301
|
Ferrari N, McDonald L, Morris JS, Cameron ER, Blyth K. RUNX2 in mammary gland development and breast cancer. J Cell Physiol 2013; 228:1137-42. [PMID: 23169547 DOI: 10.1002/jcp.24285] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 11/06/2012] [Indexed: 12/17/2022]
Abstract
Runx2 is best known as an essential factor in osteoblast differentiation and bone development but, like many other transcription factors involved in development, is known to operate over a much wider tissue range. Our understanding of these other aspects of Runx2 function is still at a relatively early stage and the importance of its role in cell fate decisions and lineage maintenance in non-osseous tissues is only beginning to emerge. One such tissue is the mammary gland, where Runx2 is known to be expressed and participate in the regulation of mammary specific genes. Furthermore, differential and temporal expression of this gene is observed during mammary epithelial differentiation in vivo, strongly indicative of an important functional role. Although the precise nature of that role remains elusive, preliminary evidence hints at possible involvement in the regulation of mammary stem and/or progenitor cells. As with many genes important in regulating cell fate, RUNX2 has also been linked to metastatic cancer where in some established breast cell lines, retention of expression is associated with a more invasive phenotype. More recently, expression analysis has been extended to primary breast cancers where high levels of RUNX2 align with a specific subtype of the disease. That RUNX2 expression correlates with the so called "Triple Negative" subtype is particularly interesting given the known cross talk between Runx2 and estrogen receptor signaling pathways. This review summaries our current understanding of Runx2 in mammary gland development and cancer, and postulates a role that may link both these processes.
Collapse
Affiliation(s)
- Nicola Ferrari
- The Beatson Institute for Cancer Research, Bearsden, Glasgow, UK
| | | | | | | | | |
Collapse
|
302
|
Raz Y, Erez N. An inflammatory vicious cycle: Fibroblasts and immune cell recruitment in cancer. Exp Cell Res 2013; 319:1596-603. [PMID: 23567181 DOI: 10.1016/j.yexcr.2013.03.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 03/12/2013] [Accepted: 03/16/2013] [Indexed: 01/07/2023]
Abstract
Cancer-associated fibroblasts (CAFs) have been established as a key component of the crosstalk between tumor cells and their microenvironment. The ability of CAFs to orchestrate tumor-promoting inflammation is central to their role in facilitating tumor growth, invasion, and metastasis. Here we review pathways by which CAFs and their soluble mediators provide multiple complex signals that modulate the recruitment, functional activation status, and retention of immune cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Yael Raz
- Lis Maternity Hospital, Tel Aviv Sorasky Medical Center, affiliated to Sackler School of Medicine, Tel Aviv, Israel; Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 69978, Israel
| | - Neta Erez
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 69978, Israel.
| |
Collapse
|
303
|
Phan-Lai V, Florczyk SJ, Kievit FM, Wang K, Gad E, Disis ML, Zhang M. Three-dimensional scaffolds to evaluate tumor associated fibroblast-mediated suppression of breast tumor specific T cells. Biomacromolecules 2013; 14:1330-7. [PMID: 23517456 DOI: 10.1021/bm301928u] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In the tumor microenvironment, the signals from tumor-associated fibroblasts (TAF) that suppress antitumor immunity remain unclear. Here, we develop and investigate an in vitro three-dimensional (3D) scaffold model for the novel evaluation of TAF interaction with breast tumor cells and breast specific, neu antigen (p98) reactive T cells. Breast cancer cells seeded on 3D chitosan-alginate (CA) scaffolds showed productive growth and formed distinct tumor spheroids. Antigen specific p98 T cells, but not naïve T cells, bound significantly better to tumor cells on scaffolds. The p98 T cells induced potent tumor cell killing but T helper cell cytokine function was impaired in the presence of TAF coseeding on scaffolds. We found that the immunosuppression was mediated, in part, by transforming growth factor beta (TGF-b) and interleukin-10 (IL-10). Therefore, TAF appear capable of inducing potent T cell suppression. CA scaffolds can provide clinically relevant findings prior to preclinical testing of novel immunotherapies.
Collapse
Affiliation(s)
- Vy Phan-Lai
- Department of Medicine, University of Washington, Seattle, Washington 98109, United States
| | | | | | | | | | | | | |
Collapse
|
304
|
The metastasis-promoting roles of tumor-associated immune cells. J Mol Med (Berl) 2013; 91:411-29. [PMID: 23515621 DOI: 10.1007/s00109-013-1021-5] [Citation(s) in RCA: 277] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 03/06/2013] [Accepted: 03/07/2013] [Indexed: 12/12/2022]
Abstract
Tumor metastasis is driven not only by the accumulation of intrinsic alterations in malignant cells, but also by the interactions of cancer cells with various stromal cell components of the tumor microenvironment. In particular, inflammation and infiltration of the tumor tissue by host immune cells, such as tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells, have been shown to support tumor growth in addition to invasion and metastasis. Each step of tumor development, from initiation through metastatic spread, is promoted by communication between tumor and immune cells via the secretion of cytokines, growth factors, and proteases that remodel the tumor microenvironment. Invasion and metastasis require neovascularization, breakdown of the basement membrane, and remodeling of the extracellular matrix for tumor cell invasion and extravasation into the blood and lymphatic vessels. The subsequent dissemination of tumor cells to distant organ sites necessitates a treacherous journey through the vasculature, which is fostered by close association with platelets and macrophages. Additionally, the establishment of the pre-metastatic niche and specific metastasis organ tropism is fostered by neutrophils and bone marrow-derived hematopoietic immune progenitor cells and other inflammatory cytokines derived from tumor and immune cells, which alter the local environment of the tissue to promote adhesion of circulating tumor cells. This review focuses on the interactions between tumor cells and immune cells recruited to the tumor microenvironment and examines the factors allowing these cells to promote each stage of metastasis.
Collapse
|
305
|
Targeting CXCL12/CXCR4 signaling with oncolytic virotherapy disrupts tumor vasculature and inhibits breast cancer metastases. Proc Natl Acad Sci U S A 2013; 110:E1291-300. [PMID: 23509246 DOI: 10.1073/pnas.1220580110] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Oncolytic viruses hold promise for the treatment of cancer, but their interaction with the tumor microenvironment needs to be elucidated for optimal tumor cell killing. Because the CXCR4 receptor for the stromal cell-derived factor-1 (SDF-1/CXCL12) chemokine is one of the key stimuli involved in signaling interactions between tumor cells and their stromal microenvironment, we used oncolytic virotherapy with a CXCR4 antagonist to target the CXCL12/CXCR4 signaling axis in a triple-negative 4T1 breast carcinoma in syngeneic mice. We show here that CXCR4 antagonist expression from an oncolytic vaccinia virus delivered intravenously to mice with orthotopic tumors attains higher intratumoral concentration than its soluble counterpart and exhibits increased efficacy over that mediated by oncolysis alone. A systemic delivery of the armed virus after resection of the primary tumor was efficacious in inhibiting the development of spontaneous metastasis and increased overall tumor-free survival. Inhibition of tumor growth with the armed virus was associated with destruction of tumor vasculature, reductions in expression of CXCL12 and VEGF, and decrease in intratumoral numbers of bone marrow-derived endothelial and myeloid cells. These changes led to induction of antitumor antibody responses and resistance to tumor rechallenge. Engineering an oncolytic virus armed with a CXCR4 antagonist represents an innovative strategy that targets multiple elements within the tumor microenvironment. As such, this approach could have a significant therapeutic impact against primary and metastatic breast cancer.
Collapse
|
306
|
Kong BH, Shin HD, Kim SH, Mok HS, Shim JK, Lee JH, Shin HJ, Huh YM, Kim EH, Park EK, Chang JH, Kim DS, Hong YK, Kim SH, Lee SJ, Kang SG. Increased in vivo angiogenic effect of glioma stromal mesenchymal stem-like cells on glioma cancer stem cells from patients with glioblastoma. Int J Oncol 2013; 42:1754-62. [PMID: 23483121 DOI: 10.3892/ijo.2013.1856] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 02/21/2013] [Indexed: 11/06/2022] Open
Abstract
The presence of glioma stromal mesenchymal stem‑like cells (GS-MSLCs) in tumors from glioma patients has been previously reported. The mechanisms through which these cells function as a part of the glioma microenvironment, however, remain incompletely understood. We investigated the biological effects of GS-MSLCs on glioma cancer stem cells (gCSCs), testing the hypothesis that GS-MSLCs alter the biological characteristics of gCSCs. GS-MSLCs and gCSCs were isolated from different glioblastoma (GBM) specimens obtained from patients. In in vitro experiments, gCSCs were cultured alone or co-cultured with GS-MSLCs, and gCSCs cell counts were compared between the two groups. In addition, two groups of orthotopic GBM xenografts in mice were created, one using gCSCs from the monoculture group and one using gCSCs isolated from the co-culture group, and tumor volume and survival were analyzed. Furthermore, in vivo proliferation, apoptosis and vessel formation were examined using immunohistochemical analyses. In vitro cell counts for gCSCs co-cultured with GS-MSLCs increased 3-fold compared to gCSCs cultured alone. In orthotopic xenograft experiments, mice injected with gCSCs isolated from the co-culture group had significantly larger tumor volume, measured on day 40 after injection, and their survival times were shorter. Immunohistochemical analysis showed increased tumor expression of CD31, indicative of enhanced microvessel formation in mice injected with gCSCs co-cultured with GS-MSLCs compared to mice injected with gCSCs cultured alone. However, proliferation (PCNA) and apoptosis (TUNEL) markers showed no significant difference between the two groups. In conclusion, GS-MSLCs may influence the biological properties of gCSCs, shifting them towards a more aggressive status; moreover, increased angiogenesis may be a critical component of this mechanism.
