based therapy. These findings could have clinical relevance in treatment of human malignancies.
Collapse
Affiliation(s)
- Antonella di Palma
- Dipartimento di Biologia e Patologia Cellulare e Molecolare L. Califano, Università Federico II, via S. Pansini 5, 80131 Naples, Italy.
| | | | | | | | | | | | | |
Collapse
|
31
|
Nishikawa H, Sato E, Briones G, Chen LM, Matsuo M, Nagata Y, Ritter G, Jäger E, Nomura H, Kondo S, Tawara I, Kato T, Shiku H, Old LJ, Galán JE, Gnjatic S. In vivo antigen delivery by a Salmonella typhimurium type III secretion system for therapeutic cancer vaccines. J Clin Invest 2006; 116:1946-54. [PMID: 16794737 PMCID: PMC1481660 DOI: 10.1172/jci28045] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2006] [Accepted: 05/09/2006] [Indexed: 12/21/2022] Open
Abstract
Bacterial vectors may offer many advantages over other antigen delivery systems for cancer vaccines. We engineered a Salmonella typhimurium vaccine strain to deliver the NY-ESO-1 tumor antigen (S. typhimurium-NY-ESO-1) through a type III protein secretion system. The S. typhimurium-NY-ESO-1 construct elicited NY-ESO-1-specific CD8+ and CD4+ T cells from peripheral blood lymphocytes of cancer patients in vitro. Oral administration of S. typhimurium-NY-ESO-1 to mice resulted in the regression of established NY-ESO-1-expressing tumors. Intratumoral inoculation of S. typhimurium-NY-ESO-1 to NY-ESO-1-negative tumors resulted in delivery of antigen in vivo and led to tumor regression in the presence of preexisting NY-ESO-1-specific CD8+ T cells. Specific T cell responses against at least 2 unrelated tumor antigens not contained in the vaccine were observed, demonstrating epitope spreading. We propose that antigen delivery through the S. typhimurium type III secretion system is a promising novel strategy for cancer vaccine development.
Collapse
Affiliation(s)
- Hiroyoshi Nishikawa
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA.
Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany.
Department of Medical Oncology and Immunology and
Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Mie, Japan
| | - Eiichi Sato
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA.
Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany.
Department of Medical Oncology and Immunology and
Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Mie, Japan
| | - Gabriel Briones
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA.
Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany.
Department of Medical Oncology and Immunology and
Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Mie, Japan
| | - Li-Mei Chen
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA.
Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany.
Department of Medical Oncology and Immunology and
Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Mie, Japan
| | - Mitsutoshi Matsuo
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA.
Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany.
Department of Medical Oncology and Immunology and
Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Mie, Japan
| | - Yasuhiro Nagata
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA.
Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany.
Department of Medical Oncology and Immunology and
Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Mie, Japan
| | - Gerd Ritter
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA.
Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany.
Department of Medical Oncology and Immunology and
Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Mie, Japan
| | - Elke Jäger
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA.
Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany.
Department of Medical Oncology and Immunology and
Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Mie, Japan
| | - Hideki Nomura
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA.
Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany.
Department of Medical Oncology and Immunology and
Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Mie, Japan
| | - Shigeto Kondo
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA.
Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany.
Department of Medical Oncology and Immunology and
Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Mie, Japan
| | - Isao Tawara
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA.
Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany.
Department of Medical Oncology and Immunology and
Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Mie, Japan
| | - Takuma Kato
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA.
Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany.
Department of Medical Oncology and Immunology and
Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Mie, Japan
| | - Hiroshi Shiku
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA.
Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany.
Department of Medical Oncology and Immunology and
Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Mie, Japan
| | - Lloyd J. Old
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA.
Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany.
Department of Medical Oncology and Immunology and
Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Mie, Japan
| | - Jorge E. Galán
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA.
Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany.
Department of Medical Oncology and Immunology and
Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Mie, Japan
| | - Sacha Gnjatic
- Ludwig Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Section of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA.
Medizinische Klinik II, Hämatologie-Onkologie, Krankenhaus Nordwest, Frankfurt, Germany.
Department of Medical Oncology and Immunology and
Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Mie, Japan
| |
Collapse
|
32
|
Nishikawa H, Kato T, Tawara I, Ikeda H, Kuribayashi K, Allen PM, Schreiber RD, Old LJ, Shiku H. IFN-γ Controls the Generation/Activation of CD4+CD25+ Regulatory T Cells in Antitumor Immune Response. THE JOURNAL OF IMMUNOLOGY 2005; 175:4433-40. [PMID: 16177085 DOI: 10.4049/jimmunol.175.7.4433] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immunization with serological identification of Ags by recombinant expression cloning (SEREX)-defined self-Ags leads to generation/activation of CD4+ CD25+ regulatory T cells with suppressive activities and enhanced expression of Foxp3. This is associated with increased susceptibility to pulmonary metastasis following challenge with syngeneic tumor cells and enhanced development of 3-methylcholanthrene-induced primary tumors. In contrast, coimmunization with the same SEREX-defined self-Ags mixed with a CTL epitope results in augmented CTL activity and heightened resistance to pulmonary metastasis, both of which depend on CD4+ Th cells. These active regulatory T cells and Th cells were derived from two distinct CD4+ T cell subsets, CD4+ CD25+ T cells and CD4+ CD25- T cells, respectively. In the present study, IFN-gamma was found to abrogate the generation/activation of CD4+ CD25+ regulatory T cells by immunization with SEREX-defined self-Ag. CD4+ CD25+ T cells from these IFN-gamma-treated mice failed to exhibit immunosuppressive activity as measured by 1) increased number of pulmonary metastasis, 2) enhanced development of 3-methylcholanthrene-induced primary tumors, 3) suppression of peptide-specific T cell proliferation, and 4) enhanced expression of Foxp3. The important role of IFN-gamma produced by CD8+ T cells was shown in experiments demonstrating that CD4+ CD25+ T cells cotransferred with CD8+ T cells from IFN-gamma(-/-) mice, but not from wild-type BALB/c mice, became immunosuppressive and enhanced pulmonary metastasis when recipient animals were subsequently immunized with a SEREX-defined self-Ag and a CTL epitope. These findings support the idea that IFN-gamma regulates the generation/activation of CD4+ CD25+ regulatory T cells.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cells, Cultured
