2301
|
Vujanovic L, Whiteside TL, Potter DM, Chu J, Ferrone S, Butterfield LH. Regulation of antigen presentation machinery in human dendritic cells by recombinant adenovirus. Cancer Immunol Immunother 2008; 58:121-33. [PMID: 18488218 DOI: 10.1007/s00262-008-0533-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Accepted: 05/05/2008] [Indexed: 02/02/2023]
Abstract
Recombinant adenoviral vectors (AdV) are potent vehicles for antigen engineering of dendritic cells (DC). DC engineered with AdV to express full length tumor antigens are capable stimulators of antigen-specific polyclonal CD8+ and CD4+ T cells. To determine the impact of AdV on the HLA class I antigen presentation pathway, we investigated the effects of AdV transduction on antigen processing machinery (APM) components in human DC. Interactions among AdV transduction, maturation, APM regulation and T cell activation were investigated. The phenotype and cytokine profile of DC transduced with AdV was intermediate, between immature (iDC) and matured DC (mDC). Statistically significant increases in expression were observed for peptide transporters TAP-1 and TAP-2, and HLA class I peptide-loading chaperone ERp57, as well as co-stimulatory surface molecule CD86 due to AdV transduction. AdV transduction enhanced the expression of APM components and surface markers on mDC, and these changes were further modulated by the timing of DC maturation. Engineering of matured DC to express a tumor-associated antigen stimulated a broader repertoire of CD8+ T cells, capable of recognizing immunodominant and subdominant epitopes. These data identify molecular changes in AdV-transduced DC (AdV/DC) that could influence T cell priming and should be considered in design of cancer vaccines.
Collapse
Affiliation(s)
- Lazar Vujanovic
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | | | | | | | | | | |
Collapse
|
2302
|
Mignot G, Ullrich E, Bonmort M, Ménard C, Apetoh L, Taieb J, Bosisio D, Sozzani S, Ferrantini M, Schmitz J, Mack M, Ryffel B, Bulfone-Paus S, Zitvogel L, Chaput N. The critical role of IL-15 in the antitumor effects mediated by the combination therapy imatinib and IL-2. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 180:6477-83. [PMID: 18453565 DOI: 10.4049/jimmunol.180.10.6477] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
The synergistic antitumor effects of the combination therapy imatinib mesylate (IM) and IL-2 depended upon NK1.1- expressing cells and were associated with the accumulation of CD11c(int)B220(+)NK1.1(+) IFN-producing killer dendritic cells (IKDC) into tumor beds. In this study, we show that the antitumor efficacy of the combination therapy was compromised in IL-15 and IFN-type 1R loss-of-function mice. IL-15Ralpha was required for the proliferation of IKDC during IM plus IL-2 therapy. Trans-presentation of IL-15/IL-15Ralpha activated IKDC to express CCR2 and to respond to type 1 IFN by producing CCL2. Moreover, the antitumor effects of the combination therapy correlated with a CCL2-dependent recruitment of IKDC, but not B220(-) NK cells, into tumor beds. Altogether, the IL-15-driven peripheral expansion and the CCL-2-dependent intratumoral chemoattraction of IKDC are two critical parameters dictating the antitumor efficacy of IM plus IL-2 in mice.
Collapse
Affiliation(s)
- Grégoire Mignot
- Institut National de la Santé et de la Recherche Médicale, Unité 805, Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2303
|
Panaretakis T, Joza N, Modjtahedi N, Tesniere A, Vitale I, Durchschlag M, Fimia GM, Kepp O, Piacentini M, Froehlich KU, van Endert P, Zitvogel L, Madeo F, Kroemer G. The co-translocation of ERp57 and calreticulin determines the immunogenicity of cell death. Cell Death Differ 2008; 15:1499-509. [PMID: 18464797 DOI: 10.1038/cdd.2008.67] [Citation(s) in RCA: 278] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The exposure of calreticulin (CRT) on the plasma membrane can precede anthracycline-induced apoptosis and is required for cell death to be perceived as immunogenic. Mass spectroscopy, immunofluorescence and immunoprecipitation experiments revealed that CRT co-translocates to the surface with another endoplasmic reticulum-sessile protein, the disulfide isomerase ERp57. The knockout and knockdown of CRT or ERp57 inhibited the anthracycline-induced translocation of ERp57 or CRT, respectively. CRT point mutants that fail to interact with ERp57 were unable to restore ERp57 translocation upon transfection into crt(-/-) cells, underscoring that a direct interaction between CRT and ERp57 is strictly required for their co-translocation to the surface. ERp57(low) tumor cells generated by retroviral introduction of an ERp57-specific shRNA exhibited a normal apoptotic response to anthracyclines in vitro, yet were resistant to anthracycline treatment in vivo. Moreover, ERp57(low) cancer cells (which failed to expose CRT) treated with anthracyclines were unable to elicit an anti-tumor response in conditions in which control cells were highly immunogenic. The failure of ERp57(low) cells to elicit immune responses and to respond to chemotherapy could be overcome by exogenous supply of recombinant CRT protein. These results indicate that tumors that possess an intrinsic defect in the CRT-translocating machinery become resistant to anthracycline chemotherapy due to their incapacity to elicit an anti-cancer immune response.
Collapse
Affiliation(s)
- T Panaretakis
- INSERM, Unit 848 'Apoptosis, Cancer and Immunity', F-94805 Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2304
|
Abstract
Apoptotic cell death is an essential and highly ordered process that contributes to both the development and the homeostasis of multicellular organisms. It is associated with dramatic biochemical and cell biological events within the dying cell, including fragmentation of the nucleus and the redistribution of intracellular proteins and membrane lipids. It has long been apparent that phagocytic clearance of the cell corpse is an integral part of the apoptotic process; apoptotic clearance also may be essential in tissue homeostasis. During the cell death process, apoptotic cells acquire new cell surface determinants for specific recognition by responder phagocytes and suppression of immune responsiveness. Recent studies indicate that these determinants are well conserved throughout metazoan evolution; remarkably, their recognition shows no species-specific restriction. Professional and non-professional phagocytes recognize and respond to apoptotic cells similarly, notably with the immediate-early transcriptional repression of a variety of specific genes including those encoding inflammatory cytokines. Secondary responses following engulfment of the apoptotic corpse, utilizing several distinct mechanisms, enhance and sustain this apoptotic suppression. In this review, we highlight the central role of apoptotic cells in innate homeostatic regulation of immunity.
