1
|
The Biology of Immune-Active Cancers and Their Regulatory Mechanisms. Cancer Treat Res 2020; 180:149-172. [PMID: 32215869 DOI: 10.1007/978-3-030-38862-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
The development of cancer results from the evolutionary balance between the proliferating aptitude of cancer cells and the response of the host's tissues. Some cancers are characterized by genetic instability dependent upon impaired DNA repair mechanisms that lead to the chaotic disruption of multiple cellular functions often in excess of the cancer survival needs and may exert broad effects on surrounding tissues, some beneficial and some detrimental to cancer growth. Among them, inflammatory processes that accompany wound healing may initiate a reaction of the host against the neo-formation. This is possibly triggered by the release by dying cancer cells of molecules known as Damage-Associated Molecular Patterns (DAMPs) following a process termed Immunogenic Cell Death (ICD) that initiates an immune response through innate and adaptive mechanisms. Indeed, analysis of large cancer data sets has shown that ICD is strictly associated with the activation of other immune effector or immune-regulatory pathways. Here, we will describe how immune activation and compensatory immune-regulatory mechanisms balance anti-cancer immune surveillance and the roles that innate and adaptive immunity play including the weight that neo-epitopes may exert as initiators and sculptors of high-affinity memory and effector immune responses against cancer. We will discuss the evolutionary basis for the existence of immune checkpoints and how several theories raised to explain cancer resistance to immunotherapy represent a facet of a similar evolutionary phenomenon that we described in the Theory of Everything. We will show how the biology of immunogenicity and counterbalancing immune regulation is widespread across cancers independent of their ontogenesis while subtle idiosyncratic differences are discernible among them. Finally, we will suggest that overcoming immune resistance implies distinct approaches relevant to the immune context of individual cancers.
Collapse
|
2
|
Payne KK, Keim RC, Graham L, Idowu MO, Wan W, Wang XY, Toor AA, Bear HD, Manjili MH. Tumor-reactive immune cells protect against metastatic tumor and induce immunoediting of indolent but not quiescent tumor cells. J Leukoc Biol 2016; 100:625-35. [PMID: 26928306 PMCID: PMC4982610 DOI: 10.1189/jlb.5a1215-580r] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 02/08/2016] [Indexed: 11/24/2022] Open
Abstract
Quiescent, but not indolent, dormant tumor cells are resistant to immunoediting, and best targets for immunotherapy of cancer. Two major barriers to cancer immunotherapy include tumor-induced immune suppression mediated by myeloid-derived suppressor cells and poor immunogenicity of the tumor-expressing self-antigens. To overcome these barriers, we reprogrammed tumor-immune cell cross-talk by combined use of decitabine and adoptive immunotherapy, containing tumor-sensitized T cells and CD25+ NKT cells. Decitabine functioned to induce the expression of highly immunogenic cancer testis antigens in the tumor, while also reducing the frequency of myeloid-derived suppressor cells and the presence of CD25+ NKT cells rendered T cells, resistant to remaining myeloid-derived suppressor cells. This combinatorial therapy significantly prolonged survival of animals bearing metastatic tumor cells. Adoptive immunotherapy also induced tumor immunoediting, resulting in tumor escape and associated disease-related mortality. To identify a tumor target that is incapable of escape from the immune response, we used dormant tumor cells. We used Adriamycin chemotherapy or radiation therapy, which simultaneously induce tumor cell death and tumor dormancy. Resultant dormant cells became refractory to additional doses of Adriamycin or radiation therapy, but they remained sensitive to tumor-reactive immune cells. Importantly, we discovered that dormant tumor cells contained indolent cells that expressed low levels of Ki67 and quiescent cells that were Ki67 negative. Whereas the former were prone to tumor immunoediting and escape, the latter did not demonstrate immunoediting. Our results suggest that immunotherapy could be highly effective against quiescent dormant tumor cells. The challenge is to develop combinatorial therapies that could establish a quiescent type of tumor dormancy, which would be the best target for immunotherapy.
Collapse
Affiliation(s)
- Kyle K Payne
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA; Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA; The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Rebecca C Keim
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA; Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Laura Graham
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA; Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Michael O Idowu
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA; Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Wen Wan
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA; Department of Biostatistics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Xiang-Yang Wang
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA; Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Amir A Toor
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA; Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA; and
| | - Harry D Bear
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA; Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Masoud H Manjili
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA; Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA;
| |
Collapse
|
3
|
Massa C, Thomas C, Wang E, Marincola F, Seliger B. Different maturation cocktails provide dendritic cells with different chemoattractive properties. J Transl Med 2015; 13:175. [PMID: 26695182 PMCID: PMC4467838 DOI: 10.1186/s12967-015-0528-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/11/2015] [Indexed: 12/16/2022] Open
Abstract
Background Dendritic cells (DC) are currently implemented as immunotherapeutic strategy for the treatment of tumor patients based on their central role in the immune system. Despite good results were obtained in vitro and in animal models, their clinical use has provided limited success suggesting the requirement to optimise the protocol for their production. Methods A cDNA array was performed on FastDC obtained from the differentiation of human peripheral blood monocytes stimulated with the clinical gold standard or with two alternative maturation cocktails combining interferon (IFN)γ and ligands for different toll like receptors (TLR). Results A stronger modulation of the DC transcriptome with respect to immature DC was found in alternatively stimulated DC when compared to DC stimulated with the clinical gold standard. A major class of molecules differentially expressed using distinct DC stimulation protocols were chemokines. Validation of their differential expression pattern at the mRNA and protein level confirmed the secretion of inflammatory chemokines by the alternative DC. Functional analyses of the chemotactic properties of DC “wash out” supernatants highlighted the ability of alternative, but not of gold standard DC to efficiently recruit immune cells with a prevalence of monocytes. Effector cells belonging to the innate as well as adaptive immunity were also attracted and the interaction with alternative DC resulted in enhanced secretion of IFNγ and induction of cytotoxic activity. Using leukocytes from cancer patients, it was demonstrated that the monocyte-attracting activity targeted cells with an inflammatory phenotype characterised by high levels of HLA-DR expression. Conclusions Despite other classes of immune modulatory genes differently expressed in the alternative DC require to be investigated and characterised regarding their functional consequences, the reduced maturation state and chemoattractive properties of the gold standard versus alternative DC clearly promote the necessity to change the clinically used maturation cocktail of DC in order to improve the outcome of patients treated with DC-based vaccines.
Collapse
Affiliation(s)
- Chiara Massa
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger str. 2, 06112, Halle (Saale), Germany.
| | - Carolin Thomas
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger str. 2, 06112, Halle (Saale), Germany.
| | - Ena Wang
- Department of Transfusion Medicine, National Institute of Health Clinical Center, Bethesda, USA. .,Sidra Medical and Research Center, Doha, Qatar.
| | - Francesco Marincola
- Department of Transfusion Medicine, National Institute of Health Clinical Center, Bethesda, USA. .,Sidra Medical and Research Center, Doha, Qatar.