- Epitopes/immunology
- Female
- Heat-Shock Proteins/immunology
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Interferon-gamma/physiology
- Lung Neoplasms/immunology
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mice, SCID
- Mice, Transgenic
- Receptors, Interleukin-2/biosynthesis
- Receptors, Interleukin-2/metabolism
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/pathology
- Sarcoma, Experimental/prevention & control
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Hiroyoshi Nishikawa
- Second Department of Internal Medicine, Mie University School of Medicine, Tsu, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
O'Mara LA, Norian LA, Kreamalmeyer D, White JM, Allen PM. T cell-mediated delay of spontaneous mammary tumor onset: increased efficacy with in vivo versus in vitro activation. THE JOURNAL OF IMMUNOLOGY 2005; 174:4662-9. [PMID: 15814690 DOI: 10.4049/jimmunol.174.8.4662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Peripheral tolerance to shared Ags expressed on both tumors and normal self-tissues presents a major barrier to T cell-based immunotherapy as a treatment for cancer. To assess the activity of tumor-specific T cells against spontaneously arising carcinomas in the context of shared Ag expression, we developed a model system whereby an identified tumor Ag, tumor ERK (tERK), is expressed transgenically on both normal mammary tissue and spontaneous mammary carcinomas. Transfer of in vitro-activated, tERK-specific DUC18 T cells delayed spontaneous tumor development in tERK-expressing mice when T cells were given before the development of palpable carcinomas. However, antitumor activity mediated by in vitro-activated DUC18 T cells, as measured by responsiveness against a transplanted tERK-expressing fibrosarcoma challenge, was lost within days of transfer. This loss was due to expression of tERK as a self-Ag on normal tissues and was independent of the presence of mammary tumors. In contrast, transferred naive DUC18 T cells maintained a long-term protective function in tERK-expressing mice. Ten-fold fewer naive T cells activated in vivo were able to replicate the delay in spontaneous tumor development achieved by in vitro-activated T cells. These results are in contrast to our earlier studies using transplanted tumors alone, in which in vitro-activated DUC18 T cells were more efficacious than naive DUC18 T cells and highlight the need to perform tumor studies in the presence of tumor Ag expression on normal self-tissue.
Collapse
Affiliation(s)
- Leigh A O'Mara
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
34
|
Liu K, Caldwell SA, Abrams SI. Immune Selection and Emergence of Aggressive Tumor Variants as Negative Consequences of Fas-Mediated Cytotoxicity and Altered IFN-γ-Regulated Gene Expression. Cancer Res 2005; 65:4376-88. [PMID: 15899830 DOI: 10.1158/0008-5472.can-04-4269] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antitumor responses can be induced in patients via active or adoptive immunotherapy, yet complete tumor eradication occurs infrequently. This paradox in tumor immunology led us to address two questions: (a) Does an antitumor response, which is intended to destroy the aberrant target population, also at the same time select for aggressive tumor variants (ATV) in vivo? (b) If this process does occur, what is the contribution of the perforin- or Fas-mediated effector mechanism in the immune selection of such ATV? Here, in an experimental mouse lung metastasis model, we showed that ATV generated either naturally in vivo or in vitro by anti-Fas selection resembled each other biologically and genetically as judged by enhanced tumor growth and genome-scale gene expression profiling, respectively. Furthermore, ATV that survived CTL adoptive immunotherapy displayed an even more profound loss of Fas expression and function as well as enhanced malignant proficiency in vivo. ATV, however, retained sensitivity to perforin-mediated lysis in vitro. Lastly, such ATV displayed a diminished responsiveness in their expression of IFN-gamma-regulated genes, including those mechanistically linked to Fas-mediated death (i.e., Fas and caspase-1). Overall, we showed that (a) immune selection did occur in vivo and played an important role in the emergence of ATV, (b) ATV bearing a Fas-resistant phenotype was a chief consequence of immune selection, and (c) an overall diminished responsiveness of IFN-gamma-regulated gene expression was characteristic of ATV. Thus, in this model, Fas-mediated cytotoxicity, in concert with IFN-gamma-regulated gene expression, mechanistically constituted significant determinants of immune selection of ATV in vivo.
Collapse
Affiliation(s)
- Kebin Liu
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland 20892-1402, USA
| | | | | |
Collapse
|
35
|
Cooperative Disengagement of Fas and Intercellular Adhesion Molecule-1 Function in Neoplastic Cells Confers Enhanced Colonization Efficiency. Cancer Res 2005. [DOI: 10.1158/0008-5472.1045.65.3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Understanding the mechanisms of tumor progression is crucial toward the development of therapeutic interventions. Although the loss of sensitivity to cell death is a hallmark of neoplastic progression, it is likely one of several essential features that underlie a malignantly proficient or aggressive tumorigenic phenotype. Here, we identified intercellular adhesion molecule-1 (ICAM-1) as a molecule with expression coordinately regulated with Fas and inversely correlated with malignant phenotype between matched pairs of differentially aggressive malignant subpopulations in three mouse models. To determine whether coordinate expression of Fas and ICAM-1 regulated malignant behavior, tumor sublines were produced that expressed either lower levels of both Fas and ICAM-1, lower levels of Fas, or lower levels of ICAM-1 and then assessed for metastatic lung tumor growth. Tumor sublines rendered both Fas incompetent and ICAM-1 incompetent displayed significantly higher numbers of tumor nodules compared with tumor sublines separately expressing low levels of Fas or ICAM-1. However, all tumor sublines regardless of their Fas and ICAM-1 levels comparably infiltrated the lung, suggesting that Fas- and ICAM-1–based interactions ultimately influenced lung colonization efficiency. Overall, these data suggested that both Fas and ICAM-1 pathways cooperated to regulate tumor progression and that the coordinate down-regulation of Fas and ICAM-1 intensified malignant progression at the level of colonization. Thus, a FasloICAM-1lo phenotype may be characteristic of at least certain advancing, immune-resistant neoplastic subpopulations.
