1
|
Sjøgren T, Islam S, Filippov I, Jebrzycka A, Sulen A, Breivik LE, Hellesen A, Jørgensen AP, Lima K, Tserel L, Kisand K, Peterson P, Ranki A, Husebye ES, Oftedal BE, Wolff AS. Single cell characterization of blood and expanded regulatory T cells in autoimmune polyendocrine syndrome type 1. iScience 2024; 27:109610. [PMID: 38632993 PMCID: PMC11022049 DOI: 10.1016/j.isci.2024.109610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 02/06/2024] [Accepted: 03/25/2024] [Indexed: 04/19/2024] Open
Abstract
Immune tolerance fails in autoimmune polyendocrine syndrome type 1 (APS-1) because of AIRE mutations. We have used single cell transcriptomics to characterize regulatory T cells (Tregs) sorted directly from blood and from in vitro expanded Tregs in APS-1 patients compared to healthy controls. We revealed only CD52 and LTB (down) and TXNIP (up) as consistently differentially expressed genes in the datasets. There were furthermore no large differences of the TCR-repertoire of expanded Tregs between the cohorts, but unique patients showed a more restricted use of specific clonotypes. We also found that in vitro expanded Tregs from APS-1 patients had similar suppressive capacity as controls in co-culture assays, despite expanding faster and having more exhausted cells. Our results suggest that APS-1 patients do not have intrinsic defects in their Treg functionality, and that their Tregs can be expanded ex vivo for potential therapeutic applications.
Collapse
Affiliation(s)
- Thea Sjøgren
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Shahinul Islam
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Igor Filippov
- QIAGEN Aarhus A/S, Aarhus, Denmark
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | | | - André Sulen
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Lars E. Breivik
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | | | | | - Kari Lima
- Department of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Liina Tserel
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Kai Kisand
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Pärt Peterson
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Annamari Ranki
- Department of Dermatology, Allergology and Venereology, University of Helsinki and Helsinki University Hospital, Inflammation Centre, Helsinki, Finland
| | - Eystein S. Husebye
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Bergithe E. Oftedal
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anette S.B. Wolff
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
2
|
Kikuchi H, Maishi N, Yu L, Jia Z, Li C, Sato M, Takeda R, Ishizuka K, Hida Y, Shinohara N, Hida K. Low-dose metronomic cisplatin as an antiangiogenic and anti-inflammatory strategy for cancer. Br J Cancer 2024; 130:336-345. [PMID: 38036665 PMCID: PMC10803316 DOI: 10.1038/s41416-023-02498-2] [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: 03/16/2023] [Revised: 10/28/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Conventional chemotherapy is based on the maximum tolerated dose (MTD) and requires treatment-free intervals to restore normal host cells. MTD chemotherapy may induce angiogenesis or immunosuppressive cell infiltration during treatment-free intervals. Low-dose metronomic (LDM) chemotherapy is defined as frequent administration at lower doses and causes less inflammatory change, whereas MTD chemotherapy induces an inflammatory change. Although several LDM regimens have been applied, LDM cisplatin (CDDP) has been rarely reported. This study addressed the efficacy of LDM CDDP on tumour endothelial cell phenotypic alteration compared to MTD CDDP. METHODS Tumour growth and metastasis were assessed in bladder cancer-bearing mice treated with LDM or MTD gemcitabine (GEM) and CDDP. To elucidate the therapeutic effects of LDM CDDP, the change of tumour vasculature, tumour-infiltrating immune cells and inflammatory changes were evaluated by histological analysis and mRNA expression in tumour tissues. RESULTS Tumour growth and bone metastasis were more suppressed by LDM CDDP + MTD GEM treatment than MTD CDDP + MTD GEM. Myeloid-derived suppressor cell accumulation was reduced by LDM CDDP, whereas inflammatory change was induced in the tumour microenvironment by MTD CDDP. CONCLUSION LDM CDDP does not cause inflammatory change unlike MTD CDDP, suggesting that it is a promising strategy in chemotherapy.
Collapse
Affiliation(s)
- Hiroshi Kikuchi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Nako Maishi
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan
| | - Li Yu
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan
| | - Zi Jia
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan
| | - Cong Li
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan
| | - Masumi Sato
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Ryo Takeda
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan
| | - Keita Ishizuka
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Advanced Robotic and Endoscopic Surgery, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Nobuo Shinohara
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kyoko Hida
- Vascular Biology, Frontier Research Unit, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Faculty of Dental Medicine, Sapporo, Japan.
| |
Collapse
|
3
|
Aristin Revilla S, Kranenburg O, Coffer PJ. Colorectal Cancer-Infiltrating Regulatory T Cells: Functional Heterogeneity, Metabolic Adaptation, and Therapeutic Targeting. Front Immunol 2022; 13:903564. [PMID: 35874729 PMCID: PMC9304750 DOI: 10.3389/fimmu.2022.903564] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/06/2022] [Indexed: 11/18/2022] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease with one of the highest rates of incidence and mortality among cancers worldwide. Understanding the CRC tumor microenvironment (TME) is essential to improve diagnosis and treatment. Within the CRC TME, tumor-infiltrating lymphocytes (TILs) consist of a heterogeneous mixture of adaptive immune cells composed of mainly anti-tumor effector T cells (CD4+ and CD8+ subpopulations), and suppressive regulatory CD4+ T (Treg) cells. The balance between these two populations is critical in anti-tumor immunity. In general, while tumor antigen-specific T cell responses are observed, tumor clearance frequently does not occur. Treg cells are considered to play an important role in tumor immune escape by hampering effective anti-tumor immune responses. Therefore, CRC-tumors with increased numbers of Treg cells have been associated with promoting tumor development, immunotherapy failure, and a poorer prognosis. Enrichment of Treg cells in CRC can have multiple causes including their differentiation, recruitment, and preferential transcriptional and metabolic adaptation to the TME. Targeting tumor-associated Treg cell may be an effective addition to current immunotherapy approaches. Strategies for depleting Treg cells, such as low-dose cyclophosphamide treatment, or targeting one or more checkpoint receptors such as CTLA-4 with PD-1 with monoclonal antibodies, have been explored. These have resulted in activation of anti-tumor immune responses in CRC-patients. Overall, it seems likely that CRC-associated Treg cells play an important role in determining the success of such therapeutic approaches. Here, we review our understanding of the role of Treg cells in CRC, the possible mechanisms that support their homeostasis in the tumor microenvironment, and current approaches for manipulating Treg cells function in cancer.
Collapse
Affiliation(s)
- Sonia Aristin Revilla
- Center Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, Netherlands
- Laboratory Translational Oncology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Onno Kranenburg
- Laboratory Translational Oncology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Paul J. Coffer
- Center Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, Netherlands
- *Correspondence: Paul J. Coffer,
| |
Collapse
|
4
|
Low-dose cyclophosphamide combined with IL-2 inhibits tumor growth by decreasing regulatory T cells and increasing CD8+ T cells and natural killer cells in mice. Immunobiology 2022; 227:152212. [DOI: 10.1016/j.imbio.2022.152212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 03/05/2022] [Accepted: 03/26/2022] [Indexed: 11/19/2022]
|
5
|
Chen W, Ye S, Wu Y, Pei X, Xiang L, Ping B, Shan B, Yang H. Changes in peripheral lymphocyte populations in patients with advanced/recurrent ovarian cancer undergoing splenectomy during cytoreductive surgery. J Ovarian Res 2021; 14:113. [PMID: 34461965 PMCID: PMC8404261 DOI: 10.1186/s13048-021-00860-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 08/14/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To investigate changes in peripheral lymphocyte subsets after splenectomy during cytoreductive surgery for advanced or recurrent ovarian cancers. METHODS We enrolled 83 patients with advanced or recurrent ovarian cancer who underwent cytoreductive surgery. Twenty patients who also underwent splenectomy were assigned to the splenectomy cohort and the rest were assigned to the non-splenectomy cohort. Flow cytometry was used to measure peripheral lymphocyte subsets consisting of T cells, regulatory T cells, natural killer cells, B cells, and activation antigens before and after surgery. RESULTS There was no difference in the number and distribution of peripheral lymphocyte subsets between the two cohorts before surgery. After surgery, we observed elevated levels of T cells (CD3+, CD3+CD8+) in the splenectomy cohort compared to those in the non-splenectomy cohort, and the difference was statistically significant. CD8+CD28+ T cells had a significant decreasing tendency (P = 0.011) while CD3+/HLA-DR+ T cells showed the opposite trend (P = 0.001) in the splenectomy cohort. The proportion of Tregs (P = 0.005) and B cells (P < 0.001) including CD3-/HLA-DR+ B cells (P = 0.007) increased after surgery, and the absolute number of T cells and NK cells decreased to different extents (P < 0.001) in the non-splenectomy cohort. The post-operative percentage of CD8+CD28+ T cells was less than the pre-operative percentage (P = 0.022), which was similar to the splenectomy cohort. There was no significant difference in progression-free survival or overall survival between the groups after a median follow-up time of 41 months. CONCLUSIONS The changes in peripheral lymphocyte populations were different between patients with and those without splenectomy during cytoreductive surgery for ovarian cancers. T cells were increased and activated in the splenectomy cohort, whereas, B cells were increased and activated in the non-splenectomy cohort.
Collapse
Affiliation(s)
- Wei Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Obstetrics and Gynecology, Minhang Hospital, Fudan University, The Central Hospital of Minhang District, Shanghai, China
| | - Shuang Ye
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yutuan Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xuan Pei
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Libing Xiang
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Bo Ping
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Boer Shan
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Huijuan Yang
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
6
|
Chalons P, Courtaut F, Limagne E, Chalmin F, Cantos-Villar E, Richard T, Auger C, Chabert P, Schini-Kerth V, Ghiringhelli F, Aires V, Delmas D. Red Wine Extract Disrupts Th17 Lymphocyte Differentiation in a Colorectal Cancer Context. Mol Nutr Food Res 2020; 64:e1901286. [PMID: 32306526 DOI: 10.1002/mnfr.201901286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/18/2020] [Indexed: 12/13/2022]
Abstract
SCOPE Scope: It is well established that immune response and inflammation promote tumoral progression. Immune cells communicate through direct contact or through cytokine secretion, and it is the pro-inflammatory status that will tip the balance toward tumor progression or anti-tumor immunity. It is demonstrated here that a red wine extract (RWE) can decrease inflammation through its action on the inflammasome complex. This study determines whether an RWE could impact other key actors of inflammation, including T helper 17 (Th17) immune cells in particular. METHODS AND RESULTS Methods and results: Using an RWE containing 4.16 g of polyphenols/liter of wine, it is shown that RWE decreases colorectal cancer cells in vitro and induces a reduction in colorectal tumor growth associated with a decrease in tumor-infiltrating lymphocytes in vivo. The process of T-lymphocyte differentiation in Th17 cells is altered by RWE, as revealed by the decrease in the expression of key actors controlling this process, such as signal transducer and activator of transcription 3 and retinoid acid-related orphan receptor γt. This disruption is associated with an inhibition of inflammatory interleukin 17 secretion. CONCLUSION The data highlights the major involvement of Th17 immune cells in the biological effects of an RWE.
