51
|
Charbe N, McCarron PA, Tambuwala MM. Three-dimensional bio-printing: A new frontier in oncology research. World J Clin Oncol 2017; 8:21-36. [PMID: 28246583 PMCID: PMC5309712 DOI: 10.5306/wjco.v8.i1.21] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/02/2016] [Accepted: 12/07/2016] [Indexed: 02/06/2023] Open
Abstract
Current research in oncology deploys methods that rely principally on two-dimensional (2D) mono-cell cultures and animal models. Although these methodologies have led to significant advancement in the development of novel experimental therapeutic agents with promising anticancer activity in the laboratory, clinicians still struggle to manage cancer in the clinical setting. The disappointing translational success is attributable mainly to poor representation and recreation of the cancer microenvironment present in human neoplasia. Three-dimensional (3D) bio-printed models could help to simulate this micro-environment, with recent bio-printing of live human cells demonstrating that effective in vitro replication is achievable. This literature review outlines up-to-date advancements and developments in the use of 3D bio-printed models currently being used in oncology research. These innovative advancements in 3D bio-printing open up a new frontier for oncology research and could herald an era of progressive clinical cancer therapeutics.
Collapse
|
52
|
Leyh B, Dittmer A, Lange T, Martens JWM, Dittmer J. Stromal cells promote anti-estrogen resistance of breast cancer cells through an insulin-like growth factor binding protein 5 (IGFBP5)/B-cell leukemia/lymphoma 3 (Bcl-3) axis. Oncotarget 2016; 6:39307-28. [PMID: 26515727 PMCID: PMC4770774 DOI: 10.18632/oncotarget.5624] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 10/09/2015] [Indexed: 12/27/2022] Open
Abstract
There is strong evidence that stromal cells promote drug resistance of cancer. Here, we show that mesenchymal stem cells (MSCs) and carcinoma-associated fibroblasts (CAFs) desensitize ERα-positive breast cancer cells to the anti-estrogen fulvestrant. In search for the mechanism, we found that MSCs and CAFs similarly increased the activity of the PI3K/AKT and the JAK/STAT3 pathways and upregulated the expression of integrin β1, IGF1R, HIF1α, CAIX and Bcl-3 in MCF-7 cells. Further analyses revealed that MSCs and CAFs coordinately induce these changes by triggering the downregulation of IGFBP5. Loss of IGFBP5 in MCF-7 cells was an early and long-lasting event in response to MSCs and CAFs and was accompanied by growth stimulation both in the absence and presence of fulvestrant. The growth-stimulatory effect in the absence of fulvestrant could be attributed to PI3K/AKT pathway activation and could be mimicked by insulin. The growth-promoting effect in the presence of fulvestrant depended upon the upregulation of Bcl-3. By cRNA microarray analysis we identified additional IGFBP5 targets, of which two (KLHL4 and SEPP1) were inversely regulated by IGFBP5 and Bcl-3. BT474 cells also responded to stromal cells by downregulating IGFBP5 and upregulating the P-AKT, Bcl-3 and IGF1R levels, whereas T47D cells did not show any of these responses. In conclusion, our data suggest that, by targeting IGFBP5 expression in ERα-positive breast cancer cells, such as MCF-7 cells, MSCs and CAFs are able to orchestrate a variety of events, particularly activation of the PI3K/AKT pathway, upregulation of Bcl-3 expression and desensitization to anti-estrogen.
Collapse
Affiliation(s)
- Benjamin Leyh
- Clinic for Gynecology, Martin Luther University Halle-Wittenberg, Halle(Saale), Germany
| | - Angela Dittmer
- Clinic for Gynecology, Martin Luther University Halle-Wittenberg, Halle(Saale), Germany
| | - Theresia Lange
- Clinic for Gynecology, Martin Luther University Halle-Wittenberg, Halle(Saale), Germany
| | - John W M Martens
- Department of Medical Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jürgen Dittmer
- Clinic for Gynecology, Martin Luther University Halle-Wittenberg, Halle(Saale), Germany
| |
Collapse
|
53
|
Akasov R, Gileva A, Zaytseva-Zotova D, Burov S, Chevalot I, Guedon E, Markvicheva E. 3D in vitro co-culture models based on normal cells and tumor spheroids formed by cyclic RGD-peptide induced cell self-assembly. Biotechnol Lett 2016; 39:45-53. [DOI: 10.1007/s10529-016-2218-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 09/13/2016] [Indexed: 01/19/2023]
|
54
|
Activation of PI3K/Akt/mTOR signaling in the tumor stroma drives endocrine therapy-dependent breast tumor regression. Oncotarget 2016; 6:22081-97. [PMID: 26098779 PMCID: PMC4673148 DOI: 10.18632/oncotarget.4203] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 05/27/2015] [Indexed: 12/21/2022] Open
Abstract
Improved efficacy of neoadjuvant endocrine-targeting therapies in luminal breast carcinomas could be achieved with optimal use of pathway targeting agents. In a mouse model of ductal breast carcinoma we identify a tumor regressive stromal reaction that is induced by neoadjuvant endocrine therapy. This reparative reaction is characterized by tumor neovascularization accompanied by infiltration of immune cells and carcinoma-associated fibroblasts that stain for phosphorylated ribosomal protein S6 (pS6), downstream the PI3K/Akt/mTOR pathway. While tumor variants with higher PI3K/Akt/mTOR activity respond well to a combination of endocrine and PI3K/Akt/mTOR inhibitors, tumor variants with lower PI3K/Akt/mTOR activity respond more poorly to the combination therapy than to the endocrine therapy alone, associated with inhibition of stromal pS6 and the reparative reaction. In human breast cancer xenografts we confirm that such differential sensitivity to therapy is primarily determined by the level of PI3K/Akt/mTOR in tumor cells. We further show that the clinical response of breast cancer patients undergoing neoadjuvant endocrine therapy is associated with the reparative stromal reaction. We conclude that tumor level and localization of pS6 are associated with therapeutic response in breast cancer and represent biomarkers to distinguish which tumors will benefit from the incorporation of PI3K/Akt/mTOR inhibitors with neoadjuvant endocrine therapy.