Collapse
|
36
|
Cicinnati VR, Dworacki G, Albers A, Beckebaum S, Tüting T, Kaczmarek E, DeLeo AB. Impact of p53-based immunization on primary chemically-induced tumors. Int J Cancer 2004; 113:961-70. [PMID: 15514940 DOI: 10.1002/ijc.20686] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In mice as well as humans, cytotoxic T lymphocytes (CTL) specific for wild-type-sequence (wt) p53 peptides have been shown to react against a wide range of tumors, but not normal cells. As such, they are attractive candidates for developing broadly applicable cancer vaccines. Of particular interest is the potential of using p53-based vaccines in high-risk individuals to prevent cancer. Methylcholanthrene, an immunosuppressive polycyclic hydrocarbon carcinogen implicated as a causative agent in human cancers, has long been used to induce murine tumors with a high incidence of genetic alterations and sensitivity to wt p53-specific CTL. To analyze the potential of p53-based vaccines on primary tumors, we evaluated the efficacy of DNA and dendritic cell vaccines targeting wt p53 peptides given to methylcholanthrene-treated mice in the protection or therapy settings. The results indicate that the efficacy of these vaccines relative to reducing tumor incidence were severely compromised by vaccine-induced tumor escape. As compared to tumors induced in non-immunized mice, a higher incidence of epitope-loss tumors was detected in tumors from the immunized mice. The increase in tumor escape arose as a consequence of either increased frequencies of mutations within/flanking p53 epitope-coding regions or downregulation of expression of the major histocompatibility complex Class I molecules that present these epitopes for T cell recognition These findings are consistent with current views of immunoselection occurring in patients receiving tumor peptide-based immunotherapy, and impact on the design and implementation of p53-based vaccines, in particular, those aimed at treating individuals at high risk for developing cancer.
Collapse
Affiliation(s)
- Vito R Cicinnati
- Division of Basic Research, University of Pittsburgh Cancer Institute, Department of Pathology, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Daftarian P, Song GY, Ali S, Faynsod M, Longmate J, Diamond DJ, Ellenhorn JDI. Two distinct pathways of immuno-modulation improve potency of p53 immunization in rejecting established tumors. Cancer Res 2004; 64:5407-14. [PMID: 15289349 DOI: 10.1158/0008-5472.can-04-0169] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The p53 gene product is overexpressed by almost 50% of cancers, making it an ideal target for cancer immunotherapy. We previously demonstrated rejection of established p53-overexpressing tumors without stimulating autoimmunity by immunization with modified vaccinia Ankara-expressing murine p53 (MVAp53). Tumor rejection was enhanced through antibody-mediated CTL-associated antigen 4 (CTLA-4) blockade. We examined the role of synthetic oligodeoxynucleotides (ODN) containing unmethylated cytosine-phosphate-guanine (CpG) motifs (CpG ODN) in enhancing MVAp53-mediated tumor rejection. CpG ODN with MVAp53 resulted in tumor rejection in BALB/c mice bearing poorly immunogenic 11A-1 murine mammary carcinomas or Meth A sarcomas and C57Bl/6 mice bearing MC-38 colon carcinomas. The effect was similar to that seen in tumor-bearing mice treated with MVAp53 along with CTLA-4 blockade. Monoclonal antibody depletion experiments demonstrated that the adjuvant effects of CpG ODN and CTLA-4 blockades were CD8 dependent. CpG ODN were partially natural killer cell dependent and ineffective in Toll-like Receptor 9(-/-) and interleukin 6(-/-) mice, whereas CTLA-4 blockade was partially CD4 dependent and functional in Toll-like Receptor 9(-/-) and interleukin 6(-/-) mice. In addition, when administered with MVAp53, both adjuvants enhanced p53-specific cytotoxicity and demonstrated an additive effect when combined. The combination of CpG ODN and CTLA-4 blockade worked synergistically to reject palpable 11A-1 and MC-38 tumors. These experiments demonstrate the potential for augmenting MVAp53-mediated antitumor immunity using CpG ODN and CTLA-4 blockade. This cell-free immunotherapy approach is a candidate for evaluation in cancer patients.
