1
|
Ali A, Gao M, Iskantar A, Wang H, Karlsson-Parra A, Yu D, Jin C. Proinflammatory allogeneic dendritic cells enhance the therapeutic efficacy of systemic anti-4-1BB treatment. Front Immunol 2023; 14:1146413. [PMID: 37654492 PMCID: PMC10466132 DOI: 10.3389/fimmu.2023.1146413] [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: 01/17/2023] [Accepted: 07/26/2023] [Indexed: 09/02/2023] Open
Abstract
As an immune adjuvant, proinflammatory allogeneic dendritic cells (AlloDCs) have demonstrated promising immune-priming effects in several preclinical and clinical studies. The effector cells, including NK cells and T cells are widely acknowledged as pivotal factors in the effectiveness of cancer immunotherapy due to their ability to selectively identify and eradicate malignant cells. 4-1BB, as a costimulatory receptor, plays a significant role in the stimulation of effector cell activation. This study evaluated the anti-tumor effects when combining intratumoral administration of the immune-adjuvant AlloDCs with systemic α4-1BB treatment directly acting on effector cells. In both the CT-26 murine colon carcinoma model and B16 murine melanoma model, AlloDCs demonstrated a significant enhancement in the therapeutic efficacy of α4-1BB antibody. This enhancement was observed through the delayed growth of tumors and prolonged survival. Analysis of the tumor microenvironment (TME) in the combined-treatment group revealed an immune-inflamed TME characterized by increased infiltration of activated endogenous DCs and IFNγ+ CD8+ T cells, showing reduced signs of exhaustion. Furthermore, there was an augmented presence of tissue-resident memory (TRM) CD8+ T cells (CD103+CD49a+CD69+). The combination treatment also led to increased infiltration of CD39+CD103+ tumor-specific CD8+ T cells and neoantigen-specific T cells into the tumor. Additionally, the combined treatment resulted in a less immunosuppressive TME, indicated by decreased infiltration of myeloid-derived suppressor cells and Tregs. These findings suggest that the combination of intratumoral AlloDCs administration with systemic agonistic α4-1BB treatment can generate a synergistic anti-tumor response, thereby warranting further investigation through clinical studies.
Collapse
Affiliation(s)
- Arwa Ali
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Menghan Gao
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Alexandros Iskantar
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Hai Wang
- Chinese Academy of Science (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | | | - Di Yu
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Chuan Jin
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
2
|
Hai Y, Hong Y, Yang Y. miR-1258 Enhances the Anti-Tumor Effect of Liver Cancer Natural Killer (NK) Cells by Stimulating Toll-Liker Receptor (TLR)7/8 to Promote Natural Killer (NK)-Dendritic Cell (DC) Interaction. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
TLR7/8 agonists are immunomodulators for treating skin cancer or virus infections. miR-1258 can activate TLR7/8. This study aims to explore the role of TLR7/8 agonists and miR-1258 in activating liver cancer NK cells. NK cells and DC cells were treated with TLR7/8 agonists R837, ssRNA40
and miR-1258 followed by analysis of hepatocellular carcinoma (HCC) cell behaviors in vivo and in vitro. TLR7/8 agonist miR-1258 activated NKs and promoted DCs maturation. In addition, DCs also assisted NKs to function and enhance the anti-HCC immune responses. The interaction
of DCs with NK cells stimulated by TLR7/8 agonist miR-1258 can significantly inhibit tumor development and metastasis in mice HCC model. TLR7 or TLR8 agonists, especially miR-1258, promoted DCs-NKs interaction by promoting the secretion of related cytokines and cell/cell contact, which increased
anti-tumor activity of NKs and promoted DC-NK cells to inhibit the growth of HCC cells. In conclusion, miR-1258 simultaneously stimulates the expression of TLR7/8, and promotes NK-DC cells to inhibit the growth of HCC cells, providing a theoretical basis for the treatment of liver cancer.
