51
|
De Monte L, Wörmann S, Brunetto E, Heltai S, Magliacane G, Reni M, Paganoni AM, Recalde H, Mondino A, Falconi M, Aleotti F, Balzano G, Algül H, Doglioni C, Protti MP. Basophil Recruitment into Tumor-Draining Lymph Nodes Correlates with Th2 Inflammation and Reduced Survival in Pancreatic Cancer Patients. Cancer Res 2016; 76:1792-803. [DOI: 10.1158/0008-5472.can-15-1801-t] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/26/2016] [Indexed: 11/16/2022]
|
52
|
Amedei A, Niccolai E, Prisco D. Pancreatic cancer: role of the immune system in cancer progression and vaccine-based immunotherapy. Hum Vaccin Immunother 2015; 10:3354-68. [PMID: 25483688 DOI: 10.4161/hv.34392] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pancreatic cancer (PC) is the 5th leading cause of cancer related death in the developed world with more than 260,000 deaths annually worldwide and with a dismal 5-year survival. Surgery is the only potential hope of cure for PC, but, unfortunately, only 20% PC patients is resectable at the time of diagnosis. Therapeutic research efforts have mainly focused on improvements in radio/ chemo treatments and to date, there are only a few chemotherapeutic agents that have shown to be effective against PC, including gemcitabine with or without abraxane as well as a combination of 5-FU, leucovorin, oxaliplatin and irinotecan (the so-called FOLFIRINOX regimen). The survival of patients treated with these regimens is marginal and hence we are in urgent need of novel therapeutic approaches to treat pancreatic cancer. The success of immunotherapeutic strategies in other cancers and various evidences that pancreatic adenocarcinoma elicits antitumor immune responses, suggest that immunotherapies can be a promising alternative treatment modality for this deadly disease. PC immunotherapy treatments include passive immunotherapeutic approaches, such as the use of effector cells generated in vitro, and active immunotherapeutic strategies, which goal is to stimulate an antitumor response in vivo, by means of vaccination. In this review, we describe the immune suppressive mechanisms of pancreatic cancer and discuss recent preclinical and clinical efforts toward PC immunotherapy, including passive approaches, such as the use of antibodies and active strategies (vaccination), with a special mention of most recent treatment with CRS-207 and GVAX.
Collapse
Key Words
- APC, Antigen Presenting Cells
- CEA, carcinoembryonic antigen
- CTL, Cytotoxic CD8 T cells
- DCs, Dendritic Cells
- ENO1, a-Enolasi
- IDO, Indoleamine 2,3-dioxygenase
- MUC1, Mucin-1
- NK, Natural Killer
- PC, pancreatic cancer
- Th, T helper
- Tregs, Regulatory T cells
- clinical trials
- immune response
- immunotherapy
- mAbs, monoclonal antibodies
- pancreatic cancer
- vaccine
Collapse
Affiliation(s)
- Amedeo Amedei
- a Department of Experimental and Clinical Internal Medicine ; University of Florence ; Florence , Italy
| | | | | |
Collapse
|
53
|
Qi Q, Geng Y, Sun M, Wang P, Chen Z. Clinical implications of systemic inflammatory response markers as independent prognostic factors for advanced pancreatic cancer. Pancreatology 2015; 15:145-50. [PMID: 25641673 DOI: 10.1016/j.pan.2014.12.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 11/11/2014] [Accepted: 12/23/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND Cancer-associated inflammation is a key molecular feature of pancreatic ductal adenocarcinoma. In this study, we systematically evaluated the prognostic relevance of systemic inflammatory response (SIR) markers in patients with advanced pancreatic cancer. METHODS A total of 321 consecutive patients with pathologically-confirmed locally advanced or metastatic pancreatic adenocarcinoma were retrospectively recruited. The patients were divided into a test set (n = 110) and a validation set (n = 211). The associations between overall survival (OS) and clinically available SIR markers including white blood cell (WBC) count, absolute neutrophil count, absolute lymphocyte count, absolute monocyte count, platelet count, neutrophil-lymphocyte ratio (NLR), platelet/lymphocyte ratio (PLR) and lymphocyte/monocyte ratio (LMR) were analyzed using Kaplan-Meier curves and multivariate Cox proportional models. RESULTS High WBC count, neutrophil count, monocyte count, NLR, PLR and low LMR were significantly associated with decreased OS in the test set. Using the validation set for confirmation, we found also in multivariate analysis an independent value of WBC count (hazard ratio (HR): 2.176, 95% confidence interval (CI): 1.560-3.035, P < 0.001), neutrophil count (HR: 2.807, 95% CI: 2.000-3.940, P < 0.001), monocyte count (HR: 1.848, 95% CI: 1.315-2.598, P < 0.001), NLR (HR: 2.204, 95% CI: 1.590-3.055, P < 0.001), PLR (HR: 1.537, 95% CI: 1.114-2.122, P = 0.009) and LMR (HR: 0.569, 95% CI: 0.412-0.784, P = 0.001) for OS in patients with advanced pancreatic cancer. CONCLUSIONS Our study confirmed that SIR markers can be used to determine optimal therapeutic strategies for individual patients and to predict pancreatic cancer prognosis.
Collapse
Affiliation(s)
- Q Qi
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong An Road, Shanghai 200032, China
| | - Y Geng
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong An Road, Shanghai 200032, China
| | - M Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai 200032, China
| | - P Wang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong An Road, Shanghai 200032, China.
| | - Z Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, 270 Dong An Road, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 130 Dong An Road, Shanghai 200032, China.
| |
Collapse
|
54
|
Protti MP, De Monte L, Monte LD, Di Lullo G, Lullo GD. Tumor antigen-specific CD4+ T cells in cancer immunity: from antigen identification to tumor prognosis and development of therapeutic strategies. ACTA ACUST UNITED AC 2014; 83:237-46. [PMID: 24641502 DOI: 10.1111/tan.12329] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Indexed: 12/22/2022]
Abstract
CD4(+) T cells comprise a large fraction of tumor infiltrating lymphocytes and it is now established that they may exert an important role in tumor immune-surveillance. Several CD4(+) T cell subsets [i.e. T helper (Th)1, Th2, T regulatory (Treg), Th17, Th22 and follicular T helper (Tfh)] have been described and differentiation of each subset depends on both the antigen presenting cells responsible for its activation and the cytokine environment present at the site of priming. Tumor antigen-specific CD4(+) T cells with different functional activity have been found in the blood of cancer patients and different CD4(+) T cell subsets have been identified at the tumor site by the expression of specific transcription factors and the profile of secreted cytokines. Importantly, depending on the subset, CD4(+) T cells may exert antitumor versus pro-tumor functions. Here we review the studies that first identified the presence of tumor-specific CD4(+) T cells in cancer patients, the techniques used to identify the tumor antigens recognized, the role of the different CD4(+) T cell subsets in tumor immunity and in cancer prognosis and the development of therapeutic strategies aimed at activating efficient antitumor CD4(+) T cell effectors.