Collapse
|
2305
|
Gullo CA, Hwang WY, Poh CK, Au M, Cow G, Teoh G. Use of ultraviolet-light irradiated multiple myeloma cells as immunogens to generate tumor-specific cytolytic T lymphocytes. JOURNAL OF IMMUNE BASED THERAPIES AND VACCINES 2008; 6:2. [PMID: 18439316 PMCID: PMC2383894 DOI: 10.1186/1476-8518-6-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2007] [Accepted: 04/28/2008] [Indexed: 12/12/2022]
Abstract
Background As the eradication of tumor cells in vivo is most efficiently performed by cytolytic T lymphocytes (CTL), various methods for priming tumor-reactive lymphocytes have been developed. In this study, a method of priming CTLs with ultraviolet (UV)-irradiated tumor cells, which results in termination of tumor cell proliferation, apoptosis, as well as upregulation of heat shock proteins (HSP) expression is described. Methods Peripheral blood mononuclear cells (PBMC) were primed weekly with UV-irradiated or mitomycin-treated RPMI 8226 multiple myeloma cells. Following three rounds of stimulation over 21 days, the lymphocytes from the mixed culture conditions were analyzed for anti-MM cell reactivity. Results By day 10 of cultures, PBMCs primed using UV-irradiated tumor cells demonstrated a higher percentage of activated CD8+/CD4- T lymphocytes than non-primed PBMCs or PBMCs primed using mitomycin-treated MM cells. Cytotoxicity assays revealed that primed PBMCs were markedly more effective (p < 0.01) than non-primed PBMCs in killing RPMI 8226 MM cells. Surface expression of glucose regulated protein 94 (Grp94/Gp96) and Grp78 were both found to be induced in UV-treated MM cells. Conclusion Since, HSP-associated peptides are known to mediate tumor rejection; these data suggest that immune-mediated eradication of MM cells could be elicited via a UV-induced HSP process. The finding that the addition of 17-allylamide-17-demethoxygeldanamycin (17AAG, an inhibitor of HSP 90-peptide interactions) resulted in decreased CTL-induced cytotoxicity supported this hypothesis. Our study, therefore, provides the framework for the development of anti-tumor CTL cellular vaccines for treating MM using UV-irradiated tumor cells as immunogens.
Collapse
Affiliation(s)
- Charles A Gullo
- Cancer Immunology Laboratory, Department of Clinical Research, Singapore General Hospital, Outram Road, 169608 Singapore.
| | | | | | | | | | | |
Collapse
|
2306
|
Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Mignot G, Ullrich E, Kroemer G, Zitvogel L. Cancer is not just a disease of a tissue: it is a host disease. How to reactivate host defense against tumors using conventional therapies of cancer? ANNALES D'ENDOCRINOLOGIE 2008; 69:151-2. [PMID: 18420177 DOI: 10.1016/j.ando.2008.02.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- L Apetoh
- Institut Gustave-Roussy, 39 rue Camille-Desmoulins, 94805 Villejuif, France
| | | | | | | | | | | | | | | |
Collapse
|
2307
|
Characterization of a single peptide derived from cytochrome P450 1B1 that elicits spontaneous human leukocyte antigen (HLA)-A1 as well as HLA-B35 restricted CD8 T-cell responses in cancer patients. Hum Immunol 2008; 69:266-72. [PMID: 18486761 DOI: 10.1016/j.humimm.2008.02.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Revised: 02/01/2008] [Accepted: 02/08/2008] [Indexed: 11/23/2022]
Abstract
Cytochrome P450 1B1 (CYP1B1) is widely expressed in human malignancies, but silent in most normal tissues. Importantly, the protein is believed to play an important role in the survival and growth of cancer cells in a stressed environment, e.g., as a result of hypoxia or chemotherapy. Thus, targeting of CYP1B1 represents a potentially successful strategy in the treatment of metastatic cancer, e.g., by therapeutic vaccination. Herein, we describe the characterization of a novel peptide from the CYP1B1 protein (CYP240), which is spontaneously recognized by CD8 T cells in cancer patients. Interestingly, the peptide binds to both human leukocyte antigen (HLA)-A1 and HLA-B35. Hence, peripheral blood lymphocytes from a total of 49 cancer patients (25 melanoma, 13 RCC, and 11 breast cancer; 41 HLA-A1 positive, 8 HLA-B35 positive) were analyzed for reactivity taking advantage of the EliSpot assay. Rare but strong responses were detected in HLA-A1-positive patients, and more frequent responses were detected in HLA-B35-positive patients. No reactivity against the peptide could be detected in healthy donors. Furthermore, we demonstrated that peptide-specific T cells were able to lyze target cells presenting the peptide on the surface. The characterized CYP240 peptide presented herein opens the avenue for more broader recruitment of patients in vaccination trials targeting CYB1B1.
Collapse
|
2308
|
Apetoh L, Mignot G, Panaretakis T, Kroemer G, Zitvogel L. Immunogenicity of anthracyclines: moving towards more personalized medicine. Trends Mol Med 2008; 14:141-51. [DOI: 10.1016/j.molmed.2008.02.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Revised: 01/29/2008] [Accepted: 02/01/2008] [Indexed: 12/22/2022]
|
2309
|
Quesnel B. Dormant tumor cells as a therapeutic target? Cancer Lett 2008; 267:10-7. [PMID: 18384938 DOI: 10.1016/j.canlet.2008.02.055] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2007] [Revised: 02/25/2008] [Accepted: 02/25/2008] [Indexed: 12/29/2022]
Abstract
Tumor dormancy is characterised by the persistence of residual tumor cells for long periods. Recurrence from minimal residual disease is a major cause of cancer death. Thus, understanding how cancer cells become and remain dormant, may lead to new strategies to prevent relapse. Evidence has emerged that a balance exists between host and dormant tumor cells. Cross-talk between tumor cells and their micro-environment, angiogenesis, and anti-tumor immune response participate in the control of dormant tumor cells. Tumor cells have several mechanisms of maintaining equilibrium, and immune escape, including expression of immuno-regulatory molecules (e.g., increased expression of B7.1 and B7-H1); epigenetic modifications (e.g., silencing of the SOCS1 gene, de-regulating the JAK/STAT pathway); and autocrine loops. These new findings offer new opportunities to design specific treatments, to modify the balance in favor of the host immune response.
Collapse
Affiliation(s)
- Bruno Quesnel
- INSERM, U837, E3 Institut de Recherche sur le Cancer de Lille, Lille, France.
| |
Collapse
|
2310
|
Rebuttal from Authors re: Christian Doehn. Immunotherapy of Prostate Cancer. Eur Urol 2008;53:681–3. Eur Urol 2008. [DOI: 10.1016/j.eururo.2007.12.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
2311
|
Barbaric D, Corthals SL, Jastaniah WA, Asalanian S, Shimizu H, Reid GSD, Schultz KR. Detection of WT1-specific T cells in paediatric acute lymphoblastic leukaemia patients in first remission. Br J Haematol 2008; 141:271-3. [DOI: 10.1111/j.1365-2141.2008.07001.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
2312
|
Hellstrom KE, Hellstrom I. Vaccines to treat cancer--an old approach whose time has arrived. J Cell Biochem 2008; 102:291-300. [PMID: 17729239 DOI: 10.1002/jcb.21468] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
There are extensive DNA changes in tumor cells and the genes of tumor cells continuously mutate at a high rate. While this can provide therapeutic targets, it makes it unlikely that an agent that is selective for a single target will work against all cells in a tumor. However, it may be possible to use tumor epitopes as sentinels to engage adaptive and innate immunological mechanisms and create a tumor destructive environment effective also against variant cells that have lost a given antigen or their ability to present it. We hypothesize that therapeutic tumor vaccines, in combination with the targeting, to tumors, of costimulatory molecules such as anti-CD137scFv, or lymphokines such as GMCSF, will expand anti-tumor responses for therapeutic benefit when used as an adjunct to surgery and chemotherapy.
