1
|
Martín-Otal C, Lasarte-Cia A, Serrano D, Casares N, Conde E, Navarro F, Sánchez-Moreno I, Gorraiz M, Sarrión P, Calvo A, De Andrea CE, Echeveste J, Vilas A, Rodriguez-Madoz JR, San Miguel J, Prosper F, Hervas-Stubbs S, Lasarte JJ, Lozano T. Targeting the extra domain A of fibronectin for cancer therapy with CAR-T cells. J Immunother Cancer 2022; 10:jitc-2021-004479. [PMID: 35918123 PMCID: PMC9351345 DOI: 10.1136/jitc-2021-004479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2022] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND One of the main difficulties of adoptive cell therapies with chimeric antigen receptor (CAR)-T cells in solid tumors is the identification of specific target antigens. The tumor microenvironment can present suitable antigens for CAR design, even though they are not expressed by the tumor cells. We have generated a CAR specific for the splice variant extra domain A (EDA) of fibronectin, which is highly expressed in the tumor stroma of many types of tumors but not in healthy tissues. METHODS EDA expression was explored in RNA-seq data from different human tumor types and by immunohistochemistry in paraffin-embedded tumor biopsies. Murine and human anti-EDA CAR-T cells were prepared using recombinant retro/lentiviruses, respectively. The functionality of EDA CAR-T cells was measured in vitro in response to antigen stimulation. The antitumor activity of EDA CAR-T cells was measured in vivo in C57BL/6 mice challenged with PM299L-EDA hepatocarcinoma cell line, in 129Sv mice-bearing F9 teratocarcinoma and in NSG mice injected with the human hepatocarcinoma cell line PLC. RESULTS EDA CAR-T cells recognized and killed EDA-expressing tumor cell lines in vitro and rejected EDA-expressing tumors in immunocompetent mice. Notably, EDA CAR-T cells showed an antitumor effect in mice injected with EDA-negative tumor cells lines when the tumor stroma or the basement membrane of tumor endothelial cells express EDA. Thus, EDA CAR-T administration delayed tumor growth in immunocompetent 129Sv mice challenged with teratocarcinoma cell line F9. EDA CAR-T treatment exerted an antiangiogenic effect and significantly reduced gene signatures associated with epithelial-mesenchymal transition, collagen synthesis, extracellular matrix organization as well as IL-6-STAT5 and KRAS pathways. Importantly, the human version of EDA CAR, that includes the human 41BB and CD3ζ endodomains, exerted strong antitumor activity in NSG mice challenged with the human hepatocarcinoma cell line PLC, which expresses EDA in the tumor stroma and the endothelial vasculature. EDA CAR-T cells exhibited a tropism for EDA-expressing tumor tissue and no toxicity was observed in tumor bearing or in healthy mice. CONCLUSIONS These results suggest that targeting the tumor-specific fibronectin splice variant EDA with CAR-T cells is feasible and offers a therapeutic option that is applicable to different types of cancer.
Collapse
Affiliation(s)
- Celia Martín-Otal
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Aritz Lasarte-Cia
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Diego Serrano
- Programa de Tumores sólidos, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Noelia Casares
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Enrique Conde
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Flor Navarro
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Inés Sánchez-Moreno
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Marta Gorraiz
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Patricia Sarrión
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Alfonso Calvo
- Programa de Tumores sólidos, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | - Carlos E De Andrea
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain.,Departamento de Patología, Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - José Echeveste
- Departamento de Patología, Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Amaia Vilas
- Programa de Hemato-Oncología, Centro de Investigación Médica Aplicada, CIMA, Pamplona, Spain
| | - Juan Roberto Rodriguez-Madoz
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain.,Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain,Cancer Center Universidad de Navarra (CCUN), Universidad de Navarra, Pamplona, Spain
| | - Jesús San Miguel
- Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain,Cancer Center Universidad de Navarra (CCUN), Universidad de Navarra, Pamplona, Spain
| | - Felipe Prosper
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain.,Programa de Hemato-Oncología, Centro de Investigación Médica Aplicada, CIMA, Pamplona, Spain.,Centro de Investigacion Biomedica en Red de Cancer (CIBERONC), Madrid, Spain,Cancer Center Universidad de Navarra (CCUN), Universidad de Navarra, Pamplona, Spain
| | - Sandra Hervas-Stubbs
- Programa de Inmunología e Inmunoterapia, Centro de Investigación Médica Aplicada, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Juan Jose Lasarte
- Departamento de Hematología, Clínica Universidad de Navarra, Pamplona, Spain,Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Teresa Lozano
- Departamento de Hematología, Clínica Universidad de Navarra, Pamplona, Spain,Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| |
Collapse
|
2
|
Tanaka T, Konishi Y, Ichise H, Tsukiji S, Matsuda M, Terai K. A Dual Promoter System to Monitor IFN-γ Signaling in vivo at Single-cell Resolution. Cell Struct Funct 2021; 46:103-111. [PMID: 34744115 PMCID: PMC10511040 DOI: 10.1247/csf.21052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/27/2021] [Indexed: 11/11/2022] Open
Abstract
IFN-γ secreted from immune cells exerts pleiotropic effects on tumor cells, including induction of immune checkpoint and antigen presentation, growth inhibition, and apoptosis induction. We combined a dual promoter system with an IFN-γ signaling responsive promoter to generate a reporter named the interferon sensing probe (ISP), which quantitates the response to IFN-γ by means of fluorescence and bioluminescence. The integration site effect of the transgene is compensated for by the PGK promoter-driven expression of a fluorescent protein. Among five potential IFN-γ-responsive elements, we found that the interferon γ-activated sequence (GAS) exhibited the best performance. When ISP-GAS was introduced into four cell lines and subjected to IFN-γ stimulation, dose-dependency was observed with an EC50 ranging from 0.2 to 0.9 ng/mL, indicating that ISP-GAS can be generally used as a sensitive biosensor of IFN-γ response. In a syngeneic transplantation model, the ISP-GAS-expressing cancer cells exhibited bioluminescence and fluorescence signals in an IFN-γ receptor-dependent manner. Thus, ISP-GAS could be used to quantitatively monitor the IFN-γ response both in vitro and in vivo.Key words: in vivo imaging, tumor microenvironment, interferon-gamma, dual promoter system.
Collapse
Affiliation(s)
- Taisei Tanaka
- Laboratory of Bioimaging and Cell Signaling, Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Yoshinobu Konishi
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Hiroshi Ichise
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shinya Tsukiji
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Nagoya, Japan
| | - Michiyuki Matsuda
- Laboratory of Bioimaging and Cell Signaling, Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan
| | - Kenta Terai
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
3
|
Bustos M, Dubrot J, Martinez-Anso E, Larequi E, Castaño D, Palazon A, Belza I, Sanmamed MF, Perez-Gracia JL, Ortiz de Solorzano C, Alfaro C, Melero I. Cardiotrophin-1 determines liver engraftment of syngenic colon carcinoma cells through an immune system-mediated mechanism. Oncoimmunology 2021; 1:1527-1536. [PMID: 23264899 PMCID: PMC3525608 DOI: 10.4161/onci.22504] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiotrophin-1 (CT-1/CTF1) is a member of the interleukin-6 (IL-6) family of cytokines that stimulates STAT-3 phosphorylation in cells bearing the cognate receptor. We report that Ctf1−/− mice (hereby referred to as CT-1−/− mice) are resistant to the hepatic engraftment of MC38 colon carcinoma cells, while these cells engraft normally in the mouse subcutaneous tissue. Tumor intake in the liver could be enhanced by the systemic delivery of a recombinant adenovirus encoding CT-1, which also partly rescued the resistance of CT-1−/− mice to the hepatic engraftment of MC38 cells. Moreover, systemic treatment of wild-type (WT) mice with a novel antibody-neutralizing mouse CT-1 also reduced engraftment of this model. Conversely, experiments with Panc02 pancreatic cancer and B16-OVA melanoma cells in CT-1−/− mice revealed rates of hepatic engraftment similar to those observed in WT mice. The mechanism whereby CT-1 renders the liver permissive for MC38 metastasis involves T lymphocytes and natural killer (NK) cells, as shown by selective depletion experiments and in genetically deficient mice. However, no obvious changes in the number or cell killing capacity of liver lymphocytes in CT-1−/− animals could be substantiated. These findings demonstrate that the seed and soil concept to understand metastasis can be locally influenced by cytokines as well as by the cellular immune system.