Collapse
Affiliation(s)
- M P Protti
- Tumor Immunology Unit, San Raffaele Scientific Institute, Milan, Italy; Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | |
Collapse
|
55
|
Xu YF, Lu Y, Cheng H, Shi S, Xu J, Long J, Liu L, Liu C, Yu X. Abnormal distribution of peripheral lymphocyte subsets induced by PDAC modulates overall survival. Pancreatology 2014; 14:295-301. [PMID: 25062880 DOI: 10.1016/j.pan.2014.05.797] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 04/29/2014] [Accepted: 05/31/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND/OBJECTIVES The impairment of the immune system is prevalent in patients with malignancies, including pancreatic ductal adenocarcinoma (PDAC). The present study aimed to evaluate alternations of peripheral lymphocyte subsets in patients with PDAC, and also to assess the prognostic value of observed changes. METHODS We recruited 160 consecutive PDAC patients who had undergone radical surgical resection between 2010 and 2013. To investigate the prognostic factors, we detected the peripheral lymphocyte subsets in PDAC by flow cytometry, including T cells (CD3(+), CD3(+)CD4(+), CD3(+)CD8(+), CD8(+)CD28(+)), regulatory T cells (Tregs, CD4(+)CD25(+)CD127(-)), natural killer cells (NK cells, CD3(-)CD56(+)) and B cells (CD19(+)). We also evaluated the clinical and pathological features of these patients. Survival analysis was performed by univariate and multivariate analyses. RESULTS Our results indicated the profile of peripheral lymphocyte subsets undergone profound changes in PDAC patients. Univariate and multivariate analysis indicated the levels of peripheral lymphocyte subsets (CD19(+) B cells, Tregs and CD8(+)CD28(+) T cells) were independent predictors for overall survival. The results also suggested that the systemic impairment of immune system in patients with PDAC, was reversed when primary tumor was removed. CONCLUSIONS The present study provided some evidences that the impairment of host immunity induced by PDAC may play a role in the survival of patients.
Collapse
Affiliation(s)
- Yong-Feng Xu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 DongAn Road, Shanghai 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China; Pancreatic Cancer Institute, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China
| | - Yu Lu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 DongAn Road, Shanghai 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China; Pancreatic Cancer Institute, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China
| | - He Cheng
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 DongAn Road, Shanghai 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China; Pancreatic Cancer Institute, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China
| | - Si Shi
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 DongAn Road, Shanghai 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China; Pancreatic Cancer Institute, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China
| | - Jin Xu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 DongAn Road, Shanghai 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China; Pancreatic Cancer Institute, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China
| | - Jiang Long
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 DongAn Road, Shanghai 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China; Pancreatic Cancer Institute, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China
| | - Liang Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 DongAn Road, Shanghai 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China; Pancreatic Cancer Institute, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China
| | - Chen Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 DongAn Road, Shanghai 200032, PR China; Pancreatic Cancer Institute, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China.
| | - Xianjun Yu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, No. 270 DongAn Road, Shanghai 200032, PR China; Department of Oncology, Shanghai Medical College, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China; Pancreatic Cancer Institute, Fudan University, No. 270 DongAn Road, Shanghai 200032, PR China.
| |
Collapse
|
56
|
Wachsmann MB, Pop LM, Vitetta ES. Pancreatic ductal adenocarcinoma: a review of immunologic aspects. J Investig Med 2014. [PMID: 22406516 DOI: 10.231/jim.0b013e31824a4d79] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
With the continued failures of both early diagnosis and treatment options for pancreatic cancer, it is now time to comprehensively evaluate the role of the immune system on the development and progression of pancreatic cancer. It is important to develop strategies that harness the molecules and cells of the immune system to treat this disease. This review will focus primarily on the role of immune cells in the development and progression of pancreatic ductal adenocarcinoma and to evaluate what is known about the interaction of immune cells with the tumor microenvironment and their role in tumor growth and metastasis. We will conclude with a brief discussion of therapy for pancreatic cancer and the potential role for immunotherapy. We hypothesize that the role of the immune system in tumor development and progression is tissue specific. Our hope is that better understanding of this process will lead to better treatments for this devastating disease.
Collapse
Affiliation(s)
- Megan B Wachsmann
- Masters Program in Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | |
Collapse
|
57
|
Abstract
Effective antitumor immunity requires the generation and persistence of functional tumor-specific T-cell responses. Among the critical factors that often control these responses is how the antigen is delivered and presented to T cells. The use of peptide-based vaccination has been found to be a promising means to induce antitumor T-cell responses but with limited effects even if the peptide is co-delivered with a potent adjuvant. This limited response could be due to cancer-induced dysfunction in dendritic cells (DC), which play a central role in shaping the quantity and quality of antitumor immunity. Therefore, DC-based peptide delivery of tumor antigen is becoming a potential approach in cancer immunotherapy. In this approach, autologous DC are generated from their precursors in bone marrow or peripheral blood mononuclear cells, loaded with tumor antigen(s) and then infused back to the tumor-bearing host in about 7 days. This DC-based vaccination can act as an antigen delivery vehicle as well as a potent adjuvant, resulting in measurable antitumor immunity in several cancer settings in preclinical and clinical studies. This chapter focuses on DC-based vaccination and how this approach can be more efficacious in cancer immunotherapy.Effective antitumor immunity requires the generation and persistence of functional tumor-specific T-cell responses. Among the critical factors that often control these responses is how the antigen is delivered and presented to T cells. The use of peptide-based vaccination has been found to be a promising means to induce antitumor T-cell responses but with limited effects even if the peptide is co-delivered with a potent adjuvant. This limited response could be due to cancer-induced dysfunction in dendritic cells (DC), which play a central role in shaping the quantity and quality of antitumor immunity. Therefore, DC-based peptide delivery of tumor antigen is becoming a potential approach in cancer immunotherapy. In this approach, autologous DC are generated from their precursors in bone marrow or peripheral blood mononuclear cells, loaded with tumor antigen(s) and then infused back to the tumor-bearing host in about 7 days. This DC-based vaccination can act as an antigen delivery vehicle as well as a potent adjuvant, resulting in measurable antitumor immunity in several cancer settings in preclinical and clinical studies. This chapter focuses on DC-based vaccination and how this approach can be more efficacious in cancer immunotherapy.
Collapse
Affiliation(s)
- Mohamed L Salem
- Immunology and Biotechnology Unit, Zoology Department, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
58
|
Sideras K, Braat H, Kwekkeboom J, van Eijck CH, Peppelenbosch MP, Sleijfer S, Bruno M. Role of the immune system in pancreatic cancer progression and immune modulating treatment strategies. Cancer Treat Rev 2013; 40:513-22. [PMID: 24315741 DOI: 10.1016/j.ctrv.2013.11.005] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 11/08/2013] [Accepted: 11/12/2013] [Indexed: 12/11/2022]
Abstract
Traditional chemotherapeutics have largely failed to date to produce significant improvements in pancreatic cancer survival. One of the reasons for the resilience of pancreatic cancer towards intensive treatment is that the cancer is capable of high jacking the immune system: during disease progression the immune system is converted from a system that attacks tumor cells into a support structure for the cancer, exerting trophic actions on the cancer cells. This turn-around of immune system action is achieved through mobilization and activation of regulatory T cells, myeloid derived suppressor cells, tumor-associated macrophages and fibroblasts, all of which suppress CD8 T cells and NK cells. This immune suppression occurs both through the expression of tolerance-inducing cell surface molecules, such as PD-L1, as well as through the production of "tolerogenic" cytokines, such as IL-10 and TGF-β. Based on the accumulating insight into the importance of the immune system for the outcome of pancreatic cancer patients multiple new immunotherapeutic approaches against pancreatic cancer are being currently tested in clinical trials. In this review we give an overview of both the immune escaping mechanisms of pancreatic cancer as well as the new immune related therapeutic strategies currently being tested in pancreatic cancer clinical trials.
