1
|
Lu SC, Hansen MJ, Hemsath JR, Parrett BJ, Zell BN, Barry MA. Modulating Oncolytic Adenovirus Immunotherapy by Driving Two Axes of the Immune System by Expressing 4-1BBL and CD40L. Hum Gene Ther 2022; 33:250-261. [PMID: 34731019 PMCID: PMC11981553 DOI: 10.1089/hum.2021.197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Oncolytic viruses (OVs) can have utility for direct killing of cancer cells, but may also serve to activate the immune system against cancer cells. While viruses alone can serve as immune stimulators, there is great interest in arming OVs with genes encoding immune stimulatory proteins to amplify their effects. In this work, we have tested the efficacy of conditionally-replicating adenoviruses (CRAds) with and without selected immunostimulatory payloads, murine CD40L (mCD40L) or 4-1BBL (m4-1BBL), in an immune competent mouse model of melanoma. When CRAd657-m4-1BBL and CRAd657-mCD40L were injected into B16-hCAR murine melanoma tumors, both single vectors delayed tumor growth and prolong survival compared to empty CRAd657. However, combined injection of both CRAd-m4-1BBL and CRAd-mCD40L mediated significantly better control of tumor growth. All of the payloads increased immune cell infiltration into tumors and notably reduced expression of PD-1 exhaustion marker on T cells. Tumor volumes were negatively associated with total infiltrating T cell population. We found that the payloads increased immune cell infiltration into tumors with some specificities: recruitment of CD8+ T cells was higher with m4-1BBL expression, while mCD40L expression induced more CD4+ T cell infiltration. Importantly, the combination of CRAd657-m4-1BBL and CRAd657-mCD40L induced the highest immune cells/T cell infiltration and the highest anti-TRP-2 tumor-associated antigen T cell responses than empty or single gene vector. This combination also caused depigmentation in areas adjacent to the tumor sites in more animals. These data indicate that driving two axes of the immune system with combined immune stimulatory payloads can lead to improved anticancer immune responses and better tumor control in an immune competent model of cancer.
Collapse
Affiliation(s)
- Shao-Chia Lu
- Virology and Gene Therapy Graduate Program, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael J. Hansen
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Jack R. Hemsath
- Department of Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Brian J. Parrett
- Virology and Gene Therapy Graduate Program, Mayo Clinic, Rochester, Minnesota, USA
| | - Brady N. Zell
- Virology and Gene Therapy Graduate Program, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael A. Barry
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
2
|
Melero I, Castanon E, Alvarez M, Champiat S, Marabelle A. Intratumoural administration and tumour tissue targeting of cancer immunotherapies. Nat Rev Clin Oncol 2021; 18:558-576. [PMID: 34006998 PMCID: PMC8130796 DOI: 10.1038/s41571-021-00507-y] [Citation(s) in RCA: 283] [Impact Index Per Article: 70.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2021] [Indexed: 02/04/2023]
Abstract
Immune-checkpoint inhibitors and chimeric antigen receptor (CAR) T cells are revolutionizing oncology and haematology practice. With these and other immunotherapies, however, systemic biodistribution raises safety issues, potentially requiring the use of suboptimal doses or even precluding their clinical development. Delivering or attracting immune cells or immunomodulatory factors directly to the tumour and/or draining lymph nodes might overcome these problems. Hence, intratumoural delivery and tumour tissue-targeted compounds are attractive options to increase the in situ bioavailability and, thus, the efficacy of immunotherapies. In mouse models, intratumoural administration of immunostimulatory monoclonal antibodies, pattern recognition receptor agonists, genetically engineered viruses, bacteria, cytokines or immune cells can exert powerful effects not only against the injected tumours but also often against uninjected lesions (abscopal or anenestic effects). Alternatively, or additionally, biotechnology strategies are being used to achieve higher functional concentrations of immune mediators in tumour tissues, either by targeting locally overexpressed moieties or engineering 'unmaskable' agents to be activated by elements enriched within tumour tissues. Clinical trials evaluating these strategies are ongoing, but their development faces issues relating to the administration methodology, pharmacokinetic parameters, pharmacodynamic end points, and immunobiological and clinical response assessments. Herein, we discuss these approaches in the context of their historical development and describe the current landscape of intratumoural or tumour tissue-targeted immunotherapies.
Collapse
Affiliation(s)
- Ignacio Melero
- Department of Immunology, Clínica Universidad de Navarra, Pamplona, Spain.
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain.
- Program for Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| | - Eduardo Castanon
- Department of Immunology, Clínica Universidad de Navarra, Pamplona, Spain
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Maite Alvarez
- Program for Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Stephane Champiat
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Université Paris Saclay, Gustave Roussy, Villejuif, France
- INSERM U1015, Gustave Roussy, Villejuif, France
- Biotherapies for In Situ Antitumor Immunization (BIOTHERIS), Centre d'Investigation Clinique INSERM CICBT1428, Villejuif, France
| | - Aurelien Marabelle
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Université Paris Saclay, Gustave Roussy, Villejuif, France.
- INSERM U1015, Gustave Roussy, Villejuif, France.
- Biotherapies for In Situ Antitumor Immunization (BIOTHERIS), Centre d'Investigation Clinique INSERM CICBT1428, Villejuif, France.
| |
Collapse
|
3
|
Heße C, Kollenda S, Rotan O, Pastille E, Adamczyk A, Wenzek C, Hansen W, Epple M, Buer J, Westendorf AM, Knuschke T. A Tumor-Peptide–Based Nanoparticle Vaccine Elicits Efficient Tumor Growth Control in Antitumor Immunotherapy. Mol Cancer Ther 2019; 18:1069-1080. [DOI: 10.1158/1535-7163.mct-18-0764] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/19/2018] [Accepted: 04/04/2019] [Indexed: 11/16/2022]
|
4
|
Manson G, Houot R. Next-generation immunotherapies for lymphoma: one foot in the future. Ann Oncol 2018; 29:588-601. [DOI: 10.1093/annonc/mdy032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
5
|
Aznar MA, Tinari N, Rullán AJ, Sánchez-Paulete AR, Rodriguez-Ruiz ME, Melero I. Intratumoral Delivery of Immunotherapy-Act Locally, Think Globally. THE JOURNAL OF IMMUNOLOGY 2017; 198:31-39. [PMID: 27994166 DOI: 10.4049/jimmunol.1601145] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 08/31/2016] [Indexed: 12/16/2022]
Abstract
Immune mechanisms have evolved to cope with local entry of microbes acting in a confined fashion but eventually inducing systemic immune memory. Indeed, in situ delivery of a number of agents into tumors can mimic in the malignant tissue the phenomena that control intracellular infection leading to the killing of infected cells. Vascular endothelium activation and lymphocyte attraction, together with dendritic cell-mediated cross-priming, are the key elements. Intratumoral therapy with pathogen-associated molecular patterns or recombinant viruses is being tested in the clinic. Cell therapies can be also delivered intratumorally, including infusion of autologous dendritic cells and even tumor-reactive T lymphocytes. Intralesional virotherapy with an HSV vector expressing GM-CSF has been recently approved by the Food and Drug Administration for the treatment of unresectable melanoma. Immunomodulatory monoclonal Abs have also been successfully applied intratumorally in animal models. Local delivery means less systemic toxicity while focusing the immune response on the malignancy and the affected draining lymph nodes.
