51
|
Polimeno M, Napolitano M, Costantini S, Portella L, Esposito A, Capone F, Guerriero E, Trotta A, Zanotta S, Pucci L, Longo N, Perdonà S, Pignata S, Castello G, Scala S. Regulatory T cells, interleukin (IL)-6, IL-8, vascular endothelial growth factor (VEGF), CXCL10, CXCL11, epidermal growth factor (EGF) and hepatocyte growth factor (HGF) as surrogate markers of host immunity in patients with renal cell carcinoma. BJU Int 2013; 112:686-96. [PMID: 23495770 DOI: 10.1111/bju.12068] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To identify a phenotype that could be informative and prognostic in patients with renal cell carcinoma (RCC) peripheral blood was evaluated for TH1, TH2, regulatory T cells (Tregs), natural killer (NK) and NKT cells and for cytokines/chemokines. PATIENTS AND METHODS Peripheral blood from 77 patients with RCC and 40 healthy controls was evaluated by flow cytometry using monoclonal antibodies against CD4, CD25, FoxP3, CD45RA, CD45RO, CD152, CD184, CD279, CD3, CD16, CD56, CD161, CD158a, CD4, CD26, CD30, CD183 and CD184. A concomitant evaluation of 38 molecules was conducted in patients' serum using a multiplex biometric ELISA-based immunoassay. RESULTS The number of NK cells CD3⁻/CD16⁺, CD3⁻/CD16⁺/CD161⁺ (NK) and CD3⁻/CD16⁺/CD161⁺/CD158a⁺ (NK- Kir 2+) was greater in the patients with RCC (P < 0.05); and the number of Treg cells CD4⁺/CD25(high+)/FOXP3⁺ and the subset CD4⁺/CD25(high+)/FOXP3⁺/CD45RA⁺ (naïve) and CD45R0⁺(memory) cells, were greater in the patients with RCC (P < 0.001). An increase in the following was observed in the serum of patients with RCC compared with healthy controls: interleukin (IL)-4, IL-6, IL-8, IL-10, G-CSF, CXCL10, CXCL11, hepatocyte growth factor (HGF) and vascular endothelial growth factor (VEGF). According to Ingenuity Pathway Analysis (IPA), CXCL10, IL-6, IL-8, epidermal growth factor (EGF), HGF and VEGF were associated with a network that controls cellular movement, tissue development and cellular growth. Kaplan-Meier analysis for disease-free survival showed that high numbers of CD4⁺/CD25(high+)/FOXP3⁺/CD45RA⁺ (Treg naïve) and low numbers of CD3⁻/CD16⁺/CD161⁺/CD158a⁺ (NK-Kir+) cells predict short disease-free survival in patients with RCC. CONCLUSION Concomitant evaluation of Treg (CD4⁺/CD25(high+)/FOXP3⁺ and CD4⁺/CD25(high+)/FOXP3⁺/CD45RA⁺) and of six soluble factors (IL-6, IL-8 ,VEGF, CXCL10, CXCL11, EGF, HGF) might be a surrogate marker of host immunity in patients with RCC.
Collapse
Affiliation(s)
- Marianeve Polimeno
- Oncological Immunology, National Cancer Institute 'G. Pascale', Mercogliano, Avellino
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Bedke J, Gouttefangeas C, Singh-Jasuja H, Stevanović S, Behnes CL, Stenzl A. Targeted therapy in renal cell carcinoma: moving from molecular agents to specific immunotherapy. World J Urol 2013; 32:31-8. [PMID: 23404195 PMCID: PMC3901931 DOI: 10.1007/s00345-013-1033-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 01/24/2013] [Indexed: 12/22/2022] Open
Abstract
Non-specific immunotherapy has been for a long time a standard treatment option for patients with metastatic renal cell carcinoma but was redeemed by specific targeted molecular therapies, namely the VEGF and mTOR inhibitors. After moving treatment for mRCC to specific molecular agents with a well-defined mode of action, immunotherapy still needs this further development to increase its accuracy. Nowadays, an evolution from a rather non-specific cytokine treatment to sophisticated targeted approaches in specific immunotherapy led to a re-launch of immunotherapy in clinical studies. Recent steps in the development of immunotherapy strategies are discussed in this review with a special focus on peptide vaccination which aims at a tumor targeting by specific T lymphocytes. In addition, different combinatory strategies with immunomodulating agents like cyclophosphamide or sunitinib are outlined, and the effects of immune checkpoint modulators as anti-CTLA-4 or PD-1 antibodies are discussed.
Collapse
Affiliation(s)
- Jens Bedke
- Department of Urology, Eberhard Karls University Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany,
| | | | | | | | | | | |
Collapse
|
53
|
Kwak M, Mu L, Lu Y, Chen JJ, Brower K, Fan R. Single-cell protein secretomic signatures as potential correlates to tumor cell lineage evolution and cell-cell interaction. Front Oncol 2013; 3:10. [PMID: 23390614 PMCID: PMC3565185 DOI: 10.3389/fonc.2013.00010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 01/11/2013] [Indexed: 12/20/2022] Open
Abstract
Secreted proteins including cytokines, chemokines, and growth factors represent important functional regulators mediating a range of cellular behavior and cell-cell paracrine/autocrine signaling, e.g., in the immunological system (Rothenberg, 2007), tumor microenvironment (Hanahan and Weinberg, 2011), or stem cell niche (Gnecchi etal., 2008). Detection of these proteins is of great value not only in basic cell biology but also for diagnosis and therapeutic monitoring of human diseases such as cancer. However, due to co-production of multiple effector proteins from a single cell, referred to as polyfunctionality, it is biologically informative to measure a panel of secreted proteins, or secretomic signature, at the level of single cells. Recent evidence further indicates that a genetically identical cell population can give rise to diverse phenotypic differences (Niepel etal., 2009). Non-genetic heterogeneity is also emerging as a potential barrier to accurate monitoring of cellular immunity and effective pharmacological therapies (Cohen etal., 2008; Gascoigne and Taylor, 2008), but can hardly assessed using conventional approaches that do not examine cellular phenotype at the functional level. It is known that cytokines, for example, in the immune system define the effector functions and lineage differentiation of immune cells. In this article, we hypothesize that protein secretion profile may represent a universal measure to identify the definitive correlate in the larger context of cellular functions to dissect cellular heterogeneity and evolutionary lineage relationship in human cancer.