Collapse
Affiliation(s)
- Yuedong Hai
- Department of Emergency Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Yu Hong
- Department of Imaging Center, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Yuzhu Yang
- Department of Emergency Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| |
Collapse
|
3
|
Veneziani I, Alicata C, Pelosi A, Landolina N, Ricci B, D'Oria V, Fagotti A, Scambia G, Moretta L, Maggi E. Toll-like receptor 8 agonists improve NK-cell function primarily targeting CD56brightCD16− subset. J Immunother Cancer 2022; 10:jitc-2021-003385. [PMID: 35091452 PMCID: PMC8804697 DOI: 10.1136/jitc-2021-003385] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 11/05/2022] Open
Abstract
Background Toll-like receptors (TLRs) are pattern-recognition sensors mainly expressed in innate immune cells that directly recognize conserved pathogen structures (pathogen-associated molecular patterns-PAMPs). Natural killer (NK) cells have been described to express different endosomal TLRs triggered by RNA and DNA sequences derived from both viruses and bacteria. This study was addressed to establish which endosomal TLR could directly mediate NK activation and function after proper stimuli. It was also important to establish the most suitable TLR agonist to be used as adjuvant in tumor vaccines or in combined cancer immunotherapies. Methods We assessed endosomal TLR expression in total NK cells by using RT-qPCR and western blotting technique. In some experiments, we purified CD56brightCD16− and CD56dimCD16+ cells subsets by using NK Cell Isolation Kit Activation marker, cytokine production, CD107a expression and cytotoxicity assay were evaluated by flow cytometry. Cytokine release was quantified by ELISA. NK cells obtained from ovarian ascites underwent the same analyses. Results Although the four endosomal TLRs (TLR3, TLR7/8, and TLR9) were uniformly expressed on CD56brightCD16− and CD56dimCD16+ cell subsets, the TLR7/8 (R848), TLR3 (polyinosinic-polycytidylic acid, Poly I:C) and TLR9 (ODN2395) ligands promoted NK-cell function only in the presence of suboptimal doses of cytokines, including interleukin (IL)-2, IL-12, IL-15, and IL-18, produced in vivo by other environmental cells. We showed that R848 rather than TLR3 and TLR9 agonists primarily activated CD56brightCD16− NK cells by increasing their proliferation, cytokine production and cytotoxic activity. Moreover, we demonstrated that R848, which usually triggers TLR7 and TLR8 on dendritic cells, macrophages and neutrophils cells, activated CD56brightCD16− NK-cell subset only via TLR8. Indeed, specific TLR8 but not TLR7 agonists increased cytokine production and cytotoxic activity of CD56brightCD16− NK cells. Importantly, these activities were also observed in peritoneal NK cells from patients with metastatic ovarian carcinoma, prevalently belonging to the CD56brightCD16− subset. Conclusion These data highlight the potential value of TLR8 in NK cells as a new target for immunotherapy in patients with cancer.
Collapse
Affiliation(s)
- Irene Veneziani
- Department of Immunology, Bambino Gesu Pediatric Hospital, Roma, Italy
| | - Claudia Alicata
- Department of Immunology, Bambino Gesu Pediatric Hospital, Roma, Italy
| | - Andrea Pelosi
- Department of Immunology, Bambino Gesu Pediatric Hospital, Roma, Italy
| | - Nadine Landolina
- Department of Immunology, Bambino Gesu Pediatric Hospital, Roma, Italy
| | - Biancamaria Ricci
- Department of Immunology, Bambino Gesu Pediatric Hospital, Roma, Italy
| | - Valentina D'Oria
- Confocal Microscopy Core Facility, Bambino Gesu Pediatric Hospital, Roma, Italy
| | - Anna Fagotti
- Department of Woman's Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Campus di Roma, Roma, Lazio, Italy
| | - Giovanni Scambia
- Department of Woman's Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Campus di Roma, Roma, Lazio, Italy
| | - Lorenzo Moretta
- Department of Immunology, Bambino Gesu Pediatric Hospital, Roma, Italy
| | - Enrico Maggi
- Department of Immunology, Bambino Gesu Pediatric Hospital, Roma, Italy
| |
Collapse
|
4
|
Sommariva M, De Cecco L, Tagliabue E, Balsari A. Modulation of DNA repair genes induced by TLR9 agonists: A strategy to eliminate "altered" cells? Oncoimmunology 2021; 1:258-259. [PMID: 22720263 PMCID: PMC3377000 DOI: 10.4161/onci.1.2.18343] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We provided evidence that the TLR9 engagement of innate immune cells present in the tumor microenvironment by CpG-oligodeoxynucleotide (CpG-ODN) induces down-modulation of DNA repair gene expression in tumor cells, sensitizing cancer cells to DNA-damaging chemotherapy. These findings expand the benefits of CpG-ODN therapy beyond induction of a strong immune response.
