1
|
Eke I, Aryankalayil MJ, Bylicky MA, Sandfort V, Vanpouille-Box C, Nandagopal S, Graves EE, Giaccia AJ, Coleman CN. Long-term expression changes of immune-related genes in prostate cancer after radiotherapy. Cancer Immunol Immunother 2022; 71:839-850. [PMID: 34435232 PMCID: PMC8873240 DOI: 10.1007/s00262-021-03036-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 08/16/2021] [Indexed: 01/14/2023]
Abstract
The expression of immune-related genes in cancer cells can alter the anti-tumor immune response and thereby impact patient outcomes. Radiotherapy has been shown to modulate immune-related genes dependent on the fractionation regimen. To identify long-term changes in gene expression after irradiation, PC3 (p53 deleted) and LNCaP (p53 wildtype) prostate cancer cells were irradiated with either a single dose (SD, 10 Gy) or a fractionated regimen (MF) of 10 fractions (1 Gy per fraction). Whole human genome arrays were used to determine gene expression at 24 h and 2 months after irradiation. Immune pathway activation was analyzed with Ingenuity Pathway Analysis software. Additionally, 3D colony formation assays and T-cell cytotoxicity assays were performed. LNCaP had a higher basal expression of immunogenic genes and was more efficiently killed by cytotoxic T-cells compared to PC3. In both cell lines, MF irradiation resulted in an increase in multiple immune-related genes immediately after irradiation, while at 2 months, SD irradiation had a more pronounced effect on radiation-induced gene expression. Both immunogenic and immunosuppressive genes were upregulated in the long term in PC3 cells by a 10 Gy SD irradiation but not in LNCaP. T-cell-mediated cytotoxicity was significantly increased in 10 Gy SD PC3 cells compared to the unirradiated control and could be further enhanced by treatment with immune checkpoint inhibitors. Irradiation impacts the expression of immune-related genes in cancer cells in a fractionation-dependent manner. Understanding and targeting these changes may be a promising strategy for primary prostate cancer and recurrent tumors.
Collapse
Affiliation(s)
- Iris Eke
- Department of Radiation Oncology, Center for Clinical Sciences Research (CCSR), Stanford University School of Medicine, 269 Campus Dr., Room 1260, Stanford, CA, 94305, USA.
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Molykutty J Aryankalayil
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Michelle A Bylicky
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Veit Sandfort
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | | | - Saravanan Nandagopal
- Department of Radiation Oncology, Center for Clinical Sciences Research (CCSR), Stanford University School of Medicine, 269 Campus Dr., Room 1260, Stanford, CA, 94305, USA
| | - Edward E Graves
- Department of Radiation Oncology, Center for Clinical Sciences Research (CCSR), Stanford University School of Medicine, 269 Campus Dr., Room 1260, Stanford, CA, 94305, USA
| | - Amato J Giaccia
- Department of Radiation Oncology, Center for Clinical Sciences Research (CCSR), Stanford University School of Medicine, 269 Campus Dr., Room 1260, Stanford, CA, 94305, USA
- Oxford Institute of Radiation Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX37DQ, UK
| | - C Norman Coleman
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Radiation Research Program, National Cancer Institute, National Institutes of Health, Rockville, MD, 20850, USA
| |
Collapse
|
2
|
The lncRNAs LINC00261 and LINC00665 are upregulated in long-term prostate cancer adaptation after radiotherapy. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 24:175-187. [PMID: 33767914 PMCID: PMC7960506 DOI: 10.1016/j.omtn.2021.02.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 02/19/2021] [Indexed: 12/17/2022]
Abstract
Long non-coding RNAs (lncRNAs) have been shown to impact important biological functions such as proliferation, survival, and genomic stability. To analyze radiation-induced lncRNA expression in human tumors, we irradiated prostate cancer cells with a single dose of 10 Gy or a multifractionated radiotherapeutic regimen of 10 fractions with a dose of 1 Gy (10 × 1 Gy) during 5 days. We found a stable upregulation of the lncRNA LINC00261 and LINC00665 at 2 months after radiotherapy that was more pronounced after single-dose irradiation. Analysis of the The Cancer Genome Atlas (TCGA) and The Atlas of Non-coding RNAs in Cancer (TANRIC) databases showed that high expression of these two lncRNAs may be a potential negative prognostic marker for overall survival of prostate cancer patients. Knockdown of LINC00261 and LINC00665 in long-term survivors decreased survival after re-irradiation and affected DNA double-strand break repair. Mechanistically, both lncRNAs showed an interdependent expression and regulated expression of the DNA repair proteins CtIP (RBBP8) and XPC as well as the microRNA miR-329. Identifying long-term tumor adaptation mechanisms can lead to the discovery of new molecular targets, in effect opening up new research directions and improving multimodal radiation oncologic treatment.