Collapse
Affiliation(s)
- Minsuk Kwak
- Department of Biomedical Engineering, Yale UniversityNew Haven, CT, USA
| | - Luye Mu
- Department of Electrical Engineering, Yale UniversityNew Haven, CT, USA
| | - Yao Lu
- Department of Biomedical Engineering, Yale UniversityNew Haven, CT, USA
| | - Jonathan J. Chen
- Department of Biomedical Engineering, Yale UniversityNew Haven, CT, USA
| | - Kara Brower
- Department of Biomedical Engineering, Yale UniversityNew Haven, CT, USA
- Isoplexis Inc.New Haven, CT, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale UniversityNew Haven, CT, USA
- Yale Comprehensive Cancer CenterNew Haven, CT, USA
| |
Collapse
|
54
|
Hadrup S, Donia M, Thor Straten P. Effector CD4 and CD8 T cells and their role in the tumor microenvironment. CANCER MICROENVIRONMENT 2012; 6:123-33. [PMID: 23242673 PMCID: PMC3717059 DOI: 10.1007/s12307-012-0127-6] [Citation(s) in RCA: 231] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 11/25/2012] [Indexed: 12/11/2022]
Abstract
T cells in tumors—the so-called tumor infiltrating lymphocytes (TIL) have been studied intensively over the past years. Compelling evidence point to a clinical relevance for high numbers of T cells at the tumor site with CD8 memory T cells as a key denominator for overall survival (OS) in patients with colo-rectal cancer (CRC), and also for others solid cancers. These data goes hand in hand with studies of clonality of TIL showing the T cells among TIL are expanded clonally, and also that tumor specific T cells of CD4 as well as CD8 type are enriched at the tumor site. The tumor microenvironment is hostile to T cell function e.g., due to expression of enzymes that depletes the amino acids tryptophan and arginine, high concentration of tumor secreted lactate, and presence innate cells or regulatory T cells both with suppressive activity. Analyses of the specificity of TILs in melanoma demonstrate that quite few known antigens are in fact recognized by these cultures underscoring patient unique and/or mutated antigens may represent important target for recognition.
Collapse
Affiliation(s)
- Sine Hadrup
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Herlev University Hospital, 2730, Herlev, Denmark
| | | | | |
Collapse
|
55
|
Wang QJ, Hanada KI, Robbins PF, Li YF, Yang JC. Distinctive features of the differentiated phenotype and infiltration of tumor-reactive lymphocytes in clear cell renal cell carcinoma. Cancer Res 2012; 72:6119-29. [PMID: 23071066 PMCID: PMC3513562 DOI: 10.1158/0008-5472.can-12-0588] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Clear cell renal cell carcinoma (RCC) is considered an immunogenic tumor, but it has been difficult to identify tumor-infiltrating lymphocytes (TIL) that show in vitro tumor recognition. We compared the characteristics of fresh RCC TIL to peripheral blood lymphocytes (PBL) or melanoma TIL. Our results showed that RCC TIL contained fewer CD27(+) T cells, and fewer naïve and central memory (CM) T cells, but more effector memory (EM) T cells than melanoma TIL or renal PBL. We hypothesized that factors in the RCC microenvironment were skewing TIL phenotype toward EM. One possibility was the expression of CD70 on nearly all human RCCs, but not melanomas. Differentiation of naïve T cells to EM cells only occurred from CD70 costimulation in concert with T-cell receptor (TCR) stimulation (signal one), suggesting that EM TIL responding to CD70 would be enriched for T cells reactive with local antigens, including those associated with RCC. Clonotypic analysis of TCRs in fresh RCCs showed that EM T cells were more clonally expanded than CM or naïve T cells, and the clonal expansion occurred at the tumor site as oligoclonal TCRs were distinct from PBL TCRs from the same patient. In addition, we found that 2 TCRs from the highly represented EM TIL clones, when reexpressed in fresh PBL, recognized an MHC-class II or MHC-class I-restricted antigens shared by multiple RCC lines. Our results suggest that RCC-reactive TIL do exist in situ, but may be difficult to recover and study because of proliferative exhaustion, driven by tumor-expressed CD70.