Collapse
Affiliation(s)
- Michele Sommariva
- Department of Human Morphology and Biomedical Sciences "Città Studi"; Università degli Studi di Milano; Milan, Italy ; Molecular Targeting Unit Fondazione IRCCS; Istituto Nazionale Tumori; Milan, Italy
| | | | | | | |
Collapse
|
5
|
Kumar SU, Telichko AV, Wang H, Hyun D, Johnson EG, Kent MS, Rebhun RB, Dahl JJ, Culp WTN, Paulmurugan R. Acoustically Driven Microbubbles Enable Targeted Delivery of microRNA-Loaded Nanoparticles to Spontaneous Hepatocellular Neoplasia in Canines. ADVANCED THERAPEUTICS 2020; 3:2000120. [PMID: 33415184 PMCID: PMC7784952 DOI: 10.1002/adtp.202000120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Indexed: 01/16/2023]
Abstract
Spatially localized microbubble cavitation by ultrasound offers an effective means of altering permeability of natural barriers (i.e. blood vessel and cell membrane) in favor of nanomaterials accumulation in the target site. In this study, a clinically relevant, minimally invasive ultrasound guided therapeutic approach is investigated for targeted delivery of anticancer microRNA loaded PLGA-b-PEG nanoparticles to spontaneous hepatocellular neoplasia in a canine model. Quantitative assessment of the delivered microRNAs revealed prominent and consistent increase in miRNAs levels (1.5-to 2.3-fold increase (p<0.001)) in ultrasound treated tumor regions compared to untreated control regions. Immunohistology of ultrasound treated tumor tissue presented a clear evidence for higher amount of nanoparticles extravasation from the blood vessels. A distinct pattern of cytokine expression supporting CD8+ T cells mediated "cold-to-hot" tumor transition was evident in all patients. On the outset, proposed platform can enhance delivery of miRNA-loaded nanoparticles to deep seated tumors in large animals to enhance chemotherapy.
Collapse
Affiliation(s)
- Sukumar Uday Kumar
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| | - Arsenii V Telichko
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| | - Huaijun Wang
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| | - Dongwoon Hyun
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| | - Eric G Johnson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, California
| | - Michael S Kent
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, California
| | - Robert B Rebhun
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, California
| | - Jeremy J Dahl
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| | - William T N Culp
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, California
| | - Ramasamy Paulmurugan
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, California; Department of Radiology, Stanford University, Stanford, California
| |
Collapse
|
6
|
Makkouk A, Chester C, Kohrt HE. Rationale for anti-CD137 cancer immunotherapy. Eur J Cancer 2016; 54:112-119. [PMID: 26751393 DOI: 10.1016/j.ejca.2015.09.026] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 09/21/2015] [Accepted: 09/21/2015] [Indexed: 11/29/2022]
Abstract
The consideration of the complex interplay between the tumour microenvironment (TME) and the immune response is the key for designing effective immunotherapies. Therapeutic strategies that harness co-stimulatory receptors have recently gained momentum in the clinic. One such strategy with promising clinical applications is the targeting of CD137, a member of the tumour necrosis factor receptor superfamily. Its expression on both innate and adaptive immune cells, coupled with its unique ability to potentiate antitumour responses through modulating the TME and to ameliorate autoimmune responses, has established it as an appealing target. In this review, we will discuss the various CD137-targeted immunotherapeutics that have reached clinical development, with a focus on recent advances and novel modalities such as CD137 chimeric antigen receptors and CD137 bispecific antibodies. We will also highlight the effect of CD137 targeting on the TME and discuss the importance of probing TME changes for predicting and testing the efficacy of CD137-mediated immunotherapy.
Collapse
Affiliation(s)
- Amani Makkouk
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA 94305 USA
| | - Cariad Chester
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA 94305 USA; Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Holbrook E Kohrt
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA 94305 USA.
| |
Collapse
|
7
|
Zhou Z, Yu X, Zhang J, Tian Z, Zhang C. TLR7/8 agonists promote NK-DC cross-talk to enhance NK cell anti-tumor effects in hepatocellular carcinoma. Cancer Lett 2015; 369:298-306. [PMID: 26433159 DOI: 10.1016/j.canlet.2015.09.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 09/17/2015] [Accepted: 09/25/2015] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is a common cancer worldwide and the third leading cause of cancer death. Immunotherapy is considered a promising treatment with the aim to boost or arouse HCC-specific immune responses. TLR7 and TLR8 agonists are effective immunomodulators and have been applied topically for the treatment of certain skin tumors and viral infections. Here, we explored the role of TLR7 and TLR8 agonists on the activation of dendritic cells (DCs) and natural killer (NK) cells. We demonstrated that these agonists could directly activate NK cells, promoting the maturation of immature DCs. Meanwhile, DCs also assisted in the function of NK cells, resulting in enhanced anti-tumor immune responses to HCC. Importantly, the combination therapy with NK cells stimulated with DCs and TLR7/8 agonist Gardiquimod (GDQ) significantly suppresses the growth of human HepG2 liver carcinoma xenografts. This study provides a new immunotherapeutic approach for human HCC based on DC-NK cross-talk and also suggests that TLR7 and/or TLR8 agonists, particularly GDQ, may serve as potent innate and adaptive immune response immunomodulators in tumor therapy.