Collapse
|
3
|
Development of potent class II transactivator gene delivery systems capable of inducing de novo MHC II expression in human cells, in vitro and ex vivo. Gene Ther 2017; 24:342-352. [PMID: 28414303 DOI: 10.1038/gt.2017.25] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 02/01/2017] [Accepted: 02/06/2017] [Indexed: 01/17/2023]
Abstract
Class II transactivator (CIITA) induces transcription of major histocompatibility complex (MHC) II genes and can potentially be used to improve genetic immunotherapies by converting non-immune cells into cells capable of presenting antigens to CD4+ T cells. However, CIITA expression is tightly controlled and it remains unclear whether distinct non-immune cells differ in this transactivator regulation. Here we describe the development of gene delivery systems capable of promoting the efficient CIITA expression in non-immune cell lines and in primary human cells of an ex vivo skin explant model. Different human cell types undergoing CIITA overexpression presented high-level de novo expression of MHC II, validating the delivery systems as suitable tools for the CIITA evaluation as a molecular adjuvant for gene therapies.
Collapse
|
4
|
Hillman GG, Reich LA, Rothstein SE, Abernathy LM, Fountain MD, Hankerd K, Yunker CK, Rakowski JT, Quemeneur E, Slos P. Radiotherapy and MVA-MUC1-IL-2 vaccine act synergistically for inducing specific immunity to MUC-1 tumor antigen. J Immunother Cancer 2017; 5:4. [PMID: 28116088 PMCID: PMC5240430 DOI: 10.1186/s40425-016-0204-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 12/12/2016] [Indexed: 11/30/2022] Open
Abstract
Background We previously demonstrated that tumor irradiation potentiates cancer vaccines using genetic modification of tumor cells in murine tumor models. To investigate whether tumor irradiation augments the immune response to MUC1 tumor antigen, we have tested the efficacy of tumor irradiation combined with an MVA-MUC1-IL2 cancer vaccine (Transgene TG4010) for murine renal adenocarcinoma (Renca) cells transfected with MUC1. Methods Established subcutaneous Renca-MUC1 tumors were treated with 8 Gy radiation on day 11 and peritumoral injections of MVA-MUC1-IL2 vector on day 12 and 17, or using a reverse sequence of vaccine followed by radiation. Growth delays were monitored by tumor measurements and histological responses were evaluated by immunohistochemistry. Specific immunity was assessed by challenge with Renca-MUC1 cells. Generation of tumor-specific T cells was detected by IFN-γ production from splenocytes stimulated in vitro with tumor lysates using ELISPOT assays. Results Tumor growth delays observed by tumor irradiation combined with MVA-MUC1-IL-2 vaccine were significantly more prolonged than those observed by vaccine, radiation, or radiation with MVA empty vector. The sequence of cancer vaccine followed by radiation two days later resulted in 55–58% complete responders and 60% mouse long-term survival. This sequence was more effective than that of radiation followed by vaccine leading to 24–30% complete responders and 30% mouse survival. Responding mice were immune to challenge with Renca-MUC1 cells, indicating the induction of specific tumor immunity. Histology studies of regressing tumors at 1 week after therapy, revealed extensive tumor destruction and a heavy infiltration of CD45+ leukocytes including F4/80+ macrophages, CD8+ cytotoxic T cells and CD4+ helper T cells. The generation of tumor-specific T cells by combined therapy was confirmed by IFN-γ secretion in tumor-stimulated splenocytes. An abscopal effect was measured by rejection of an untreated tumor on the contralateral flank to the tumor treated with radiation and vaccine. Conclusions These findings suggest that cancer vaccine given prior to local tumor irradiation augments an immune response targeted at tumor antigens that results in specific anti-tumor immunity. These findings support further exploration of the combination of radiotherapy with cancer vaccines for the treatment of cancer. Electronic supplementary material The online version of this article (doi:10.1186/s40425-016-0204-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gilda G Hillman
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Hudson Webber Cancer Research Center, room 515, 4100 John R, Detroit, MI 48201 USA ; Radiation Oncology Division, Immunology & Microbiology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI 48201 USA
| | - Lyndsey A Reich
- Radiation Oncology Division, Immunology & Microbiology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI 48201 USA
| | - Shoshana E Rothstein
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Hudson Webber Cancer Research Center, room 515, 4100 John R, Detroit, MI 48201 USA
| | - Lisa M Abernathy