Collapse
MESH Headings
- Adjuvants, Immunologic/pharmacology
- Adjuvants, Immunologic/therapeutic use
- Animals
- Antibodies, Monoclonal/metabolism
- Antigens, CD
- Antigens, Differentiation/chemistry
- CD8-Positive T-Lymphocytes/immunology
- CTLA-4 Antigen
- Cells, Cultured
- Colonic Neoplasms/genetics
- Colonic Neoplasms/immunology
- Colonic Neoplasms/therapy
- Combined Modality Therapy
- Cricetinae
- Cytotoxicity Tests, Immunologic
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/physiology
- Drug Synergism
- Female
- Homozygote
- Humans
- Immunization
- Interleukin-6/genetics
- Interleukin-6/physiology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/immunology
- Mammary Neoplasms, Animal/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Oligodeoxyribonucleotides/pharmacology
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/physiology
- Sarcoma, Experimental/genetics
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/therapy
- Signal Transduction
- Toll-Like Receptor 9
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/pharmacology
Collapse
Affiliation(s)
- Pirouz Daftarian
- Laboratory of Vaccine Research, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Norian LA, Allen PM. No intrinsic deficiencies in CD8+ T cell-mediated antitumor immunity with aging. THE JOURNAL OF IMMUNOLOGY 2004; 173:835-44. [PMID: 15240670 DOI: 10.4049/jimmunol.173.2.835] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Aging is associated with a decline in immune function, particularly within the T cell compartment. Because CD8(+) T cells are critical mediators of protective immunity against cancer, which arises more frequently with advancing age, it is important to understand how aging affects T cell-based antitumor responses. We used our DUC18 T cell/CMS5 tumor model system to examine the ability of both aged APCs and aged, tumor-specific CD8(+) T cells to mount protective responses to tumors in vivo. An assessment of aged DUC18 T cells in vitro showed a naive phenotype, but impaired proliferation in response to anti-CD3 and anti-CD28 stimulation. We found that DCs from young and old recipient mice are comparable phenotypically, and endogenous APCs in these mice are equally able to prime adoptively transferred young DUC18 T cells. Even when aged DUC18 T cells are transferred into aged CMS5-challenged mice, Ag-specific proliferation and CD25 expression are similar to those found when young DUC18 T cells are transferred into young mice. Although trafficking to tumor sites appears unequal, old and young DUC18 T cells reject primary CMS5 challenges to the same degree and with similar kinetics. Overall, we found no loss of endogenous APC function or intrinsic defects in CD8(+) DUC18 T cells with advanced age. Therefore, when young and old tumor-specific T cell populations are equivalently sized, CD8(+) T cell-mediated antitumor immunity in our system is not impaired by age, a finding that has positive implications for T cell-based immunotherapies.
Collapse
Affiliation(s)
- Lyse A Norian
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | |
Collapse
|
39
|
Hanson HL, Kang SS, Norian LA, Matsui K, O'Mara LA, Allen PM. CD4-directed peptide vaccination augments an antitumor response, but efficacy is limited by the number of CD8+ T cell precursors. THE JOURNAL OF IMMUNOLOGY 2004; 172:4215-24. [PMID: 15034034 DOI: 10.4049/jimmunol.172.7.4215] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Peptide vaccination is an immunotherapeutic strategy being pursued as a method of enhancing Ag-specific antitumor responses. To date, most studies have focused on the use of MHC class I-restricted peptides, and have not shown a correlation between Ag-specific CD8(+) T cell expansion and the generation of protective immune responses. We investigated the effects of CD4-directed peptide vaccination on the ability of CD8(+) T cells to mount protective antitumor responses in the DUC18/CMS5 tumor model system. To accomplish this, we extended the amino acid sequence of the known MHC class I-restricted DUC18 rejection epitope from CMS5 to allow binding to MHC class II molecules. Immunization with this peptide (tumor-derived extracellular signal-regulated kinase-II (tERK-II)) induced Ag-specific CD4(+) T cell effector function, but did not directly prime CD8(+) T cells. Approximately 31% of BALB/c mice immunized with tERK-II were protected from subsequent tumor challenge in a CD40-dependent manner. Priming of endogenous CD8(+) T cells in immunized mice was detected only after CMS5 challenge. Heightened CD4(+) Th cell function in response to tERK II vaccination allowed a 12-fold reduction in the number of adoptively transferred CD8(+) DUC18 T cells needed to protect recipients against tumor challenge as compared with previous studies using unimmunized mice. Furthermore, tERK-II immunization led to a more rapid and transient expansion of transferred DUC18 T cells than was seen in unimmunized mice. These findings illustrate that CD4-directed peptide vaccination augments antitumor immunity, but that the number of tumor-specific precursor CD8(+) T cells will ultimately dictate the success of immunotherapy.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Amino Acid Sequence
- Animals
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/immunology
- CD4 Antigens/administration & dosage
- CD4 Antigens/immunology
- CD40 Antigens/physiology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/transplantation
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cell Division/genetics
- Cell Division/immunology
- Cell Line, Tumor
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/immunology
- Female
- Fibrosarcoma/immunology
- Fibrosarcoma/pathology
- Fibrosarcoma/prevention & control
- Injections, Subcutaneous
- Interphase/genetics
- Interphase/immunology
- Lymphocyte Activation/immunology
- Lymphocyte Count
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mice, SCID
- Mice, Transgenic
- Molecular Sequence Data
- Neoplasm Transplantation
- Stem Cells/cytology
- Stem Cells/immunology
- T-Lymphocytes, Regulatory/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Holly L Hanson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
40
|
Zuberek K, Ling V, Wu P, Ma HL, Leonard JP, Collins M, Dunussi-Joannopoulos K. Comparable in vivo efficacy of CD28/B7, ICOS/GL50, and ICOS/GL50B costimulatory pathways in murine tumor models: IFNgamma-dependent enhancement of CTL priming, effector functions, and tumor specific memory CTL. Cell Immunol 2004; 225:53-63. [PMID: 14643304 DOI: 10.1016/j.cellimm.2003.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Increasing evidence suggests that B7/CD28 interactions are important in clonal expansion and effector function of nai;ve CD4(+) T cells, whereas ICOS/GL50 interactions may optimize the responses of recently activated T(H) cells. In tumor models, it has been shown that engagement of ICOS, like CD28, by its ligands can be effective in enhancing tumor immunity. In this report, we have directly compared the in vivo efficacy of CD28 vs ICOS activation in the MethA fibrosarcoma and B16F1 melanoma tumor models. We studied the efficacy of systemic treatment of tumors with murine B7.2-IgG or GL50-IgG fusion proteins, and the therapeutic potential of B7.1 or GL50 vaccines given during various phases of the antitumor responses. In addition, we compare the efficacy of ICOS-ligand splice variants GL50 and GL50B in promoting tumor immunity. We find that each of these pathways is equally effective in promoting tumor immunity and that the efficacy of both GL50 and B7.1 vaccines is IFN-gamma but not IL-10 dependent. Our results suggest that CD28 or ICOS costimulation-based strategies may be equally efficacious as adjuvants to conventional cancer treatment.