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger str. 2, 06112, Halle (Saale), Germany.
| |
Collapse
|
4
|
Hannesdóttir L, Tymoszuk P, Parajuli N, Wasmer MH, Philipp S, Daschil N, Datta S, Koller JB, Tripp CH, Stoitzner P, Müller-Holzner E, Wiegers GJ, Sexl V, Villunger A, Doppler W. Lapatinib and doxorubicin enhance the Stat1-dependent antitumor immune response. Eur J Immunol 2013; 43:2718-29. [DOI: 10.1002/eji.201242505] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 05/02/2013] [Accepted: 06/06/2013] [Indexed: 12/22/2022]
Affiliation(s)
- Lára Hannesdóttir
- Division of Medical Biochemistry; Biocenter, Innsbruck Medical University; Innsbruck Austria
| | - Piotr Tymoszuk
- Division of Medical Biochemistry; Biocenter, Innsbruck Medical University; Innsbruck Austria
| | - Nirmala Parajuli
- Division of Medical Biochemistry; Biocenter, Innsbruck Medical University; Innsbruck Austria
| | - Marie-Helene Wasmer
- Division of Medical Biochemistry; Biocenter, Innsbruck Medical University; Innsbruck Austria
| | - Sonja Philipp
- Division of Medical Biochemistry; Biocenter, Innsbruck Medical University; Innsbruck Austria
| | - Nina Daschil
- Division of Medical Biochemistry; Biocenter, Innsbruck Medical University; Innsbruck Austria
| | - Sebak Datta
- Division of Medical Biochemistry; Biocenter, Innsbruck Medical University; Innsbruck Austria
| | - Johann-Benedikt Koller
- Division of Medical Biochemistry; Biocenter, Innsbruck Medical University; Innsbruck Austria
| | - Christoph H. Tripp
- Department of Dermatology; Innsbruck Medical University; Innsbruck Austria
- Oncotyrol - Center for Personalized Cancer Medicine; Innsbruck Austria
| | - Patrizia Stoitzner
- Department of Dermatology; Innsbruck Medical University; Innsbruck Austria
| | | | - Gerrit Jan Wiegers
- Division of Developmental Immunology, Biocenter; Innsbruck Medical University; Innsbruck Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology; Veterinary University Vienna; Vienna Austria
| | - Andreas Villunger
- Division of Developmental Immunology, Biocenter; Innsbruck Medical University; Innsbruck Austria
| | - Wolfgang Doppler
- Division of Medical Biochemistry; Biocenter, Innsbruck Medical University; Innsbruck Austria
| |
Collapse
|
5
|
Busse A, Rapion J, Fusi A, Suciu S, Nonnenmacher A, Santinami M, Kruit WHJ, Testori A, Punt CJA, Dalgleish AG, Spatz A, Eggermont AMM, Keilholz U. Analysis of surrogate gene expression markers in peripheral blood of melanoma patients to predict treatment outcome of adjuvant pegylated interferon alpha 2b (EORTC 18991 side study). Cancer Immunol Immunother 2013; 62:1223-33. [PMID: 23624802 PMCID: PMC11028414 DOI: 10.1007/s00262-013-1428-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 04/10/2013] [Indexed: 10/26/2022]
Abstract
We analysed mRNA levels of interferon response genes (ISG15, STAT1, CXCL10) of inhibitors of the JAK/STAT pathway (STAT3, SOCS1, SOCS3) and of cytokines (TNFα, IL10, TGFß1) in peripheral blood of 91 stage III melanoma patients enrolled in EORTC 18991 trial to find biomarkers indicative for disease stage and predictive for efficacy of pegylated interferon alpha-2b (PEG-IFNα-2b) therapy. mRNA levels were analysed at baseline and after 6 months. Univariate and multivariate analyses were performed to estimate the prognostic and predictive role of mRNA levels for distant metastasis-free survival (DMFS) and relapse-free survival (RFS). Compared to healthy controls, melanoma patients showed significantly higher TGFβ1 mRNA levels. In a multivariate model, increasing SOCS1 and SOCS3 mRNA levels were associated with worse RFS (P = 0.02 and P = 0.04, respectively) and DMFS (P = 0.05 and P = 0.05, respectively) due to negative correlation between, respectively, SOCS1/SOCS3 mRNA levels and ulceration or Breslow thickness. No impact of PEG-IFNα-2b on mRNA levels was observed except for ISG15 mRNA levels, which decreased in the treatment arm (P = 0.001). It seems that patients with a decrease >60 % of ISG15 mRNA levels during 6 months PEG-IFNα-2b had inferior outcome.
Collapse
Affiliation(s)
- Antonia Busse
- Department of Medicine III, Charité-CBF, Hindenburgdamm 30, 12200, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Petro M, Kish D, Guryanova OA, Ilyinskaya G, Kondratova A, Fairchild RL, Gorbachev AV. Cutaneous Tumors Cease CXCL9/Mig Production as a Result of IFN-γ–Mediated Immunoediting. THE JOURNAL OF IMMUNOLOGY 2012; 190:832-41. [DOI: 10.4049/jimmunol.1201906] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
7
|
Reinboth J, Ascierto ML, Chen NG, Zhang Q, Yu YA, Aguilar RJ, Carretero R, Worschech A, Zhao Y, Wang E, Marincola FM, Szalay AA. Correlates between host and viral transcriptional program associated with different oncolytic vaccinia virus isolates. Hum Gene Ther Methods 2012; 23:285-96. [PMID: 23131031 DOI: 10.1089/hgtb.2012.057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Vaccinia virus (VACV) has emerged as an attractive tool in oncolytic virotherapy. VACV replication efficiency plays a crucial role in the therapeutic outcome. However, little is known about the influence of host factors on viral replication efficiency and permissiveness of a host cell line to infection and oncolysis. In this study, replication of the attenuated VACV GLV-1h68 strain and three wild-type VACV isolates was determined in two autologous human melanoma cell lines (888-MEL and 1936-MEL). Host gene expression and viral gene expression in infected cells were evaluated via respective expression array platforms. Microarray analyses followed by sequential statistical approaches characterized human genes that change specifically due to virus infection. Viral gene transcription correlated with viral replication in a time-dependent manner. A set of human genes revealed strong correlations with the respective viral gene expression. Finally we identified a set of human genes with possible predictive value for viral replication in an independent dataset. The results demonstrate a probable correlation between viral replication, early gene expression, and the respective host response, and thus a possible involvement of human host factors in viral early replication. The characterization of human target genes that influence viral replication could help answer the question of host cell permissiveness to oncolytic virotherapy and provide important information for the development of novel recombinant vaccinia viruses with improved features to enhance replication rate and hence trigger therapeutic outcome.
Collapse
Affiliation(s)
- Jennifer Reinboth
- Genelux Corporation, San Diego Science Center, San Diego, CA 92109, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Reinboth J, Ascierto ML, Chen NG, Zhang Q, Yu YA, Aguilar RJ, Carretero R, Worschech A, Zhao Y, Wang E, Marincola FM, Szalay AA. Correlates between host and viral transcriptional program associated with different oncolytic vaccinia virus isolates. Hum Gene Ther Methods 2012. [DOI: 10.1089/hum.2012.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
9
|
Payne KK, Manjili MH. Adaptive immune responses associated with breast cancer relapse. Arch Immunol Ther Exp (Warsz) 2012; 60:345-50. [PMID: 22911133 DOI: 10.1007/s00005-012-0185-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 05/28/2012] [Indexed: 12/21/2022]
Abstract
The generation, survival, and differentiation of breast cancer stem cells (BCSC) in immunocompetent hosts remain elusive. Some investigators have shown that BCSC can be induced from epithelial tumor cells by the pathologic epithelial to mesenchymal transition (EMT). Emerging evidence suggests that the induction of EMT among epithelial tumor cells originates from signals produced by the non-tumor cells that constitute the tumor microenvironment, including the immune effectors that infiltrate the tumors. Thus, this suggests that the immune system not only has anti-tumor function, but also paradoxically immunoedits tumors, facilitating tumor escape and progression. Indeed, many studies in human breast cancers show both positive and negative associations between the infiltration of various immune effectors (e.g., CD4 and CD8 T cells) and the propensity to relapse with metastatic disease. These observations suggest that distinct types of immune effector cells may induce or inhibit tumor relapse. This review focuses on recent advances in identifying components of the immune system that may directly induce tumor escape and relapse. We propose that levels of interferon (IFN)-γ production or levels of the expression of IFN-γ receptor α on tumor cells may determine whether tumor inhibitory or relapse-promoting effect of IFN-γ may prevail.