Collapse
|
55
|
Paulsson J, Rydén L, Strell C, Frings O, Tobin NP, Fornander T, Bergh J, Landberg G, Stål O, Östman A. High expression of stromal PDGFRβ is associated with reduced benefit of tamoxifen in breast cancer. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2016; 3:38-43. [PMID: 28138400 PMCID: PMC5259559 DOI: 10.1002/cjp2.56] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/06/2016] [Accepted: 07/12/2016] [Indexed: 12/20/2022]
Abstract
Cancer‐associated fibroblasts (CAFs) regulate tumour growth, metastasis and response to treatment. Recent studies indicate the existence of functionally distinct CAF subsets. Suggested mechanisms whereby CAFs can impact on treatment response include paracrine signalling affecting cancer cell drug sensitivity and effects on tumour drug uptake. PDGFRβ is an important regulator of fibroblasts. Experimental studies have linked PDGFRβ‐positive fibroblasts to metastasis and also to reduced tumour drug uptake. This study has investigated the potential role of PDGFRβ‐positive fibroblasts in response to adjuvant tamoxifen treatment of breast cancer. Analyses of two breast cancer collections from randomised studies analysing adjuvant tamoxifen treatment in early breast cancer demonstrated significant benefit of tamoxifen in the group with low stromal PDGFRβ, which was not observed in the group with high stromal PDGFRβ. In general terms these findings provide novel evidence, derived from analyses of randomised clinical studies, of response‐predictive capacity of a marker‐defined subset of CAFs and, more specifically, identify stromal PDGFRβ as a marker related to tamoxifen benefit in early breast cancer.
Collapse
Affiliation(s)
- Janna Paulsson
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet Stockholm Sweden
| | - Lisa Rydén
- Division of Surgery, Department of Clinical SciencesLund UniversityLundSweden; Department of SurgerySkåne University HospitalLundSweden
| | - Carina Strell
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet Stockholm Sweden
| | - Oliver Frings
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet Stockholm Sweden
| | - Nicholas P Tobin
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet Stockholm Sweden
| | - Tommy Fornander
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet Stockholm Sweden
| | - Jonas Bergh
- Department of Oncology-PathologyCancer Center Karolinska, Karolinska InstitutetStockholmSweden; Radiumhemmet, Karolinska University HospitalStockholmSweden
| | - Göran Landberg
- Department of Pathology Sahlgrenska Cancer Centre, University of Gothenburg Gothenburg Sweden
| | - Olle Stål
- Department of Clinical and Experimental Medicine Oncology, Linköping University Linköping Sweden
| | - Arne Östman
- Department of Oncology-Pathology Cancer Center Karolinska, Karolinska Institutet Stockholm Sweden
| |
Collapse
|
56
|
Abstract
Estrogens are important modulators of a broad spectrum of physiological functions in humans. However, despite their beneficial actions, a number of lines of evidence correlate the sustained exposure to exogenous estrogen with increased risk of the onset of various cancers. Mainly these steroid hormones induce their effects by binding and activating estrogen receptors (ERα and ERβ). These receptors belong to the family of ligand-regulated transcription factors, and upon activation they regulate the expression of different target genes by binding directly to specific DNA sequences. On the other hand, in recent years it has become clear that the G protein-coupled estrogen receptor 30 (GPR30/GPER) is able to mediate non-genomic action of estrogens in different cell contexts. In particular, GPER has been shown to specifically bind estrogens, and in turn to functionally cross-react with diverse cell signaling systems such as the epidermal growth factor receptor (EGFR) pathway, the Notch signaling pathway and the mitogen-activated protein kinases (MAPK) pathway. In this chapter we will present some of the different experimental techniques currently used to demonstrate the functional role of GPER in mediating non-genomic actions of estrogens, such as the dual luciferase assay, assessment of the involvement of GPER in the stimulation of cell migration in breast cancer cell lines and in cancer-associated fibroblasts, and chromatin immunoprecipitation assay. Overall, the experimental procedures described herein represent key instruments for assessing the biological role of GPER in mediating non-genomic signals of estrogen.
Collapse
Affiliation(s)
- Marco Pupo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Anna Maria Musti
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
- Institute for Clinical Neurobiology, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
57
|
Kalluri R. The biology and function of fibroblasts in cancer. NATURE REVIEWS. CANCER 2016. [PMID: 27550820 DOI: 10.1038/nrc.2016.73.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Among all cells, fibroblasts could be considered the cockroaches of the human body. They survive severe stress that is usually lethal to all other cells, and they are the only normal cell type that can be live-cultured from post-mortem and decaying tissue. Their resilient adaptation may reside in their intrinsic survival programmes and cellular plasticity. Cancer is associated with fibroblasts at all stages of disease progression, including metastasis, and they are a considerable component of the general host response to tissue damage caused by cancer cells. Cancer-associated fibroblasts (CAFs) become synthetic machines that produce many different tumour components. CAFs have a role in creating extracellular matrix (ECM) structure and metabolic and immune reprogramming of the tumour microenvironment with an impact on adaptive resistance to chemotherapy. The pleiotropic actions of CAFs on tumour cells are probably reflective of them being a heterogeneous and plastic population with context-dependent influence on cancer.
Collapse
Affiliation(s)
- Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| |
Collapse
|
58
|
Abstract
Among all cells, fibroblasts could be considered the cockroaches of the human body. They survive severe stress that is usually lethal to all other cells, and they are the only normal cell type that can be live-cultured from post-mortem and decaying tissue. Their resilient adaptation may reside in their intrinsic survival programmes and cellular plasticity. Cancer is associated with fibroblasts at all stages of disease progression, including metastasis, and they are a considerable component of the general host response to tissue damage caused by cancer cells. Cancer-associated fibroblasts (CAFs) become synthetic machines that produce many different tumour components. CAFs have a role in creating extracellular matrix (ECM) structure and metabolic and immune reprogramming of the tumour microenvironment with an impact on adaptive resistance to chemotherapy. The pleiotropic actions of CAFs on tumour cells are probably reflective of them being a heterogeneous and plastic population with context-dependent influence on cancer.