Collapse
Affiliation(s)
- K Sideras
- Erasmus University Medical Center, Department of Gastroenterology and Hepatology, NA-0621's, Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - H Braat
- Erasmus University Medical Center, Department of Gastroenterology and Hepatology, Hs-510's, Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - J Kwekkeboom
- Erasmus University Medical Center, Laboratory of Gastroenterology and Hepatology, NA-1009's, Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - C H van Eijck
- Erasmus University Medical Center, Department of Surgery, Room H-818k's Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - M P Peppelenbosch
- Erasmus University Medical Center, Laboratory of Gastroenterology and Hepatology, Na-1007's, Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - S Sleijfer
- Erasmus University Medical Center, Department of Oncology, He-116's Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - M Bruno
- Erasmus University Medical Center, Department of Gastroenterology and Hepatology, H-358's, Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| |
Collapse
|
59
|
Potent CD4+ T-cell epitope P30 enhances HER2/neu-engineered dendritic cell-induced immunity against Tg1-1 breast cancer in transgenic FVBneuN mice by enhanced CD4+ T-cell-stimulated CTL responses. Cancer Gene Ther 2013; 20:590-8. [PMID: 24052129 DOI: 10.1038/cgt.2013.60] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 08/13/2013] [Accepted: 08/18/2013] [Indexed: 12/29/2022]
Abstract
One of the major obstacles in human epidermal growth factor receptor (HER)-2/neu-specific trastuzumab immunotherapy of HER2/neu-positive breast cancer is the development of trastuzumab resistance, warranting the search for other therapeutic strategies. Although dendritic cell (DC) vaccines have been extensively applied in clinical trials for cancer treatment, the vaccination efficacy is still limited, mostly because DC vaccines are not sufficient to break tumor-associated antigen-specific self-immune tolerance in cancer patients. P30 (FNNFTVSFWLRVPKVSASHLE) derived from tetanus toxin is a universally potent CD4(+) T helper epitope capable of enhancing CD8(+) cytotoxic T-lymphocyte (CTL) responses. In this study, we constructed two recombinant adenoviral vectors (AdVs), AdVOVA-P30 and AdVHER2/neu-P30, expressing ovalbumin (OVA)-P30 and HER2/neu-P30. In order to enhance DC vaccine efficacy, we transfected mouse bone marrow (BM)-derived DCs with AdVOVA-P30 and AdVHER2/neu-P30 to generate engineered DCOVA-P30 and DCHER2/neu-P30 vaccines, respectively. We, then, compared CD4(+) and CD8(+) T-cell responses and antitumor immunity derived from DCOVA-P30 and DCHER2/neu-P30 vaccination in wild-type C57BL/6 and transgenic FVBneuN mice, respectively. We demonstrate that engineered DCOVA-P30 vaccine stimulates more efficient CD4(+) and CD8(+) T-cell responses than DCOVA in C57BL/6 mice. Interestingly, the increased DCOVA-P30-induced CTL responses are mainly contributed by enhanced CD4(+) T-cell-stimulated CTL proliferation. We show that DCOVA-P30 vaccine also stimulates more efficient therapeutic immunity against OVA-expressing BL6-10OVA melanoma than DCOVA in C57BL/6 mice. In addition, we demonstrate that DCHER2/neu-P30 vaccine stimulates more efficient CD4(+) and CD8(+) T-cell responses and protective immunity against HER2/neu-expressing Tg1-1 breast cancer than DCHER2/neu in transgenic FVBneuN mice with HER2/neu-specific self-immune tolerance. Therefore, the engineered DCHER2/neu-P30 vaccine may provide a new immunotherapy alternative for women with HER2/neu(+) breast cancer, especially for trastuzumab-resistant HER2/neu(+) breast cancer patients.
Collapse
|
60
|
Scurr M, Bloom A, Pembroke T, Srinivasan R, Brown C, Smart K, Bridgeman H, Davies M, Hargest R, Phillips S, Christian A, Hockey T, Gallimore A, Godkin A. Escalating regulation of 5T4-specific IFN-γ + CD4 + T cells distinguishes colorectal cancer patients from healthy controls and provides a target for in vivo therapy. Cancer Immunol Res 2013; 1. [PMID: 24409450 DOI: 10.1158/2326-6066.cir-13-0035] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The relationship between the adaptive CD4+ T cell response and human cancer is unclear. The oncofetal antigen 5T4 is expressed on many human carcinomas, including colorectal cancer (CRC) cells, but has limited expression on normal tissues. We previously identified anti-5T4 CD4+ T cells in a proportion of CRC patients, and we extended this study to examine whether the quality or quantity of the T cell response reflects tumor stage. An overlapping peptide library spanning 5T4 was used as a target to enumerate cognate IFN-γ+CD4+ T-cells (measured as spot forming cells [SFC]/105 cultured T cells) in peripheral blood-derived lymphocytes following a 12-day in vitro culture period comparing patients pre-operatively (n = 27) to healthy controls (n = 17). Robust 5T4-specific T cell responses were present in 100% of healthy donors. There was a steady loss of T cell responses with advancing tumors with a significant negative correlation from stage I to III (P = 0.008). The predictability of the decline meant < 200 SFC/105 was only found in subjects with stage III CRC. The mechanism of loss of T cell response is independent of HLA-DR type or patient age, but does correspond to increases in Foxp3+ regulatory T cells (Tregs). Using low-dose cyclophosphamide to reduce the proportion of Tregs in vivo resulted in increased anti-5T4 T cell responses in CRC patients. The selective loss of 5T4-specific IFN-γ+CD4+ T cell responses implies a link between tumor stage and antitumor Th1 effector function; depleting Tregs can enhance such responses.
Collapse
Affiliation(s)
- Martin Scurr
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Anja Bloom
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Tom Pembroke
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Rohit Srinivasan
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Clare Brown
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Kathryn Smart
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Hayley Bridgeman
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Mike Davies
- Department of Surgery, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff, UK
| | - Rachel Hargest
- Department of Surgery, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff, UK
| | - Simon Phillips
- Department of Surgery, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff, UK
| | - Adam Christian
- Department of Histopathology, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff, UK
| | - Tom Hockey
- Department of Histopathology, Cardiff and Vale University Health Board, University Hospital of Wales, Cardiff, UK
| | - Awen Gallimore
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Andrew Godkin
- Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
61
|
Protti MP, De Monte L. Immune infiltrates as predictive markers of survival in pancreatic cancer patients. Front Physiol 2013; 4:210. [PMID: 23950747 PMCID: PMC3738865 DOI: 10.3389/fphys.2013.00210] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 07/23/2013] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer is a devastating disease with dismal prognosis. The tumor microenvironment is composed by multiple cell types, molecular factors, and extracellular matrix forming a strong desmoplastic reaction, which is a hallmark of the disease. A complex cross-talk between tumor cells and the stroma exists with reciprocal influence that dictates tumor progression and ultimately the clinical outcome. In this context, tumor infiltrating immune cells through secretion of chemokine and cytokines exert an important regulatory role. Here we review the correlation between the immune infiltrates, evaluated on tumor samples of pancreatic cancer patients underwent surgical resection, and disease free and/or overall survival after surgery. Specifically, we focus on tumor infiltrating lymphocytes (TILs), mast cells (MCs) and macrophages that all contribute to a Th2-type inflammatory and immunosuppressive microenvironment. In these patients tumor immune infiltrates not only do not contribute to disease eradication but rather the features of Th2-type inflammation and immunosuppression is significantly associated with more rapid disease progression and reduced survival.