Collapse
Affiliation(s)
- Zhixia Zhou
- Institute of Immunopharmacology & Immunotherapy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Xin Yu
- Institute of Immunopharmacology & Immunotherapy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Jian Zhang
- Institute of Immunopharmacology & Immunotherapy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Zhigang Tian
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei 230027, Anhui, China.
| | - Cai Zhang
- Institute of Immunopharmacology & Immunotherapy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China.
| |
Collapse
|
8
|
Devaud C, John LB, Westwood JA, Yong CS, Beavis PA, Schwendener RA, Darcy PK, Kershaw MH. Cross-talk between tumors can affect responses to therapy. Oncoimmunology 2015; 4:e975572. [PMID: 26140251 DOI: 10.4161/2162402x.2014.975572] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 10/07/2014] [Indexed: 12/26/2022] Open
Abstract
Advanced stages of cancer often involve multiple tumors in different locations in the body. These tumors are associated with a microenvironment that can influence tumor responses to immunotherapy. Whether tumors and their disparate microenvironment can interact together at distance in a multiple tumor setting, through a form of cross-talk, and affect their responses to immunotherapy has never been described. Our study investigated the cross-talk between two tumors with disparate microenvironments in a mouse model. We demonstrated that immunosuppressive visceral tumors could influence distant subcutaneous (SC) tumors to render them resistant to immunotherapy. We observed distinct modifications in the SC tumor microenvironment following cross-talk with kidney tumors that exhibit a type-2 macrophage-related immunosuppressive microenvironment. Indeed, when a concomitant kidney tumor was present in the mouse, the SC tumors were highly infiltrated with M2 macrophages and had a reduced T cell and NK cell effector immune profile. Finally, blocking the M2-associated chemokine CCL2 or depleting macrophages, significantly improved the effect of immunotherapy on growth of SC tumors in the presence of concomitant kidney tumors. This work emphasizes the potential negative influence that a tumor, with a strong immunosuppressive microenvironment, can exert on distant tumors that would normally be treatment-responsive. This report may lead to a new vision of the prioritization in the treatment of advanced metastatic cancer.
Collapse
Affiliation(s)
- Christel Devaud
- Cancer Immunology Research Program; Sir Peter MacCallum Department of Oncology; University of Melbourne ; Parkville, Australia
| | - Liza B John
- Cancer Immunology Research Program; Sir Peter MacCallum Department of Oncology; University of Melbourne ; Parkville, Australia
| | - Jennifer A Westwood
- Cancer Immunology Research Program; Sir Peter MacCallum Department of Oncology; University of Melbourne ; Parkville, Australia
| | - Carmen Sm Yong
- Cancer Immunology Research Program; Sir Peter MacCallum Department of Oncology; University of Melbourne ; Parkville, Australia
| | - Paul A Beavis
- Cancer Immunology Research Program; Sir Peter MacCallum Department of Oncology; University of Melbourne ; Parkville, Australia
| | - Reto A Schwendener
- Institute of Molecular Cancer Research; University of Zurich ; Zurich, Switzerland
| | - Phillip K Darcy
- Cancer Immunology Research Program; Sir Peter MacCallum Department of Oncology; University of Melbourne ; Parkville, Australia ; ; Department of Immunology; Monash University ; Prahran, Victoria, Australia
| | - Michael H Kershaw
- Cancer Immunology Research Program; Sir Peter MacCallum Department of Oncology; University of Melbourne ; Parkville, Australia ; ; Department of Immunology; Monash University ; Prahran, Victoria, Australia
| |
Collapse
|
9
|
Westwood JA, Darcy PK, Kershaw MH. The potential impact of mouse model selection in preclinical evaluation of cancer immunotherapy. Oncoimmunology 2014; 3:e946361. [PMID: 25610731 DOI: 10.4161/21624011.2014.946361] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 06/06/2014] [Indexed: 11/19/2022] Open
Abstract
This commentary provides the authors' perspective on the article "Routes of delivery for CpG and anti-CD137 for the treatment of orthotopic kidney tumors in mice", published in PLoS ONE. It also discusses the caveats of using subcutaneous tumors to model the treatment of human cancers versus orthotopic mouse models that more closely mimic human disease.