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Hudson Webber Cancer Research Center, room 515, 4100 John R, Detroit, MI 48201 USA ; Radiation Oncology Division, Immunology & Microbiology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI 48201 USA ; Present address: Department of Microbiology and Immunology, Indiana University School of Medicine at Notre Dame, South Bend, IN 46617 USA
| | - Matthew D Fountain
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Hudson Webber Cancer Research Center, room 515, 4100 John R, Detroit, MI 48201 USA ; Radiation Oncology Division, Immunology & Microbiology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, MI 48201 USA
| | - Kali Hankerd
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Hudson Webber Cancer Research Center, room 515, 4100 John R, Detroit, MI 48201 USA
| | - Christopher K Yunker
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Hudson Webber Cancer Research Center, room 515, 4100 John R, Detroit, MI 48201 USA
| | - Joseph T Rakowski
- Department of Oncology, Wayne State University School of Medicine, Karmanos Cancer Institute, Hudson Webber Cancer Research Center, room 515, 4100 John R, Detroit, MI 48201 USA
| | - Eric Quemeneur
- Transgene SA, Parc d'Innovation, CS80166, 67405 Illkirch-Graffenstaden Cedex, France
| | - Philippe Slos
- Transgene SA, Parc d'Innovation, CS80166, 67405 Illkirch-Graffenstaden Cedex, France ; Present address: Oncodesign, 20, rue Jean Mazen, 21076 Dijon Cedex, France
| |
Collapse
|
5
|
Robson T, Worthington J, McKeown SR, Hirst DG. Radiogenic Therapy: Novel Approaches for Enhancing Tumor Radiosensitivity. Technol Cancer Res Treat 2016; 4:343-61. [PMID: 16029055 DOI: 10.1177/153303460500400404] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy (RT) is a well established modality for treating many forms of cancer. However, despite many improvements in treatment planning and delivery, the total radiation dose is often too low for tumor cure, because of the risk of normal tissue damage. Gene therapy provides a new adjunctive strategy to enhance the effectiveness of RT, offering the potential for preferential killing of cancer cells and sparing of normal tissues. This specificity can be achieved at several levels including restricted vector delivery, transcriptional targeting and specificity of the transgene product. This review will focus on those gene therapy strategies that are currently being evaluated in combination with RT, including the use of radiation sensitive promoters to control the timing and location of gene expression specifically within tumors. Therapeutic transgenes chosen for their radiosensitizing properties will also be reviewed, these include: gene correction therapy, in which normal copies of genes responsible for radiation-induced apoptosis are transfected to compensate for the deletions or mutated variants in tumor cells (p53 is the most widely studied example). enzymes that synergize the radiation effect, by generation of a toxic species from endogenous precursors ( e.g., inducible nitric oxide synthase) or by activation of non toxic prodrugs to toxic species ( e.g., herpes simplex virus thymidine kinase/ganciclovir) within the target tissue. conditionally replicating oncolytic adenoviruses that synergize the radiation effect. membrane transport proteins ( e.g., sodium iodide symporter) to facilitate uptake of cytotoxic radionuclides. The evidence indicates that many of these approaches are successful for augmenting radiation induced tumor cell killing with clinical trials currently underway.
Collapse
Affiliation(s)
- T Robson
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| | | | | | | |
Collapse
|
6
|
Heery CR, Hodge JW, Gulley JL. Combining radiation and therapeutic cancer vaccines: a synergistic approach. BREAST CANCER MANAGEMENT 2012. [DOI: 10.2217/bmt.12.52] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY The role of radiotherapy for prevention of recurrence of breast cancer is well established. Preclinical data indicate that the combination of therapeutic cancer vaccines and radiotherapy may be synergistic. Radiation can induce immunogenic cell death, or, at sublethal doses, immunogenic modulation in tumor cells, making them more amenable to T-cell-mediated death. Radiation also stimulates microenvironment effects that attract immune cells and improve their functional capacity. The capacity of radiation to induce the abscopal effect appears to be immune mediated and may be related to the other effects described. This phenomenon may indicate the capacity of radiation to induce antigen spreading, causing broader and deeper immune activation than a vaccine alone. This review discusses preclinical and clinical findings of radiation-induced immune modulation, preclinical evidence of synergy with vaccine therapy, and the rationale for clinical trials combining these treatment modalities in breast cancer.