Collapse
Affiliation(s)
- Pauline Chalons
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France
| | - Flavie Courtaut
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France
| | - Emeric Limagne
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France.,Plateforme de Transfert en Biologie du Cancer (PTBC) Centre Georges François Leclerc, Dijon, F-21000, France
| | - Fanny Chalmin
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France
| | - Emma Cantos-Villar
- Instituto de Investigación y Formación Agraria y Pesquera (IFAPA) Rancho de La Merced, Ctra. Trebujena, 11.471 Jerez de la Frontera, Cadiz, Spain
| | - Tristan Richard
- Université de Bordeaux, Unité de Recherche Œnologie, EA 4577, USC 1366 INRA, Equipe Molécules d'Intérêt Biologique - ISVV, Villenave-d'Ornon, F-33882, France
| | - Cyril Auger
- UMR 1260 INSERM Nanomédecine Régénérative, Université de Strasbourg, Illkirch, F-67401, France
| | - Philippe Chabert
- UMR CNRS 7021 - Laboratoire de Bioimagerie et Pathologies, Université de Strasbourg, Illkirch-Graffenstaden, F-67401, France
| | - Valérie Schini-Kerth
- UMR 1260 INSERM Nanomédecine Régénérative, Université de Strasbourg, Illkirch, F-67401, France
| | - François Ghiringhelli
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France.,Plateforme de Transfert en Biologie du Cancer (PTBC) Centre Georges François Leclerc, Dijon, F-21000, France.,Department of Medical Oncology, Centre Georges François Leclerc, Dijon, F-21000, France
| | - Virginie Aires
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France
| | - Dominique Delmas
- Université de Bourgogne Franche-Comté, Dijon, F-21000, France.,INSERM Research Center U1231-Cancer and Adaptive Immune Response Team, Dijon, F-21000, France.,Department of Medical Oncology, Centre Georges François Leclerc, Dijon, F-21000, France
| |
Collapse
|
7
|
Sag D, Ayyildiz ZO, Gunalp S, Wingender G. The Role of TRAIL/DRs in the Modulation of Immune Cells and Responses. Cancers (Basel) 2019; 11:cancers11101469. [PMID: 31574961 PMCID: PMC6826877 DOI: 10.3390/cancers11101469] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/09/2019] [Accepted: 09/20/2019] [Indexed: 12/26/2022] Open
Abstract
Expression of TRAIL (tumor necrosis factor–related apoptosis–inducing ligand) by immune cells can lead to the induction of apoptosis in tumor cells. However, it becomes increasingly clear that the interaction of TRAIL and its death receptors (DRs) can also directly impact immune cells and influence immune responses. Here, we review what is known about the role of TRAIL/DRs in immune cells and immune responses in general and in the tumor microenvironment in particular.
Collapse
Affiliation(s)
- Duygu Sag
- Izmir Biomedicine and Genome Center (IBG), 35340 Balcova/Izmir, Turkey.
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Zeynep Ozge Ayyildiz
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Sinem Gunalp
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Gerhard Wingender
- Izmir Biomedicine and Genome Center (IBG), 35340 Balcova/Izmir, Turkey.
- Department of Biomedicine and Health Technologies, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| |
Collapse
|
8
|
de Looff M, de Jong S, Kruyt FAE. Multiple Interactions Between Cancer Cells and the Tumor Microenvironment Modulate TRAIL Signaling: Implications for TRAIL Receptor Targeted Therapy. Front Immunol 2019; 10:1530. [PMID: 31333662 PMCID: PMC6617985 DOI: 10.3389/fimmu.2019.01530] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/19/2019] [Indexed: 01/09/2023] Open
Abstract
Tumor necrosis factor (TNF) related apoptosis-inducing ligand (TRAIL) signaling is far more complex than initially anticipated and can lead to either anti- or protumorigenic effects, hampering the successful clinical use of therapeutic TRAIL receptor agonists. Cell autonomous resistance mechanisms have been identified in addition to paracrine factors that can modulate apoptosis sensitivity. The tumor microenvironment (TME), consisting of cellular and non-cellular components, is a source for multiple signals that are able to modulate TRAIL signaling in tumor and stromal cells. Particularly immune effector cells, also part of the TME, employ the TRAIL/TRAIL-R system whereby cell surface expressed TRAIL can activate apoptosis via TRAIL receptors on tumor cells, which is part of tumor immune surveillance. In this review we aim to dissect the impact of the TME on signaling induced by endogenous and exogenous/therapeutic TRAIL, thereby distinguishing different components of the TME such as immune effector cells, neutrophils, macrophages, and non-hematopoietic stromal cells. In addition, also non-cellular biochemical and biophysical properties of the TME are considered including mechanical stress, acidity, hypoxia, and glucose deprivation. Available literature thus far indicates that tumor-TME interactions are complex and often bidirectional leading to tumor-enhancing or tumor-reducing effects in a tumor model- and tumor type-dependent fashion. Multiple signals originating from different components of the TME simultaneously affect TRAIL receptor signaling. We conclude that in order to unleash the full clinical potential of TRAIL receptor agonists it will be necessary to increase our understanding of the contribution of different TME components on outcome of therapeutic TRAIL receptor activation in order to identify the most critical mechanism responsible for resistance, allowing the design of effective combination treatments.
Collapse
Affiliation(s)
- Margot de Looff
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Steven de Jong
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Frank A E Kruyt
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
9
|
Expression of costimulatory and inhibitory receptors in FoxP3 + regulatory T cells within the tumor microenvironment: Implications for combination immunotherapy approaches. Adv Cancer Res 2019; 144:193-261. [PMID: 31349899 DOI: 10.1016/bs.acr.2019.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The unprecedented success of immune checkpoint inhibitors has given rise to a rapidly growing number of immuno-oncology agents undergoing preclinical and clinical development and an exponential increase in possible combinations. Defining a clear rationale for combinations by identifying synergies between immunomodulatory pathways has therefore become a high priority. Immunosuppressive regulatory T cells (Tregs) within the tumor microenvironment (TME) represent a major roadblock to endogenous and therapeutic tumor immunity. However, Tregs are also essential for the maintenance of immunological self-tolerance, and share many molecular pathways with conventional T cells including cytotoxic T cells, the primary mediators of tumor immunity. Hence the inability to specifically target and neutralize Tregs within the TME of cancer patients without globally compromising self-tolerance poses a significant challenge. Here we review recent advances in the characterization of tumor-infiltrating Tregs with a focus on costimulatory and inhibitory receptors. We discuss receptor expression patterns, their functional role in Treg biology and mechanistic insights gained from targeting these receptors in preclinical models to evaluate their potential as clinical targets. We further outline a framework of parameters that could be used to refine the assessment of Tregs in cancer patients and increase their value as predictive biomarkers. Finally, we propose modalities to integrate our increasing knowledge on Treg phenotype and function for the rational design of checkpoint inhibitor-based combination therapies. Such combinations have great potential for synergy, as they could concomitantly enhance cytotoxic T cells and inhibit Tregs within the TME, thereby increasing the efficacy of current cancer immunotherapies.
Collapse
|
10
|
Collignon A, Silvy F, Robert S, Trad M, Germain S, Nigri J, André F, Rigot V, Tomasini R, Bonnotte B, Lombardo D, Mas E, Beraud E. Dendritic cell-based vaccination: powerful resources of immature dendritic cells against pancreatic adenocarcinoma. Oncoimmunology 2018; 7:e1504727. [PMID: 30524902 DOI: 10.1080/2162402x.2018.1504727] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 07/19/2018] [Accepted: 07/21/2018] [Indexed: 02/06/2023] Open
Abstract
Pancreatic adenocarcinoma (PAC) has a poor prognosis. One treatment approach, investigated here, is to reinforce antitumor immunity. Dendritic cells (DCs) are essential for the development and regulation of adaptive host immune responses against tumors. A major role for DCs may be as innate tumoricidal effector cells. We explored the efficacy of vaccination with immature (i)DCs, after selecting optimal conditions for generating immunostimulatory iDCs. We used two models, C57BL/6Jrj mice with ectopic tumors induced by the PAC cell line, Panc02, and genetically engineered (KIC) mice developing PAC. Therapeutic iDC-vaccination resulted in a significant reduction in tumor growth in C57BL/6Jrj mice and prolonged survival in KIC mice. Prophylactic iDC-vaccination prevented subcutaneous tumor development. These protective effects were long-lasting in Panc02-induced tumor development, but not in melanoma. iDC-vaccination impacted the immune status of the hosts by greatly increasing the percentage of CD8+ T-cells, and natural killer (NK)1.1+ cells, that express granzyme B associated with Lamp-1 and IFN-γ. Efficacy of iDC-vaccination was CD8+ T-cell-dependent but NK1.1+ cell-independent. We demonstrated the ability of DCs to produce peroxynitrites and to kill tumor cells; this killing activity involved peroxynitrites. Altogether, these findings make killer DCs the pivotal actors in the beneficial clinical outcome that accompanies antitumor immune responses. We asked whether efficacy can be improved by combining DC-vaccination with the FOLFIRINOX regimen. Combined treatment significantly increased the lifespan of KIC mice with PAC. Prolonged treatment with FOLFIRINOX clearly augmented this beneficial effect. Combining iDC-vaccination with FOLFIRINOX may therefore represent a promising therapeutic option for patients with PAC.
Collapse
Affiliation(s)
- Aurélie Collignon
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Marseille, France
| | - Françoise Silvy
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Marseille, France
| | | | - Malika Trad
- CHU Dijon-Bocage, Médecine interne et Immunologie Clinique, Dijon, France
| | - Sébastien Germain
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Marseille, France
| | - Jérémy Nigri
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Cancer Research Center of Marseille, Marseille, France
| | - Frédéric André
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Marseille, France
| | - Véronique Rigot
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Marseille, France
| | - Richard Tomasini
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Cancer Research Center of Marseille, Marseille, France
| | - Bernard Bonnotte
- CHU Dijon-Bocage, Médecine interne et Immunologie Clinique, Dijon, France
| | - Dominique Lombardo
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Marseille, France
| | - Eric Mas
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Marseille, France
| | - Evelyne Beraud
- Aix Marseille Univ, INSERM, CRO2, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Marseille, France
| |
Collapse
|
11
|
Changes in regulatory T cells in patients with ovarian cancer undergoing surgery: Preliminary results. Int Immunopharmacol 2017; 47:244-250. [PMID: 28437737 DOI: 10.1016/j.intimp.2017.04.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/31/2017] [Accepted: 04/05/2017] [Indexed: 01/15/2023]
Abstract
Regulatory T cells (Treg) suppress immune responses in patients with cancer. Surgery is the most effective therapeutic strategy for ovarian cancer (OC). However, the interplay between the Treg population and surgical resection remains unclear. 61 patients with OC who received no prior treatment were enrolled in the study. Treg percentages were characterized from peripheral blood mononuclear cells. We investigated CD4+CD25+, CD4+CD25+Foxp3+, CD8+CD28-, and CD8+Foxp3+ Tregs in OC patients and their postoperative changes using flow cytometry. Treg percentages were significantly higher in OC patients than those in benign ovarian tumors (BOT) and healthy controls. Higher percentages of Tregs were found in patients with stage III/IV than stage I/II OC. Treg percentages were significantly decreased postoperatively. The postoperative Treg percentages in patients with stage I/II OC were similar to those in BOT patients, while postoperative Treg percentages in patients with stage III/IV OC remained higher. Tregs were markedly lower on postoperative day (POD) 3 than preoperatively. They increased slightly after 7days, but remained lower than preoperative levels. These data suggested that Tregs continued to decline from POD 7 to POD 30. Treg percentages are correlated with the tumor burden and could be a key factor in monitoring the immunological status of patients with OC.