Collapse
Affiliation(s)
- Byung Ho Kong
- Department of Medical Science, The Catholic University of Korea College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
307
|
Shipp C, Weide B, Derhovanessian E, Pawelec G. Hsps are up-regulated in melanoma tissue and correlate with patient clinical parameters. Cell Stress Chaperones 2013; 18:145-54. [PMID: 22872370 PMCID: PMC3581625 DOI: 10.1007/s12192-012-0363-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 07/19/2012] [Accepted: 07/23/2012] [Indexed: 12/22/2022] Open
Abstract
Heat shock proteins (hsps) have been studied in numerous cancer types, but a clear view of their clinical relevance in melanoma remains elusive. Therefore, the aim of this study was to investigate the expression of hsps in melanoma with respect to patient clinical parameters. Using Western immunoblotting, hsps 90, 70, 60, 40 and 32 were observed to be widely expressed in metastatic melanomas (n = 31), while immunofluorescence demonstrated that in the majority of samples these hsps, apart from hsp32, were increased in expression in melanoma cells compared with surrounding non-melanoma cells in situ (n = 8). Correlating hsp expression with patient clinical parameters indicated that greater hsp90 (P < 0.02) and hsp40 (P < 0.03) expression correlated with advanced stage (stage III Vs stage IV), while in the case of hsp40, this was additionally associated with reduced patient survival (P < 0.05). In contrast, higher hsp32 expression was associated with improved patient survival (P < 0.007). On the other hand, the expression of the other hsps did not correlate with any obtainable patient clinical parameters. This study provides further evidence for the importance of hsps in melanoma and for their use as therapeutic targets and biomarkers, but larger-scale follow-up studies are required to confirm these results.
Collapse
Affiliation(s)
- Christopher Shipp
- Tübingen Ageing and Tumour Immunology Group, Section for Transplantation Immunology, Department of Internal Medicine II, Centre for Medical Research, University of Tübingen, Waldhörnlestr. 22, 72072 Tübingen, Germany.
| | | | | | | |
Collapse
|
308
|
Murugaiyan G, Saha B. IL-27 in tumor immunity and immunotherapy. Trends Mol Med 2013; 19:108-16. [DOI: 10.1016/j.molmed.2012.12.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Revised: 11/30/2012] [Accepted: 12/07/2012] [Indexed: 01/23/2023]
|
309
|
Serini G, Bussolino F, Maione F, Giraudo E. Class 3 semaphorins: physiological vascular normalizing agents for anti-cancer therapy. J Intern Med 2013. [PMID: 23198760 DOI: 10.1111/joim.12017] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Findings from preclinical and clinical studies show that vascular normalization represents a novel strategy to enhance the efficacy of and overcome the acquired resistance to anti-angiogenic therapies in cancer. Several mechanisms of tumour vessel normalization have been revealed. Amongst them, secreted class 3 semaphorins (Sema3), which regulate axon guidance and angiogenesis, have been recently identified as novel vascular normalizing agents that inhibit metastatic dissemination by restoring vascular function. Here, we discuss the different biological functions and mechanisms of action of Sema3 in the context of tumour vascular normalization, and their impact on the different cellular components of the tumour microenvironment.
Collapse
Affiliation(s)
- G Serini
- Institute for Cancer Research at Candiolo (IRCC), University of Torino, Turin, Italy
| | | | | | | |
Collapse
|
310
|
Carcinoma-associated fibroblasts are a promising therapeutic target. Cancers (Basel) 2013; 5:149-69. [PMID: 24216702 PMCID: PMC3730310 DOI: 10.3390/cancers5010149] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 01/21/2013] [Accepted: 01/23/2013] [Indexed: 12/21/2022] Open
Abstract
Human carcinomas frequently exhibit significant stromal reactions such as the so-called "desmoplastic stroma" or "reactive stroma", which is characterised by the existence of large numbers of stromal cells and extracellular matrix proteins. Carcinoma-associated fibroblasts (CAFs), which are rich in activated fibroblast populations exemplified by myofibroblasts, are among the predominant cell types present within the tumour-associated stroma. Increased numbers of stromal myofibroblasts are often associated with high-grade malignancies with poor prognoses in humans. CAF myofibroblasts possess abilities to promote primary tumour development, growth and progression by stimulating the processes of neoangiogenesis as well as tumour cell proliferation, survival, migration and invasion. Moreover, it has been demonstrated that CAFs serve as a niche supporting the metastatic colonisation of disseminated carcinoma cells in distant organs. Their contribution to primary and secondary malignancies makes these fibroblasts a potential therapeutic target and they also appear to be relevant to the development of drug resistance and tumour recurrence. This review summarises our current knowledge of tumour-promoting CAFs and discusses the therapeutic feasibility of targeting these cells as well as disrupting heterotypic interactions with other cell types in tumours that may improve the efficacy of current anti-tumour therapies.
Collapse
|
311
|
Abstract
Human carcinomas frequently exhibit significant stromal reactions such as the so-called "desmoplastic stroma" or "reactive stroma", which is characterised by the existence of large numbers of stromal cells and extracellular matrix proteins. Carcinoma-associated fibroblasts (CAFs), which are rich in activated fibroblast populations exemplified by myofibroblasts, are among the predominant cell types present within the tumour-associated stroma. Increased numbers of stromal myofibroblasts are often associated with high-grade malignancies with poor prognoses in humans. CAF myofibroblasts possess abilities to promote primary tumour development, growth and progression by stimulating the processes of neoangiogenesis as well as tumour cell proliferation, survival, migration and invasion. Moreover, it has been demonstrated that CAFs serve as a niche supporting the metastatic colonisation of disseminated carcinoma cells in distant organs. Their contribution to primary and secondary malignancies makes these fibroblasts a potential therapeutic target and they also appear to be relevant to the development of drug resistance and tumour recurrence. This review summarises our current knowledge of tumour-promoting CAFs and discusses the therapeutic feasibility of targeting these cells as well as disrupting heterotypic interactions with other cell types in tumours that may improve the efficacy of current anti-tumour therapies.
Collapse
|
312
|
Shibahara Y, Miki Y, Ishida T, Nakamura Y, Suzuki T, Ohuchi N, Sasano H. Immunohistochemical analysis of aromatase in metastatic lymph nodes of breast cancer. Pathol Int 2012; 63:20-8. [DOI: 10.1111/pin.12015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 11/08/2012] [Indexed: 11/30/2022]
Affiliation(s)
- Yukiko Shibahara
- Department of Pathology; Tohoku University Graduate School of Medicine; Sendai; Miyagi; Japan
| | - Yasuhiro Miki
- Department of Pathology; Tohoku University Graduate School of Medicine; Sendai; Miyagi; Japan
| | - Takanori Ishida
- Division of Surgical Oncology; Tohoku University Graduate School of Medicine; Sendai; Miyagi; Japan
| | - Yasuhiro Nakamura
- Department of Pathology; Tohoku University Graduate School of Medicine; Sendai; Miyagi; Japan
| | - Takashi Suzuki
- Department of Pathology; Tohoku University Graduate School of Medicine; Sendai; Miyagi; Japan
| | - Noriaki Ohuchi
- Division of Surgical Oncology; Tohoku University Graduate School of Medicine; Sendai; Miyagi; Japan
| | - Hironobu Sasano
- Department of Pathology; Tohoku University Graduate School of Medicine; Sendai; Miyagi; Japan
| |
Collapse
|
313
|
Angelucci C, Maulucci G, Lama G, Proietti G, Colabianchi A, Papi M, Maiorana A, De Spirito M, Micera A, Balzamino OB, Di Leone A, Masetti R, Sica G. Epithelial-stromal interactions in human breast cancer: effects on adhesion, plasma membrane fluidity and migration speed and directness. PLoS One 2012; 7:e50804. [PMID: 23251387 PMCID: PMC3519494 DOI: 10.1371/journal.pone.0050804] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Accepted: 10/25/2012] [Indexed: 02/04/2023] Open
Abstract
Interactions occurring between malignant cells and the stromal microenvironment heavily influence tumor progression. We investigated whether this cross-talk affects some molecular and functional aspects specifically correlated with the invasive phenotype of breast tumor cells (i.e. adhesion molecule expression, membrane fluidity, migration) by co-culturing mammary cancer cells exhibiting different degrees of metastatic potential (MDA-MB-231>MCF-7) with fibroblasts isolated from breast healthy skin (normal fibroblasts, NFs) or from breast tumor stroma (cancer-associated fibroblasts, CAFs) in 2D or 3D (nodules) cultures. Confocal immunofluorescence analysis of the epithelial adhesion molecule E-cadherin on frozen nodule sections demonstrated that NFs and CAFs, respectively, induced or inhibited its expression in MCF-7 cells. An increase in the mesenchymal adhesion protein N-cadherin was observed in CAFs, but not in NFs, as a result of the interaction with both kinds of cancer cells. CAFs, in turn, promoted N-cadherin up-regulation in MDA-MB-231 cells and its de novo expression in MCF-7 cells. Beyond promotion of “cadherin switching”, another sign of the CAF-triggered epithelial-mesenchymal transition (EMT) was the induction of vimentin expression in MCF-7 cells. Plasma membrane labeling of monolayer cultures with the fluorescent probe Laurdan showed an enhancement of the membrane fluidity in cancer cells co-cultured with NFs or CAFs. An increase in lipid packing density of fibroblast membranes was promoted by MCF-7 cells. Time-lapsed cell tracking analysis of mammary cancer cells co-cultured with NFs or CAFs revealed an enhancement of tumor cell migration velocity, even with a marked increase in the directness induced by CAFs. Our results demonstrate a reciprocal influence of mammary cancer and fibroblasts on various adhesiveness/invasiveness features. Notably, CAFs' ability to promote EMT, reduction of cell adhesion, increase in membrane fluidity, and migration velocity and directness in mammary cancer cells can be viewed as an overall progression- and invasion-promoting effect.
