1
|
Highfill SL, Cui Y, Giles AJ, Smith JP, Zhang H, Morse E, Kaplan RN, Mackall CL. Disruption of CXCR2-mediated MDSC tumor trafficking enhances anti-PD1 efficacy. Sci Transl Med 2014; 6:237ra67. [PMID: 24848257 DOI: 10.1126/scitranslmed.3007974] [Citation(s) in RCA: 595] [Impact Index Per Article: 54.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Suppression of the host's immune system plays a major role in cancer progression. Tumor signaling of programmed death 1 (PD1) on T cells and expansion of myeloid-derived suppressor cells (MDSCs) are major mechanisms of tumor immune escape. We sought to target these pathways in rhabdomyosarcoma (RMS), the most common soft tissue sarcoma of childhood. Murine RMS showed high surface expression of PD-L1, and anti-PD1 prevented tumor growth if initiated early after tumor inoculation; however, delayed anti-PD1 had limited benefit. RMS induced robust expansion of CXCR2(+)CD11b(+)Ly6G(hi) MDSCs, and CXCR2 deficiency prevented CD11b(+)Ly6G(hi) MDSC trafficking to the tumor. When tumor trafficking of MDSCs was inhibited by CXCR2 deficiency, or after anti-CXCR2 monoclonal antibody therapy, delayed anti-PD1 treatment induced significant antitumor effects. Thus, CXCR2(+)CD11b(+)Ly6G(hi) MDSCs mediate local immunosuppression, which limits the efficacy of checkpoint blockade in murine RMS. Human pediatric sarcomas also produce CXCR2 ligands, including CXCL8. Patients with metastatic pediatric sarcomas display elevated serum CXCR2 ligands, and elevated CXCL8 is associated with diminished survival in this population. We conclude that accumulation of MDSCs in the tumor bed limits the efficacy of checkpoint blockade in cancer. We also identify CXCR2 as a novel target for modulating tumor immune escape and present evidence that CXCR2(+)CD11b(+)Ly6G(hi) MDSCs are an important suppressive myeloid subset in pediatric sarcomas. These findings present a translatable strategy to improve the efficacy of checkpoint blockade by preventing trafficking of MDSCs to the tumor site.
Collapse
Affiliation(s)
- Steven L Highfill
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yongzhi Cui
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Amber J Giles
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jillian P Smith
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hua Zhang
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elizabeth Morse
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rosandra N Kaplan
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Crystal L Mackall
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
2
|
Cintolo JA, Datta J, Mathew SJ, Czerniecki BJ. Dendritic cell-based vaccines: barriers and opportunities. Future Oncol 2013; 8:1273-99. [PMID: 23130928 DOI: 10.2217/fon.12.125] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) have several characteristics that make them an ideal vehicle for tumor vaccines, and with the first US FDA-approved DC-based vaccine in use for the treatment of prostate cancer, this technology has become a promising new therapeutic option. However, DC-based vaccines face several barriers that have limited their effectiveness in clinical trials. A major barrier includes the activation state of the DC. Both DC lineage and maturation signals must be selected to optimize the antitumor response and overcome immunosuppressive effects of the tumor microenvironment. Another barrier to successful vaccination is the selection of target antigens that will activate both CD8(+) and CD4(+) T cells in a potent, immune-specific manner. Finally, tumor progression and immune dysfunction limit vaccine efficacy in advanced stages, which may make DC-based vaccines more efficacious in treating early-stage disease. This review underscores the scientific basis and advances in the development of DC-based vaccines, focuses on current barriers to success and highlights new research opportunities to address these obstacles.