Collapse
|
12
|
Abu Eid R, Razavi GSE, Mkrtichyan M, Janik J, Khleif SN. Old-School Chemotherapy in Immunotherapeutic Combination in Cancer, A Low-cost Drug Repurposed. Cancer Immunol Res 2016; 4:377-82. [PMID: 27196429 DOI: 10.1158/2326-6066.cir-16-0048] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer immunotherapy has proven to be a potent treatment modality. Although often successful in generating antitumor immune responses, cancer immunotherapy is frequently hindered by tumor immune-escape mechanisms. Among immunosuppressive strategies within the tumor microenvironment, suppressive immune regulatory cells play a key role in promoting tumor progression through inhibiting the effector arm of the immune response. Targeting these suppressive cells can greatly enhance antitumor immune therapies, hence augmenting a highly effective therapeutic antitumor response. Several approaches are being tested to enhance the effector arm of the immune system while simultaneously inhibiting the suppressor arm. Some of these approaches are none other than traditional drugs repurposed as immune modulators. Cyclophosphamide, an old-school chemotherapeutic agent used across a wide range of malignancies, was found to be a potent immune modulator that targets suppressive regulatory immune cells within the tumor microenvironment while enhancing effector cells. Preclinical and clinical findings have confirmed the ability of low doses of cyclophosphamide to selectively deplete regulatory T cells while enhancing effector and memory cytotoxic T cells within the tumor microenvironment. These immune effects translate to suppressed tumor growth and enhanced survival, evidence of antitumor therapeutic efficacy. This article discusses the reincarnation of cyclophosphamide as an immune modulator that augments novel immunotherapeutic approaches. Cancer Immunol Res; 4(5); 377-82. ©2016 AACR.
Collapse
Affiliation(s)
- Rasha Abu Eid
- Georgia Cancer Center, Augusta University (Previously Georgia Regents University), Augusta, Georgia
| | - Ghazaleh Shoja E Razavi
- Georgia Cancer Center, Augusta University (Previously Georgia Regents University), Augusta, Georgia
| | - Mikayel Mkrtichyan
- Georgia Cancer Center, Augusta University (Previously Georgia Regents University), Augusta, Georgia
| | - John Janik
- Georgia Cancer Center, Augusta University (Previously Georgia Regents University), Augusta, Georgia
| | - Samir N Khleif
- Georgia Cancer Center, Augusta University (Previously Georgia Regents University), Augusta, Georgia.
| |
Collapse
|
13
|
Yamazaki T, Pitt JM, Vétizou M, Marabelle A, Flores C, Rekdal Ø, Kroemer G, Zitvogel L. The oncolytic peptide LTX-315 overcomes resistance of cancers to immunotherapy with CTLA4 checkpoint blockade. Cell Death Differ 2016; 23:1004-15. [PMID: 27082453 DOI: 10.1038/cdd.2016.35] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 02/29/2016] [Accepted: 03/08/2016] [Indexed: 02/08/2023] Open
Abstract
Intratumoral immunotherapies aim at reducing local immunosuppression, as well as reinstating and enhancing systemic anticancer T-cell functions, without inducing side effects. LTX-315 is a first-in-class oncolytic peptide-based local immunotherapy that meets these criteria by inducing a type of malignant cell death that elicits anticancer immune responses. Here, we show that LTX-315 rapidly reprograms the tumor microenvironment by decreasing the local abundance of immunosuppressive Tregs and myeloid-derived suppressor cells and by increasing the frequency of polyfunctional T helper type 1/type 1 cytotoxic T cells with a concomitant increase in cytotoxic T-lymphocyte antigen-4 (CTLA4) and drop in PD-1 expression levels. Logically, in tumors that were resistant to intratumoral or systemic CTLA4 blockade, subsequent local inoculation of LTX-315 cured the animals or caused tumor regressions with abscopal effects. This synergistic interaction between CTLA4 blockade and LTX-315 was reduced upon blockade of the β-chain of the interleukin-2 receptor (CD122). This preclinical study provides a strong rationale for administering the oncolytic peptide LTX-315 to patients who are receiving treatment with the CTLA4 blocking antibody ipilimumab.
Collapse
Affiliation(s)
- T Yamazaki
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1015, Equipe Labellisée Ligue Nationale Contre le Cancer, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Université Paris Sud, Paris Saclay, Le Kremlin-Bicêtre, France
| | - J M Pitt
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1015, Equipe Labellisée Ligue Nationale Contre le Cancer, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Université Paris Sud, Paris Saclay, Le Kremlin-Bicêtre, France
| | - M Vétizou
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1015, Equipe Labellisée Ligue Nationale Contre le Cancer, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Université Paris Sud, Paris Saclay, Le Kremlin-Bicêtre, France
| | - A Marabelle
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1015, Equipe Labellisée Ligue Nationale Contre le Cancer, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - C Flores
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1015, Equipe Labellisée Ligue Nationale Contre le Cancer, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Université Paris Sud, Paris Saclay, Le Kremlin-Bicêtre, France
| | - Ø Rekdal
- University of Tromsø, Tromsø, Norway.,Lytix Biopharma AS, Tromsø, Norway
| | - G Kroemer
- Université Paris Sud, Paris Saclay, Le Kremlin-Bicêtre, France.,Equipe 11 Labellisée par la Ligue Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Cell Biology and Metabolomics platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,INSERM, U1138, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT1428), Villejuif, France
| | - L Zitvogel
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1015, Equipe Labellisée Ligue Nationale Contre le Cancer, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Université Paris Sud, Paris Saclay, Le Kremlin-Bicêtre, France.,Université Pierre et Marie Curie, Paris, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT1428), Villejuif, France
| |
Collapse
|
14
|
Garg AD, Romano E, Rufo N, Agostinis P. Immunogenic versus tolerogenic phagocytosis during anticancer therapy: mechanisms and clinical translation. Cell Death Differ 2016; 23:938-51. [PMID: 26891691 DOI: 10.1038/cdd.2016.5] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 12/21/2015] [Accepted: 01/03/2016] [Indexed: 12/15/2022] Open
Abstract
Phagocytosis of dying cells is a major homeostatic process that represents the final stage of cell death in a tissue context. Under basal conditions, in a diseased tissue (such as cancer) or after treatment with cytotoxic therapies (such as anticancer therapies), phagocytosis has a major role in avoiding toxic accumulation of cellular corpses. Recognition and phagocytosis of dying cancer cells dictate the eventual immunological consequences (i.e., tolerogenic, inflammatory or immunogenic) depending on a series of factors, including the type of 'eat me' signals. Homeostatic clearance of dying cancer cells (i.e., tolerogenic phagocytosis) tends to facilitate pro-tumorigenic processes and actively suppress antitumour immunity. Conversely, cancer cells killed by immunogenic anticancer therapies may stimulate non-homeostatic clearance by antigen-presenting cells and drive cancer antigen-directed immunity. On the other hand, (a general) inflammatory clearance of dying cancer cells could have pro-tumorigenic or antitumorigenic consequences depending on the context. Interestingly, the immunosuppressive consequences that accompany tolerogenic phagocytosis can be reversed through immune-checkpoint therapies. In the present review, we discuss the pivotal role of phagocytosis in regulating responses to anticancer therapy. We give particular attention to the role of phagocytosis following treatment with immunogenic or immune-checkpoint therapies, the clinical prognostic and predictive significance of phagocytic signals for cancer patients and the therapeutic strategies that can be employed for direct targeting of phagocytic determinants.
Collapse
Affiliation(s)
- A D Garg
- Cell Death Research and Therapy (CDRT) Laboratory, Department for Cellular and Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - E Romano
- Cell Death Research and Therapy (CDRT) Laboratory, Department for Cellular and Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - N Rufo
- Cell Death Research and Therapy (CDRT) Laboratory, Department for Cellular and Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - P Agostinis
- Cell Death Research and Therapy (CDRT) Laboratory, Department for Cellular and Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Rossi AH, Farias A, Fernández JE, Bonomi HR, Goldbaum FA, Berguer PM. Brucella spp. Lumazine Synthase Induces a TLR4-Mediated Protective Response against B16 Melanoma in Mice. PLoS One 2015; 10:e0126827. [PMID: 25973756 PMCID: PMC4431812 DOI: 10.1371/journal.pone.0126827] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 04/07/2015] [Indexed: 02/03/2023] Open
Abstract
Brucella Lumazine Synthase (BLS) is a highly immunogenic decameric protein which can accept the fusion of foreign proteins at its ten N-termini. These chimeras are very efficient to elicit systemic and oral immunity without adjuvants. BLS signaling via Toll-Like Receptor 4 (TLR4) regulates innate and adaptive immune responses, inducing dendritic cell maturation and CD8+ T-cell cytotoxicity. In this work we study the effect induced by BLS in TLR4-expressing B16 melanoma. In order to evaluate the effectiveness of BLS as a preventive vaccine, C57BL/6J mice were immunized with BLS or BLS-OVA, and 35 days later were subcutaneously inoculated with B16-OVA melanoma. BLS or BLS-OVA induced a significant inhibition of tumor growth, and 50% of mice immunized with the highest dose of BLS did not develop visible tumors. This effect was not observed in TLR4-deficient mice. For treatment experiments, mice were injected with BLS or BLS-OVA 2 days after the inoculation of B16 cells. Both treatments induced significant and equal tumor growth delay and increased survival. Moreover, BLS and BLS-OVA stimulation were also effective in TLR4-deficient mice. In order to study whether BLS has a direct effect on tumor cells, B16 cells were preincubated with BLS, and after 48h, cells were inoculated. Tumors induced by BLS-stimulated cells had inhibited growth and survival was increased. In the BLS group, 40% of mice did not develop tumors. This effect was abolished by the addition of TLR4/MD2 blocking antibody to cells before BLS stimulation. Our work demonstrates that BLS immunization induces a preventive antitumor response that depends on mice TLR4. We also show that BLS generates a therapeutic effect in mice inoculated with B16 cells. Our results show that BLS acts directly in cultured tumor cells via TLR4, highly suggesting that BLS elicits its therapeutic effects acting on the TLR4 from B16 melanoma cells.