Collapse
Affiliation(s)
- Jennifer A Westwood
- Division of Cancer Research; Peter MacCallum Cancer Center ; East Melbourne, Australia
| | - Phillip K Darcy
- Division of Cancer Research; Peter MacCallum Cancer Center ; East Melbourne, Australia ; Sir Peter MacCallum Department of Oncology; University of Melbourne ; Parkville, Australia ; Department of Immunology; Monash University ; Prahran, Australia
| | - Michael H Kershaw
- Division of Cancer Research; Peter MacCallum Cancer Center ; East Melbourne, Australia ; Sir Peter MacCallum Department of Oncology; University of Melbourne ; Parkville, Australia ; Department of Immunology; Monash University ; Prahran, Australia
| |
Collapse
|
10
|
Westwood JA, Potdevin Hunnam TCU, Pegram HJ, Hicks RJ, Darcy PK, Kershaw MH. Routes of delivery for CpG and anti-CD137 for the treatment of orthotopic kidney tumors in mice. PLoS One 2014; 9:e95847. [PMID: 24788789 PMCID: PMC4008493 DOI: 10.1371/journal.pone.0095847] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 04/01/2014] [Indexed: 12/24/2022] Open
Abstract
We have found previously that the tumor cell lines, Renca (a renal cancer) and MC38 (a colon tumor) which had been injected subcutaneously in mice, could be successfully treated with a combination therapy of an oligodeoxynucleotide (CpG1826) (injected intratumorally) and anti-CD137 antibody (injected intraperitoneally). Thus the combination treatment was expected to initiate a “danger” signal via TLR9 on immune cells, and the anti-CD137 was expected to further activate T cells. In the present study, we found that several other tumor types injected subcutaneously could also be successfully treated with this combination therapy. In addition, we wished to determine if the treatment could work as effectively in an orthotopic metastatic model, which is more physiologically relevant to cancer in humans. Renca was selected as we were familiar with injecting this orthotopically into the outer cortex of the kidney in mice, and it spontaneously metastasizes to lung and abdominal sites. We tested various routes of delivery of CpG combined with intraperitoneal delivery of anti-CD137. Orthotopic tumors were injected with CpG intratumorally, using ultrasound-guided delivery on multiple occasions, combined with anti-CD137 intraperitoneally. A reduction in primary tumor size was observed following intratumoral injection of CpG compared to other treatments. We found that there was a statistically significant increase in survival of mice with orthotopic Renca tumor following intratumoral injection of CpG. However, we determined that the most effective route of delivery of CpG was intravenous, which led to further significantly enhanced survival of mice when combined with anti-CD137 intraperitoneally, likely due to inhibition of metastatic disease. Our data supports future development of this combination therapy for cancer.
Collapse
Affiliation(s)
- Jennifer A. Westwood
- Division of Cancer Research, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Australia
| | | | - Hollie J. Pegram
- Division of Cancer Research, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Australia
| | - Rodney J. Hicks
- Centre for Cancer Imaging, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Australia
| | - Phillip K. Darcy
- Division of Cancer Research, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
- Department of Immunology, Monash University, Prahran, Australia
| | - Michael H. Kershaw
- Division of Cancer Research, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
- Department of Immunology, Monash University, Prahran, Australia
- * E-mail:
| |
Collapse
|
11
|
Devaud C, John LB, Westwood JA, Darcy PK, Kershaw MH. Immune modulation of the tumor microenvironment for enhancing cancer immunotherapy. Oncoimmunology 2013; 2:e25961. [PMID: 24083084 PMCID: PMC3782527 DOI: 10.4161/onci.25961] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 07/29/2013] [Indexed: 12/21/2022] Open
Abstract
There is much promise in the use of immunotherapy for the treatment of cancer. Approaches such as those using antibodies or adoptive cell transfer can mediate complete tumor regression in a proportion of patients. However, the tumor microenvironment can inhibit immune responses leading to ineffective or suboptimal responses of tumors to immunotherapy in the majority of cases. As our knowledge of the tumor microenvironment increases, many strategies are emerging for changing the immunosuppressive nature of the tumor toward a microenvironment able to support immunity. These strategies aim to enhance the ability of immunotherapies to initiate effective immune responses able to destroy tumors. In this article, we review approaches that use immunomodulators specifically to modify the tumor microenvironment, and their use in combination with other immune-based strategies for cancer therapy.