Collapse
Affiliation(s)
- Cristiana Angelucci
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Giuseppe Maulucci
- Istituto di Fisica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Gina Lama
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Gabriella Proietti
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Anna Colabianchi
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Massimiliano Papi
- Istituto di Fisica, Università Cattolica del Sacro Cuore, Roma, Italia
| | | | - Marco De Spirito
- Istituto di Fisica, Università Cattolica del Sacro Cuore, Roma, Italia
- * E-mail:
| | - Alessandra Micera
- Istituto di Ricovero e Cura a Carattere Scientifico - Fondazione G.B. Bietti, Roma, Italia
| | - Omar Bijorn Balzamino
- Istituto di Ricovero e Cura a Carattere Scientifico - Fondazione G.B. Bietti, Roma, Italia
| | - Alba Di Leone
- Dipartimento per la Tutela della Salute della Donna e della Vita Nascente, del Bambino e dell'Adolescente - Unità Operativa di Chirurgia Senologica, Facoltà di Medicina e Chirurgia “A. Gemelli”, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Riccardo Masetti
- Dipartimento per la Tutela della Salute della Donna e della Vita Nascente, del Bambino e dell'Adolescente - Unità Operativa di Chirurgia Senologica, Facoltà di Medicina e Chirurgia “A. Gemelli”, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Gigliola Sica
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Roma, Italia
| |
Collapse
|
314
|
Liu W, Chen JR, Hsu CH, Li YH, Chen YM, Lin CY, Huang SJ, Chang ZK, Chen YC, Lin CH, Gong HY, Lin CC, Kawakami K, Wu JL. A zebrafish model of intrahepatic cholangiocarcinoma by dual expression of hepatitis B virus X and hepatitis C virus core protein in liver. Hepatology 2012; 56:2268-76. [PMID: 22729936 DOI: 10.1002/hep.25914] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2011] [Accepted: 06/09/2012] [Indexed: 12/25/2022]
Abstract
UNLABELLED The mechanisms that mediate the initiation and development of intrahepatic cholangiocarcinoma (ICC) associated with hepatitis B and C virus (HBV and HCV, respectively) infection remain largely unclear. In this study we conditionally coexpressed hepatitis B virus X (HBx) and hepatitis C virus core (HCP) proteins in zebrafish livers, which caused fibrosis and consequently contributed to ICC formation at the age of 3 months. Suppressing the transgene expression by doxycycline (Dox) treatment resulted in the loss of ICC formation. The biomarker networks of zebrafish ICC identified by transcriptome sequencing and analysis were also frequently involved in the development of human neoplasms. The profiles of potential biomarker genes of zebrafish ICC were similar to those of human cholangiocarcinoma. Our data also showed that the pSmad3L oncogenic pathway was activated in HBx and HCP-induced ICC and included phosphorylation of p38 mitogen-activated proteinbase (MAPK) and p44/42 mitogen-activated protein kinase (ERK1/2), indicating the association with transforming growth factor beta 1 (TGF-β1) signaling pathway in ICC. Bile duct proliferation, fibrosis, and ICC were markedly reduced by knockdown of TGF-β1 by in vivo morpholinos injections. CONCLUSION These results reveal that TGF-β1 plays an important role in HBx- and HCP-induced ICC development. This in vivo model is a potential approach to study the molecular events of fibrosis and ICC occurring in HBV and HCV infection.
Collapse
Affiliation(s)
- Wangta Liu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
315
|
Gomes FG, Nedel F, Alves AM, Nör JE, Tarquinio SBC. Tumor angiogenesis and lymphangiogenesis: tumor/endothelial crosstalk and cellular/microenvironmental signaling mechanisms. Life Sci 2012. [PMID: 23178150 DOI: 10.1016/j.lfs.2012.10.008] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumor angiogenesis and lymphangiogenesis are key features of tumor progression and metastasis. The role of tumor cells-derived factors in the promotion of associated angiogenesis and lymphangiogenesis is much studied and, no doubt, very important for the understanding of cancer progression. This review aims to present and discuss the work done on the pro-angiogenic and lymphangiogenic cellular interactions within the tumor microenvironment and the signaling pathways that regulate this crosstalk. Such multifactor studies are critical for the development of future therapeutic approaches for cancer because they take into account the complexities of cellular interactions within the tumor microenvironment.
Collapse
Affiliation(s)
- Fausto Gueths Gomes
- Nucleus of Cellular and Tecidual Biology (NCTBio), Department of Oral Pathology, School of Dentistry, Federal University of Pelotas, Brazil
| | | | | | | | | |
Collapse
|
316
|
Servais C, Erez N. From sentinel cells to inflammatory culprits: cancer-associated fibroblasts in tumour-related inflammation. J Pathol 2012; 229:198-207. [DOI: 10.1002/path.4103] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 08/27/2012] [Accepted: 09/07/2012] [Indexed: 12/11/2022]
Affiliation(s)
- Charlotte Servais
- Department of Pathology, Sackler School of Medicine; Tel Aviv University; Tel-Aviv Israel 69978
| | - Neta Erez
- Department of Pathology, Sackler School of Medicine; Tel Aviv University; Tel-Aviv Israel 69978
| |
Collapse
|
317
|
Abstract
Immunotherapy for solid tumors has shown promise in preclinical as well as early clinical studies. However, its efficacy remains limited. The hindrance to achieving objective, long-lasting therapeutic responses in solid tumors is, in part, mediated by the dynamic nature of the tumor and its complex microenvironment. Tumor-directed therapies fail to eliminate components of the microenvironment, which can reinstate a tumorigenic milieu and contribute to recurrence. Cancer-associated fibroblasts (CAFs) form the most preponderant cell type in the solid tumor microenvironment. Given their pervasive role in facilitating tumor growth and metastatic dissemination, CAFs have emerged as attractive therapeutic targets in the tumor microenvironment. In this article, we highlight the cross-talk between CAFs and cancer cells, and discuss how targeting CAFs has the potential to improve current immunotherapy approaches for cancer.
Collapse
Affiliation(s)
- Sunitha Kakarla
- Center for Cell & Gene Therapy, Texas Children’s Hospital, The Methodist Hospital, Baylor College of Medicine, 1102 Bates Street, Suite 1770, Houston, TX 77030, USA
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
- Interdepartmental Program in Translational Biology & Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Xiao-Tong Song
- Center for Cell & Gene Therapy, Texas Children’s Hospital, The Methodist Hospital, Baylor College of Medicine, 1102 Bates Street, Suite 1770, Houston, TX 77030, USA
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
- Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Stephen Gottschalk
- Center for Cell & Gene Therapy, Texas Children’s Hospital, The Methodist Hospital, Baylor College of Medicine, 1102 Bates Street, Suite 1770, Houston, TX 77030, USA
- Texas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
- Interdepartmental Program in Translational Biology & Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
318
|
Marsh T, Pietras K, McAllister SS. Fibroblasts as architects of cancer pathogenesis. Biochim Biophys Acta Mol Basis Dis 2012; 1832:1070-8. [PMID: 23123598 DOI: 10.1016/j.bbadis.2012.10.013] [Citation(s) in RCA: 189] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 10/16/2012] [Accepted: 10/19/2012] [Indexed: 12/19/2022]
Abstract
Studies of epithelial cancers (i.e., carcinomas) traditionally focused on transformation of the epithelium (i.e., the cancer cells) and how aberrant signaling within the cancer cells modulates the surrounding tissue of origin. In more recent decades, the normal cells, blood vessels, molecules, and extracellular components that surround the tumor cells, collectively known as the "tumor microenvironment" or "stroma", have received increasing attention and are now thought to be key regulators of tumor initiation and progression. Of particular relevance to the work reviewed herein are the fibroblasts, which make up the major cell type within the microenvironment of most carcinomas. Due to their inherent heterogeneity, plasticity, and function, it is perhaps not surprising that fibroblasts are ideal modulators of normal and cancerous epithelium; however, these aspects also present challenges if we are to interrupt their tumor-supportive functions. Here, we review the current body of knowledge and the many questions that still remain about the special entity known as the cancer-associated fibroblast. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
Affiliation(s)
- Timothy Marsh
- Hematology Division, Brigham & Women's Hospital, Boston, MA 02115, USA
| | | | | |
Collapse
|
319
|
Subramanian A, Shu-Uin G, Kae-Siang N, Gauthaman K, Biswas A, Choolani M, Bongso A, Chui-Yee F. Human umbilical cord Wharton's jelly mesenchymal stem cells do not transform to tumor-associated fibroblasts in the presence of breast and ovarian cancer cells unlike bone marrow mesenchymal stem cells. J Cell Biochem 2012; 113:1886-95. [PMID: 22234854 DOI: 10.1002/jcb.24057] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Human bone marrow mesenchymal stem cells (hBMMSCs) were shown to transform into tumor-associated fibroblasts (TAFs) when in the vicinity of breast cancer tumors and played an important role in tumor enhancement and metastasis. In early human development MSCs migrating from the yolk sac and aorta-gonad-mesonephros (AGM) via the umbilical cord to the placenta and back to the fetal bone marrow were shown to get trapped in the gelatinous Wharton's jelly of the umbilical cord. The common origin of the Wharton's jelly MSCs and the finally homed hBMMSCs prompted us to evaluate whether hWJSCs are also involved in TAF transformation. hWJSCs and hBMMSCs were grown in the presence of breast and ovarian cancer cell conditioned medium (MDA-TCM, TOV-TCM) for 30 days. No changes were observed in the hWJSCs but the hBMMSCs transformed from short to thin long fibroblasts, their proliferation rates increased and CD marker expression decreased. The transformed hBMMSCs showed positive staining for the tumor-associated markers FSP, VEGF, EGF, and Tn-C. Real-time PCR and multiplex luminex bead analysis showed upregulation of TAF-related genes (FSP, FAP, Tn-C, Tsp-1, EGF, bFGF, IL-6, α-SMA, VEGF, and TGF-β) for hBMMSCs with low expression for hWJSCs. The luciferase assay showed that hWJSCs previously exposed to MDA-TCM or TOV-TCM had no stimulatory growth effect on luciferase-tagged MDA or TOV cells unlike hBMMSCs. The results confirmed that hWJSCs do not transform to the TAF phenotype and may therefore not be associated with enhanced growth of solid tumors making them a safe MSC for cell based therapies.