Collapse
Affiliation(s)
- M Angela Aznar
- Division of Immunology and Immunotherapy, Center for Applied Medical Research, University of Navarra, Pamplona 31008, Spain
| | - Nicola Tinari
- Department of Experimental and Clinical Sciences, G. D'Annunzio University and Foundation, Chieti 66100, Italy
| | - Antonio J Rullán
- Department of Medical Oncology, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona 08908, Spain; and
| | - Alfonso R Sánchez-Paulete
- Division of Immunology and Immunotherapy, Center for Applied Medical Research, University of Navarra, Pamplona 31008, Spain
| | - María E Rodriguez-Ruiz
- Division of Immunology and Immunotherapy, Center for Applied Medical Research, University of Navarra, Pamplona 31008, Spain.,University Clinic, University of Navarra and Health Research Institute of Navarra, 31008 Pamplona, Spain
| | - Ignacio Melero
- Division of Immunology and Immunotherapy, Center for Applied Medical Research, University of Navarra, Pamplona 31008, Spain; .,University Clinic, University of Navarra and Health Research Institute of Navarra, 31008 Pamplona, Spain
| |
Collapse
|
6
|
Rainone V, Martelli C, Ottobrini L, Biasin M, Borelli M, Lucignani G, Trabattoni D, Clerici M. Immunological Characterization of Whole Tumour Lysate-Loaded Dendritic Cells for Cancer Immunotherapy. PLoS One 2016; 11:e0146622. [PMID: 26795765 PMCID: PMC4721657 DOI: 10.1371/journal.pone.0146622] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/18/2015] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Dendritic cells play a key role as initiators of T-cell responses, and even if tumour antigen-loaded dendritic cells can induce anti-tumour responses, their efficacy has been questioned, suggesting a need to enhance immunization strategies. MATHERIALS & METHODS We focused on the characterization of bone marrow-derived dendritic cells pulsed with whole tumour lysate (TAA-DC), as a source of known and unknown antigens, in a mouse model of breast cancer (MMTV-Ras). Dendritic cells were evaluated for antigen uptake and for the expression of MHC class I/II and costimulatory molecules and markers associated with maturation. RESULTS Results showed that antigen-loaded dendritic cells are characterized by a phenotypically semi-mature/mature profile and by the upregulation of genes involved in antigen presentation and T-cell priming. Activated dendritic cells stimulated T-cell proliferation and induced the production of high concentrations of IL-12p70 and IFN-γ but only low levels of IL-10, indicating their ability to elicit a TH1-immune response. Furthermore, administration of Antigen loaded-Dendritic Cells in MMTV-Ras mice evoked a strong anti-tumour response in vivo as demonstrated by a general activation of immunocompetent cells and the release of TH1 cytokines. CONCLUSION Data herein could be useful in the design of antitumoral DC-based therapies, showing a specific activation of immune system against breast cancer.
Collapse
Affiliation(s)
- Veronica Rainone
- Department of Biomedical and Clinical Sciences “L. Sacco”, Chair of Immunology, University of Milan, Milan, Italy
| | - Cristina Martelli
- Department of Pathophysiology and Transplantation, University of Milan, Segrate, Milan, Italy
- Centre of Molecular and Cellular Imaging—IMAGO, University of Milan, Milan, Italy
| | - Luisa Ottobrini
- Department of Pathophysiology and Transplantation, University of Milan, Segrate, Milan, Italy
- Centre of Molecular and Cellular Imaging—IMAGO, University of Milan, Milan, Italy
- Institute for Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Segrate, Milan, Italy
| | - Mara Biasin
- Department of Biomedical and Clinical Sciences “L. Sacco”, Chair of Immunology, University of Milan, Milan, Italy
| | - Manuela Borelli
- Department of Biomedical and Clinical Sciences “L. Sacco”, Chair of Immunology, University of Milan, Milan, Italy
| | - Giovanni Lucignani
- Centre of Molecular and Cellular Imaging—IMAGO, University of Milan, Milan, Italy
- Departments of Health Sciences, University of Milan, Milan, Italy
- Department of Diagnostic Services, Unit of Nuclear Medicine, San Paolo Hospital, Milan, Italy
| | - Daria Trabattoni
- Department of Biomedical and Clinical Sciences “L. Sacco”, Chair of Immunology, University of Milan, Milan, Italy
| | - Mario Clerici
- Department of Pathophysiology and Transplantation, University of Milan, Segrate, Milan, Italy
- Don C. Gnocchi Foundation IRCCS, Milan, Italy
| |
Collapse
|
7
|
Van der Jeught K, Bialkowski L, Daszkiewicz L, Broos K, Goyvaerts C, Renmans D, Van Lint S, Heirman C, Thielemans K, Breckpot K. Targeting the tumor microenvironment to enhance antitumor immune responses. Oncotarget 2015; 6:1359-81. [PMID: 25682197 PMCID: PMC4359300 DOI: 10.18632/oncotarget.3204] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 12/24/2014] [Indexed: 12/16/2022] Open
Abstract
The identification of tumor-specific antigens and the immune responses directed against them has instigated the development of therapies to enhance antitumor immune responses. Most of these cancer immunotherapies are administered systemically rather than directly to tumors. Nonetheless, numerous studies have demonstrated that intratumoral therapy is an attractive approach, both for immunization and immunomodulation purposes. Injection, recruitment and/or activation of antigen-presenting cells in the tumor nest have been extensively studied as strategies to cross-prime immune responses. Moreover, delivery of stimulatory cytokines, blockade of inhibitory cytokines and immune checkpoint blockade have been explored to restore immunological fitness at the tumor site. These tumor-targeted therapies have the potential to induce systemic immunity without the toxicity that is often associated with systemic treatments. We review the most promising intratumoral immunotherapies, how these affect systemic antitumor immunity such that disseminated tumor cells are eliminated, and which approaches have been proven successful in animal models and patients.