Collapse
Affiliation(s)
- Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| |
Collapse
|
59
|
Al-Howail HA, Hakami HA, Al-Otaibi B, Al-Mazrou A, Daghestani MH, Al-Jammaz I, Al-Khalaf HH, Aboussekhra A. PAC down-regulates estrogen receptor alpha and suppresses epithelial-to-mesenchymal transition in breast cancer cells. BMC Cancer 2016; 16:540. [PMID: 27465411 PMCID: PMC4964287 DOI: 10.1186/s12885-016-2583-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 07/19/2016] [Indexed: 12/31/2022] Open
Abstract
Background Triple-negative breast cancer (TNBC) is an aggressive histological subtype with limited treatment options and very poor prognosis following progression after standard chemotherapeutic regimens. Therefore, novel molecules and therapeutic options are urgently needed for this category of patients. Recently, we have identified PAC as a curcumin analogue with potent anti-cancer features. Methods HPLC was used to evaluate the stability of PAC and curcumin in PBS and also in circulating blood. Cytotoxicity/apoptosis was assessed in different breast cancer cell lines using propidium iodide/annexinV associated with flow cytometry. Furthermore, immunoblotting analysis determined the effects of PAC on different oncogenic proteins and pathways. Additionally, the real time xCELLigence RTCA technology was applied to investigate the effect of PAC on the cellular proliferation, migration and invasion capacities. Results PAC is more stable than curcumin in PBS and in circulating blood. Furthermore, we have shown differential sensitivity of estrogen receptor-alfa positive (ERα+) and estrogen receptor alfa negative (ERα−) breast cancer cells to PAC, which down-regulated ERα in both cell types. This led to complete disappearance of ERα in ERα− cells, which express very low level of this receptor. Interestingly, specific down-regulation of ERα in receptor positive cells increased the apoptotic response of these cells to PAC, confirming that ERα inhibits PAC-dependent induction of apoptosis, which could be mediated through ERα down-regulation. Additionally, PAC inhibited the proliferation and suppressed the epithelial-to-mesenchymal transition process in breast cancer cells, with higher efficiency on the TNBC subtype. This effect was also observed in vivo on tumor xenografts. Additionally, PAC suppressed the expression/secretion of 2 important cytokines IL-6 and MCP-1, and consequently inhibited the paracrine procarcinogenic effects of breast cancer cells on breast stromal fibroblasts. Conclusion These results indicate that PAC could be considered as important candidate for future therapeutic options against the devastating TNBC subtype.
Collapse
Affiliation(s)
- Huda A Al-Howail
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, MBC # 03, PO BOX 3354, Riyadh, 11211, Kingdom of Saudi Arabia
| | - Hana A Hakami
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, MBC # 03, PO BOX 3354, Riyadh, 11211, Kingdom of Saudi Arabia.,Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia.,Present Address: McGill University Health Center, Montreal, QC, Canada
| | - Basem Al-Otaibi
- Department of Cyclotron and Radiopharmaceuticals, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Kingdom of Saudi Arabia
| | - Amer Al-Mazrou
- Stem Cell Therapy Program, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Kingdom of Saudi Arabia
| | - Maha H Daghestani
- Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Ibrahim Al-Jammaz
- Department of Cyclotron and Radiopharmaceuticals, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Kingdom of Saudi Arabia
| | - Huda H Al-Khalaf
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, MBC # 03, PO BOX 3354, Riyadh, 11211, Kingdom of Saudi Arabia.,The National Center for Genomics Research, King Abdulaziz City for Science and Technology, Riyadh, 11211, Kingdom of Saudi Arabia
| | - Abdelilah Aboussekhra
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, MBC # 03, PO BOX 3354, Riyadh, 11211, Kingdom of Saudi Arabia.
| |
Collapse
|
60
|
Nass N, Dittmer A, Hellwig V, Lange T, Beyer JM, Leyh B, Ignatov A, Weiβenborn C, Kirkegaard T, Lykkesfeldt AE, Kalinski T, Dittmer J. Expression of transmembrane protein 26 (TMEM26) in breast cancer and its association with drug response. Oncotarget 2016; 7:38408-38426. [PMID: 27224909 PMCID: PMC5122400 DOI: 10.18632/oncotarget.9493] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/29/2016] [Indexed: 12/18/2022] Open
Abstract
We have previously shown that stromal cells desensitize breast cancer cells to the anti-estrogen fulvestrant and, along with it, downregulate the expression of TMEM26 (transmembrane protein 26). In an effort to study the function and regulation of TMEM26 in breast cancer cells, we found that breast cancer cells express non-glycosylated and N-glycosylated isoforms of the TMEM26 protein and demonstrate that N-glycosylation is important for its retention at the plasma membrane. Fulvestrant induced significant changes in expression and in the N-glycosylation status of TMEM26. In primary breast cancer, TMEM26 protein expression was higher in ERα (estrogen receptor α)/PR (progesterone receptor)-positive cancers. These data suggest that ERα is a major regulator of TMEM26. Significant changes in TMEM26 expression and N-glycosylation were also found, when MCF-7 and T47D cells acquired fulvestrant resistance. Furthermore, patients who received aromatase inhibitor treatment tend to have a higher risk of recurrence when tumoral TMEM26 protein expression is low. In addition, TMEM26 negatively regulates the expression of integrin β1, an important factor involved in endocrine resistance. Data obtained by spheroid formation assays confirmed that TMEM26 and integrin β1 can have opposite effects in breast cancer cells. These data are consistent with the hypothesis that, in ERα-positive breast cancer, TMEM26 may function as a tumor suppressor by impeding the acquisition of endocrine resistance. In contrast, in ERα-negative breast cancer, particularly triple-negative cancer, high TMEM26 expression was found to be associated with a higher risk of recurrence. This implies that TMEM26 has different functions in ERα-positive and -negative breast cancer.