Collapse
Affiliation(s)
- Arjunan Subramanian
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge 119074, Singapore
| | | | | | | | | | | | | | | |
Collapse
|
320
|
Fang D, Sun L, Lin S, Zhou L, Su N, Yuan S, Yu B. Vinorelbine inhibits angiogenesis and 95D migration via reducing hypoxic fibroblast stromal cell-derived factor 1 secretion. Exp Biol Med (Maywood) 2012; 237:1045-55. [PMID: 22946087 DOI: 10.1258/ebm.2012.012037] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tumor stroma plays a prominent role in cancer progression. Fibroblasts constitute a majority of the stromal cells in tumor, and yet the functional contributions of these cells to tumor angiogenesis and invasion are poorly understood, especially the anticancer drug interference to these processes. To estimate the effects of vinorelbine (VNR) on fibroblast-associated tumor invasion and angiogenesis, we evaluated the response of 95D and human umbilical vein endothelial cell (HUVEC) migration, tube formation in vitro, as well as capillary formation of rat thoracic aorta rings to hypoxic MRC-5 conditioned medium (CM) by VNR pretreatment. Our results demonstrated that VNR significantly inhibited 95D and HUVEC migration and angiogenesis induced by hypoxic MRC-5 cells. We also showed that hypoxic MRC-5 CM (Hypo-CM) had a higher level of stromal cell-derived factor 1 (SDF-1) secretion, while Hypo-CM up-regulated the CXCR4 expression in HUVECs and 95Ds. This increased activity of SDF-1/CXCR4 paracrine was clearly attenuated by VNR pretreatment. It was further found that pretreating HUVECs and 95Ds with AMD3100, a CXCR4 antagonist, markedly reversed the Hypo-CM promoting cell migration and angiogenesis, while adding exogenous SDF-1 attenuated the inhibition effects of CM collected from VNR-pretreated hypoxic MRC-5 (Hypo-CMV). These data indicate that VNR indirectly decreased 95D migration and angiogenesis through its effect on hypoxic MRC-5, via impacting SDF-1/CXCR4 paracrine, suggesting that VNR could interrupt the influence of fibroblasts on HUVECs and 95Ds to exert an anticancer role. Therefore, fibroblasts should be taken into consideration when evaluating and developing anticancer drugs.
Collapse
Affiliation(s)
- Dongdong Fang
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
321
|
The pro-metastatic role of bone marrow-derived cells: a focus on MSCs and regulatory T cells. EMBO Rep 2012; 13:412-22. [PMID: 22473297 DOI: 10.1038/embor.2012.41] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 03/05/2012] [Indexed: 12/15/2022] Open
Abstract
Several bone marrow-derived cells have been shown to promote tumour growth and progression. These cells can home to the primary tumour and become active components of the tumour microenvironment. Recent studies have also identified bone marrow-derived cells—such as mesenchymal stem cells and regulatory T cells—as contributors to cancer metastasis. The innate versatility of these cells provides diverse functional aid to promote malignancy, ranging from structural support to signal-mediated suppression of the host immune response. Here, we review the role of mesenchymal stem cells and regulatory T cells in cancer metastasis. A better understanding of the bipolar nature of these bone marrow-derived cells in physiological and malignant contexts could pave the way for new therapeutics against metastatic disease.
Collapse
|
322
|
Brennen WN, Rosen DM, Wang H, Isaacs JT, Denmeade SR. Targeting carcinoma-associated fibroblasts within the tumor stroma with a fibroblast activation protein-activated prodrug. J Natl Cancer Inst 2012; 104:1320-34. [PMID: 22911669 DOI: 10.1093/jnci/djs336] [Citation(s) in RCA: 146] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Fibroblasts undergo a morphological transformation to a reactive phenotype in the tumor microenvironment characterized by the expression of proteins such as fibroblast activation protein (FAP), a post-prolyl endopeptidase with expression largely restricted to carcinoma-associated fibroblasts. Thapsigargin (TG) is a highly toxic natural plant product that triggers a rise in intracellular calcium levels and apoptosis. FAP is therefore a provocative target for the activation of prodrugs consisting of a FAP-specific peptide coupled to a potent cytotoxic analog of TG. METHODS The efficacy of FAP-activated peptidyl-TG prodrugs was tested in vitro in cell proliferation assays and effects on intracellular calcium in human cancer cell lines. The effects of FAP-activated prodrugs on tumor growth and host toxicity were tested in Balb-C nude MCF-7 and LNCaP xenograft mice (n = 9-11 per group). P values were calculated using permutation tests based on 50 000 permutations. Mixed effects models were used to account for correlations among replicate measures. All statistical tests were two-sided. RESULTS FAP-activated prodrugs killed human cancer cells at low nanomolar concentrations (MCF-7 cells: IC(50) = 3.5 nM). Amino acid-12ADT analogs from FAP-cleaved prodrugs, but not uncleaved prodrugs, produced a rapid rise in intracellular calcium within minutes of exposure. Immunohistochemical analysis of xenografts exposed to FAP-prodrugs documented stromal-selective cell death of fibroblasts, pericytes, and endothelial cells of sufficient magnitude to inhibit growth of MCF-7 and LNCaP xenografts with minimal systemic toxicity, whereas non-FAP cleavable prodrugs were inactive. MCF-7 and LNCaP xenografts treated with a FAP-activated prodrug had maximal treated-to-control tumor volume ratios of 0.36 (treated: mean = 0.206 mm(3), 95% CI = 0.068 to 0.344 mm(3); control: mean = 0.580 mm(3), 95% CI = 0.267 to 0.893 mm(3)) and 0.24 (treated: mean = 0.131 mm(3), 95% CI = 0.09 to 0.180 mm(3); control: mean = 0.543 mm(3), 95% CI = 0.173 to 0.913 mm(3)), respectively, on day 21 after therapy. CONCLUSIONS This study validates the proteolytic activity of FAP as a target for the activation of a systemically delivered cytotoxic prodrug and demonstrates that targeted killing of cells within the stromal compartment of the tumor microenvironment can produce a therapeutic response.
Collapse
Affiliation(s)
- W Nathaniel Brennen
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore, MD, USA
| | | | | | | | | |
Collapse
|
323
|
Kim YS, Ahn YH, Song KJ, Kang JG, Lee JH, Jeon SK, Kim HC, Yoo JS, Ko JH. Overexpression and β-1,6-N-acetylglucosaminylation-initiated aberrant glycosylation of TIMP-1: a "double whammy" strategy in colon cancer progression. J Biol Chem 2012; 287:32467-78. [PMID: 22859303 DOI: 10.1074/jbc.m112.370064] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
There has been ongoing debate over whether tissue inhibitor of metalloproteinase-1 (TIMP-1) is pro- or anti-oncogenic. We confirmed that TIMP-1 reinforced cell proliferation in an αvβ3 integrin-dependent manner and conferred resistance against cytotoxicity triggered by TNF-α and IL-2 in WiDr colon cancer cells. The cell-proliferative effects of TIMP-1 contributed to clonogenicity and tumor growth during the onset and early phase of tumor formation in vivo and in vitro. However, mass-produced TIMP-1 impeded further tumor growth by tightly inhibiting the activities of collagenases, which are critical for tumor growth and malignant transformation. Tumor cells could overcome this impasse by overexpression of N-acetylglucosaminyltransferase V, which deteriorates TIMP-1 into an aberrant glycoform. The aberrant glycoform of TIMP-1 was responsible for the mitigated inhibition of collagenases. The outbalanced activities of collagenases can degrade the basement membrane and the interstitial matrix, which act as a physical barrier for tumor growth and progression more efficiently. The concomitant overexpression of TIMP-1 and N-acetylglucosaminyltransferase V enabled WiDr cells to show a higher tumor growth rate as well as more malignant behaviors in a three-dimensional culture system.
Collapse
Affiliation(s)
- Yong-Sam Kim
- Cancer Biomarkers Development Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
324
|
Khalili JS, Liu S, Rodríguez-Cruz TG, Whittington M, Wardell S, Liu C, Zhang M, Cooper ZA, Frederick DT, Li Y, Zhang M, Joseph RW, Bernatchez C, Ekmekcioglu S, Grimm E, Radvanyi LG, Davis RE, Davies MA, Wargo JA, Hwu P, Lizée G. Oncogenic BRAF(V600E) promotes stromal cell-mediated immunosuppression via induction of interleukin-1 in melanoma. Clin Cancer Res 2012; 18:5329-40. [PMID: 22850568 DOI: 10.1158/1078-0432.ccr-12-1632] [Citation(s) in RCA: 255] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE In this study, we assessed the specific role of BRAF(V600E) signaling in modulating the expression of immune regulatory genes in melanoma, in addition to analyzing downstream induction of immune suppression by primary human melanoma tumor-associated fibroblasts (TAF). EXPERIMENTAL DESIGN Primary human melanocytes and melanoma cell lines were transduced to express WT or V600E forms of BRAF, followed by gene expression analysis. The BRAF(V600E) inhibitor vemurafenib was used to confirm targets in BRAF(V600E)-positive melanoma cell lines and in tumors from melanoma patients undergoing inhibitor treatment. TAF lines generated from melanoma patient biopsies were tested for their ability to inhibit the function of tumor antigen-specific T cells, before and following treatment with BRAF(V600E)-upregulated immune modulators. Transcriptional analysis of treated TAFs was conducted to identify potential mediators of T-cell suppression. RESULTS Expression of BRAF(V600E) induced transcription of interleukin 1 alpha (IL-1α) and IL-1β in melanocytes and melanoma cell lines. Further, vemurafenib reduced the expression of IL-1 protein in melanoma cell lines and most notably in human tumor biopsies from 11 of 12 melanoma patients undergoing inhibitor treatment. Treatment of melanoma-patient-derived TAFs with IL-1α/β significantly enhanced their ability to suppress the proliferation and function of melanoma-specific cytotoxic T cells, and this inhibition was partially attributable to upregulation by IL-1 of COX-2 and the PD-1 ligands PD-L1 and PD-L2 in TAFs. CONCLUSIONS This study reveals a novel mechanism of immune suppression sensitive to BRAF(V600E) inhibition, and indicates that clinical blockade of IL-1 may benefit patients with BRAF wild-type tumors and potentially synergize with immunotherapeutic interventions.