Collapse
Affiliation(s)
- Kevin Van der Jeught
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Lukasz Bialkowski
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Lidia Daszkiewicz
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Katrijn Broos
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Cleo Goyvaerts
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Dries Renmans
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Sandra Van Lint
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Carlo Heirman
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| |
Collapse
|
8
|
Abstract
Dendritic cells (DCs) are the most powerful professional antigen-presenting cells and are unique in their capability to initiate, maintain and regulate the intensity of primary immune responses, including specific antitumor responses. Development of practical procedures to prepare sufficient numbers of functional human DCs in culture from the peripheral blood precursors, paved the way for clinical trials to evaluate various DC-based strategies in patients with malignant diseases. However, no definite conclusions regarding the clinical and even immunological efficacy of DC vaccination can be stated, despite the fact that 12 years have passed since the first clinical trial utilizing DCs in cancer patients. Many unanswered questions hamper the development of DC-based vaccines, including the source of DC preparation and protocols for DC generation, activation and loading with tumor antigens, source of tumor antigens, route of vaccine administration and methods of immunomonitoring. Fortunately, in spite of the many obstacles, DC vaccines continue to hold promise for cancer therapy.
Collapse
Affiliation(s)
- Hua Zhong
- Shanghai Jiao Tong University, Shanghai Chest Hospital, 241 Huaihai Road (w), Shanghai 200030, China.
| | | | | |
Collapse
|
9
|
Kaur P, Asea A. Radiation-induced effects and the immune system in cancer. Front Oncol 2012; 2:191. [PMID: 23251903 PMCID: PMC3523399 DOI: 10.3389/fonc.2012.00191] [Citation(s) in RCA: 162] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 11/23/2012] [Indexed: 12/31/2022] Open
Abstract
Chemotherapy and radiation therapy (RT) are standard therapeutic modalities for patients with cancers, and could induce various tumor cell death modalities, releasing tumor-derived antigens as well as danger signals that could either be captured for triggering anti-tumor immune response. Historic studies examining tissue and cellular responses to RT have predominantly focused on damage caused to proliferating malignant cells leading to their death. However, there is increasing evidence that RT also leads to significant alterations in the tumor microenvironment, particularly with respect to effects on immune cells and infiltrating tumors. This review will focus on immunologic consequences of RT and discuss the therapeutic reprogramming of immune responses in tumors and how it regulates efficacy and durability to RT.
Collapse
Affiliation(s)
- Punit Kaur
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine Atlanta, GA, USA
| | | |
Collapse
|
10
|
KHRANOVSKAYA NATALYA, OREL VALERII, GRINEVICH YURIY, ALEKSEENKO OXANA, ROMANOV ANDRIY, SKACHKOVA OXANA, DZYATKOVSKAYA NATALYA, BURLAKA ANATOLIY, LUKIN SERGEY. MECHANICAL HETEROGENIZATION OF LEWIS LUNG CARCINOMA CELLS CAN IMPROVE ANTIMETASTATIC EFFECT OF DENDRITIC CELLS. J MECH MED BIOL 2012. [DOI: 10.1142/s0219519411004757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The effect of mechanically heterogenized (MCH) microparticles of tumor cells (TCs) on antimetastatic action of dendritic cells (DCs) is studied in C57BL/6 mice with Lewis' carcinoma. DCs isolated from mice spleens and loaded with MCH-TCs are analyzed with flow cytometry methods. MCH-TCs are analyzed with optical and/or electron microscopy. The paper describes an original high-precision medical microvibromill with high-acceleration linear induction motor that generates magnetic levitation to produce mechanical heterogenization of TCs. MCH-TCs have a more asymmetric morphology, larger surface and higher internal structure heterogeneity, and higher concentration of free radicals with respect to conventionally treated TCs. The rate of DCs maturity, being affected by pre-incubation with MCH-TCs is found to be higher than its counterpart treated with conventional TCs. DCs loaded with MCH-TCs show a significantly higher ability to induce proliferation of allogeneic lymphocytes in mixed leukocyte reaction. The inhibition index of metastases formation increases from 42% (conventional TCs) to 66% when DCs are treated with MCH-TCs. The present results demonstrate the feasibility of increasing antimetastatic activity of DCs-based vaccines when MCH-TCs is used for their loading. Mathematical model is developed in order to simulate the processes of capture, processing and presentation of tumor antigens by DCs when using conventional TCs or MCH-TCs.
Collapse
Affiliation(s)
- NATALYA KHRANOVSKAYA
- Experimental Oncology Laboratory, National Cancer Institute, 33/43 Lomonosov Street, Kyiv, 03022, Ukraine
| | - VALERII OREL
- Medical Physics & Bioengineering Laboratory, National Cancer Institute, 33/43 Lomonosov Street, Kyiv, 03022, Ukraine
| | - YURIY GRINEVICH
- Clinical Immunology Laboratory, National Cancer Institute, 33/43 Lomonosov Street, Kyiv, 03022, Ukraine
| | - OXANA ALEKSEENKO
- Cytology Laboratory, National Cancer Institute, 33/43 Lomonosov Street, Kyiv, 03022, Ukraine
| | - ANDRIY ROMANOV
- Medical Physics & Bioengineering Laboratory, National Cancer Institute, 33/43 Lomonosov Street, Kyiv, 03022, Ukraine
| | - OXANA SKACHKOVA
- Experimental Oncology Laboratory, National Cancer Institute, 33/43 Lomonosov Street, Kyiv, 03022, Ukraine
| | - NATALYA DZYATKOVSKAYA
- Medical Physics & Bioengineering Laboratory, National Cancer Institute, 33/43 Lomonosov Street, Kyiv, 03022, Ukraine
| | - ANATOLIY BURLAKA
- R. E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of National Academy of Sciences of Ukraine, 45, Vasil'kovsky Street, Kyiv, 03022, Ukraine
| | - SERGEY LUKIN
- R. E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology of National Academy of Sciences of Ukraine, 45, Vasil'kovsky Street, Kyiv, 03022, Ukraine
| |
Collapse
|
11
|
Carcinoma-derived interleukin-8 disorients dendritic cell migration without impairing T-cell stimulation. PLoS One 2011; 6:e17922. [PMID: 21423807 PMCID: PMC3056721 DOI: 10.1371/journal.pone.0017922] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Accepted: 02/17/2011] [Indexed: 01/14/2023] Open
Abstract
Background Interleukin-8 (IL-8, CXCL8) is readily produced by human malignant cells.
Dendritic cells (DC) both produce IL-8 and express the IL-8 functional
receptors CXCR1 and CXCR2. Most human colon carcinomas produce IL-8. IL-8
importance in malignancies has been ascribed to angiogeneis promotion. Principal Findings IL-8 effects on human monocyte-derived DC biology were explored upon DC
exposure to recombinant IL-8 and with the help of an IL-8 neutralizing mAb.