Collapse
Affiliation(s)
- Matilde Bustos
- Gene Therapy and Hepatology Unit; Center for Applied Medical Research; University Clinic of Navarra; Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Sánchez-Paulete AR, Teijeira Á, Quetglas JI, Rodríguez-Ruiz ME, Sánchez-Arráez Á, Labiano S, Etxeberria I, Azpilikueta A, Bolaños E, Ballesteros-Briones MC, Casares N, Quezada SA, Berraondo P, Sancho D, Smerdou C, Melero I. Intratumoral Immunotherapy with XCL1 and sFlt3L Encoded in Recombinant Semliki Forest Virus-Derived Vectors Fosters Dendritic Cell-Mediated T-cell Cross-Priming. Cancer Res 2018; 78:6643-6654. [PMID: 30297531 DOI: 10.1158/0008-5472.can-18-0933] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/01/2018] [Accepted: 09/25/2018] [Indexed: 11/16/2022]
Abstract
: Multiple lines of evidence indicate a critical role of antigen cross-presentation by conventional BATF3-dependent type 1 classical dendritic cells (cDC1) in CD8-mediated antitumor immunity. Flt3L and XCL1, respectively, constitute a key growth/differentiation factor and a potent and specific chemoattractant for cDC1. To exploit their antitumor functions in local immunotherapy, we prepared Semliki Forest Virus (SFV)-based vectors encoding XCL1 and soluble Flt3L (sFlt3L). These vectors readily conferred transgene expression to the tumor cells in culture and when engrafted as subcutaneous mouse tumor models. In syngeneic mice, intratumoral injection of SFV-XCL1-sFlt3L (SFV-XF) delayed progression of MC38- and B16-derived tumors. Therapeutic activity was observed and exerted additive effects in combination with anti-PD-1, anti-CD137, or CTLA-4 immunostimulatory mAbs. Therapeutic effects were abolished by CD8β T-cell depletion and were enhanced by CD4 T-cell depletion, but not by T regulatory cell predepletion with anti-CD25 mAb. Antitumor effects were also abolished in BATF3- and IFNAR-deficient mice. In B16-OVA tumors, SFV-XF increased the number of infiltrating CD8 T cells, including those recognizing OVA. Consistently, following the intratumoral SFV-XF treatment courses, we observed increased BATF3-dependent cDC1 among B16-OVA tumor-infiltrating leukocytes. Such an intratumoral increase was not seen in MC38-derived tumors, but both resident and migratory cDC1 were boosted in SFV-XF-treated MC38 tumor-draining lymph nodes. In conclusion, viral gene transfer of sFlt3L and XCL1 is feasible, safe, and biologically active in mice, exerting antitumor effects that can be potentiated by CD4 T-cell depletion. SIGNIFICANCE: These findings demonstrate that transgenic expression of sFLT3L and XCL1 in tumor cells mediates cross-priming of, and elicits potent antitumor activity from, CD8 T lymphocytes, particularly in combination with CD4 T-cell depletion.
Collapse
Affiliation(s)
- Alfonso R Sánchez-Paulete
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Álvaro Teijeira
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - José I Quetglas
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - María E Rodríguez-Ruiz
- University Clinic, University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Álvaro Sánchez-Arráez
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Sara Labiano
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Iñaki Etxeberria
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Arantza Azpilikueta
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Elixabet Bolaños
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
- CIBERONC, Instituto de Investigación Carlos III, Madrid, Spain
| | - María Cristina Ballesteros-Briones
- Division of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical Research (CIMA), University of Navarra, and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Noelia Casares
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Sergio A Quezada
- Cancer Immunology Unit, University College London Cancer Institute, University College London, London, United Kingdom
| | - Pedro Berraondo
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
- CIBERONC, Instituto de Investigación Carlos III, Madrid, Spain
| | - David Sancho
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Cristian Smerdou
- Division of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical Research (CIMA), University of Navarra, and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Ignacio Melero
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain.
- University Clinic, University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
- CIBERONC, Instituto de Investigación Carlos III, Madrid, Spain
| |
Collapse
|
5
|
Shin DS, Zaretsky JM, Escuin-Ordinas H, Garcia-Diaz A, Hu-Lieskovan S, Kalbasi A, Grasso CS, Hugo W, Sandoval S, Torrejon DY, Palaskas N, Rodriguez GA, Parisi G, Azhdam A, Chmielowski B, Cherry G, Seja E, Berent-Maoz B, Shintaku IP, Le DT, Pardoll DM, Diaz LA, Tumeh PC, Graeber TG, Lo RS, Comin-Anduix B, Ribas A. Primary Resistance to PD-1 Blockade Mediated by JAK1/2 Mutations. Cancer Discov 2017; 7:188-201. [PMID: 27903500 PMCID: PMC5296316 DOI: 10.1158/2159-8290.cd-16-1223] [Citation(s) in RCA: 924] [Impact Index Per Article: 132.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 11/28/2016] [Accepted: 11/28/2016] [Indexed: 01/05/2023]
Abstract
Loss-of-function mutations in JAK1/2 can lead to acquired resistance to anti-programmed death protein 1 (PD-1) therapy. We reasoned that they may also be involved in primary resistance to anti-PD-1 therapy. JAK1/2-inactivating mutations were noted in tumor biopsies of 1 of 23 patients with melanoma and in 1 of 16 patients with mismatch repair-deficient colon cancer treated with PD-1 blockade. Both cases had a high mutational load but did not respond to anti-PD-1 therapy. Two out of 48 human melanoma cell lines had JAK1/2 mutations, which led to a lack of PD-L1 expression upon interferon gamma exposure mediated by an inability to signal through the interferon gamma receptor pathway. JAK1/2 loss-of-function alterations in The Cancer Genome Atlas confer adverse outcomes in patients. We propose that JAK1/2 loss-of-function mutations are a genetic mechanism of lack of reactive PD-L1 expression and response to interferon gamma, leading to primary resistance to PD-1 blockade therapy. SIGNIFICANCE A key functional result from somatic JAK1/2 mutations in a cancer cell is the inability to respond to interferon gamma by expressing PD-L1 and many other interferon-stimulated genes. These mutations result in a genetic mechanism for the absence of reactive PD-L1 expression, and patients harboring such tumors would be unlikely to respond to PD-1 blockade therapy. Cancer Discov; 7(2); 188-201. ©2016 AACR.See related commentary by Marabelle et al., p. 128This article is highlighted in the In This Issue feature, p. 115.
