1
|
Guerin MV, Regnier F, Thoreau M, Vimeux L, Benard M, Dransart E, Penny HL, Johannes L, Trautmann A, Bercovici N. Local IFNα enhances the anti-tumoral efficacy of systemic anti-PD1 to prevent tumor relapse. J Immunother Cancer 2020; 8:jitc-2020-000996. [PMID: 33239415 PMCID: PMC7689071 DOI: 10.1136/jitc-2020-000996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2020] [Indexed: 11/23/2022] Open
Abstract
Background Tumor relapse constitutes a major challenge for anti-tumoral treatments, including immunotherapies. Indeed, most cancer-related deaths occur during the tumor relapse phase. Methods We designed a mouse model of tumor relapse in which mice transplanted with E7+ TC1 tumor cells received a single therapeutic vaccination of STxB-E7+IFNα. Unlike the complete regression observed after two vaccinations, such a treatment induced a transient shrinkage of the tumor mass, followed by a rapid tumor outgrowth. To prevent this relapse, we tested the efficacy of a local administration of IFNα together with a systemic therapy with anti-PD1 Ab. The immune response was analyzed during both the tumor regression and relapse phases. Results We show that, during the regression phase, tumors of mice treated with a single vaccination of STxB-E7 + IFNα harbor fewer activated CD8 T cells and monocytes than tumors doomed to fully regress after two vaccinations. In contrast, the systemic injection of an anti-PD1 Ab combined with the peri-tumoral injection of IFNα in this time frame promotes infiltration of activated CD8 T cells and myeloid cells, which, together, exert a high cytotoxicity in vitro against TC1 cells. Moreover, the IFNα and anti-PD1 Ab combination was found to be more efficient than IFNα or anti-PD1 used alone in preventing tumor relapse and was better able to prolong mice survival. Conclusions Together, these results indicate that the local increase of IFNα in combination with an anti-PD1 therapy is an effective way to promote efficient and durable innate and adaptive immune responses preventing tumor relapse.
Collapse
Affiliation(s)
- Marion V Guerin
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Fabienne Regnier
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Maxime Thoreau
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Lene Vimeux
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Matthieu Benard
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Estelle Dransart
- Institut Curie, PSL Research University, Cellular and Chemical Biology unit, UMR3666 CNRS, U1143 INSERM, 75248, CEDEX 05, Paris, France
| | | | - Ludger Johannes
- Institut Curie, PSL Research University, Cellular and Chemical Biology unit, UMR3666 CNRS, U1143 INSERM, 75248, CEDEX 05, Paris, France
| | - Alain Trautmann
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| | - Nadege Bercovici
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014, Paris, France
| |
Collapse
|
2
|
Ohfuji S. A Secretory Variant of Cerebral Meningioma with Partial Spontaneous Regression in a Cow. J Comp Pathol 2020; 180:86-90. [PMID: 33222879 DOI: 10.1016/j.jcpa.2020.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/16/2020] [Accepted: 08/28/2020] [Indexed: 10/23/2022]
Abstract
A 3.5-year-old Holstein cow presented with a 2-week history of weakness, ataxia and difficulty in rising, followed by recumbency. At necropsy, a 3 × 2.5 cm tumour mass was found in the subdural area of the left parietal lobe of the cerebrum. Histopathology revealed the tumour to be a variant of secretory meningeal tumour. Immunohistochemically, all of the neoplastic cells expressed vimentin and some were immunopositive for cytokeratin. There were variably sized, scattered pseudocysts containing eosinophilic material that stained positively with Alcian blue and periodic acid-Schiff stains. The marginal zone of the tumour exhibited regressive change characterized by lymphocytic infiltration and extensive fibrotic areas enclosing large numbers of tumour cell nests in which most tumour cells were apoptotic. This tumour was considered to be a secretory variant of cerebral meningioma with partial spontaneous regression.
Collapse
Affiliation(s)
- Susumu Ohfuji
- Department of Histopathology, Diagnostic Animal Pathology Office, Hokkaido, Japan.