Collapse
Affiliation(s)
- Kyle K Payne
- Department of Microbiology and Immunology, Massey Cancer Center, Virginia Commonwealth University, 401 College Street, Box 980035, Richmond, VA 23298, USA
| | | |
Collapse
|
10
|
Carretero R, Wang E, Rodriguez AI, Reinboth J, Ascierto ML, Engle AM, Liu H, Camacho FM, Marincola FM, Garrido F, Cabrera T. Regression of melanoma metastases after immunotherapy is associated with activation of antigen presentation and interferon-mediated rejection genes. Int J Cancer 2012; 131:387-95. [PMID: 21964766 PMCID: PMC3504975 DOI: 10.1002/ijc.26471] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Accepted: 09/12/2011] [Indexed: 01/11/2023]
Abstract
We present the results of a comparative gene expression analysis of 15 metastases (10 regressing and 5 progressing) obtained from 2 melanoma patients with mixed response following different forms of immunotherapy. Whole genome transcriptional analysis clearly indicate that regression of melanoma metastases is due to an acute immune rejection mediated by the upregulation of genes involved in antigen presentation and interferon mediated response (STAT-1/IRF-1) in all the regressing metastases from both patients. In contrast, progressing metastases showed low transcription levels of genes involved in these pathways. Histological analysis showed T cells and HLA-DR positive infiltrating cells in the regressing but not in the progressing metastases. Quantitative expression analysis of HLA-A,B and C genes on microdisected tumoral regions indicate higher HLA expression in regressing than in progressing metastases. The molecular signature obtained in melanoma rejection appeared to be similar to that observed in other forms of immune-mediated tissue-specific rejection such as allograft, pathogen clearance, graft versus host or autoimmune disease, supporting the immunological constant of rejection. We favor the idea that the major factor determining the success or failure of immunotherapy is the nature of HLA Class I alterations in tumor cells and not the type of immunotherapy used. If the molecular alteration is reversible by the immunotherapy, the HLA expression will be upregulated and the lesion will be recognized and rejected. In contrast, if the defect is structural the MHC Class I expression will remain unchanged and the lesion will progress.
Collapse
Affiliation(s)
- Rafael Carretero
- Departamento de Bioquímica, Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
- Departamento de Análisis Clínicos e Inmunología, Hospital Virgen de las Nieves, Granada, Spain
| | - Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD
| | - Ana I. Rodriguez
- Departamento de Análisis Clínicos e Inmunología, Hospital Virgen de las Nieves, Granada, Spain
| | - Jennifer Reinboth
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD
- Department of Biochemistry, Biocenter, University of Wuerzburg, Wuerzburg, Germany
- Genelux Corporation, San Diego Science Center, San Diego, CA
| | - Maria L. Ascierto
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD
| | - Alyson M. Engle
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD
| | - Hui Liu
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD
| | | | - Francesco M. Marincola
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine and Trans-NIH Center for Human Immunology (CHI), National Institutes of Health, Bethesda, MD
| | - Federico Garrido
- Departamento de Bioquímica, Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
- Departamento de Análisis Clínicos e Inmunología, Hospital Virgen de las Nieves, Granada, Spain
| | - Teresa Cabrera
- Departamento de Bioquímica, Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
- Departamento de Análisis Clínicos e Inmunología, Hospital Virgen de las Nieves, Granada, Spain
| |
Collapse
|
11
|
Sun M, Hoffman D, Sundaresan G, Yang L, Lamichhane N, Zweit J. Synthesis and characterization of intrinsically radiolabeled quantum dots for bimodal detection. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2012; 2:122-135. [PMID: 23133807 PMCID: PMC3477726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 02/01/2012] [Indexed: 06/01/2023]
Abstract
A novel approach was developed to synthesize radioactive quantum dots (r-QDs) thereby enabling both optical and radionuclide signals to be detected from the same intrinsic bimodal probe. This proof-of-concept is exemplified by the incorporation of the radionuclide (109)Cadmium into the core/shell of the nanoparticle. Green and near infrared (NIR) emission intrinsic r-QDs were synthesized and characterized. Zwitterionic and Poly-polyethlene glycol (PEGylated) ligands were synthesized and used to coat r-QDs. Zwitterionic NIR r-QDs (quantum yield = 11%) and PEGylated NIR r-QDs (quantum yield = 14%) with an average size of 13.8 nm and 16.8 nm were obtained respectively. The biodistribution of NIR zwitterionic and PEGylated r-QDs in nude mice was investigated and zwitterionic r-QDs showed longer blood circulation (t(1/2) = 21.4±1.1 hrs) than their PEGylated counterparts (t(1/2) = 6.4±0.5 min). Both zwitterionic and PEGylated r-QDs exhibited progressive accumulation in the liver and spleen, but the magnitude of the accumulation (%ID/g) was about 3-6 fold higher with the PEGylated r-QDs at all the time points. The results demonstrated the feasibility of r-QDs synthesis in quantitative yield and retention of fluorescence following incorporation of radioactivity into the core/shell of the nanoparticle. The gamma signal from the same fluorescent elemental material enabled quantitative and robust pharmacokinetic measurements and how these changed depended on the type of coating ligands used. This strategy for intrinsically radio-labeling the QDs is currently being implemented in our laboratory for the incorporation of other radiometals.
Collapse
Affiliation(s)
- Minghao Sun
- Center for Molecular Imaging, Department of Radiology, Virginia Commonwealth University Richmond, Virginia, 23298, USA
| | | | | | | | | | | |
Collapse
|
12
|
Gowda M, Godder K, Kmieciak M, Worschech A, Ascierto ML, Wang E, Marincola FM, Manjili MH. Distinct signatures of the immune responses in low risk versus high risk neuroblastoma. J Transl Med 2011; 9:170. [PMID: 21978632 PMCID: PMC3195752 DOI: 10.1186/1479-5876-9-170] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 10/06/2011] [Indexed: 12/29/2022] Open
Abstract
Background Over 90% of low risk (LR) neuroblastoma patients survive whereas less than 30% of high risk (HR) patients are long term survivors. Age (children younger than 18 months old) is associated with LR disease. Considering that adaptive immune system is well developed in older children, and that T cells were shown to be involved in tumor escape and progression of cancers, we sought to determine whether HR patients may tend to show a signature of adaptive immune responses compared to LR patients who tend to have diminished T-cell responses but an intact innate immune response. Methods We performed microarray analysis of RNA extracted from the tumor specimens of HR and LR patients. Flow cytometry was performed to determine the cellular constituents in the blood while multiplex cytokine array was used to detect the cytokine profile in patients' sera. A HR tumor cell line, SK-N-SH, was also used for detecting the response to IL-1β, a cytokines which is involved in the innate immune responses. Results Distinct patterns of gene expression were detected in HR and LR patients indicating an active T-cell response and a diminished adaptive immune response, respectively. A diminished adaptive immune response in LR patients was evident by higher levels of IL-10 in the sera. In addition, HR patients had lower levels of circulating myeloid derived suppressor cells (MDSC) compared with a control LR patient. LR patients showed slightly higher levels of cytokines of the innate immune responses. Treatment of the HR tumor line with IL-1β induced expression of cytokines of the innate immune responses. Conclusions This data suggests that adaptive immune responses may play an important role in the progression of HR disease whereas innate immune responses may be active in LR patients.
Collapse
Affiliation(s)
- Madhu Gowda
- Department of Pediatrics, Children's Hospital of Richmond, Richmond, VA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Idowu MO, Kmieciak M, Dumur C, Burton RS, Grimes MM, Powers CN, Manjili MH. CD44(+)/CD24(-/low) cancer stem/progenitor cells are more abundant in triple-negative invasive breast carcinoma phenotype and are associated with poor outcome. Hum Pathol 2011; 43:364-73. [PMID: 21835433 DOI: 10.1016/j.humpath.2011.05.005] [Citation(s) in RCA: 195] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 05/03/2011] [Accepted: 05/04/2011] [Indexed: 12/16/2022]
Abstract
Women classified as having triple-negative tumors have a poor prognosis. The importance of CD44(+)/CD24(-/low) (stem/progenitor cell-phenotype) in breast cancer patients has also been appreciated. However, correlation between triple negativity and CD44(+)/CD24(-/low) with tumor recurrence remains elusive. In the present study, we evaluated tumor specimens of 50 breast cancer patients with known hormone receptor status for whom we had follow-up information and outcome data available, and performed immunohistochemistry analysis to determine CD44 and CD24 expression. Gene expression arrays were also independently performed on 52 breast cancer specimens with banked frozen tissue. Lastly, we used FVBN202 transgenic mouse model of breast carcinoma and determined the hormone receptor status, the proportion of CD44(+)/CD24(-/low) breast cancer stem-like cells, and the behavior of the tumor. We determined that patients with triple-negative tumors had significantly higher incidence of recurrence or distant metastasis associated with increased frequency of breast cancer stem cell phenotypes compared with those with non-triple-negative tumors. Preclinical studies in FVBN202 transgenic mice confirmed these findings by showing that relapsed tumors were triple negative and had significantly higher frequency of breast cancer stem cells compared with their related primary tumors. Unlike non-triple-negative primary tumors, relapsed triple-negative tumors were tumorigenic at low doses when inoculated into FVBN202 transgenic mice. These findings suggest that CD44(+)/CD24(-/low) breast cancer stem-like cells play an important role in the clinical behavior of triple-negative breast cancer and that development of therapeutic targets directed to breast cancer stem-like cells may lead to reduction in the aggressiveness of triple-negative breast cancers.