In vivo experiments were performed in immunodeficient
mice xenografted with IL-8-producing human colon carcinomas and
comparatively with cell lines that do not produce IL-8. Allogenic T
lymphocyte stimulation by DC was explored under the influence of IL-8. DC
and neutrophil chemotaxis were measured by transwell-migration assays. Sera
from tumor-xenografted mice contained increasing concentrations of IL-8 as
the tumors progress. IL-8 production by carcinoma cells can be modulated by
low doses of cyclophosphamide at the transcription level. If human DC are
injected into HT29 or CaCo2 xenografted tumors, DC are retained
intratumorally in an IL-8-dependent fashion. However, IL-8 did not modify
the ability of DC to stimulate T cells. Interestingly, pre-exposure of DC to
IL-8 desensitizes such cells for IL-8-mediated in vitro or
in vivo chemoattraction. Thereby DC become disoriented
to subsequently follow IL-8 chemotactic gradients towards malignant or
inflamed tissue. Conclusions IL-8 as produced by carcinoma cells changes DC migration cues, without
directly interfering with DC-mediated T-cell stimulation.
Collapse
|
12
|
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: 1.8] [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
|
13
|
Dubrot J, Palazón A, Alfaro C, Azpilikueta A, Ochoa MC, Rouzaut A, Martinez-Forero I, Teijeira A, Berraondo P, Le Bon A, Hervás-Stubbs S, Melero I. Intratumoral injection of interferon-α and systemic delivery of agonist anti-CD137 monoclonal antibodies synergize for immunotherapy. Int J Cancer 2010; 128:105-18. [PMID: 20309938 DOI: 10.1002/ijc.25333] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
CD137 artificial costimulation results in complete tumor rejection in several mouse models. Type I interferons (IFN) exert antitumor effects through an array of molecular functions on malignant cells, tumor stroma and immune system cells. The fact that agonist anti-CD137 mAb induce tumor regressions in mice deficient in the unique receptor for Type I IFNs (IFNAR(-/-) ) indicated potential for treatment combinations. Indeed, combination of intratumor injections of mouse IFN-α and intraperitoneal injections of anti-CD137 mAb synergized as seen on subcutaneous lesions derived from the MC38 colon carcinoma, which is resistant to each treatment if given separately. Therapeutic activity was achieved both against lesions directly injected with IFN-α and against distant concomitant tumors. Experiments in bone marrow chimeras prepared with IFNAR(-/-) and WT mice concluded that expression of the receptor for Type I interferons is mainly required on cells of the hematopoietic compartment. Synergistic effects correlated with a remarkable cellular hyperplasia of the tumor draining lymph nodes (TDLNs). Enlarged TDLNs contained more plasmacytoid and conventional dendritic cells (DC) that more readily cross-presented. Importantly, numbers of both DC subtypes inversely correlated with the tumor size. Numbers of CD8 T cells specific for a dominant tumor antigen were increased at TDLNs by each separate treatment but only with slight augments due to the combination. Combined antitumor effects of the therapeutic strategy were also seen on subcutaneous TC-1 tumors established for 24 days before treatment onset. The described strategy is realistic because (i) agents of each kind are clinically available and (ii) equivalent procedures in humans are feasible.
Collapse
Affiliation(s)
- Juan Dubrot
- CIMA and Clinica Universitaria, Universidad de Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Dubrot J, Milheiro F, Alfaro C, Palazón A, Martinez-Forero I, Perez-Gracia JL, Morales-Kastresana A, Romero-Trevejo JL, Ochoa MC, Hervás-Stubbs S, Prieto J, Jure-Kunkel M, Chen L, Melero I. Treatment with anti-CD137 mAbs causes intense accumulations of liver T cells without selective antitumor immunotherapeutic effects in this organ. Cancer Immunol Immunother 2010; 59:1223-33. [PMID: 20336294 PMCID: PMC11030554 DOI: 10.1007/s00262-010-0846-9] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Accepted: 03/04/2010] [Indexed: 11/26/2022]
Abstract
BACKGROUND/AIMS Cancer therapy with agonist anti-CD137 mAbs has been shown to induce immune-mediated tumor rejections in mice, and equivalent agents of this kind are currently being tested in cancer patients. Previous reports indicated that CD137 stimulation induced polyclonal infiltrates of T lymphocytes in the liver. This study characterizes the liver infiltrates and the target dependency of the phenomena and addresses the question of whether tumors nested in the liver are a more favorable target for CD137-based immunotherapy. METHODS Liver infiltrates were studied with conventional histology and multiple color flow cytometry of total liver leukocytes. CD137(-/-) mice, mice with a single rearrangement of the TCR (OT-1 mice) and Rag(-/-) mice were used to clarify molecular requirements. Mice implanted with MC38 colon carcinomas either subcutaneously or inside the liver were used for comparative studies under treatment with agonist anti-CD137 mAbs. RESULTS CD137 treatment caused mononuclear inflammation in the portal spaces of the liver, which gave rise to moderate increases in transaminases without signs of cholestasis. Marked increases in the numbers of CD8+ T cells were observed, including CD8+ T lymphocytes co-expressing CD11c. Infiltrates were absent in CD137(-/-) mice and mitigated in mice harboring a single transgenic TCR on their CD8 T cells. Despite the tumor-independent accumulation of T cells in the liver, immunotherapeutic effects were not more prominent against tumors located in this organ. CONCLUSIONS Target-dependent effects of CD137 stimulation lead to liver infiltration with T cells, but lymphocyte enrichment in this organ does not privilege this site for immunotherapeutic effects against transplanted tumors.