| |
Collapse
|
3
|
Omene C, Ma L, Moore J, Ouyang H, Illa-Bochaca I, Chou W, Patel MS, Sebastiano C, Demaria S, Mao JH, Karagoz K, Gatza ML, Barcellos-Hoff MH. Aggressive Mammary Cancers Lacking Lymphocytic Infiltration Arise in Irradiated Mice and Can Be Prevented by Dietary Intervention. Cancer Immunol Res 2019; 8:217-229. [PMID: 31831632 DOI: 10.1158/2326-6066.cir-19-0253] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/26/2019] [Accepted: 11/27/2019] [Indexed: 01/06/2023]
Abstract
Because the incidence of breast cancer increases decades after ionizing radiation exposure, aging has been implicated in the evolution of the tumor microenvironment and tumor progression. Here, we investigated radiation-induced carcinogenesis using a model in which the mammary glands of 10-month-old BALB/c mice were transplanted with Trp53-null mammary tissue 3 days after exposure to low doses of sparsely ionizing γ-radiation or densely ionizing particle radiation. Mammary transplants in aged, irradiated hosts gave rise to significantly more tumors that grew more rapidly than those in sham-irradiated mice, with the most pronounced effects seen in mice irradiated with densely ionizing particle radiation. Tumor transcriptomes identified a characteristic immune signature of these aggressive cancers. Consistent with this, fast-growing tumors exhibited an immunosuppressive tumor microenvironment with few infiltrating lymphocytes, abundant immunosuppressive myeloid cells, and high COX-2 and TGFβ. Only irradiated hosts gave rise to tumors lacking cytotoxic CD8+ lymphocytes (defined here as immune desert), which also occurred in younger irradiated hosts. These data suggest that host irradiation may promote immunosuppression. To test this, young chimera mice were fed chow containing a honeybee-derived compound with anti-inflammatory and immunomodulatory properties, caffeic acid phenethyl ester (CAPE). CAPE prevented the detrimental effects of host irradiation on tumor growth rate, immune signature, and immunosuppression. These data indicated that low-dose radiation, particularly densely ionizing exposure of aged mice, promoted more aggressive cancers by suppressing antitumor immunity. Dietary intervention with a nontoxic immunomodulatory agent could prevent systemic effects of radiation that fuel carcinogenesis, supporting the potential of this strategy for cancer prevention.
Collapse
Affiliation(s)
- Coral Omene
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Lin Ma
- University of California, San Francisco, San Francisco, California
| | - Jade Moore
- University of California, San Francisco, San Francisco, California
| | - Haoxu Ouyang
- New York University School of Medicine, New York, New York
| | | | - William Chou
- University of California, San Francisco, San Francisco, California
| | - Manan S Patel
- New York University School of Medicine, New York, New York
| | | | - Sandra Demaria
- New York University School of Medicine, New York, New York
| | - Jian-Hua Mao
- Lawrence Berkeley National Laboratory, Berkeley, California
| | - Kubra Karagoz
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Michael L Gatza
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | | |
Collapse
|
4
|
Implementation of Mass Cytometry for Immunoprofiling of Patients with Solid Tumors. J Immunol Res 2019; 2019:6705949. [PMID: 30886872 PMCID: PMC6388349 DOI: 10.1155/2019/6705949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/12/2018] [Accepted: 11/21/2018] [Indexed: 11/22/2022] Open
Abstract
Monitoring immune responses to solid cancers may be a better prognostic tool than conventional staging criteria, and it can also serve as an important criterion for the selection of individualized therapy. Multiparametric phenotyping by mass cytometry extended possibilities for immunoprofiling. However, careful optimization of each step of such method is necessary for obtaining reliable results. Also, with respect to procedure length and costs, sample preparation, staining, and storage should be optimized. Here, we designed a panel of 31 antibodies which allows for identification of several subpopulations of lymphoid and myeloid cells in a solid tumor and peripheral blood simultaneously. For sample preparation, disaggregation of tumor tissue with two different collagenases combined with DNase I was compared, and removal of dead or tumor cells by magnetic separation was evaluated. Two possible procedures of barcoding for single-tube staining of several samples were examined. While the palladium-based barcoding affected the stability of several antigens, the staining with two differently labeled CD45 antibodies was suitable for cells isolated from a patient's blood and tumor. The storage of samples in the intercalation solution for up to two weeks did not influence results of the analysis, which allowed the measurement of samples collected within this interval on the same day. This procedure optimized on samples from patients with head and neck squamous cell carcinoma enabled identification of various immune cells including rare subpopulations.