Collapse
MESH Headings
- Animals
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Differentiation, T-Lymphocyte/therapeutic use
- B7-1 Antigen/immunology
- B7-1 Antigen/therapeutic use
- CD28 Antigens/immunology
- CD28 Antigens/therapeutic use
- Cell Line, Tumor
- Enzyme-Linked Immunosorbent Assay
- Female
- Flow Cytometry
- Immunologic Memory/immunology
- Inducible T-Cell Co-Stimulator Ligand
- Inducible T-Cell Co-Stimulator Protein
- Interferon-gamma/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, SCID
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/therapy
- Proteins/immunology
- Proteins/therapeutic use
- Recombinant Proteins/immunology
- Recombinant Proteins/therapeutic use
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Krystyna Zuberek
- Wyeth Research, 200 Cambridgepark Drive, Cambridge, MA 02140, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
O'Mara LA, Allen PM. Pulmonary Tumors Inefficiently Prime Tumor-Specific T Cells. THE JOURNAL OF IMMUNOLOGY 2003; 172:310-7. [PMID: 14688339 DOI: 10.4049/jimmunol.172.1.310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The lung is a common site of metastatic and primary tumor growth, and has been shown to be an immunosuppressive environment. We tested the impact of the lung environment on the development of tumor-specific T cell responses against the CMS5 fibrosarcoma, and found a deficit in the efficacy of naive tumor-specific DUC18 T cells against tumors established in the lung. One hundred-fold more naive tumor-specific T cells were required to protect against tumor development or reject established tumors in the lung than an identical tumor challenge delivered s.c. in the flank. Importantly, CMS5 growing in the flank facilitated the rejection of tumors present in the lungs. In the presence of flank tumors, transferred T cells were not phenotypically altered but were present in much greater numbers in the parabronchial lymph nodes, bronchoalveolar lavage, and lung parenchyma than in mice bearing lung tumors alone. We hypothesized that APC present in the lung and skin draining lymph nodes were differentially initiating T cell proliferation, leading to differences in the size of the final effector populations. A direct comparison of DUC18 T cell proliferation against APC from flank or lung draining lymph nodes showed profoundly greater proliferation to flank draining lymph node APC. The impaired stimulation of naive T cell proliferation by lung draining APC provides one mechanistic explanation for the lower overall immune response, and inability to effectively reject tumors, in the lung.
Collapse
Affiliation(s)
- Leigh A O'Mara
- Department of Pathology and Immunology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | | |
Collapse
|
42
|
Uenaka A, Nakayama E. Murine leukemia RL male 1 and sarcoma Meth A antigens recognized by cytotoxic T lymphocytes (CTL). Cancer Sci 2003; 94:931-6. [PMID: 14611667 PMCID: PMC11160269 DOI: 10.1111/j.1349-7006.2003.tb01380.x] [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: 07/29/2003] [Revised: 09/02/2003] [Accepted: 09/05/2003] [Indexed: 11/29/2022] Open
Abstract
Peptide elution and expression cloning methods have been used to identify T cell-recognized antigens for which no molecular information is available. We identified a unique tumor antigen peptide pRL1a, IPGLPLSL that is recognized by CTL on BALB/c RL male 1 leukemia by peptide elution. The sequence of the peptide corresponded to the normally untranslated 5' region of akt. Cytotoxicity was generated in BALB/c spleen cells by in vivo and in vitro sensitization with pRL1a peptide in the form of multiple antigen peptide (MAP), but not the original form. pRL1a MAP immunization had a significant growth-inhibitory effect. pRL1a MAP was mostly internalized into the endosomal compartment of antigen-presenting cells, leaked to the cytosol, and degraded, and the pRL1a peptide produced was presented through the MHC class I pathway. In vivo depletion of CD4 T cells from tumor-inoculated BALB/c mice caused RL male 1 regression. Overexpression of the RLakt molecule seemed to induce CD4 immunoregulatory cells, which resulted in progressive RL male 1 growth in BALB/c mice. In vivo administration of anti-CD25 mAb (PC61) caused regression of RL male 1, suggesting that CD4(+) CD25(+) immunoregulatory cells were involved in the tumor growth. Recently, we improved the sensitivity and the efficacy of T cell antigen cloning from cDNA expression libraries by using large- and small-scale ELISPOT assays. Using the IFN-gamma ELISPOT method, we obtained a cDNA clone S35 of 937 bp recognized by AT-1 CTL on BALB/c Meth A sarcoma. S35 was a part of the retinoic acid-regulated nuclear matrix-associated protein (ramp). AT-1 CTL recognized the peptide LGAEAIFRL, which was derived from a newly created open reading frame due to the exon 14 extension.
Collapse
Affiliation(s)
- Akiko Uenaka
- Department of Immunology, Okayama University Graduate School of Medicine and Dentistry, Okayama 700-8558.