Collapse
Affiliation(s)
- Michael O Idowu
- Department of Pathology, Virginia Commonwealth University Medical Center, Richmond, VA 23298, USA.
| | | | | | | | | | | | | |
Collapse
|
14
|
O'Meara MM, Disis ML. Therapeutic cancer vaccines and translating vaccinomics science to the global health clinic: emerging applications toward proof of concept. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2011; 15:579-88. [PMID: 21732821 DOI: 10.1089/omi.2010.0149] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
As vaccines evolve to be a more common treatment for some cancers, further research is needed to improve the process of developing vaccines and assessing response to treatment. Vaccinomics involves a wide-ranging integration of multiple high throughput technologies including transcriptional, translational, and posttranslational population-based assessments of the human genome, transcriptome, proteome, and immunome. Vaccinomics combines the fields of immunogenetics, immunogenomics, immunoproteomics, and basic immunology to create vaccines that are tailor made to an individual or groups of individuals. This broad range of omics applications to tumor immunology includes antigen discovery, diagnostic biomarkers, cancer vaccine development, predictors of immune response, and clinical response biomarkers. These technologies have aided in the advancement of cancer vaccine development, as illustrated in examples including NY-ESO-1 originally defined by SEREX, and HER2/neu peptides analyzed via high-throughput epitope prediction methods. As technology improves, it presents an opportunity to improve cancer immunotherapy on a global scale, and attention must also be given to utilize these high-throughput methods for the understanding of cancer and immune signatures across populations.
Collapse
Affiliation(s)
- Megan M O'Meara
- Tumor Vaccine Group, Center for Translational Medicine in Women's Health, University of Washington, Seattle, Washington 98195-8050, USA
| | | |
Collapse
|
15
|
Pakravan N, Hassan ZM. Comparison of adjuvant activity of N- and C-terminal domain of gp96 in a Her2-positive breast cancer model. Cell Stress Chaperones 2011; 16:449-57. [PMID: 21359667 PMCID: PMC3118821 DOI: 10.1007/s12192-011-0258-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2010] [Revised: 01/22/2011] [Accepted: 01/26/2011] [Indexed: 10/18/2022] Open
Abstract
It has been frequently reported that gp96 acts as a strong biologic adjuvant. Some studies have even investigated adjuvant activity of the gp96 C- or N-terminal domain. The controversy surrounding adjuvant activity of gp96 terminal domains prompted us to compare adjuvant activity of gp96 C- or N-terminal domain toward Her2/neu, as DNA vaccine in a Her2/neu-positive breast cancer model. To do so, mice were immunized with DNA vaccine consisting of transmembrane and extracellular domain (TM + ECD) of rat Her2/neu alone or fused to N- or C-terminal domain of gp96. Treatment with Her2/neu fused to N-terminal domain of gp96 resulted in tumor progression, compared to the groups vaccinated with pCT/Her2 or pHer2. Immunological examination revealed that treatment with Her2/neu fused to N-terminal domain of gp96 led to significantly lower survival rates, higher interferon-γ secretion, and induced infiltration of CD4(+)/CD8(+) cells to the tumor site. However, it could not induce cytotoxic T lymphocyte activity, did not decrease regulatory T cell percentage at the tumor site, and eventually led to tumor progression. Our results reveal that gp96 N-terminal domain does not have adjuvant activity toward Her2/neu. It is also proposed that adjuvant activity and the resultant immune response of gp96 terminal domains may be directed by the antigen applied.
Collapse
Affiliation(s)
- Nafiseh Pakravan
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zuhair Mohammad Hassan
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Ale-Ahmad Avenue, P.O. Box 14115-331, Tehran, Iran
| |
Collapse
|
16
|
Kmieciak M, Basu D, Payne KK, Toor A, Yacoub A, Wang XY, Smith L, Bear HD, Manjili MH. Activated NKT cells and NK cells render T cells resistant to myeloid-derived suppressor cells and result in an effective adoptive cellular therapy against breast cancer in the FVBN202 transgenic mouse. THE JOURNAL OF IMMUNOLOGY 2011; 187:708-17. [PMID: 21670315 DOI: 10.4049/jimmunol.1100502] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Attempts to cure breast cancer by adoptive cellular therapy (ACT) have not been successful. This is primarily due to the presence of tumor-induced immune-suppressive mechanisms as well as the failure of tumor-reactive T cells to provide long-term memory responses in vivo. To address these clinically important challenges, we developed an ex vivo protocol for the expansion of tumor-reactive immune cells obtained from tumor-bearing animals prior to or after local radiation therapy. We used an Ag-free protocol that included bryostatin 1/ionomycin and sequential common γ-chain cytokines (IL-7/IL-15 + IL-2). The proposed protocol expanded tumor-reactive T cells as well as activated non-T cells, including NKT cells, NK cells, and IFN-γ-producing killer dendritic cells. Antitumor efficacy of T cells depended on the presence of non-T cells. The effector non-T cells also rendered T cells resistant to myeloid-derived suppressor cells. Radiation therapy altered phenotypic distribution and differentiation of T cells as well as their ability to generate central memory T cells. ACT by means of the expanded cells protected animals from tumor challenge and generated long-term memory responses against the tumor, provided that leukocytes were derived from tumor-bearing animals prior to radiation therapy. The ex vivo protocol was also able to expand HER-2/neu-specific T cells derived from the PBMC of a single patient with breast carcinoma. These data suggest that the proposed ACT protocol should be studied further in breast cancer patients.
Collapse
Affiliation(s)
- Maciej Kmieciak
- Department of Microbiology and Immunology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23298, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Manjili MH. Revisiting cancer immunoediting by understanding cancer immune complexity. J Pathol 2011; 224:5-9. [PMID: 21480229 PMCID: PMC3092128 DOI: 10.1002/path.2865] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 01/20/2011] [Indexed: 01/07/2023]
Abstract
Since 1909, the cancer immunosurveillance concept has undergone four distinct eras. These include a general acceptance during 1957–1974, an abandonment during 1974–1996, resurrection during 1996–2001 in the form of an elegant theory of tumour immunoediting proposed by Robert Schreiber, and a retreat since 2006. Recently, in the Journal of Pathology, Ciampricotti et al reported an elegant experimental model designed by establishing RAG2−/−/MMTV-NeuT mice. Using this, they demonstrated that the development and metastasis of HER-2/neu-positive spontaneous mammary carcinoma were not altered by the presence or absence of the adaptive immune system. Their fascinating results are a call to revisit controversial reports as to an effective role of the adaptive immune system in tumour inhibition versus tumour promotion or tolerance in the development of spontaneous carcinomas. Ciampricotti and colleagues present a strong case for revising our ideas of cancer immunoediting and appreciating the complexity of the interaction between cancer and the immune system.