Collapse
Affiliation(s)
- Karl Erik Hellstrom
- Department of Pathology, Harborview Medical Center, University of Washington, Seattle, Washington 98104-2499, USA.
| | | |
Collapse
|
2313
|
Bellone M, Mondino A, Corti A. Vascular targeting, chemotherapy and active immunotherapy: teaming up to attack cancer. Trends Immunol 2008; 29:235-41. [PMID: 18375183 DOI: 10.1016/j.it.2008.02.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2007] [Revised: 01/30/2008] [Accepted: 02/01/2008] [Indexed: 01/08/2023]
Abstract
Chemotherapy has been combined with therapeutic tumor-specific vaccination in an attempt to simultaneously debulk tumors, increase the effector lymphocyte:tumor cell ratio, and favor immune-mediated tumor rejection. However, chemotherapy is often inadequate because of insufficient and uneven drug penetration into tumors, and because it might also cause, in some instances, undesirable side effects and immunosuppression. Here, we suggest a combined approach based on targeted alteration of the endothelial barrier function with vascular disrupting agents, such as tumor necrosis factor-alpha (TNF-alpha), before chemotherapy and tumor-specific vaccination. This approach has the potential to empower chemoimmunotherapeutic strategies by improving cytotoxic drug penetration into tumors while exploiting the proinflammatory and immunostimulating activities of TNF-alpha and active immunotherapy.
Collapse
Affiliation(s)
- Matteo Bellone
- Cancer Immunotherapy and Gene Therapy Program, San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy.
| | | | | |
Collapse
|
2314
|
Formenti SC, Demaria S. Effects of chemoradiation on tumor-host interactions: the immunologic side. J Clin Oncol 2008; 26:1562-3; author reply 1563. [PMID: 18349411 DOI: 10.1200/jco.2007.15.5499] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
2315
|
Exploiting dendritic cells and natural killer T cells in immunotherapy against malignancies. Trends Immunol 2008; 29:242-9. [PMID: 18372215 DOI: 10.1016/j.it.2008.02.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2007] [Revised: 01/28/2008] [Accepted: 02/13/2008] [Indexed: 01/09/2023]
Abstract
A primary focus of tumor immunotherapy research is to change the immune system so that it becomes immunized and not tolerized to the presentation of antigens by or from tumor cells. Dendritic cells (DCs) are the logical target for the development of immunotherapies because DCs instruct the ensuing immune response. Upon activation, invariant natural killer T (iNKT) cells have direct antitumor effects and also induce in situ DC maturation, linking the innate and adaptive arms of the immune system in an immunogenic form. The characterization and manipulation of DC function in tumor-bearing hosts will provide new insights into mechanisms of tumor immunology and lead to the development of successful clinical strategies.
Collapse
|
2316
|
Demaria S, Formenti SC. Sensors of ionizing radiation effects on the immunological microenvironment of cancer. Int J Radiat Biol 2008; 83:819-25. [PMID: 17852561 DOI: 10.1080/09553000701481816] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
PURPOSE When cancer develops in an immunocompetent host it represents the result of a successful deception of the immune system as to the nature of the danger and the type of response needed to reject the neoplastic tissue. We will briefly review some of the recently emerged evidence that irradiation of the tumor and its microenvironment can induce essential molecular signals required for an effective response of the immune system to the tumor. CONCLUSIONS The subversion of a highly organized tissue architecture is a hallmark of cancer, and results in uneven distribution of oxygen and nutrients, interstitial pressure gradients and areas of patchy necrosis and inflammation. In this microenvironment, cancer cells that carry mutations favoring survival rather than cell death in response to stress find a selection advantage. Importantly, the signals derived from the disruption of orderly physiology within tissues are also what the immune system has evolved to respond to. The type of response is tuned to be adequate to the cause of the disruption. An infectious organism will carry or elicit from the involved tissue a number of 'danger signals' leading to development of cell mediated and humoral responses to both eliminating the invader and preventing future infections. In contrast, a simple wound will call for a repair response. The sensors of the type of damage are complex molecular interactions between the damaged organ and cells of the innate and adaptive immune system. Progress in the identification of these interactions elucidates which pathways are specifically altered in cancer. It also provides a novel understanding of the radiation-induced effects on tumor immunogenicity. We propose that specific radiation-induced effects could be successfully exploited to improve the effectiveness of immunotherapy.
Collapse
Affiliation(s)
- Sandra Demaria
- Department of Pathology, NYU Cancer Institute, New York University School of Medicine, New York 10016, USA.
| | | |
Collapse
|
2317
|
Netea-Maier RT, Hunsucker SW, Hoevenaars BM, Helmke SM, Slootweg PJ, Hermus AR, Haugen BR, Duncan MW. Discovery and Validation of Protein Abundance Differences between Follicular Thyroid Neoplasms. Cancer Res 2008; 68:1572-80. [DOI: 10.1158/0008-5472.can-07-5020] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
2318
|
Prendergast GC. Immune escape as a fundamental trait of cancer: focus on IDO. Oncogene 2008; 27:3889-900. [PMID: 18317452 DOI: 10.1038/onc.2008.35] [Citation(s) in RCA: 250] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Immune escape is a critical gateway to malignancy. The emergence of this fundamental trait of cancer represents the defeat of immune surveillance, a potent, multi-armed and essential mode of cancer suppression that may influence the ultimate clinical impact of an early stage tumor. Indeed, immune escape may be a central modifier of clinical outcomes, by affecting tumor dormancy versus progression, licensing invasion and metastasis and impacting therapeutic response. Although relatively little studied until recently, immune suppression and escape in tumors are now hot areas with clinical translation of several new therapeutic agents already under way. The interconnections between signaling pathways that control immune escape and those that control proliferation, senescence, apoptosis, metabolic alterations, angiogenesis, invasion and metastasis remain virtually unexplored, offering rich new areas for investigation. Here, an overview of this area is provided with a focus on the tryptophan catabolic enzyme indoleamine 2,3-dioxygenase (IDO) and its recently discovered relative IDO2 that are implicated in suppressing T-cell immunity in normal and pathological settings including cancer. Emerging evidence suggests that during cancer progression activation of the IDO pathway might act as a preferred nodal modifier pathway for immune escape, for example analogous to the PI3K pathway for survival or the VEGF pathway for angiogenesis. Small molecule inhibitors of IDO and IDO2 heighten chemotherapeutic efficacy in mouse models of cancer in a nontoxic fashion and an initial lead compound entered phase I clinical trials in late 2007. New modalities in this area offer promising ways to broaden the combinatorial attack on advanced cancers, where immune escape mechanisms likely provide pivotal support.