Collapse
Affiliation(s)
- Christopher R Heery
- Laboratory of Tumor Immunology & Biology, National Cancer Institute, National Institutes of Health, Room 13N208, MSC-1750, 10 Center Drive, Building 10, Bethesda, MD 20892, USA
| | - James W Hodge
- Laboratory of Tumor Immunology & Biology, National Cancer Institute, National Institutes of Health, Room 13N208, MSC-1750, 10 Center Drive, Building 10, Bethesda, MD 20892, USA
| | - James L Gulley
- Laboratory of Tumor Immunology & Biology, National Cancer Institute, National Institutes of Health, Room 13N208, MSC-1750, 10 Center Drive, Building 10, Bethesda, MD 20892, USA
| |
Collapse
|
7
|
Gough MJ, Crittenden MR. Combination approaches to immunotherapy: the radiotherapy example. Immunotherapy 2011; 1:1025-37. [PMID: 20635917 DOI: 10.2217/imt.09.64] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The marquee cell of the immune response, the T lymphocyte, is exquisitely sensitive to ionizing radiation. Moreover, the majority of cells of the hematopoietic lineage, with their high turnover and radiation sensitivity, are effectively ablated by relatively low radiation doses, such that total-body irradiation is an effective immunosuppressive therapy. Despite and sometimes because of these facts, the immune system may underlie some portion of the therapeutic effects of radiation. In this review, we will discuss these processes, and go on to discuss current therapies combining radiation therapy with immunotherapy for the treatment of cancer.
Collapse
Affiliation(s)
- Michael J Gough
- Robert W Franz Cancer Center, Earle A Chiles Research Institute, Providence Portland Medical Center, Portland, OR 97213, USA.
| | | |
Collapse
|
8
|
Serduc R, Christen T, Laissue J, Farion R, Bouchet A, Sanden BVD, Segebarth C, Bräuer-Krisch E, Le Duc G, Bravin A, Rémy C, Barbier EL. Brain tumor vessel response to synchrotron microbeam radiation therapy: a short-termin vivostudy. Phys Med Biol 2008; 53:3609-22. [DOI: 10.1088/0031-9155/53/13/015] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
9
|
Rosenthal KS, Zimmerman DH. Vaccines: all things considered. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2006; 13:821-9. [PMID: 16893980 PMCID: PMC1539119 DOI: 10.1128/cvi.00152-06] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Ken S Rosenthal
- Northeastern Ohio Universities College of Medicine, 4209 SR 44, Rootstown, OH 44272, USA.
| | | |
Collapse
|
10
|
Lu X, Wu S, Blackwell CE, Humphreys RE, von Hofe E, Xu M. Suppression of major histocompatibility complex class II-associated invariant chain enhances the potency of an HIV gp120 DNA vaccine. Immunology 2006; 120:207-16. [PMID: 17116173 PMCID: PMC2265863 DOI: 10.1111/j.1365-2567.2006.02492.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Summary One function of the major histocompatibility complex (MHC) class II-associated invariant chain (Ii) is to prevent MHC class II molecules from binding endogenously generated antigenic epitopes. Ii inhibition leads to MHC class II presentation of endogenous antigens by APC without interrupting MHC class I presentation. We present data that in vivo immunization of BALB/c mice with HIV gp120 cDNA plus an Ii suppressive construct significantly enhances the activation of both gp120-specific T helper (Th) cells and cytotoxic T lymphocytes (CTL). Our results support the concept that MHC class II-positive/Ii-negative (class II(+)/Ii(-)) antigen-presenting cells (APC) present endogenously synthesized vaccine antigens simultaneously by MHC class II and class I molecules, activating both CD4(+) and CD8(+) T cells. Activated CD4(+) T cells locally strengthen the response of CD8(+) CTL, thus enhancing the potency of a DNA vaccine.