Collapse
Affiliation(s)
- Norbert Nass
- Otto-von-Guericke-Universität Magdeburg, Institut für Pathologie, Magdeburg, Germany
| | - Angela Dittmer
- Klinik für Gynäkologie, Martin-Luther-Universität Halle-Wittenberg, Halle/Saale, Germany
| | - Vicky Hellwig
- Klinik für Gynäkologie, Martin-Luther-Universität Halle-Wittenberg, Halle/Saale, Germany
| | - Theresia Lange
- Klinik für Gynäkologie, Martin-Luther-Universität Halle-Wittenberg, Halle/Saale, Germany
| | - Johanna Mirjam Beyer
- Klinik für Gynäkologie, Martin-Luther-Universität Halle-Wittenberg, Halle/Saale, Germany
| | - Benjamin Leyh
- Klinik für Gynäkologie, Martin-Luther-Universität Halle-Wittenberg, Halle/Saale, Germany
| | - Atanas Ignatov
- Otto-von-Guericke-Universität Magdeburg, Universitätsfrauenklinik, Magdeburg, Germany
| | - Christine Weiβenborn
- Otto-von-Guericke-Universität Magdeburg, Universitätsfrauenklinik, Magdeburg, Germany
| | - Tove Kirkegaard
- Breast Cancer Group, Cell Death and Metabolism, Danish Cancer Society Research Center, Copenhagen, Denmark.,Present address: Department of Surgery, Koege Hospital, Koege, Denmark
| | - Anne E Lykkesfeldt
- Breast Cancer Group, Cell Death and Metabolism, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Thomas Kalinski
- Otto-von-Guericke-Universität Magdeburg, Institut für Pathologie, Magdeburg, Germany
| | - Jürgen Dittmer
- Klinik für Gynäkologie, Martin-Luther-Universität Halle-Wittenberg, Halle/Saale, Germany
| |
Collapse
|
61
|
Regier MC, Alarid ET, Beebe DJ. Progress towards understanding heterotypic interactions in multi-culture models of breast cancer. Integr Biol (Camb) 2016; 8:684-92. [PMID: 27097801 PMCID: PMC4993016 DOI: 10.1039/c6ib00001k] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Microenvironments in primary tumors and metastases include multiple cell types whose dynamic and reciprocal interactions are central to progression of the disease. However, the literature involving breast cancer studied in vitro is dominated by cancer cells in mono-culture or co-cultured with one other cell type. For in vitro studies of breast cancer the inclusion of multiple cell types has led to models that are more representative of in vivo behaviors and functions as compared to more traditional monoculture. Here, we review foundational co-culture techniques and their adaptation to multi-culture (including three or more cell types). Additionally, while macroscale methods involving conditioned media, direct contact, and indirect interactions have been informative, we examined many advances that have been made more recently using microscale systems with increased control over cellular and structural complexity. Throughout this discussion we consider the benefits and limitations of current multi-culture methods and the significant results they have produced.
Collapse
Affiliation(s)
- Mary C Regier
- Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| | | | | |
Collapse
|
62
|
De Souza A, Tinguely M, Burghart DR, Berisha A, Mertz KD, Kempf W. Characterization of the tumor microenvironment in primary cutaneous CD30-positive lymphoproliferative disorders: a predominance of CD163-positive M2 macrophages. J Cutan Pathol 2016; 43:579-88. [PMID: 27080437 DOI: 10.1111/cup.12719] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 01/13/2016] [Accepted: 04/11/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND The tumor microenvironment is essential for tumor survival, growth and progression. There are only a few studies on the tumor microenvironment in cutaneous CD30-positive lymphoproliferative disorders. METHODS We assessed the composition of the tumor microenvironment using immunohistochemistry studies in skin biopsies from cases diagnosed with lymphomatoid papulosis (LyP: 18 specimens), primary cutaneous anaplastic large-cell lymphoma (PC-ALCL: 8 specimens), and reactive diseases harboring CD30-positive cells (18 specimens). RESULTS The predominant cells present in LyP and PC-ALCL were CD163+ M2 macrophages (44.7%, 35%), followed by CD8+ tumor infiltrating lymphocytes (11%, 15%), FOXP3+ T-regulatory cells (9%, 4.5%) and programmed cell death 1(PD-1) + lymphocytes (2.2%, 6.8%). In contrast, CD30-positive reactive inflammatory and infectious disorders were characterized by higher numbers of CD123+ plasmacytoid dendritic cells (6.3%) when compared to LyP (1%), and PC-ALCL (1.1%). CONCLUSIONS Key differences exist between the microenvironment of CD30-positive lymphoproliferative disorders and reactive conditions harboring CD30-positive lymphocytes. The high number of tumor associated macrophages, and the close vicinity of these immune cells to the CD30-positive tumor cells might suggest that tumor associated macrophages have direct influence on tumorigenesis in LyP and ALCL. Therefore, modulation of M2 macrophages may represent a new therapeutic strategy in cutaneous CD30-positive lymphoproliferative disorders.
Collapse
Affiliation(s)
- Aieska De Souza
- Kempf and Pfaltz Histologische Diagnostik, Zürich, Switzerland.,Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | | | - Daniel R Burghart
- Kempf and Pfaltz Histologische Diagnostik, Zürich, Switzerland.,Department of Economics, California State University Sacramento, Sacramento, CA, USA
| | | | - Kirsten D Mertz
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Werner Kempf
- Kempf and Pfaltz Histologische Diagnostik, Zürich, Switzerland
| |
Collapse
|
63
|
Brodsky AS, Xiong J, Yang D, Schorl C, Fenton MA, Graves TA, Sikov WM, Resnick MB, Wang Y. Identification of stromal ColXα1 and tumor-infiltrating lymphocytes as putative predictive markers of neoadjuvant therapy in estrogen receptor-positive/HER2-positive breast cancer. BMC Cancer 2016; 16:274. [PMID: 27090210 PMCID: PMC4835834 DOI: 10.1186/s12885-016-2302-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 04/07/2016] [Indexed: 12/15/2022] Open
Abstract
Background The influence of the tumor microenvironment and tumor-stromal interactions on the heterogeneity of response within breast cancer subtypes have just begun to be explored. This study focuses on patients with estrogen receptor-positive/human epidermal growth factor receptor 2-positive (ER+/HER2+) breast cancer receiving neoadjuvant chemotherapy and HER2-targeted therapy (NAC+H), and was designed to identify novel predictive biomarkers by combining gene expression analysis and immunohistochemistry with pathologic response. Methods We performed gene expression profiling on pre-NAC+H tumor samples from responding (no or minimal residual disease at surgery) and non-responding patients. Gene set enrichment analysis identified potentially relevant pathways, and immunohistochemical staining of pre-treatment biopsies was used to measure protein levels of those pathways, which were correlated with pathologic response in both univariate and multivariate analysis. Results Increased expression of genes encoding for stromal collagens, including Col10A1, and reduced expression of immune-associated genes, reflecting lower levels of total tumor-infiltrating lymphocytes (TILs), were strongly associated with poor pathologic response. Lower TILs in tumor biopsies correlated with reduced likelihood of achieving an optimal pathologic response, but increased expression of the Col10A1 gene product, colXα1, had greater predictive value than stromal abundance for poor response (OR = 18.9, p = 0.003), and the combination of increased colXα1 expression and low TILs was significantly associated with poor response in multivariate analysis. ROC analysis suggests strong specificity and sensitivity for this combination in predicting treatment response. Conclusions Increased expression of stromal colXα1 and low TILs correlate with poor pathologic response in ER+/HER2+ breast tumors. Further studies are needed to confirm their predictive value and impact on long-term outcomes, and to determine whether this collagen exerts a protective effect on the cancer cells or simply reflects other factors within the tumor microenvironment. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2302-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexander S Brodsky
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, USA. .,Department of Pathology, Rhode Island Hospital and Lifespan Medical Center, Providence, RI, 02903, USA.