Collapse
MESH Headings
- Amidinotransferases/immunology
- Amidinotransferases/metabolism
- Animals
- Antibodies, Monoclonal/administration & dosage
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cell Count
- Cell Line, Tumor
- Cell Movement/drug effects
- Colonic Neoplasms/immunology
- Colonic Neoplasms/pathology
- Colonic Neoplasms/therapy
- Immunotherapy
- Liver/drug effects
- Liver/immunology
- Liver/metabolism
- Liver/pathology
- Lymphocytes, Tumor-Infiltrating/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Neoplasm Transplantation
- Organ Specificity
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
Collapse
Affiliation(s)
- Juan Dubrot
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Av. Pio XII, 55, 31008 Pamplona, Spain
| | - Francisca Milheiro
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Av. Pio XII, 55, 31008 Pamplona, Spain
| | - Carlos Alfaro
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Av. Pio XII, 55, 31008 Pamplona, Spain
| | - Asis Palazón
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Av. Pio XII, 55, 31008 Pamplona, Spain
| | - Ivan Martinez-Forero
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Av. Pio XII, 55, 31008 Pamplona, Spain
| | | | - Aizea Morales-Kastresana
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Av. Pio XII, 55, 31008 Pamplona, Spain
| | - José L. Romero-Trevejo
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Av. Pio XII, 55, 31008 Pamplona, Spain
| | - María C. Ochoa
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Av. Pio XII, 55, 31008 Pamplona, Spain
| | - Sandra Hervás-Stubbs
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Av. Pio XII, 55, 31008 Pamplona, Spain
| | - Jesús Prieto
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Av. Pio XII, 55, 31008 Pamplona, Spain
- Clínica Universitaria, Universidad de Navarra, Pamplona, Spain
| | - Maria Jure-Kunkel
- Bristol Myers-Squibb Pharmaceutical Research Institute, Princeton, NJ USA
| | - Lieping Chen
- Sidney Kimmel Cancer Center, Johns Hopkins Medical School, Baltimore, MD USA
| | - Ignacio Melero
- Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Av. Pio XII, 55, 31008 Pamplona, Spain
- Clínica Universitaria, Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
15
|
Abstract
Advances in the understanding of the immunoregulatory functions of dendritic cells (DCs) in animal models and humans have led to their exploitation as anticancer vaccines. Although DC-based immunotherapy has proven clinically safe and efficient to induce tumor-specific immune responses, only a limited number of objective clinical responses have been reported in cancer patients. These relatively disappointing results have prompted the evaluation of multiple approaches to improve the efficacy of DC vaccines. The topic of this review focuses on personalized DC-based anticancer vaccines, which in theory have the potential to present to the host immune system the entire repertoire of antigens harbored by autologous tumor cells. We also discuss the implementation of these vaccines in cancer therapeutic strategies, their limitations and the future challenges for effective immunotherapy against cancer.
Collapse
Affiliation(s)
- Nona Janikashvili
- Department of Pediatrics, Steele Children’s Research Center, Arizona 85724, USA
| | - Nicolas Larmonier
- Department of Pediatrics, Steele Children’s Research Center, Arizona 85724, USA
- Department of Immunobiology, BIO5 Institute & Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Emmanuel Katsanis
- Department of Pediatrics, Steele Children’s Research Center, Arizona 85724, USA
- Department of Immunobiology, BIO5 Institute & Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
- University of Arizona, Department of Pediatrics, 1501 N Campbell Ave, PO Box 245073, Tucson, AZ 85724-85073, USA
| |
Collapse
|
16
|
Fu W, Lan H, Liang S, Gao T, Ren D. Suicide gene/prodrug therapy using salmonella-mediated delivery of Escherichia coli purine nucleoside phosphorylase gene and 6-methoxypurine 2'-deoxyriboside in murine mammary carcinoma 4T1 model. Cancer Sci 2008; 99:1172-9. [PMID: 18429958 PMCID: PMC11158249 DOI: 10.1111/j.1349-7006.2008.00808.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2007] [Revised: 02/18/2008] [Accepted: 02/20/2008] [Indexed: 11/26/2022] Open
Abstract
Attenuated salmonella have been reported to be capable of both selectively growing in tumors and expressing exogenous genes for tumor-targeted therapy. As 6-methoxypurine 2'-deoxyriboside (MoPdR) is similar to 6-methylpurine 2'-deoxyriboside in structure, we aimed to evaluate the antitumoral effect of the Escherichia coli purine nucleoside phosphorylase (ePNP) gene, using an attenuated salmonella-mediated delivery system, in combination with MoPdR. A novel mutant serovar Typhimurium (SC36) was used to carry the pEGFP-C1-ePNP vector that contains an enhanced green fluorescent protein and an ePNP gene under the control of the cytomegalovirus promoter. The function of the ePNP expression vector was confirmed in vitro using the enzymic conversion of MoPdR into methoxypurine. We also observed a high bystander effect induced by the ePNP/MoPdR system with a very low proportion (1%) of ePNP-positive cells and 5 microg/mL MoPdR, although the growth of parental cells was affected appreciably by MoPdR. The killing effect and increased apoptosis induced by SC36 carrying the ePNP expression vector (SC/ePNP) were detected by cytotoxicity assay and propidium iodide staining flow cytometry analysis, in combination with MoPdR. SC/ePNP was given orally to mice bearing mammary carcinomas, and its antitumor effect was evaluated. SC/ePNP plus MoPdR significantly inhibited tumor growth by approximately 86.6-88.7% and prolonged the survival of tumor-hosting mice. Our data support the view that MoPdR combined with the ePNP gene could be used in gene-directed enzyme prodrug therapy. Attenuated salmonella could be a promising strategy to improve ePNP/MoPdR bystander killing due to its preferential accumulation and anticancer activity in tumors.
Collapse
MESH Headings
- Animals
- Apoptosis/physiology
- Blotting, Western
- Bystander Effect
- Caspase 3/metabolism
- Cell Proliferation
- Combined Modality Therapy
- Escherichia coli/enzymology
- Female
- Genes, Transgenic, Suicide
- Genetic Therapy
- Genetic Vectors
- Green Fluorescent Proteins/genetics
- Humans
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Prodrugs/therapeutic use
- Promoter Regions, Genetic
- Purine Nucleosides/therapeutic use
- Purine-Nucleoside Phosphorylase/genetics
- Salmonella/genetics
- Transcription, Genetic
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Wei Fu
- The State Key Laboratory of Genetic Engineering, Institute of Genetics, Fudan University, 220 Han Dan Road, Shanghai 200433, China
| | | | | | | | | |
Collapse
|
17
|
Shurin MR, Shurin GV, Lokshin A, Yurkovetsky ZR, Gutkin DW, Chatta G, Zhong H, Han B, Ferris RL. Intratumoral cytokines/chemokines/growth factors and tumor infiltrating dendritic cells: friends or enemies? Cancer Metastasis Rev 2007; 25:333-56. [PMID: 17029028 DOI: 10.1007/s10555-006-9010-6] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The tumor microenvironment consists of a variable combination of tumor cells, stromal fibroblasts, endothelial cells and infiltrating leukocytes, such as macrophages, T lymphocytes, and dendritic cells. A variety of cytokines, chemokines and growth factors are produced in the local tumor environment by different cells accounting for a complex cell interaction and regulation of differentiation, activation, function and survival of multiple cell types. The interaction between cytokines, chemokines, growth factors and their receptors forms a comprehensive network at the tumor site, which is primary responsible for overall tumor progression and spreading or induction of antitumor immune responses and tumor rejection. Although the general thought is that dendritic cells are among the first cells migrating to the tumor site and recognizing tumor cells for the induction of specific antitumor immunity, the clinical relevance of dendritic cells at the site of the tumor remains a matter of debate regarding their role in the generation of successful antitumor immune responses in human cancers. While several lines of evidence suggest that intratumoral dendritic cells play an important role in antitumor immune responses, understanding the mechanisms of dendritic cell/tumor cell interaction and modulation of activity and function of different dendritic cell subtypes at the tumor site is incomplete. This review is limited to discussing the role of intratumoral cytokine network in the understanding immunobiology of tumor-associated dendritic cells, which seems to possess different regulatory functions at the tumor site.