Collapse
Affiliation(s)
- Xueqing Lu
- Antigen Express, Inc., Worcester, MA 01606-2758, USA
| | | | | | | | | | | |
Collapse
|
11
|
Thompson JA, Dissanayake SK, Ksander BR, Knutson KL, Disis ML, Ostrand-Rosenberg S. Tumor cells transduced with the MHC class II Transactivator and CD80 activate tumor-specific CD4+ T cells whether or not they are silenced for invariant chain. Cancer Res 2006; 66:1147-54. [PMID: 16424052 DOI: 10.1158/0008-5472.can-05-2289] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The specificity and potency of the immune system make immunotherapy a potential strategy for the treatment of cancer. To exploit this potential, we have developed cell-based cancer vaccines consisting of tumor cells expressing syngeneic MHC class II and costimulatory molecules. The vaccines mediate tumor regression in mice and activate human CD4+ T cells in vitro. Previous vaccines were generated by transducing MHC II negative tumor cells with a single HLA-DR allele. Because expression of multiple MHC II alleles would facilitate presentation of a broader repertoire of tumor antigens, we have now transduced tumor cells with the MHC class II transactivator (CIITA), a regulatory gene that coordinately increases expression of all MHC II alleles. Previous studies in mice indicated that coexpression of the MHC II accessory molecule invariant chain (Ii) inhibited presentation of endogenously synthesized tumor antigens and reduced vaccine efficacy. To determine if Ii expression affects presentation of MHC class II-restricted endogenously synthesized tumor antigens in human tumor cells, HLA-DR-MCF10 breast cancer cells were transduced with the CIITA, CD80 costimulatory molecule gene, and with or without small interfering RNAs (siRNA) specific for Ii. Ii expression is silenced >95% in CIITA/CD80/siRNA transductants; down-regulation of Ii does not affect HLA-DR expression or stability; and Ii(+) and Ii(-) transductants activate human CD4+ T cells to DRB1*0701-restricted HER-2/neu epitopes. Therefore, tumor cells transduced with the CIITA, CD80, and with or without Ii siRNA present endogenously synthesized tumor antigens and are potential vaccines for activating tumor-specific CD4+ T cells.
Collapse
Affiliation(s)
- James A Thompson
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD 21250, USA
| | | | | | | | | | | |
Collapse
|
12
|
Chakravarty PK, Guha C, Alfieri A, Beri V, Niazova Z, Deb NJ, Fan Z, Thomas EK, Vikram B. Flt3L Therapy following Localized Tumor Irradiation Generates Long-Term Protective Immune Response in Metastatic Lung Cancer: Its Implication in Designing a Vaccination Strategy. Oncology 2006; 70:245-54. [PMID: 17047396 DOI: 10.1159/000096288] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2005] [Accepted: 05/15/2006] [Indexed: 11/19/2022]
Abstract
Flt3 ligand (Flt3L) therapy that expands dendritic cells in vivo in combination with local tumor radiotherapy (RT) significantly improved survival and induced a long-term tumor-specific immune response in a murine model of Lewis lung carcinoma (3LL). The irradiated tumor cells were able to significantly restimulate the splenocytes of the RT + Flt3L cohort in vitro. The restimulated splenocytes demonstrated increased cytotoxic response, lymphocytic proliferation and elevated levels of Th type I cytokines (IL-2, IL-12, IFN-gamma and TNF-alpha). The combination therapy of RT + Flt3L induced a long-term protective immunity in the disease-free animals. The protective effect was further enhanced when the disease-free animals were vaccinated with irradiated tumor cells. The vaccinated animals had significantly greater protection compared to the nonvaccinated group against subsequent challenge with 3LL cells. Taken together, these results indicate that the release of tumor antigens by irradiated dying tumors and concomitant administration of Flt3L was able to facilitate the generation of a tumor-specific long-term immune response against a poorly immunogenic tumor. This effect was further boosted by vaccination with irradiated tumor cells.
Collapse
Affiliation(s)
- Prabir K Chakravarty
- Department of Radiation Oncology, Albert Einstein College of Medicine and Montefiore Medical Center, New York, NY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Reith W, LeibundGut-Landmann S, Waldburger JM. Regulation of MHC class II gene expression by the class II transactivator. Nat Rev Immunol 2005; 5:793-806. [PMID: 16200082 DOI: 10.1038/nri1708] [Citation(s) in RCA: 327] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
MHC class II molecules are pivotal for the adaptive immune system, because they guide the development and activation of CD4+ T helper cells. Fulfilling these functions requires that the genes encoding MHC class II molecules are transcribed according to a strict cell-type-specific and quantitatively modulated pattern. This complex gene-expression profile is controlled almost exclusively by a single master regulatory factor, which is known as the class II transactivator. As we discuss here, differential activation of the three independent promoters that drive expression of the gene encoding the class II transactivator ultimately determines the exquisitely regulated pattern of MHC class II gene expression.
Collapse
Affiliation(s)
- Walter Reith
- Department of Pathology and Immunology, University of Geneva Medical School, Centre Médical Universitaire, 1 Rue Michel-Servet, CH-1211, Geneva, Switzerland.
| | | | | |
Collapse
|