Collapse
|
5
|
Experimental Combined Immunotherapy of Tumours with Major Histocompatibility Complex Class I Downregulation. Int J Mol Sci 2018; 19:ijms19113693. [PMID: 30469401 PMCID: PMC6274939 DOI: 10.3390/ijms19113693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/14/2018] [Accepted: 11/17/2018] [Indexed: 12/20/2022] Open
Abstract
Combined immunotherapy constitutes a novel, advanced strategy in cancer treatment. In this study, we investigated immunotherapy in the mouse TC-1/A9 model of human papillomavirus type 16 (HPV16)-associated tumors characterized by major histocompatibility complex class I (MHC-I) downregulation. We found that the induction of a significant anti-tumor response required a combination of DNA vaccination with the administration of an adjuvant, either the synthetic oligodeoxynucleotide ODN1826, carrying immunostimulatory CpG motifs, or α-galactosylceramide (α-GalCer). The most profound anti-tumor effect was achieved when these adjuvants were applied in a mix with a one-week delay relative to DNA immunization. Combined immunotherapy induced tumor infiltration with various subsets of immune cells contributing to tumor regression, of which cluster of differentiation (CD) 8⁺ T cells were the predominant subpopulation. In contrast, the numbers of tumor-associated macrophages (TAMs) were not markedly increased after immunotherapy but in vivo and in vitro results showed that they could be repolarized to an anti-tumor M1 phenotype. A blockade of T cell immunoglobulin and mucin-domain containing-3 (Tim-3) immune checkpoint had a negligible effect on anti-tumor immunity and TAMs repolarization. Our results demonstrate a benefit of combined immunotherapy comprising the activation of both adaptive and innate immunity in the treatment of tumors with reduced MHC-I expression.
Collapse
|
6
|
Trautmann A. From kinetics and cellular cooperations to cancer immunotherapies. Oncotarget 2018; 7:44779-44789. [PMID: 27014912 PMCID: PMC5190134 DOI: 10.18632/oncotarget.8242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/04/2016] [Indexed: 12/26/2022] Open
Abstract
In this review will be underlined two simple ideas of potential interest for the design of cancer immunotherapies. One concerns the importance of kinetics, with the key notion that a single cause may trigger two opposite effects with different kinetics. The importance of this phenomenon will be underlined in neurobiology, transcription networks and the immune system. The second idea is that efficient immune responses have been selected against pathogens, throughout evolution. They are never due to a single cell type, but always require multiple, complex cellular cooperations. One cannot recognize this fact and persist in the presently dominant T-cell centered view of cancer immunotherapies. Suggestions will be made to incorporate these simple ideas for improving these therapies.
Collapse
Affiliation(s)
- Alain Trautmann
- INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Equipe Labellisée "Ligue contre le Cancer", Paris, France
| |
Collapse
|
7
|
Abstract
The therapy of different advanced-stage malignancies with monoclonal antibodies blocking programmed cell death protein 1 (PD-1)/PD-1 ligand 1 (PD-L1) signaling has had an impressive long-lasting effect in a portion of patients, but in most cases, this therapy was not successful, or a secondary resistance developed. To enhance its efficacy in treated patients, predictive biomarkers are searched for and various combination treatments are intensively investigated. As the downregulation of major histocompatibility complex (MHC) class I molecules is one of the most frequent mechanisms of tumor escape from the host’s immunity, it should be considered in PD-1/PD-L1 checkpoint inhibition. The potential for the use of a PD-1/PD-L1 blockade in the treatment of tumors with aberrant MHC class I expression is discussed, and some strategies of combination therapy are suggested.
Collapse
|
8
|
Vaccine-induced tumor regression requires a dynamic cooperation between T cells and myeloid cells at the tumor site. Oncotarget 2016; 6:27832-46. [PMID: 26337837 PMCID: PMC4695029 DOI: 10.18632/oncotarget.4940] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 07/29/2015] [Indexed: 12/31/2022] Open
Abstract
Most cancer immunotherapies under present investigation are based on the belief that cytotoxic T cells are the most important anti-tumoral immune cells, whereas intra-tumoral macrophages would rather play a pro-tumoral role. We have challenged this antagonistic point of view and searched for collaborative contributions by tumor-infiltrating T cells and macrophages, reminiscent of those observed in anti-infectious responses. We demonstrate that, in a model of therapeutic vaccination, cooperation between myeloid cells and T cells is indeed required for tumor rejection. Vaccination elicited an early rise of CD11b+ myeloid cells that preceded and conditioned the intra-tumoral accumulation of CD8+ T cells. Conversely, CD8+ T cells and IFNγ production activated myeloid cells were required for tumor regression. A 4-fold reduction of CD8+ T cell infiltrate in CXCR3KO mice did not prevent tumor regression, whereas a reduction of tumor-infiltrating myeloid cells significantly interfered with vaccine efficiency. We show that macrophages from regressing tumors can kill tumor cells in two ways: phagocytosis and TNFα release. Altogether, our data suggest new strategies to improve the efficiency of cancer immunotherapies, by promoting intra-tumoral cooperation between macrophages and T cells.