Collapse
Affiliation(s)
- G C Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA.
| |
Collapse
|
2319
|
Skak K, Kragh M, Hausman D, Smyth MJ, Sivakumar PV. Interleukin 21: combination strategies for cancer therapy. Nat Rev Drug Discov 2008; 7:231-40. [DOI: 10.1038/nrd2482] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
2320
|
Cabezón C, Cabrera G, Paredes R, Ferreira A, Galanti N. Echinococcus granulosus calreticulin: Molecular characterization and hydatid cyst localization. Mol Immunol 2008; 45:1431-8. [DOI: 10.1016/j.molimm.2007.08.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Revised: 08/23/2007] [Accepted: 08/29/2007] [Indexed: 10/22/2022]
|
2321
|
Salaun B, Romero P, Lebecque S. Toll-like receptors' two-edged sword: when immunity meets apoptosis. Eur J Immunol 2008; 37:3311-8. [PMID: 18034428 DOI: 10.1002/eji.200737744] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Toll-like receptors (TLR) have emerged as key players in the detection of pathogens and the induction of anti-microbial immune response. TLR recognize pathogen-associated molecular patterns, and trigger anti-microbial innate immune responses ranging from the secretion of pro-inflammatory mediators to the increase of natural killer cell cytotoxicity. Besides activating the innate immune response, TLR engagement also shapes the adaptive immune response. Indeed, the broad diversity of signaling pathways initiated by TLR is progressively unraveled. Recent reports suggested that among the anti-microbial defenses they initiate, members of the TLR family can induce apoptosis. This review focuses on this newly described function of TLR, and emphasizes the similarities and differences between the different apoptosis-signaling pathways described downstream of TLR. The functional relevance of TLR-triggered apoptosis is also discussed, as therapeutic applications are likely to ensue in the near future.
Collapse
Affiliation(s)
- Bruno Salaun
- Ludwig Institute for Cancer Research, Lausanne Branch, Epalinges, Switzerland
| | | | | |
Collapse
|
2322
|
Pucci-Minafra I, Cancemi P, Albanese NN, Di Cara G, Marabeti MR, Marrazzo A, Minafra S. New Protein Clustering of Breast Cancer Tissue Proteomics Using Actin Content as a Cellularity Indicator. J Proteome Res 2008; 7:1412-8. [DOI: 10.1021/pr700748m] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Ida Pucci-Minafra
- Dipartimento di Oncologia Sperimentale e Applicazioni Cliniche (DOSAC), Università di Palermo, Patermo, Italy, Centro di Oncobiologia Sperimentale (COBS), Palermo, Italy, and Ospedale “La Maddalena” D.O. III livello, Palermo, Italy
| | - Patrizia Cancemi
- Dipartimento di Oncologia Sperimentale e Applicazioni Cliniche (DOSAC), Università di Palermo, Patermo, Italy, Centro di Oncobiologia Sperimentale (COBS), Palermo, Italy, and Ospedale “La Maddalena” D.O. III livello, Palermo, Italy
| | - Nadia Ninfa Albanese
- Dipartimento di Oncologia Sperimentale e Applicazioni Cliniche (DOSAC), Università di Palermo, Patermo, Italy, Centro di Oncobiologia Sperimentale (COBS), Palermo, Italy, and Ospedale “La Maddalena” D.O. III livello, Palermo, Italy
| | - Gianluca Di Cara
- Dipartimento di Oncologia Sperimentale e Applicazioni Cliniche (DOSAC), Università di Palermo, Patermo, Italy, Centro di Oncobiologia Sperimentale (COBS), Palermo, Italy, and Ospedale “La Maddalena” D.O. III livello, Palermo, Italy
| | - Maria Rita Marabeti
- Dipartimento di Oncologia Sperimentale e Applicazioni Cliniche (DOSAC), Università di Palermo, Patermo, Italy, Centro di Oncobiologia Sperimentale (COBS), Palermo, Italy, and Ospedale “La Maddalena” D.O. III livello, Palermo, Italy
| | - Antonio Marrazzo
- Dipartimento di Oncologia Sperimentale e Applicazioni Cliniche (DOSAC), Università di Palermo, Patermo, Italy, Centro di Oncobiologia Sperimentale (COBS), Palermo, Italy, and Ospedale “La Maddalena” D.O. III livello, Palermo, Italy
| | - Salvatore Minafra
- Dipartimento di Oncologia Sperimentale e Applicazioni Cliniche (DOSAC), Università di Palermo, Patermo, Italy, Centro di Oncobiologia Sperimentale (COBS), Palermo, Italy, and Ospedale “La Maddalena” D.O. III livello, Palermo, Italy
| |
Collapse
|
2323
|
Andersen MH, Sørensen RB, Schrama D, Svane IM, Becker JC, Thor Straten P. Cancer treatment: the combination of vaccination with other therapies. Cancer Immunol Immunother 2008; 57:1735-43. [PMID: 18286284 PMCID: PMC2522294 DOI: 10.1007/s00262-008-0480-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Accepted: 02/05/2008] [Indexed: 12/22/2022]
Abstract
Harnessing of the immune system by the development of ‘therapeutic’ vaccines, for the battle against cancer has been the focus of tremendous research efforts over the past two decades. As an illustration of the impressive amounts of data gathered over the past years, numerous antigens expressed on the surface of cancer cells, have been characterized. To this end, recent years research has focussed on characterization of antigens that play an important role for the growth and survival of cancer cells. Anti-apoptotic molecules like survivin that enhance the survival of cancer cells and facilitate their escape from cytotoxic therapies represent prime vaccination candidates. The characterization of a high number of tumor antigens allow the concurrent or serial immunological targeting of different proteins associated with such cancer traits. Moreover, while vaccination in itself is a promising new approach to fight cancer, the combination with additional therapy could create a number of synergistic effects. Herein we discuss the possibilities and prospects of vaccination when combined with other treatments. In this regard, cell death upon drug exposure may be immunogenic or non-immunogenic depending on the specific chemotherapeutics. Also, chemotherapy represents one of several options available for clearance of CD4+ Foxp3+ regulatory T cells. Moreover, therapies based on monoclonal antibodies may have synergistic potential in combination with vaccination, both when used for targeting of tumor cells and endothelial cells. The efficacy of therapeutic vaccination against cancer will over the next few years be studied in settings taking advantage of strategies in which vaccination is combined with other treatment modalities. These combinations should be based on current knowledge not only regarding the biology of the cancer cell per se, but also considering how treatment may influence the malignant cell population as well as the immune system.
Collapse
Affiliation(s)
- Mads Hald Andersen
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), Herlev University Hospital, 54P4, Herlev Ringvej 75, 2730 Herlev, Denmark
| | | | | | | | | | | |
Collapse
|
2324
|
Schmitz F, Heit A. Protective cancer immunotherapy: what can the innate immune system contribute? Expert Opin Biol Ther 2008; 8:31-43. [PMID: 18081535 DOI: 10.1517/14712598.8.1.31] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Despite significant efforts to induce protection against malignant diseases, the clinical effects of antitumour vaccines are poor. However, recent studies on a quadrivalent human papilloma virus vaccine suggest that protection against secondary tumour development is feasible. While this scenario benefits rather from antiviral protection than from direct antitumour responses, immunisation against cancers of non-viral origin demands strategies that rely on the circumvention of intrinsic regulatory mechanisms. Strong activation of innate immune cells seems to be key and, thus, the choice of adjuvant determines vaccination efficacy. The recently acquired knowledge about molecular and cellular recognition of microbial molecules suggests how one can modulate innate and adaptive immune reactions to potentially induce robust T- and B-cell reactions capable of prohibiting tumour development and progression. Here, the authors review the present knowledge of innate immune reactions, which may help to define rationales on the design of novel antitumour vaccines.