Collapse
Affiliation(s)
- Christel Devaud
- Cancer Immunology Research Program; Sir Peter MacCallum Department of Oncology; University of Melbourne; Parkville, VIC Australia
| | | | | | | | | |
Collapse
|
12
|
Moossavi S, Rezaei N. Toll-like receptor signalling and their therapeutic targeting in colorectal cancer. Int Immunopharmacol 2013; 16:199-209. [PMID: 23602501 DOI: 10.1016/j.intimp.2013.03.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Revised: 02/13/2013] [Accepted: 03/13/2013] [Indexed: 12/16/2022]
Abstract
Intestinal homeostasis is dependent on the proper host/microbiota interaction via pattern recognition receptors. Toll-like receptors are a specialised group of membrane receptors which detect pathogen-associated conserved structures. They are present in the intestinal tract and are required for intestinal homeostasis. Dysregulation in the Toll-like receptor signalling can conceivably result in a dysregulated immune response which could contribute to major intestinal pathologies including colorectal cancer. Evidence for the role of microbiota and toll-like receptors in colorectal cancer is emerging. In this report the evidence for the contribution of toll-like receptors to the pathogenesis of colorectal cancer; potential mechanisms affecting toll-like receptor signalling; and their therapeutic targeting in colorectal cancer are reviewed.
Collapse
Affiliation(s)
- Shirin Moossavi
- Digestive Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
13
|
Zonneveld-Huijssoon E, van Wijk F, Roord S, Delemarre E, Meerding J, de Jager W, Klein M, Raz E, Albani S, Kuis W, Boes M, Prakken BJ. TLR9 agonist CpG enhances protective nasal HSP60 peptide vaccine efficacy in experimental autoimmune arthritis. Ann Rheum Dis 2012; 71:1706-15. [PMID: 22562976 DOI: 10.1136/annrheumdis-2011-201131] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Peptide-based immune tolerance induction is considered an attractive treatment option for autoimmune diseases. The authors have developed a novel method that can enhance the induction of protective peptide-specific T-cell responses, using a rat arthritis model. The authors focused on the Toll-like receptor 9 ligand CpG, which was shown to stimulate regulatory T-cell proliferation when added to plasmacytoid dendritic cells (pDC) using in-vitro cultures. METHODS The peptide used is a heat shock protein 60 epitope (p1) that elicits tolerogenic peptide-specific immune responses in human arthritis patients and was recently shown to have protective capacity as a bystander antigen in the rat adjuvant arthritis model. Rats were treated with three nasal doses of p1, CpG or a combination of p1 and CpG. Antigen-presenting cells were studied in nose-draining lymph nodes (mandibular lymph nodes; MLN) after nasal treatment, and T-cell responses were analysed in joint-draining lymph nodes after arthritis induction. RESULTS Nasal co-administration of p1/CpG significantly augmented the arthritis-protective effect of p1, while CpG treatment alone did not. Co-treatment of p1/CpG increased both the number and activation status of pDC in draining MLN, which was accompanied by amplified p1-specific T-cell proliferation and interleukin (IL)-10 production. During early arthritis, p1-specific IL-10 production was identified at the site of inflammation. P1 and p1/CpG-treated rats showed a greater amount of CD4+FoxP3+ regulatory T cells in the joint-draining lymph nodes, which correlated with lower arthritis scores. CONCLUSIONS These clinical and immunological data suggest the use of CpG as a potent adjuvant for mucosal peptide-specific immune therapy in arthritis.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Administration, Intranasal
- Animals
- Arthritis, Experimental/immunology
- Arthritis, Rheumatoid/immunology
- Chaperonin 60/administration & dosage
- Chaperonin 60/immunology
- Dendritic Cells/immunology
- Disease Models, Animal
- Epitopes, T-Lymphocyte/immunology
- Lymphocyte Activation/immunology
- Male
- Oligodeoxyribonucleotides/administration & dosage
- Oligodeoxyribonucleotides/immunology
- Rats
- Rats, Inbred Lew
- T-Lymphocytes, Regulatory/immunology
- Toll-Like Receptor 9/agonists
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Evelien Zonneveld-Huijssoon
- Department of Pediatric Immunology, University Medical Centre Utrecht, Centre for Molecular and Cellular Intervention, Lundlaan 6, 3584 EA Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Maito FLDM, Souza APDD, Pereira L, Smithey M, Hinrichs D, Bouwer A, Bonorino C. Intratumoral TLR-4 Agonist Injection Is Critical for Modulation of Tumor Microenvironment and Tumor Rejection. ACTA ACUST UNITED AC 2012. [DOI: 10.5402/2012/926817] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The tumor microenvironment shelters a complex network of mechanisms that enables local Immunosuppression to support tumor growth. In this study we found that, B16F10 melanoma growth is inversely correlated with peritumoral infiltrate cell number and with cell numbers in draining lymph nodes. Tumor growth ensued even when a foreign antigen was expressed by B16F10 cells in the presence of naïve specific CD8+ T cells. Treatment with TLR agonists has shown to sometimes result in tumor regression, however, not always with long-lasting effects. We compared the relevance of different injection regimens of lipopolysaccharide (LPS). Tumor growth was arrested only by intratumoral LPS injection after the tumor was already established. This result was accompanied by a dramatic change in DC activation inside the tumor. Intratumoral LPS also enhanced antigen presentation and tumor-specific CD4+ T cell production of IFN-γ. Injection of LPS before tumor challenge or codelivery of tumor cells and LPS did not have any effect on tumor progression. Our results suggest that an efficient antitumor immune response leading to tumor regression can be achieved with proper TLR4 activation inside the tumor tissue, impacting the tumor microenvironment. These findings are relevant for the design of treatment for patients with malignant melanomas.