Collapse
Affiliation(s)
| | - Jesse M Zaretsky
- University of California, Los Angeles (UCLA), Los Angeles, California
| | | | - Angel Garcia-Diaz
- University of California, Los Angeles (UCLA), Los Angeles, California
| | | | - Anusha Kalbasi
- University of California, Los Angeles (UCLA), Los Angeles, California
| | | | - Willy Hugo
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Salemiz Sandoval
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Davis Y Torrejon
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Nicolaos Palaskas
- University of California, Los Angeles (UCLA), Los Angeles, California
| | | | - Giulia Parisi
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Ariel Azhdam
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Bartosz Chmielowski
- University of California, Los Angeles (UCLA), Los Angeles, California
- Jonsson Comprehensive Cancer Center, Los Angeles, California
| | - Grace Cherry
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Elizabeth Seja
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Beata Berent-Maoz
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - I Peter Shintaku
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Dung T Le
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Drew M Pardoll
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Luis A Diaz
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Paul C Tumeh
- University of California, Los Angeles (UCLA), Los Angeles, California
| | - Thomas G Graeber
- University of California, Los Angeles (UCLA), Los Angeles, California
- Jonsson Comprehensive Cancer Center, Los Angeles, California
| | - Roger S Lo
- University of California, Los Angeles (UCLA), Los Angeles, California
- Jonsson Comprehensive Cancer Center, Los Angeles, California
| | - Begoña Comin-Anduix
- University of California, Los Angeles (UCLA), Los Angeles, California
- Jonsson Comprehensive Cancer Center, Los Angeles, California
| | - Antoni Ribas
- University of California, Los Angeles (UCLA), Los Angeles, California.
- Jonsson Comprehensive Cancer Center, Los Angeles, California
| |
Collapse
|
6
|
Abstract
Loss-of-function mutations in JAK1/2 can lead to acquired resistance to anti-programmed death protein 1 (PD-1) therapy. We reasoned that they may also be involved in primary resistance to anti-PD-1 therapy. JAK1/2-inactivating mutations were noted in tumor biopsies of 1 of 23 patients with melanoma and in 1 of 16 patients with mismatch repair-deficient colon cancer treated with PD-1 blockade. Both cases had a high mutational load but did not respond to anti-PD-1 therapy. Two out of 48 human melanoma cell lines had JAK1/2 mutations, which led to a lack of PD-L1 expression upon interferon gamma exposure mediated by an inability to signal through the interferon gamma receptor pathway. JAK1/2 loss-of-function alterations in The Cancer Genome Atlas confer adverse outcomes in patients. We propose that JAK1/2 loss-of-function mutations are a genetic mechanism of lack of reactive PD-L1 expression and response to interferon gamma, leading to primary resistance to PD-1 blockade therapy. SIGNIFICANCE A key functional result from somatic JAK1/2 mutations in a cancer cell is the inability to respond to interferon gamma by expressing PD-L1 and many other interferon-stimulated genes. These mutations result in a genetic mechanism for the absence of reactive PD-L1 expression, and patients harboring such tumors would be unlikely to respond to PD-1 blockade therapy. Cancer Discov; 7(2); 188-201. ©2016 AACR.See related commentary by Marabelle et al., p. 128This article is highlighted in the In This Issue feature, p. 115.
Collapse
|
7
|
Ribas A, Hu-Lieskovan S. What does PD-L1 positive or negative mean? J Exp Med 2016; 213:2835-2840. [PMID: 27903604 PMCID: PMC5154949 DOI: 10.1084/jem.20161462] [Citation(s) in RCA: 247] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/13/2016] [Accepted: 11/21/2016] [Indexed: 12/13/2022] Open
Abstract
Ribas and Hu-Lieskovan show that different processes may lead to the expression of PD-L1 on cancer cells, and each one of them may have a different meaning to interpret the results of clinical trials with anti–PD-1/L1 antibodies. Expression of the programmed death-1 (PD-1) ligand 1 (PD-L1) is used to select patients and analyze responses to anti–PD-1/L1 antibodies. The expression of PD-L1 is regulated in different ways, which leads to a different significance of its presence or absence. PD-L1 positivity may be a result of genetic events leading to constitutive PD-L1 expression on cancer cells or inducible PD-L1 expression on cancer cells and noncancer cells in response to a T cell infiltrate. A tumor may be PD-L1 negative because it has no T cell infiltrate, which may be reversed with an immune response. Finally, a tumor that is unable to express PD-L1 because of a genetic event will always be negative for PD-L1 on cancer cells.
Collapse
Affiliation(s)
- Antoni Ribas
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, CA 90095 .,Jonsson Comprehensive Cancer Center (JCCC), University of California, Los Angeles (UCLA), Los Angeles, CA 90095
| | - Siwen Hu-Lieskovan
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles (UCLA), Los Angeles, CA 90095.,Jonsson Comprehensive Cancer Center (JCCC), University of California, Los Angeles (UCLA), Los Angeles, CA 90095
| |
Collapse
|
8
|
Son CH, Bae JH, Shin DY, Lee HR, Yang K, Park YS. Antitumor effect of dendritic cell loaded ex vivo and in vivo with tumor-associated antigens in lung cancer model. Immunol Invest 2014; 43:447-62. [PMID: 24654594 DOI: 10.3109/08820139.2014.884576] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Various ex vivo or in vivo loading protocols have been developed or evaluated for the delivery of tumor antigens to dendritic cells (DCs). We compared the antitumor effect of mature DCs electroporation-pulsed (EP/mDC) ex vivo with tumor cell lysate and immature DCs (iDCs) injected into the tumor apoptosed by ionizing radiation (IR/iDC) in lung cancer model. DCs were generated from bone marrow of C57BL/6 mice. Ionizing radiation (IR) was applied at a dose of 10 Gy to the tumor on the right thigh. iDCs were intratumorally injected into the irradiated tumor and EP/mDC was injected subcutaneously in the right flank. DC injection induced strong tumor-specific immunity against Lewis lung carcinoma, as compared with the tumor-bearing control and IR only treated mice. The growth of a distant tumor on the right and left flank was inhibited by IR/iDC and EP/mDC. Particularly, IR/iDC resulted in a more significant inhibition of tumor growth and prolonged survival time. It was related to increase of tumor-specific interferon-gamma, cytotoxicity, and decrease of regulatory T-cells. The results indicate that DCs electroporation-pulsed with tumor cell lysate induce a potent antitumor effect, but that iDCs intratumoral injected into the irradiated tumor induce a more potent antitumor effect.
