1
|
Doctor A, Laube M, Meister S, Kiss OC, Kopka K, Hauser S, Pietzsch J. Combined PET Radiotracer Approach Reveals Insights into Stromal Cell-Induced Metabolic Changes in Pancreatic Cancer In Vitro and In Vivo. Cancers (Basel) 2024; 16:3393. [PMID: 39410013 PMCID: PMC11475921 DOI: 10.3390/cancers16193393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objective Pancreatic stellate cells (PSCs) in pancreatic adenocarcinoma (PDAC) are producing extracellular matrix, which promotes the formation of a dense fibrotic microenvironment. This makes PDAC a highly heterogeneous tumor-stroma-driven entity, associated with reduced perfusion, limited oxygen supply, high interstitial fluid pressure, and limited bioavailability of therapeutic agents. Methods In this study, spheroid and tumor xenograft models of human PSCs and PanC-1 cells were characterized radiopharmacologically using a combined positron emission tomography (PET) radiotracer approach. [18F]FDG, [18F]FMISO, and [18F]FAPI-74 were employed to monitor metabolic activity, hypoxic metabolic state, and functional expression of fibroblast activation protein alpha (FAPα), a marker of activated PSCs. Results In vitro, PanC-1 and multi-cellular tumor spheroids demonstrated comparable glucose uptake and hypoxia, whereas FAPα expression was significantly higher in PSC spheroids. In vivo, glucose uptake as well as the transition to hypoxia were comparable in PanC-1 and multi-cellular xenograft models. In mice injected with PSCs, FAPα expression decreased over a period of four weeks post-injection, which was attributed to the successive death of PSCs. In contrast, FAPα expression increased in both PanC-1 and multi-cellular xenograft models over time due to invasion of mouse fibroblasts. Conclusion The presented models are suitable for subsequently characterizing stromal cell-induced metabolic changes in tumors using noninvasive molecular imaging techniques.
Collapse
Affiliation(s)
- Alina Doctor
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| | - Markus Laube
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
| | - Sebastian Meister
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
| | - Oliver C. Kiss
- Department of Targetry, Target Chemistry and Radiopharmacy, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany;
| | - Klaus Kopka
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, Partner Site Dresden, University Cancer Center (UCC), Fetscherstraße 74, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| |
Collapse
|
2
|
Castro-Abril H, Heras J, Del Barrio J, Paz L, Alcaine C, Aliácar MP, Garzón-Alvarado D, Doblaré M, Ochoa I. The Role of Mechanical Properties and Structure of Type I Collagen Hydrogels on Colorectal Cancer Cell Migration. Macromol Biosci 2023; 23:e2300108. [PMID: 37269065 DOI: 10.1002/mabi.202300108] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/05/2023] [Indexed: 06/04/2023]
Abstract
Mechanical interactions between cells and their microenvironment play an important role in determining cell fate, which is particularly relevant in metastasis, a process where cells invade tissue matrices with different mechanical properties. In vitro, type I collagen hydrogels have been commonly used for modeling the microenvironment due to its ubiquity in the human body. In this work, the combined influence of the stiffness of these hydrogels and their ultrastructure on the migration patterns of HCT-116 and HT-29 spheroids are analyzed. For this, six different types of pure type I collagen hydrogels by changing the collagen concentration and the gelation temperature are prepared. The stiffness of each sample is measured and its ultrastructure is characterized. Cell migration studies are then performed by seeding the spheroids in three different spatial conditions. It is shown that changes in the aforementioned parameters lead to differences in the mechanical stiffness of the matrices as well as the ultrastructure. These differences, in turn, lead to distinct cell migration patterns of HCT-116 and HT-29 spheroids in either of the spatial conditions tested. Based on these results, it is concluded that the stiffness and the ultrastructural organization of the matrix can actively modulate cell migration behavior in colorectal cancer spheroids.
Collapse
Affiliation(s)
- Hector Castro-Abril
- Tissue Microenvironment lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, 50018, Spain
- Biomimetics Lab, National University of Colombia, Bogotá, 111321, Colombia
| | - Jónathan Heras
- Grupo de Informática, University of La Rioja, La Rioja, 26006, Spain
| | - Jesús Del Barrio
- Instituto de Nanociencia y Materiales de Aragón (INMA), Department of Organic Chemistry, CSIC-University of Zaragoza, Zaragoza, 50018, Spain
| | - Laura Paz
- Tissue Microenvironment lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, 50018, Spain
- Centro Investigación Biomédica en Red. Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, 50018, Spain
| | - Clara Alcaine
- Tissue Microenvironment lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, 50018, Spain
| | - Marina Pérez Aliácar
- Tissue Microenvironment lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, 50018, Spain
- Centro Investigación Biomédica en Red. Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, 50018, Spain
| | | | - Manuel Doblaré
- Tissue Microenvironment lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, 50018, Spain
- Centro Investigación Biomédica en Red. Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, 50018, Spain
- Nanjing Tech University, Nanjing, 50018, China
| | - Ignacio Ochoa
- Tissue Microenvironment lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, 50018, Spain
- Centro Investigación Biomédica en Red. Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, 50018, Spain
- Nanjing Tech University, Nanjing, 50018, China
| |
Collapse
|
3
|
Sourouni M, Opitz C, Radke I, Kiesel L, Tio J, Götte M, von Wahlde M. Establishment of a
3D
co‐culture model to investigate the role of primary fibroblasts in ductal carcinoma in situ of the breast. Cancer Rep (Hoboken) 2022; 6:e1771. [PMID: 36534078 PMCID: PMC10075300 DOI: 10.1002/cnr2.1771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 12/01/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Ductal carcinoma in situ (DCIS) is a precursor form of breast cancer. 13%-50% of these lesions will progress to invasive breast cancer, but the individual progression risk cannot be estimated. Therefore, all patients receive the same therapy, resulting in potential overtreatment of a large proportion of patients. AIMS The role of the tumor microenvironment (TME) and especially of fibroblasts appears to be critical in DCIS development and a better understanding of their role may aid individualized treatment. METHODS AND RESULTS Primary fibroblasts isolated from benign or malignant punch biopsies of the breast and MCF10DCIS.com cells were seeded in a 3D cell culture system. The fibroblasts were cultured in a type I collagen layer beneath a Matrigel layer with MCF10DCIS.com cells. Dye-quenched (DQ) fluorescent collagen I and IV were used in collagen and Matrigel layer respectively to demonstrate proteolysis. Confocal microscopy was performed on day 2, 7, and 14 to reveal morphological changes, which could indicate the transition to an invasive phenotype. MCF10DCIS.com cells form smooth, round spheroids in co-culture with non-cancer associated fibroblasts (NAFs). Spheroids in co-culture with tumor-associated fibroblasts (TAFs) appear irregularly shaped and with an uneven surface; similar to spheroids formed from invasive cells. Therefore, these morphological changes represent the progression of an in situ to an invasive phenotype. In addition, TAFs show a higher proteolytic activity compared to NAFs. The distance between DCIS cells and fibroblasts decreases over time. CONCLUSION The TAFs seem to play an important role in the progression of DCIS to invasive breast cancer. The better characterization of the TME could lead to the identification of DCIS lesions with high or low risk of progression. This could enable personalized oncological therapy, prevention of overtreatment and individualized hormone replacement therapy after DCIS.