Collapse
Affiliation(s)
- Jahan S Khalili
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
325
|
Fernandez-Valdivia R, Lydon JP. From the ranks of mammary progesterone mediators, RANKL takes the spotlight. Mol Cell Endocrinol 2012; 357:91-100. [PMID: 21964466 PMCID: PMC3253322 DOI: 10.1016/j.mce.2011.09.030] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 09/16/2011] [Accepted: 09/16/2011] [Indexed: 12/18/2022]
Abstract
Whether during the diestrus phase of the estrous cycle or with pregnancy onset, the mitogenic effects of progesterone are well-established in the murine mammary epithelium. Importantly, progesterone-induced mitogenicity is critical for mammary tumor promotion, providing one explanation for the increase in breast cancer-risk observed with prolonged progestin-based hormone therapy. At the cellular level, progesterone projects its mitogenic influence through an evolutionary conserved paracrine mechanism of action. In this regard, recent studies provide compelling support for receptor activator of NF-kB ligand (RANKL) as a key paracrine mediator of the progesterone mitogenic signal. Induction of RANKL is sufficient to elicit mammary ductal side-branching and alveologenesis, the very morphogenetic responses elicited by progesterone during pregnancy and at diestrus. Significantly, the proliferative and pro-survival signals triggered by RANKL are also required for progestin-promotion of mammary tumorigenesis, underscoring a dual role for RANKL in progesterone-dependent mammary morphogenesis and tumorigenesis. Recently, RANKL has been shown to be critical for progesterone-induced expansion of the mammary stem cell population (and its lineal descendents), thereby advancing our conceptual understanding not only of RANKL's involvement in normal mammary morphogenesis but also in breast cancer risk associated with sustained hormone exposure. Finally, these studies together suggest that chemotherapeutic intervention of RANKL signaling represents a feasible approach for the effective prevention and/or treatment of hormone-responsive breast cancers.
Collapse
Affiliation(s)
- Rodrigo Fernandez-Valdivia
- Brown Foundation, Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, 77030
| | - John P. Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, 77030
- Corresponding Author: Telephone: 713-798-3534 Fax: 713-790-1275
| |
Collapse
|
326
|
Brennen WN, Isaacs JT, Denmeade SR. Rationale behind targeting fibroblast activation protein-expressing carcinoma-associated fibroblasts as a novel chemotherapeutic strategy. Mol Cancer Ther 2012; 11:257-66. [PMID: 22323494 DOI: 10.1158/1535-7163.mct-11-0340] [Citation(s) in RCA: 239] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The tumor microenvironment has emerged as a novel chemotherapeutic strategy in the treatment of cancer. This is most clearly exemplified by the antiangiogenesis class of compounds. Therapeutic strategies that target fibroblasts within the tumor stroma offer another treatment option. However, despite promising data obtained in preclinical models, such strategies have not been widely used in the clinical setting, largely due to a lack of effective treatments that specifically target this population of cells. The identification of fibroblast activation protein α (FAP) as a target selectively expressed on fibroblasts within the tumor stroma or on carcinoma-associated fibroblasts led to intensive efforts to exploit this novel cellular target for clinical benefit. FAP is a membrane-bound serine protease of the prolyl oligopeptidase family with unique post-prolyl endopeptidase activity. Until recently, the majority of FAP-based therapeutic approaches focused on the development of small-molecule inhibitors of enzymatic activity. Evidence suggests, however, that FAP's pathophysiological role in carcinogenesis may be highly contextual, depending on both the exact nature of the tumor microenvironment present and the cancer type in question to determine its tumor-promoting or tumor-suppressing phenotype. As an alternative strategy, we are taking advantage of FAP's restricted expression and unique substrate preferences to develop a FAP-activated prodrug to target the activation of a cytotoxic compound within the tumor stroma. Of note, this strategy would be effective independently of FAP's role in tumor progression because its therapeutic benefit would rely on FAP's localization and activity within the tumor microenvironment rather than strictly on inhibition of its function.
Collapse
Affiliation(s)
- W Nathaniel Brennen
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | |
Collapse
|
327
|
Multifaceted tumor stromal fibroblasts. CANCER MICROENVIRONMENT 2012; 5:187-93. [PMID: 22627670 DOI: 10.1007/s12307-012-0109-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 04/25/2012] [Indexed: 01/01/2023]
Abstract
Tumors are highly complex tissues composed of neoplastic cells and different kinds of stromal cells. Tumor stromal cells, especially fibroblasts, play important roles during the multistep development of tumors. In this review, the two-faced characteristics of tumor stromal fibroblasts are discussed in the light of our current knowledge. For one thing, fibroblasts act as an "inflammation regulator" by secretion of cytokines and regulation of tumor immunity; for another, they act as a "damage healer" for cure of wounds by remodeling extracellular matrix or taking a part in the "foreign body reaction". Since the properties of fibroblasts are complicated, both aspects of fibroblasts for tumor development should be considered carefully in clinical studies to target cancer-associated fibroblasts.
Collapse
|
328
|
Ding ZY, Zou XL, Wei YQ. Cancer microenvironment and cancer vaccine. CANCER MICROENVIRONMENT 2012; 5:333-44. [PMID: 22562695 DOI: 10.1007/s12307-012-0107-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 04/03/2012] [Indexed: 02/05/2023]
Abstract
The cancer microenvironment is constituted of non-transformed host stromal cells such as endothelial cells, fibroblasts, various immune cells, and a complex extra-cellular matrix secreted by both the normal and neoplastic cells embedded in it. The importance of the microenvironment and its potential in cancer therapy is just being established. Among modalities that target the microenvironment, cancer vaccine is a unique strategy which is aimed to elicit specific immunity against components in the microenvironment. Most, if not all, components can be targeted by the vaccines. The most extensively studied are the endothelial cells, fibroblasts and macrophages as well as ECM. Vaccines are in development for each of them. All the vaccines were proved to be effective at providing protective or therapeutic anti-tumor effects in the pre-clinical models. A few of them have been tested in the clinical trials. The mechanisms of the vaccines were mainly related to the cellular immune response such as CD8+ cytotoxic T cells, and in some instances CD4+ Th cells were involved as well. The present review also discussed the hurdles associated with the microenvironment-based vaccines such as the selection of suitable patients with appropriate biomarkers. With the rapid increase of our knowledge in the cancer microenvironment, the proof-of-concept of microenvironment-based cancer vaccines will surely expand our armamentarium against cancer.
Collapse
Affiliation(s)
- Zhen-Yu Ding
- Cancer Center, West China Hospital, West China Medical School, State Key Laboratory of Biotherapy, Sichuan Univerisity, Guoxue Lane 37, Chengdu, 610041, China
| | | | | |
Collapse
|
329
|
Schmidt T, Ben-Batalla I, Schultze A, Loges S. Macrophage-tumor crosstalk: role of TAMR tyrosine kinase receptors and of their ligands. Cell Mol Life Sci 2012; 69:1391-414. [PMID: 22076650 PMCID: PMC11115155 DOI: 10.1007/s00018-011-0863-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2011] [Revised: 10/14/2011] [Accepted: 10/14/2011] [Indexed: 02/07/2023]
Abstract
Ample clinical and preclinical evidence indicates that macrophages interact with tumor cells as well as with virtually all populations of host cells present in the tumor microenvironment. This crosstalk can strongly promote malignancy, but also has in principle the potential to inhibit tumor growth. Thus, it is of the utmost importance to improve our understanding of the mechanisms driving the pro- and antimalignant behavior of tumor-associated macrophages (TAMs) in order to develop better anticancer therapies. In this review, we discuss the biological consequences of reciprocal interactions between TAMs, cancer cells, endothelial cells, fibroblasts and other leukocyte subfractions within tumors. It was recently elucidated that tumors specifically educate macrophages to secrete growth arrest-specific gene 6 (Gas6), the common ligand of the Tyro3, Axl, Mer receptor (TAMR) family. In turn, Gas6 fosters tumor growth by promoting cancer cell proliferation. Therefore, the Gas6-TAMR axis might represent a novel target for disrupting tumor-macrophage crosstalk. We summarize here what is known about TAMR and their ligands in (human) cancer biology. In order to shed more light on the role of macrophages in human cancer, we additionally provide an overview of what is currently known about the prognostic impact of TAMs in human cancer.
Collapse
Affiliation(s)
- Thomas Schmidt
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Isabel Ben-Batalla
- Department of Hematology and Oncology with Sections BMT and Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
- Institute of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexander Schultze
- Department of Hematology and Oncology with Sections BMT and Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
- Institute of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sonja Loges
- Department of Hematology and Oncology with Sections BMT and Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
- Institute of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
330
|
Barar J. Targeting tumor microenvironment: the key role of immune system. BIOIMPACTS : BI 2012; 2:1-3. [PMID: 23678436 DOI: 10.5681/bi.2012.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 02/27/2012] [Accepted: 03/03/2012] [Indexed: 12/21/2022]
Abstract
In recent years, huge investigations on cancer progression and invasion have led to under-stand the pivotal role of tumor microenvironment. The current era of cancer therapy is based on the concept of simply targeting precise mechanisms to kill or to suppress the growth and expansion of malignant cells. Clinical data clearly correlate with in-vitro re-sults, emphasizing the direct impact of cancer environment on disease progression. This provides the opportunity to advance cancer therapy by virtue of targeting cancerous cells and non-cancerous component of tumor in a combinatorial manner. This tailor-made strategy demands the profound knowledge of cross talk between the bio-factors of tumor environment and corresponding pharmacology of drug candidates. The neighborhood of tumor is critical for how cancer cells grow and invade surrounding tissues. It appears that the tumor microenvironment as a "co-op" includes malignant cells, blood vessels, im-mune/inflammatory factors and extracellular matrix. As a longstanding dilemma, it is well-proved that immune system plays a direct role in the existence and progression of such coop. In some cases, immune cells e.g. tumor associated macrophages (TAMs) infiltrate into tumor and instead of fighting cancer cells, support them to grow. As an important fact, this tumor complexity should not be taken as granted where it can be advantageous in cancer therapy as well as early detection and prevention. The central aim of this editorial article is to highlight the importance of tumor microenvironment for successful cancer therapy.