| | - Jinjun Xiong
- Department of Pathology, Women and Infants Hospital, Warren Alpert Medical School of Brown University, Providence, USA
| | - Dongfang Yang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, USA
| | - Christoph Schorl
- Molecular Biology, Cell Biology, & Biochemistry, Brown University, Providence, USA
| | - Mary Anne Fenton
- Department of Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, USA
| | - Theresa A Graves
- Department of Surgery, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, USA
| | - William M Sikov
- Program in Women's Oncology, Women and Infants Hospital, Warren Alpert Medical School of Brown University, Providence, USA
| | - Murray B Resnick
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, USA
| | - Yihong Wang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Lifespan Medical Center, Warren Alpert Medical School of Brown University, Providence, USA. .,Department of Pathology, Rhode Island Hospital and Lifespan Medical Center, Providence, RI, 02903, USA.
| |
Collapse
|
64
|
Abstract
The tumor microenvironment (TME) is a complex, heterogeneous, and dominant component of solid tumors. Cancer imaging strategies of a subset of characteristics of the TME are under active development, and currently used modalities and novel approaches are summarized in this article. Understanding the dynamic and evolving functions of the TME is critical to accurately inform imaging and clinical care of cancer. Novel insights into distinct roles of the TME in cancer progression urge careful interpretation of imaging data and impel the development of novel modalities.
Collapse
Affiliation(s)
- Valerie S LeBleu
- From the Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
65
|
Abstract
Cancer cells are distinguished from normal cells by increased proliferation and metabolism, loss of polarity control, and the potential to invade other tissues of the body. As hubs of signaling transduction, primary cilia have been linked to diverse developmental and degenerative disorders. Interestingly, loss of cilia has been observed in multiple malignant tumors, suggesting a potential suppressive role of cilia in cancer development. More recently, emerging studies began to unveil the bidirectional interaction of cilia and autophagy, a basic cellular clearance and recycling mechanism to regulate cell homeostasis. Here, we summarize the interplay between cilia and autophagy and discuss the roles of cilia in both autophagy and cancer.
Collapse
Affiliation(s)
- Muqing Cao
- Center for Autophagy Research; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX USA
| | - Qing Zhong
- Center for Autophagy Research; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX USA
| |
Collapse
|
66
|
Skolekova S, Matuskova M, Bohac M, Toro L, Durinikova E, Tyciakova S, Demkova L, Gursky J, Kucerova L. Cisplatin-induced mesenchymal stromal cells-mediated mechanism contributing to decreased antitumor effect in breast cancer cells. Cell Commun Signal 2016; 14:4. [PMID: 26759169 PMCID: PMC4710002 DOI: 10.1186/s12964-016-0127-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 01/06/2016] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Cells of the tumor microenvironment are recognized as important determinants of the tumor biology. The adjacent non-malignant cells can regulate drug responses of the cancer cells by secreted paracrine factors and direct interactions with tumor cells. RESULTS Human mesenchymal stromal cells (MSC) actively contribute to tumor microenvironment. Here we focused on their response to chemotherapy as during the treatment these cells become affected. We have shown that the secretory phenotype and behavior of mesenchymal stromal cells influenced by cisplatin differs from the naïve MSC. MSC were more resistant to the concentrations of cisplatin, which was cytotoxic for tumor cells. They did not undergo apoptosis, but a part of MSC population underwent senescence. However, MSC pretreatment with cisplatin led to changes in phosphorylation profiles of many kinases and also increased secretion of IL-6 and IL-8 cytokines. These changes in cytokine and phosphorylation profile of MSC led to increased chemoresistance and stemness of breast cancer cells. CONCLUSION Taken together here we suggest that the exposure of the chemoresistant cells in the tumor microenvironment leads to substantial alterations and might lead to promotion of acquired microenvironment-mediated chemoresistance and stemness.
Collapse
Affiliation(s)
- Svetlana Skolekova
- Laboratory of Molecular Oncology, Cancer Research Institute, Slovak Academy of Sciences, Vlarska 7, 833 91, Bratislava, Slovakia.
| | - Miroslava Matuskova
- Laboratory of Molecular Oncology, Cancer Research Institute, Slovak Academy of Sciences, Vlarska 7, 833 91, Bratislava, Slovakia.
| | - Martin Bohac
- Department of Plastic, Aesthetic and Reconstructive Surgery, University Hospital, Bratislava, Slovakia.
| | - Lenka Toro
- Laboratory of Molecular Oncology, Cancer Research Institute, Slovak Academy of Sciences, Vlarska 7, 833 91, Bratislava, Slovakia.
| | | | | | - Lucia Demkova
- Laboratory of Molecular Oncology, Cancer Research Institute, Slovak Academy of Sciences, Vlarska 7, 833 91, Bratislava, Slovakia.
| | - Jan Gursky
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 5, Olomouc, Czech Republic.
| | - Lucia Kucerova
- Laboratory of Molecular Oncology, Cancer Research Institute, Slovak Academy of Sciences, Vlarska 7, 833 91, Bratislava, Slovakia.
| |
Collapse
|
67
|
A Role for Notch Signalling in Breast Cancer and Endocrine Resistance. Stem Cells Int 2016; 2016:2498764. [PMID: 26880941 PMCID: PMC4736972 DOI: 10.1155/2016/2498764] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/20/2015] [Indexed: 12/16/2022] Open
Abstract
Over the past decade, there has been growing interest in the Notch signalling pathway within the breast cancer field. This interest stemmed initially from the observation that Notch signalling is aberrantly activated in breast cancer and its effects on various cellular processes including proliferation, apoptosis, and cancer stem cell activity. However more recently, elevated Notch signalling has been correlated with therapy resistance in oestrogen receptor-positive breast cancer. As a result, inhibiting Notch signalling with therapeutic agents is being explored as a promising treatment option for breast cancer patients.