Collapse
Affiliation(s)
- Marina Sourouni
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Carl Opitz
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Isabel Radke
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Ludwig Kiesel
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Joke Tio
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Martin Götte
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| | - Marie‐Kristin von Wahlde
- Department of Obstetrics and Gynecology, Breast Center University Hospital Münster Münster Germany
| |
Collapse
|
4
|
Nowińska K, Jabłońska K, Ciesielska U, Piotrowska A, Haczkiewicz-Leśniak K, Pawełczyk K, Podhorska-Okołów M, Dzięgiel P. Association of Irisin/FNDC5 with ERRα and PGC-1α Expression in NSCLC. Int J Mol Sci 2022; 23:ijms232214204. [PMID: 36430689 PMCID: PMC9694131 DOI: 10.3390/ijms232214204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
The rapid growth and division of cancer cells are associated with mitochondrial biogenesis or switching to glycolysis. ERRα, PGC-1α and irisin/FNDC5 are some of the proteins that can influence these processes. The aim of this study was to determine the correlation of these proteins in non-small cell lung cancer (NSCLC) and to investigate their association with clinicopathological parameters. Immunohistochemistry reactions were performed on tissue microarrays (860 NSCLC, 140 non-malignant lung tissue). The normal fibroblast cell line (IMR-90) and lung cancer cell lines (NCI-H1703 and NCI-H522) were used as co-cultures. The mRNA levels of FNDC5 and ESRRA (encoding ERRα) were assessed in IMR-90 cells after co-culture with lung cancer cells. We observed a decreased level of ERRα with an increase in tumor size (T), stages of the disease, and lymph node metastases (N). In the adenocarcinoma (AC) subtype, patients with a higher ERRα expression had significantly longer overall survival. A moderate positive correlation was observed between FNDC5 mRNA and ESRRA mRNA in NSCLCs. The expression of FNDC5 mRNA in IMR-90 cells increased after 24 h, and ESRRA gene expression increased after 48 h of co-culture. The ERRα receptor with PGC-1α participates in the control of FNDC5/irisin expression. Normal fibroblasts revealed an upregulation of the FNDC5 and ESRRA genes under the influence of lung cancer cells.
Collapse
Affiliation(s)
- Katarzyna Nowińska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Correspondence:
| | - Karolina Jabłońska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Urszula Ciesielska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Aleksandra Piotrowska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | | | - Konrad Pawełczyk
- Lower Silesian Centre of Oncology, Pulmonology and Haematology, 53-439 Wroclaw, Poland
| | - Marzenna Podhorska-Okołów
- Division of Ultrastructural Research, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Physiotherapy, Wroclaw University School of Physical Education, 51-612 Wroclaw, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Physiotherapy, Wroclaw University School of Physical Education, 51-612 Wroclaw, Poland
| |
Collapse
|
5
|
Hu H, Piotrowska Z, Hare PJ, Chen H, Mulvey HE, Mayfield A, Noeen S, Kattermann K, Greenberg M, Williams A, Riley AK, Wilson JJ, Mao YQ, Huang RP, Banwait MK, Ho J, Crowther GS, Hariri LP, Heist RS, Kodack DP, Pinello L, Shaw AT, Mino-Kenudson M, Hata AN, Sequist LV, Benes CH, Niederst MJ, Engelman JA. Three subtypes of lung cancer fibroblasts define distinct therapeutic paradigms. Cancer Cell 2021; 39:1531-1547.e10. [PMID: 34624218 PMCID: PMC8578451 DOI: 10.1016/j.ccell.2021.09.003] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/27/2021] [Accepted: 09/03/2021] [Indexed: 12/20/2022]
Abstract
Cancer-associated fibroblasts (CAFs) are highly heterogeneous. With the lack of a comprehensive understanding of CAFs' functional distinctions, it remains unclear how cancer treatments could be personalized based on CAFs in a patient's tumor. We have established a living biobank of CAFs derived from biopsies of patients' non-small lung cancer (NSCLC) that encompasses a broad molecular spectrum of CAFs in clinical NSCLC. By functionally interrogating CAF heterogeneity using the same therapeutics received by patients, we identify three functional subtypes: (1) robustly protective of cancers and highly expressing HGF and FGF7; (2) moderately protective of cancers and highly expressing FGF7; and (3) those providing minimal protection. These functional differences among CAFs are governed by their intrinsic TGF-β signaling, which suppresses HGF and FGF7 expression. This CAF functional classification correlates with patients' clinical response to targeted therapies and also associates with the tumor immune microenvironment, therefore providing an avenue to guide personalized treatment.