Collapse
Affiliation(s)
- Maria Pia Protti
- Tumor Immunology Unit, Transplantation and Infectious Diseases, San Raffaele Scientific Institute Milan, Italy ; Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute Milan, Italy
| | | |
Collapse
|
62
|
Roshani R, McCarthy F, Hagemann T. Inflammatory cytokines in human pancreatic cancer. Cancer Lett 2013; 345:157-63. [PMID: 23879960 DOI: 10.1016/j.canlet.2013.07.014] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 07/10/2013] [Accepted: 07/15/2013] [Indexed: 12/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal types of cancer with poor prognosis. Despite extensive efforts, the current treatment methods have limited success. Therefore, novel therapeutic approaches are required. The pancreatic tumor microenvironment is rich in growth factors and inflammatory cytokines that support tumor growth, and it is highly immunosuppressive. Up-regulation of cytokine pathways has been shown to modulate PDAC progression and immune evasion; therefore targeting cytokines may have therapeutic benefits. In this review we provide an overview of current understanding of pro- and anti-inflammatory cytokines in pancreatic cancer and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Rozita Roshani
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK.
| | - Fiona McCarthy
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK.
| | - Thorsten Hagemann
- Centre for Cancer and Inflammation, Barts Cancer Institute, Queen Mary, University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
63
|
Wörmann SM, Diakopoulos KN, Lesina M, Algül H. The immune network in pancreatic cancer development and progression. Oncogene 2013; 33:2956-67. [PMID: 23851493 DOI: 10.1038/onc.2013.257] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 04/22/2013] [Accepted: 04/22/2013] [Indexed: 02/07/2023]
Abstract
The presence of stromal desmoplasia is a hallmark of spontaneous pancreatic ductal adenocarcinoma, forming a unique microenvironment that comprises many cell types. Only recently, the immune system has entered the pathophysiology of pancreatic ductal adenocarcinoma development. Tumor cells in the pancreas seem to dysbalance the immune system, thus facilitating spontaneous cancer development. This review will try to assemble all relevant data to demonstrate the implications of the immune network on spontaneous cancer development.
Collapse
Affiliation(s)
- S M Wörmann
- Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - K N Diakopoulos
- Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - M Lesina
- Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - H Algül
- Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
64
|
Zheng L, Xue J, Jaffee EM, Habtezion A. Role of immune cells and immune-based therapies in pancreatitis and pancreatic ductal adenocarcinoma. Gastroenterology 2013; 144:1230-40. [PMID: 23622132 PMCID: PMC3641650 DOI: 10.1053/j.gastro.2012.12.042] [Citation(s) in RCA: 235] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 12/12/2012] [Accepted: 12/21/2013] [Indexed: 12/22/2022]
Abstract
Immune cells are important in the pathogenesis of acute pancreatitis and determine disease severity. Results from cytokine-based clinical trials for acute pancreatitis have been disappointing, so strategies that target and alter the behavior of infiltrating immune cells require consideration. Recurrent acute pancreatitis can progress to chronic pancreatitis, which is a well-described risk factor for pancreatic ductal adenocarcinoma (PDA). However, most patients with chronic pancreatitis do not develop PDA, and most patients with PDA do not have a history of pancreatitis. Interestingly, chronic pancreatitis and PDA tissues have similarities in their desmoplasia and inflammatory infiltrates, indicating overlapping inflammatory responses. Further studies are needed to determine the differences and similarities of these responses, improve our understanding of PDA pathogenesis, and develop specific immune-based therapies. Immune cells in PDA produce immunosuppressive signals that allow tumors to evade the immune response. Unlike single therapeutic agent studies that block immunosuppressive mechanisms, studies of combination therapies that include therapeutic vaccines have provided promising results.
Collapse
Affiliation(s)
- Lei Zheng
- Stanford University School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, California and The Sidney Kimmel Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jing Xue
- Stanford University School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, California and The Sidney Kimmel Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth M. Jaffee
- Stanford University School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, California and The Sidney Kimmel Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aida Habtezion
- Stanford University School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, Stanford, California and The Sidney Kimmel Cancer Center at Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
65
|
Vonderheide RH, Bayne LJ. Inflammatory networks and immune surveillance of pancreatic carcinoma. Curr Opin Immunol 2013; 25:200-5. [PMID: 23422836 DOI: 10.1016/j.coi.2013.01.006] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 01/21/2013] [Accepted: 01/25/2013] [Indexed: 02/07/2023]
Abstract
Cancer-associated inflammation plays an important role in restraining anti-tumor immunity, particularly in pancreatic ductal adenocarcinoma (PDA) for which a massive infiltration of immunosuppressive leukocytes into the tumor stroma is an early and consistent event in oncogenesis. Intratumoral effector T cells are rare. This pathophysiology is in contrast to many other solid tumors for which infiltration of effector T cells is often prominent, associated with improved clinical outcomes, and mechanistically contributes to tumor immunoediting that ultimately can mediate immune escape. In PDA, increasing evidence suggests that the ras oncogene drives an inflammatory program that establishes immune privilege in the tumor microenvironment. Indeed, PDA cells might remain intrinsically sensitive to T cell killing because they have never been exposed to T cell selective pressure in vivo. In support of this hypothesis, recent studies demonstrate that derailing immune suppressive pathways in the PDA microenvironment, such as tumor derived GM-CSF, facilitates T-cell mediated tumor rejection. These findings carry major implications for the development of novel, combination immunotherapies for pancreatic cancer.