Collapse
Affiliation(s)
- Frank Schmitz
- Technical University Munich, Institute of Medical Microbiology, Immunology and Hygiene, Trogerstrasse 30, 81675 Munich, Germany.
| | | |
Collapse
|
2325
|
Zitvogel L, Apetoh L, Ghiringhelli F, Kroemer G. Immunological aspects of cancer chemotherapy. Nat Rev Immunol 2008; 8:59-73. [PMID: 18097448 DOI: 10.1038/nri2216] [Citation(s) in RCA: 1224] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Accumulating evidence indicates that the innate and adaptive immune systems make a crucial contribution to the antitumour effects of conventional chemotherapy-based and radiotherapy-based cancer treatments. Moreover, the molecular and cellular bases of the immunogenicity of cell death that is induced by cytotoxic agents are being progressively unravelled, challenging the guidelines that currently govern the development of anticancer drugs. Here, we review the immunological aspects of conventional cancer treatments and propose that future successes in the fight against cancer will rely on the development and clinical application of combined chemo- and immunotherapies.
Collapse
Affiliation(s)
- Laurence Zitvogel
- INSERM, U805, Institut Gustave Roussy, 39 Rue Camille Desmoulins, F-94805 Villejuif, Paris, France.
| | | | | | | |
Collapse
|
2326
|
Nonapoptotic role for Apaf-1 in the DNA damage checkpoint. Mol Cell 2008; 28:624-37. [PMID: 18042457 DOI: 10.1016/j.molcel.2007.09.030] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Revised: 05/11/2007] [Accepted: 09/24/2007] [Indexed: 12/22/2022]
Abstract
Apaf-1 is an essential factor for cytochrome c-driven caspase activation during mitochondrial apoptosis but has also an apoptosis-unrelated function. Knockdown of Apaf-1 in human cells, knockout of apaf-1 in mice, and loss-of-function mutations in the Caenorhabditis elegans apaf-1 homolog ced-4 reveal the implication of Apaf-1/CED-4 in DNA damage-induced cell-cycle arrest. Apaf-1 loss compromised the DNA damage checkpoints elicited by ionizing irradiation or chemotherapy. Apaf-1 depletion reduced the activation of the checkpoint kinase Chk1 provoked by DNA damage, and knockdown of Chk1 abrogated the Apaf-1-mediated cell-cycle arrest. Nuclear translocation of Apaf-1, induced in vitro by exogenous DNA-damaging agents, correlated in non-small cell lung cancer (NSCLC) with the endogenous activation of Chk-1, suggesting that this pathway is clinically relevant. Hence, Apaf-1 exerts two distinct, phylogenetically conserved roles in response to mitochondrial membrane permeabilization and DNA damage. These data point to a role for Apaf-1 as a bona fide tumor suppressor.
Collapse
|
2327
|
Palucka AK, Ueno H, Fay JW, Banchereau J. Taming cancer by inducing immunity via dendritic cells. Immunol Rev 2008; 220:129-50. [PMID: 17979844 DOI: 10.1111/j.1600-065x.2007.00575.x] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Immunotherapy seeks to mobilize a patient's immune system for therapeutic benefit. It can be passive, i.e. transfer of immune effector cells (T cells) or proteins (antibodies), or active, i.e. vaccination. In cancer, passive immunotherapy can lead to some objective clinical responses, thus demonstrating that the immune system can reject tumors. However, passive immunotherapy is not expected to yield long-lived memory T cells that might control tumor outgrowth. Active immunotherapy with dendritic cell (DC)-based vaccines has the potential to induce both tumor-specific effector and memory T cells. Early clinical trials testing vaccination with ex vivo-generated DCs pulsed with tumor antigens provide a proof-of-principle that therapeutic immunity can be elicited. Yet, there is a need to improve their efficacy. The next generation of DC vaccines is expected to generate large numbers of high-avidity effector CD8(+) T cells and to overcome regulatory T cells. Therapeutic vaccination protocols will combine improved ex vivo DC vaccines with therapies that offset the suppressive environment established by tumors.
Collapse
Affiliation(s)
- A Karolina Palucka
- Baylor Institute for Immunology Research and Baylor Research Institute, Dallas, TX, USA.
| | | | | | | |
Collapse
|
2328
|
Obeid M, Tesniere A, Panaretakis T, Tufi R, Joza N, van Endert P, Ghiringhelli F, Apetoh L, Chaput N, Flament C, Ullrich E, de Botton S, Zitvogel L, Kroemer G. Ecto-calreticulin in immunogenic chemotherapy. Immunol Rev 2008; 220:22-34. [PMID: 17979837 DOI: 10.1111/j.1600-065x.2007.00567.x] [Citation(s) in RCA: 171] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The conventional treatment of cancer relies upon radiotherapy and chemotherapy. Such treatments supposedly mediate their effects via the direct elimination of tumor cells. Nonetheless, there are circumstances in which conventional anti-cancer therapy can induce a modality of cellular demise that elicits innate and cognate immune responses, which in turn mediate part of the anti-tumor effect. Although different chemotherapeutic agents may kill tumor cells through an apparently homogeneous apoptotic pathway, they differ in their capacity to stimulate immunogenic cell death. We discovered that the pre-apoptotic translocation of intracellular calreticulin (endo-CRT) to the plasma membrane surface (ecto-CRT) is critical for the recognition and engulfment of dying tumor cells by dendritic cells. Thus, anthracyclines and gamma-irradiation that induce ecto-CRT cause immunogenic cell death, while other pro-apoptotic agents (such as mitomycin C and etoposide) induce neither ecto-CRT nor immunogenic cell death. Depletion of CRT abolishes the immunogenicity of cell death elicited by anthracyclines, while exogenous supply of CRT or enforcement of CRT exposure by pharmacological agents that favor CRT translocation can enhance the immunogenicity of cell death. For optimal anti-tumor vaccination and immunogenic chemotherapy, the same cells have to expose ecto-CRT and to succumb to apoptosis; if these events affect different cells, no anti-tumor immune response is elicited. These results may have far reaching implications for tumor immunology because (i) ecto-CRT exposure by tumor cells allows for the prediction of therapeutic outcome and because (ii) the re-establishment of ecto-CRT may ameliorate the efficacy of chemotherapy.
Collapse
|
2329
|
Apetoh L, Ghiringhelli F, Tesniere A, Criollo A, Ortiz C, Lidereau R, Mariette C, Chaput N, Mira JP, Delaloge S, André F, Tursz T, Kroemer G, Zitvogel L. The interaction between HMGB1 and TLR4 dictates the outcome of anticancer chemotherapy and radiotherapy. Immunol Rev 2008; 220:47-59. [PMID: 17979839 DOI: 10.1111/j.1600-065x.2007.00573.x] [Citation(s) in RCA: 440] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
For the last four decades, the treatment of cancer has relied on four treatment modalities, namely surgery, radiotherapy, cytotoxic chemotherapy, and hormonotherapy. Most of these therapies are believed to directly attack and eradicate tumor cells. The emerging concept that cancer is not just a disease of a tissue or an organ but also a host disease relies on evidence of tumor-induced immunosuppression and polymorphisms in genes involved in host protection against tumors. This theory is now gaining new impetus, based on our recent data showing that optimal therapeutic effects require the immunoadjuvant effect of tumor cell death induced by cytotoxic anticancer agents. Here, we show that the release of the high mobility group box 1 protein (HMGB1) by dying tumor cells is mandatory to license host dendritic cells (DCs) to process and present tumor antigens. HMGB1 interacts with Toll-like receptor 4 (TLR4) on DCs, which are selectively involved in the cross-priming of anti-tumor T lymphocytes in vivo. A TLR4 polymorphism that affects the binding of HMGB1 to TLR4 predicts early relapse after anthracycline-based chemotherapy in breast cancer patients. This knowledge may be clinically exploited to predict the immunogenicity and hence the efficacy of chemotherapeutic regimens.