Collapse
Affiliation(s)
- Qiong J Wang
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | | | |
Collapse
|
56
|
Lin YC, Mahalingam J, Chiang JM, Su PJ, Chu YY, Lai HY, Fang JH, Huang CT, Chiu CT, Lin CY. Activated but not resting regulatory T cells accumulated in tumor microenvironment and correlated with tumor progression in patients with colorectal cancer. Int J Cancer 2012; 132:1341-50. [PMID: 22907255 DOI: 10.1002/ijc.27784] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2012] [Revised: 06/27/2012] [Accepted: 07/31/2012] [Indexed: 12/28/2022]
Abstract
Activated T regulatory (T(reg)) cells are potent suppressors that mediate immune tolerance. We investigated the relationship between activated T(reg) cells and the progression of human colon cancer. We designed a cross-sectional study of CD4(+) Foxp3(+) T cells from peripheral blood, primary tumor and nontumor colon tissue of 42 patients with colon cancer and correlated the percentages of different subgroups of T(reg) cells with colon cancer stage. The phenotypes, cytokine-release patterns and suppression ability of these T(reg) cells were analyzed. We found that T(reg) cells increased significantly in both peripheral blood and cancer tissue. In addition, the T(reg) cells expressed significantly lower levels of CCR7, CD62L and CD45RA in comparison to normal volunteers. Further dividing T(reg) cells into subgroups based on Foxp3 and CD45RA expression revealed that both activated T(reg) cells (Foxp3(hi) CD45RA(-)) and nonsuppressive T(reg) cells (Foxp3(lo) CD45RA(-)), but not resting T(reg) cells (Foxp3(low) CD45RA(+)), increased in the peripheral blood and cancer tissue of patients with colon cancer. Only the activated T(reg) cells expressed significantly higher levels of tumor necrosis factor receptor 2 and cytotoxic T-cell antigen-4. Activated T(reg) cells, however, secreted significantly lower levels of effector cytokines (interleukin-2, tumor necrosis factor-α and interferon-γ) than did resting T(reg) cells and nonsuppressive cells upon ex vivo stimulation. Activated, but not resting, T(reg) cells in cancer tissue correlated with tumor metastases. In summary, we confirmed that activated T(reg) cells are a distinct subgroup with effector memory phenotype and fully functional regulatory activity against human colorectal cancer immunity.
Collapse
Affiliation(s)
- Yung-Chang Lin
- Department of Hematology-Oncology, Linkou Medical Center, Chang Gung Memorial Hospital, Kweishan, Tayouan, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Mahalingam J, Lin YC, Chiang JM, Su PJ, Fang JH, Chu YY, Huang CT, Chiu CT, Lin CY. LAP+CD4+ T cells are suppressors accumulated in the tumor sites and associated with the progression of colorectal cancer. Clin Cancer Res 2012; 18:5224-33. [PMID: 22879386 DOI: 10.1158/1078-0432.ccr-12-0211] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
PURPOSE Suppressor T cells are one of the determinants of colorectal cancer (CRC) clinical outcome. LAP(+)CD4(+) T cell is a recently identified subset of suppressor T cells. This study was designed to investigate their clinical relevance in patients with CRC. EXPERIMENTAL DESIGN Sixty patients with CRC and 24 healthy donors (HD) were enrolled in this study. The percentages of LAP(+)CD4(+) T cells in peripheral blood and tumor tissue were measured. The phenotype and functional relevance of LAP(+)CD4(+) T cells were analyzed subsequently. RESULTS The percentages of LAP(+)CD4(+) T cells in peripheral blood of patients with CRC were significantly higher than HD (HD vs. CRC: 3.1% ± 0.78% vs. 8.8% ± 5.8%, P < 0.0001) and in tumor tissue when compared with nontumor tissue (nontumor vs. tumor: 3.2% ± 1.1% vs. 9.5% ± 5.5%, P = 0.0002). In addition, LAP(+)CD4(+) T cells with effector memory (EM) phenotype were more likely to accumulate in the tumor sites than in peripheral blood. These LAP(+)CD4(+) T cells produced significantly higher levels of IFN-γ, IL-17 and comparatively lower IL-2 and very few IL-10. LAP(+)CD4(+) T cells could suppress the proliferation of LAP(-)CD4(+) T cells that were partially mediated by TGF-β. Furthermore, these LAP(+)CD4(+) T cells accumulated in tumor site and increased further in the peripheral blood in patients with metastasis. CONCLUSIONS LAP(+)CD4(+) T cells as a suppressor subset could accumulate in the tumor microenvironment and circulated more in the peripheral blood with tumor progression in patients with CRC.
Collapse
Affiliation(s)
- Jayashri Mahalingam
- Department of Gastroenterology-Hepatology, Chang Gung University, Kweishan, Tayouan, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Chaves KCB, Peron JPS, Chammas R, Turaça LT, Pesquero JB, Braga MS, Foguer K, Schor N, Bellini MH. Endostatin gene therapy stimulates upregulation of ICAM-1 and VCAM-1 in a metastatic renal cell carcinoma model. Cancer Gene Ther 2012; 19:558-65. [PMID: 22699868 DOI: 10.1038/cgt.2012.32] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
One of the greatest challenges in urological oncology is renal cell carcinoma (RCC), which is the third leading cause of death in genitourinary cancers. RCCs are highly vascularized and respond positively to antiangiogenic therapy. Endostatin (ES) is a fragment of collagen XVIII that possesses antiangiogenic activity. In this study, we examined the potential of ES-based antiangiogenic therapy to activate tumor-associated endothelial cells in metastatic RCC (mRCC). Balb/c-bearing Renca cells were treated with NIH/3T3-LendSN or, as a control, with NIH/3T3-LXSN cells. The T-cell subsets and lymphocyte populations of tumors, mediastinal lymph nodes and the spleen were assessed by flow cytometry. The expression of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) was assessed by real-time PCR, flow cytometry and immunohistochemistry analysis. ES gene therapy led to an increase in the percentage of infiltrating CD4-interferon (IFN)-γ cells (P<0.05), CD8-IFN-γ cells (P<0.01) and CD49b-tumor necrosis factor-α cells (P<0.01). In addition, ES therapy caused an increase at the mRNA level of ICAM-1 (1.4-fold; P<0.01) and VCAM-1 (1.5-fold) (control vs treated group; P<0.001). Through flow cytometry, we found a significant increase in the CD34/ICAM-1 cells (8.1-fold; P<0.001) and CD34/VCAM-1 cells (1.6-fold; P<0.05). ES gene therapy induced a significant increase in both T CD4 and CD8 cells in the lymph nodes and the spleen, suggesting that ES therapy may facilitate cell survival or clonal expansion. CD49b cells were also present in increased quantities in all of these organs. In this study, we demonstrate an antitumor inflammatory effect of ES in an mRCC model, and this effect is mediated by an increase in ICAM-1 and VCAM-1 expression in tumor-associated endothelial cells.