Collapse
Affiliation(s)
- Robert H Vonderheide
- 8-121 Smilow Center for Translational Research, 3400 Civic Center Blvd, Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| | | |
Collapse
|
66
|
Shimato S, Maier LM, Maier R, Bruce JN, Anderson RCE, Anderson DE. Profound tumor-specific Th2 bias in patients with malignant glioma. BMC Cancer 2012. [PMID: 23186108 PMCID: PMC3537750 DOI: 10.1186/1471-2407-12-561] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Background Vaccination against tumor-associated antigens is one promising approach to immunotherapy against malignant gliomas. While previous vaccine efforts have focused exclusively on HLA class I-restricted peptides, class II-restricted peptides are necessary to induce CD4+ helper T cells and sustain effective anti-tumor immunity. In this report we investigated the ability of five candidate peptide epitopes derived from glioma-associated antigens MAGE and IL-13 receptor α2 to detect and characterize CD4+ helper T cell responses in the peripheral blood of patients with malignant gliomas. Methods Primary T cell responses were determined by stimulating freshly isolated PBMCs from patients with primary glioblastoma (GBM) (n = 8), recurrent GBM (n = 5), meningioma (n = 7), and healthy controls (n = 6) with each candidate peptide, as well as anti-CD3 monoclonal antibody (mAb) and an immunodominant peptide epitope derived from myelin basic protein (MBP) serving as positive and negative controls, respectively. ELISA was used to measure IFN-γ and IL-5 levels, and the ratio of IFN-γ/IL-5 was used to determine whether the response had a predominant Th1 or Th2 bias. Results We demonstrate that novel HLA Class-II restricted MAGE-A3 and IL-13Rα2 peptides can detect T cell responses in patients with GBMs as well as in healthy subjects. Stimulation with a variety of peptide antigens over-expressed by gliomas is associated with a profound reduction in the IFN-γ/IL-5 ratio in GBM patients relative to healthy subjects. This bias is more pronounced in patients with recurrent GBMs. Conclusions Therapeutic vaccine strategies to shift tumor antigen-specific T cell response to a more immunostimulatory Th1 bias may be needed for immunotherapeutic trials to be more successful clinically.
Collapse
Affiliation(s)
- Shinji Shimato
- Department of Neurosurgery, Gabriele Bartoli Brain Tumor Research Laboratory, Columbia University, New York City, NY, USA
| | | | | | | | | | | |
Collapse
|
67
|
Kubuschok B, Pfreundschuh M, Breit R, Hartmann F, Sester M, Gärtner B, König J, Murawski N, Held G, Zwick C, Neumann F. Mutated Ras-transfected, EBV-transformed lymphoblastoid cell lines as a model tumor vaccine for boosting T-cell responses against pancreatic cancer: a pilot trial. Hum Gene Ther 2012; 23:1224-36. [PMID: 22966960 DOI: 10.1089/hum.2011.153] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Genetically modified lymphoblastoid cell lines (LCL) have been shown to be an attractive alternative source of antigen-presenting cells for cancer vaccination in vitro. We tested their application in patients with pancreatic cancer in a phase I clinical trial. As a model tumor antigen, we selected the point-mutated (codon 12) Ki-Ras p21 oncogene (muRas) frequently (∼85%) present in pancreatic adenocarcinoma. Autologous LCLs were established in vitro by spontaneous outgrowth from peripheral blood lymphocytes of seven pancreatic carcinoma patients and were genetically modified with an episomal Epstein-Barr virus (EBV)-based expression vector to express muRas (muRas-LCL). Weekly vaccinations with subcutaneous injection of 5×10(6) muRas-LCL were done. In six of seven patients, therapeutic vaccination elicited a T-cell response with an increase in the frequency of muRas-specific precursor cytotoxic T lymphocytes in the peripheral blood and positive delayed-type hypersensitivity reactions at the injection site. Besides local reactions and flu-like symptoms, there were no signs of toxicity and no acute EBV infection, onset of EBV-associated lymphoma, or other severe complications. A clinical response (stable disease) was observed for a short time period (2-4 months) in four of seven patients (57%), mostly in earlier tumor stages. Our results indicate that LCL presenting genetically modified antigen represent a valuable and easily available tool for in vivo autologous tumor vaccination. LCL can be transfected with any known tumor antigen and therefore should be further clinically investigated.
Collapse
Affiliation(s)
- Boris Kubuschok
- Department of Internal Medicine I, Jose-Carreras Centrum for Immuno- and Gene Therapy, University of Saarland Medical School, D-66421 Homburg/Saar, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Di Lullo G, Ieva F, Longhi R, Paganoni AM, Protti MP. Estimating point and interval frequency of antigen-specific CD4+ T cells based on short in vitro expansion and improved poisson distribution analysis. PLoS One 2012; 7:e42340. [PMID: 22879946 PMCID: PMC3413706 DOI: 10.1371/journal.pone.0042340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 07/03/2012] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Knowledge of antigen-specific CD4(+) T cells frequencies is pivotal to the choice of the antigen to be used in anti-viral and anti-tumor vaccination procedures and for monitoring of immune responses. Methods that employ small cell numbers from patient samples, are easy to perform and do not require complex techniques/instrumentations and therefore standardization are desirable. METHODOLOGY/PRINCIPAL FINDINGS Purified blood CD4(+) T cells from healthy donors were cultured with autologous antigen presenting cells in several replicate wells in equal numbers in the absence (un-stimulated wells) or in the presence of synthetic peptides corresponding to viral antigens promiscuous HLA-DR epitopes (antigen-stimulated wells). At day 7 of culture low dose IL-2 was added and at day 14 IFN-γ and IL-5 release in the supernatant was measured. A statistical analysis approach, based on Poisson distribution, was then implemented to calculate the frequency of viral-specific CD4(+) T cells. We first determined a patient-specific exceptionality threshold of cytokine release in the un-stimulated wells and then, based on this threshold, we counted the inactive/active wells within the antigen-stimulated wells. This number, along with the number of cells per well, allowed the point and interval estimates of frequencies. A ready-to-use Excel worksheet template with automatic calculations for frequencies estimate was developed and is provided as a supplemental file (Table S9). CONCLUSIONS/SIGNIFICANCE We report a simple experimental procedure combining short term in vitro cell culture with statistical analysis to calculate the frequency of antigen-specific CD4(+) T cells. The detailed experimental procedure along with the Excel applicative are a valuable tool for monitoring immune responses in the clinical practice.
Collapse
Affiliation(s)
- Giulia Di Lullo
- Tumor Immunology Unit, San Raffaele Scientific Institute, Milan, Italy
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Ieva
- Laboratorty for Modeling and Scientific Computing (MOX), Dipartimento di Matematica, Politecnico di Milano, Milan, Italy
| | - Renato Longhi
- Consiglio Nazionale delle Ricerche (CNR), Istituto di Chimica del Riconoscimento Molecolare, Milan, Italy
| | - Anna Maria Paganoni
- Laboratorty for Modeling and Scientific Computing (MOX), Dipartimento di Matematica, Politecnico di Milano, Milan, Italy
| | - Maria Pia Protti
- Tumor Immunology Unit, San Raffaele Scientific Institute, Milan, Italy
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
- * E-mail:
| |
Collapse
|
69
|
Evans A, Costello E. The role of inflammatory cells in fostering pancreatic cancer cell growth and invasion. Front Physiol 2012; 3:270. [PMID: 22969725 PMCID: PMC3431795 DOI: 10.3389/fphys.2012.00270] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 06/26/2012] [Indexed: 12/11/2022] Open
Abstract
The pancreatic ductal adenocarcinoma (PDAC) microenvironment accommodates a variety of cell types and a plethora of complex interactions between tumor cells, host cells and extracellular matrix (ECM) components. Here we review the role of inflammatory cells, in particular mast cells, myeloid-derived suppressor cells, macrophages, T regulatory cells, T helper cells and neutrophils. The picture that emerges is that of a tumor microenvironment, in which the immune response is actively suppressed, and inflammatory cells contribute in a variety of ways to tumor progression.