Collapse
Affiliation(s)
- Haichuan Hu
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA.
| | - Zofia Piotrowska
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Patricia J Hare
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Huidong Chen
- Massachusetts General Hospital and Department of Pathology, Harvard Medical School, Boston, MA 02114, USA; Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Hillary E Mulvey
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Aislinn Mayfield
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Sundus Noeen
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Krystina Kattermann
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Max Greenberg
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - August Williams
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Amanda K Riley
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | | | - Ying-Qing Mao
- RayBiotech Inc, Norcross, GA 30092, USA; RayBiotech Inc, Guangzhou, Guangdong 510630, China
| | - Ruo-Pan Huang
- RayBiotech Inc, Norcross, GA 30092, USA; RayBiotech Inc, Guangzhou, Guangdong 510630, China; Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong 510095, China
| | - Mandeep K Banwait
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Jeffrey Ho
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Giovanna S Crowther
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Lida P Hariri
- Massachusetts General Hospital and Department of Pathology, Harvard Medical School, Boston, MA 02114, USA
| | - Rebecca S Heist
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - David P Kodack
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Luca Pinello
- Massachusetts General Hospital and Department of Pathology, Harvard Medical School, Boston, MA 02114, USA; Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Alice T Shaw
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Mari Mino-Kenudson
- Massachusetts General Hospital and Department of Pathology, Harvard Medical School, Boston, MA 02114, USA
| | - Aaron N Hata
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Lecia V Sequist
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Cyril H Benes
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, MA 02114, USA.
| | | | | |
Collapse
|
6
|
Biological Mechanisms and Therapeutic Opportunities in Mammographic Density and Breast Cancer Risk. Cancers (Basel) 2021; 13:cancers13215391. [PMID: 34771552 PMCID: PMC8582527 DOI: 10.3390/cancers13215391] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 12/13/2022] Open
Abstract
Mammographic density is an important risk factor for breast cancer; women with extremely dense breasts have a four to six fold increased risk of breast cancer compared to women with mostly fatty breasts, when matched with age and body mass index. High mammographic density is characterised by high proportions of stroma, containing fibroblasts, collagen and immune cells that suggest a pro-tumour inflammatory microenvironment. However, the biological mechanisms that drive increased mammographic density and the associated increased risk of breast cancer are not yet understood. Inflammatory factors such as monocyte chemotactic protein 1, peroxidase enzymes, transforming growth factor beta, and tumour necrosis factor alpha have been implicated in breast development as well as breast cancer risk, and also influence functions of stromal fibroblasts. Here, the current knowledge and understanding of the underlying biological mechanisms that lead to high mammographic density and the associated increased risk of breast cancer are reviewed, with particular consideration to potential immune factors that may contribute to this process.
Collapse
|
7
|
Bhardwaj P, Goda JS, Pai V, Chaudhari P, Mohanty B, Pai T, Vishwakarma K, Thorat R, Wadasadawala T, Banerjee R. Ultrasound augments on-demand breast tumor radiosensitization and apoptosis through a tri-responsive combinatorial delivery theranostic platform. NANOSCALE 2021; 13:17077-17092. [PMID: 34622906 DOI: 10.1039/d1nr04211d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Advanced inoperable triple-negative breast cancer (TNBC) comprises aggressive tumors with a modest pathological response to neoadjuvant chemotherapy. The concomitant use of chemoradiotherapy improves the pathological response rates. However, the dose-dependent systemic toxicity of clinical radiosensitizers with poor circulation half-life and limited passive bioavailability limits their clinical utility. We address these challenges by rationally designing a stealth and tumor microenvironment responsive nano-conjugate platform for the ultrasound-mediated on-demand spatio-temporal delivery of plant flavonoid curcumin as a combinatorial regimen with clinically approved paclitaxel for the neoadjuvant chemoradiotherapy of locally advanced triple-negative breast cancer (TNBC). Interestingly, the focused application of ultrasound at the orthotopic TNBC xenograft of NOD-SCID mice facilitated the immediate infiltration of nano-conjugates at the tumor interstitium, and conferred in vivo safety over marketed paclitaxel formulation. In addition, curcumin significantly potentiated the in vivo chemoradiotherapeutic efficacy of paclitaxel upon loading into nano-conjugates. This gets further enhanced by the concurrent pulse of ultrasound, as confirmed by PET-CT imaging, along with a significant improvement in the mice survival. The quadrapeutic apoptotic effect by the combination of paclitaxel, curcumin, radiation, and ultrasound, along with a reduction in the tumor microvessel density and cell proliferation marker, confers the broad chemo-radiotherapeutic potential of this regimen for radio-responsive solid tumors, as well as metastatic niches.