Collapse
Affiliation(s)
- Jessica A Cintolo
- Department of Surgery & Harrison Department of Surgical Research, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
3
|
Izhak L, Ambrosino E, Kato S, Parish ST, O’Konek JJ, Weber H, Xia Z, Venzon D, Berzofsky JA, Terabe M. Delicate balance among three types of T cells in concurrent regulation of tumor immunity. Cancer Res 2013; 73:1514-23. [PMID: 23319803 PMCID: PMC3622595 DOI: 10.1158/0008-5472.can-12-2567] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The nature of the regulatory cell types that dominate in any given tumor is not understood at present. Here, we addressed this question for regulatory T cells (Treg) and type II natural killer T (NKT) cells in syngeneic models of colorectal and renal cancer. In mice with both type I and II NKT cells, or in mice with neither type of NKT cell, Treg depletion was sufficient to protect against tumor outgrowth. Surprisingly, in mice lacking only type I NKT cells, Treg blockade was insufficient for protection. Thus, we hypothesized that type II NKT cells may be neutralized by type I NKT cells, leaving Tregs as the primary suppressor, whereas in mice lacking type I NKT cells, unopposed type II NKT cells could suppress tumor immunity even when Tregs were blocked. We confirmed this hypothesis in 3 ways by reconstituting type I NKT cells as well as selectively blocking or activating type II NKT cells with antibody or the agonist sulfatide, respectively. In this manner, we showed that blockade of both type II NKT cells and Tregs is necessary to abrogate suppression of tumor immunity, but a third cell, the type I NKT cell, determines the balance between these regulatory mechanisms. As patients with cancer often have deficient type I NKT cell function, managing this delicate balance among 3 T-cell subsets may be critical for the success of immunotherapy for human cancer.
Collapse
Affiliation(s)
- Liat Izhak
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Elena Ambrosino
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Shingo Kato
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Stanley T. Parish
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Jessica J. O’Konek
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Hannah Weber
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Zheng Xia
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - David Venzon
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Jay A. Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Masaki Terabe
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| |
Collapse
|
4
|
Mabuchi A, Nagao T, Koshio O, Ishiwata T, Yano A, Suzuki K, Yokomuro K, Wheatley AM. Role of F4/80Mac-1 adherent non-parenchymal liver cells in concanavalin A-induced hepatic injury in mice. Hepatol Res 2008; 38:1040-9. [PMID: 18513334 DOI: 10.1111/j.1872-034x.2008.00362.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AIM Non-parenchymal liver cells (NPLC) play an important role in the regulation of immune responses and the inflammatory process. In this study, we hypothesized that F4/80(+)Mac-1(high+) cells were involved in the regulative feedback-modulated regulation of inflammatory responses during concanavalin A (Con A)-induced hepatitis. METHODS Hepatitis was induced in BALB/c mice by the intravenous injection of Con A. Liver injury was assessed using serum aminotransferase and pathology. The function of NPLC was assessed by FACS analysis. Accessory cell function of adherent Con A NPLC was performed with an ovalbumin specific T-helper 1 (Th1) clone proliferation assay. The culture supernatant nitric oxide (NO) content was quantified by the Griess reaction. Inducible NO synthase (iNOS) expression was demonstrated by immunohistochemistry and Western blot analysis. RESULTS The number of hepatic F4/80(+)Mac-1(high+)cells increased in a time-dependent manner after Con A administration, which consequently suppressed Th1 cell proliferation by a mechanism likely to involve NO. The iNOS expression of NPLC was elevated at 24 h post-Con A injection. In nude mice, F4/80(+)Mac-1(high+)cells did not increase in the Con A-treated liver; the NPLC did not suppress Th1 clone proliferation. CONCLUSION These findings suggest that the in vivo activation of F4/80(+)Mac-1(high+)cells by Con A administration suppresses Th1 cell proliferation by increasing NO, and subsequently reducing liver injury.