Collapse
Affiliation(s)
- Masoud H Manjili
- Department of Microbiology and Immunology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23298, USA.
| |
Collapse
|
18
|
Ascierto ML, Kmieciak M, Idowu MO, Manjili R, Zhao Y, Grimes M, Dumur C, Wang E, Ramakrishnan V, Wang XY, Bear HD, Marincola FM, Manjili MH. A signature of immune function genes associated with recurrence-free survival in breast cancer patients. Breast Cancer Res Treat 2011; 131:871-80. [PMID: 21479927 DOI: 10.1007/s10549-011-1470-x] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2011] [Accepted: 03/17/2011] [Indexed: 12/11/2022]
Abstract
The clinical significance of tumor-infiltrating immune cells has been reported in a variety of human carcinomas including breast cancer. However, molecular signature of tumor-infiltrating immune cells and their prognostic value in breast cancer patients remain elusive. We hypothesized that a distinct network of immune function genes at the tumor site can predict a low risk versus high risk of distant relapse in breast cancer patients regardless of the status of ER, PR, or HER-2/neu in their tumors. We conducted retrospective studies in a diverse cohort of breast cancer patients with a 1-5 year tumor relapse versus those with up to 7 years relapse-free survival. The RNAs were extracted from the frozen tumor specimens at the time of diagnosis and subjected to microarray analysis and real-time RT-PCR. Paraffin-embedded tissues were also subjected to immunohistochemistry staining. We determined that a network of immune function genes involved in B cell development, interferon signaling associated with allograft rejection and autoimmune reaction, antigen presentation pathway, and cross talk between adaptive and innate immune responses were exclusively upregulated in patients with relapse-free survival. Among the 299 genes, five genes which included B cell response genes were found to predict with >85% accuracy relapse-free survival. Real-time RT-PCR confirmed the 5-gene prognostic signature that was distinct from an FDA-cleared 70-gene signature of MammaPrint panel and from the Oncotype DX recurrence score assay panel. These data suggest that neoadjuvant immunotherapy in patients with high risk of relapse may reduce tumor recurrence by inducing the immune function genes.
Collapse
Affiliation(s)
- Maria Libera Ascierto
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine and Center for Human Immunology, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Wiseman M D C. Questions from the fourth son: a clinician reflects on immunomonitoring, surrogate markers and systems biology. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2011; 8:279-287. [PMID: 21631130 DOI: 10.3934/mbe.2011.8.279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
The fourth son is the one who doesn't even know how to ask a question. Tumor immunology is challenged by the failure to identify reliable surrogate markers in vaccine and other experimental therapies for cancer; perhaps investigators haven't yet asked the right questions. Unlike prophylactic vaccines for infectious disease, where the development of antibody is considered a satisfactory endpoint, no such endpoint exists for human therapeutic vaccines. Why is this? Despite an extensive roster of in vitro assays that correlate immune responses to favorable clinical outcomes, no assay is sufficiently reliable to be usefully predictive for vaccine therapy. The discussion reviews some of the historical developments in tumor immunology and the problem of defining a causal relationship when strong correlations are identified. The development of mathematical models from empirical data may help inform the clinician/scientist about underlying mechanisms and help frame new testable hypotheses.
Collapse
|
20
|
Kmieciak M, Payne KK, Idowu MO, Grimes MM, Graham L, Ascierto ML, Wang E, Wang XY, Bear HD, Manjili MH. Tumor escape and progression of HER-2/neu negative breast cancer under immune pressure. J Transl Med 2011; 9:35. [PMID: 21453513 PMCID: PMC3076247 DOI: 10.1186/1479-5876-9-35] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Accepted: 03/31/2011] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Emerging data from pre-clinical and clinical studies suggest that HER-2/neu-specific T cell responses could induce HER-2/neu antigen loss in the tumor cells. These data suggest that patients with HER-2/neu negative breast cancer might have had HER-2/neu positive premalignant lesions in the past that progressed to HER-2/neu negative breast cancer under HER-2/neu-specific immune pressure. METHODS We conducted a pilot study in patients with HER-2/neu positive and HER-2/neu negative breast cancers as well as a patient with ductal carcinoma in situ (DCIS). HER-2/neu expression was determined by FISH. HER-2/neu-specific T cell responses were determined by using IFN-γ ELISA. Expression of IFN-γ Rα in the tumors was determined by immunohistochemistry analysis of paraffin-embedded tissues. RESULTS We determined that majority of (10 of 12) patients with HER-2/neu negative breast cancer had HER-2/neu-specific IFN-γ producing T cell responses which was stronger than those in patients with HER-2/neu positive tumors. Such immune responses were associated with nuclear translocation of IFN-γ Rα in their tumor cells. Patient with DCIS also showed HER-2/neu-specific T cell responses. CONCLUSION These data suggest that conducting retrospective studies in patients with HER-2/neu negative breast cancers and prospective studies in patients with HER-2/neu positive DCIS can determine whether HER-2/neu negative invasive carcinomas arise from HER-2/neu positive DCIS under the immune pressure.
Collapse
Affiliation(s)
- Maciej Kmieciak
- Department of Microbiology & Immunology, Virginia Commonwealth University Massey Cancer Center, 401 College Street, Richmond VA 23298, USA
| | - Kyle K Payne
- Department of Microbiology & Immunology, Virginia Commonwealth University Massey Cancer Center, 401 College Street, Richmond VA 23298, USA
| | - Michael O Idowu
- Department of Pathology, Virginia Commonwealth University Massey Cancer Center, 1200 E. Marshall Street, Richmond VA 980662, USA
| | - Margaret M Grimes
- Department of Pathology, Virginia Commonwealth University Massey Cancer Center, 1200 E. Marshall Street, Richmond VA 980662, USA
| | - Laura Graham
- Department of Surgery, Virginia Commonwealth University Massey Cancer Center, 1200 E. Broad Street, Richmond VA 980011, USA
| | - Maria-Libera Ascierto
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Center for Human Immunology (CHI), National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Center for Human Immunology (CHI), National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Virginia Commonwealth University Massey Cancer Center, 401 College Street, Richmond VA 23298, USA
| | - Harry D Bear
- Department of Surgery, Virginia Commonwealth University Massey Cancer Center, 1200 E. Broad Street, Richmond VA 980011, USA
| | - Masoud H Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University Massey Cancer Center, 401 College Street, Richmond VA 23298, USA
| |
Collapse
|
21
|
Seubert CM, Stritzker J, Hess M, Donat U, Sturm JB, Chen N, Hof JMV, Krewer B, Tietze LF, Gentschev I, Szalay AA. Enhanced tumor therapy using vaccinia virus strain GLV-1h68 in combination with a β-galactosidase-activatable prodrug seco-analog of duocarmycin SA. Cancer Gene Ther 2011; 18:42-52. [PMID: 20829890 PMCID: PMC3007590 DOI: 10.1038/cgt.2010.49] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2009] [Revised: 05/10/2010] [Accepted: 07/19/2010] [Indexed: 12/24/2022]
Abstract
Breast cancer is the most common cause of cancer-related death worldwide, thus remaining a crucial health problem among women despite advances in conventional therapy. Therefore, new alternative strategies are needed for effective diagnosis and treatment. One approach is the use of oncolytic viruses for gene-directed enzyme prodrug therapy. Here, the lacZ-carrying vaccinia virus (VACV) strain GLV-1h68 was used in combination with a β-galactosidase-activatable prodrug derived from a seco-analog of the natural antibiotic duocarmycin SA. Tumor cell infection with the VACV strain GLV-1h68 led to production of β-galactosidase, essential for the conversion of the prodrug to the toxic compound. Furthermore, drug-dependent cell kill and induction of the intrinsic apoptosis pathway in tumor cells was also observed on combination therapy using the prodrug and the GLV-1h68 strain, despite the fact that VACV strains encode antiapoptotic proteins. Moreover, GI-101A breast cancer xenografts were effectively treated by the combination therapy. In conclusion, the combination of a β-galactosidase-activatable prodrug with a tumor-specific vaccinica virus strain encoding this enzyme, induced apoptosis in cultures of the human GI-101A breast cancer cells, in which a synergistic oncolytic effect was observed. Moreover, in vivo, additional prodrug treatment had beneficial effects on tumor regression in GLV-1h68-treated GI-101A-xenografted mice.