Collapse
Affiliation(s)
- Prateek Bhardwaj
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India.
| | - Jayant Sastri Goda
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India.
| | - Venkatesh Pai
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India.
| | - Pradip Chaudhari
- Animal house facility, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India
| | - Bhabani Mohanty
- Animal house facility, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India
| | - Trupti Pai
- Department of Pathology, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India
| | - Komal Vishwakarma
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India.
| | - Rahul Thorat
- Animal house facility, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India
| | - Tabassum Wadasadawala
- Department of Radiation Oncology, Tata Memorial Centre, Mumbai, & Homi Bhabha National Institute, Maharashtra, India.
| | - Rinti Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India.
| |
Collapse
|
8
|
3D Modeling of Epithelial Tumors-The Synergy between Materials Engineering, 3D Bioprinting, High-Content Imaging, and Nanotechnology. Int J Mol Sci 2021; 22:ijms22126225. [PMID: 34207601 PMCID: PMC8230141 DOI: 10.3390/ijms22126225] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022] Open
Abstract
The current statistics on cancer show that 90% of all human cancers originate from epithelial cells. Breast and prostate cancer are examples of common tumors of epithelial origin that would benefit from improved drug treatment strategies. About 90% of preclinically approved drugs fail in clinical trials, partially due to the use of too simplified in vitro models and a lack of mimicking the tumor microenvironment in drug efficacy testing. This review focuses on the origin and mechanism of epithelial cancers, followed by experimental models designed to recapitulate the epithelial cancer structure and microenvironment, such as 2D and 3D cell culture models and animal models. A specific focus is put on novel technologies for cell culture of spheroids, organoids, and 3D-printed tissue-like models utilizing biomaterials of natural or synthetic origins. Further emphasis is laid on high-content imaging technologies that are used in the field to visualize in vitro models and their morphology. The associated technological advancements and challenges are also discussed. Finally, the review gives an insight into the potential of exploiting nanotechnological approaches in epithelial cancer research both as tools in tumor modeling and how they can be utilized for the development of nanotherapeutics.
Collapse
|
9
|
Three-Dimensional Culture System of Cancer Cells Combined with Biomaterials for Drug Screening. Cancers (Basel) 2020; 12:cancers12102754. [PMID: 32987868 PMCID: PMC7601447 DOI: 10.3390/cancers12102754] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary For the research and development of drug discovery, it is of prime importance to construct the three-dimensional (3D) tissue models in vitro. To this end, the enhancement design of cell function and activity by making use of biomaterials is essential. In this review, 3D culture systems of cancer cells combined with several biomaterials for anticancer drug screening are introduced. Abstract Anticancer drug screening is one of the most important research and development processes to develop new drugs for cancer treatment. However, there is a problem resulting in gaps between the in vitro drug screening and preclinical or clinical study. This is mainly because the condition of cancer cell culture is quite different from that in vivo. As a trial to mimic the in vivo cancer environment, there has been some research on a three-dimensional (3D) culture system by making use of biomaterials. The 3D culture technologies enable us to give cancer cells an in vitro environment close to the in vivo condition. Cancer cells modified to replicate the in vivo cancer environment will promote the biological research or drug discovery of cancers. This review introduces the in vitro research of 3D cell culture systems with biomaterials in addition to a brief summary of the cancer environment.
Collapse
|
10
|
Force-dependent extracellular matrix remodeling by early-stage cancer cells alters diffusion and induces carcinoma-associated fibroblasts. Biomaterials 2020; 234:119756. [PMID: 31954229 DOI: 10.1016/j.biomaterials.2020.119756] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/28/2019] [Accepted: 01/02/2020] [Indexed: 12/21/2022]
Abstract
It is known cancer cells secrete cytokines inducing normal fibroblasts (NFs) to become carcinoma-associated fibroblasts (CAFs). However, it is not clear how the CAF-promoting cytokines can effectively navigate the dense ECM, a diffusion barrier, in the tumor microenvironment to reach NFs during the early stages of cancer development. In this study, we devised a 3D coculture system to investigate the possible mechanism of CAF induction at early stages of breast cancer. We found that in a force-dependent manner, ECM fibrils are radially aligned relative to the tumor spheroid. The fibril alignment enhances the diffusion of exosomes containing CAF-promoting cytokines towards NFs. Suppression of force generation or ECM remodeling abolishes the enhancement of exosome diffusion and the subsequent CAF induction. In summary, our finding suggests that early-stage, pre-metastatic cancer cells can generate high forces to align the ECM fibrils, thereby enhancing the diffusion of CAF-promoting exosomes to reach the stroma and induce CAFs.
Collapse
|
11
|
Xin X, Yang ST. A Dual Fluorescent 3-D Multicellular Coculture of Breast Cancer MCF-7 and Fibroblast NIH-3T3 Cells for High Throughput Cancer Drug Screening. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2019.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
12
|
Yoriki K, Mori T, Kokabu T, Matsushima H, Umemura S, Tarumi Y, Kitawaki J. Estrogen-related receptor alpha induces epithelial-mesenchymal transition through cancer-stromal interactions in endometrial cancer. Sci Rep 2019; 9:6697. [PMID: 31040369 PMCID: PMC6491648 DOI: 10.1038/s41598-019-43261-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/17/2019] [Indexed: 02/06/2023] Open
Abstract
Estrogen-related receptor alpha (ERRα), which shares structural similarities with estrogen receptors, is associated with tumor progression in endometrial cancer, but little is known about the detailed underlying mechanism. We investigated whether ERRα, in cooperation with peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), could participate in epithelial-mesenchymal transition (EMT) in endometrial cancer through cancer-stromal interactions. Two endometrial cancer cell lines, Ishikawa and HEC-1A, transfected with ERRα/PGC-1α expression plasmids or silenced for ERRα expression, were co-cultured with telomerase-transformed human endometrial stromal cells (T-HESCs). We found that EMT-associated factors including vimentin, Snail, and zinc finger E-box binding homeobox 1 were upregulated in cancer cells overexpressing ERRα/PGC-1α and that transforming growth factor-beta (TGF-β) was induced in T-HESCs in the same conditions. In contrast, ERRα knockdown suppressed EMT-associated factors in cancer cells and TGF-β in T-HESCs. ERRα/PGC-1α overexpression increased the expression of EMT-associated factors after TGF-β exposure; however, it decreased E-cadherin at protein level. ERRα knockdown suppressed EMT-associated factors in the presence of TGF-β, whereas E-cadherin remained unchanged. Matrigel invasion assays revealed that ERRα knockdown attenuated the stimulation of migration and invasion by TGF-β. These findings suggest that ERRα is a potential target for inhibiting TGF-β-induced EMT through cancer-stromal interactions in endometrial cancer.