Collapse
|
2330
|
Abolition of stress-induced protein synthesis sensitizes leukemia cells to anthracycline-induced death. Blood 2008; 111:2866-77. [PMID: 18182573 DOI: 10.1182/blood-2007-07-103242] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Anthracycline action has been thought to involve the neosynthesis of proapoptotic gene products and to therefore depend on protein synthesis for optimal effect. We found that inhibition of general, but not rapamycin-sensitive (cap-dependent), protein synthesis in the preapoptotic period enhanced anthracycline-induced acute myelogenous leukemia (AML) cell death, both in vitro and in several animal AML models. Pre-apoptotic anthracycline-exposed AML cells had altered translational specificity, with enhanced synthesis of a subset of proteins, including endoplasmatic reticulum chaperones. The altered translational specificity could be explained by perturbation (protein degradation, truncation, or dephosphorylation) of the cap-dependent translation initiation machinery and of proteins control-ing translation of specific mRNAs. We propose that judiciously timed inhibition of cap-independent translation is considered for combination therapy with anthracyclines in AML.
Collapse
|
2331
|
Srivastava RM, Varalakshmi C, Khar A. The Ischemia-Responsive Protein 94 (Irp94) Activates Dendritic Cells through NK Cell Receptor Protein-2/NK Group 2 Member D (NKR-P2/NKG2D) Leading to Their Maturation. THE JOURNAL OF IMMUNOLOGY 2008; 180:1117-30. [DOI: 10.4049/jimmunol.180.2.1117] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
2332
|
Emens LA. Chemotherapy and tumor immunity: an unexpected collaboration. FRONT BIOSCI-LANDMRK 2008; 13:249-57. [PMID: 17981543 DOI: 10.2741/2675] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chemotherapy directly targets the transformed tumor cell, and has long been a key component of therapy for most early and advanced cancers. However, its utility is ultimately limited by unavoidable toxicity to normal tissues, and by drug resistance pathways deeply embedded within the biology of the tumor cell itself. These limitations strongly argue for innovative strategies to treat and manage cancer. Engaging the power of the patient's own immune system is a highly attractive way to complement the activity of standard cancer treatment. Tumor vaccines offer the potential for preventing cancer in those at high risk for disease development, preventing relapse in those diagnosed with early cancer, and treating advanced disease. Notably, the barriers to tumor vaccine efficacy are distinct from the limitations of combination chemotherapy. The ability of vaccines to induce a response robust enough to mediate tumor rejection is limited by the extent of disease burden, the suppressive effect of the local tumor micronenvironment, and multiple layers of systemic immune tolerance established to keep the immune response turned off. Chemotherapy can be used with tumor vaccines in unexpected ways, breaking down these barriers and unleashing the full potential of the antitumor immune response.
Collapse
Affiliation(s)
- Leisha A Emens
- Department of Oncology, The Johns Hopkins University and the Sidney Kimmel Comprehensive Cancer Center, 1650 Orleans Street, Room 4M90, Bunting Blaustein Cancer Research Building, Baltimore, MD 21231-1000, USA.
| |
Collapse
|
2333
|
Vitale I, Galluzzi L, Vivet S, Nanty L, Dessen P, Senovilla L, Olaussen KA, Lazar V, Prudhomme M, Golsteyn RM, Castedo M, Kroemer G. Inhibition of Chk1 kills tetraploid tumor cells through a p53-dependent pathway. PLoS One 2007; 2:e1337. [PMID: 18159231 PMCID: PMC2131784 DOI: 10.1371/journal.pone.0001337] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2007] [Accepted: 11/16/2007] [Indexed: 11/19/2022] Open
Abstract
Tetraploidy constitutes an adaptation to stress and an intermediate step between euploidy and aneuploidy in oncogenesis. Tetraploid cells are particularly resistant against genotoxic stress including radiotherapy and chemotherapy. Here, we designed a strategy to preferentially kill tetraploid tumor cells. Depletion of checkpoint kinase-1 (Chk1) by siRNAs, transfection with dominant-negative Chk1 mutants or pharmacological Chk1 inhibition killed tetraploid colon cancer cells yet had minor effects on their diploid counterparts. Chk1 inhibition abolished the spindle assembly checkpoint and caused premature and abnormal mitoses that led to p53 activation and cell death at a higher frequency in tetraploid than in diploid cells. Similarly, abolition of the spindle checkpoint by knockdown of Bub1, BubR1 or Mad2 induced p53-dependent apoptosis of tetraploid cells. Chk1 inhibition reversed the cisplatin resistance of tetraploid cells in vitro and in vivo, in xenografted human cancers. Chk1 inhibition activated p53-regulated transcripts including Puma/BBC3 in tetraploid but not in diploid tumor cells. Altogether, our results demonstrate that, in tetraploid tumor cells, the inhibition of Chk1 sequentially triggers aberrant mitosis, p53 activation and Puma/BBC3-dependent mitochondrial apoptosis.
Collapse
Affiliation(s)
- Ilio Vitale
- INSERM, U848, Cancer and Immunity, Villejuif, France
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
| | - Lorenzo Galluzzi
- INSERM, U848, Cancer and Immunity, Villejuif, France
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
| | - Sonia Vivet
- INSERM, U848, Cancer and Immunity, Villejuif, France
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
| | - Lisa Nanty
- INSERM, U848, Cancer and Immunity, Villejuif, France
- Institut Gustave Roussy,Villejuif, France
| | - Philippe Dessen
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
- Centre National de la Recherche Scientifique (CNRS), FRE2939, Villejuif, France
- Unité de Génomique Fonctionnelle, Institut Gustave Roussy,Villejuif, France
| | - Laura Senovilla
- INSERM, U848, Cancer and Immunity, Villejuif, France
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
| | - Ken A. Olaussen
- INSERM, U848, Cancer and Immunity, Villejuif, France
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
| | - Vladimir Lazar
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
- Centre National de la Recherche Scientifique (CNRS), FRE2939, Villejuif, France
- Unité de Génomique Fonctionnelle, Institut Gustave Roussy,Villejuif, France
| | - Michelle Prudhomme
- Université Blaise Pascal, Synthèse et Etude de Systèmes à Intérêt Biologique, UMR 6504 Centre National de la Recherche Scientifique (CNRS), Aubière, France
| | | | - Maria Castedo
- INSERM, U848, Cancer and Immunity, Villejuif, France
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
| | - Guido Kroemer
- INSERM, U848, Cancer and Immunity, Villejuif, France
- Institut Gustave Roussy,Villejuif, France
- Université Paris Sud-11, Villejuif, France
| |
Collapse
|
2334
|
|
2335
|
Ueno H, Hawrylowicz CM, Banchereau J. Immunological intervention in human diseases. J Transl Med 2007; 5:59. [PMID: 18036229 PMCID: PMC2176053 DOI: 10.1186/1479-5876-5-59] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Accepted: 11/23/2007] [Indexed: 02/11/2023] Open
Abstract
A recent Keystone Symposium Meeting on "Immunological Intervention in Human Disease" was held in Big Sky, Montana on January, 6–11, 2007, organized by Jacques Banchereau, Federica Sallusto and Robert Coffman. It brought together basic scientists and clinicians from both academia and the pharmaceutical industry to discuss how the immune system is involved in the development of human diseases, including cancer, allergy, autoimmunity, and infectious diseases. We highlight advances in our understanding of the pathogenesis of immune-mediated diseases and future approaches in the immune therapeutic interventions. Considerable progress in the development of model systems and methodologies to monitor human immune responses will help to develop and to evaluate new immune-based therapies at pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Hideki Ueno
- Baylor Institute for Immunology Research and Baylor Research Institute, 3434 Live Oak St, Dallas, TX 75204, USA.