| | | |
Collapse
|
43
|
Nishikawa H, Kato T, Tanida K, Hiasa A, Tawara I, Ikeda H, Ikarashi Y, Wakasugi H, Kronenberg M, Nakayama T, Taniguchi M, Kuribayashi K, Old LJ, Shiku H. CD4+ CD25+ T cells responding to serologically defined autoantigens suppress antitumor immune responses. Proc Natl Acad Sci U S A 2003; 100:10902-6. [PMID: 12947044 PMCID: PMC196900 DOI: 10.1073/pnas.1834479100] [Citation(s) in RCA: 125] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
A variety of tumor-derived antigens have been defined by IgG antibodies in tumor bearers' sera with serological identification of antigens by recombinant expression cloning (SEREX), a serological expression cloning method. The majority of these antigens show no structural abnormality and seem to be wild-type autoantigens. Coimmunization with DNA encoding these autoantigens and tumor-specific cytotoxic T lymphocytes epitopes heightened CD8+ T cell responses and increased resistance to tumor challenge in a CD4+ T cell-dependent manner. In contrast, immunization with these SEREX-defined autoantigens alone leads to heightened susceptibility to tumor challenge. This suppressive effect of immunization is mediated by CD4+ CD25+ T cells. In mice immunized with one of the SEREX-defined autoantigens, Dna J-like 2, the number of alpha-GalCer/CD1d tetramer+ CD3+ T cells [representing natural killer T (NKT) cells] was reduced in the pulmonary compartment, whereas no evident change in the number of other T cell subsets was observed. Experiments with Jalpha281-/- mice lacking most NKT cells indicate that NKT cells are primarily responsible for metastasis suppression and that their activity is inhibited by immunization with Dna J-like 2. We propose that SEREX identifies a pool of autoantigens that maintains and regulates immunological homeostasis via CD4+ CD25+ regulatory T cells.
Collapse
Affiliation(s)
- Hiroyoshi Nishikawa
- Second Department of Internal Medicine, Mie University School of Medicine, Mie 514-8507, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Caldwell SA, Ryan MH, McDuffie E, Abrams SI. The Fas/Fas ligand pathway is important for optimal tumor regression in a mouse model of CTL adoptive immunotherapy of experimental CMS4 lung metastases. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:2402-12. [PMID: 12928387 DOI: 10.4049/jimmunol.171.5.2402] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The mechanisms of CTL-mediated tumor regression in vivo remain to be fully understood. If CTL do mediate tumor regression in vivo by direct cytotoxicity, this may occur via two major effector mechanisms involving the secretion of perforin/granzymes and/or engagement of Fas by Fas ligand (FasL) expressed by the activated CTL. Although the perforin pathway has been considered the dominant player, it is unclear whether Fas-mediated cytotoxicity is additionally required for optimal tumor rejection. Previously, we produced H-2L(d)-restricted CTL reactive against the CMS4 sarcoma, which expresses a naturally occurring rejection Ag recognized by these CTL and harbors a cytokine (IFN-gamma plus TNF)-inducible, Fas-responsive phenotype. The adoptive transfer of these CTL to syngeneic BALB/c mice with minimal (day 3 established) or extensive (day 10 established) experimental pulmonary metastases resulted in strong antitumor responses. Here we investigated whether a FasL-dependent CTL effector mechanism was important for optimal tumor regression in this adoptive immunotherapy model. The approach taken was to compare the therapeutic efficacy of wild-type to FasL-deficient (gld) CTL clones by adoptive transfer. In comparison with wild-type CTL, gld-CTL efficiently mediated tumor cytolysis and produced comparable amounts of IFN-gamma, after tumor-specific stimulation, as in vitro assessments of Ag recognition. Moreover, gld-CTL mediated comparably potent antitumor effects in a minimal disease setting, but were significantly less effective under conditions of an extensive tumor burden. Overall, under conditions of extensive lung metastases, these data revealed for the first time an important role for a FasL-dependent CTL effector mechanism in optimal tumor regression.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Clone Cells
- Cytotoxicity, Immunologic/genetics
- Disease Models, Animal
- Fas Ligand Protein
- Female
- Immunotherapy, Adoptive/methods
- Injections, Intravenous
- Ligands
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Lymphocyte Activation/genetics
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Neoplasm Transplantation
- Perforin
- Pore Forming Cytotoxic Proteins
- Sarcoma, Experimental/genetics
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/secondary
- Sarcoma, Experimental/therapy
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/transplantation
- fas Receptor/biosynthesis
- fas Receptor/physiology
Collapse
Affiliation(s)
- Sheila A Caldwell
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
45
|
Busund LTR, Killie MK, Bartnes K, Seljelid R. Spontaneously formed tumorigenic hybrids of Meth A sarcoma cells and macrophages in vivo. Int J Cancer 2003; 106:153-9. [PMID: 12800188 DOI: 10.1002/ijc.11210] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We have recently demonstrated that malignant cells can hybridize with tissue macrophages in vitro, giving rise to tumorigenic hybrids. We now demonstrate that this can occur spontaneously in vivo as a result of fusion between inoculated Meth A sarcoma cells and host cells, presumably macrophages. Thus, from tumor cell suspensions prepared by collagenase perfusion and density centrifugation, hybrid cells could be isolated that were neoplastic but in contrast to Meth A expressed macrophage markers and had phagocytic capacity. Their morphologic features were intermediate between Meth A and macrophages. By taking advantage of a semiallogeneic experimental system by inoculation of Meth A cells from BALB/c (H-2 K(d)) into (BALB.K x BALB/c) F(1) (H-2(k/d)), hybrid cells from these tumors could be shown to express MHC antigens of both the Meth A and the host haplotypes. Hybrid cells grew faster than Meth A cells in vivo, indicating acquisition of growth-promoting properties through heterotypic cell fusion.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/analysis
- Cell Division
- Chromosomes/genetics
- Female
- Hybrid Cells/pathology
- Macrophages/pathology
- Methylcholanthrene
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Microscopy, Electron, Scanning
- Phagocytosis
- Sarcoma, Experimental/chemically induced
- Sarcoma, Experimental/pathology
- Sarcoma, Experimental/ultrastructure
- Transplantation, Homologous
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Lill-Tove R Busund
- Department of Morphology, Institute of Medical Biology, University of Tromsø, Tromsø, Norway.