Collapse
Affiliation(s)
- Kaori Yoriki
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Taisuke Mori
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Tetsuya Kokabu
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hiroshi Matsushima
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Shiori Umemura
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yosuke Tarumi
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Jo Kitawaki
- Department of Obstetrics and Gynecology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
13
|
Williams GP, Darbre PD. Low-dose environmental endocrine disruptors, increase aromatase activity, estradiol biosynthesis and cell proliferation in human breast cells. Mol Cell Endocrinol 2019; 486:55-64. [PMID: 30817981 DOI: 10.1016/j.mce.2019.02.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/07/2019] [Accepted: 02/18/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Phenolic endocrine-disrupting compounds (EDCs) have long been suspected of increasing human breast cancer risk, via aromatase up-regulation; however, the metabolic effects upon aromatase in human breast cells exposed to environmentally relevant concentrations of phenolic compounds, have not been addressed. OBJECTIVES To examine the mechanistic responses of aromatase CYP19A1 mRNA, aromatase activity, estradiol biosynthesis and cellular proliferation, in three human breast cell lines, exposed to seven phenolic compounds, at environmentally relevant concentrations. METHODS MCF-7 and ZR-75-1 breast cancer cells, and HMF3A breast fibroblasts were treated with specific concentrations of p,p'-DDT, methoxychlor, benzophenone-2, bisphenol A, bisphenol S, 4-phenylphenol and n-butylparaben, with and without the presence of aromatase inhibitors and estrogen receptor inhibitors. RESULTS All test EDCs up-regulated aromatase mRNA, increased aromatase activity, significantly increased the aromatase-induced biosynthesis of the breast carcinogen 17β-estradiol, and increased ERα-positive breast cell proliferation. CONCLUSION Inadvertent exposures to 'phenolic' EDCs, increase estradiol biosynthesis, and estrogen-sensitive breast cancer proliferation.
Collapse
Affiliation(s)
- Graeme P Williams
- Molecular and Cellular Medicine Group, School of Biological Sciences, University of Reading, Reading, Berkshire, RG6 6UB, United Kingdom.
| | - Philippa D Darbre
- Molecular and Cellular Medicine Group, School of Biological Sciences, University of Reading, Reading, Berkshire, RG6 6UB, United Kingdom
| |
Collapse
|
14
|
|
15
|
Zhang X, Ding K, Wang J, Li X, Zhao P. Chemoresistance caused by the microenvironment of glioblastoma and the corresponding solutions. Biomed Pharmacother 2018; 109:39-46. [PMID: 30391707 DOI: 10.1016/j.biopha.2018.10.063] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/03/2018] [Accepted: 10/12/2018] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary human brain tumor. Although comprehensive therapies combining radiotherapy and chemotherapy after surgery can prolong survival, the prognosis is still poor with a median survival of only 14.6 months. Chemoresistance is one of the major causes of relapse as well as poor survival in glioma patients. Therefore, novel strategies to overcome chemoresistance are desperately needed for improved treatment of human GBM. Recent studies have demonstrated that the tumor microenvironment plays a critical role in the chemoresistance of various tumor types, which makes it a suitable target in anti-cancer therapies, as well as a valuable biomarker for prognostic purposes. This review focuses on chemoresistance in GBM induced by stromal cells, including the endothelium of blood vessels, astrocytes, and myeloid cells, as well as non-cellular factors in the tumor microenvironment. Corresponding therapies are discussed, including progressive strategies involving 3-dimensional models integrating engineering as well as biological advances.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China
| | - Kaikai Ding
- Shandong Key Laboratory of Brain Function Remodeling, PR China; Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, 250012, PR China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China; Department of Biomedicine, University of Bergen, 5009, Bergen, Norway
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China
| | - Peng Zhao
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China.
| |
Collapse
|
16
|
Balachander GM, Talukdar PM, Debnath M, Rangarajan A, Chatterjee K. Inflammatory Role of Cancer-Associated Fibroblasts in Invasive Breast Tumors Revealed Using a Fibrous Polymer Scaffold. ACS APPLIED MATERIALS & INTERFACES 2018; 10:33814-33826. [PMID: 30207687 DOI: 10.1021/acsami.8b07609] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Inflammation in cancer fuels metastasis and worsens prognosis. Cancer-associated fibroblasts (CAFs) present in the tumor stroma play a vital role in mediating the cascade of cancer inflammation that drives metastasis by enhancing angiogenesis, tissue remodeling, and invasion. In vitro models that faithfully recapitulate CAF-mediated inflammation independent of coculturing with cancer cells are nonexistent. We have engineered fibrous matrices of poly(ε-caprolactone) (PCL) that can maintain the manifold tumor-promoting properties of patient-derived CAFs, which would otherwise require repetitive isolation and complex coculturing with cancer cells. On these fibrous matrices, CAFs proliferated and remodeled the extracellular matrix (ECM) in a parallel-patterned manner mimicking the ECM of high-grade breast tumors and induced stemness in breast cancer cells. The response of the fibroblasts was observed to be sensitive to the scaffold architecture and not the polymer composition. The CAFs cultured on fibrous matrices exhibited increased activation of the NF-κB pathway and downstream proinflammatory gene expression compared to CAFs cultured on conventional two-dimensional (2D) dishes and secreted higher levels of proinflammatory cytokines such as IL-6, GM-CSF, and MIP-3α. Consistent with this, we observed increased infiltration of inflammatory cells to the tumor site and enhanced invasiveness of the tumor in vivo when tumor cells were injected admixed with CAFs grown on fibrous matrices. These data suggest that CAFs better retain their tumor-promoting proinflammatory properties on fibrous polymeric matrices, which could serve as a unique model to investigate the mechanisms of stroma-induced inflammation in cancer progression.