Collapse
Affiliation(s)
- C M Seubert
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany
| | - J Stritzker
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany
- Genelux Corporation, San Diego, CA, USA
| | - M Hess
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany
| | - U Donat
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany
| | - J B Sturm
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany
| | - N Chen
- Genelux Corporation, San Diego, CA, USA
| | - J M von Hof
- Institut für Organische und Biomolekulare Chemie, Georg-August-Universität Göttingen, Göttingen, Germany
| | - B Krewer
- Institut für Organische und Biomolekulare Chemie, Georg-August-Universität Göttingen, Göttingen, Germany
| | - L F Tietze
- Institut für Organische und Biomolekulare Chemie, Georg-August-Universität Göttingen, Göttingen, Germany
| | - I Gentschev
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany
- Genelux Corporation, San Diego, CA, USA
| | - A A Szalay
- Department of Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany
- Genelux Corporation, San Diego, CA, USA
- Department of Radiation Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
22
|
Klinke DJ. A multiscale systems perspective on cancer, immunotherapy, and Interleukin-12. Mol Cancer 2010; 9:242. [PMID: 20843320 PMCID: PMC3243044 DOI: 10.1186/1476-4598-9-242] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Accepted: 09/15/2010] [Indexed: 12/05/2022] Open
Abstract
Monoclonal antibodies represent some of the most promising molecular targeted immunotherapies. However, understanding mechanisms by which tumors evade elimination by the immune system of the host presents a significant challenge for developing effective cancer immunotherapies. The interaction of cancer cells with the host is a complex process that is distributed across a variety of time and length scales. The time scales range from the dynamics of protein refolding (i.e., microseconds) to the dynamics of disease progression (i.e., years). The length scales span the farthest reaches of the human body (i.e., meters) down to the range of molecular interactions (i.e., nanometers). Limited ranges of time and length scales are used experimentally to observe and quantify changes in physiology due to cancer. Translating knowledge obtained from the limited scales observed experimentally to predict patient response is an essential prerequisite for the rational design of cancer immunotherapies that improve clinical outcomes. In studying multiscale systems, engineers use systems analysis and design to identify important components in a complex system and to test conceptual understanding of the integrated system behavior using simulation. The objective of this review is to summarize interactions between the tumor and cell-mediated immunity from a multiscale perspective. Interleukin-12 and its role in coordinating antibody-dependent cell-mediated cytotoxicity is used illustrate the different time and length scale that underpin cancer immunoediting. An underlying theme in this review is the potential role that simulation can play in translating knowledge across scales.
Collapse
Affiliation(s)
- David J Klinke
- Department of Chemical Engineering and Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506-6102, USA.
| |
Collapse
|
23
|
Pakravan N, Soudi S, Hassan ZM. N-terminally fusion of Her2/neu to HSP70 decreases efficiency of Her2/neu DNA vaccine. Cell Stress Chaperones 2010; 15:631-8. [PMID: 20224916 PMCID: PMC3006617 DOI: 10.1007/s12192-010-0175-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Revised: 01/27/2010] [Accepted: 01/29/2010] [Indexed: 01/23/2023] Open
Abstract
DNA vaccines consisted of tumor-associated antigen (TAA) are well suited for immunotherapy against tumor. The construct can contain TAA fused to an appropriate molecule (biologic adjuvant) to improve the efficacy of anti-tumor immune response. Heat shock protein 70 (HSP70) has been shown to be an excellent candidate, capable of cross-priming TAA by antigen presenting cells leading to a robust T-cell response. However, the relationship between strong T-cell responses and tumor rejection is not always mutually exclusive, for which TAA loss or activation of suppressive mechanisms may occur. HSP70 fused to downstream of Her2/neu as DNA vaccine has been shown to be efficient against Her2-expressing tumors. In this study, we examined if N-terminally fusion of Her2/neu to HSP70 could also improve efficiency of Her2/neu DNA vaccine. Therefore, mice with an established Her2/neu expressing tumor were immunized with DNA vaccine consisting of extracellular and trans-membrane domain (EC+TM) of rat Her2/neu alone or N-terminally fused to HSP70 and immune response was evaluated. Administration of rat Her2/neu led to partial control of tumor progression. Surprisingly, fusion of HSP70 to N-terminal of rat Her2/neu led to tumor progression. Our result proposes that fusion direction of biologic adjuvant is an important consideration when Her2/neu is used.
Collapse
Affiliation(s)
- Nafiseh Pakravan
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Ale-Ahmad Avenue, P.O. Box 14115-331, Tehran, Iran.
| | | | | |
Collapse
|
24
|
CD4+ T cells inhibit the neu-specific CD8+ T-cell exhaustion during the priming phase of immune responses against breast cancer. Breast Cancer Res Treat 2010; 126:385-94. [PMID: 20480224 DOI: 10.1007/s10549-010-0942-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Accepted: 05/06/2010] [Indexed: 10/24/2022]
Abstract
Studies conducted in animal model of infectious diseases or H-Y antigen model suggest a crucial role for CD4+ T cells in providing help for CD8+ T-cell memory responses. This concept suggests that inclusion of T helper epitopes in vaccine formulation will result in improved CD8+ T-cell responses. Although this concept has been applied to cancer vaccine design, the role of CD4+ T cells in the memory differentiation of CD8+ T cells and retention of their anti-tumor function have never been tested in breast cancer model. Using the FVB mouse model of neu-positive breast carcinoma we report for the first time that helpless T cells showed cytostatic or tumor inhibitory effects during primary tumor challenge whereas, helped T cells showed cytotoxic effects and resulted in complete tumor rejection. Such differential effects, in vivo, were associated with higher frequency of CD8+PD-L1+ and CD8+PD-1+ T cells in animals harboring helpless T cells as well as higher titer of IL-2 in the sera of animals harboring helped T cells. However, depletion of CD4+ T cells did not alter the ability of neu-specific CD8+ T cells to differentiate into memory cells and to retain their effector function against the tumor during recall challenge. These results suggest the inhibitory role of CD4+ T cells on CD8+ T-cell exhaustion without substantial effects on the differentiation of memory T cells during priming phase of the immune responses against breast cancer.
Collapse
|
25
|
Abe F, Dafferner AJ, Donkor M, Westphal SN, Scholar EM, Solheim JC, Singh RK, Hoke TA, Talmadge JE. Myeloid-derived suppressor cells in mammary tumor progression in FVB Neu transgenic mice. Cancer Immunol Immunother 2010; 59:47-62. [PMID: 19449184 PMCID: PMC11030983 DOI: 10.1007/s00262-009-0719-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Accepted: 04/22/2009] [Indexed: 01/13/2023]
Abstract
Female mice transgenic for the rat proto-oncogene c-erb-B2, under control of the mouse mammary tumor virus (MMTV) promoter (neuN), spontaneously develop metastatic mammary carcinomas. The development of these mammary tumors is associated with increased number of GR-1(+)CD11b(+) myeloid derived suppressor cells (MDSCs) in the peripheral blood (PB), spleen and tumor. We report a complex relationship between tumor growth, MDSCs and immune regulatory molecules in non-mutated neu transgenic mice on a FVB background (FVB-neuN). The first and second tumors in FVB-neuN mice develop at a median of 265 (147-579) and 329 (161-523) days, respectively, resulting in a median survival time (MST) of 432 (201 to >500) days. During tumor growth, significantly increased number of MDSCs is observed in the PB and spleen, as well as, in infiltrating the mammary tumors. Our results demonstrate a direct correlation between tumor size and the number of MDSCs infiltrating the tumor and an inverse relationship between the frequency of CD4(+) T-cells and MDSCs in the spleen. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) assessment of enzyme and cytokine transcript levels in the spleen, tumor, tumor-infiltrating non-parenchymal cells (NPCs) and mammary glands revealed a significant increase in transcript levels from grossly normal mammary glands and tumor-infiltrating NPCs during tumor progression. Tumor NPCs, as compared to spleen cells from wild-type (w/t) mice, expressed significantly higher levels of arginase-1 (ARG-1), nitric oxide synthase (NOS-2), vascular endothelial growth factor (VEGF-A) and significantly lower levels of interferon (IFN)-gamma, interleukin (IL)-2 and fms-like tyrosine kinase-3 ligand (Flt3L) transcript levels. Transcript levels in the spleens of tumor-bearing (TB) mice also differed from normal mice, although to a lesser extent than transcript levels from tumor-infiltrating NPCs. Furthermore, both spleen cells and NPCs from TB mice, but not control mice, suppressed alloantigen responses by syngeneic control spleen cells. Correlative studies revealed that the number of MDSCs in the spleen was directly associated with granulocyte colony stimulating factor (G-CSF) transcript levels in the spleen; while the number of MDSCs in the tumors was directly correlated with splenic granulocyte macrophage stimulating factor (GM-CSF) transcript levels, tumor volume and tumor cell number. Together our results support a role for MDSCs in tumor initiation and progressive, T-cell depression and loss of function provide evidence which support multiple mechanisms of MDSC expansion in a site-dependent manner.