Collapse
Affiliation(s)
- Cheol-Hun Son
- Dongnam Institute of Radiological & Medical Sciences , Busan 619-953 , South Korea
| | | | | | | | | | | |
Collapse
|
9
|
Quetglas JI, Dubrot J, Bezunartea J, Sanmamed MF, Hervas-Stubbs S, Smerdou C, Melero I. Immunotherapeutic synergy between anti-CD137 mAb and intratumoral administration of a cytopathic Semliki Forest virus encoding IL-12. Mol Ther 2012; 20:1664-75. [PMID: 22735380 DOI: 10.1038/mt.2012.56] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Intratumoral injection of Semliki Forest virus encoding interleukin-12 (SFV-IL-12) combines acute expression of IL-12 and stressful apoptosis of infected malignant cells. Agonist antibodies directed to costimulatory receptor CD137 (4-1BB) strongly amplify pre-existing cellular immune responses toward weak tumor antigens. In this study, we provide evidence for powerful synergistic effects of a combined strategy consisting of intratumoral injection of SFV-IL-12 and systemic delivery of agonist anti-CD137 monoclonal antibodies (mAbs), which was substantiated against poorly immunogenic B16 melanomas (B16-OVA and B16.F10) and TC-1 lung carcinomas. Effector CD8(β)(+) T cells were sufficient to mediate complete tumor eradications. Accordingly, there was an intensely synergistic in vivo enhancement of cytotoxic T lymphocytes (CTL)-mediated immunity against the tumor antigens OVA and tyrosine-related protein-2 (TRP-2). This train of phenomena led to long-lasting tumor-specific immunity against rechallenge, attained transient control of the progression of concomitant tumor lesions that were not directly treated with SFV-IL-12 and caused autoimmune vitiligo. Importantly, we found that SFV-IL-12 intratumoral injection induces bright expression of CD137 on most tumor-infiltrating CD8(+) T lymphocytes, thereby providing more abundant targets for the action of the agonist antibody. This efficacious combinatorial immunotherapy strategy offers feasibility for clinical translation since anti-CD137 mAbs are already undergoing clinical trials and development of clinical-grade SFV-IL-12 vectors is in progress.
Collapse
Affiliation(s)
- José I Quetglas
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Navarra, Spain
| | | | | | | | | | | | | |
Collapse
|
10
|
Perez-Gracia JL, Berraondo P, Martinez-Forero I, Alfaro C, Suarez N, Gurpide A, Sangro B, Hervas-Stubbs S, Ochoa C, Melero JA, Melero I. Clinical development of combination strategies in immunotherapy: are we ready for more than one investigational product in an early clinical trial? Immunotherapy 2011; 1:845-53. [PMID: 20636027 DOI: 10.2217/imt.09.51] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Stimulating the innate and adaptive immunity against cancer necessitates the tricking of a system evolved to fight microbial pathogens and directing its activity towards transformed self-tissue. Efficacious interventions to start and sustain the response will probably require a number of agents to tamper simultaneously or sequentially with several immune mechanisms. Although master switches controlling various functions may exist, the goal of a curative immune response will probably demand the combined actions of several therapeutic components. Synergy occurs when drugs interact in ways that enhance or magnify one or more effects or side effects. In cancer immunotherapy, two agents that have minor or no therapeutic effects as single agents can be powerful when combined. Mouse experimentation provides multiple examples of synergistic combinations. Elements to be combined include chiefly: tumor vaccines, adoptive T-cell therapies, cytokines, costimulatory molecules, molecular deactivation of immunosuppressive or tolerogenic pathways and immunostimulatory monoclonal antibodies. These novel therapies, even as single agents, are extremely complex products to be developed owing to the associated biomolecules, cell therapies or gene therapies. At present, drug-development programs are run individually for each immunotherapeutic agent and combinations are considered only at a later stage in clinical development, even in the absence of formal compulsory regulations to prevent clinical trials with combinations. As a result, instead of the search for maximal efficacy, ease of combination with standard treatments, intellectual property management, regulations and business-based decisions often guide the way. Even though the maximal effort must be made in order to prevent adverse effects in patients, it seems reasonable that combination pilot trials should be performed at an early stage, following safe completion of Phase I trials. These trials should be performed based on evidence for synergy in animal models and be simplified in terms of regulatory requirements. Such 'short-cut' combination immunotherapy trials can bring much needed efficacy earlier to the bedside.
Collapse
Affiliation(s)
- Jose L Perez-Gracia
- Centro de Investigación Médica Aplicada y Clinica Universitaria. Universidad de Navarra, Avenida de Pio XII 55, 31008 Pamplona, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Saha A, Chatterjee SK. Combination of CTL-associated antigen-4 blockade and depletion of CD25 regulatory T cells enhance tumour immunity of dendritic cell-based vaccine in a mouse model of colon cancer. Scand J Immunol 2010; 71:70-82. [PMID: 20384858 DOI: 10.1111/j.1365-3083.2009.02355.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Immune regulation has been shown to be involved in the progressive growth of some murine tumours. Interruption of immune regulatory pathways via CTL-associated antigen-4 (CTLA-4) blockade or removal of CD4(+) CD25(+) regulatory T (Treg) cells appears to be a promising strategy for cancer immunotherapy. In this study, we tested the hypothesis that the combination of CTLA-4 blockade and depletion of Treg cells would improve the potency of dendritic cell (DC)-based vaccine in a clinically relevant mouse model, which is transgenic for both carcinoembryonic antigen (CEA) and HLA-A2 for the treatment of colon carcinoma in a therapeutic setting. We found that administration of anti-CD25 antibody prior to vaccination or systemic administration of anti-CTLA-4 antibody with the vaccine improved tumour-free survival against CEA-expressing tumours compared with mice immunized with DC-based vaccine alone. However, the efficacy of the vaccine proved to be most effective when anti-CTLA-4 antibody was combined with Treg inhibition. This vaccination strategy dramatically improved the tumour-free survival and allowed the development of long-lasting immune responses. The combined vaccination strategy resulted in increased secretion of IFN-gamma and enhanced HLA-A2-restricted CEA-specific CTL responses. Furthermore, coadministration of anti-CD25 and anti-CTLA-4 antibodies along with the vaccine was effective against more advanced tumours. These results provide evidence that simultaneous blockade of T-cell regulatory pathways is a promising approach for the induction of therapeutic antitumour immunity against CEA(+) colon carcinoma.
Collapse
Affiliation(s)
- Asim Saha
- Department of Internal Medicine, University of Cincinnati Medical Center, Cincinnati, OH, USA.
| | | |
Collapse
|
12
|
Doi C, Egashira N, Kawabata A, Maurya DK, Ohta N, Uppalapati D, Ayuzawa R, Pickel L, Isayama Y, Troyer D, Takekoshi S, Tamura M. Angiotensin II type 2 receptor signaling significantly attenuates growth of murine pancreatic carcinoma grafts in syngeneic mice. BMC Cancer 2010; 10:67. [PMID: 20181281 PMCID: PMC2846883 DOI: 10.1186/1471-2407-10-67] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Accepted: 02/24/2010] [Indexed: 12/19/2022] Open
Abstract
Background Pancreatic cancer is one of the most aggressive human malignancies, with a very poor prognosis. To evaluate the effect of angiotensin II (Ang II) type 2 receptor (AT2) expression in the host's body on the growth of pancreatic carcinoma, we have investigated the growth of mouse pancreatic ductal carcinoma grafts in syngeneic wild type and AT2 receptor-deficient (AT2-KO) mice. Methods The role of AT2 receptor-signaling in stromal cells on the growth of murine pancreatic carcinoma cells (PAN02) was studied using various in vitro and in vivo assays. In vivo cell proliferation, apoptosis, and vasculature in tumors were monitored by Ki-67 immunostaining, TUNEL assay, and von Willebrand factor immunostaining, respectively. In the co-culture study, cell proliferation was measured by MTT cell viability assay. All the data were analyzed using t-test and data were treated as significant when p < 0.05. Results Our results show that the growth of subcutaneously transplanted syngeneic xenografts of PAN02 cells, mouse pancreatic ductal carcinoma cells derived from the C57/BL6 strain, was significantly faster in AT2-KO mice compared to control wild type mice. Immunohistochemical analysis of tumor tissue revealed significantly more Ki-67 positive cells in xenografts grown in AT2-KO mice than in wild type mice. The index of apoptosis is slightly higher in wild type mice than in AT2-KO mice as evaluated by TUNEL assay. Tumor vasculature number was significantly higher in AT2-KO mice than in wild type mice. In vitro co-culture studies revealed that the growth of PAN02 cells was significantly decreased when grown with AT2 receptor gene transfected wild type and AT2-KO mouse-derived fibroblasts. Faster tumor growth in AT2-KO mice may be associated with higher VEGF production in stromal cells. Conclusions These results suggest that Ang II regulates the growth of pancreatic carcinoma cells through modulating functions of host stromal cells; Moreover, Ang II AT2 receptor signaling is a negative regulator in the growth of pancreatic carcinoma cells. These findings indicate that the AT2 receptor in stromal fibroblasts is a potentially important target for chemotherapy for pancreatic cancer.