Collapse
Affiliation(s)
- K C B Chaves
- Department of Medicine, Nephrology Division, Federal University of São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Prinz PU, Mendler AN, Masouris I, Durner L, Oberneder R, Noessner E. High DGK-α and Disabled MAPK Pathways Cause Dysfunction of Human Tumor-Infiltrating CD8+ T Cells That Is Reversible by Pharmacologic Intervention. THE JOURNAL OF IMMUNOLOGY 2012; 188:5990-6000. [DOI: 10.4049/jimmunol.1103028] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
60
|
Oldham KA, Parsonage G, Bhatt RI, Wallace DMA, Deshmukh N, Chaudhri S, Adams DH, Lee SP. T lymphocyte recruitment into renal cell carcinoma tissue: a role for chemokine receptors CXCR3, CXCR6, CCR5, and CCR6. Eur Urol 2012; 61:385-94. [PMID: 22079021 DOI: 10.1016/j.eururo.2011.10.035] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Accepted: 10/26/2011] [Indexed: 01/13/2023]
Abstract
BACKGROUND Evidence suggests that some patients with renal cell carcinoma (RCC) respond to immunomodulatory therapies that activate T lymphocytes. A prerequisite for effective T cell therapy is efficient targeting of effector T cells to the tumour site, yet the molecular basis of T cell recruitment to RCC is unknown. Furthermore, some T cells that naturally infiltrate this cancer are regulatory T cells (Tregs) that may suppress antitumour immune responses. OBJECTIVE Determine the mechanisms of effector and regulatory T cell recruitment to RCC to allow targeted therapy that promotes local anti-tumour immunity. DESIGN, SETTING, AND PARTICIPANTS Tumour-infiltrating and peripheral blood T cells were collected from 70 patients undergoing nephrectomy for RCC. MEASUREMENTS T cells were analysed by multicolour flow cytometry for expression of 19 chemokine receptors and 7 adhesion molecules. Receptors that were expressed at higher levels on tumour-infiltrating lymphocytes (TILs) compared with matched peripheral blood lymphocytes (PBLs) were analysed further for their ability to mediate migration responses in TILs and for expression of corresponding ligands in tumour tissue. RESULTS AND LIMITATIONS Three chemokine receptors-CCR5, CXCR3, and CXCR6-were significantly overexpressed on TILs compared with matched PBLs (n=16 cases) and were capable of promoting migration in vitro. Their corresponding ligands CCL4-5, CXCL9-11, and CXCL16 were all detected in RCC tissue. However, since they were present in all cases studied, it was not possible to correlate ligand expression with levels of T cell infiltration. Foxp3(+) Tregs were enriched within TILs compared with matched PBLs and expressed high levels of CCR5, CXCR3, and CXCR6, as well as CCR6, the ligand for which (CCL20) was detectable in RCC tissue. CONCLUSIONS Our data support a role for CCR5, CXCR3, and CXCR6 in the selective recruitment of T cells into RCC tissue and, together with CCR6, in the recruitment of Tregs.
Collapse
Affiliation(s)
- Kimberley A Oldham
- School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | | | | | | | | | | | | | | |
Collapse
|
61
|
Sasada T, Suekane S. Variation of tumor-infiltrating lymphocytes in human cancers: controversy on clinical significance. Immunotherapy 2012; 3:1235-51. [PMID: 21995574 DOI: 10.2217/imt.11.106] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tumors develop and progress under the influence of a microenvironment comprising a variety of immune cell subsets and their products. Recent studies have shown that tumor-infiltrating lymphocytes (TILs) are not randomly distributed, but organized to accumulate more or less densely in different regions within tumors, and interact with each other. Substantial evidence has suggested that not only CD8(+) and/or CD4(+) αβ T cells but also other lymphocyte subsets, including γδ T cells, B cells, NK cells, and NKT cells, infiltrate tumor tissues in variable quantities and play a key role in the regulation of antitumor immunity. In this article, we summarize available information regarding the diversity and composition of TILs, which may positively or negatively affect tumor growth and patient clinical outcomes. The clinical significance of TILs in human cancers remains unclear and is a subject of considerable controversy; largely due to the lack of functional data for TILs, as well as due to enormous variability of TILs in different tumors. A great deal more functional data about TILs needs to be obtained for individual tumors before TILs can be considered as a prognostic parameter in human cancers.
Collapse
Affiliation(s)
- Tetsuro Sasada
- Department of Immunology & Immunotherapy, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka 830-0011, Japan.
| | | |
Collapse
|
62
|
The CD3-zeta chimeric antigen receptor overcomes TCR Hypo-responsiveness of human terminal late-stage T cells. PLoS One 2012; 7:e30713. [PMID: 22292024 PMCID: PMC3264628 DOI: 10.1371/journal.pone.0030713] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Accepted: 12/20/2011] [Indexed: 12/17/2022] Open
Abstract
Adoptive therapy of malignant diseases with tumor-specific cytotoxic T cells showed remarkable efficacy in recent trials. Repetitive T cell receptor (TCR) engagement of target antigen, however, inevitably ends up in hypo-responsive cells with terminally differentiated KLRG-1+ CD57+ CD7− phenotype limiting their therapeutic efficacy. We here revealed that hypo-responsiveness of CMV-specific late-stage CD8+ T cells is due to reduced TCR synapse formation compared to younger cells. Membrane anchoring of TCR components contributes to T cell hypo-responsiveness since dislocation of galectin-3 from the synapse by swainsonine restored both TCR synapse formation and T cell response. Transgenic expression of a CD3-zeta signaling chimeric antigen receptor (CAR) recovered hypo-responsive T cells to full effector functions indicating that the defect is restricted to TCR membrane components while synapse formation of the transgenic CAR was not blocked. CAR engineered late-stage T cells released cytokines and mediated redirected cytotoxicity as efficiently as younger effector T cells. Our data provide a rationale for TCR independent, CAR mediated activation in the adoptive cell therapy to avoid hypo-responsiveness of late-stage T cells upon repetitive antigen encounter.