Collapse
Affiliation(s)
| | - Pinku Mani Talukdar
- Department of Human Genetics , National Institute of Mental Health and Neurosciences , Bangalore 560029 , India
| | - Monojit Debnath
- Department of Human Genetics , National Institute of Mental Health and Neurosciences , Bangalore 560029 , India
| | | | | |
Collapse
|
17
|
Lang L, Shay C, Xiong Y, Thakkar P, Chemmalakuzhy R, Wang X, Teng Y. Combating head and neck cancer metastases by targeting Src using multifunctional nanoparticle-based saracatinib. J Hematol Oncol 2018; 11:85. [PMID: 29925404 PMCID: PMC6011403 DOI: 10.1186/s13045-018-0623-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/29/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Inhibition of metastasis of head and neck squamous cell carcinoma (HNSCC) is one of the most important challenges in cancer treatment. Src, a non-receptor tyrosine kinase, has been implicated as a key promoter in tumor progression and metastasis of HNSCC. However, Src therapy for HNSCC is limited by lack of efficient in vivo delivery and underlying mechanisms remain elusive. METHODS Src knockdown cells were achieved by lentiviral-mediated interference. Cell migration and invasion were examined by wound healing and Transwell assays. Protein levels were determined by Western blot and/or immunohistochemistry. The Src inhibitor saracatinib was loaded into self-assembling nanoparticles by the solvent evaporation method. An experimental metastasis mouse model was generated to investigate the drug efficacy in metastasis. RESULTS Blockade of Src kinase activity by saracatinib effectively suppressed invasion and metastasis of HNSCC. Mechanistic assessment of the drug effects in HNSCC cells showed that saracatinib induced suppression of Src-dependent invasion/metastasis through downregulating the expression levels of Vimentin and Snail proteins. In tests in mice, saracatinib loaded into the novel multifunctional nanoparticles exhibited superior effects on suppression of HNSCC metastasis compared with the free drug, which is mainly attributed to highly specific and efficient tumor-targeted drug delivery system. CONCLUSIONS These findings and advances are of great importance to the development of Src-targeted nanomedicine as a more effective therapy for metastatic HNSCC.
Collapse
Affiliation(s)
- Liwei Lang
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA USA
| | - Chloe Shay
- Department of Pediatrics, Emory Children’s Center, Emory University, Atlanta, GA USA
| | - Yuanping Xiong
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA USA
| | - Parth Thakkar
- Department of Biology, College of Science and Mathematics, Augusta University, Augusta, GA USA
| | - Ron Chemmalakuzhy
- Department of Biology, College of Science and Mathematics, Augusta University, Augusta, GA USA
| | - Xuli Wang
- Department of Radiology and Imaging Sciences, School of Medicine, University of Utah, 201 Presidents Cir, Salt Lake City, UT 84112 USA
| | - Yong Teng
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA USA
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA USA
- Department of Medical Laboratory, Imaging and Radiologic Sciences, College of Allied Health, Augusta University, 1120 15th Street, Augusta, GA 30912 USA
| |
Collapse
|
18
|
Recapitulating spatiotemporal tumor heterogeneity in vitro through engineered breast cancer microtissues. Acta Biomater 2018; 73:236-249. [PMID: 29679778 DOI: 10.1016/j.actbio.2018.04.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 01/14/2023]
Abstract
Tumor and microenvironmental heterogeneity hinders the study of breast cancer biology and the assessment of therapeutic strategies, being associated with high variability and drug resistance. In this context, it is mandatory to develop three-dimensional breast tumor models able to reproduce this heterogeneity and the dynamic interaction occurring between tumor cells and microenvironment. Here we show a new breast cancer microtissue model (T-µTP) uniquely able to present intra-tumor morphological heterogeneity in a dynamic and responsive endogenous matrix. T-µTP consists of adenocarcinoma cells, endothelial cells and stromal fibroblasts. These three kinds of cells are totally embedded into an endogenous matrix which is rich in collagen and hyaluronic acid and it is directly produced by human fibroblasts. In this highly physiologically relevant environment, tumor cells evolve in different cluster morphologies recapitulating tumor spatiotemporal heterogeneity. Moreover they activate the desmoplastic and vascular reaction with affected collagen content, assembly and organization and the presence of aberrant capillary-like structures (CLS). Thus, T-µTP allows to outline main crucial events involved in breast cancer progression into a single model overcoming the limit of artificial extra cellular matrix surrogates. We strongly believe that T-µTP is a suitable model for the study of breast cancer and for drug screening assays following key parameters of clinical interest. STATEMENT OF SIGNIFICANCE Tumor and microenvironmental heterogeneity makes very hurdle to find a way to study and treat breast cancer. Here we develop an innovative 3D tumor microtissue model recapitulating in vitro tumor heterogeneity. Tumor microtissues are characterized by the activation of the stromal and vascular reaction too. We underline the importance to mimic different microenvironmental tumor features in the same time and in a single tissue in order to obtain a model of spatiotemporal tumor genesis and progression, suitable for the study of tumor treatment and resistance.