Collapse
Affiliation(s)
- Fuminori Abe
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| | - Alicia J. Dafferner
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| | - Moses Donkor
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| | - Sherry N. Westphal
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| | - Eric M. Scholar
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| | - Joyce C. Solheim
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| | - Rakesh K. Singh
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| | - Traci A. Hoke
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| | - James E. Talmadge
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| |
Collapse
|
26
|
Zhao Y, Wang QJ, Yang S, Kochenderfer JN, Zheng Z, Zhong X, Sadelain M, Eshhar Z, Rosenberg SA, Morgan RA. A herceptin-based chimeric antigen receptor with modified signaling domains leads to enhanced survival of transduced T lymphocytes and antitumor activity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 183:5563-74. [PMID: 19843940 PMCID: PMC6292203 DOI: 10.4049/jimmunol.0900447] [Citation(s) in RCA: 230] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
To generate chimeric Ag receptors (CARs) for the adoptive immunotherapy of cancer patients with ErbB2-expressing tumors, a single-chain Ab derived from the humanized mAb 4D5 Herceptin (trastuzumab) was initially linked to T cell signaling domains derived from CD28 and the CD3zeta to generate a CAR against ErbB2. Human PBLs expressing the 4D5 CAR demonstrated Ag-specific activities against ErbB2(+) tumors. However, a gradual loss of transgene expression was noted for PBLs transduced with this 4D5 CAR. When the CD3zeta signaling domain of the CAR was truncated or mutated, loss of CAR expression was not observed, suggesting that the CD3zeta signaling caused the transgene decrease, which was supported by the finding that T cells expressing 4D5 CARs with CD3zeta ITAM mutations were less prone to apoptosis. By adding 4-1BB cytoplasmic domains to the CD28-CD3zeta signaling moieties, we found increased transgene persistence in 4D5 CAR-transduced PBLs. Furthermore, constructs with 4-1BB sequences demonstrated increased cytokine secretion and lytic activity in 4D5 CAR-transduced T cells. More importantly, PBLs expressing this new version of the 4D5 CAR could not only efficiently lyse the autologous fresh tumor digests, but they could strongly suppress tumor growth in a xenogenic mouse model.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents/chemical synthesis
- Antineoplastic Agents/therapeutic use
- Cell Line
- Cell Line, Tumor
- Cell Survival/genetics
- Cell Survival/immunology
- Coculture Techniques
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Immunotherapy, Adoptive/methods
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/therapy
- Mice
- Mice, SCID
- Protein Structure, Tertiary/genetics
- Receptor, ErbB-2/biosynthesis
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Receptors, Antigen, T-Cell/biosynthesis
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/therapeutic use
- Recombinant Fusion Proteins/chemical synthesis
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/therapeutic use
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
- Transduction, Genetic
- Trastuzumab
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Yangbing Zhao
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Qiong J. Wang
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Shicheng Yang
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - James N. Kochenderfer
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Zhili Zheng
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Xiaosong Zhong
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Michel Sadelain
- Center for Cell Engineering, Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10021
| | - Zelig Eshhar
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Steven A. Rosenberg
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Richard A. Morgan
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
27
|
Vo DD, Prins RM, Begley JL, Donahue TR, Morris LF, Bruhn KW, de la Rocha P, Yang MY, Mok S, Garban HJ, Craft N, Economou JS, Marincola FM, Wang E, Ribas A. Enhanced antitumor activity induced by adoptive T-cell transfer and adjunctive use of the histone deacetylase inhibitor LAQ824. Cancer Res 2009; 69:8693-9. [PMID: 19861533 DOI: 10.1158/0008-5472.can-09-1456] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumors grow in the presence of antigen-specific T cells, suggesting the existence of intrinsic cancer cell escape mechanisms. We hypothesized that a histone deacetylase (HDAC) inhibitor could sensitize tumor cells to immunotherapy because this class of agents has been reported to increase tumor antigen expression and shift gene expression to a proapoptotic milieu in cancer cells. To test this question, we treated B16 murine melanoma with the combination of the HDAC inhibitor LAQ824 and the adoptive transfer of gp100 melanoma antigen-specific pmel-1 T cells. The combined therapy significantly improved antitumor activity through several mechanisms: (a) increase in MHC and tumor-associated antigen expression by tumor cells; (b) decrease in competing endogenous lymphocytes in recipient mice, resulting in a proliferative advantage for the adoptively transferred cells; and (c) improvement in the functional activity of the adoptively transferred lymphocytes. We confirmed the beneficial effects of this HDAC inhibitor as a sensitizer to immunotherapy in a different model of prophylactic prime-boost vaccination with the melanoma antigen tyrosinase-related protein 2, which also showed a significant improvement in antitumor activity against B16 melanoma. In conclusion, the HDAC inhibitor LAQ824 significantly enhances tumor immunotherapy through effects on target tumor cells as well as improving the antitumor activity of tumor antigen-specific lymphocytes.
Collapse
Affiliation(s)
- Dan D Vo
- Department of Medicine, Division of Hematology/Oncology, Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Parajuli N, Müller-Holzner E, Böck G, Werner ER, Villunger A, Doppler W. Infiltrating CD11b+CD11c+cells have the potential to mediate inducible nitric oxide synthase-dependent cell death in mammary carcinomas of HER-2/neu transgenic mice. Int J Cancer 2009; 126:896-908. [DOI: 10.1002/ijc.24805] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
29
|
Worschech A, Chen N, Yu YA, Zhang Q, Pos Z, Weibel S, Raab V, Sabatino M, Monaco A, Liu H, Monsurró V, Buller RM, Stroncek DF, Wang E, Szalay AA, Marincola FM. Systemic treatment of xenografts with vaccinia virus GLV-1h68 reveals the immunologic facet of oncolytic therapy. BMC Genomics 2009; 10:301. [PMID: 19583830 PMCID: PMC2713268 DOI: 10.1186/1471-2164-10-301] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Accepted: 07/07/2009] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND GLV-1h68 is an attenuated recombinant vaccinia virus (VACV) that selectively colonizes established human xenografts inducing their complete regression. RESULTS Here, we explored xenograft/VACV/host interactions in vivo adopting organism-specific expression arrays and tumor cell/VACV in vitro comparing VACV replication patterns. There were no clear-cut differences in vitro among responding and non-responding tumors, however, tumor rejection was associated in vivo with activation of interferon-stimulated genes (ISGs) and innate immune host's effector functions (IEFs) correlating with VACV colonization of the xenografts. These signatures precisely reproduce those observed in humans during immune-mediated tissue-specific destruction (TSD) that causes tumor or allograft rejection, autoimmunity or clearance of pathogens. We recently defined these common pathways in the "immunologic constant of rejection" hypothesis (ICR). CONCLUSION This study provides the first prospective validation of a universal mechanism associated with TSD. Thus, xenograft infection by oncolytic VACV, beyond offering a promising therapy of established cancers, may represent a reliable pre-clinical model to test therapeutic strategies aimed at modulating the central pathways leading to TSD; this information may lead to the identification of principles that could refine the treatment of cancer and chronic infection by immune stimulation or autoimmunity and allograft rejection through immune tolerance.