Collapse
Affiliation(s)
- Ayako Mabuchi
- Department of Physiology, Otago University, Dunedin, New Zealand; and Department of Microbiology and Immunology, Nippon Medical School, Japan
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Talmadge JE. Pathways mediating the expansion and immunosuppressive activity of myeloid-derived suppressor cells and their relevance to cancer therapy. Clin Cancer Res 2007; 13:5243-8. [PMID: 17875751 DOI: 10.1158/1078-0432.ccr-07-0182] [Citation(s) in RCA: 218] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer immunotherapy has focused on inducing and expanding CTLs and improving the immune recognition of weak antigenic determinants expressed by tumors. However, few positive clinical outcomes have been reported due, in part, to tumor-associated immunologic tolerance, supporting the need for an emphasis on overcoming immunosuppression. Systemic immunosuppression is associated with abnormal myelopoiesis secondary to tumor growth, myelosuppressive therapy, and growth factor administration and subsequent expansion/mobilization of bone marrow-derived immunosuppressive cells. These myeloid-derived suppressor cells (MDSC) reduce activated T-cell number and inhibit their function by multiple mechanisms, including depletion of l-arginine by arginase-1 (ARG1) production of nitric oxide, reactive oxygen species, and reactive nitrogen oxide species by inducible nitric oxide synthase. Increased numbers of MDSCs are associated with neoplastic, inflammatory, infectious, and graft-versus-host diseases where they restrain exuberant or novel T-cell responses. In this review, we discuss critical components of MDSC-mediated suppression of T-cell function, including cellular expansion and activation-induced secretion of immunosuppressive mediators. Both components of MDSC bioactivity are amenable to pharmacologic intervention as discussed herein. We also focus on the relationship between MDSCs, tumor growth, therapeutic responses, and the mechanisms of cellular expansion, activation, and immunosuppression.
Collapse
Affiliation(s)
- James E Talmadge
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska 68198-7660, USA.
| |
Collapse
|
6
|
Bel'skaya NV, Bel'skii YP, Danilets MG, Trofimova ES, Sherstoboev EY, Agafonov VI. Effects of natural suppressor cells on production of Th1 and Th2 cytokines and proliferation of polarized T cells. Bull Exp Biol Med 2006; 141:440-3. [PMID: 17152366 DOI: 10.1007/s10517-006-0194-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Natural suppressor cells from the bone marrow of intact animals and mice with Th1- and Th2-dependent immune response largely suppressed the production of Th1 (IFN-gamma, IL-2) and Th2 cytokines (IL-4). Antiproliferative activity of natural suppressor cells from intact and immunized animals was mediated by NO. Proliferation of Th1 cells was suppressed greater than that of Th2 cells. This selective effect of natural suppressor cells on the proliferation of target lymphocytes was due to higher stimulatory potential of Th1 cells (in comparison with Th2) towards natural suppressor cells.
Collapse
Affiliation(s)
- N V Bel'skaya
- Institute of Pharmacology, Tomsk Research Center, Siberian Division of Russian Academy of Medical Sciences.
| | | | | | | | | | | |
Collapse
|
7
|
Kamihira T, Shimoda S, Nakamura M, Yokoyama T, Takii Y, Kawano A, Handa M, Ishibashi H, Gershwin ME, Harada M. Biliary epithelial cells regulate autoreactive T cells: implications for biliary-specific diseases. Hepatology 2005; 41:151-9. [PMID: 15619239 DOI: 10.1002/hep.20494] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The biliary epithelial cell (BEC) is the target for several human immune mediated liver diseases, including primary biliary cirrhosis, but it is not always clear whether the BEC functions as an accessory cell or an antigen presenting cell, although it is well documented that BECs express high levels of human leukocyte antigen Class II, intercellular adhesion molecule-1, and lymphocyte function-associated antigen-3. To examine this issue, we established autoreactive T-cell clones from human leukocyte antigen-DR53 patients with primary biliary cirrhosis and characterized BEC function as a function of the ability of BECs to regulate T-cell activation. We report herein that BEC-mediated T-cell activation occurs partially via programmed death 1 ligands in a cell-contact-dependent manner. Further, such activation occurs via prostaglandin E2 production in a cell-contact-independent fashion. Moreover, the production of prostaglandin E2 was partially controlled by interleukin-1beta and tumor necrosis factor alpha. In conclusion, the regulatory activities of BECs are important for the maintenance of peripheral immune tolerance. Further, modulation of BEC function may be used for therapeutic modulation.