| | | | | | | |
Collapse
|
46
|
Matsui K, O'Mara LA, Allen PM. Successful elimination of large established tumors and avoidance of antigen-loss variants by aggressive adoptive T cell immunotherapy. Int Immunol 2003; 15:797-805. [PMID: 12807818 DOI: 10.1093/intimm/dxg078] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Utilization of ex vivo-expanded epitope-specific cytotoxic T lymphocytes has become a clinical standard in the adoptive immunotherapy of tumors. One of the obstacles faced by T cell-based immunotherapy is the development of tumor immune-escape variants. Using our previously reported CMS5 tumor/DUC18 CD8(+) TCR transgenic system, we sought to investigate whether large established tumors can be successfully eliminated before the development of escape variants. Using BALB/c mice that were s.c. transplanted with two tumors that had been growing for 8 days (double 8-day tumors), we assessed the in vivo anti-tumor activity of in vitro peptide-stimulated DUC18 T cells. A single infusion of activated DUC18 T cells showed a modest effect against the double 8-day tumors, whereas two and three administrations led to regression of both tumors within 10 days. However, in some mice, the tumors re-grew approximately 10 days after the regression. We found these tumors to be antigen-loss variants. These relapsed tumor cells progressively grew in DUC18 transgenic mice and did not express tERK-specific message. When four doses of activated DUC18 T cells were infused, the double 8-day tumors were successfully eliminated and the tumors did not grow out in any mice. Our results demonstrate that mono-specific CD8(+) T cells can effectively eliminate large established tumors before the development of antigen-loss variants when a high number of T cells is rapidly administered.
Collapse
MESH Headings
- Animals
- Antigenic Variation
- Antigens, Neoplasm/analysis
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Apoptosis
- Cell Line, Tumor
- Epitopes
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Genetic Variation
- Immunotherapy, Adoptive
- Lymphocyte Activation
- Mice
- Mice, Inbred BALB C
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/therapy
- Peptides/genetics
- Peptides/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/transplantation
Collapse
Affiliation(s)
- Ken Matsui
- Department of Pathology and Immunology, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO 63110, USA
| | | | | |
Collapse
|
47
|
Liu K, Abrams SI. Alterations in Fas expression are characteristic of, but not solely responsible for, enhanced metastatic competence. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:5973-80. [PMID: 12794124 DOI: 10.4049/jimmunol.170.12.5973] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dysregulation of the Fas pathway has been implicated in tumor progression; however, how alterations in Fas expression influence metastatic behavior remains unresolved. In this study, we investigated the link between Fas expression and metastatic capacity in two mouse tumor models: one was a sarcoma, which was used to analyze the consequences of loss of Fas function in experimental pulmonary metastases, and the other was a mammary carcinoma, where Fas expression was examined in matched pairs of primary and metastatic cell lines as well as by immunohistochemistry of tissues taken from primary and metastatic sites of spontaneous tumor development. In the sarcoma model, a Fas-resistant/refractory subline was produced in vitro from the parental line by biologic selection against Fas-responsive cells, and it was then compared with the poorly metastatic parental line and to an in vivo-derived subline that was highly metastatic for growth in the lungs. In both tumor models, an inverse correlation was demonstrated between Fas expression and metastatic phenotype. Subsequent studies in the sarcoma model revealed that although the Fas-resistant/refractory subline displayed significant metastatic ability, the parental line from which it was derived exhibited little to no additional metastatic activity if experimentally rendered Fas-resistant by molecular-based strategies or transplanted into a Fas ligand-deficient host. Therefore, these findings suggested that down-regulation of Fas was associated with the metastatic phenotype, but alterations in Fas expression alone were insufficient for acquisition of full metastatic potential. Rather, the ability of such Fas-resistant neoplastic subpopulations to achieve metastatic competence apparently required co-possession of additional malignant characteristics.
Collapse
MESH Headings
- Adjuvants, Immunologic/biosynthesis
- Adjuvants, Immunologic/genetics
- Adjuvants, Immunologic/metabolism
- Adjuvants, Immunologic/physiology
- Animals
- Apoptosis/genetics
- Apoptosis/immunology
- Carcinoma/genetics
- Carcinoma/immunology
- Carcinoma/pathology
- Carcinoma/secondary
- Disease Progression
- Fas Ligand Protein
- Female
- Gene Expression Regulation, Neoplastic/immunology
- Ligands
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/secondary
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasm Transplantation/immunology
- Phenotype
- Sarcoma, Experimental/genetics
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/pathology
- Sarcoma, Experimental/secondary
- Tumor Cells, Cultured
- fas Receptor/biosynthesis
- fas Receptor/genetics
- fas Receptor/metabolism
- fas Receptor/physiology
Collapse
Affiliation(s)
- Kebin Liu
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
48
|
Uenaka A, Hirano Y, Hata H, Win S, Aji T, Tanaka M, Ono T, Skipper JCA, Shimizu K, Nakayama E. Cryptic CTL epitope on a murine sarcoma Meth A generated by exon extension as a novel mechanism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4862-8. [PMID: 12707369 DOI: 10.4049/jimmunol.170.9.4862] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Using the recently developed ELISPOT cloning methodology, we obtained cDNA clone S35 coding for the Ag epitope recognized by a murine sarcoma Meth A-specific CTL clone AT-1. Analysis of truncated S35 constructs and overlapping peptides revealed that the peptide epitope was LGAEAIFRL. AT-1 CTL lysed peptide-pulsed CMS8 cells at a nanomolar concentration, and the peptide strongly stimulated IFN-gamma production in AT-1 CTL. Sequence homology indicated that the S35 was derived from a mouse homologue of human retinoic acid-regulated nuclear matrix-associated protein (ramp). The ramp gene consisted of 15 exons. The majority of the ramp mRNA was the transcript normally spliced between exons 14 and 15, but a minor population of mRNA with an extended exon 14 was also present in Meth A cells. The epitope was derived from the newly created open reading frame, which resulted from extension of exon 14 after splicing of the adjacent intronic sequence.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/isolation & purification