Collapse
Affiliation(s)
- Andrea Worschech
- Genelux Corporation, San Diego Science Center, San Diego, California, USA
- Virchow Center for Experimental Biomedicine and Institute for Biochemistry, University of Würzburg, Am Hubland, Würzburg, Germany
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Nanhai Chen
- Genelux Corporation, San Diego Science Center, San Diego, California, USA
| | - Yong A Yu
- Genelux Corporation, San Diego Science Center, San Diego, California, USA
| | - Qian Zhang
- Genelux Corporation, San Diego Science Center, San Diego, California, USA
| | - Zoltan Pos
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephanie Weibel
- Genelux Corporation, San Diego Science Center, San Diego, California, USA
- Virchow Center for Experimental Biomedicine and Institute for Biochemistry, University of Würzburg, Am Hubland, Würzburg, Germany
| | - Viktoria Raab
- Genelux Corporation, San Diego Science Center, San Diego, California, USA
- Virchow Center for Experimental Biomedicine and Institute for Biochemistry, University of Würzburg, Am Hubland, Würzburg, Germany
| | - Marianna Sabatino
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Alessandro Monaco
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Hui Liu
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Vladia Monsurró
- Department of Pathology, Immunology Section, University of Verona Medical School, Verona, Italy
| | - R Mark Buller
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St Louis, MO, USA
| | - David F Stroncek
- Cellular Processing Section, Department of Transfusion Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Aladar A Szalay
- Genelux Corporation, San Diego Science Center, San Diego, California, USA
- Virchow Center for Experimental Biomedicine and Institute for Biochemistry, University of Würzburg, Am Hubland, Würzburg, Germany
| | - Francesco M Marincola
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
30
|
Santisteban M, Reiman JM, Asiedu MK, Behrens MD, Nassar A, Kalli KR, Haluska P, Ingle JN, Hartmann LC, Manjili MH, Radisky DC, Ferrone S, Knutson KL. Immune-induced epithelial to mesenchymal transition in vivo generates breast cancer stem cells. Cancer Res 2009; 69:2887-95. [PMID: 19276366 DOI: 10.1158/0008-5472.can-08-3343] [Citation(s) in RCA: 306] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The breast cancer stem cell (BCSC) hypotheses suggest that breast cancer is derived from a single tumor-initiating cell with stem-like properties, but the source of these cells is unclear. We previously observed that induction of an immune response against an epithelial breast cancer led in vivo to the T-cell-dependent outgrowth of a tumor, the cells of which had undergone epithelial to mesenchymal transition (EMT). The resulting mesenchymal tumor cells had a CD24(-/lo)CD44(+) phenotype, consistent with BCSCs. In the present study, we found that EMT was induced by CD8 T cells and the resulting tumors had characteristics of BCSCs, including potent tumorigenicity, ability to reestablish an epithelial tumor, and enhanced resistance to drugs and radiation. In contrast to the hierarchal cancer stem cell hypothesis, which suggests that breast cancer arises from the transformation of a resident tissue stem cell, our results show that EMT can produce the BCSC phenotype. These findings have several important implications related to disease progression and relapse.
Collapse
Affiliation(s)
- Marta Santisteban
- Department of Oncology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ney JT, Schmidt T, Kurts C, Zhou Q, Eckert D, Felsher DW, Schorle H, Knolle P, Tüting T, Barchet W, Büttner R, Limmer A, Gütgemann I. Autochthonous liver tumors induce systemic T cell tolerance associated with T cell receptor down-modulation. Hepatology 2009; 49:471-81. [PMID: 19105207 DOI: 10.1002/hep.22652] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The reason the adaptive immune system fails in advanced liver tumors is largely unclear. To address this question, we have developed a novel murine model that combines c-myc-induced autochthonous tumorigenesis with expression of a cognate antigen, ovalbumin (OVA). When c-myc/OVA transgenic mice were crossed with liver-specific inducer mice, multifocal hepatocellular carcinomas co-expressing OVA developed in a tetracycline-dependent manner with a short latency and 100% penetrance. Transferred OVA-specific T cells, although infiltrating the tumor at high numbers, were hyporesponsive, as evidenced by a lack of in vivo cytotoxicity and interferon gamma production. This allowed the tumor to progress even in the presence of large numbers of antigen-specific T cells and even after vaccination (OVA+CpG-DNA). Interestingly, T cell receptor down-modulation was observed, which may explain antigen-specific hyporesponsiveness. This model is helpful in understanding liver cancer-specific mechanisms of T cell tolerance and dissection of antigen-specific and nonspecific mechanisms of immunotherapies in the preclinical phase.
Collapse
Affiliation(s)
- Jasmin T Ney
- Department of Pathology, University of Bonn, Bonn, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Adoptive transfer of HER2/neu-specific T cells expanded with alternating gamma chain cytokines mediate tumor regression when combined with the depletion of myeloid-derived suppressor cells. Cancer Immunol Immunother 2008; 58:941-53. [PMID: 18979098 DOI: 10.1007/s00262-008-0609-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 10/01/2008] [Accepted: 10/03/2008] [Indexed: 12/12/2022]
Abstract
Adoptive immunotherapy (AIT) using ex vivo-expanded HER-2/neu-specific T cells has shown initial promising results against disseminated tumor cells in the bone marrow. However, it has failed to promote objective responses against primary tumors. We report for the first time that alternating gamma chain cytokines (IL-2, IL-7 and IL-15) ex vivo can expand the neu-specific lymphocytes that can kill breast tumors in vitro. However, the anti-tumor efficacy of these neu-specific T cells was compromised by the increased levels of myeloid-derived suppressor cells (MDSC) during the premalignant stage in FVBN202 transgenic mouse model of breast carcinoma. Combination of AIT with the depletion of MDSC, in vivo, resulted in the regression of neu positive primary tumors. Importantly, neu-specific antibody responses were restored only when AIT was combined with the depletion of MDSC. In vitro studies determined that MDSC caused inhibition of T cell proliferation in a contact-dependent manner. Together, these results suggest that combination of AIT with depletion or inhibition of MDSC could lead to the regression of mammary tumors.
Collapse
|
33
|
Manjili MH, Kmieciak M. Does HER-2/neu antigen loss in metastatic breast tumors occur under immune pressure? Int J Cancer 2008; 123:1476-7; author reply 1478-9. [PMID: 18566999 PMCID: PMC3732034 DOI: 10.1002/ijc.23663] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
34
|
Radiofrequency thermal ablation of breast tumors combined with intralesional administration of IL-7 and IL-15 augments anti-tumor immune responses and inhibits tumor development and metastasis. Breast Cancer Res Treat 2008; 114:423-31. [PMID: 18425677 DOI: 10.1007/s10549-008-0024-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2008] [Accepted: 04/10/2008] [Indexed: 01/27/2023]
Abstract
Tumor development or recurrence is always a matter of concern following radiofrequency thermal ablation (RFA) of tumors. To determine whether combining RFA with immunologically active cytokines might induce tumor-specific immune responses against mammary carcinoma and inhibit tumor development or metastasis, we evaluated intralesional injection of IL-7 and IL-15 in RFA-treated murine tumors. We used two different breast carcinoma models: neu-overexpressing mouse mammary carcinoma (MMC) in FVBN202 transgenic mouse and 4T1 tumors in Balb/c mouse. MMC tend to relapse even in the presence of neu-specific immune responses, and 4T1 is a weakly immunogenic, aggressive and highly metastatic transplantable tumor. In vivo growth of both of these tumors is also associated with increased numbers of CD11b+Gr1+ myeloid-derived suppressor cells (MDSC). We showed for the first time that unlike RFA alone, RFA combined with the administration of intralesional IL-7 and IL-15 (after RFA), induced immune responses to tumors, inhibited tumor development and lung metastasis, and reduced MDSC.
Collapse
|