Collapse
Affiliation(s)
- Andrés H. Rossi
- Fundación Instituto Leloir, IIBBA, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ana Farias
- Fundación Instituto Leloir, IIBBA, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Javier E. Fernández
- Fundación Instituto Leloir, IIBBA, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Hernán R. Bonomi
- Fundación Instituto Leloir, IIBBA, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Fernando A. Goldbaum
- Fundación Instituto Leloir, IIBBA, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Paula M. Berguer
- Fundación Instituto Leloir, IIBBA, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
16
|
Wei H, Zhang Y, Xu F. A Minireview on the Supportive Care in Cancer with Traditional Chinese Medicine. INT J PHARMACOL 2015. [DOI: 10.3923/ijp.2015.222.225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
17
|
Zhang X, Gao L, Liang X, Guo M, Wang R, Pan Y, Liu P, Zhang F, Guo C, Zhu F, Qu C, Ma C. HBV preS2 transactivates FOXP3 expression in malignant hepatocytes. Liver Int 2015; 35:1087-94. [PMID: 25047684 DOI: 10.1111/liv.12642] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 07/11/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Recent data reported the increased expression of forkhead box protein 3 (FOXP3), the well known master regulator of CD4(+) C25(+) regulatory T cells, in hepatocellular carcinoma (HCC) cells. However, the mechanisms remain unknown. We previously showed that preS2, one of important regulatory proteins encoded by HBV, triggers transactivation of hTERT in malignant hepatocytes. Here, we aimed to explore the role of preS2 in regulating FOXP3 expression in HCC. METHODS FOXP3 expression was detected by RT-PCR, Western blot and immunohistochemical staining. Cotransfection and siRNA knockdown were involved to study the regulation effects of preS2 on FOXP3 expression in cultured HCC cell lines. Luciferase reporter assay and EMSA assay were performed to explore the mechanism of preS2-mediated FOXP3 upregulation. RESULTS Immunohistochemical staining detected significant increased FOXP3 expression in malignant hepatocytes from sections of HCC patients. The total FOXP3 expression in hepatocytes from patients with HBsAg-positive HCC was significantly increased compared to that of HBV-negative HCCs (P = 0.002). In accordance, preS2 overexpression enhanced FOXP3 expression in HCC cell lines, while preS2 knockdown significantly reduced FOXP3 expression in HBV-integrated HepG2.2.15 cells. Results of cotransfection and luciferase report assay showed that preS2 transactivated FOXP3 promoter in a dose-dependent manner. Further study identified the AP-1 binding site at 20 bp region from -465 bp to -445 bp of FOXP3 promoter was responsible for preS2-induced FOXP3 transcriptional activation. CONCLUSIONS Our data here, for the first time, provided direct evidence to demonstrate that preS2 oncoprotein encoded by HBV transactivated FOXP3 transcription in HCC cells.
Collapse
Affiliation(s)
- Xiaoning Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Shandong Provincial Key Laboratory of Infection & Immunity, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Tongu M, Harashima N, Tamada K, Chen L, Harada M. Intermittent chemotherapy can retain the therapeutic potential of anti-CD137 antibody during the late tumor-bearing state. Cancer Sci 2014; 106:9-17. [PMID: 25363339 PMCID: PMC4317772 DOI: 10.1111/cas.12568] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/20/2014] [Accepted: 10/28/2014] [Indexed: 01/08/2023] Open
Abstract
Immunomodulating monoclonal antibodies (mAb) can evoke antitumor T-cell responses, which are attenuated by regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSC). Treatment with cyclophosphamide (CP) and gemcitabine (GEM) can mitigate the immunosuppression by Treg and MDSC, respectively. In the current study, we examined the antitumor effects of a combination of local injection with anti-CD137 mAb and intermittent low-dose chemotherapy using CP and GEM in subcutaneously established CT26 colon carcinoma. Although a significant antitumor effect was observed when local anti-CD137 mAb therapy (5 μg) was started early in the tumor-bearing stage (day 10), no therapeutic efficacy was observed when the mAb therapy was started at a later tumor-bearing stage (day 17). Analyses of the tumor-infiltrating immune cells revealed that the number of Gr-1(high/low) CD11b(+) MDSC started to increase 13 days after tumor inoculation, whereas injection with low-dose (50 mg/kg) CP and GEM mitigated this increase. In addition, although intermittent injections with low-dose CP and GEM on days 10 and 18 suppressed tumor growth significantly, additional local injections of anti-CD137 mAb on days 19, 21, and 23 further augmented the therapeutic efficacy. Cytotoxic T lymphocytes reactive to CT26 and a tumor antigen peptide were induced successfully from the spleen cells of tumor-cured or tumor-stable mice. In a bilateral tumor inoculation model, this combination therapy achieved systemic therapeutic effects and suppressed the growth of mAb-untreated tumors. These results suggest that intermittent immunochemotherapy using CP and GEM could retain the therapeutic potential of anti-CD137 mAb that is normally impaired during the late tumor-bearing stage.
Collapse
Affiliation(s)
- Miki Tongu
- Department of Experimental Animals, Center for Integrated Research in Science, Shimane University, Izumo, Japan
| | | | | | | | | |
Collapse
|
19
|
Ghiringhelli F, Apetoh L. The interplay between the immune system and chemotherapy: emerging methods for optimizing therapy. Expert Rev Clin Immunol 2013; 10:19-30. [DOI: 10.1586/1744666x.2014.865520] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
20
|
Ghiringhelli F, Apetoh L. Chemotherapy and immunomodulation: from immunogenic chemotherapies to novel therapeutic strategies. Future Oncol 2013; 9:469-72. [PMID: 23560368 DOI: 10.2217/fon.12.207] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
21
|
Dudek AM, Garg AD, Krysko DV, De Ruysscher D, Agostinis P. Inducers of immunogenic cancer cell death. Cytokine Growth Factor Rev 2013; 24:319-33. [DOI: 10.1016/j.cytogfr.2013.01.005] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 01/09/2013] [Indexed: 02/07/2023]
|
22
|
Natural Killer Dendritic Cells Enhance Immune Responses Elicited by α -Galactosylceramide-Stimulated Natural Killer T Cells. BIOMED RESEARCH INTERNATIONAL 2013; 2013:460706. [PMID: 23878807 PMCID: PMC3708385 DOI: 10.1155/2013/460706] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 04/28/2013] [Accepted: 05/13/2013] [Indexed: 12/24/2022]
Abstract
Natural killer dendritic cells (NKDCs) possess potent anti-tumor activity, but the cellular effect of NKDC interactions with other innate immune cells is unclear. In this study, we demonstrate that the interaction of NKDCs and natural killer T (NKT) cells is required for the anti-tumor immune responses that are elicited by α-galactosylceramide (α-GC) in mice. The rapid and strong expression of interferon-γ by NKDCs after α-GC stimulation was dependent on NKT cells. Various NK and DC molecular markers and cytotoxic molecules were up-regulated following α-GC administration. This up-regulation could improve NKDC presentation of tumor antigens and increase cytotoxicity against tumor cells. NKDCs were required for the stimulation of DCs, NK cells, and NKT cells. The strong anti-tumor immune responses elicited by α-GC may be due to the down-regulation of regulatory T cells. Furthermore, the depletion of NKDCs dampened the tumor clearance mediated by α-GC-stimulated NKT cells in vivo. Taken together, these results indicate that complex interactions of innate immune cells might be required to achieve optimal anti-tumor immune responses during the early stages of tumorigenesis.
Collapse
|
23
|
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
|
24
|
Tongu M, Harashima N, Monma H, Inao T, Yamada T, Kawauchi H, Harada M. Metronomic chemotherapy with low-dose cyclophosphamide plus gemcitabine can induce anti-tumor T cell immunity in vivo. Cancer Immunol Immunother 2013; 62:383-91. [PMID: 22926062 PMCID: PMC11029128 DOI: 10.1007/s00262-012-1343-0] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 08/13/2012] [Indexed: 02/07/2023]
Abstract
Several chemotherapeutic drugs have immune-modulating effects. For example, cyclophosphamide (CP) and gemcitabine (GEM) diminish immunosuppression by regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), respectively. Here, we show that intermittent (metronomic) chemotherapy with low-dose CP plus GEM can induce anti-tumor T cell immunity in CT26 colon carcinoma-bearing mice. Although no significant growth suppression was observed by injections of CP (100 mg/kg) at 8-day intervals or those of CP (50 mg/kg) at 4-day intervals, CP injection (100 mg/kg) increased the frequency of tumor peptide-specific T lymphocytes in draining lymph nodes, which was abolished by two injections of CP (50 mg/kg) at a 4-day interval. Alternatively, injection of GEM (50 mg/kg) was superior to that of GEM (100 mg/kg) in suppressing tumor growth in vivo, despite the smaller dose. When CT26-bearing mice were treated with low-dose (50 mg/kg) CP plus (50 mg/kg) GEM at 8-day intervals, tumor growth was suppressed without impairing T cell function; the effect was mainly T cell dependent. The metronomic combination chemotherapy cured one-third of CT26-bearing mice that acquired tumor-specific T cell immunity. The combination therapy decreased Foxp3 and arginase-1 mRNA levels but increased IFN-γ mRNA expression in tumor tissues. The percentages of tumor-infiltrating CD45(+) cells, especially Gr-1(high) CD11b(+) MDSCs, were decreased. These results indicate that metronomic chemotherapy with low-dose CP plus GEM is a promising protocol to mitigate totally Treg- and MDSC-mediated immunosuppression and elicit anti-tumor T cell immunity in vivo.
Collapse
Affiliation(s)
- Miki Tongu
- Department of Experimental Animals, Center for Integrated Research in Science, Shimane University, Izumo, Shimane Japan
| | - Nanae Harashima
- Department of Immunology, Faculty of Medicine, Shimane University, Izumo, Shimane 693-8501 Japan
| | - Hiroyuki Monma
- Department of Immunology, Faculty of Medicine, Shimane University, Izumo, Shimane 693-8501 Japan
- Department of Surgery, Faculty of Medicine, Shimane University, Izumo, Shimane Japan
| | - Touko Inao
- Department of Surgery, Faculty of Medicine, Shimane University, Izumo, Shimane Japan
| | - Takaya Yamada
- Department of Experimental Animals, Center for Integrated Research in Science, Shimane University, Izumo, Shimane Japan
| | - Hideyuki Kawauchi
- Department of Otorhinolaryngology, Faculty of Medicine, Shimane University, Izumo, Shimane Japan
| | - Mamoru Harada
- Department of Immunology, Faculty of Medicine, Shimane University, Izumo, Shimane 693-8501 Japan
| |
Collapse
|
25
|
Toiyama D, Takaha N, Shinnoh M, Ueda T, Kimura Y, Nakamura T, Hongo F, Mikami K, Kamoi K, Kawauchi A, Miki T. Significance of serum tumor necrosis factor-related apoptosis-inducing ligand as a prognostic biomarker for renal cell carcinoma. Mol Clin Oncol 2012; 1:69-74. [PMID: 24649125 DOI: 10.3892/mco.2012.35] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2012] [Accepted: 09/28/2012] [Indexed: 12/25/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is endogenously expressed in immune cells and contributes to immunosurveillance for cancer. TRAIL induces apoptosis preferentially in various cancer cells, including renal cell carcinoma (RCC) cells. In this study, the serum TRAIL level was examined using an enzyme-linked immunosorbent assay in 52 healthy controls and in 84 RCC patients prior to surgery and its significance as a biomarker was evaluated. The median serum TRAIL level was lower in RCC patients compared to the healthy controls (55.9 vs. 103.1 pg/ml; P=0.019). RCC with lymph node metastasis (N1-2), distant metastasis (M1), stage III-IV, or microscopic venous invasion was associated with decreased serum TRAIL levels (P=0.032, 0.067, 0.020 and 0.011). When comparing serum TRAIL levels in the same RCC patients prior and subsequent to surgery (n=11), the levels were significantly higher after surgery (P=0.031). The cause-specific survival rate was significantly higher in RCC patients with high serum TRAIL levels compared to those with low serum TRAIL levels (P=0.0451). TRAIL was estimated to contribute 64 and 13% of the lymphocyte-mediated cytotoxicity against human RCC ACHN and Caki-1 cells, respectively. These data suggest that the serum TRAIL level may be useful as a prognostic biomarker in RCC patients.