Collapse
|
9
|
Fend L, Remy-Ziller C, Foloppe J, Kempf J, Cochin S, Barraud L, Accart N, Erbs P, Fournel S, Préville X. Oncolytic virotherapy with an armed vaccinia virus in an orthotopic model of renal carcinoma is associated with modification of the tumor microenvironment. Oncoimmunology 2015; 5:e1080414. [PMID: 27057460 PMCID: PMC4801465 DOI: 10.1080/2162402x.2015.1080414] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 07/31/2015] [Accepted: 07/31/2015] [Indexed: 12/21/2022] Open
Abstract
Oncolytic virotherapy is an emergent promising therapeutic approach for the treatment of cancer. We have constructed a vaccinia virus (WR strain) deleted for thymidine kinase (TK) and ribonucleotide reductase (RR) genes that expressed the fusion suicide gene FCU1 derived from the yeast cytosine deaminase and uracil phosphoribosyltransferase genes. We evaluated this construct (VV-FCU1) in the orthotopic model of renal carcinoma (RenCa). Systemic administration of VV-FCU1 resulted in orthotopic tumor growth inhibition, despite temporary expression of viral proteins. VV-FCU1 treatment was associated with an infiltration of tumors by CD8+ T lymphocytes and a decrease in the proportion of infiltrating Tregs, thus modifying the ratio of CD8+/CD4+ Treg in favor of CD8+cytotoxic T cells. We demonstrated that VV-FCU1 treatment prolonged survival of animals implanted with RenCa cells in kidney. Depletion of CD8+ T cells abolished the therapeutic effect of VV-FCU1 while depletion of CD4+ T cells enhanced its protective activity. Administration of the prodrug 5-fluorocytosine (5-FC) resulted in a sustained control of tumor growth but did not extend survival. This study shows the importance of CD4+ and CD8+ T cells in vaccinia virus-mediated oncolytic virotherapy and suggests that this approach may be evaluated for the treatment of human renal cell carcinoma.
Collapse
Affiliation(s)
- Laetitia Fend
- Transgene S.A., 400 boulevard Gonthier d'Andernach, Parc d'innovation, CS80166, Illkirch-Graffenstaden Cedex, France and Institut Gustave Roussy, Unité INSERM 1015 114 rue Edouard-Vaillant , 94805 Villejuif Cedex, France
| | - Christelle Remy-Ziller
- Transgene S.A., 400 boulevard Gonthier d'Andernach, Parc d'innovation, CS80166, Illkirch-Graffenstaden Cedex, France and Institut Gustave Roussy, Unité INSERM 1015 114 rue Edouard-Vaillant , 94805 Villejuif Cedex, France
| | - Johann Foloppe
- Transgene S.A., 400 boulevard Gonthier d'Andernach, Parc d'innovation, CS80166, Illkirch-Graffenstaden Cedex, France and Institut Gustave Roussy, Unité INSERM 1015 114 rue Edouard-Vaillant , 94805 Villejuif Cedex, France
| | - Juliette Kempf
- Transgene S.A., 400 boulevard Gonthier d'Andernach, Parc d'innovation, CS80166, Illkirch-Graffenstaden Cedex, France and Institut Gustave Roussy, Unité INSERM 1015 114 rue Edouard-Vaillant , 94805 Villejuif Cedex, France
| | - Sandrine Cochin
- Transgene S.A., 400 boulevard Gonthier d'Andernach, Parc d'innovation, CS80166, Illkirch-Graffenstaden Cedex, France and Institut Gustave Roussy, Unité INSERM 1015 114 rue Edouard-Vaillant , 94805 Villejuif Cedex, France
| | - Luc Barraud
- Transgene S.A., 400 boulevard Gonthier d'Andernach, Parc d'innovation, CS80166, Illkirch-Graffenstaden Cedex, France and Institut Gustave Roussy, Unité INSERM 1015 114 rue Edouard-Vaillant , 94805 Villejuif Cedex, France
| | - Nathalie Accart
- Novartis Institutes for Biomedical Research, Analytical Sciences and Imaging , WSJ386, Basel, Switzerland
| | - Philippe Erbs
- Transgene S.A., 400 boulevard Gonthier d'Andernach, Parc d'innovation, CS80166, Illkirch-Graffenstaden Cedex, France and Institut Gustave Roussy, Unité INSERM 1015 114 rue Edouard-Vaillant , 94805 Villejuif Cedex, France
| | - Sylvie Fournel
- Laboratoire de Conception et Application de Molécules Bioactives, Equipe de Biovectorologie, UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie , 74 Route du Rhin- BP60024 , Illkirch-Graffenstaden Cedex, France
| | - Xavier Préville
- Transgene S.A., 400 boulevard Gonthier d'Andernach, Parc d'innovation, CS80166, Illkirch-Graffenstaden Cedex, France and Institut Gustave Roussy, Unité INSERM 1015 114 rue Edouard-Vaillant , 94805 Villejuif Cedex, France
| |
Collapse
|
10
|
Vacchelli E, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Monoclonal antibodies in cancer therapy. Oncoimmunology 2014; 2:e22789. [PMID: 23482847 PMCID: PMC3583934 DOI: 10.4161/onci.22789] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