| | | | | |
Collapse
|
2336
|
Reduction of endoplasmic reticulum Ca2+ levels favors plasma membrane surface exposure of calreticulin. Cell Death Differ 2007; 15:274-82. [PMID: 18034188 DOI: 10.1038/sj.cdd.4402275] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Some chemotherapeutic agents can elicit apoptotic cancer cell death, thereby activating an anticancer immune response that influences therapeutic outcome. We previously reported that anthracyclins are particularly efficient in inducing immunogenic cell death, correlating with the pre-apoptotic exposure of calreticulin (CRT) on the plasma membrane surface of anthracyclin-treated tumor cells. Here, we investigated the role of cellular Ca(2+) homeostasis on CRT exposure. A neuroblastoma cell line (SH-SY5Y) failed to expose CRT in response to anthracyclin treatment. This defect in CRT exposure could be overcome by the overexpression of Reticulon-1C, a manipulation that led to a decrease in the Ca(2+) concentration within the endoplasmic reticulum lumen. The combination of Reticulon-1C expression and anthracyclin treatment yielded more pronounced endoplasmic reticulum Ca(2+) depletion than either of the two manipulations alone. Chelation of intracellular (and endoplasmic reticulum) Ca(2+), targeted expression of the ligand-binding domain of the IP(3) receptor and inhibition of the sarco-endoplasmic reticulum Ca(2+)-ATPase pump reduced endoplasmic reticulum Ca(2+) load and promoted pre-apoptotic CRT exposure on the cell surface, in SH-SY5Y and HeLa cells. These results provide evidence that endoplasmic reticulum Ca(2+) levels control the exposure of CRT.
Collapse
|
2337
|
Abstract
Apoptotic cell death is initiated by a morphologically homogenous entity that was considered to be non-immunogenic and non-inflammatory in nature. However, recent advances suggest that apoptosis, under certain circumstances, can be immunogenic. In particular, some characteristics of the plasma membrane, acquired at preapoptotic stage, can cause immune effectors to recognize and attack preapoptotic tumor cells. The signals that mediate the immunogenicity of tumor cells involve elements of the DNA damage response (such as ataxia telangiectasia mutated and p53 activation), elements of the endoplasmic reticulum stress response (such as eukaryotic initiation factor 2alpha phosphorylation), as well as elements of the apoptotic response (such as caspase activation). Depending on the signal-transduction pathway, tumor cells responding to chemotherapy or radiotherapy can express 'danger' and 'eat me' signals on the cell surface (such as NKG2D ligands, heat-shock proteins and calreticulin) or can secrete/release immunostimulatory factors (such as cytokines and high-mobility group box 1) to stimulate innate immune effectors. Likewise, the precise sequence of such events influences the 'decision' of the immune system to mount a cognate response or not. We therefore anticipate that the comprehension of the mechanisms governing the immunogenicity of cell death will have a profound impact on the design of anticancer therapies.
Collapse
|
2338
|
From regulation of dying cell engulfment to development of anti-cancer therapy. Cell Death Differ 2007; 15:29-38. [DOI: 10.1038/sj.cdd.4402271] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
2339
|
Decoding dangerous death: how cytotoxic chemotherapy invokes inflammation, immunity or nothing at all. Cell Death Differ 2007; 15:13-20. [PMID: 18007666 DOI: 10.1038/sj.cdd.4402255] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Chemotherapy and immunotherapy can be either synergistic or antagonistic modalities in the treatment of cancer. Cytotoxic chemotherapy not only affects the tumor but also targets dividing lymphocytes, the very cells that are required to develop an immune response. For this reason, chemo- and immunotherapy have been seen as antagonistic. However, cell death can be immunogenic and the way in which chemotherapeutic drug kills a tumor cell is likely to be an important determinant of how that dying cell interacts with the immune system and whether the interaction will lead to an immune response. When a cell dies as the result of infection, the immune system responds rapidly and the system of Toll-like receptors (TLR) plays a key role in this process. In this review, we will briefly summarize the intracellular signaling pathways that link TLR ligation with immune activation and we will address the questions where and how TLRs recognize their targets.
Collapse
|
2340
|
Ullrich E, Bonmort M, Mignot G, Kroemer G, Zitvogel L. Tumor stress, cell death and the ensuing immune response. Cell Death Differ 2007; 15:21-8. [PMID: 17992190 DOI: 10.1038/sj.cdd.4402266] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A cornucopia of physiological and pathological circumstances including anticancer chemotherapy and radiotherapy can induce cell death. However, the immunological consequences of tumor cell demise have remained largely elusive. The paradigm opposing 'apoptosis versus necrosis' as to their respective immunogenicity does not currently hold to predict long-term immunity. Moreover, the notion that tumor cells may be 'stressed' before death to be recognized by immune cells deserves to be underlined. 'Eat-me', 'danger' and 'killing' signals released by stressed tumor under the pressure of cytotoxic compounds may serve as links between the chemotherapy-elicited response of tumor cells and subsequent immune responses. This review will summarize the state-of-the-art of cancer immunity and describe how tumor cell death dictates the links between innate and acquired immunity.
Collapse
Affiliation(s)
- E Ullrich
- INSERM U805, Institut Gustave Roussy, Pavillon de Recherche 1, 39 rue Camille Desmoulins, Villejuif, France
| | | | | | | | | |
Collapse
|
2341
|
Calreticulin expression in the clonal plasma cells of patients with systemic light-chain (AL-) amyloidosis is associated with response to high-dose melphalan. Blood 2007; 111:549-57. [PMID: 17982021 DOI: 10.1182/blood-2007-05-090852] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In high doses with stem-cell transplantation, melphalan is an effective but toxic therapy for patients with systemic light-chain (AL-) amyloidosis, a protein deposition and monoclonal plasma cell disease. Melphalan can eliminate the indolent clonal plasma cells that cause the disease, an achievement called a complete response. Such a response is usually associated with extended survival, while no response (a less than 50% reduction) is not. Gene-expression studies and a stringently supervised analysis identified calreticulin as having significantly higher expression in the pretreatment plasma cells of patients with systemic AL-amyloidosis who then had a complete response to high-dose melphalan. Calreticulin is a pleiotropic calcium-binding protein found in the endoplasmic reticulum and the nucleus whose overexpression is associated with increased sensitivity to apoptotic stimuli. Real-time PCR and immunohistochemical staining also showed that expression of calreticulin was higher in the plasma cells of those with a complete response. Furthermore, wild-type murine embryonic fibroblasts were significantly more sensitive to melphalan than calreticulin knock-out murine embryonic fibroblasts. These data have important implications for understanding the activity of melphalan in plasma-cell diseases and support further investigation of calreticulin and its modulation in patients with systemic AL-amyloidosis receiving high-dose melphalan.