Collapse
Affiliation(s)
| | | | | | - Takashi Ueda
- Departments of Urology and ; Translational Cancer Drug Development, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kyoto 602-8566, Japan
| | | | | | | | | | | | | | - Tsuneharu Miki
- Departments of Urology and ; Translational Cancer Drug Development, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kyoto 602-8566, Japan
| |
Collapse
|
26
|
Cytotoxic chemotherapy and CD4+ effector T cells: an emerging alliance for durable antitumor effects. Clin Dev Immunol 2012; 2012:890178. [PMID: 22400040 PMCID: PMC3286900 DOI: 10.1155/2012/890178] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 11/01/2011] [Accepted: 11/05/2011] [Indexed: 12/18/2022]
Abstract
Standard cytotoxic chemotherapy can initially achieve high response rates, but relapses often occur in patients and represent a severe clinical problem. As increasing numbers of chemotherapeutic agents are found to have immunostimulatory effects, there is a growing interest to combine chemotherapy and immunotherapy for synergistic antitumor effects and improved clinical benefits. Findings from recent studies suggest that highly activated, polyfunctional CD4+ effector T cells have tremendous potential in strengthening and sustaining the overall host antitumor immunity in the postchemotherapy window. This review focuses on the latest progresses regarding the impact of chemotherapy on CD4+ T-cell phenotype and function and discusses the prospect of exploiting CD4+ T cells to control tumor progression and prevent relapse after chemotherapy.
Collapse
|
27
|
Martin F, Apetoh L, Ghiringhelli F. Role of myeloid-derived suppressor cells in tumor immunotherapy. Immunotherapy 2012; 4:43-57. [DOI: 10.2217/imt.11.154] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are immature myeloid cells that infiltrate human and experimental tumors and strongly inhibit anticancer immune response directly or by inducing regulatory T-lymphocyte activity. Consequently, MDSCs are important actors of cancer-induced immune tolerance and a major obstacle to efficiency of cancer immunotherapy. Several means of preventing MDSCs accumulation or inhibiting their immunosuppressive effect were recently discovered in cancer-bearing hosts, contributing to restoring antitumor immunity and consequently to control of tumor growth. In experimental tumor models, targeting MDSCs can enhance the effects of active or passive immunotherapy. While similar effects have not yet been noted in cancer-bearing patients, recent preclinical findings demonstrating that the selective toxicity of conventional chemotherapies such as gemcitabine and 5-fluorouracil on MDSCs might contribute to their anticancer effect provide impetus to pursue investigations to unravel novel therapeutics that target MDSCs in humans.
Collapse
Affiliation(s)
- François Martin
- Institut National de la Santé et de la Recherche Médicale, Avenir Team INSERM, CRI-866 University of Burgundy, Dijon, France
| | - Lionel Apetoh
- Institut National de la Santé et de la Recherche Médicale, Avenir Team INSERM, CRI-866 University of Burgundy, Dijon, France
- Department of Medical Oncology, Georges François Leclerc Center, 21000, Dijon, France
| | | |
Collapse
|
28
|
Proietti E, Moschella F, Capone I, Belardelli F. Exploitation of the propulsive force of chemotherapy for improving the response to cancer immunotherapy. Mol Oncol 2011; 6:1-14. [PMID: 22177803 DOI: 10.1016/j.molonc.2011.11.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 11/11/2011] [Indexed: 12/19/2022] Open
Abstract
Since the early clinical studies of cancer immunotherapy, the question arose as to whether it was possible to combine it with standard cancer treatments, mostly chemotherapy. The answer, now, is past history. The combined use of immunotherapy and chemotherapy is not only possible but, in certain cases, can be advantageous, depending on the drug, the dose and the combination modalities. In order to find the best synergisms between the two treatments and to turn weak immunotherapeutic interventions into potent anticancer instruments, it is mandatory to understand the complex mechanisms responsible for the positive interactions between chemotherapy and immunotherapy. In this article, we review the current knowledge on mechanisms involved in the immunostimulating activity of chemotherapy and summarize the main studies in both mouse models and patients aimed at exploiting such mechanisms for enhancing the response to cancer immunotherapy.
Collapse
Affiliation(s)
- Enrico Proietti
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | | | | | | |
Collapse
|
29
|
Toll-like receptor 4 activation in cancer progression and therapy. Clin Dev Immunol 2011; 2011:609579. [PMID: 22110526 PMCID: PMC3216292 DOI: 10.1155/2011/609579] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 09/01/2011] [Indexed: 12/12/2022]
Abstract
Cancer immunotherapy has been the focus of intense research since the late 19th century when Coley observed that bacterial components can contribute to cancer regression by eliciting an antitumor immune response. Successful activation and maturation of tumor-specific immune cells is now known to be mediated by bacterial endotoxin, which activates Toll-like receptor 4 (TLR4). TLR4 is expressed on a variety of immune as well as tumor cells, but its activation can have opposing effects. While TLR4 activation can promote antitumor immunity, it can also result in increased tumor growth and immunosuppression. Nevertheless, TLR4 engagement by endotoxin as well as by endogenous ligands represents notable contribution to the outcome of different cancer treatments, such as radiation or chemotherapy. Further research of the role and mechanisms of TLR4 activation in cancer may provide novel antitumor vaccine adjuvants as well as TLR4 inhibitors that could prevent inflammation-induced carcinogenesis.
Collapse
|
30
|
Ma Y, Conforti R, Aymeric L, Locher C, Kepp O, Kroemer G, Zitvogel L. How to improve the immunogenicity of chemotherapy and radiotherapy. Cancer Metastasis Rev 2011; 30:71-82. [PMID: 21298323 DOI: 10.1007/s10555-011-9283-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chemotherapy or radiotherapy could induce various tumor cell death modalities, releasing tumor-derived antigen as well as danger signals that could either be captured for triggering antitumor immune response or ignored. Exploring the interplay among therapeutic drugs, tumor cell death and the immune cells should improve diagnostic, prognostic, predictive, and therapeutic management of tumor. We summarized some of the cell death-derived danger signals and the mechanism for host to sense and response to cell death in the tumor microenvironment. Based on the recent clinical or experimental findings, several strategies have been suggested to improve the immunogenicity of cell death and augment antitumor immunity.
Collapse
Affiliation(s)
- Yuting Ma
- INSERM, U1015, 94805 Villejuif, France
| | | | | | | | | | | | | |
Collapse
|
31
|
Sistigu A, Viaud S, Chaput N, Bracci L, Proietti E, Zitvogel L. Immunomodulatory effects of cyclophosphamide and implementations for vaccine design. Semin Immunopathol 2011; 33:369-83. [PMID: 21611872 DOI: 10.1007/s00281-011-0245-0] [Citation(s) in RCA: 227] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 01/12/2011] [Indexed: 12/11/2022]
Abstract
Drug repositioning refers to the utilization of a known compound in a novel indication underscoring a new mode of action that predicts innovative therapeutic options. Since 1959, alkylating agents, such as the lead compound cyclophosphamide (CTX), have always been conceived, at high dosages, as potent cytotoxic and lymphoablative drugs, indispensable for dose intensity and immunosuppressive regimen in the oncological and internal medicine armamentarium. However, more recent work highlighted the immunostimulatory and/or antiangiogenic effects of low dosing CTX (also called "metronomic CTX") opening up novel indications in the field of cancer immunotherapy. CTX markedly influences dendritic cell homeostasis and promotes IFN type I secretion, contributing to the induction of antitumor cytotoxic T lymphocytes and/or the proliferation of adoptively transferred T cells, to the polarization of CD4(+) T cells into TH1 and/or TH17 lymphocytes eventually affecting the Treg/Teffector ratio in favor of tumor regression. Moreover, CTX has intrinsic "pro-immunogenic" activities on tumor cells, inducing the hallmarks of immunogenic cell death on a variety of tumor types. Fifty years after its Food and Drug Administration approval, CTX remains a safe and affordable compound endowed with multifaceted properties and plethora of clinical indications. Here we review its immunomodulatory effects and advocate why low dosing CTX could be successfully combined to new-generation cancer vaccines.
Collapse
|
32
|
Lee JC, Hayman E, Pegram HJ, Santos E, Heller G, Sadelain M, Brentjens R. In vivo inhibition of human CD19-targeted effector T cells by natural T regulatory cells in a xenotransplant murine model of B cell malignancy. Cancer Res 2011; 71:2871-81. [PMID: 21487038 DOI: 10.1158/0008-5472.can-10-0552] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human T cells genetically modified to express chimeric antigen receptors (CAR) specific to the B cell tumor antigen CD19 can successfully eradicate systemic human CD19(+) tumors in immunocompromised SCID (severe combined immunodeficient)-Beige mice. However, in the clinical setting, CD4(+) CD25(hi) T regulatory cells (Treg) present within the tumor microenvironment may be potent suppressors of tumor-targeted effector T cells. In order to assess the impact of Tregs on CAR-modified T cells in the SCID-Beige xenotransplant model, we isolated, genetically targeted and expanded natural T regulatory cells (nTreg). In vitro nTregs modified to express CD19-targeted CARs efficiently inhibited the proliferation of activated human T cells, as well as the capacity of CD19-targeted 19-28z(+) effector T cells to lyse CD19(+) Raji tumor cells. Intravenous infusion of CD19-targeted nTregs into SCID-Beige mice with systemic Raji tumors traffic to sites of tumor and recapitulate a clinically relevant hostile tumor microenvironment. Antitumor efficacy of subsequently infused 19-28z(+) effector T cells was fully abrogated as assessed by long-term survival of treated mice. Optimal suppression by genetically targeted nTregs was dependent on nTreg to effector T-cell ratios and in vivo nTreg activation. Prior infusion of cyclophosphamide in the setting of this nTreg-mediated hostile microenvironment was able to restore the antitumor activity of subsequently infused 19-28z(+) effector T cells through the eradication of tumor-targeted nTregs. These findings have significant implications for the design of future clinical trials utilizing CAR-based adoptive T-cell therapies of cancer.