Collapse
Affiliation(s)
- Chiyo Doi
- Department of Anatomy & Physiology, Kansas State University, College of Veterinary Medicine, Manhattan, KS 66506, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Matar P, Alaniz L, Rozados V, Aquino JB, Malvicini M, Atorrasagasti C, Gidekel M, Silva M, Scharovsky OG, Mazzolini G. Immunotherapy for liver tumors: present status and future prospects. J Biomed Sci 2009; 16:30. [PMID: 19272130 PMCID: PMC2662798 DOI: 10.1186/1423-0127-16-30] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Accepted: 03/06/2009] [Indexed: 12/22/2022] Open
Abstract
Increasing evidence suggests that immune responses are involved in the control of cancer and that the immune system can be manipulated in different ways to recognize and attack tumors. Progress in immune-based strategies has opened new therapeutic avenues using a number of techniques destined to eliminate malignant cells. In the present review, we overview current knowledge on the importance, successes and difficulties of immunotherapy in liver tumors, including preclinical data available in animal models and information from clinical trials carried out during the lasts years. This review shows that new options for the treatment of advanced liver tumors are urgently needed and that there is a ground for future advances in the field.
Collapse
Affiliation(s)
- Pablo Matar
- Institute of Experimental Genetics, School of Medical Sciences, National University of Rosario, Santa Fe 3100, (2000) Rosario, Argentina.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Chen C, Zhang C, Zhuang G, Luo H, Su J, Yin P, Wang J. Decoy receptor 3 overexpression and immunologic tolerance in hepatocellular carcinoma (HCC) development. Cancer Invest 2009; 26:965-74. [PMID: 19093253 DOI: 10.1080/07357900801975256] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The recently identified decoy receptor 3 (DcR3) inhibits FasL-induced apoptosis by binding to FasL, and it is considered to play a key role in the immune escape system of neoplastic cells. In order to examine the involvement of DcR3 in the immunologic tolerance of hepatocellular carcinoma (HCC), we investigated the amplification and expression of DcR3, FasL, and Fas in an HCC mice model using RT-PCR, western blotting, and ELISA, and analyzed the space-time relationship with various cytokines including the forkhead transcription factor forkhead/winged helix transcription factor gene (Foxp3), CTLA-4, TGF-beta, IL-10, TNF-alpha, and IFN-gamma. The RT-PCR results revealed that Fas expression preceded that of DcR3 during the early phases of tumorigenesis. Thereafter, the expression of DcR3 was up-regulated; however, the expression of Fas was down-regulated and eventually ceased. DcR3 and FasL were expressed and amplified simultaneously in muscle tumor. CTLA-4 expression was earlier than Foxp3, and both CTLA-4 and Foxp3 amplification and expression were consistent with that of DcR3. The results suggest that the elevated levels of DcR3, Foxp3, and CTLA-4 in tissue were positively correlated with tumor growth. The partial tumor immunoregulation inclined to negative modulation, and DcR3 may play an important role in inducing immunologic tolerance.
Collapse
Affiliation(s)
- Caixia Chen
- Anti-Cancer Research Center, Xiamen University Medical College, XiaMen, China
| | | | | | | | | | | | | |
Collapse
|
15
|
Murillo O, Arina A, Hervas-Stubbs S, Gupta A, McCluskey B, Dubrot J, Palazón A, Azpilikueta A, Ochoa MC, Alfaro C, Solano S, Pérez-Gracia JL, Oyajobi BO, Melero I. Therapeutic antitumor efficacy of anti-CD137 agonistic monoclonal antibody in mouse models of myeloma. Clin Cancer Res 2008; 14:6895-906. [PMID: 18980984 DOI: 10.1158/1078-0432.ccr-08-0285] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Eradication of post-treatment residual myeloma cells is needed to prevent relapses, and immunostimulatory monoclonal antibodies (mAb) such as anti-CD137, CTLA-4, CD40, etc., which enhance the immune response against malignancies, represent a means of achieving this purpose. This study explores anti-CD137 mAbs for multiple myeloma treatment in preclinical models of the disease because they safely augment tumor immunity and are in clinical trials for other cancers. EXPERIMENTAL DESIGN The antitumor effect of anti-CD137 mAb on mouse plasmacytomas derived from HOPC and NS0 cell lines was studied and compared with that of anti-CTLA-4, anti-CD40, and anti-ICAM-2 mAbs. The antitumor effect of anti-CD137 mAb was also examined in a mouse syngeneic disseminated myeloma (5TGM1) model, which more closely resembles human multiple myeloma. Depletions of specific cell populations and gene-targeted mice were used to unravel the requirements for tumor rejection. RESULTS Agonistic mAb against CD137 and blocking anti-CTLA-4 mAb showed activity against i.p. HOPC tumors, resulting in extended survival of mice that also became immune to rechallenge. Anti-CD137 mAbs induced complete eradications of established s.c. NS0-derived tumors that were dependent on IFN-gamma, natural killer cells, and CD8(+) T lymphocytes. Natural killer cells accumulated in tumor draining lymph nodes and showed increased IFN-gamma production. Antitumor efficacy of anti-CD137 mAb was preserved in CD28-deficient mice despite the fact that CD28 signaling increases the expression of CD137 on CD8(+) T cells. Importantly, anti-CD137 mAb treatment significantly decreased systemic tumor burden in the disseminated 5TGM1 model. CONCLUSIONS The immune-mediated antitumor activity of anti-CD137 mAb in mouse models holds promise for myeloma treatment in humans.
Collapse
Affiliation(s)
- Oihana Murillo
- Gene Therapy Unit, Centro de Investigación Médica Aplicada, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Mazzolini G, Murillo O, Atorrasagasti C, Dubrot J, Tirapu I, Rizzo M, Arina A, Alfaro C, Azpilicueta A, Berasain C, Perez-Gracia JL, Gonzalez A, Melero I. Immunotherapy and immunoescape in colorectal cancer. World J Gastroenterol 2007; 13:5822-31. [PMID: 17990348 PMCID: PMC4205429 DOI: 10.3748/wjg.v13.i44.5822] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy encompasses a variety of interventions and techniques with the common goal of eliciting tumor cell destructive immune responses. Colorectal carcinoma often presents as metastatic disease that impedes curative surgery. Novel strategies such as active immunization with dendritic cells (DCs), gene transfer of cytokines into tumor cells or administration of immunostimulatory monoclonal antibodies (such as anti-CD137 or anti-CTLA-4) have been assessed in preclinical studies and are at an early clinical development stage. Importantly, there is accumulating evidence that chemotherapy and immunotherapy can be combined in the treatment of some cases with colorectal cancer, with synergistic potentiation as a result of antigens cross-presented by dendritic cells and/or elimination of competitor or suppressive T lymphocyte populations (regulatory T-cells). However, genetic and epigenetic unstable carcinoma cells frequently evolve mechanisms of immunoevasion that are the result of either loss of antigen presentation, or an active expression of immunosuppressive substances. Some of these actively immunosuppressive mechanisms are inducible by cytokines that signify the arrival of an effector immune response. For example, induction of 2, 3 indoleamine dioxygenase (IDO) by IFNγ in colorectal carcinoma cells. Combinational and balanced strategies fostering antigen presentation, T-cell costimulation and interference with immune regulatory mechanisms will probably take the stage in translational research in the treatment of colorectal carcinoma.