Collapse
Affiliation(s)
- Fabio Luiz Dal Moro Maito
- Faculdade de Biociências e Instituto de Pesquisas Biomédicas, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Avenida Ipiranga 6690, 2° Andar, 90680-001 Porto Alegre, RS, Brazil
| | - Ana Paula Duarte de Souza
- Faculdade de Biociências e Instituto de Pesquisas Biomédicas, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Avenida Ipiranga 6690, 2° Andar, 90680-001 Porto Alegre, RS, Brazil
| | - Luciana Pereira
- Faculdade de Biociências e Instituto de Pesquisas Biomédicas, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Avenida Ipiranga 6690, 2° Andar, 90680-001 Porto Alegre, RS, Brazil
| | - Megan Smithey
- Departments of Molecular Microbiology and Immunology, Veterans Affairs Medical Center, Oregon Health and Science University, Portland, OR 97239, USA
| | - David Hinrichs
- Departments of Molecular Microbiology and Immunology, Veterans Affairs Medical Center, Oregon Health and Science University, Portland, OR 97239, USA
| | - Archie Bouwer
- Departments of Molecular Microbiology and Immunology, Veterans Affairs Medical Center, Oregon Health and Science University, Portland, OR 97239, USA
| | - Cristina Bonorino
- Faculdade de Biociências e Instituto de Pesquisas Biomédicas, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Avenida Ipiranga 6690, 2° Andar, 90680-001 Porto Alegre, RS, Brazil
| |
Collapse
|
15
|
Oncolytic Virus and Anti–4-1BB Combination Therapy Elicits Strong Antitumor Immunity against Established Cancer. Cancer Res 2012; 72:1651-60. [DOI: 10.1158/0008-5472.can-11-2788] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
16
|
Sommariva M, De Cecco L, De Cesare M, Sfondrini L, Ménard S, Melani C, Delia D, Zaffaroni N, Pratesi G, Uva V, Tagliabue E, Balsari A. TLR9 agonists oppositely modulate DNA repair genes in tumor versus immune cells and enhance chemotherapy effects. Cancer Res 2011; 71:6382-90. [PMID: 21878529 DOI: 10.1158/0008-5472.can-11-1285] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Synthetic oligodeoxynucleotides expressing CpG motifs (CpG-ODN) are a Toll-like receptor 9 (TLR9) agonist that can enhance the antitumor activity of DNA-damaging chemotherapy and radiation therapy in preclinical mouse models. We hypothesized that the success of these combinations is related to the ability of CpG-ODN to modulate genes involved in DNA repair. We conducted an in silico analysis of genes implicated in DNA repair in data sets obtained from murine colon carcinoma cells in mice injected intratumorally with CpG-ODN and from splenocytes in mice treated intraperitoneally with CpG-ODN. CpG-ODN treatment caused downregulation of DNA repair genes in tumors. Microarray analyses of human IGROV-1 ovarian carcinoma xenografts in mice treated intraperitoneally with CpG-ODN confirmed in silico findings. When combined with the DNA-damaging drug cisplatin, CpG-ODN significantly increased the life span of mice compared with individual treatments. In contrast, CpG-ODN led to an upregulation of genes involved in DNA repair in immune cells. Cisplatin-treated patients with ovarian carcinoma as well as anthracycline-treated patients with breast cancer who are classified as "CpG-like" for the level of expression of CpG-ODN modulated DNA repair genes have a better outcome than patients classified as "CpG-untreated-like," indicating the relevance of these genes in the tumor cell response to DNA-damaging drugs. Taken together, the findings provide evidence that the tumor microenvironment can sensitize cancer cells to DNA-damaging chemotherapy, thereby expanding the benefits of CpG-ODN therapy beyond induction of a strong immune response.