Collapse
Affiliation(s)
- Michael R Shurin
- Department of Pathology, University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Chen Z, Xia D, Bi X, Saxena A, Sidhu N, El-Gayed A, Xiang J. Combined radiation therapy and dendritic cell vaccine for treating solid tumors with liver micro-metastasis. J Gene Med 2005; 7:506-17. [PMID: 15580588 DOI: 10.1002/jgm.692] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Tumor metastasis and relapse are major obstacles in combating human malignant diseases. Neither radiotherapy alone nor injection of dendritic cells (DCs) can successfully overcome this problem. Radiation induces tumor cell apoptosis and necrosis, resulting in the release of tumor antigen and danger signals, which are favorable for DC capturing antigens and maturation. Hence, the strategy of combined irradiation and DC vaccine may be a novel approach for treating human malignancies and early metastasis. METHODS To develop an effective combined therapeutic approach, we established a novel concomitant local tumor and liver metastases model through subcutaneous (s.c.) and intravenous (i.v.) injection. We selected the optimal time for DC injection after irradiation and investigated the antitumor effect of combining irradiation with DC intratumoral injection and the related mechanism. RESULTS Combined treatment with radiotherapy and DC vaccine could induce a potent antitumor immune response, resulting in a significant decrease in the rate of local tumor relapse and the numbers of liver metastases. The related mechanisms for this strong antitumor immunity of this combined therapy might be associated with the production of apoptotic and necrotic tumor antigens and heat shock proteins after irradiation, phagocytosis, migration and maturation of DCs, and induction of more efficient tumor-specific cytotoxic T lymphocyte activity through a cross-presentation pathway. CONCLUSIONS Co-administration of local irradiation and intratumoral DC injection may be a promising strategy for treating radiosensitive tumors and eliminating metastasis in the clinic.
Collapse
Affiliation(s)
- Zhuang Chen
- Department of Oncology, Saskatoon Cancer Center, College of Medicine, University of Saskatchewan, 20 Campus Drive, Saskatoon, Saskatchewan S7N 4H4, Canada
| | | | | | | | | | | | | |
Collapse
|
19
|
Graner MW, Bigner DD. Chaperone proteins and brain tumors: potential targets and possible therapeutics. Neuro Oncol 2005; 7:260-78. [PMID: 16053701 PMCID: PMC1871914 DOI: 10.1215/s1152851704001188] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Chaperone proteins are most notable for the proteo- and cyotoprotective capacities they afford during cellular stress. Under conditions of cellular normalcy, chaperones still play integral roles in the folding of nascent polypeptides into functional entities, in assisting in intracellular/intraorganellar transport, in assembly and maintenance of multi-subunit protein complexes, and in aiding and abetting the degradation of senescent proteins. Tumors frequently have relatively enhanced needs for chaperone number and activity because of the stresses of rapid proliferation, increased metabolism, and overall genetic instability. Thus, it may be possible to take advantage of this reliance that tumor cells have on chaperones by pharmacologic and biologic means. Certain chaperones are abundant in the brain, which implies important roles for them. While it is presumed that the requirements of brain tumors for chaperone proteins are similar to those of any other cell type, tumor or otherwise, very little inquiry has been directed at the possibility of using chaperone proteins as therapeutic targets or even as therapeutic agents against central nervous system malignancies. This review highlights some of the research on the functions of chaperone proteins, on what can be done to modify those functions, and on the physiological responses that tumors and organisms can have to chaperone-targeted or chaperone-based therapies. In particular, this review will also underscore areas of research where brain tumors have been part of the field, although in general those instances are few and far between. This relative dearth of research devoted to chaperone protein targets and therapeutics in brain tumors reveals much untrodden turf to explore for potential treatments of these dreadfully refractive diseases.
Collapse
Affiliation(s)
- Michael W Graner
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA.
| | | |
Collapse
|
20
|
Feijoó E, Alfaro C, Mazzolini G, Serra P, Peñuelas I, Arina A, Huarte E, Tirapu I, Palencia B, Murillo O, Ruiz J, Sangro B, Richter JA, Prieto J, Melero I. Dendritic cells delivered inside human carcinomas are sequestered by interleukin-8. Int J Cancer 2005; 116:275-81. [PMID: 15800914 DOI: 10.1002/ijc.21046] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
In the course of a clinical trial consisting of intratumoral injections of dendritic cells (DCs) transfected to produce interleukin-12, the use of (111)In-labeled tracing doses of DCs showed that most DCs remained inside tumor tissue, instead of migrating out. In search for factors that could explain this retention, it was found that tumors from patients suffering hepatocellular carcinoma, colorectal or pancreatic cancer were producing IL-8 and that this chemokine attracted monocyte-derived dendritic cells that uniformly express both IL-8 receptors CXCR1 and CXCR2. Accordingly, neutralizing antihuman IL-8 monoclonal antibodies blocked the chemotactic attraction of DCs by recombinant IL-8, as well as by the serum of the patients or culture supernatants of human colorectal carcinomas. In addition, tissue culture supernatants of colon carcinoma cells inhibited DC migration induced by MIP-3beta in an IL-8-dependent fashion. IL-8 production in malignant tissue and the responsiveness of DCs to IL-8 are a likely explanation of the clinical images, which suggest retention of DCs inside human malignant lesions. Impairment of DC migration toward lymphoid tissue could be involved in cancer immune evasion.
Collapse
Affiliation(s)
- Esperanza Feijoó
- Division of Hepatology and Gene Therapy, Clínica Universitaria/School of Medicine, Foundation for Applied Medical Research, Universidad de Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Crittenden MR, Thanarajasingam U, Vile RG, Gough MJ. Intratumoral immunotherapy: using the tumour against itself. Immunology 2005; 114:11-22. [PMID: 15606790 PMCID: PMC1782057 DOI: 10.1111/j.1365-2567.2004.02001.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2004] [Revised: 09/16/2004] [Accepted: 09/17/2004] [Indexed: 12/23/2022] Open
Abstract
Summary Diverse immunotherapy approaches have achieved success in controlling individual aspects of immune responses in animal models. Transfer of such immunotherapies to clinical trials has obtained some success in patients, with clinical responses observed or effective antigen specific immune responses achieved, but has had limited impact on patient survival. Key elements required to generate de novo cell-mediated antitumour immune responses in vivo include recruitment of antigen-presenting cells to the tumour site, loading these cells with antigen, and their migration and maturation to full antigen-presenting function. In addition, it is essential for antigen-specific T cells to locate the tumour to mediate cytotoxicity, emphasizing the need for local inflammation to target effector cell recruitment. We review those therapies that involve the tumour site as a target and source of antigen for the initiation of immune responses, and discuss strategies to generate and co-ordinate an optimal cell-mediated immune response to control tumours locally.