Collapse
|
63
|
Daurkin I, Eruslanov E, Stoffs T, Perrin GQ, Algood C, Gilbert SM, Rosser CJ, Su LM, Vieweg J, Kusmartsev S. Tumor-associated macrophages mediate immunosuppression in the renal cancer microenvironment by activating the 15-lipoxygenase-2 pathway. Cancer Res 2011; 71:6400-9. [PMID: 21900394 DOI: 10.1158/0008-5472.can-11-1261] [Citation(s) in RCA: 166] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Renal cell carcinoma (RCC), the most common human kidney cancer, is frequently infiltrated with tumor-associated macrophages (TAM) that can promote malignant progression. Here, we show that TAMs isolated from human RCC produce substantial amounts of the proinflammatory chemokine CCL2 and immunosuppressive cytokine IL-10, in addition to enhanced eicosanoid production via an activated 15-lipoxygenase-2 (15-LOX2) pathway. TAMs isolated from RCC tumors had a high 15-LOX2 expression and secreted substantial amounts of 15(S)-hydroxyeicosatetraenoic acid, its major bioactive lipid product. Inhibition of lipoxygenase activity significantly reduced production of CCL2 and IL-10 by RCC TAMs. In addition, TAMs isolated from RCC were capable of inducing in T lymphocytes, the pivotal T regulatory cell transcription factor forkhead box P3 (FOXP3), and the inhibitory cytotoxic T-lymphocyte antigen 4 (CTLA-4) coreceptor. However, this TAM-mediated induction of FOXP3 and CTLA-4 in T cells was independent of lipoxygenase and could not be reversed by inhibiting lipoxygenase activity. Collectively, our results show that TAMs, often present in RCCs, display enhanced 15-LOX2 activity that contributes to RCC-related inflammation, immunosuppression, and malignant progression. Furthermore, we show that TAMs mediate the development of immune tolerance through both 15-LOX2-dependent and 15-LOX2-independent pathways. We propose that manipulating LOX-dependent arachidonic acid metabolism in the tumor microenvironment could offer new strategies to block cancer-related inflammation and immune escape in patients with RCC.
Collapse
Affiliation(s)
- Irina Daurkin
- Department of Urology, University of Florida, Gainesville, Florida, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Klein O, Schmidt C, Knights A, Davis ID, Chen W, Cebon J. Melanoma vaccines: developments over the past 10 years. Expert Rev Vaccines 2011; 10:853-73. [PMID: 21692705 DOI: 10.1586/erv.11.74] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Decades of preclinical evaluation and clinical trials into melanoma vaccines have yielded spectacular progress in our understanding of melanoma antigens and the immune mechanisms of tumor rejection. Key insights and the results of their clinical evaluation are reviewed in this article. Unfortunately, durable clinical benefit following vaccination remains uncommon. Two recent clinical advances that will impact on melanoma vaccine development are trials with inhibitors of CTLA-4 and oncogenic BRAF. Long-term therapeutic control of melanoma will require integration of specific active immunotherapy with these emerging successful therapies from the disparate fields of immune regulation and signal transduction.
Collapse
Affiliation(s)
- Oliver Klein
- Ludwig Institute for Cancer Research, Austin Branch, Austin Hospital, Studley Road, Heidelberg, Victoria, 3084, Australia
| | | | | | | | | | | |
Collapse
|
65
|
Widenmeyer M, Griesemann H, Stevanović S, Feyerabend S, Klein R, Attig S, Hennenlotter J, Wernet D, Kuprash DV, Sazykin AY, Pascolo S, Stenzl A, Gouttefangeas C, Rammensee HG. Promiscuous survivin peptide induces robust CD4+ T-cell responses in the majority of vaccinated cancer patients. Int J Cancer 2011; 131:140-9. [PMID: 21858810 DOI: 10.1002/ijc.26365] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 07/21/2011] [Indexed: 12/22/2022]
Abstract
CD4(+) T cells have been shown to be crucial for the induction and maintenance of cytotoxic T cell responses and to be also capable of mediating direct tumor rejection. Therefore, the anticancer therapeutic efficacy of peptide-based vaccines may be improved by addition of HLA class II epitopes to stimulate T helper cells. Survivin is an apoptosis inhibiting protein frequently overexpressed in tumors. Here we describe the first immunological evaluation of a survivin-derived CD4(+) T cell epitope in a multipeptide immunotherapy trial for prostate carcinoma patients. The survivin peptide is promiscuously presented by several human HLA-DRB1 molecules and, most importantly, is naturally processed by dendritic cells. In vaccinated patients, it was able to induce frequent, robust and multifunctional CD4(+) T cell responses, as monitored by IFN-γ ELISPOT and intracellular cytokine staining. Thus, this HLA-DR restricted epitope is broadly immunogenic and should be valuable for stimulating T helper cells in patients suffering from a wide range of tumors.