Collapse
Affiliation(s)
- Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran ; Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
331
|
Ji RC. Macrophages are important mediators of either tumor- or inflammation-induced lymphangiogenesis. Cell Mol Life Sci 2012; 69:897-914. [PMID: 21984600 PMCID: PMC11114502 DOI: 10.1007/s00018-011-0848-6] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 07/26/2011] [Accepted: 09/22/2011] [Indexed: 01/13/2023]
Abstract
The lymphatic system provides important functions for tissue fluid homeostasis and immune response. Lymphangiogenesis, the formation of new lymphatics, comprises a series of complex cellular events in vitro or in vivo, e.g., proliferation, differentiation, and sprouting. Recent evidence has implied that macrophages act as a direct structural contributor to lymphatic endothelial walls or secret VEGF-C/-D and VEGF-A to initiate lymphangiogenesis in inflamed or tumor tissues. Bone marrow-derived macrophages are versatile cells that express different functional programs in response to exposure to microenvironmental signals, and can be identified by specific expression of a number of proteins, F4/80, CD11b, and CD68. Several causative factors, e.g., NF-κB, IL-1β, TNF-α, SDF-1, M-CSF, especially TonEBP/VEGF-C signaling, may be actively involved in macrophage-induced lymphangiogenesis. Alteration of macrophage phenotype and function has a profound effect on the development and progression of inflammation and malignancy, and macrophage depletion for controlling lymphangiogenesis may provide a novel approach for prevention and treatment of lymphatic-associated diseases.
Collapse
Affiliation(s)
- Rui-Cheng Ji
- Department of Human Anatomy, Oita University Faculty of Medicine, Oita 879-5593, Japan.
| |
Collapse
|
332
|
Tarasenko N, Cutts SM, Phillips DR, Inbal A, Nudelman A, Kessler-Icekson G, Rephaeli A. Disparate impact of butyroyloxymethyl diethylphosphate (AN-7), a histone deacetylase inhibitor, and doxorubicin in mice bearing a mammary tumor. PLoS One 2012; 7:e31393. [PMID: 22384017 PMCID: PMC3285631 DOI: 10.1371/journal.pone.0031393] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 01/06/2012] [Indexed: 02/01/2023] Open
Abstract
The histone deacetylase inhibitor (HDACI) butyroyloxymethyl diethylphosphate (AN-7) synergizes the cytotoxic effect of doxorubicin (Dox) and anti-HER2 on mammary carcinoma cells while protecting normal cells against their insults. This study investigated the concomitant changes occurring in heart tissue and tumors of mice bearing a subcutaneous 4T1 mammary tumor following treatment with AN-7, Dox, or their combination. Dox or AN-7 alone led to inhibition of both tumor growth and lung metastases, whereas their combination significantly increased their anticancer efficacy and attenuated Dox- toxicity. Molecular analysis revealed that treatment with Dox, AN-7, and to a greater degree, AN-7 together with Dox increased tumor levels of γH2AX, the marker for DNA double-strand breaks and decreased the expression of Rad51, a protein needed for DNA repair. These events culminated in increased apoptosis, manifested by the appearance of cytochrome-c in the cytosol. In the myocardium, Dox-induced cardiomyopathy was associated with an increase in γH2AX expression and a reduction in Rad51 and MRE11 expression and increased apoptosis. The addition of AN-7 to the Dox treatment protected the heart from Dox insults as was manifested by a decrease in γH2AX levels, an increase in Rad51 and MRE11 expression, and a diminution of cytochrome-c release. Tumor fibrosis was high in untreated mice but diminished in Dox- and AN-7-treated mice and was almost abrogated in AN-7+Dox-treated mice. By contrast, in the myocardium, Dox alone induced a dramatic increase in fibrosis, and AN7+Dox attenuated it. The high expression levels of c-Kit, Ki-67, c-Myc, lo-FGF, and VEGF in 4T1 tumors were significantly reduced by Dox or AN-7 and further attenuated by AN-7+Dox. In the myocardium, Dox suppressed these markers, whereas AN-7+Dox restored their expression. In conclusion, the combination of AN-7 and Dox results in two beneficial effects, improved anticancer efficacy and cardioprotection.
Collapse
Affiliation(s)
- Nataly Tarasenko
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Beilinson Campus, Petach-Tikva, Israel
- Thrombosis and Hemostasis Unit, Rabin Medical Center, Beilinson Hospital, Petach-Tikva, Israel
| | - Suzanne M. Cutts
- Department of Biochemistry, La Trobe University, Victoria, Australia
| | - Don R. Phillips
- Department of Biochemistry, La Trobe University, Victoria, Australia
| | - Aida Inbal
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Beilinson Campus, Petach-Tikva, Israel
- Thrombosis and Hemostasis Unit, Rabin Medical Center, Beilinson Hospital, Petach-Tikva, Israel
| | | | - Gania Kessler-Icekson
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Beilinson Campus, Petach-Tikva, Israel
| | - Ada Rephaeli
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Beilinson Campus, Petach-Tikva, Israel
- * E-mail:
| |
Collapse
|
333
|
Khamis ZI, Sahab ZJ, Sang QXA. Active roles of tumor stroma in breast cancer metastasis. Int J Breast Cancer 2012; 2012:574025. [PMID: 22482059 PMCID: PMC3296264 DOI: 10.1155/2012/574025] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 11/04/2011] [Accepted: 11/11/2011] [Indexed: 12/21/2022] Open
Abstract
Metastasis is the major cause of death for breast cancer patients. Tumors are heterogenous cellular entities composed of cancer cells and cells of the microenvironment in which they reside. A reciprocal dynamic interaction occurs between the tumor cells and their surrounding stroma under physiological and pathological conditions. This tumor-host communication interface mediates the escape of tumor cells at the primary site, survival of circulating cancer cells in the vasculature, and growth of metastatic cancer at secondary site. Each step of the metastatic process is accompanied by recruitment of stromal cells from the microenvironment and production of unique array of growth factors and chemokines. Stromal microenvironment may play active roles in breast cancer metastasis. Elucidating the types of cells recruited and signal pathways involved in the crosstalk between tumor cells and stromal cells will help identify novel strategies for cotargeting cancer cells and tumor stromal cells to suppress metastasis and improve patient outcome.
Collapse
Affiliation(s)
- Zahraa I. Khamis
- Department of Chemistry and Biochemistry and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306-4390, USA
| | - Ziad J. Sahab
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Qing-Xiang Amy Sang
- Department of Chemistry and Biochemistry and Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306-4390, USA
| |
Collapse
|
334
|
The Microenvironmental Effect in the Progression, Metastasis, and Dormancy of Breast Cancer: A Model System within Bone Marrow. Int J Breast Cancer 2012; 2012:721659. [PMID: 22482060 PMCID: PMC3296216 DOI: 10.1155/2012/721659] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 11/12/2011] [Accepted: 11/15/2011] [Indexed: 01/14/2023] Open
Abstract
Despite diagnostic advances, breast cancer remains the most prevalent cancer among women in the United States. The armamentarium of treatment options for metastatic disease is limited and mostly ineffective with regards to eradicating cancer. However, there have been novel findings in the recent literature that substantiate the function of the microenvironment in breast cancer progression and the support of metastasis to tertiary sites such as bone marrow. The uncovered significance of the microenvironment in the pathophysiology of breast cancer metastasis has served to challenge previously widespread theories and introduce new perspectives for the future research to eradicate breast cancer. This paper delineates the current understanding of the molecular mechanisms involved in the interactions between breast cancer cells and the microenvironment in progression, metastasis, and dormancy. The information, in addition to other mechanisms described in bone marrow, is discussed in the paper.
Collapse
|
335
|
Liu R, Li H, Liu L, Yu J, Ren X. Fibroblast activation protein: A potential therapeutic target in cancer. Cancer Biol Ther 2012; 13:123-9. [PMID: 22236832 DOI: 10.4161/cbt.13.3.18696] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The concept of targeting antigens selectively expressed on the surface of tumor capillary endothelial cells or in tumor stroma has emerged as a promising strategy for cancer therapeutics. Identification of stromal targets for anticancer therapy and development of selective inhibitors of these targets are of great clinical interest. Fibroblast activation protein (FAP), a member of the serine protease family, selectively expressed in the stromal fibroblasts associated with epithelial cancers, whereas with low or undetectable expression in the resting fibroblasts of normal adult tissues. The proteolytic activity of FAP has been shown to support tumor growth and proliferation, making it a potential target for novel anticancer therapies, such as those by immune-based approaches.
Collapse
Affiliation(s)
- Rui Liu
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | | | | | | | | |
Collapse
|
336
|
Porta C, Riboldi E, Totaro MG, Strauss L, Sica A, Mantovani A. Macrophages in cancer and infectious diseases: the 'good' and the 'bad'. Immunotherapy 2012; 3:1185-202. [PMID: 21995571 DOI: 10.2217/imt.11.116] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Macrophages are crucial orchestrators of host defence and tissue homeostasis. Macrophages are heterogeneous and plastic cells that in response to different microenvironmental signals can mount a broad spectrum of different programs of polarized activation. In different pathological contexts including cancer and infectious diseases, macrophages diversity and plasticity may act as a double-edged sword. The elucidation of the molecular mechanisms underlying macrophages recruitment and functional activation allows the identification of valuable targets for the development of innovative therapeutic approaches.