Collapse
|
22
|
Shrivastava P, Singh SM, Singh N. Effect of thymosin alpha 1 on the antitumor activity of tumor-associated macrophage-derived dendritic cells. J Biomed Sci 2004; 11:623-30. [PMID: 15316138 DOI: 10.1007/bf02256128] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2003] [Accepted: 04/09/2004] [Indexed: 11/25/2022] Open
Abstract
We have previously suggested that thymosin alpha(1) (thyalpha1), an immunomodulating thymic hormone, can activate tumor-associated macrophages to a tumoricidal state in a murine model bearing a transplantable T-cell lymphoma of spontaneous origin designated as Dalton's lymphoma (DL). Since tumor-infiltrating dendritic cells (DC) also play an important role in the host's antitumor response and are as such in an immunocompromised state in a tumor-bearing host, in the present investigation we studied if thyalpha1 is able to influence the differentiation of tumor-associated macrophages (TAM) into DC with granulocyte macrophage colony stimulating factor (GM-CSF), interleukin (IL)-4 and tumor necrosis factor (TNF) and whether these TAM-derived DC show enhanced antitumor activity. It was observed that DC generated from thyalpha1-administered tumor-bearing mice showed augmented antitumor activity in vitro. Adoptive immunotherapy using TAM-derived DC showed a significant delay in the tumor growth and a prolongation of the survival time in tumor-bearing mice. DC obtained from TAM of thyalpha1-administered mice also produced an enhanced amount of cytokines like IL-1 and TNF-alpha. This is the first study of its kind regarding the effect of thyalpha1 on the differentiation of DC from TAM and the role of TAM-derived DC in tumor progression.
Collapse
|
23
|
Critchley RJ, Jezzard S, Radford KJ, Goussard S, Lemoine NR, Grillot-Courvalin C, Vassaux G. Potential therapeutic applications of recombinant, invasive E. coli. Gene Ther 2004; 11:1224-33. [PMID: 15141160 DOI: 10.1038/sj.gt.3302281] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
An invasive Escherichia coli expressing the inv gene from Yersinia pseudotuberculosis was used as a vector for protein delivery to mammalian epithelial cells. Upon incubation with beta1-integrin-expressing mammalian cells, the bacteria are internalized, allowing bacteria-encoded proteins to function from within the mammalian cell. These bacteria are eventually processed in the host phagosome where they are destroyed. Expression of listeriolysin O from Listeria monocytogenes in the bacterium and its subsequent release into the phagosome triggers the breakdown of the membrane, allowing the release of the bacterial content into the cytosol of host cells. Using this vector, we demonstrate delivery of a gene and intact, functional proteins into mammalian cells in which beta1-integrin is expressed and accessible. At a ratio of bacteria/mammalian cells compatible with the survival of the mammalian cells, protein delivery can be observed in the entire cell population in vitro, while gene transfer is far less efficient. Protein delivery can also be achieved in vivo in mouse tumour models and can be detected at least 96 h after inoculation. Functional, natural E. coli proteins are delivered in the process and can provide therapeutic benefit in vivo, when associated with prodrugs. This therapeutic effect is associated with infiltration of neutrophils, eosinophils, macrophages and to a lesser extent dendritic cells in the tumour mass.
Collapse
Affiliation(s)
- R J Critchley
- Cancer Research UK Molecular Oncology Unit, ICSM at Hammersmith Hospital, London, UK
| | | | | | | | | | | | | |
Collapse
|
24
|
Tirapu I, Arina A, Mazzolini G, Duarte M, Alfaro C, Feijoo E, Qian C, Chen L, Prieto J, Melero I. Improving efficacy of interleukin-12-transfected dendritic cells injected into murine colon cancer with anti-CD137 monoclonal antibodies and alloantigens. Int J Cancer 2004; 110:51-60. [PMID: 15054868 DOI: 10.1002/ijc.20093] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Intralesional administration of cultured dendritic cells (DCs) engineered to produce IL-12 by in vitro infection with recombinant adenovirus frequently displays eradicating efficacy against established subcutaneous tumors derived from the CT26 murine colon carcinoma cell line. The elicited response is mainly mediated by cytolytic T lymphocytes. In order to search for strategies that would enhance the efficacy of the therapeutic procedure against less immunogenic tumors, we moved onto malignancies derived from the inoculation of MC38 colon cancer cells that are less prone to undergo complete regression upon a single intratumoral injection of IL-12-secreting DCs. In this model, we found that repeated injections of such DCs, as opposed to a single injection, achieved better efficacy against both the injected and a distantly implanted tumor; that the use of semiallogeneic DCs that are mismatched in one MHC haplotype with the tumor host showed slightly better efficacy; and that the combination of this treatment with systemic injections of immunostimulatory anti-CD137 (4-1BB) monoclonal antibody achieved potent combined effects that correlated with the antitumor immune response measured in IFN-gamma ELISPOT assays. The elicited systemic immune response eradicates concomitant untreated lesions in most cases. Curative efficacy was also found against some tumors established for 2 weeks when these strategies were used in combination. These are preclinical pieces of evidence to be considered in order to enhance the therapeutic benefit of a strategy that is currently being tested in clinical trials. Supplementary Material for this article can be found on the International Journal of Cancer website at http://www.interscience.wiley.com/jpages/0020-7136/suppmat/index.html.
Collapse
Affiliation(s)
- Iñigo Tirapu
- Gene Therapy Division, Fundación para la Investigación Médica Aplicada, University of Navarre, Irunlarrea s/n 31008 Pamplona, Navarre, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Pastorino S, Carta L, Puppo M, Melillo G, Bosco MC, Varesio L. Picolinic acid- or desferrioxamine-inducible autocrine activation of macrophages engineered to produce IFNγ: an approach for gene therapy. Gene Ther 2004; 11:560-8. [PMID: 14961065 DOI: 10.1038/sj.gt.3302217] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Macrophage (Mphi)-based vectors are highly mobile cellular shuttles designed to deliver therapeutic genes within the tissues. We engineered a mouse Mphi cell line to express the murine interferon-gamma (IFNgamma) under the control of an inducible promoter containing the hypoxia-responsive element, which can be triggered by hypoxia and other stimuli. We show that this Mphi vector can be induced to produce IFNgamma under normoxic conditions by stimulation with picolinic acid (PA), a catabolite of tryptophan, or desferrioxamine (DFX), an iron-chelating drug. The Mphi vector responds to IFNgamma with the induction of IRF-1 and of other IFNgamma-inducible genes, the expression of Ia antigens and induction of phagocytic activity. Inducible nitric oxygen synthase gene expression, nitric oxide production, as well as TNFalpha secretion were enhanced by PA or DFX as the sole stimuli. None of the above responses could be triggered individually by PA or DFX in control, normal Mphi, indicating that the Mphi vector overcame the need for costimulatory molecules derived from the immune system for its full activation. Furthermore, we demonstrate that extracellular iron can downregulate such response, thereby identifying an additional tool for the fine tuning of the Mphi vector response to stimulation.