Collapse
Affiliation(s)
- Melanie Widenmeyer
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, Tübingen 72076, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Wang J, Cai D, Ma B, Wu G, Wu J. Skewing the Balance of Regulatory T-Cells and T-Helper 17 Cells in Breast Cancer Patients. J Int Med Res 2011; 39:691-701. [PMID: 21819700 DOI: 10.1177/147323001103900301] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
This study investigated the distribution of interleukin (IL)-17-producing CD4+ T-cells (T-helper [Th17] cells) in relation to CD4+CD25+CD127− cells (regulatory T-cells [Treg]) in tumour-infiltrating lymphocytes (TILs) and peripheral blood mononuclear cells (PBMCs) from breast cancer patients. The Th17 and Treg cells were evaluated by flow cytometry and reported as a percentage of total CD4+ cells. In TILs from early breast cancer patients ( n = 12), the frequency of Th17 cells was significantly higher than in PBMCs (14.5 ± 7.2% versus 6.9 ± 2.1%). In TILs from patients with advanced breast cancer ( n = 15), the frequency of Th17 cells was also significantly higher than that in PBMCs (9.1 ± 5.7% versus 3.2 ± 2.3%) but lower compared with early disease. The Th17/Treg ratio in TILs was markedly increased in early versus advanced disease. In conclusion, Th17 and Treg cell accumulation in the tumour microenvironment of breast cancer occurred in early disease; Th17 cell infiltration gradually decreased and Treg cells accumulated with disease progression.
Collapse
Affiliation(s)
- J Wang
- Department of Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - D Cai
- Department of Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - B Ma
- Department of Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - G Wu
- Department of Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - J Wu
- Department of Surgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
67
|
Ma C, Fan R, Ahmad H, Shi Q, Comin-Anduix B, Chodon T, Koya RC, Liu CC, Kwong GA, Radu CG, Ribas A, Heath JR. A clinical microchip for evaluation of single immune cells reveals high functional heterogeneity in phenotypically similar T cells. Nat Med 2011; 17:738-43. [PMID: 21602800 DOI: 10.1038/nm.2375] [Citation(s) in RCA: 330] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2010] [Accepted: 01/12/2011] [Indexed: 11/09/2022]
Abstract
Cellular immunity has an inherent high level of functional heterogeneity. Capturing the full spectrum of these functions requires analysis of large numbers of effector molecules from single cells. We report a microfluidic platform designed for highly multiplexed (more than ten proteins), reliable, sample-efficient (∼1 × 10(4) cells) and quantitative measurements of secreted proteins from single cells. We validated the platform by assessment of multiple inflammatory cytokines from lipopolysaccharide (LPS)-stimulated human macrophages and comparison to standard immunotechnologies. We applied the platform toward the ex vivo quantification of T cell polyfunctional diversity via the simultaneous measurement of a dozen effector molecules secreted from tumor antigen-specific cytotoxic T lymphocytes (CTLs) that were actively responding to tumor and compared against a cohort of healthy donor controls. We observed profound, yet focused, functional heterogeneity in active tumor antigen-specific CTLs, with the major functional phenotypes quantitatively identified. The platform represents a new and informative tool for immune monitoring and clinical assessment.
Collapse
Affiliation(s)
- Chao Ma
- NanoSystems Biology Cancer Center, California Institute of Technology, Pasadena, California, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Figel AM, Brech D, Prinz PU, Lettenmeyer UK, Eckl J, Turqueti-Neves A, Mysliwietz J, Anz D, Rieth N, Muenchmeier N, Buchner A, Porubsky S, Siegert SI, Segerer S, Nelson PJ, Noessner E. Human renal cell carcinoma induces a dendritic cell subset that uses T-cell crosstalk for tumor-permissive milieu alterations. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:436-51. [PMID: 21703422 DOI: 10.1016/j.ajpath.2011.03.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 03/17/2011] [Accepted: 03/21/2011] [Indexed: 12/20/2022]
Abstract
Tissue dendritic cells (DCs) may influence the progression of renal cell carcinoma (RCC) by regulating the functional capacity of antitumor effector cells. DCs and their interaction with T cells were analyzed in human RCC and control kidney tissues. The frequency of CD209(+) DCs in RCCs was found to be associated with an unfavorable T(H)1 cell balance in the tissue and advanced tumor stages. The CD209(+) DCs in RCC were unusual because most of them co-expressed macrophage markers (CD14, CD163). The phenotype of these enriched-in-renal-carcinoma DCs (ercDCs) could be reiterated in vitro by carcinoma-secreted factors (CXCL8/IL-8, IL-6, and vascular endothelial growth factor). ErcDCs resembled conventional DCs in costimulatory molecule expression and antigen cross-presentation. They did not suppress cognate cytotoxic T-lymphocyte function and did not cause CD3ζ down-regulation, FOXP3 induction, or T-cell apoptosis in situ or in vitro; thus, they are different from classic myeloid-derived suppressor cells. ErcDCs secreted high levels of metalloproteinase 9 and used T-cell crosstalk to increase tumor-promoting tumor necrosis factor α and reduce chemokines relevant for T(H)1-polarized lymphocyte recruitment. This modulation of the tumor environment exerted by ercDCs suggests an immunologic mechanism by which tumor control can fail without involving cytotoxic T-lymphocyte inhibition. Pharmacologic targeting of the deviated DC differentiation could improve the efficacy of immunotherapy against RCC.
Collapse
Affiliation(s)
- Ainhoa-M Figel
- Institute of Molecular Immunology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Singer K, Kastenberger M, Gottfried E, Hammerschmied CG, Büttner M, Aigner M, Seliger B, Walter B, Schlösser H, Hartmann A, Andreesen R, Mackensen A, Kreutz M. Warburg phenotype in renal cell carcinoma: high expression of glucose-transporter 1 (GLUT-1) correlates with low CD8(+) T-cell infiltration in the tumor. Int J Cancer 2011; 128:2085-95. [PMID: 20607826 DOI: 10.1002/ijc.25543] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Many tumor cells are characterized by a dysregulated glucose metabolism associated with increased glycolysis in the presence of oxygen ("Warburg Effect"). Here, we analyzed for the first time a possible link between glucose metabolism and immune cell infiltration in renal cell carcinoma (RCC). RCC specimens revealed a highly significant increase in the expression of lactate dehydrogenase A (LDHA) and glucose-transporter 1 (GLUT-1) compared to the corresponding normal kidney tissue on mRNA level. Accordingly, tumor cell lines of different origin such as RCC, melanoma and hepatocellular carcinoma strongly expressed LDHA and GLUT-1 compared to their nonmalignant counterparts. In line with this finding, tumor cells secreted high amounts of lactate. High expression of GLUT-1 and LDH5, a tetramer of 4 LDHA subunits, was confirmed by tissue microarray analysis of 249 RCC specimens. Overall, 55/79 (69.6%) and 46/71 (64.7%) cases of clear cell carcinoma showed a constitutive, but heterogeneous expression of GLUT-1 and LDH5, respectively. The number of CD3(+), CD8(+) and FOXP3(+) T cells was significantly elevated in RCC lesions compared to normal kidney epithelium, but effector molecules such as granzyme B and perforin were decreased in tumor infiltrating T cells. Of interest, further analysis revealed an inverse correlation between GLUT-1 expression and the number of CD8(+) T cells in RCC lesions. Together, our data suggest that an accelerated glucose metabolism in RCC tissue is associated with a low infiltration of CD8(+) effector T cells. Targeting the glucose metabolism may represent an interesting tool to improve the efficacy of specific immunotherapeutic approaches in RCC.