Collapse
|
17
|
Weiss JM, Subleski JJ, Wigginton JM, Wiltrout RH. Immunotherapy of cancer by IL-12-based cytokine combinations. Expert Opin Biol Ther 2007; 7:1705-21. [PMID: 17961093 DOI: 10.1517/14712598.7.11.1705] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Cancer is a multi-faceted disease comprising complex interactions between neoplastic and normal cells. Over the past decade, there has been considerable progress in defining the molecular, cellular and environmental contributions to the pathophysiology of tumor development. Despite these advances, the conventional treatment of patients still generally involves surgery, radiotherapy and/or chemotherapy, and the clinical outcome for many of these efforts remains unsatisfactory. Recent studies have highlighted the feasibility of using immunotherapeutic approaches that seek to enhance host immune responses to developing tumors. These strategies include immunomodulatory cytokines, with TNF-alpha, type I or type II IFNs, IL-2, IL-12, IL-15 and IL-18 being among the most potent inducers of anti-tumor activity in a variety of preclinical studies. More recently, some exciting new cytokines have been characterized, such as IL-21, IL-23, IL-27 and their immunomodulatory and antitumor effects in vitro and in vivo suggest that they may have considerable promise for future immunotherapy protocols. The promise of cytokine therapy does indeed derive from the identification of these novel cytokines but even more fundamentally, the field is greatly benefiting from the ever-expanding amount of preclinical data that convincingly demonstrate synergistic and/or novel biologic effects, which may be achieved through the use of several combinations of cytokines with complementary immune-stimulating capabilities. One cytokine in particular, IL-12, holds considerable promise by virtue of the fact that it plays a central role in regulating both innate and adaptive immune responses, can by itself induce potent anticancer effects, and synergizes with several other cytokines for increased immunoregulatory and antitumor activities. This review discusses the antitumor activity of IL-12, with a special emphasis on its ability to synergize with other cytokines for enhancement of immune effector cell populations and regulation of host-tumor cell interactions and the overall tumor microenvironment.
Collapse
Affiliation(s)
- Jonathan M Weiss
- National Cancer Institute, Cancer and Inflammation Program, Laboratory of Experimental Immunology, Center for Cancer Research, Frederick, MD 21702, USA
| | | | | | | |
Collapse
|
18
|
Zhong S, Xie D. Gene Ontology analysis in multiple gene clusters under multiple hypothesis testing framework. Artif Intell Med 2007; 41:105-15. [PMID: 17913480 DOI: 10.1016/j.artmed.2007.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2006] [Revised: 08/02/2007] [Accepted: 08/03/2007] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Gene Ontology (GO) has become a routine resource for functional analysis of gene lists. Although a number of tools have been provided to identify enriched GO terms in one or two gene lists, two technical challenges remain. First, how to handle multiple hypothesis testing in the analysis given that the tests are heavily correlated; second, how to identify GO terms that are enriched in a gene cluster, as compared to multiple other gene clusters. We provide a statistical procedure to rigorously treat these problems and offer a software tool for applying GO to the analysis of gene clusters. METHODS We previously introduced a statistical procedure that handles hypothesis testing in a two-group comparison scenario. In this paper we extend the two-group comparison procedure into a general procedure that enables the analysis of any number of gene lists/clusters. This new procedure enables identification of GO terms enriched in any gene cluster, while it controls for multiple hypothesis testing. This procedure is implemented into a user-friendly analysis tool: GoSurfer. The current version of GoSurfer takes one or several gene lists as input, and it identifies the GO terms that are enriched in any of the input gene lists. GoSurfer estimates a conservative false discovery rate (FDR) for every GO term. The FDR estimation procedure in GoSurfer has two advantages: it does not rely on independence assumption, and it does not assume all the hypotheses are null hypothesis (complete null). Thus GoSurfer's FDR estimates are mildly conservative rather than overly conservative. RESULTS We implemented the new procedure for GO analysis in multiple gene clusters into the GoSurfer software. We provide three examples on using GoSurfer to analyze time course gene expression data sets on the differentiation of embryonic stem cells. In the example of analysis of multiple gene clusters, we first used a typical clustering algorithm and identified five gene clusters, representing up-regulation, down-regulation and other patterns in the differentiation time course. Taking all the five gene clusters as input data, GoSurfer reports "cell adhesion" and "muscle contraction" as significant GO terms for the up-regulated cluster, "amino acids metabolism" as a significant GO term for the down-regulated gene cluster, and GoSurfer reports a number of GO terms related to RNA processing and RNA transport as significant terms to a cluster that is up-regulated in both early and late time points. This may suggest that genes for RNA processing and genes for RNA transport are coregulated in the differentiation process of embryonic stem cells. CONCLUSION The GoSurfer software is provided to analyze multiple gene clusters and identify GO terms that are enriched in any gene cluster. Gosurfer is available at: www.gosurfer.org.
Collapse
Affiliation(s)
- Sheng Zhong
- Department of Bioengineering, University of Illinois at Urbana Champaign, IL 61801, United States.
| | | |
Collapse
|
19
|
Abstract
The application of gene transfer technologies to the treatment of cancer has led to the development of new experimental approaches like gene directed enzyme/pro-drug therapy (GDEPT), inhibition of oncogenes and restoration of tumor-suppressor genes. In addition, gene therapy has a big impact on other fields like cancer immunotherapy, anti-angiogenic therapy and virotherapy. These strategies are being evaluated for the treatment of primary and metastatic liver cancer and some of them have reached clinical phases. We present a review on the basis and the actual status of gene therapy approaches applied to liver cancer.
Collapse
|
20
|
Abstract
Hepatocellular carcinoma (HCC), one of the most common cancers worldwide, is often diagnosed at an advanced stage when most potentially curative therapies such as resection, transplantation or percutaneous and transarterial interventions are of limited efficacy. The fact that HCC is resistant to conventional chemotherapy, and is rarely amenable to radiotherapy, leaves this disease with no effective therapeutic options and a very poor prognosis. Therefore, the development of more effective therapeutic tools and strategies is much needed. HCCs are phenotypically and genetically heterogeneous tumors that commonly emerge on a background of chronic liver disease. However, in spite of this heterogeneity recent insights into the biology of HCC suggest that certain signaling pathways and molecular alterations are likely to play essential roles in HCC development by promoting cell growth and survival. The identification of such mechanisms may open new avenues for the prevention and treatment of HCC through the development of targeted therapies. In this review we will describe the new potential therapeutic targets and clinical developments that have emerged from progress in the knowledge of HCC biology, In addition, recent advances in gene therapy and combined cell and gene therapy, together with new radiotherapy techniques and immunotherapy in patients with HCC will be discussed.