Collapse
Affiliation(s)
- Anthony Evans
- Liverpool Cancer Research UK Centre, University of Liverpool Liverpool, UK
| | | |
Collapse
|
70
|
Abstract
With the continued failures of both early diagnosis and treatment options for pancreatic cancer, it is now time to comprehensively evaluate the role of the immune system on the development and progression of pancreatic cancer. It is important to develop strategies that harness the molecules and cells of the immune system to treat this disease. This review will focus primarily on the role of immune cells in the development and progression of pancreatic ductal adenocarcinoma and to evaluate what is known about the interaction of immune cells with the tumor microenvironment and their role in tumor growth and metastasis. We will conclude with a brief discussion of therapy for pancreatic cancer and the potential role for immunotherapy. We hypothesize that the role of the immune system in tumor development and progression is tissue specific. Our hope is that better understanding of this process will lead to better treatments for this devastating disease.
Collapse
Affiliation(s)
- Megan B. Wachsmann
- Masters Program in Clinical Sciences, University of Texas Southwestern Medical Center at Dallas, 6000 Harry Hines Blvd, Dallas, Texas 75390-8576, USA
| | - Laurentiu M. Pop
- The Cancer Immunobiology Center, University of Texas Southwestern Medical Center at Dallas, 6000 Harry Hines Blvd, Dallas, Texas 75390-8576, USA
| | - Ellen S. Vitetta
- The Cancer Immunobiology Center, University of Texas Southwestern Medical Center at Dallas, 6000 Harry Hines Blvd, Dallas, Texas 75390-8576, USA
- The Departments of Microbiology and Immunology, University of Texas Southwestern Medical Center at Dallas, 6000 Harry Hines Blvd, Dallas, Texas 75390-8576, USA
| |
Collapse
|
71
|
Fridman WH, Pagès F, Sautès-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer 2012; 12:298-306. [PMID: 22419253 DOI: 10.1038/nrc3245] [Citation(s) in RCA: 3419] [Impact Index Per Article: 284.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tumours grow within an intricate network of epithelial cells, vascular and lymphatic vessels, cytokines and chemokines, and infiltrating immune cells. Different types of infiltrating immune cells have different effects on tumour progression, which can vary according to cancer type. In this Opinion article we discuss how the context-specific nature of infiltrating immune cells can affect the prognosis of patients.
Collapse
Affiliation(s)
- Wolf Herman Fridman
- INSERM UMRS872, Laboratory of Immune microenvironment and tumours, Paris F75006, France
| | | | | | | |
Collapse
|
72
|
Abstract
OBJECTIVES To verify whether the dysregulation of CD4 T cells concurs in worsening the outcome of pancreatic cancer, we compared the effects of pancreatic cancer and other gastrointestinal cancer cell-conditioned media on the (1) proliferation, migration, and differentiation of CD4 T cells and (2) expansion of CD4 memory (CD45RO), naive (CD45RA), activated (CD69), and regulatory (CD25) subsets. METHODS After culture of CD4 T cells in control, pancreatic (BxPC3, Capan1, MiaPaCa2), or gastrointestinal cancer (AGS, HepG2, HT29) cell-conditioned media, we evaluated proliferation, migration, interferon γ (IFNγ) production, and CD45RA, CD45RO, CD69, and CD25 membrane expression in control and conditioned CD4 T cells. RESULTS Only pancreatic cancer-conditioned media (1) inhibited CD4 T-cell proliferation (P < 0.001) and migration under human stromal cell-derived factor-α chemotaxis (P < 0.001) and (2) induced CD4 T-cell IFNγ production (P < 0.05) and the expansion of the CD69-positive subset (P < 0.001) with respect to the control, with no changes being found in the CD45RA, CD45RO, and CD25 subsets. CONCLUSIONS The in vitro findings achieved in the present study demonstrate that pancreatic cancer cells inhibit CD4 T-cell proliferation and migration, induce IFNγ production, and favor a CD69 subset expansion, suggesting that CD4 T cells play an important role in pancreatic cancer immune evasion.
Collapse
|
73
|
Inhibition and promotion of tumor growth with adeno-associated virus carcinoembryonic antigen vaccine and Toll-like receptor agonists. Cancer Gene Ther 2011; 18:850-8. [PMID: 21869824 PMCID: PMC4143190 DOI: 10.1038/cgt.2011.54] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Carcinoembryonic antigen (CEA) is a cancer vaccines target. Several feature of recombinant adeno-associated virus (rAAV) are attractive for vaccine applications. Combining other viral vector vaccines with Toll like receptor (TLR) agonists enhances antitumor immunity. Wild-type and CEA transgenic (Tg) mice were immunized with rAAV expressing CEA, the TLR9 agonist, ODN1826, and the TLR7 agonist, imiquimod. Mice were challenged with MC38 colon tumor cells and MC38 cells expressing CEA. rAAV-CEA immunization combined with ODN1826 or imiquimod enhanced CEA-specific T-helper-1 immunity and protected against tumor challenge in wild-type but not in CEA-Tg mice. In contrast, immunization with rAAV-CEA in CEA-Tg mice could abrogate the antitumor effects of ODN1826 and promote tumor growth. Compared to wild-type, CEA-Tg mice were characterized by a greater myeloid suppressor cell and T-helper 2 response to TLR agonists and to syngeneic tumors. Depleting PDCA1+ plasmacytoid dendritic cells and Gr1+ myeloid cells increased anti-CEA immune responses in CEA-Tg mice to rAAV-CEA-ODN1826 immunization; depleting CD25+ T cells did not. There are differences in the response of wild-type and CEA-Tg mice to rAAV-CEA, TLR agonists, and syngeneic tumor. In CEA-Tg mice tumor growth can be promoted with rAAV-CEA and TLR agonists. Dendritic and myeloid cells play a regulatory role.
Collapse
|
74
|
De Monte L, Reni M, Tassi E, Clavenna D, Papa I, Recalde H, Braga M, Di Carlo V, Doglioni C, Protti MP. Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. ACTA ACUST UNITED AC 2011; 208:469-78. [PMID: 21339327 PMCID: PMC3058573 DOI: 10.1084/jem.20101876] [Citation(s) in RCA: 530] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Expression of TSLP in pancreatic cancer correlates with Th2 deviation of antitumor immunity that is associated with decrease of patient survival. Pancreatic cancer is a very aggressive disease characterized by a marked desmoplasia with a predominant Th2 (GATA-3+) over Th1 (T-bet+) lymphoid infiltrate. We found that the ratio of GATA-3+/T-bet+ tumor-infiltrating lymphoid cells is an independent predictive marker of patient survival. Patients surgically treated for stage IB/III disease with a ratio inferior to the median value had a statistically significant prolonged overall survival, implying an active role for Th2 responses in disease progression. Thymic stromal lymphopoietin (TSLP), which favors Th2 cell polarization through myeloid dendritic cell (DC) conditioning, was secreted by cancer-associated fibroblasts (CAFs) after activation with tumor-derived tumor necrosis factor α and interleukin 1β. TSLP-containing supernatants from activated CAFs induced in vitro myeloid DCs to up-regulate the TSLP receptor (TSLPR), secrete Th2-attracting chemokines, and acquire TSLP-dependent Th2-polarizing capability in vitro. In vivo, Th2 chemoattractants were expressed in the tumor and in the stroma, and TSLPR-expressing DCs were present in the tumor stroma and in tumor-draining but not in nondraining lymph nodes. Collectively, this study identifies in pancreatic cancer a cross talk between tumor cells and CAFs, resulting in a TSLP-dependent induction of Th2-type inflammation which associates with reduced patient survival. Thus, blocking TSLP production by CAFs might help to improve prognosis in pancreatic cancer.