Collapse
Affiliation(s)
- James C Lee
- Center for Cell Engineering, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Maglioco A, Machuca D, Mundiñano J, Cabrera G, Camicia G, Bruzzo J, Camerano G, Costa H, Ruggiero RA, Dran GI. Lymphadenectomy exacerbates tumor growth while lymphadenectomy plus the adoptive transfer of autologous cytotoxic cells and low-dose cyclophosphamide induces regression of an established murine fibrosarcoma. Cancer Immunol Immunother 2011; 60:389-99. [PMID: 21153814 PMCID: PMC11029519 DOI: 10.1007/s00262-010-0949-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Accepted: 11/19/2010] [Indexed: 01/18/2023]
Abstract
Tumor-draining lymph node (TDLN) ablation is routinely performed in the management of cancer; nevertheless, its usefulness is at present a matter of debate. TDLN are central sites where T cell priming to tumor antigens and onset of the antitumor immune response occur. However, tumor-induced immunosuppression has been demonstrated at TDLN, leading to downregulation of antitumor reaction and tolerance induction. Tolerance in turn is a main impairment for immunotherapy trials. We used a murine immunogenic fibrosarcoma that evolves to a tolerogenic state, to study the cellular and molecular mechanisms underlying tolerance induction at the level of TDLN and to design an appropriate immunotherapy. We determined that following a transient activation, the established tumor induces signs of immunosuppression at TDLN that coexist with local and systemic evidences of antitumor response. Therefore, we evaluated the feasibility of removing TDLN in order to eliminate a focus of immunosuppression and favor tumor rejection; but instead, a marked exacerbation of tumor growth was induced. Combining TDLN ablation with the in vivo depletion of regulatory cells by low-dose cyclophosphamide and the restoring of the TDLN-derived cells into the donor mouse by adoptive transference, resulted in lowered tumor growth, enhanced survival and a considerable degree of tumor regression. Our results demonstrate that important antitumor elements can be eliminated by lymphadenectomy and proved that the concurrent administration of low-dose chemotherapy along with the reinoculation of autologous cytotoxic cells provides protection. We suggest that this protocol may be useful, especially in the cases where lymphadenectomy is mandatory.
Collapse
Affiliation(s)
- Andrea Maglioco
- División Medicina Experimental, Academia Nacional de Medicina de Buenos Aires, ILEX-CONICET, Buenos Aires, Argentina
| | - Damián Machuca
- División Medicina Experimental, Academia Nacional de Medicina de Buenos Aires, ILEX-CONICET, Buenos Aires, Argentina
| | - Juliana Mundiñano
- División Medicina Experimental, Academia Nacional de Medicina de Buenos Aires, ILEX-CONICET, Buenos Aires, Argentina
| | - Gabriel Cabrera
- División Medicina Experimental, Academia Nacional de Medicina de Buenos Aires, ILEX-CONICET, Buenos Aires, Argentina
| | - Gabriela Camicia
- División Medicina Experimental, Academia Nacional de Medicina de Buenos Aires, ILEX-CONICET, Buenos Aires, Argentina
| | - Juan Bruzzo
- División Medicina Experimental, Academia Nacional de Medicina de Buenos Aires, ILEX-CONICET, Buenos Aires, Argentina
| | - Gabriela Camerano
- División Medicina Experimental, Academia Nacional de Medicina de Buenos Aires, ILEX-CONICET, Buenos Aires, Argentina
| | - Héctor Costa
- División Medicina Experimental, Academia Nacional de Medicina de Buenos Aires, ILEX-CONICET, Buenos Aires, Argentina
| | - Raúl A. Ruggiero
- División Medicina Experimental, Academia Nacional de Medicina de Buenos Aires, ILEX-CONICET, Buenos Aires, Argentina
| | - Graciela I. Dran
- División Medicina Experimental, Academia Nacional de Medicina de Buenos Aires, ILEX-CONICET, Buenos Aires, Argentina
| |
Collapse
|
34
|
Apetoh L, Locher C, Ghiringhelli F, Kroemer G, Zitvogel L. Harnessing dendritic cells in cancer. Semin Immunol 2011; 23:42-9. [DOI: 10.1016/j.smim.2011.01.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 01/05/2011] [Indexed: 01/28/2023]
|
35
|
CXCL12 Mediates Immunosuppression in the Lymphoma Microenvironment after Allogeneic Transplantation of Hematopoietic Cells. Cancer Res 2010; 70:10170-81. [DOI: 10.1158/0008-5472.can-10-1943] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
36
|
Medina-Echeverz J, Fioravanti J, Zabala M, Ardaiz N, Prieto J, Berraondo P. Successful Colon Cancer Eradication after Chemoimmunotherapy Is Associated with Profound Phenotypic Change of Intratumoral Myeloid Cells. THE JOURNAL OF IMMUNOLOGY 2010; 186:807-15. [DOI: 10.4049/jimmunol.1001483] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
37
|
Rajecki M, Raki M, Escutenaire S, Pesonen S, Cerullo V, Helminen A, Hannuksela P, Partanen K, Laasonen L, Joensuu T, Kangasniemi L, Haavisto E, Kanerva A, Ahtiainen L, Hemminki A. Safety of glucocorticoids in cancer patients treated with oncolytic adenoviruses. Mol Pharm 2010; 8:93-103. [PMID: 20964369 DOI: 10.1021/mp1002174] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Oncolytic adenoviruses are an emerging treatment option for advanced and refractory cancer. Such patients are often treated with corticosteroids to ameliorate tumor associated symptoms. Thus, it is important to evaluate whether safety is affected by immunosuppression possibly induced by corticosteroids. Concurrent low-dose cyclophosphamide, appealing for its immunomodulatory effects, could also impact safety. In a retrospective case-control study, we evaluated the effect of systemic corticosteroid use in cancer patients receiving oncolytic virotherapy. Four treatment groups were identified: (1) oncolytic adenovirus with oral glucocorticoids, (2) virus alone, (3) virus with glucocorticoids and cyclophosphamide and (4) virus with cyclophosphamide. Adverse events, neutralizing antibody titers, viral DNA in circulation and tumor responses were evaluated. The most common adverse effects were grade 1-2 fatigue, nausea, fever and abdominal pain. Common asymptomatic findings included self-limiting grade 1-3 hyponatremia and aspartate aminotransferase increase. Safety was good and no significant differences were observed between the groups. All patients had an increase in neutralizing antibody titers post-treatment, and no trends for differences between groups were observed. There were fewer post-treatment virus genomes circulating in patients receiving glucocorticoids when compared to their control groups. Overall, glucocorticoid use in cancer patients receiving oncolytic adenovirus, with or without low-dose cyclophosphamide, seems safe.
Collapse
Affiliation(s)
- Maria Rajecki
- Cancer Gene Therapy Group, Transplantation Laboratory and Haartman Institute and Finnish Institute of Molecular Medicine, University of Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Sharabi A, Laronne-Bar-On A, Meshorer A, Haran-Ghera N. Chemoimmunotherapy reduces the progression of multiple myeloma in a mouse model. Cancer Prev Res (Phila) 2010; 3:1265-76. [PMID: 20719903 DOI: 10.1158/1940-6207.capr-10-0138] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Multiple myeloma (MM) is a B-cell malignancy characterized by clonal proliferation of malignant plasma cells in the bone marrow. Recently, we showed a correlation between increased ratios of functional regulatory T cells (Treg) and disease progression in a unique mouse model that mimics the human disease. Cyclophosphamide (CYC) is a cytotoxic alkylating agent widely used in chemotherapeutic regimens. Low-dose CYC was previously reported to selectively reduce Treg levels and to contribute to immunostimulation. Our objectives were (a) to determine whether treatment using a low-dose CYC could reduce MM progression and (b) to further characterize the modes of action underlying these effects. We found that both low- and high-dose CYC given to sick mice with hind limb paralysis resulted in the disappearance of the paralysis, the replacement of plasma tumor cells in the bone marrow by normal cell populations, and a significant prolongation of survival. However, only low-dose CYC treatment decreased the incidence of MM. Low-dose CYC rendered Tregs susceptible to apoptosis because of the downregulation of Bcl-xL and CTLA-4 in these cells, and a decreased production of interleukin 2 by effector CD4 cells. Moreover, using this treatment, we noted the recovery of IFN-γ-producing natural killer T cells and maturation of dendritic cells. Treatment of tumor-bearing mice with repeated administrations of low-dose CYC at longer time intervals (coinciding with the blocked renewal of Tregs) resulted in reduced tumor load, and the prevention or delay of disease recurrence, thereby breaking immune tolerance against MM tumor cells.
Collapse
Affiliation(s)
- Amir Sharabi
- Department of Immunology, The Weizmann Institute of Science, 240 Hertzl Street, Rehovot 76100, Israel.
| | | | | | | |
Collapse
|
39
|
Characterisation of the cutaneous pathology in non-small cell lung cancer (NSCLC) patients treated with the EGFR tyrosine kinase inhibitor erlotinib. Eur J Cancer 2010; 46:2010-9. [PMID: 20621734 DOI: 10.1016/j.ejca.2010.04.028] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Accepted: 04/28/2010] [Indexed: 12/18/2022]
Abstract
INTRODUCTION EGFR inhibitors (EGFRIs) have been shown to be clinically effective in various cancers. Unique skin toxicity is commonly observed with EGFRIs and a correlation between the clinical benefit of EGFRIs and this characteristic rash has been reported. Erlotinib is a potent EGFRI approved for treatment of non-small cell lung cancer (NSCLC) and pancreatic cancer. METHODS This is the first time in which patients were given increasing doses of an EGFRI to induce a mechanistic rash and study its associated pathology in skin. Biopsies were collected during treatment from both rash-affected and unaffected skin of 23 NSCLC patients and compared with pre-treatment biopsies. RESULTS Altered differentiation of appendegeal epithelium (hair follicles and sebaceous glands) was remarkable in both affected and unaffected skin, although epidermal growth was not significantly reduced. A predominantly mononuclear leucocyte infiltrate was detected in the interfollicular dermis or around skin appendages. This infiltrate included TRAIL-positive cells with a dendritic cell (DC) morphology, although T-cells, antigen-presenting DCs and macrophages were also evident. This is the first report showing the involvement of a dendritic cell subtype with EGFRI skin toxicity. CONCLUSIONS Altered differentiation of pilosebaceous epithelium is evident in both rash-affected and unaffected skin and constitutes the primary process of EGFRI in human skin. We propose that this eventually triggers inflammation and the EGFRI rash. TRAIL-positive inflammatory cells could link rash development and immune-triggered apoptosis of epithelial cells, including those of underlying carcinomas.
Collapse
|
40
|
Abstract
FOXP3 is a transcription factor necessary and sufficient for induction of the immunosuppressive functions in regulatory T lymphocytes. Its expression was first considered as specific of this cell type, but FOXP3 can also be transiently expressed in T-cell antigen receptor-activated human nonregulatory T cells. Recent data indicate that FOXP3 is also expressed by some nonlymphoid cells, in which it can repress various oncogenes that are restored following FOXP3 deletion or mutation. This review summarizes major advances in (1) the understanding of Foxp3 functions in human regulatory T cells, (2) the prognostic significance of Foxp3-expressing T cells in human malignancies and (3) the significance of Foxp3 expression in human tumor cells.