Collapse
Affiliation(s)
- M A Avila
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | | | | | | |
Collapse
|
21
|
Zhong S, Tian L, Li C, Storch KF, Wong WH. Comparative analysis of gene sets in the Gene Ontology space under the multiple hypothesis testing framework. PROCEEDINGS. IEEE COMPUTATIONAL SYSTEMS BIOINFORMATICS CONFERENCE 2006:425-35. [PMID: 16448035 DOI: 10.1109/csb.2004.1332455] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The Gene Ontology (GO) resource can be used as a powerful tool to uncover the properties shared among, and specific to, a list of genes produced by high-throughput functional genomics studies, such as microarray studies. In the comparative analysis of several gene lists, researchers maybe interested in knowing which GO terms are enriched in one list of genes but relatively depleted in another. Statistical tests such as Fisher's exact test or Chi-square test can be performed to search for such GO terms. However, because multiple GO terms are tested simultaneously, individual p-values from individual tests do not serve as good indicators for picking GO terms. Furthermore, these multiple tests are highly correlated, usual multiple testing procedures that work under an independence assumption are not applicable. In this paper we introduce a procedure, based on False Discovery Rate (FDR), to treat this correlated multiple testing problem. This procedure calculates a moderately conserved estimator of q-value for every GO term. We identify the GO terms with q-values that satisfy a desired level as the significant GO terms. This procedure has been implemented into the GoSurfer software. GoSurfer is a windows based graphical data mining tool. It is freely available at http://www.gosurfer.org.
Collapse
Affiliation(s)
- Sheng Zhong
- Department of Biostatistics, Harvard University, USA
| | | | | | | | | |
Collapse
|
22
|
Huarte E, Larrea E, Hernández-Alcoceba R, Alfaro C, Murillo O, Arina A, Tirapu I, Azpilicueta A, Hervás-Stubbs S, Bortolanza S, Pérez-Gracia JL, Civeira MP, Prieto J, Riezu-Boj JI, Melero I. Recombinant adenoviral vectors turn on the type I interferon system without inhibition of transgene expression and viral replication. Mol Ther 2006; 14:129-38. [PMID: 16627004 DOI: 10.1016/j.ymthe.2006.02.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2005] [Revised: 01/26/2006] [Accepted: 02/14/2006] [Indexed: 01/12/2023] Open
Abstract
Recombinant adenovirus administration gives rise to transgene-independent effects caused by the ability of the vector to activate innate immunity mechanisms. We show that recombinant adenoviruses encoding reporter genes trigger IFN-alpha and IFN-beta transcription from both plasmacytoid and myeloid mouse dendritic cells. Interestingly, IFN-beta and IFN-alpha5 are the predominant transcribed type I IFN genes both in vitro and in vivo. In human peripheral blood leukocytes type I IFNs are induced by adenoviral vectors, with a preponderance of IFN-beta together with IFN-alpha1 and IFN-alpha5 subtypes. Accordingly, functional type I IFN is readily detected in serum samples from human cancer patients who have been treated intratumorally with a recombinant adenovirus encoding thymidine kinase. Despite inducing functional IFN-alpha release in both mice and humans, gene transfer by recombinant adenoviruses is not interfered with by type I IFNs either in vitro or in vivo. Moreover, IFN-alpha does not impair replication of wild-type adenovirus. As a consequence, cancer gene therapy strategies with defective or replicative-competent adenoviruses are not expected to be hampered by the effect of the type I IFNs induced by the vector itself. However, type I IFN might modulate antitumor and antiadenoviral immune responses and thus influence the outcome of gene immunotherapy.
Collapse
Affiliation(s)
- Eduardo Huarte
- Center for Applied Medical Research, School of Medicine, and University Clinic, University of Navarra, Avenida Pio XII, 55, 31008 Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Kipshidze N, Tsapenko M, Iversen P, Burger D. Antisense therapy for restenosis following percutaneous coronary intervention. Expert Opin Biol Ther 2006; 5:79-89. [PMID: 15709911 DOI: 10.1517/14712598.5.1.79] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Recent advances in vascular gene transfer have shown potential new treatment modalities for cardiovascular disease, particularly in the treatment of vascular restenosis. The antisense approach to inhibiting gene expression involves introducing oligonucleotides complementary to mRNA into cells in order to block any one of the following processes: uncoiling of DNA, transcription of DNA, export of RNA, DNA splicing, RNA stability, or RNA translation involved in the synthesis of proteins in cellular proliferation. The approach includes the use of antisense oligonucleotides, antisense mRNA, autocatalytic ribozymes, and the insertion of a section of DNA to form a triple helix. Proof of principle has been established that inhibition of several cellular proto-oncogenes, including DNA binding protein c-myb, non-muscle myosin heavy chain, PCNA proliferating-cell nuclear antigen, platelet-derived growth factor, basic fibroblast growth factor and c-myc, inhibits smooth muscle cell proliferation in vitro and in several animal models. The first clinical study demonstrated the safety and feasibility of local delivery of antisense in the treatment and prevention of restenosis; another randomised clinical trial (AVAIL) with local delivery of c-myc morpholino compound in patients with coronary artery disease demonstrated its long-term effect on reducing neointimal formation, as well as its safety. These preliminary findings from the small cohort of patients require confirmation in a larger trial utilising more sophisticated drug-eluting technologies.
Collapse
Affiliation(s)
- Nicholas Kipshidze
- Lenox Hill Hospital, Department of Interventional Cardiac & Vascular Services, 130 East 77th Street, New York, NY 10021, USA.
| | | | | | | |
Collapse
|
24
|
Shibata MA, Ito Y, Morimoto J, Kusakabe K, Yoshinaka R, Otsuki Y. In vivo electrogene transfer ofinterleukin-12 inhibits tumor growth and lymph node and lung metastases in mouse mammary carcinomas. J Gene Med 2006; 8:335-52. [PMID: 16345101 DOI: 10.1002/jgm.854] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Human breast cancer metastasizes mainly to lymph nodes, lungs, liver, and bone; in the majority of cases, it is the development of metastases which leads to death. In order to suppress mammary cancer metastasis, we applied in vivo electrogene transfer (non-viral method) as a means of interleukin-12 (IL-12) gene therapy on highly metastatic murine mammary cancer model. METHODS Metastatic mammary tumors induced by inoculation in BALB/c female mice were treated by intratumoral injections of either a plasmid vector containing IL-12 or empty vector and then subjected to in vivo electrogene transfer once a week for 8 weeks. RESULTS Treatment with IL-12 resulted in elevation of both IL-12 and IFNgamma levels in mammary tumors and in serum and intratumoral levels of CD4 and CD8 proteins were also increased. Tumor volumes and lymphatic and pulmonary metastases were significantly reduced. The histopathological changes induced by IL-12 characteristically included marked inflammation, increased apoptosis, decreased DNA synthesis, peripheral influx of significantly greater numbers of active macrophages, and reduced blood microvessel density, and apoptotic vascular endothelial cells were frequently seen. Western blotting showed decreases in VEGFR-3 of tumors exposed to IL-12 gene therapy. In adjuvant immunofluorescence studies, the CD31-positive endothelial cells of microvessels showed decreased VEGFR-3 expression in IL-12-treated tumors. However, apparent alterations in VEGFR-3 expression of podoplanin-positive lymphatic endothelial cells were not observed in IL-12-treated tumors. Although recombinant IL-12 did not inhibit tubular formation of human umbilical vein endothelial cells in a Matrigel assay, recombinant IFNgamma did completely suppress the tubular formation. CONCLUSIONS In vivo electrogene transfer of IL-12 exerts strong anti-tumorigenic and anti-metastatic effects likely due to T-cell-mediated immune responses as well as anti-angiogenic action.