Collapse
Affiliation(s)
- Takashi Kamihira
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Delarbre C, Gachelin G. Injection of the immuno-modulatory drug alpha-galactosylceramide results in the recruitment of a large population of antigen-presenting cells into the liver of C57BL/6 mice. Microbes Infect 2004; 6:360-8. [PMID: 15050963 DOI: 10.1016/j.micinf.2003.11.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2003] [Accepted: 11/29/2003] [Indexed: 11/17/2022]
Abstract
Injection of the immuno-modulatory drug alpha-galactosylceramide into C57BL/6 mice leads to the already known apoptosis of natural killer T (NKT) cells and to thus far undescribed large changes in the leukocyte populations of the liver. These changes are characterized by the recruitment of neutrophils and that of a population of large monocytic cells. The latter cells display the morphological and immunological features of natural suppressor cells. Their recruitment in the liver depends on the presence of NKT cells, most probably through the local release of cytokines and chemokines by activated NKT cells. We discuss the ubiquitous, long-term effects of alpha-galactosylceramide injection on immuno-pathological processes mediated through the NKT-triggered recruitment of a subset of large macrophages/monocytes.
Collapse
Affiliation(s)
- Christiane Delarbre
- Département d'immunologie, Unité de Biologie moléculaire du gene, Inserm U 277, Institut Pasteur, 25, rue du Dr Roux, 75015 Paris, France
| | | |
Collapse
|
9
|
Terabe M, Matsui S, Park JM, Mamura M, Noben-Trauth N, Donaldson DD, Chen W, Wahl SM, Ledbetter S, Pratt B, Letterio JJ, Paul WE, Berzofsky JA. Transforming growth factor-beta production and myeloid cells are an effector mechanism through which CD1d-restricted T cells block cytotoxic T lymphocyte-mediated tumor immunosurveillance: abrogation prevents tumor recurrence. ACTA ACUST UNITED AC 2004; 198:1741-52. [PMID: 14657224 PMCID: PMC2194133 DOI: 10.1084/jem.20022227] [Citation(s) in RCA: 427] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Our previous work demonstrated that cytotoxic T lymphocyte (CTL)-mediated tumor immunosurveillance of the 15-12RM tumor could be suppressed by a CD1d-restricted lymphocyte, most likely a natural killer (NK) T cell, which produces interleukin (IL)-13. Here we present evidence for the effector elements in this suppressive pathway. T cell–reconstituted recombination activating gene (RAG)2 knockout (KO) and RAG2/IL-4 receptor α double KO mice showed that inhibition of immunosurveillance requires IL-13 responsiveness by a non–T non–B cell. Such nonlymphoid splenocytes from tumor-bearing mice produced more transforming growth factor (TGF)-β, a potent inhibitor of CTL, ex vivo than such cells from naive mice, and this TGF-β production was dependent on the presence in vivo of both IL-13 and CD1d-restricted T cells. Ex vivo TGF-β production was also abrogated by depleting either CD11b+ or Gr-1+ cells from the nonlymphoid cells of tumor-bearing mice. Further, blocking TGF-β or depleting Gr-1+ cells in vivo prevented the tumor recurrence, implying that TGF-β made by a CD11b+ Gr-1+ myeloid cell, in an IL-13 and CD1d-restricted T cell–dependent mechanism, is necessary for down-regulation of tumor immunosurveillance. Identification of this stepwise regulation of immunosurveillance, involving CD1-restricted T cells, IL-13, myeloid cells, and TGF-β, explains previous observations on myeloid suppressor cells or TGF-β and provides insights for targeted approaches for cancer immunotherapy, including synergistic blockade of TGF-β and IL-13.
Collapse
Affiliation(s)
- Masaki Terabe
- Molecular Immunogenetics and Vaccine Research Section, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|