Collapse
Affiliation(s)
- Chiara Porta
- DiSCAFF, University of Piemonte Orientale A. Avogadro, Novara, Italy
| | | | | | | | | | | |
Collapse
|
337
|
Görgün G, Anderson KC. Intrinsic modulation of lymphocyte function by stromal cell network: advance in therapeutic targeting of cancer. Immunotherapy 2012; 3:1253-64. [PMID: 21995575 DOI: 10.2217/imt.11.124] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Advances in tumor biology have demonstrated a point of critical importance: tumor are established as an intersection of malignant clone cells and surrounding stromal cells. The stroma is composed of nonhematopoietic cells, including connective tissue cells, blood vessels, nerves, fat and smooth muscle cells, in the extracellular matrix niche. Recent studies have demonstrated that stromal cells regulate immune responses by: coordinating lymphocyte homing, differentiation, activation and antigen responses; inducing tolerance; and maintaining immunologic memory. Hence, elucidation of the interaction between stromal cells and lymphocytes is essential for generating effective immunotherapies. In this article, we summarize what is currently known about the interactions between stromal cells and lymphocytes in the tumor microenvironment, as well as potential immunotherapeutic approaches targeting stroma-lymphocyte interactions; both in the context of our work on multiple myeloma, and of recent literature in both solid tumors and hematologic malignancies.
Collapse
Affiliation(s)
- Güllü Görgün
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
338
|
Hielscher AC, Qiu C, Gerecht S. Breast cancer cell-derived matrix supports vascular morphogenesis. Am J Physiol Cell Physiol 2012; 302:C1243-56. [PMID: 22277754 DOI: 10.1152/ajpcell.00011.2012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The extracellular matrix (ECM), important for maintaining tissue homeostasis, is abnormally expressed in mammary tumors and additionally plays a crucial role in angiogenesis. We hypothesize that breast cancer cells (BCCs) deposit ECM that supports unique patterns of vascular morphogenesis of endothelial cells (ECs). Evaluation of ECM expression revealed that a nontumorigenic cell line (MCF10A), a tumorigenic cell line (MCF7), and a metastatic cell line (MDA-MB-231) express collagens I and IV, fibronectin, and laminin, with tenascin-C limited to MCF10A and MCF7. The amount of ECM deposited by BCCs was found to be higher in MCF10A compared with MCF7 and MDA231, with all ECM differing in their gross structure but similar in mean fiber diameter. Nonetheless, deposition of ECM from BCC lines was overall difficult to detect and insufficient to support capillary-like structure (CLS) formation of ECs. Therefore, a coculture approach was undertaken in which individual BCC lines were cocultured with fibroblasts. Variation in abundance of deposited ECM, deposition of ECM proteins, such as absent collagen I deposition from MDA231-fibroblast cocultures, and fibril organization was found. Deposited ECM from fibroblasts and each coculture supported rapid CLS formation of ECs. Evaluation of capillary properties revealed that CLS grown on ECM deposited from MDA231-fibroblast cocultures possessed significantly larger lumen diameters, occupied the greatest percentage of area, expressed the highest levels of von Willebrand factor, and expressed the greatest amount of E-selectin, which was upregulated independent of exposure to TNF-α. To our knowledge, this is the first study to report tumor cell ECM-mediated differences in vascular capillary features, and thus offers the framework for future investigations interrogating the role of the tumor ECM in supporting vascular morphogenesis.
Collapse
Affiliation(s)
- Abigail C Hielscher
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | |
Collapse
|
339
|
Thapa D, Kang Y, Park PH, Noh SK, Lee YR, Han SS, Ku SK, Jung Y, Kim JA. Anti-tumor Activity of the Novel Hexahydrocannabinol Analog LYR-8 in Human Colorectal Tumor Xenograft Is Mediated through the Inhibition of Akt and Hypoxia-Inducible Factor-1α Activation. Biol Pharm Bull 2012; 35:924-32. [DOI: 10.1248/bpb.35.924] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | - Youra Kang
- College of Pharmacy, Yeungnam University
| | | | - Seok Kyun Noh
- School of Chemical Engineering and Technology, Yeungnam University
| | - Yong Rok Lee
- School of Chemical Engineering and Technology, Yeungnam University
| | - Sung Soo Han
- Department of Nano, Medical & Polymer Materials, College of Engineering, Yeungnam University
| | - Sae Kwang Ku
- College of Oriental Medicine, Daegu Hanny University
| | - Yunjin Jung
- College of Pharmacy, Pusan National University
| | | |
Collapse
|
340
|
Employing Live Microbes for Vaccine Delivery. DEVELOPMENT OF NOVEL VACCINES 2012. [PMCID: PMC7123214 DOI: 10.1007/978-3-7091-0709-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/29/2022]
|
341
|
|
342
|
Tchou J, Conejo-Garcia J. Targeting the Tumor Stroma as a Novel Treatment Strategy for Breast Cancer. ADVANCES IN PHARMACOLOGY 2012; 65:45-61. [DOI: 10.1016/b978-0-12-397927-8.00003-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
343
|
Engels B, Rowley DA, Schreiber H. Targeting stroma to treat cancers. Semin Cancer Biol 2011; 22:41-9. [PMID: 22212863 DOI: 10.1016/j.semcancer.2011.12.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Accepted: 12/15/2011] [Indexed: 01/29/2023]
Abstract
All cancers depend on stroma for support of growth. Leukemias, solid tumors, cancer cells causing effusions, metastases as well as micro-disseminated cancer cells release factors that stimulate stromal cells, which in turn produce ligands that stimulate cancer cells. Therefore, elimination of stromal support by destroying the stromal cells or by inhibiting feedback stimulation of cancer growth is in the focus of many evolving therapies. A stringent evaluation of the efficacy of stromal targeting requires testing in animal models. Most current studies emphasize the successes of stromal targeting rather than deciphering its limitations. Here we show that many of the stromal targeting approaches, while often reducing tumor growth rates, are rarely curative. Therefore, we will also discuss conditions where stromal targeting can eradicate large established tumors. Finally, we will examine still unanswered questions of this promising and exciting area of cancer research.
Collapse
Affiliation(s)
- Boris Engels
- Department of Pathology, Committee on Cancer Biology, Committee on Immunology, The University of Chicago, Chicago, IL 60637-5420, USA.
| | | | | |
Collapse
|
344
|
Interleukin 1α sustains the expression of inflammatory factors in human pancreatic cancer microenvironment by targeting cancer-associated fibroblasts. Neoplasia 2011; 13:664-75. [PMID: 21847358 DOI: 10.1593/neo.11332] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 05/17/2011] [Accepted: 05/20/2011] [Indexed: 02/06/2023] Open
Abstract
The tumor microenvironment in pancreatic ductal adenocarcinoma (PDAC) is dynamic, with an extensive interaction between the stroma and tumor cells. The aim of this study was to delineate the cross talk between PDAC and cancer-associated fibroblasts (CAFs), with a focus on the mechanism creating the chronic inflammatory tumor milieu. We assessed the effects of the cross talk between PDAC and CAF cell lines on the creation and sustenance of the inflammatory tumor microenvironment in pancreatic cancer. The coculture of PDAC and CAF cell lines enhanced the levels of inflammatory factors including IL-1α, IL-6, CXCL8, VEGF-A, CCL20, and COX-2. CAFs were superior to tumor cells regarding the production of most inflammatory factors, and tumor cell-associated IL-1α was established as the initiator of the enhanced production of inflammatory factors through the binding of IL-1α to IL-1 receptor 1 (IL-1R1) expressed predominantly by CAFs. Furthermore, we found a correlation between IL-1α and CXCL8 expression levels in PDAC tissues and correlation between IL-1α expression and the clinical outcome of the patients. This confirmed an important role for the IL-1 signaling cascade in the creation and sustenance of a tumor favorable microenvironment. Neutralization of the IL-1α signaling efficiently diminished the cross talk-induced production of inflammatory factors. These data suggest that the cross talk between PDAC cells and the main stroma cell type, i.e. CAFs, is one essential factor in the formation of the inflammatory tumor environment, and we propose that neutralization of the IL-1α signaling might be a potential therapy for this cancer.
Collapse
|
345
|
Martinez-Outschoorn UE, Goldberg A, Lin Z, Ko YH, Flomenberg N, Wang C, Pavlides S, Pestell RG, Howell A, Sotgia F, Lisanti MP. Anti-estrogen resistance in breast cancer is induced by the tumor microenvironment and can be overcome by inhibiting mitochondrial function in epithelial cancer cells. Cancer Biol Ther 2011; 12:924-38. [PMID: 22041887 DOI: 10.4161/cbt.12.10.17780] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Here, we show that tamoxifen resistance is induced by cancer-associated fibroblasts (CAFs). Coculture of estrogen receptor positive (ER+) MCF7 cells with fibroblasts induces tamoxifen and fulvestrant resistance with 4.4 and 2.5-fold reductions, respectively, in apoptosis compared with homotypic MCF7 cell cultures. Treatment of MCF7 cells cultured alone with high-energy mitochondrial "fuels" (L-lactate or ketone bodies) is sufficient to confer tamoxifen resistance, mimicking the effects of coculture with fibroblasts. To further demonstrate that epithelial cancer cell mitochondrial activity is the origin of tamoxifen resistance, we employed complementary pharmacological and genetic approaches. First, we studied the effects of two mitochondrial "poisons," namely metformin and arsenic trioxide (ATO), on fibroblast-induced tamoxifen resistance. We show here that treatment with metformin or ATO overcomes fibroblast-induced tamoxifen resistance in MCF7 cells. Treatment with the combination of tamoxifen plus metformin or ATO leads to increases in glucose uptake in MCF7 cells, reflecting metabolic uncoupling between epithelial cancer cells and fibroblasts. In coculture, tamoxifen induces the upregulation of TIGAR (TP53-induced glycolysis and apoptosis regulator), a p53 regulated gene that simultaneously inhibits glycolysis, autophagy and apoptosis and reduces ROS generation, thereby promoting oxidative mitochondrial metabolism. To genetically mimic the effects of coculture, we next recombinantly overexpressed TIGAR in MCF7 cells. Remarkably, TIGAR overexpression protects epithelial cancer cells from tamoxifen-induced apoptosis, providing genetic evidence that increased mitochondrial function confers tamoxifen resistance. Finally, CAFs also protect MCF7 cells against apoptosis induced by other anticancer agents, such as the topoisomerase inhibitor doxorubicin (adriamycin) and the PARP-1 inhibitor ABT-888. These results suggest that the tumor microenvironment may be a general mechanism for conferring drug resistance. In summary, we have discovered that mitochondrial activity in epithelial cancer cells drives tamoxifen resistance in breast cancer and that mitochondrial "poisons" are able to re-sensitize these cancer cells to tamoxifen. In this context, TIGAR may be a key "druggable" target for preventing drug resistance in cancer cells, as it protects cancer cells against the onset of stress-induced mitochondrial dys-function and aerobic glycolysis.