Collapse
Affiliation(s)
- Masa-Aki Shibata
- Department of Anatomy and Biology, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | | | | | | | | | | |
Collapse
|
25
|
Liu XY, Qiu SB, Zou WG, Pei ZF, Gu JF, Luo CX, Ruan HM, Chen Y, Qi YP, Qian C. Effective gene-virotherapy for complete eradication of tumor mediated by the combination of hTRAIL (TNFSF10) and plasminogen k5. Mol Ther 2005; 11:531-41. [PMID: 15771956 DOI: 10.1016/j.ymthe.2004.12.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2004] [Accepted: 12/06/2004] [Indexed: 11/27/2022] Open
Abstract
Virotherapy with oncolytic viruses is a highly promising approach for cancer therapy. To improve further the therapeutic effect of oncolytic viruses, therapeutic genes have been incorporated into these types of vectors. In this study, we have inserted hTRAIL (approved gene symbol TNFSF10) into the ZD55 vector, which was based on deletion of the adenoviral E1B 55-kDa gene and could replicate in and lyse p53-deficient tumors. Our data shows that infection of colorectal carcinoma cells with ZD55-hTRAIL resulted in tumor cell death that was much greater than that induced by ZD55 vector or replication-defective adenovirus expressing hTRAIL. In contrast to these, ZD55-hTRAIL did not induce any cytopathic effect in normal cells. Treatment of established tumor with ZD55-hTRAIL resulted in dramatic inhibition of tumor growth in an animal model of colorectal carcinoma. However, when the established tumors were treated by coadministration of ZD55-hTRAIL and Ad-k5, we observed complete eradication of the established tumors in all animals treated with the combined therapy. This strong anti-tumor activity was due to the fact that two genes may act with compensative (or synergic) effect through different mechanisms to kill tumors. Therefore, targeting dual gene-virotherapy may be one of the best strategies for cancer therapy if two suitable genes are chosen.
Collapse
Affiliation(s)
- Xin-Yuan Liu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Prieto J, Qian C, Hernandez-Alcoceba R, Gonzalez-Aseguinolaza G, Mazzolini G, Sangro B, Kramer MG. Gene therapy of liver diseases. Expert Opin Biol Ther 2005; 4:1073-91. [PMID: 15268675 DOI: 10.1517/14712598.4.7.1073] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Many liver diseases lack satisfactory treatment and alternative therapeutic options are urgently needed. Gene therapy is a new mode of treatment for both inherited and acquired diseases, based on the transfer of genetic material to the tissues. Genes are incorporated into appropriate vectors in order to facilitate their entrance and function inside the target cells. Gene therapy vectors can be constructed on the basis of viral or non-viral molecular structures. Viral vectors are frequently used, due to their higher transduction efficiency. Both the type of vector and the expression cassette determine the duration, specificity and inducibility of gene expression. A considerable number of preclinical studies indicate that a great variety of liver diseases, including inherited metabolic defects, chronic viral hepatitis, liver cirrhosis and primary and metastatic liver cancer, are amenable to gene therapy. Gene transfer to the liver can also be used to convert this organ into a factory of secreted proteins needed to treat conditions that do not affect the liver itself. Clinical trials of gene therapy for the treatment of inherited diseases and liver cancer have been initiated but human gene therapy is still in its infancy. Recent progress in vector technology and imaging techniques, allowing in vivo assessment of gene expression, will facilitate the development of clinical applications of gene therapy.
Collapse
Affiliation(s)
- Jesus Prieto
- Department of Internal Medicine, Clinica Universitaria de Navarra, Avda. Pio XII 36, 31008 Pamplona, Spain
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
A large proportion of patients with advanced hepatocellular carcinoma (HCC) lack effective therapy. Due to chemoresistance, hope has focused on other approaches including targeted therapies, immune stimulants, and the emerging area of gene therapy. Increasing efforts in basic and clinical development of these approaches will hopefully result in more efficient therapies against HCC.
Collapse
Affiliation(s)
- Bruno Sangro
- The Liver Unit, Clínica Universitaria de Navarra, Fundación para la Investigación Médica Aplicada, University of Navarra, Pamplona, Spain.
| | | | | |
Collapse
|
28
|
Abstract
Nonresectable primary and metastatic liver tumors are common malignancies that lack therapies allowing substantial prolongation of survival. Recent progress in molecular and cell biology has opened the way to novel therapies based on biological modifiers, gene transfer, and autologous stem cells. It is now possible to transfer therapeutic genes to the tumor or pericancerous tissue, and to control their expression for long periods of time. It is also feasible to generate autologous endothelial progenitor cells that can be recruited by tumoral vessels acting as vehicles to convey therapeutic genes to the interior of the tumor mass. Combination of biological modifiers, gene therapy, and cell therapy will hopefully provide efficient means to combat inoperable neoplasms in a not-very-distant future.
Collapse
Affiliation(s)
- Jesus Prieto
- Division of Hepatology and Gene Therapy, Fundación para la Investigación Médica Aplicada (FIMA), University of Navarra, Avenida Pio XII s/n, 31080 Pamplona, Spain.
| | | | | | | | | |
Collapse
|
29
|
Casares N, Arribillaga L, Sarobe P, Dotor J, Lopez-Diaz de Cerio A, Melero I, Prieto J, Borrás-Cuesta F, Lasarte JJ. CD4+/CD25+ Regulatory Cells Inhibit Activation of Tumor-Primed CD4+ T Cells with IFN-γ-Dependent Antiangiogenic Activity, as well as Long-Lasting Tumor Immunity Elicited by Peptide Vaccination. THE JOURNAL OF IMMUNOLOGY 2003; 171:5931-9. [PMID: 14634104 DOI: 10.4049/jimmunol.171.11.5931] [Citation(s) in RCA: 165] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD25(+) regulatory T (T reg) cells suppress the activation/proliferation of other CD4(+) or CD8(+) T cells in vitro. Also, down-regulation of CD25(+) T reg cells enhance antitumor immune responses. In this study, we show that depletion of CD25(+) T reg cells allows the host to induce both CD4(+) and CD8(+) antitumoral responses following tumor challenge. Simultaneous depletion of CD25(+) and CD8(+) cells, as well as adoptive transfer experiments, revealed that tumor-specific CD4(+) T cells, which emerged in the absence of CD25(+) T reg cells, were able to reject CT26 colon cancer cells, a MHC class II-negative tumor. The antitumoral effect mediated by CD4(+) T cells was dependent on IFN-gamma production, which exerted a potent antiangiogenic activity. The capacity of the host to mount this antitumor response is lost once the number of CD25(+) T reg cells is restored over time. However, CD25(+) T reg cell depletion before immunization with AH1 (a cytotoxic T cell determinant from CT26 tumor cells) permits the induction of a long-lasting antitumoral immune response, not observed if immunization is conducted in the presence of regulatory cells. A study of the effect of different levels of depletion of CD25(+) T reg cells before immunization with the peptide AH1 alone, or in combination with a Th determinant, unraveled that Th cells play an important role in overcoming the suppressive effect of CD25(+) T reg on the induction of long-lasting cellular immune responses.
Collapse
Affiliation(s)
- Noelia Casares
- Department of Internal Medicine, Medical School, University Clinic and Fundación para la Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|