Collapse
|
19
|
Increased phosphorylation of eIF2α in chronic myeloid leukemia cells stimulates secretion of matrix modifying enzymes. Oncotarget 2018; 7:79706-79721. [PMID: 27802179 PMCID: PMC5346746 DOI: 10.18632/oncotarget.12941] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 10/17/2016] [Indexed: 12/14/2022] Open
Abstract
Recent studies underscore the role of the microenvironment in therapy resistance of chronic myeloid leukemia (CML) cells and leukemia progression. We previously showed that sustained mild activation of endoplasmic reticulum (ER) stress in CML cells supports their survival and resistance to chemotherapy. We now demonstrate, using dominant negative non-phosphorylable mutant of eukaryotic initiation factor 2 α subunit (eIF2α), that phosphorylation of eIF2α (eIF2α-P), which is a hallmark of ER stress in CML cells, substantially enhances their invasive potential and modifies their ability to secrete extracellular components, including the matrix-modifying enzymes cathepsins and matrix metalloproteinases. These changes are dependent on the induction of activating transcription factor-4 (ATF4) and facilitate extracellular matrix degradation by CML cells. Conditioned media from CML cells with constitutive activation of the eIF2α-P/ATF4 pathway induces invasiveness of bone marrow stromal fibroblasts, suggesting that eIF2α-P may be important for extracellular matrix remodeling and thus leukemia cells-stroma interactions. Our data show that activation of stress response in CML cells may contribute to the disruption of bone marrow niche components by cancer cells and in this way support CML progression.
Collapse
|
20
|
Thomsen AR, Aldrian C, Bronsert P, Thomann Y, Nanko N, Melin N, Rücker G, Follo M, Grosu AL, Niedermann G, Layer PG, Heselich A, Lund PG. A deep conical agarose microwell array for adhesion independent three-dimensional cell culture and dynamic volume measurement. LAB ON A CHIP 2017; 18:179-189. [PMID: 29211089 DOI: 10.1039/c7lc00832e] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Multicellular spheroids represent a well-established 3D model to study healthy and diseased cells in vitro. The use of conventional 3D cell culture platforms for the generation of multicellular spheroids is limited to cell types that easily self-assemble into spheroids because less adhesive cells fail to form stable aggregates. A high-precision micromoulding technique developed in our laboratory produces deep conical agarose microwell arrays that allow the cultivation of uniform multicellular aggregates, irrespective of the spheroid formation capacity of the cells. Such hydrogel arrays warrant a steady nutrient supply for several weeks, permit live volumetric measurements to monitor cell growth, enable immunohistochemical staining, fluorescence-based microscopy, and facilitate immediate harvesting of cell aggregates. This system also allows co-cultures of two distinct cell types either in direct cell-cell contact or at a distance as the hydrogel permits diffusion of soluble compounds. Notably, we show that co-culture of a breast cancer cell line with bone marrow stromal cells enhances 3D growth of the cancer cells in this system.
Collapse
Affiliation(s)
- Andreas R Thomsen
- Department of Radiation Oncology, Medical Center - University of Freiburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Zhao L, Guo T, Wang L, Liu Y, Chen G, Zhou H, Zhang M. Tape-Assisted Photolithographic-Free Microfluidic Chip Cell Patterning for Tumor Metastasis Study. Anal Chem 2017; 90:777-784. [DOI: 10.1021/acs.analchem.7b03225] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Liang Zhao
- Research Center for Bioengineering
and Sensing Technology, School of Chemistry and Biological Engineering,
Institute of Precision Medicine and Health, Beijing Key Laboratory
for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China
| | - Tengfei Guo
- Research Center for Bioengineering
and Sensing Technology, School of Chemistry and Biological Engineering,
Institute of Precision Medicine and Health, Beijing Key Laboratory
for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China
| | - Lirong Wang
- Research Center for Bioengineering
and Sensing Technology, School of Chemistry and Biological Engineering,
Institute of Precision Medicine and Health, Beijing Key Laboratory
for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China
| | - Yang Liu
- Research Center for Bioengineering
and Sensing Technology, School of Chemistry and Biological Engineering,
Institute of Precision Medicine and Health, Beijing Key Laboratory
for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China
| | - Ganyu Chen
- Research Center for Bioengineering
and Sensing Technology, School of Chemistry and Biological Engineering,
Institute of Precision Medicine and Health, Beijing Key Laboratory
for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China
| | - Hao Zhou
- Research Center for Bioengineering
and Sensing Technology, School of Chemistry and Biological Engineering,
Institute of Precision Medicine and Health, Beijing Key Laboratory
for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China
| | - Meiqin Zhang
- Research Center for Bioengineering
and Sensing Technology, School of Chemistry and Biological Engineering,
Institute of Precision Medicine and Health, Beijing Key Laboratory
for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China
| |
Collapse
|
22
|
Fontanil T, Álvarez-Teijeiro S, Villaronga MÁ, Mohamedi Y, Solares L, Moncada-Pazos A, Vega JA, García-Suárez O, Pérez-Basterrechea M, García-Pedrero JM, Obaya AJ, Cal S. Cleavage of Fibulin-2 by the aggrecanases ADAMTS-4 and ADAMTS-5 contributes to the tumorigenic potential of breast cancer cells. Oncotarget 2017; 8:13716-13729. [PMID: 28099917 PMCID: PMC5355132 DOI: 10.18632/oncotarget.14627] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 01/04/2017] [Indexed: 11/30/2022] Open
Abstract
Fibulin-2 participates in the assembly of extracellular matrix components through interactions with multiple ligands and promotes contacts between cells and their surrounding environment. Consequently, identification of processes that could lead to an altered Fibulin-2 could have a major impact not only in the maintenance of tissue architecture and morphogenesis but also in pathological situations including cancer. Herein, we have investigated the ability of the secreted metalloproteases ADAMTS-4 and ADAMTS-5 to digest Fibulin-2. Using in vitro approaches and cultured breast cancer cell lines we demonstrate that Fibulin-2 is a better substrate for ADAMTS-5 than it is for ADAMTS-4. Moreover, Fibulin-2 degradation is associated to an enhancement of the invasive potential of T47D, MCF-7 and SK-BR-3 cells. We have also found that conditioned medium from MCF-7 cells that simultaneously overexpress Fibulin-2 and ADAMTS-5 significantly induced the migratory and invasive ability of normal breast fibroblasts using 3D collagen matrices. Immunohistochemical analysis highlights the close proximity or partial overlap of both Fibulin-2 and ADAMTS-5 in breast tumor samples. Additionally, proteolytic products derived from a potential degradation of Fibulin-2 by ADAMTS-5 were also identified in these samples. Finally, we also show that the cleavage of Fibulin-2 by ADAMTS-5 is counteracted by ADAMTS-12, a metalloprotease that interacts with Fibulin-2. Overall, our results provide direct evidence indicating that Fibulin-2 is a novel substrate of ADAMTS-5 and that this proteolysis could alter the cellular microenvironment affecting the balance between protumor and antitumor effects associated to both Fibulin-2 and the ADAMTSs metalloproteases.