Collapse
|
68
|
Leonard F, Godin B. 3D In Vitro Model for Breast Cancer Research Using Magnetic Levitation and Bioprinting Method. Methods Mol Biol 2016; 1406:239-51. [PMID: 26820961 DOI: 10.1007/978-1-4939-3444-7_21] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor microenvironment composition and architecture are known as a major factor in orchestrating the tumor growth and its response to various therapies. In this context, in vivo studies are necessary to evaluate the responses. However, while tumor cells can be of human origin, tumor microenvironment in the in vivo models is host-based. On the other hand, in vitro studies in a flat monoculture of tumor cells (the most frequently used in vitro tumor model) are unable to recapitulate the complexity of tumor microenvironment. Three-dimensional (3D) in vitro cell cultures of tumor cells have been proven to be an important experimental tool in understanding mechanisms of tumor growth, response to therapeutics, and transport of nutrients/drugs. We have recently described a novel tool to create 3D co-cultures of tumor cells and cells in the tumor microenvironment. Our method utilizes magnetic manipulation/levitation of the specific ratios of tumor cells and cells in the tumor microenvironment (from human or animal origin) aiding in the formation of tumor spheres with defined cellular composition and density, as quickly as within 24 h. This chapter describes the experimental protocols developed to model the 3D structure of the cancer environment using the above method.
Collapse
Affiliation(s)
- Fransisca Leonard
- Department of Nanomedicine, Houston Methodist Research Institute, R8-213, 6670 Bertner st, Houston, TX, 77030, USA
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, R8-213, 6670 Bertner st, Houston, TX, 77030, USA.
| |
Collapse
|
69
|
Recouvreux S, Sampayo R, Bessone MID, Simian M. Microenvironment and endocrine resistance in breast cancer: Friend or foe? World J Clin Oncol 2015; 6:207-211. [PMID: 26677432 PMCID: PMC4675904 DOI: 10.5306/wjco.v6.i6.207] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 08/18/2015] [Accepted: 10/13/2015] [Indexed: 02/06/2023] Open
Abstract
Breast cancer affects one in eight women around the world. Seventy five percent of these patients have tumors that are estrogen receptor positive and as a consequence receive endocrine therapy. However, about one third eventually develop resistance and cancer reappears. In the last decade our vision of cancer has evolved to consider it more of a tissue-related disease than a cell-centered one. This editorial argues that we are only starting to understand the role the tumor microenvironment plays in therapy resistance in breast cancer. The development of new therapeutic strategies that target the microenvironment will come when we clearly understand this extremely complicated scenario. As such, and as a scientific community, we have extremely challenging work ahead. We share our views regarding these matters.
Collapse
|
70
|
Reinert T, Barrios CH. Optimal management of hormone receptor positive metastatic breast cancer in 2016. Ther Adv Med Oncol 2015; 7:304-20. [PMID: 26557899 PMCID: PMC4622303 DOI: 10.1177/1758834015608993] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hormone receptor positive tumors represent the most common form of breast cancer and account for most of the deaths from the disease. Endocrine therapy represents the main initial therapeutic strategy for these patients and has been associated with significant clinical benefits in a majority of patients. While in early stages endocrine therapy is administered as part of a curative approach once clinical metastases develop, the disease is considered incurable and the main management objectives are tumor control and quality of life. The two major clinical paradigms of always indicating endocrine therapy in the absence of visceral crises and sequencing endocrine treatments have been guiding our therapeutic approach to these patients. However, for many decades, we have delivered endocrine therapy with a 'one size fits all' approach by applying agents that interfere with hormone receptor signaling equally in every clinical patient scenario. We have been unable to incorporate the well-known biologic principle of different degrees of hormone receptor dependency in our therapeutic recommendations. Recent developments in the understanding of molecular interactions of hormone signaling with other important growth factor, metabolic and cell division pathways have opened the possibility of improving results by modulating hormone signaling and interfering with resistance mechanisms yet to be fully understood. Unfortunately, limitations in the design of trials conducted in this area have made it difficult to develop predictive biomarkers and most of the new combinations with targeted agents, even though showing improvements in clinical endpoints, have been directed to an unselected population of patients. In this review we explore some of the current and most relevant literature in the management of hormone receptor positive advance breast cancer.
Collapse
Affiliation(s)
- Tomas Reinert
- Instituto do Câncer, Sistema de Saúde Mãe de Deus, Porto Alegre, RS, Brazil
| | - Carlos H. Barrios
- PUCRS School of Medicine, Department of Medicine, Padre Chagas 66/203, CEP 90 570 080, Porto Alegre, RS, Brazil
| |
Collapse
|
71
|
Giussani M, Merlino G, Cappelletti V, Tagliabue E, Daidone MG. Tumor-extracellular matrix interactions: Identification of tools associated with breast cancer progression. Semin Cancer Biol 2015; 35:3-10. [PMID: 26416466 DOI: 10.1016/j.semcancer.2015.09.012] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 09/23/2015] [Indexed: 12/18/2022]
Abstract
Several evidences support the concept that cancer development and progression are not entirely cancer cell-autonomous processes, but may be influenced, and possibly driven, by cross-talk between cancer cells and the surrounding microenvironment in which, besides immune cells, stromal cells and extracellular matrix (ECM) play a major role in regulating distinct biologic processes. Stroma and ECM-related signatures proved to influence breast cancer progression, and to contribute to the identification of tumor phenotypes resistant to cytotoxic and hormonal treatments. The possible clinical implications of the interplay between tumor cells and the microenvironment, with special reference to ECM remodelling, will be discussed in this review.