- Antigens, Neoplasm/metabolism
- Base Sequence
- Carrier Proteins/biosynthesis
- Carrier Proteins/genetics
- Carrier Proteins/isolation & purification
- Carrier Proteins/metabolism
- Clone Cells
- Cloning, Molecular
- Cytotoxicity Tests, Immunologic
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/isolation & purification
- DNA-Binding Proteins/metabolism
- Enzyme-Linked Immunosorbent Assay
- Epitopes, T-Lymphocyte/biosynthesis
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/isolation & purification
- Epitopes, T-Lymphocyte/metabolism
- Exons/genetics
- Exons/immunology
- Gene Library
- Humans
- Interferon-gamma/analysis
- Methylcholanthrene
- Mice
- Mice, Inbred BALB C
- Mice, Inbred DBA
- Molecular Sequence Data
- Sarcoma, Experimental/chemically induced
- Sarcoma, Experimental/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Transcription Factors
Collapse
Affiliation(s)
- Akiko Uenaka
- Department of Immunology, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Espenschied J, Lamont J, Longmate J, Pendas S, Wang Z, Diamond DJ, Ellenhorn JDI. CTLA-4 blockade enhances the therapeutic effect of an attenuated poxvirus vaccine targeting p53 in an established murine tumor model. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:3401-7. [PMID: 12626601 DOI: 10.4049/jimmunol.170.6.3401] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
p53 is overexpressed by half of all cancers, and is an attractive target for a vaccine approach to immunotherapy. p53 overexpression is frequently the result of point mutations, which leaves the majority of the protein in its wild-type form. Therefore, the majority of p53 sequence is wild type, making it a self-protein for which tolerance plays a role in limiting immune responses. To overcome tolerance to p53, we have expressed wild-type murine p53 in the nonpathogenic attenuated poxvirus, modified vaccinia virus Ankara (recombinant modified vaccinia virus Ankara expressing wild-type murine p53 (rMVAp53)). Mice immunized with rMVAp53 vaccine developed vigorous p53-specific CTL responses. rMVAp53 vaccine was evaluated for its ability to inhibit the outgrowth of the syngeneic murine sarcoma Meth A, which overexpresses mutant p53. Mice were inoculated with a lethal dose (5 x 10(5) cells injected s.c.) of Meth A tumor cells and vaccinated by i.p. injection 3 days later with 5 x 10(7) PFU of rMVAp53. The majority of mice remained tumor free and resistant to rechallenge with Meth A tumor cells. We wished to determine whether rMVAp53 immunization could effect the rejection of an established, palpable Meth A tumor. In subsequent experiments, mice were injected with 10(6) Meth A tumor cells, and treated 6 days later with anti-CTLA-4 Ab (9H10) and rMVAp53. The majority of treated mice had complete tumor regression along with lasting tumor immunity. In vivo Ab depletion confirmed that the antitumor effect was primarily CD8 and to a lesser extent CD4 dependent. These experiments demonstrate the potential of a novel cell-free vaccine targeting p53 in malignancy.
Collapse
MESH Headings
- Abatacept
- Adjuvants, Immunologic/pharmacology
- Animals
- Antibodies, Blocking/pharmacology
- Antigens, CD
- Antigens, Differentiation/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- CTLA-4 Antigen
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Cell Line
- Cricetinae
- Female
- Fibrosarcoma/immunology
- Fibrosarcoma/mortality
- Fibrosarcoma/prevention & control
- Genetic Vectors
- Humans
- Immunoconjugates
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Interferon-gamma/physiology
- Killer Cells, Natural/immunology
- Lymphocyte Depletion
- Methylcholanthrene
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/mortality
- Sarcoma, Experimental/prevention & control
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Cells, Cultured
- Tumor Suppressor Protein p53/biosynthesis
- Tumor Suppressor Protein p53/genetics
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/therapeutic use
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/therapeutic use
- Vaccinia virus/genetics
- Vaccinia virus/immunology
- Viral Vaccines/genetics
- Viral Vaccines/immunology
- Viral Vaccines/therapeutic use
Collapse
Affiliation(s)
- Jonathan Espenschied
- Division of General and Oncologic Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Ara G, Baher A, Storm N, Horan T, Baikalov C, Brisan E, Camacho R, Moore A, Goldman H, Kohno T, Cattley RC, Van G, Gaida K, Zhang M, Whoriskey JS, Fong D, Yoshinaga SK. Potent activity of soluble B7RP-1-Fc in therapy of murine tumors in syngeneic hosts. Int J Cancer 2003; 103:501-7. [PMID: 12478666 DOI: 10.1002/ijc.10831] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
We have characterized a receptor:ligand pair, ICOS:B7RP-1, that is structurally and functionally related to CD28:B7.1/2. We reported previously that B7RP-1 costimulates T cell proliferation and immune responses (Yoshinaga et al., Nature 1999;402:827-32; Guo et al., J Immunol 2001;166:5578-84; Yoshinaga et al., Int Immunol 2000;12:1439-47). We report that B7RP-1-Fc causes rejection or growth inhibition of Meth A, SA-1 and EMT6 tumors in syngeneic mice. Established Meth A tumors were rejected effectively with a single dose of B7RP-1-Fc, however, the treatment was less effective on larger tumors. Mice that rejected Meth A tumors previously by Day 30, also rejected a subsequent Meth A challenge on Day 60, without additional B7RP-1-Fc treatment, indicating a long-lived memory response. Tumor cells believed to be less immunogenic, such as P815 and EL-4 cells, were less responsive to this treatment. The EL-4 responsiveness to the B7RP-1-Fc treatment was enhanced, however, by pre-treatment of the mice with cyclophosphamide. As expected, T cells appeared to be targeted by B7RP-1-Fc treatment. Thus, the administration of soluble B7RP-1-Fc may have therapeutic value in generating or enhancing anti-tumor activity in a clinical setting.
Collapse
Affiliation(s)
- Gulshan Ara
- Department of Pharmacology, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|