Collapse
|
41
|
Tongu M, Harashima N, Yamada T, Harada T, Harada M. Immunogenic chemotherapy with cyclophosphamide and doxorubicin against established murine carcinoma. Cancer Immunol Immunother 2010; 59:769-77. [PMID: 19940990 PMCID: PMC11030296 DOI: 10.1007/s00262-009-0797-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Accepted: 11/10/2009] [Indexed: 12/22/2022]
Abstract
Mitigation of regulatory T cell-mediated immunosuppression and elicitation of immunogenic tumor cell death are crucial events for optimal anti-tumor immune activity in vivo. This study was designed to investigate the potential synergistic activity of the combined use of cyclophosphamide (CP) and doxorubicin (DR), both of which are known to resolve these two issues. BALB/c mice were inoculated subcutaneously with CT-26 carcinoma cells in the bilateral flank and treated with an intraperitoneal injection of a low dose of CP followed by an intratumoral injection of DR into one side of the tumor. We found that, in addition to a significant suppression of growth on the DR-treated side of the tumor, combination therapy suppressed the growth of DR-untreated remote tumors in both tumor-specific and T cell-dependent manners. Mitomycin C showed no such synergistic anti-tumor activity with CP treatment. Combination therapy increased the frequency of interferon (IFN)-gamma-producing T lymphocytes specific to a CT-26-associated class I-binding tumor peptide in the tumor-draining lymph nodes. Real-time PCR analysis revealed that combination therapy led to an increase in IFN-gamma and tumor necrosis factor-alpha mRNA expression; however, levels of Foxp3 and transforming growth factor-beta within the remote tumor tissues were decreased. In addition, knock down of calreticulin expression in CT-26 cells using small interfering RNA attenuated anti-tumor vaccine effects induced by DR-treated CT-26 cells. These results provide an immunological rationale for the combined use of chemotherapeutic drugs, i.e., CP and DR, and further recommend their use with current cancer vaccines.
Collapse
Affiliation(s)
- Miki Tongu
- Department of Immunology, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501 Japan
- Experimental Animal Center, Shimane University, Izumo, Shimane 693-8501 Japan
| | - Nanae Harashima
- Department of Immunology, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501 Japan
| | - Takaya Yamada
- Experimental Animal Center, Shimane University, Izumo, Shimane 693-8501 Japan
| | - Takayuki Harada
- Department of Pathology, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501 Japan
| | - Mamoru Harada
- Department of Immunology, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501 Japan
| |
Collapse
|
42
|
Ma Y, Kepp O, Ghiringhelli F, Apetoh L, Aymeric L, Locher C, Tesniere A, Martins I, Ly A, Haynes NM, Smyth MJ, Kroemer G, Zitvogel L. Chemotherapy and radiotherapy: cryptic anticancer vaccines. Semin Immunol 2010; 22:113-24. [PMID: 20403709 DOI: 10.1016/j.smim.2010.03.001] [Citation(s) in RCA: 163] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2009] [Accepted: 03/15/2010] [Indexed: 02/07/2023]
Abstract
An attractive, yet hitherto unproven concept predicts that the promotion of tumor regression should elicit the host's immune response against residual tumor cells to achieve an optimal therapeutic effect. In a way, chemo- or radiotherapy must trigger "danger signals" emitted from immunogenic cell death and hence elicit "danger associated molecular patterns" to stimulate powerful anticancer immune responses. Here, based on the recent experimental and clinical evidence, we will discuss the molecular identity of the multiple checkpoints that dictate the success of "immunogenic chemotherapy" at the levels of the drug, of the tumor cell and of the host immune system.
Collapse
Affiliation(s)
- Yuting Ma
- INSERM, U805, F-94805 Villejuif, France; Institut Gustave Roussy, F-94805 Villejuif, France; Université Paris-Sud, F-94805 Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Vincent J, Mignot G, Chalmin F, Ladoire S, Bruchard M, Chevriaux A, Martin F, Apetoh L, Rébé C, Ghiringhelli F. 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res 2010; 70:3052-61. [PMID: 20388795 DOI: 10.1158/0008-5472.can-09-3690] [Citation(s) in RCA: 946] [Impact Index Per Article: 63.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Myeloid-derived suppressor cells (MDSC) accumulate in the spleen and tumor bed during tumor growth. They contribute to the immune tolerance of cancer notably by inhibiting the function of CD8(+) T cells. Thus, their elimination may hamper tumor growth by enhancing antitumor T-cell functions. We have previously reported that some anticancer agents relied on T cell-dependent anticancer responses to achieve maximal efficacy. However, the effect of anticancer agents on MDSC has remained largely unexplored. In this study, we observed that gemcitabine and 5-fluorouracil (5FU) were selectively cytotoxic on MDSC. In vivo, the treatment of tumor-bearing mice with 5FU led to a major decrease in the number of MDSC in the spleens and tumor beds of animals whereas no significant effect on T cells, natural killer cells, dendritic cells, or B cells was noted. Interestingly, 5FU showed a stronger efficacy over gemcitabine to deplete MDSC and selectively induced MDSC apoptotic cell death in vitro and in vivo. The elimination of MDSC by 5FU increased IFN-gamma production by tumor-specific CD8(+) T cells infiltrating the tumor and promoted T cell-dependent antitumor responses in vivo. Altogether, these findings suggest that the antitumor effect of 5FU is mediated, at least in part, by its selective cytotoxic action on MDSC.
Collapse
Affiliation(s)
- Julie Vincent
- INSERM Research Center 866, AVENIR Team, Faculty of Medicine, University of Burgundy, Anti-Cancer Center, Georges François Leclerc, Dijon, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Presence of Foxp3 expression in tumor cells predicts better survival in HER2-overexpressing breast cancer patients treated with neoadjuvant chemotherapy. Breast Cancer Res Treat 2010; 125:65-72. [PMID: 20229175 DOI: 10.1007/s10549-010-0831-1] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Accepted: 03/03/2010] [Indexed: 12/22/2022]
Abstract
The Forkhead Box Protein 3 is highly expressed not only in regulatory T cells, but also in tumor cells, acting as a transcriptional repressor of breast oncogenes including HER2. We investigated the prognostic significance of Foxp3 expression in cancer cells in a large cohort of patients with HER2-overexpressing breast carcinoma treated with neoadjuvant chemotherapy. Foxp3-positive tumor cells were detected by immunohistochemistry in 103 patients with primary invasive HER2-overexpressing breast carcinoma, and treated with neoadjuvant chemotherapy, with or without trastuzumab. Kaplan-Meier analysis and Cox regression model were used to assess relapse-free and overall survival, respectively, and according to the presence or the absence of Foxp3 expression in tumor cells. Breast cancer cells were Foxp3+ in 57% of tumors. Foxp3 expression in breast cancer cells was associated with better relapse-free (P = 0.005) and overall survival (P = 0.03). By multivariate analysis, the presence of Foxp3+ tumor cells produced an independent prognostic factor for both better relapse-free (P = 0.006) and overall survival (P = 0.03). These findings indicate that the presence of Foxp3+ tumor cells represents a new independent prognostic factor of improved outcome in HER2-overexpressing breast carcinoma, which could help identify high-risk patients for additional therapies after neoadjuvant chemotherapy.
Collapse
|
45
|
Zheng J, Liu Y, Lau YL, Tu W. CD40-activated B cells are more potent than immature dendritic cells to induce and expand CD4(+) regulatory T cells. Cell Mol Immunol 2010; 7:44-50. [PMID: 20081875 DOI: 10.1038/cmi.2009.103] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
CD4(+) regulatory T cells (Tregs) play an important role in maintaining immune tolerance by suppressing pathologic immune responses. The generation of large numbers of antigen-specific Tregs ex vivo is critical for the development of clinical immunotherapy based on the adoptive transfer of Tregs. Both CD40-activated B cells (CD40-B) and immature dendritic cells (imDCs) have been used as professional antigen-presenting cells (APCs) to generate antigen-specific Tregs. However, the efficiencies of CD40-B and imDCs to generate CD4(+) Tregs have not been compared directly and the mechanism driving the generation of these Tregs remains largely unknown. In this study, we found that CD40-B exhibited mature phenotypes and were more able to induce and expand CD4(high)CD25(+) Tregs than imDCs. Moreover, Tregs induced by CD40-B had greater suppressive capacity than those induced by imDCs. The generation of CD4(high)CD25(+) Tregs by CD40-B and imDCs is cell-cell contact dependent and partially relies on the expression of human leukocyte antigen (HLA)-DR and CD80/86. Differences in CD4(high)CD25(+) Treg generation efficiency were largely explained by the production of endogenous IL-2 by CD40-B. Our results suggest that CD40-B is better able to generate large numbers of antigen-specific Tregs than imDCs. Additionally, using CD40-B to generate Tregs may accelerate the clinical use of Treg-based immunotherapy in the treatment of allograft rejection, graft versus host disease (GVHD) and autoimmune diseases.
Collapse
Affiliation(s)
- Jian Zheng
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | | | | | | |
Collapse
|
46
|
Peddareddigari VG, Wang D, DuBois RN. The tumor microenvironment in colorectal carcinogenesis. CANCER MICROENVIRONMENT : OFFICIAL JOURNAL OF THE INTERNATIONAL CANCER MICROENVIRONMENT SOCIETY 2010; 3:149-66. [PMID: 21209781 PMCID: PMC2990487 DOI: 10.1007/s12307-010-0038-3] [Citation(s) in RCA: 159] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Accepted: 01/03/2010] [Indexed: 02/07/2023]
Abstract
Colorectal cancer is the second leading cause of cancer-related mortality in the United States. Therapeutic developments in the past decade have extended life expectancy in patients with metastatic disease. However, metastatic colorectal cancers remain incurable. Numerous agents that were demonstrated to have significant antitumor activity in experimental models translated into disappointing results in extending patient survival. This has resulted in more attention being focused on the contribution of tumor microenvironment to the progression of a number of solid tumors including colorectal cancer. A more complete understanding of interactions between tumor epithelial cells and their stromal elements will enhance therapeutic options and improve clinical outcome. Here we will review the role of various stromal components in colorectal carcinogenesis and discuss the potential of targeting these components for the development of future therapeutic agents.
Collapse
Affiliation(s)
- Vijay G. Peddareddigari
- Department of Cancer Biology, The University of Texas, M. D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Dingzhi Wang
- Department of Cancer Biology, The University of Texas, M. D. Anderson Cancer Center, Houston, TX 77030 USA
| | - Raymond N. DuBois
- Department of Cancer Biology, The University of Texas, M. D. Anderson Cancer Center, Houston, TX 77030 USA
- Gastrointestinal Medical Oncology, The University of Texas, M. D. Anderson Cancer Center, Unit 118, 1515 Holcombe Boulevard, Houston, TX 77030-4009 USA
| |
Collapse
|
47
|
Fraszczak J, Trad M, Janikashvili N, Cathelin D, Lakomy D, Granci V, Morizot A, Audia S, Micheau O, Lagrost L, Katsanis E, Solary E, Larmonier N, Bonnotte B. Peroxynitrite-dependent killing of cancer cells and presentation of released tumor antigens by activated dendritic cells. THE JOURNAL OF IMMUNOLOGY 2010; 184:1876-84. [PMID: 20089706 DOI: 10.4049/jimmunol.0900831] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DCs), essential for the initiation and regulation of adaptive immune responses, have been used as anticancer vaccines. DCs may also directly trigger tumor cell death. In the current study, we have investigated the tumoricidal and immunostimulatory activities of mouse bone marrow-derived DCs. Our results indicate that these cells acquire killing capabilities toward tumor cells only when activated with LPS or Pam3Cys-SK4. Using different transgenic mouse models including inducible NO synthase or GP91 knockout mice, we have further established that LPS- or Pam3Cys-SK4-activated DC killing activity involves peroxynitrites. Importantly, after killing of cancer cells, DCs are capable of engulfing dead tumor cell fragments and of presenting tumor Ags to specific T lymphocytes. Thus, upon specific stimulation, mouse bone marrow-derived DCs can directly kill tumor cells through a novel peroxynitrite-dependent mechanism and participate at virtually all levels of antitumor immune responses, which reinforces their interest in immunotherapy.