Collapse
Affiliation(s)
- Marta Giussani
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G.A. Amadeo, 42, 20133 Milan, Italy.
| | - Giuseppe Merlino
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G.A. Amadeo, 42, 20133 Milan, Italy.
| | - Vera Cappelletti
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G.A. Amadeo, 42, 20133 Milan, Italy.
| | - Elda Tagliabue
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G.A. Amadeo, 42, 20133 Milan, Italy.
| | - Maria Grazia Daidone
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G.A. Amadeo, 42, 20133 Milan, Italy.
| |
Collapse
|
72
|
Dittmer J. The role of the transcription factor Ets1 in carcinoma. Semin Cancer Biol 2015; 35:20-38. [PMID: 26392377 DOI: 10.1016/j.semcancer.2015.09.010] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/16/2015] [Accepted: 09/16/2015] [Indexed: 12/12/2022]
Abstract
Ets1 belongs to the large family of the ETS domain family of transcription factors and is involved in cancer progression. In most carcinomas, Ets1 expression is linked to poor survival. In breast cancer, Ets1 is primarily expressed in the triple-negative subtype, which is associated with unfavorable prognosis. Ets1 contributes to the acquisition of cancer cell invasiveness, to EMT (epithelial-to-mesenchymal transition), to the development of drug resistance and neo-angiogenesis. The aim of this review is to summarize the current knowledge on the functions of Ets1 in carcinoma progression and on the mechanisms that regulate Ets1 activity in cancer.
Collapse
Affiliation(s)
- Jürgen Dittmer
- Clinic for Gynecology, Martin Luther University Halle-Wittenberg, Germany.
| |
Collapse
|
73
|
Kim SA, Lee EK, Kuh HJ. Co-culture of 3D tumor spheroids with fibroblasts as a model for epithelial-mesenchymal transition in vitro. Exp Cell Res 2015; 335:187-96. [PMID: 26022665 DOI: 10.1016/j.yexcr.2015.05.016] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 04/21/2015] [Accepted: 05/18/2015] [Indexed: 12/20/2022]
Abstract
Epithelial-mesenchymal transition (EMT) acts as a facilitator of metastatic dissemination in the invasive margin of malignant tumors where active tumor-stromal crosstalks take place. Co-cultures of cancer cells with cancer-associated fibroblasts (CAFs) are often used as in vitro models of EMT. We established a tumor-fibroblast proximity co-culture using HT-29 tumor spheroids (TSs) with CCD-18 co fibroblasts. When co-cultured with TSs, CCD-18 co appeared activated, and proliferative activity as well as cell migration increased. Expression of fibronectin increased whereas laminin and type I collagen decreased in TSs co-cultured with fibroblasts compared to TSs alone, closely resembling the margin of in vivo xenograft tissue. Active TGFβ1 in culture media significantly increased in TS co-cultures but not in 2D co-cultures of cancer cells-fibroblasts, indicating that 3D context-associated factors from TSs may be crucial to crosstalks between cancer cells and fibroblasts. We also observed in TSs co-cultured with fibroblasts increased expression of α-SMA, EGFR and CTGF; reduced expression of membranous β-catenin and E-cadherin, together suggesting an EMT-like changes similar to a marginal region of xenograft tissue in vivo. Overall, our in vitro TS-fibroblast proximity co-culture mimics the EMT-state of the invasive margin of in vivo tumors in early metastasis.
Collapse
Affiliation(s)
- Sun-Ah Kim
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 137-701, Republic of Korea.
| | - Eun Kyung Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 137-701, Republic of Korea; Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul 137-701, Republic of Korea.
| | - Hyo-Jeong Kuh
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 137-701, Republic of Korea; Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul 137-701, Republic of Korea.
| |
Collapse
|
74
|
Abstract
Oestrogen receptor-positive (ER(+)) breast cancer is a major cause of cancer death in women. Although aromatase inhibitors suppress the function of ER and reduce the risk of recurrence, therapeutic resistance is common and essentially inevitable in advanced disease. This Review considers both genomic and cell biological explanations as to why ER(+) breast cancer cells persist, progress and cause an incurable, lethal, systemic disease. The design and outcomes of clinical trials are considered with the perspective that resistance mechanisms are heterogeneous, and therefore biomarker and somatic mutation-based stratification and eligibility will be essential for improvements in patient outcomes.
Collapse
Affiliation(s)
- Cynthia X Ma
- Division of Oncology, Department of Medicine, Siteman Cancer Center, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, Missouri 63110, USA
| | - Tomás Reinert
- Department of Medical Oncology, Instituto Nacional de Câncer (INCA), Praça da Cruz Vermelha, 23, 20230-130, Rio de Janeiro, Brazil
| | - Izabela Chmielewska
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8 St., 20-954, Lublin, Poland
| | - Matthew J Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston 77030, Texas, USA
| |
Collapse
|
75
|
Han B, Wang TD, Shen SM, Yu Y, Mao C, Yao ZJ, Wang LS. Annonaceous acetogenin mimic AA005 induces cancer cell death via apoptosis inducing factor through a caspase-3-independent mechanism. BMC Cancer 2015; 15:139. [PMID: 25885900 PMCID: PMC4379763 DOI: 10.1186/s12885-015-1133-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 02/24/2015] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Annonaceous acetogenins are a family of natural products with antitumor activities. Annonaceous acetogenin mimic AA005 reportedly inhibits mammalian mitochondrial NADH-ubiquinone reductase (Complex I) and induces gastric cancer cell death. However, the mechanisms underlying its cell-death-inducing activity are unclear. METHODS We used SW620 colorectal adenocarcinoma cells to study AA005 cytotoxic activity. Cell deaths were determined by Trypan blue assay and flow cytometry, and related proteins were characterized by western blot. Immunofluorescence and subcellular fractionation were used to evaluate AIF nuclear translocation. Reactive oxygen species were assessed by using redox-sensitive dye DCFDA. RESULTS AA005 induces a unique type of cell death in colorectal adenocarcinoma cells, characterized by lack of caspase-3 activation or apoptotic body formation, sensitivity to poly (ADP-ribose) polymerase inhibitor Olaparib (AZD2281) but not pan-caspase inhibitor Z-VAD.fmk, and dependence on apoptosis-inducing factor (AIF). AA005 treatment also reduced expression of mitochondrial Complex I components, and leads to accumulation of intracellular reactive oxygen species (ROS) at the early stage. Blocking ROS formation significantly suppresses AA005-induced cell death in SW620 cells. Moreover, blocking activation of RIP-1 by necroptosis inhibitor necrotatin-1 inhibits AIF translocation and partially suppresses AA005-induced cell death in SW620 cells demonstrating that RIP-1 protein may be essential for cell death. CONCLUSIONS AA005 may trigger the cell death via mediated by AIF through caspase-3 independent pathway. Our work provided new mechanisms for AA005-induced cancer cell death and novel clues for cancer treatment via AIF dependent cell death.