Collapse
Affiliation(s)
- Michele Sommariva
- Department of Human Morphology and Biomedical Sciences Città Studi, Università degli Studi di Milano, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Anti-ErbB-2 mAb therapy requires type I and II interferons and synergizes with anti-PD-1 or anti-CD137 mAb therapy. Proc Natl Acad Sci U S A 2011; 108:7142-7. [PMID: 21482773 DOI: 10.1073/pnas.1016569108] [Citation(s) in RCA: 389] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Trastuzumab, a monoclonal antibody targeting human epidermal growth factor receptor-2 (HER2/ErbB-2), has become the mainstay of treatment for HER2-positive breast cancer. Nevertheless, its exact mechanism of action has not been fully elucidated. Although several studies suggest that Fc receptor-expressing immune cells are involved in trastuzumab therapy, the relative contribution of lymphocyte-mediated cellular cytotoxicity and antitumor cytokines remains unknown. We report here that anti-ErbB-2 mAb therapy is dependent on the release of type I and type II IFNs but is independent of perforin or FasL. Our study thus challenges the notion that classical antibody-dependent, lymphocyte-mediated cellular cytotoxicity is important for trastuzumab. We demonstrate that anti-ErbB-2 mAb therapy of experimental tumors derived from MMTV-ErbB-2 transgenic mice triggers MyD88-dependent signaling and primes IFN-γ-producing CD8+ T cells. Adoptive cell transfer of purified T cell subsets confirmed the essential role of IFN-γ-producing CD8+ T cells. Notably, anti-ErbB-2 mAb therapy was independent of IL-1R or IL-17Ra signaling. Finally, we investigated whether immunostimulatory approaches with antibodies against programmed death-1 (PD-1) or 41BB (CD137) could be used to capitalize on the immune-mediated effects of trastuzumab. We demonstrate that anti-PD-1 or anti-CD137 mAb can significantly improve the therapeutic activity of anti-ErbB-2 mAb in immunocompetent mice.
Collapse
|
18
|
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: 37] [Impact Index Per Article: 2.5] [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
|
19
|
Haynes NM, Hawkins ED, Li M, McLaughlin NM, Hämmerling GJ, Schwendener R, Winoto A, Wensky A, Yagita H, Takeda K, Kershaw MH, Darcy PK, Smyth MJ. CD11c+Dendritic Cells and B Cells Contribute to the Tumoricidal Activity of Anti-DR5 Antibody Therapy in Established Tumors. THE JOURNAL OF IMMUNOLOGY 2010; 185:532-41. [DOI: 10.4049/jimmunol.0903624] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
20
|
Westwood JA, Darcy PK, Guru PM, Sharkey J, Pegram HJ, Amos SM, Smyth MJ, Kershaw MH. Three agonist antibodies in combination with high-dose IL-2 eradicate orthotopic kidney cancer in mice. J Transl Med 2010; 8:42. [PMID: 20426873 PMCID: PMC2873376 DOI: 10.1186/1479-5876-8-42] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 04/28/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Combination immunotherapies can be effective against subcutaneous tumors in mice but the effect against orthotopic malignant disease is less well characterized. In particular, a combination of three agonist antibodies, termed Tri-mAb, consisting of anti-DR5, anti-CD40 and anti-CD137 has previously been demonstrated to eradicate a large proportion of subcutaneous renal cell carcinoma (Renca) tumors (75% long-term survival), but the effect against orthotopic disease is not known. PURPOSE To determine the relative response of orthotopic tumors, we inoculated Renca into the kidney followed by treatment with Tri-mAb. RESULTS We found that orthotopic tumors responded much less to treatment (approximately 13% survival), but a significant improvement in survival was achieved through the addition of IL-2 to the treatment regimen (55% survival). All three agonist antibodies and high dose IL-2, 100,000 IU for up to six doses, were required. CD8+ T cells were also required for optimal anti-tumor responses. Coadministration of IL-2 led to enhanced T cell activity as demonstrated by an increased frequency of IFN-gamma-producing T cells in tumor-draining lymph nodes, which may have contributed to the observed improvement of therapy against kidney tumors. IMPLICATIONS Responses of subcutaneous tumors to immunotherapy do not necessarily reflect how orthotopic tumors respond. The use of combination immunotherapy stimulating multiple facets of immunity and including cytokine support for T cells can induce effective anti-tumor responses against orthotopic and metastatic tumors.
Collapse
Affiliation(s)
- Jennifer A Westwood
- Cancer Immunology Research Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | | | | | | | | | | | | |
Collapse
|