Collapse
Affiliation(s)
- Jennifer Fraszczak
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 866, Institut de Recherche Fédératif 100, Université de Bourgogne, Dijon, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Chalmin F, Ladoire S, Mignot G, Vincent J, Bruchard M, Remy-Martin JP, Boireau W, Rouleau A, Simon B, Lanneau D, De Thonel A, Multhoff G, Hamman A, Martin F, Chauffert B, Solary E, Zitvogel L, Garrido C, Ryffel B, Borg C, Apetoh L, Rébé C, Ghiringhelli F. Membrane-associated Hsp72 from tumor-derived exosomes mediates STAT3-dependent immunosuppressive function of mouse and human myeloid-derived suppressor cells. J Clin Invest 2010; 120:457-71. [PMID: 20093776 DOI: 10.1172/jci40483] [Citation(s) in RCA: 539] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Accepted: 12/02/2009] [Indexed: 12/13/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) have been identified in humans and mice as a population of immature myeloid cells with the ability to suppress T cell activation. They accumulate in tumor-bearing mice and humans and have been shown to contribute to cancer development. Here, we have isolated tumor-derived exosomes (TDEs) from mouse cell lines and shown that an interaction between TDE-associated Hsp72 and MDSCs determines the suppressive activity of the MDSCs via activation of Stat3. In addition, tumor-derived soluble factors triggered MDSC expansion via activation of Erk. TDE-associated Hsp72 triggered Stat3 activation in MDSCs in a TLR2/MyD88-dependent manner through autocrine production of IL-6. Importantly, decreasing exosome production using dimethyl amiloride enhanced the in vivo antitumor efficacy of the chemotherapeutic drug cyclophosphamide in 3 different mouse tumor models. We also demonstrated that this mechanism is relevant in cancer patients, as TDEs from a human tumor cell line activated human MDSCs and triggered their suppressive function in an Hsp72/TLR2-dependent manner. Further, MDSCs from cancer patients treated with amiloride, a drug used to treat high blood pressure that also inhibits exosome formation, exhibited reduced suppressor functions. Collectively, our findings show in both mice and humans that Hsp72 expressed at the surface of TDEs restrains tumor immune surveillance by promoting MDSC suppressive functions.
Collapse
|
49
|
Cubillos-Ruiz JR, Engle X, Scarlett UK, Martinez D, Barber A, Elgueta R, Wang L, Nesbeth Y, Durant Y, Gewirtz AT, Sentman CL, Kedl R, Conejo-Garcia JR. Polyethylenimine-based siRNA nanocomplexes reprogram tumor-associated dendritic cells via TLR5 to elicit therapeutic antitumor immunity. J Clin Invest 2009; 119:2231-44. [PMID: 19620771 PMCID: PMC2719935 DOI: 10.1172/jci37716] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Accepted: 05/27/2009] [Indexed: 02/06/2023] Open
Abstract
The success of clinically relevant immunotherapies requires reversing tumor-induced immunosuppression. Here we demonstrated that linear polyethylenimine-based (PEI-based) nanoparticles encapsulating siRNA were preferentially and avidly engulfed by regulatory DCs expressing CD11c and programmed cell death 1-ligand 1 (PD-L1) at ovarian cancer locations in mice. PEI-siRNA uptake transformed these DCs from immunosuppressive cells to efficient antigen-presenting cells that activated tumor-reactive lymphocytes and exerted direct tumoricidal activity, both in vivo and in situ. PEI triggered robust and selective TLR5 activation in vitro and elicited the production of hallmark TLR5-inducible cytokines in WT mice, but not in Tlr5-/- littermates. Thus, PEI is a TLR5 agonist that, to our knowledge, was not previously recognized. In addition, PEI-complexed nontargeting siRNA oligonucleotides stimulated TLR3 and TLR7. The nonspecific activation of multiple TLRs (specifically, TLR5 and TLR7) reversed the tolerogenic phenotype of human and mouse ovarian tumor-associated DCs. In ovarian carcinoma-bearing mice, this induced T cell-mediated tumor regression and prolonged survival in a manner dependent upon myeloid differentiation primary response gene 88 (MyD88; i.e., independent of TLR3). Furthermore, gene-specific siRNA-PEI nanocomplexes that silenced immunosuppressive molecules on mouse tumor-associated DCs elicited discernibly superior antitumor immunity and enhanced therapeutic effects compared with nontargeting siRNA-PEI nanocomplexes. Our results demonstrate that the intrinsic TLR5 and TLR7 stimulation of siRNA-PEI nanoparticles synergizes with the gene-specific silencing activity of siRNA to transform tumor-infiltrating regulatory DCs into DCs capable of promoting therapeutic antitumor immunity.
Collapse
Affiliation(s)
- Juan R. Cubillos-Ruiz
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, USA.
Nanostructured Polymers Research Center, University of New Hampshire, Durham, New Hampshire, USA.
Department of Pathology, Emory University, Atlanta, Georgia, USA.
Department of Immunology, University of Colorado, Denver, Colorado, USA.
Department of Medicine, Dartmouth Medical School, Lebanon, New Hampshire, USA
| | - Xavier Engle
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, USA.
Nanostructured Polymers Research Center, University of New Hampshire, Durham, New Hampshire, USA.
Department of Pathology, Emory University, Atlanta, Georgia, USA.
Department of Immunology, University of Colorado, Denver, Colorado, USA.
Department of Medicine, Dartmouth Medical School, Lebanon, New Hampshire, USA
| | - Uciane K. Scarlett
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, USA.
Nanostructured Polymers Research Center, University of New Hampshire, Durham, New Hampshire, USA.
Department of Pathology, Emory University, Atlanta, Georgia, USA.
Department of Immunology, University of Colorado, Denver, Colorado, USA.
Department of Medicine, Dartmouth Medical School, Lebanon, New Hampshire, USA
| | - Diana Martinez
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, USA.
Nanostructured Polymers Research Center, University of New Hampshire, Durham, New Hampshire, USA.
Department of Pathology, Emory University, Atlanta, Georgia, USA.
Department of Immunology, University of Colorado, Denver, Colorado, USA.
Department of Medicine, Dartmouth Medical School, Lebanon, New Hampshire, USA
| | - Amorette Barber
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, USA.
Nanostructured Polymers Research Center, University of New Hampshire, Durham, New Hampshire, USA.
Department of Pathology, Emory University, Atlanta, Georgia, USA.
Department of Immunology, University of Colorado, Denver, Colorado, USA.
Department of Medicine, Dartmouth Medical School, Lebanon, New Hampshire, USA
| | - Raul Elgueta
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, USA.
Nanostructured Polymers Research Center, University of New Hampshire, Durham, New Hampshire, USA.
Department of Pathology, Emory University, Atlanta, Georgia, USA.
Department of Immunology, University of Colorado, Denver, Colorado, USA.
Department of Medicine, Dartmouth Medical School, Lebanon, New Hampshire, USA
| | - Li Wang
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, USA.
Nanostructured Polymers Research Center, University of New Hampshire, Durham, New Hampshire, USA.
Department of Pathology, Emory University, Atlanta, Georgia, USA.
Department of Immunology, University of Colorado, Denver, Colorado, USA.
Department of Medicine, Dartmouth Medical School, Lebanon, New Hampshire, USA
| | - Yolanda Nesbeth
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, USA.
Nanostructured Polymers Research Center, University of New Hampshire, Durham, New Hampshire, USA.
Department of Pathology, Emory University, Atlanta, Georgia, USA.
Department of Immunology, University of Colorado, Denver, Colorado, USA.
Department of Medicine, Dartmouth Medical School, Lebanon, New Hampshire, USA
| | - Yvon Durant
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, USA.
Nanostructured Polymers Research Center, University of New Hampshire, Durham, New Hampshire, USA.
Department of Pathology, Emory University, Atlanta, Georgia, USA.
Department of Immunology, University of Colorado, Denver, Colorado, USA.
Department of Medicine, Dartmouth Medical School, Lebanon, New Hampshire, USA
| | - Andrew T. Gewirtz
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, USA.
Nanostructured Polymers Research Center, University of New Hampshire, Durham, New Hampshire, USA.
Department of Pathology, Emory University, Atlanta, Georgia, USA.
Department of Immunology, University of Colorado, Denver, Colorado, USA.
Department of Medicine, Dartmouth Medical School, Lebanon, New Hampshire, USA
| | - Charles L. Sentman
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, USA.
Nanostructured Polymers Research Center, University of New Hampshire, Durham, New Hampshire, USA.
Department of Pathology, Emory University, Atlanta, Georgia, USA.
Department of Immunology, University of Colorado, Denver, Colorado, USA.
Department of Medicine, Dartmouth Medical School, Lebanon, New Hampshire, USA
| | - Ross Kedl
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, USA.
Nanostructured Polymers Research Center, University of New Hampshire, Durham, New Hampshire, USA.
Department of Pathology, Emory University, Atlanta, Georgia, USA.
Department of Immunology, University of Colorado, Denver, Colorado, USA.
Department of Medicine, Dartmouth Medical School, Lebanon, New Hampshire, USA
| | - Jose R. Conejo-Garcia
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire, USA.
Nanostructured Polymers Research Center, University of New Hampshire, Durham, New Hampshire, USA.
Department of Pathology, Emory University, Atlanta, Georgia, USA.
Department of Immunology, University of Colorado, Denver, Colorado, USA.
Department of Medicine, Dartmouth Medical School, Lebanon, New Hampshire, USA
| |
Collapse
|
50
|
Liu Z, Kim JH, Falo LD, You Z. Tumor regulatory T cells potently abrogate antitumor immunity. THE JOURNAL OF IMMUNOLOGY 2009; 182:6160-7. [PMID: 19414769 DOI: 10.4049/jimmunol.0802664] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Regulatory T cell (Treg) from mice bearing a breast tumor were elevated (tumor Treg). In vitro, whereas tumor Treg ability to inhibit tumor-primed CD4(+) T cell activity is comparable to Treg from naive mice (naive Treg), only tumor Treg suppress naive CD8(+) T cell activation and DC function. Neither tumor Treg nor naive Treg can suppress antitumor immunity at the effector phase of the immune response induced by adoptively transferred tumor-primed CD4(+) T cells. This is consistent with the observation that, in this model, neither tumor Treg nor naive Treg can inhibit effectors in vitro or in vivo. However, tumor Treg abrogate tumor-specific CD8(+) T cell responses in tumor-draining lymph nodes and antitumor immunity at the early stage of the immune response induced by adoptively transferred tumor-primed CD4(+) T cells. These data indicate that, in this model, tumor Treg potently abrogate tumor-specific CD8(+) T cell responses in tumor-draining lymph nodes, thereby suppressing antitumor immunity at the early stage of the immune response induced by adoptively transferred tumor-primed CD4(+) T cells.
Collapse
Affiliation(s)
- Zuqiang Liu
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|