Collapse
Affiliation(s)
- Bing Han
- Center for Molecular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Tong-Dan Wang
- Center for Molecular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Shao-Ming Shen
- Center for Molecular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Yun Yu
- Center for Molecular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Chan Mao
- State Key Laboratory of Coordination Chemistry, Institute of Chemical Biology and Drug Innovation, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, P. R. China.
| | - Zhu-Jun Yao
- State Key Laboratory of Coordination Chemistry, Institute of Chemical Biology and Drug Innovation, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, P. R. China. .,Shanghai Universities E-Institute for Chemical Biology, Shanghai, 200025, China.
| | - Li-Shun Wang
- Center for Molecular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Shanghai Universities E-Institute for Chemical Biology, Shanghai, 200025, China.
| |
Collapse
|
76
|
Valta M, Fagerlund K, Suominen M, Halleen J, Tuomela J. Importance of microenvironment in preclinical models of breast and prostate cancer. World J Pharmacol 2015; 4:47-57. [DOI: 10.5497/wjp.v4.i1.47] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 11/18/2014] [Accepted: 01/19/2015] [Indexed: 02/06/2023] Open
Abstract
The majority of cancer drugs entering clinical trials fail to reach the market due to poor efficacy. Preclinical efficacy has been traditionally tested using subcutaneous xenograft models that are cheap, fast and easy to perform. However, these models lack the correct tumor microenvironment, leading to poor clinical predictivity. Selecting compounds for clinical trials based on efficacy results obtained from subcutaneous xenograft models may therefore be one important reason for the high failure rates. In this review we concentrate in describing the role and importance of the tumor microenvironment in progression of breast and prostate cancer, and describe some breast and prostate cancer cell lines that are widely used in preclinical studies. We go through different preclinical efficacy models that incorporate the tissue microenvironment and should therefore be clinically more predictive than subcutaneous xenografts. These include three-dimensional cell culture models, orthotopic and metastasis models, humanized and transgenic mouse models, and patient-derived xenografts. Different endpoint measurements and applicable imaging techniques are also discussed. We conclude that models that incorporate the tissue microenvironment should be increasingly used in preclinical efficacy studies to reduce the current high attrition rates of cancer drugs in clinical trials.
Collapse
|
77
|
Chen F, Zhuang X, Lin L, Yu P, Wang Y, Shi Y, Hu G, Sun Y. New horizons in tumor microenvironment biology: challenges and opportunities. BMC Med 2015; 13:45. [PMID: 25857315 PMCID: PMC4350882 DOI: 10.1186/s12916-015-0278-7] [Citation(s) in RCA: 522] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/16/2015] [Indexed: 02/07/2023] Open
Abstract
The tumor microenvironment (TME) is being increasingly recognized as a key factor in multiple stages of disease progression, particularly local resistance, immune-escaping, and distant metastasis, thereby substantially impacting the future development of frontline interventions in clinical oncology. An appropriate understanding of the TME promotes evaluation and selection of candidate agents to control malignancies at both the primary sites as well as the metastatic settings. This review presents a timely outline of research advances in TME biology and highlights the prospect of targeting the TME as a critical strategy to overcome acquired resistance, prevent metastasis, and improve therapeutic efficacy. As benign cells in TME niches actively modulate response of cancer cells to a broad range of standard chemotherapies and targeted agents, cancer-oriented therapeutics should be combined with TME-targeting treatments to achieve optimal clinical outcomes. Overall, a body of updated information is delivered to summarize recently emerging and rapidly progressing aspects of TME studies, and to provide a significant guideline for prospective development of personalized medicine, with the long term aim of providing a cure for cancer patients.
Collapse
Affiliation(s)
- Fei Chen
- />Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiaotong University School of Medicine, Shanghai, 200031 China
| | - Xueqian Zhuang
- />Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiaotong University School of Medicine, Shanghai, 200031 China
| | - Liangyu Lin
- />Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiaotong University School of Medicine, Shanghai, 200031 China
| | - Pengfei Yu
- />Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiaotong University School of Medicine, Shanghai, 200031 China
| | - Ying Wang
- />Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiaotong University School of Medicine, Shanghai, 200031 China
| | - Yufang Shi
- />Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiaotong University School of Medicine, Shanghai, 200031 China
- />Soochow Institutes for Translational Medicine, Soochow University, Suzhou, 215123 China
| | - Guohong Hu
- />Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiaotong University School of Medicine, Shanghai, 200031 China
| | - Yu Sun
- />Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiaotong University School of Medicine, Shanghai, 200031 China
- />VA Seattle Medical Center, Seattle, WA 98108 USA
- />Department of Medicine, University of Washington, Seattle, WA 98195 USA
- />Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) and Shanghai Jiaotong University School of Medicine (SJTUSM), 320 Yue Yang Road, Biological Research Building A, Shanghai, 200031 China
| |
Collapse
|
78
|
Skvortsova I. It is well established that affected intracellular signaling is associated with carcinogenesis, cancer progression and tumor sensitivity to currently existing therapeutic approaches. Semin Cancer Biol 2015; 31:1-2. [PMID: 25559282 DOI: 10.1016/j.semcancer.2014.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Ira Skvortsova
- Laboratory for Experimental and Translational Research on Radiation Oncology, Department of Therapeutic Radiology and Oncology, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|