Collapse
Affiliation(s)
- Katrin Singer
- Department of Internal Medicine 5, Haematology/Oncology, University of Erlangen, Erlangen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Wang SF, Fouquet S, Chapon M, Salmon H, Regnier F, Labroquère K, Badoual C, Damotte D, Validire P, Maubec E, Delongchamps NB, Cazes A, Gibault L, Garcette M, Dieu-Nosjean MC, Zerbib M, Avril MF, Prévost-Blondel A, Randriamampita C, Trautmann A, Bercovici N. Early T cell signalling is reversibly altered in PD-1+ T lymphocytes infiltrating human tumors. PLoS One 2011; 6:e17621. [PMID: 21408177 PMCID: PMC3049782 DOI: 10.1371/journal.pone.0017621] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Accepted: 02/02/2011] [Indexed: 12/18/2022] Open
Abstract
To improve cancer immunotherapy, a better understanding of the weak efficiency of tumor-infiltrating T lymphocytes (TIL) is necessary. We have analyzed the functional state of human TIL immediately after resection of three types of tumors (NSCLC, melanoma and RCC). Several signalling pathways (calcium, phosphorylation of ERK and Akt) and cytokine secretion are affected to different extents in TIL, and show a partial spontaneous recovery within a few hours in culture. The global result is an anergy that is quite distinct from clonal anergy induced in vitro, and closer to adaptive tolerance in mice. PD-1 (programmed death -1) is systematically expressed by TIL and may contribute to their anergy by its mere expression, and not only when it interacts with its ligands PD-L1 or PD-L2, which are not expressed by every tumor. Indeed, the TCR-induced calcium and ERK responses were reduced in peripheral blood T cells transfected with PD-1. Inhibition by sodium stibogluconate of the SHP-1 and SHP-2 phosphatases that associate with several inhibitory receptors including PD-1, relieves part of the anergy apparent in TIL or in PD-1-transfected T cells. This work highlights some of the molecular modifications contributing to functional defects of human TIL.
Collapse
Affiliation(s)
- Shu-Fang Wang
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Stéphane Fouquet
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Maxime Chapon
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Hélène Salmon
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Fabienne Regnier
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Karine Labroquère
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Cécile Badoual
- Inserm U970, Univ Paris Descartes, PARCC, Paris, France
- Service d'Anatomie-Pathologique, Hôpital Européen Georges Pompidou, APHP, Paris, France
| | - Diane Damotte
- Laboratoire Microenvironnement immunitaire et tumeurs, INSERM U872, Centre de Recherche des Cordeliers, Paris, France
- Univ Pierre et Marie Curie, UMR S872, Paris, France
- Univ Paris Descartes, UMR S872, Paris, France
- Service d'Anatomie-Pathologie, Hôpital Hôtel Dieu, AP-HP, Paris, France
| | - Pierre Validire
- Service d'Anatomie-Pathologie, Institut Mutualiste Montsouris, Paris, France
| | - Eve Maubec
- APHP, UnivParis Diderot, Service de Dermatologie, Hôpital Bichat, Paris, France
| | | | - Aurélie Cazes
- Service d'Anatomie-Pathologique, Hôpital Européen Georges Pompidou, APHP, Paris, France
- Inserm U833, Collège de France, Université Paris Descartes, Paris, France
| | - Laure Gibault
- Service d'anatomie et cytologie pathologiques, Groupe Hospitalier Cochin-Saint Vincent de Paul, Univ Paris Descartes, Paris, France
| | - Marylène Garcette
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Marie-Caroline Dieu-Nosjean
- Laboratoire Microenvironnement immunitaire et tumeurs, INSERM U872, Centre de Recherche des Cordeliers, Paris, France
- Univ Pierre et Marie Curie, UMR S872, Paris, France
- Univ Paris Descartes, UMR S872, Paris, France
| | - Marc Zerbib
- APHP, Hôpital Cochin, service d'Urologie, Paris, France
| | - Marie-Françoise Avril
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
- APHP, Hôpital Cochin, Service de Dermatologie, Paris, France
| | - Armelle Prévost-Blondel
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Clotilde Randriamampita
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
| | - Alain Trautmann
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
- * E-mail: (AT); (NB)
| | - Nadège Bercovici
- Inserm, U1016, Institut Cochin, Paris, France
- Cnrs, UMR8104, Paris, France
- Univ Paris Descartes, Paris, France
- * E-mail: (AT); (NB)
| |
Collapse
|
71
|
Singer K, Gottfried E, Kreutz M, Mackensen A. Suppression of T-cell responses by tumor metabolites. Cancer Immunol Immunother 2011; 60:425-31. [PMID: 21240484 PMCID: PMC11029601 DOI: 10.1007/s00262-010-0967-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2010] [Accepted: 12/28/2010] [Indexed: 12/29/2022]
Abstract
Tumor cells have developed multiple mechanisms to escape T-cell-mediated immune recognition. Recent work has revealed that the altered tumor metabolism depletes essential nutrients or leads to the accumulation of immunosuppressive metabolites in the tumor microenvironment. In this review, we discuss the suppressive activity of some metabolic key players, which are upregulated in human tumor cells, including indolamine-2,3-dioxygenase (IDO), arginase, inducible nitric oxide synthetase (iNOS), and lactate dehydrogenase (LDH)-A, on the adaptive immune system. A better understanding of the impact of metabolic alterations of tumor cells on effector T-cell functions could lead to new therapeutic strategies to improve the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Katrin Singer