Collapse
Affiliation(s)
- Ubaldo E Martinez-Outschoorn
- The Jefferson Stem Cell Biology and Regenerative Medicine Center, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
346
|
Leibovici J, Itzhaki O, Huszar M, Sinai J. Targeting the tumor microenvironment by immunotherapy: part 2. Immunotherapy 2011; 3:1385-408. [DOI: 10.2217/imt.11.112] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cancer therapy was traditionally centered on the neoplastic cells. This included mainly surgery, radiation, and chemotherapy, in some cases hormone therapy and to a lesser extent immunotherapy – all traditionally targeted to the highly proliferating mutated tumor cells. In view of our present understanding of the powerfull influence of the tumor microenvironment (TME) on cancer behavior and response – and lack of response – to treatment, this previously ignored constituent of cancer now has to be considered as an important, even indispensable target for therapy. The TME may be targeted both to its immune and to its nonimmune components. The various immune evasion elements of the TME should be targeted as well.
Collapse
Affiliation(s)
| | - Orit Itzhaki
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, 69978 Tel-Aviv, Israel
| | - Monica Huszar
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, 69978 Tel-Aviv, Israel
| | - Judith Sinai
- Department of Pathology, Sackler Faculty of Medicine, Tel-Aviv University, 69978 Tel-Aviv, Israel
| |
Collapse
|
347
|
Sica A, Porta C, Morlacchi S, Banfi S, Strauss L, Rimoldi M, Totaro MG, Riboldi E. Origin and Functions of Tumor-Associated Myeloid Cells (TAMCs). CANCER MICROENVIRONMENT 2011; 5:133-49. [PMID: 21948460 DOI: 10.1007/s12307-011-0091-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 09/13/2011] [Indexed: 02/06/2023]
Abstract
The construction of an inflammatory microenvironment provides the fuel for cancer development and progression. Hence, solid tumors promote the expansion and the recruitment of leukocyte populations, among which tumor-associated myeloid cells (TAMCs) represent a paradigm for cancer-promoting inflammation. TAMCs group heterogeneous phagocytic populations stemming from a common myeloid progenitor (CMP), that orchestrate various aspects of cancer, including: diversion and skewing of adaptive responses; immunosuppression; cell growth; angiogenesis; matrix deposition and remodelling; construction of a metastatic niche and actual metastasis. Several evidence indicate that TAMCs show plasticity and/or functional heterogeneity, suggesting that tumour-derived factors promote their functional "reprogramming" towards protumoral activities. While recent studies have attempted to address the role of microenvironment signals, the interplay between cancer cells, innate and adaptive immunity is now emerging as a crucial step of the TAMCs reprogramming. Here we discuss the evidence for the differentiation of TAMCs during the course of tumor progression and the molecular mechanisms that regulate such event.
Collapse
Affiliation(s)
- Antonio Sica
- DiSCAFF, University of Piemonte Orientale A. Avogadro, via Bovio 6, 28100, Novara, Italy,
| | | | | | | | | | | | | | | |
Collapse
|
348
|
Rudnick JA, Arendt LM, Klebba I, Hinds JW, Iyer V, Gupta PB, Naber SP, Kuperwasser C. Functional heterogeneity of breast fibroblasts is defined by a prostaglandin secretory phenotype that promotes expansion of cancer-stem like cells. PLoS One 2011; 6:e24605. [PMID: 21957456 PMCID: PMC3177828 DOI: 10.1371/journal.pone.0024605] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 08/14/2011] [Indexed: 12/31/2022] Open
Abstract
Fibroblasts are important in orchestrating various functions necessary for maintaining normal tissue homeostasis as well as promoting malignant tumor growth. Significant evidence indicates that fibroblasts are functionally heterogeneous with respect to their ability to promote tumor growth, but markers that can be used to distinguish growth promoting from growth suppressing fibroblasts remain ill-defined. Here we show that human breast fibroblasts are functionally heterogeneous with respect to tumor-promoting activity regardless of whether they were isolated from normal or cancerous breast tissues. Rather than significant differences in fibroblast marker expression, we show that fibroblasts secreting abundant levels of prostaglandin (PGE2), when isolated from either reduction mammoplasty or carcinoma tissues, were both capable of enhancing tumor growth in vivo and could increase the number of cancer stem-like cells. PGE2 further enhanced the tumor promoting properties of fibroblasts by increasing secretion of IL-6, which was necessary, but not sufficient, for expansion of breast cancer stem-like cells. These findings identify a population of fibroblasts which both produce and respond to PGE2, and that are functionally distinct from other fibroblasts. Identifying markers of these cells could allow for the targeted ablation of tumor-promoting and inflammatory fibroblasts in human breast cancers.
Collapse
Affiliation(s)
- Jenny A. Rudnick
- Graduate Program in Cell, Molecular and Developmental Biology Program, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Lisa M. Arendt
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Ina Klebba
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - John W. Hinds
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Vandana Iyer
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Piyush B. Gupta
- Whitehead Institute for Biomedical Research, Broad Institute, Department of Biology, Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, United States of America
| | - Stephen P. Naber
- Department of Pathology, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Charlotte Kuperwasser
- Graduate Program in Cell, Molecular and Developmental Biology Program, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
349
|
Fernando RI, Castillo MD, Litzinger M, Hamilton DH, Palena C. IL-8 signaling plays a critical role in the epithelial-mesenchymal transition of human carcinoma cells. Cancer Res 2011; 71:5296-306. [PMID: 21653678 PMCID: PMC3148346 DOI: 10.1158/0008-5472.can-11-0156] [Citation(s) in RCA: 310] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The switch of tumor cells from an epithelial to a mesenchymal-like phenotype [designated as epithelial-to-mesenchymal transition (EMT)] is known to induce tumor cell motility and invasiveness, therefore promoting metastasis of solid carcinomas. Although multiple studies have focused on elucidating the signaling events that initiate this phenotypic switch, there has been so far no characterization of the pattern of soluble mediators released by tumor cells undergoing EMT, and the potential impact that this phenotypic switch could have on the remodeling of the tumor microenvironment. Here we show that induction of EMT in human carcinoma cells via overexpression of the transcription factor Brachyury is associated with enhanced secretion of multiple cytokines, chemokines, and angiogenic factors and, in particular, with the induction of the IL-8/IL-8R axis. Our results also indicate the essential role of interleukin 8 (IL-8) signaling for the acquisition and/or maintenance of the mesenchymal and invasive features of Brachyury-overexpressing tumor cells and show that IL-8 secreted by tumor cells undergoing EMT could potentiate tumor progression by inducing adjacent epithelial tumor cells into EMT. Altogether, our results emphasize the potential role of EMT in the modulation of the tumor microenvironment via secretion of multiple soluble mediators and suggest that IL-8 signaling blockade may provide a means of targeting mesenchymal-like, invasive tumor cells.
Collapse
Affiliation(s)
- Romaine I Fernando
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
350
|
Liao D, Liu Z, Wrasidlo WJ, Luo Y, Nguyen G, Chen T, Xiang R, Reisfeld RA. Targeted therapeutic remodeling of the tumor microenvironment improves an HER-2 DNA vaccine and prevents recurrence in a murine breast cancer model. Cancer Res 2011; 71:5688-96. [PMID: 21784871 DOI: 10.1158/0008-5472.can-11-1264] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The tumor microenvironment (TME) mediates immunosuppression resulting in tumor cell escape from immune surveillance and cancer vaccine failure. Immunosuppression is mediated by the STAT-3 transcription factor, which potentiates signaling in tumor and immune cells. Because immunosuppression continues to be a major inhibitor of cancer vaccine efficacy, we examined in this study whether therapeutically targeted delivery of a synthetic STAT-3 inhibitor to the TME, combined with an HER-2 DNA vaccine can improve immune surveillance against HER-2(+) breast cancer and prevent its recurrence. To this end, we developed a novel ligand-targeted nanoparticle (NP) encapsulating a CDDO-Im payload capable of specific delivery to the TME, which showed an effective therapeutic inhibition of STAT-3 activation in primary tumors. Furthermore, we showed that treatment with these NPs resulted in priming of the immune TME, characterized by increased IFN-γ, p-STAT-1, GM-CSF, IL-2, IL-15, and IL-12b and reduced TGF-β, IL-6, and IL-10 protein expression. In addition, we found significantly increased tumor infiltration by activated CD8(+) T cells, M1 macrophages, and dendritic cells. These changes correlated with delayed growth of orthotopic 4TO7 breast tumors and, when combined with an HER-2 DNA vaccine, prevented HER-2(+) primary tumor recurrence in immunocompetent mice. Furthermore, antitumor T-cell responses were enhanced in splenocytes isolated from mice treated with this combination therapy. Together, these data show effective protection from cancer recurrence through improved immune surveillance against a tumor-specific antigen.
Collapse
Affiliation(s)
- Debbie Liao
- Department of Immunology and Microbial Science, The Scripps Research Institute, University of California, San Diego, La Jolla, California, USA
| | | | | | | | | | | | | | | |
Collapse
|