Collapse
|
2342
|
Abstract
Dendritic cells (DCs) orchestrate a repertoire of immune responses that bring about resistance to infection and silencing or tolerance to self. In the settings of infection and cancer, microbes and tumours can exploit DCs to evade immunity, but DCs also can generate resistance, a capacity that is readily enhanced with DC-targeted vaccines. During allergy, autoimmunity and transplant rejection, DCs instigate unwanted responses that cause disease, but, again, DCs can be harnessed to silence these conditions with novel therapies. Here we present some medical implications of DC biology that account for illness and provide opportunities for prevention and therapy.
Collapse
|
2343
|
Duus K, Pagh RT, Holmskov U, Højrup P, Skov S, Houen G. Interaction of Calreticulin with CD40 Ligand, TRAIL and Fas Ligand. Scand J Immunol 2007; 66:501-7. [DOI: 10.1111/j.1365-3083.2007.01999.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
2344
|
Hayashi E, Matsuzaki Y, Hasegawa G, Yaguchi T, Kurihara S, Fujita T, Kageshita T, Sano M, Kawakami Y. Identification of a Novel Cancer-Testis Antigen CRT2 Frequently Expressed in Various Cancers Using Representational Differential Analysis. Clin Cancer Res 2007; 13:6267-74. [DOI: 10.1158/1078-0432.ccr-07-1374] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
2345
|
Beachy S, Kisailus A, Repasky E, Subjeck J, Wang X, Kazim A. Engineering secretable forms of chaperones for immune modulation and vaccine development. Methods 2007; 43:184-93. [PMID: 17920514 DOI: 10.1016/j.ymeth.2007.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Accepted: 06/25/2007] [Indexed: 12/22/2022] Open
|
2346
|
Obeid M, Panaretakis T, Tesniere A, Joza N, Tufi R, Apetoh L, Ghiringhelli F, Zitvogel L, Kroemer G. Leveraging the immune system during chemotherapy: moving calreticulin to the cell surface converts apoptotic death from "silent" to immunogenic. Cancer Res 2007; 67:7941-4. [PMID: 17804698 DOI: 10.1158/0008-5472.can-07-1622] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In contrast to prior belief, tumor cell apoptosis is not necessarily silent but can be immunogenic. By tracing how anthracyclines and gamma-irradiation trigger immunogenic cell deaths, we found that they were causally connected to the exposure of calreticulin on the tumor cell surface, before apoptosis in the tumor cell itself occurred. Furthermore, we showed that calreticulin exposure was necessary and sufficient to increase proimmunogenic killing by other chemotherapies. Our findings suggest that calreticulin could serve as a biomarker to predict therapy-associated immune responses, and that tactics to expose calreticulin might improve the clinical efficacy of many cancer therapies.
Collapse
|
2347
|
Cao T, Ueno H, Glaser C, Fay JW, Palucka AK, Banchereau J. Both Langerhans cells and interstitial DC cross-present melanoma antigens and efficiently activate antigen-specific CTL. Eur J Immunol 2007; 37:2657-67. [PMID: 17683111 DOI: 10.1002/eji.200636499] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dendritic cells (DC) have a unique capacity to present external antigens to CD8(+) T cells, i.e. cross-presentation. However, it is not fully established whether the ability to cross-presentation is restricted to a unique subset of DC in humans. Here, we show that two major myeloid DC subsets, i.e. Langerhans cells (LC) and interstitial DC (Int-DC), have the ability to cross-present antigens to CD8(+) T cells in vitro. LC and Int-DC were obtained from DC generated by culturing human CD34(+)-hematopoietic progenitor cells with GM-CSF, FLT3-L, and TNF-alpha (CD34-DC). Both DC subsets were able to capture necrotic/apoptotic allogeneic melanoma cells and present antigens to CD8(+) T cells, resulting in efficient priming of naive CD8(+) T cells into CTL capable of killing melanoma cells. Strikingly, a single stimulation with either subset (LC or Int-DC) or total CD34-DC loaded with necrotic/apoptotic melanoma cells was sufficient to activate melanoma-specific memory CD8(+) T cells obtained from patients with metastatic melanoma to become effective CTL. Thus, this study provides the rationale to use CD34-DC loaded with necrotic/apoptotic allogeneic melanoma cells in a clinical trial.
Collapse
Affiliation(s)
- Tinghua Cao
- Baylor Institute for Immunology Research, Dallas, TX 75204, USA
| | | | | | | | | | | |
Collapse
|
2348
|
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells (APCs) specialized in the stimulation of naïve T lymphocytes, which are key components of antiviral and antitumor immunity. DCs are 'sentinels' of the immune system endowed with the mission to (1) sense invading pathogens as well as any form of tissue distress and (2) alert the effectors of the immune response. They represent a very heterogeneous population including subsets characterized by their anatomical locations and specific missions. Beyond their unique APC features, DCs exhibit a large array of effector functions that play critical roles in the induction and regulation of the cell-mediated as well as humoral immune responses. In the course of the antitumor immune response, DCs are unique in engulfing tumor cells killed by natural killer (NK) cells and cross-presenting tumor-associated antigens to cytotoxic T lymphocytes (CTLs). However, while DCs mediate antitumor immune responses by stimulating tumor-specific CTLs and NK cells, direct tumoricidal mechanisms have been recently evoked. This review addresses the other face of DCs to directly deliver apoptotic signals to stressed cells, their role in tumor cell death, and its implication in the design of DC-based cancer immunotherapies.
Collapse
|
2349
|
Interactions of tumor cells with dendritic cells: balancing immunity and tolerance. Cell Death Differ 2007; 15:39-50. [PMID: 17948027 DOI: 10.1038/sj.cdd.4402247] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Dendritic cells (DCs) are antigen-presenting cells specialized to initiate and maintain immunity and tolerance. DCs initiate immune responses in a manner that depends on signals they receive from pathogens, surrounding cells and their products. Most tumors are infiltrated by DCs. Thus, interactions between DCs and dying tumor cells may determine the balance between immunity and tolerance to tumor cells. In addition, DCs also display non-immunologic effects on tumors and the tumor microenvironment. Therefore, improved understanding of the cross talk between tumor cells and DCs may suggest new approaches to improve cancer therapy.
Collapse
|
2350
|
Ling S, Pi X, Holoshitz J. The Rheumatoid Arthritis Shared Epitope Triggers Innate Immune Signaling via Cell Surface Calreticulin. THE JOURNAL OF IMMUNOLOGY 2007; 179:6359-67. [DOI: 10.4049/jimmunol.179.9.6359] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|