Collapse
Affiliation(s)
- Lucia De Monte
- Tumor Immunology Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
de Souza APD, de Jesus Borges T, Pillat MM, Bonorino C. CD4+ T cell response against a non-tumor antigen is unaffected in melanoma-bearing mice. Cancer Immunol Immunother 2011; 60:145-51. [PMID: 20963412 PMCID: PMC11029038 DOI: 10.1007/s00262-010-0922-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Accepted: 09/28/2010] [Indexed: 01/01/2023]
Abstract
The tumor microenvironment is complex and creates an immunosuppressive network to tolerize tumor-specific immune responses; however, little information is available regarding the response against non-tumor antigens in tumor-bearing individuals. The goal of the present study was to evaluate if tumor burden could influence a CD4(+) T cell response against a soluble protein, not expressed by the tumor, in the absence of in vitro stimulation. Using an experimental system in which we can compare CD4(+) T cell responses to the Ea antigen when it is either expressed by B16F10 melanoma cells (B16EaRFP cells) or is an exogenous, non-tumor antigen (soluble EaRFP protein), in immunizations of B16F10 tumor-bearing mice, we observed that the tumor can modulate the CD4(+) T cell-specific response to the antigen when it is expressed by the tumor cells. TEa cells proliferated poorly and produced less IFN-γ in mice bearing B16F10 melanoma expressing Ea peptide, and tumor growth was impervious to this response. However, in mice bearing 7 days B16F10 tumors, not expressing the Ea antigen, priming of TEa cells was similar to that observed in tumor-free mice, based on the total number of cells recovered and proliferation assessed by CFSE dilution after EaRFP immunization. We also investigated if tumor burden could influence recall responses of already differentiated effector cells. We immunized mice with EaRFP antigen and after a few days injected B16F10 cells. After 10 days of tumor growth, we challenged the mice with the non-tumor antigen. We found that the number of TEa cells producing IFN-γ in tumor-bearing mice was not different compared to tumor-free mice. No differences in antigen presentation, assessed by YAe antibody staining, were verified in the draining lymph node of these two groups. Collectively, our data indicate that tumor burden does not affect immune responses to non-tumor antigens. These results have important implications in the design of anti-cancer therapy.
Collapse
Affiliation(s)
- Ana Paula Duarte de Souza
- Departamento de Biologia Celular e Molecular (FABIO) and Instituto de Pesquisas Biomédicas, PUCRS, Av. Ipiranga, 6690 2o andar, Porto Alegre, RS 90610-000 Brazil
| | - Thiago de Jesus Borges
- Departamento de Biologia Celular e Molecular (FABIO) and Instituto de Pesquisas Biomédicas, PUCRS, Av. Ipiranga, 6690 2o andar, Porto Alegre, RS 90610-000 Brazil
| | - Micheli M. Pillat
- Departamento de Biologia Celular e Molecular (FABIO) and Instituto de Pesquisas Biomédicas, PUCRS, Av. Ipiranga, 6690 2o andar, Porto Alegre, RS 90610-000 Brazil
| | - Cristina Bonorino
- Departamento de Biologia Celular e Molecular (FABIO) and Instituto de Pesquisas Biomédicas, PUCRS, Av. Ipiranga, 6690 2o andar, Porto Alegre, RS 90610-000 Brazil
| |
Collapse
|
76
|
Kudela P, Sun Z, Fourcade J, Janjic B, Kirkwood JM, Maillere B, Zarour HM. Epitope hierarchy of spontaneous CD4+ T cell responses to LAGE-1. THE JOURNAL OF IMMUNOLOGY 2010; 186:312-22. [PMID: 21131422 DOI: 10.4049/jimmunol.1001989] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
NY-ESO-1 and LAGE-1 represent highly homologous cancer-germline Ags frequently coexpressed by many human cancers, but not by normal tissues, except testis. In contrast to NY-ESO-1, little is known about spontaneous immune responses to LAGE-1. In the current study, we report on spontaneous LAGE-1-specific CD4(+) T cells isolated from PBLs of patients with advanced LAGE-1(+)/NY-ESO-1(+) melanoma and directed against three promiscuous and immunodominant epitopes. Strikingly, although the three LAGE-1-derived epitopes are highly homologous to NY-ESO-1-derived epitopes, LAGE-1-specific CD4(+) T cells did not cross-react with NY-ESO-1. LAGE-1-specific CD4(+) T cells produced Th1-type and/or Th2-type cytokines and did not exert inhibitory effects on allogenic T cells. We observed that most patients with spontaneous NY-ESO-1-specific responses exhibited spontaneous CD4(+) T cell responses to at least one of the three immunodominant LAGE-1 epitopes. Additionally, nearly half of the patients with spontaneous LAGE-1-specific CD4(+) T cell responses had circulating LAGE-1-specific Abs that recognized epitopes located in the C-terminal portion of LAGE-1, which is distinct from NY-ESO-1. Collectively, our findings define the hierarchy of immunodominance of spontaneous LAGE-1-specific CD4(+) T cell responses in patients with advanced melanoma. These findings demonstrate the capability of LAGE-1 to stimulate integrated cellular and humoral immune responses that do not cross-react with NY-ESO-1. Therefore, they provide a strong rationale for the inclusion of LAGE-1 peptides or protein in vaccine trials for patients with NY-ESO-1(+)/LAGE-1(+) tumors.
Collapse
Affiliation(s)
- Pavol Kudela
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | |
Collapse
|
77
|
Patra CR, Bhattacharya R, Mukhopadhyay D, Mukherjee P. Fabrication of gold nanoparticles for targeted therapy in pancreatic cancer. Adv Drug Deliv Rev 2010; 62:346-61. [PMID: 19914317 PMCID: PMC2827658 DOI: 10.1016/j.addr.2009.11.007] [Citation(s) in RCA: 238] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2009] [Accepted: 10/17/2009] [Indexed: 12/18/2022]
Abstract
The targeted delivery of a drug should result in enhanced therapeutic efficacy with low to minimal side effects. This is a widely accepted concept, but limited in application due to lack of available technologies and process of validation. Biomedical nanotechnology can play an important role in this respect. Biomedical nanotechnology is a burgeoning field with myriads of opportunities and possibilities for advancing medical science and disease treatment. Cancer nanotechnology (1-100 nm size range) is expected to change the very foundations of cancer treatment, diagnosis and detection. Nanomaterials, especially gold nanoparticles (AuNPs) have unique physico-chemical properties, such as ultra small size, large surface area to mass ratio, and high surface reactivity, presence of surface plasmon resonance (SPR) bands, biocompatibility and ease of surface functionalization. In this review, we will discuss how the unique physico-chemical properties of gold nanoparticles may be utilized for targeted drug delivery in pancreatic cancer leading to increased efficacy of traditional chemotherapeutics.