During the past 20 years, dozens-if not hundreds-of monoclonal antibodies have been developed and characterized for their capacity to mediate antineoplastic effects, either as they activate/enhance tumor-specific immune responses, either as they interrupt cancer cell-intrinsic signal transduction cascades, either as they specifically delivery toxins to malignant cells or as they block the tumor-stroma interaction. Such an intense research effort has lead to the approval by FDA of no less than 14 distinct molecules for use in humans affected by hematological or solid malignancies. In the inaugural issue of OncoImmunology, we briefly described the scientific rationale behind the use of monoclonal antibodies in cancer therapy and discussed recent, ongoing clinical studies investigating the safety and efficacy of this approach in patients. Here, we summarize the latest developments in this exciting area of clinical research, focusing on high impact studies that have been published during the last 15 months and clinical trials launched in the same period to investigate the therapeutic profile of promising, yet hitherto investigational, monoclonal antibodies.
Collapse
Affiliation(s)
- Erika Vacchelli
- Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France ; INSERM; U848; Villejuif, France
| | | | | | | | | | | | | |
Collapse
|
11
|
In vivo imaging-based mathematical modeling techniques that enhance the understanding of oncogene addiction in relation to tumor growth. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2013; 2013:802512. [PMID: 23573174 PMCID: PMC3616361 DOI: 10.1155/2013/802512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 02/15/2013] [Indexed: 11/18/2022]
Abstract
The dependence on the overexpression of a single oncogene constitutes an exploitable weakness for molecular targeted therapy. These drugs can produce dramatic tumor regression by targeting the driving oncogene, but relapse often follows. Understanding the complex interactions of the tumor's multifaceted response to oncogene inactivation is key to tumor regression. It has become clear that a collection of cellular responses lead to regression and that immune-mediated steps are vital to preventing relapse. Our integrative mathematical model includes a variety of cellular response mechanisms of tumors to oncogene inactivation. It allows for correct predictions of the time course of events following oncogene inactivation and their impact on tumor burden. A number of aspects of our mathematical model have proven to be necessary for recapitulating our experimental results. These include a number of heterogeneous tumor cell states since cells following different cellular programs have vastly different fates. Stochastic transitions between these states are necessary to capture the effect of escape from oncogene addiction (i.e., resistance). Finally, delay differential equations were used to accurately model the tumor growth kinetics that we have observed. We use this to model oncogene addiction in MYC-induced lymphoma, osteosarcoma, and hepatocellular carcinoma.
Collapse
|
12
|
Immune microenvironment in tumor progression: characteristics and challenges for therapy. JOURNAL OF ONCOLOGY 2012; 2012:608406. [PMID: 22927846 PMCID: PMC3423944 DOI: 10.1155/2012/608406] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 06/18/2012] [Accepted: 07/02/2012] [Indexed: 02/08/2023]
Abstract
The tumor microenvironment plays a critical role in cancer development, progression, and control. The molecular and cellular nature of the tumor immune microenvironment influences disease outcome by altering the balance of suppressive versus cytotoxic responses in the vicinity of the tumor. Recent developments in systems biology have improved our understanding of the complex interactions between tumors and their immunological microenvironment in various human cancers. Effective tumor surveillance by the host immune system protects against disease, but chronic inflammation and tumor “immunoediting” have also been implicated in disease development and progression. Accordingly, reactivation and maintenance of appropriate antitumor responses within the tumor microenvironment correlate with a good prognosis in cancer patients. Improved understanding of the factors that shape the tumor microenvironment will be critical for the development of effective future strategies for disease management. The manipulation of these microenvironmental factors is already emerging as a promising tool for novel cancer treatments. In this paper, we summarize the various roles of the tumor microenvironment in cancer, focusing on immunological mediators of tumor progression and control, as well as the significant challenges for future therapies.
Collapse
|