- Department of Internal Medicine 5, Hematology/Oncology, University of Erlangen, Krankenhausstrasse 12, 91054 Erlangen, Germany
| | - Eva Gottfried
- Department of Hematology and Oncology, University Medical School, Regensburg, Germany
| | - Marina Kreutz
- Department of Hematology and Oncology, University Medical School, Regensburg, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology/Oncology, University of Erlangen, Krankenhausstrasse 12, 91054 Erlangen, Germany
| |
Collapse
|
72
|
Abstract
T-cell therapy involves the ex vivo isolation and expansion of antigen-specific T cells for adoptive transfer. The use of T-cell clones represents one embodiment of this approach and provides a uniform population of effector cells, so that parameters contributing to an effective response can be rigorously evaluated. T cells of defined specificity, phenotype, and function are isolated and expanded; when infused into patients, these intrinsic factors can be considered in light of extrinsic factors such as the type of conditioning regimen, cytokine support, and immunomodulatory reagents. In this chapter, 2 topics related to the use of antigen-specific T-cell clones are discussed: first, advances enabling the isolation and expansion of antigen-specific T-cell clones for human trials of adoptive therapy, and second, a contextual framework of advantages and limitations in which the use of adoptively transferred T-cell clones can be judiciously applied as a means to dissect the requirements for effective therapy.
Collapse
|
73
|
Widenmeyer M, Shebzukhov Y, Haen SP, Schmidt D, Clasen S, Boss A, Kuprash DV, Nedospasov SA, Stenzl A, Aebert H, Wernet D, Stevanović S, Pereira PL, Rammensee HG, Gouttefangeas C. Analysis of tumor antigen-specific T cells and antibodies in cancer patients treated with radiofrequency ablation. Int J Cancer 2010; 128:2653-62. [PMID: 20715115 DOI: 10.1002/ijc.25601] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Accepted: 07/20/2010] [Indexed: 01/22/2023]
Abstract
Radiofrequency (RF) ablation is a minimally invasive technique routinely applied for the treatment of primary and secondary liver tumors. It induces cell death by thermal coagulative necrosis of tumor tissues, whereas cellular metabolism can still take place in a transition zone surrounding the necrotic area. An increase in heat shock protein expression occurs shortly after treatment, suggesting that the induction of activating signals may stimulate the host immune system. In addition, various effects on immune effectors have also been observed, including stimulation of tumor-directed T lymphocytes. Here, we prospectively assessed the activation of tumor antigen-specific antibodies, as well as antigen-specific CD4(+) and CD8(+) T cells in patients suffering from primary or secondary malignancies and treated by RF ablation with or without concomitant chemotherapy. An increase of antibodies (in 4 patients of 49), CD4(+) T cells or CD8(+) T cells (in 2 patients of 49) could be detected several weeks to months following intervention. These findings suggest that in addition to the local control of tumor growth, RF ablation can provide the appropriate conditions for activating tumor-antigen specific immune responses.
Collapse
Affiliation(s)
- Melanie Widenmeyer
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, Auf der Morgenstelle 15, 72076 Tuebingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Johann PD, Vaegler M, Gieseke F, Mang P, Armeanu-Ebinger S, Kluba T, Handgretinger R, Müller I. Tumour stromal cells derived from paediatric malignancies display MSC-like properties and impair NK cell cytotoxicity. BMC Cancer 2010; 10:501. [PMID: 20858262 PMCID: PMC2949810 DOI: 10.1186/1471-2407-10-501] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 09/21/2010] [Indexed: 12/11/2022] Open
Abstract
Background Tumour growth and metastatic infiltration are favoured by several components of the tumour microenvironment. Bone marrow-derived multipotent mesenchymal stromal cells (MSC) are known to contribute to the tumour stroma. When isolated from healthy bone marrow, MSC exert potent antiproliferative effects on immune effector cells. Due to phenotypic and morphological similarities of MSC and tumour stromal cells (TStrC), we speculated that immunotherapeutic approaches may be hampered if TStrC may still exhibit immunomodulatory properties of MSC. Methods In order to compare immunomodulatory properties of MSC and tumour stromal cells (TStrC), we established and analyzed TStrC cultures from eleven paediatric tumours and MSC preparations from bone marrow aspirates. Immunophenotyping, proliferation assays and NK cell cytotoxicity assays were employed to address the issue. Results While TStrC differed from MSC in terms of plasticity, they shared surface expression of CD105, CD73 and other markers used for MSC characterization. Furthermore, TStrC displayed a strong antiproliferative effect on peripheral blood mononuclear cells (PBMC) in coculture experiments similar to MSC. NK cell cytotoxicity was significantly impaired after co-culture with TStrC and expression of the activating NK cell receptors NKp44 and NKp46 was reduced. Conclusions Our data show that TStrC and MSC share important phenotypic and functional characteristics. The inhibitory effect of TStrC on PBMC and especially on NK cells may facilitate the immune evasion of paediatric tumours.
Collapse
Affiliation(s)
- Pascal-David Johann
- Department of General Paediatrics, University Children's Hospital, Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
75
|
|