Collapse
Affiliation(s)
- Chitta Ranjan Patra
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
78
|
de Souza AP, Bonorino C. Tumor immunosuppressive environment: effects on tumor-specific and nontumor antigen immune responses. Expert Rev Anticancer Ther 2009; 9:1317-32. [PMID: 19761435 DOI: 10.1586/era.09.88] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The interactions between cancer cells and host immune cells in tumoral microenvironments create an immunosuppressive network that promotes tumor growth, protects the tumor from immune attack and attenuates the efficacy of immunotherapeutic approaches. The development of immune tolerance becomes predominant in the immune system of patients with advanced-stage tumors. Several mechanisms have been described by which tumors can suppress the immune system, including secretion of cytokines, alterations in antigen-presenting cell subsets, costimulatory and coinhibitory molecule alterations and altered ratios of Tregs to effector T cells. It is well demonstrated that these mechanisms of immunosuppression can impair tumor specific immune responses. However, it is not well established whether this immunosuppressive environment can affect immune responses to nontumor antigens, specifically in regard to priming and the development of memory. The few existing studies indicate that responses to nontumor antigens seem unaffected, although there is still a deep lack of understanding of this phenomenon. This is an important issue regarding patient endurance and quality of life. Here, we review the existing evidence on immunosuppression promoted by tumors, with particular attention to its impact on specific immune responses. Understanding these interactions can help us subvert tumor-induced tolerance and optimize anti-tumor therapy.
Collapse
Affiliation(s)
- Ana Paula de Souza
- Departamento de Biologia Celular e Molecular (FABIO) and Instituto de Pesquisas Biomédicas, PUCRS. Av. Ipiranga, 6690 2o andar; 90610-90000 Porto Alegre, RS, Brazil.
| | | |
Collapse
|
79
|
Tassi E, Braga M, Longhi R, Gavazzi F, Parmiani G, Di Carlo V, Protti MP. Non-redundant role for IL-12 and IL-27 in modulating Th2 polarization of carcinoembryonic antigen specific CD4 T cells from pancreatic cancer patients. PLoS One 2009; 4:e7234. [PMID: 19798410 PMCID: PMC2749205 DOI: 10.1371/journal.pone.0007234] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Accepted: 09/09/2009] [Indexed: 01/23/2023] Open
Abstract
Background Pancreatic cancer is a very aggressive disease with dismal prognosis; peculiar is the tumor microenvironment characterized by an extensive fibrotic stroma, which favors rapid tumor progression. We previously reported that pancreatic cancer patients have a selective Th2 skew in the anti-carcinoembryonic antigen (CEA) CD4+ T cell immunity, which correlates with the presence of a predominant GATA-3+ tumor lymphoid infiltrate. This has negative effects in both effective anti-tumor immunity and further favoring fibrinogenesis. Aim of this study was to evaluate whether the Th2 polarization of CEA-specific CD4+ T cells from pancreatic cancer patients is stable or can be reverted by immunomodulating cytokines. Methodology/Principal Findings We first evaluated the influence of IL-12 and IL-27, as single agents and in association, on the polarization of CEA-specific Th2 CD4+ T cell clones from a pancreatic cancer patient. We found that only the combination of IL-12 and IL-27 modified the polarization of Th2 effectors by both reduction of IL-5, GM-CSF and IL-13 and induction of IFN-γ production, which lasted after cytokine removal. Second, we evaluated the effect of the combined treatment on polyclonal CEA-specific CD4+ T cells in short-time re-stimulation assays. In agreement with the data obtained with the clones, we found that the combined treatment functionally modulated the Th2 polarization of CEA-specific CD4+ T cells and enhanced pre-existing Th1 type immunity. Conclusions/Significance Collectively, our results demonstrate that tumor antigen specific Th2 CD4+ T cells in pancreatic cancer are endowed with functional plasticity. Hence, loco-regional cytokines delivery or targeted therapy based on antibodies or molecules directed to the tumor stroma might improve anti-tumor immunity and ameliorate fibrosis, without systemic toxicity.
Collapse
Affiliation(s)
- Elena Tassi
- Tumor Immunology Unit, San Raffaele Scientific Institute, Milan, Italy
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Marco Braga
- Pancreas Unit, San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Renato Longhi
- CNR-Istituto di Chimica del Riconoscimento Molecolare, Milan, Italy
| | - Francesca Gavazzi
- Pancreas Unit, San Raffaele Scientific Institute, Milan, Italy
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Giorgio Parmiani
- Immuno-Biotherapy of Melanoma and Solid Tumors Unit, San Raffaele Scientific Institute, Milan, Italy
- Division of Molecular Oncology, San Raffaele Scientific Institute, Milan, Italy
| | - Valerio Di Carlo
- Pancreas Unit, San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Maria Pia Protti
- Tumor Immunology Unit, San Raffaele Scientific Institute, Milan, Italy
- Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
- * E-mail:
| |
Collapse
|
80
|
Karyampudi L, Krco CJ, Kalli KR, Erskine CL, Hartmann LC, Goodman K, Ingle JN, Maurer MJ, Nassar A, Yu C, Disis ML, Wettstein PJ, Fikes JD, Beebe M, Ishioka G, Knutson KL. Identification of a broad coverage HLA-DR degenerate epitope pool derived from carcinoembryonic antigen. Cancer Immunol Immunother 2009; 59:161-71. [PMID: 19621224 DOI: 10.1007/s00262-009-0738-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Accepted: 07/02/2009] [Indexed: 12/19/2022]
Abstract
CD4 T cells are important for anti-tumor immune responses. Aside from their role in the activation of CD8 T cells, CD4 T cells also mediate anti-tumor immune responses by recruiting innate immune effectors into the tumor microenvironment. Thus, the search for strategies to boost CD4 T cell immunity is an active area of research. Our goal in this study was to identify HLA-DR epitopes of carcinoembryonic antigen (CEA), a commonly over-expressed tumor antigen. HLA-DR epitopes of CEA were identified using the epitope prediction program, PIC (predicted IC(50)) and tested using in vitro HLA-DR binding assays. Following CEA epitope confirmation, IFN-gamma ELIspot assays were used to detect existing immunity against the HLA-DR epitope panel of CEA in breast and ovarian cancer patients. In vitro generated peptide-specific CD4 T cells were used to determine whether the epitopes are naturally processed from CEA protein. Forty-three epitopes of CEA were predicted, 15 of which had high binding affinity for 8 or more common HLA-DR molecules. A degenerate pool of four, HLA-DR restricted 15 amino acid epitopes (CEA.24, CEA.176/354, CEA.488, and CEA.653) consisting of two novel epitopes (CEA.24 and CEA.488) was identified against which 40% of breast and ovarian cancer patients had pre-existent T cell immunity. All four epitopes are naturally processed by antigen-presenting cells. Hardy-Weinberg analysis showed that the pool is useful in approximately 94% of patients. Patients with breast or ovarian cancer demonstrate pre-existent immune responses to the tumor antigen CEA. The degenerate pool of CEA peptides may be useful for augmenting CD4 T cell immunity.
Collapse
Affiliation(s)
- Lavakumar Karyampudi
- Department of Immunology, College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|