Collapse
Affiliation(s)
- S Pastorino
- Laboratory of Molecular Biology, G Gaslini Institute, Largo Gaslini, Genova, Italy
| | | | | | | | | | | |
Collapse
|
26
|
Arina A, Tirapu I, Alfaro C, Rodríguez-Calvillo M, Mazzolini G, Inogés S, López A, Feijoo E, Bendandi M, Melero I. Clinical implications of antigen transfer mechanisms from malignant to dendritic cells. exploiting cross-priming. Exp Hematol 2002; 30:1355-64. [PMID: 12482496 DOI: 10.1016/s0301-472x(02)00956-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Expansion and activation of cytolytic T lymphocytes bearing high-affinity T-cell receptors specific for tumor antigens is a major goal of active cancer immunotherapy. Physiologically, T cells receive promitotic and activating signals from endogenous professional antigen-presenting cells (APC) rather than directly from malignant cells. This phenomenon fits with the broader concept of cross-presentation that earlier was demonstrated for minor histocompatibility and viral antigens. Many mechanisms have been found to be capable of transferring antigenic material from malignant cells to APC so that it can be processed and subsequently presented by MHC class I molecules expressed on APC. Dendritic cells (DC) are believed to be the most relevant APC mediating cross-presentation because they can take up antigens from apoptotic, necrotic, and even intact tumor cells. There exist specific molecular mechanisms that ensure this transfer of antigenic material: 1) opsonization of apoptotic bodies; 2) receptors for released heat shock proteins carrying peptides processed intracellularly; 3) Fc receptors that uptake immunocomplexes and immunoglobulins; and 4) pinocytosis. DC have the peculiar capability of reentering the exogenously captured material into the MHC class I pathway. Exploitation of these pieces of knowledge is achieved by providing DC with complex mixtures of tumor antigens ex vivo and by agents and procedures that promote infiltration of malignant tissue by DC. The final outcome of DC cross-presentation could be T-cell activation (cross-priming) but also, and importantly, T-cell tolerance contingent upon the activation/maturation status of DC. Artificial enhancement of tumor antigen cross-presentation and control of the immune-promoting status of the antigen-presenting DC will have important therapeutic implications in the near future.
Collapse
Affiliation(s)
- Ainhoa Arina
- Centro de Investigación Médica Aplicada (CIMA), School of Medicine and Cell Therapy Area (Clínica Universitaria), University of Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
A better understanding of the biology of malignant cells and of the host immune system together with dramatic advances in technology have led to the design of innovative immune-mediated approaches to control neoplastic clones, including various haematological malignancies. One of the major problems with conventional cancer therapies is their inability to eradicate residual cancer cells that are resistant to therapy, hence immune intervention might improve the clinical outcome of patients. This mini-review will focus mainly on immunological approaches to the therapy of acute myeloid leukaemia (AML), a subset of a much larger family of leukaemias. Immune-mediated approaches ranging from allogeneic lymphocyte transplants to cytokine therapy, immune-gene therapy and vaccination by dendritic-cell-based vaccines will be discussed.
Collapse
Affiliation(s)
- Joanna Galea-Lauri
- Department of Molecular Medicine, GKT, School of Medicine, The Rayne Institute, London, UK.
| |
Collapse
|
28
|
Rodríguez-Calvillo M, Duarte M, Tirapu I, Berraondo P, Mazzolini G, Qian C, Prieto J, Melero I. Upregulation of natural killer cells functions underlies the efficacy of intratumorally injected dendritic cells engineered to produce interleukin-12. Exp Hematol 2002; 30:195-204. [PMID: 11882356 DOI: 10.1016/s0301-472x(01)00792-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Injection of dendritic cells (DC) engineered with recombinant adenoviral vectors to produce interleukin-12 (IL-12) inside experimental murine tumors frequently achieves complete regressions. In such a system the function of CD8(+) T cells has been shown to be an absolute requirement, in contrast to observations made upon depletion of CD4(+) T cells, which minimally affected the outcome. The aim of this work was to study the possible involvement of natural killer (NK) cells in this setting. MATERIALS, METHODS, AND RESULTS Depletions with anti-AsialoGM1 antiserum showed only a small decrease in the proportion of complete regressions obtained that correlated with induction of NK activities in lymphatic tissues into which DC migrate, whereas combined depletions of CD4(+) and NK cells completely eliminated the antitumor effects. Likewise in vivo neutralization of interferon-gamma (IFN-gamma) also eliminated those therapeutic effects. Trying to define the cellular role played by NK cells in vivo, it was observed that injection of cultured DC inside the spleen of T- and B-cell-deficient (Rag1(-/-)) mice induced upregulation of NK activity only if DC had been adenovirally engineered to produce IL-12. In addition, identically transfected fibroblasts also activated NK cells, indicating that IL-12 transfection was the unique requirement. Equivalent human DC only activated in vitro the cytolytic and cytokine-secreting functions of autologous NK cells if transfected to express human IL-12. CONCLUSIONS Overall, these results point out an important role played by NK cell activation in the potent immunotherapeutic effects elicited by intratumoral injection of IL-12--secreting DC and that NK activation under these conditions is mainly, if not only, dependent on IL-12.
Collapse
|
29
|
Halloran CM, Ghaneh P, Neoptolemos JP, Costello E. Gene therapy for pancreatic cancer--current and prospective strategies. Surg Oncol 2000; 9:181-91. [PMID: 11476989 DOI: 10.1016/s0960-7404(01)00011-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pancreatic ductal adenocarcinoma is one of the most common causes of cancer death in the developed world. Long-term survival is currently only achieved through surgical resection. Most patients have locally advanced or metastatic disease at the time of diagnosis and are therefore not amenable to resection, whilst chemotherapy and radiotherapy are by and large ineffective. Gene therapy offers an alternative to current adjuvant strategies. With approximately two-thirds of all gene therapy trials worldwide directed at cancer, the gene therapy approaches that are currently being explored for pancreatic cancer are specifically examined. Gene delivery systems, genetic targets, and combined gene delivery with chemotherapy are discussed in the context of pancreatic cancer treatment.
Collapse
Affiliation(s)
- C M Halloran
- Department of Surgery, Royal Liverpool University Hospital, 5th Floor UCD Building, Daulby Street, Liverpool, L69 3GA, UK.
| | | | | | | |
Collapse
|