Collapse
Affiliation(s)
- Tania Fontanil
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Asturias Spain.,Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain
| | - Saúl Álvarez-Teijeiro
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain.,Hospital Universitario Central de Asturias, Universidad de Oviedo, Asturias, and CIBERONC, Madrid, Spain
| | - M Ángeles Villaronga
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain.,Hospital Universitario Central de Asturias, Universidad de Oviedo, Asturias, and CIBERONC, Madrid, Spain
| | - Yamina Mohamedi
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Asturias Spain.,Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain
| | - Laura Solares
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Asturias Spain.,Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain
| | - Angela Moncada-Pazos
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Asturias Spain.,Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain.,Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - José A Vega
- Department of Morphology and Cellular Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain.,Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Chile
| | - Olivia García-Suárez
- Department of Morphology and Cellular Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain
| | - Marcos Pérez-Basterrechea
- Unidad de Trasplantes, Terapia Celular y Medicina Regenerativa, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Juana M García-Pedrero
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain.,Hospital Universitario Central de Asturias, Universidad de Oviedo, Asturias, and CIBERONC, Madrid, Spain
| | - Alvaro J Obaya
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain.,Departamento de Biología Funcional, Area de Fisiología, Universidad de Oviedo, Asturias, Spain
| | - Santiago Cal
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Asturias Spain.,Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, Asturias, Spain
| |
Collapse
|
23
|
Rijal G, Li W. A versatile 3D tissue matrix scaffold system for tumor modeling and drug screening. SCIENCE ADVANCES 2017; 3:e1700764. [PMID: 28924608 PMCID: PMC5597314 DOI: 10.1126/sciadv.1700764] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/14/2017] [Indexed: 05/19/2023]
Abstract
Most of the anticancer drug candidates entering preclinical trials fail to be approved for clinical applications. The following are among the main causes of these failures: studying molecular mechanisms of cancer development, identifying therapeutic targets, and testing drug candidates using inappropriate tissue culture models, which do not recapitulate the native microenvironment where the cancer cells originate. It has become clear that three-dimensional (3D) cell cultures are more biologically and clinically relevant than 2D models. The spatial and mechanical conditions of 3D cultures enable the cancer cells to display heterogeneous growth, assume diverse phenotypes, express distinct gene and protein products, and attain metastatic potential and resistance to drugs that are reminiscent of tumors in humans. However, the current 3D culture systems using synthetic polymers or selected components of the extracellular matrix (ECM) are defective (particularly the biophysical and biochemical properties of the native ECM) and remain distant to optimally support the signaling cue-oriented cell survival and growth. We introduce a reconstitutable tissue matrix scaffold (TMS) system fabricated using native tissue ECM, with tissue-like architecture and resilience. The structural and compositional properties of TMS favor robust cell survival, proliferation, migration, and invasion in culture and vascularized tumor formation in animals. The combination of porous and hydrogel TMS allows compartmental culture of cancerous and stromal cells, which are distinguishable by biomarkers. The response of the cancer cells grown on TMS to drugs well reflects animal and clinical observations. TMS enables more biologically relevant studies and is suitable for preclinical drug screening.
Collapse
|
24
|
Roberts S, Speirs V. Advances in the development of improved animal-free models for use in breast cancer biomedical research. Biophys Rev 2017; 9:321-327. [PMID: 28748520 PMCID: PMC5578919 DOI: 10.1007/s12551-017-0276-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/03/2017] [Indexed: 02/08/2023] Open
Abstract
Through translational research, the outcomes for women (and men) diagnosed with breast cancer have improved significantly, with now over 80% of women surviving for at least 5 years post-diagnosis. Much of this success has been translated from the bench to the bedside using laboratory models. Here, we outline the types of laboratory models that have helped achieve this and discuss new approaches as we move towards animal-free disease modelling.
Collapse
Affiliation(s)
- Sophie Roberts
- Leeds Institute of Cancer & Pathology, University of Leeds, St James's University Hospital, Wellcome Trust Brenner Building, Leeds, LS9 7TF, UK
| | - Valerie Speirs
- Leeds Institute of Cancer & Pathology, University of Leeds, St James's University Hospital, Wellcome Trust Brenner Building, Leeds, LS9 7TF, UK.
| |
Collapse
|
25
|
Cai D, Wu X, Hong T, Mao Y, Ge X, Hua D. CD61+ and CAF+ were found to be good prognosis factors for invasive breast cancer patients. Pathol Res Pract 2017; 213:1296-1301. [PMID: 28935175 DOI: 10.1016/j.prp.2017.06.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 06/01/2017] [Accepted: 06/24/2017] [Indexed: 11/18/2022]
Abstract
BACKGROUND During the epithelial tumor development process, changes in tumor cell genes are an important driving factor for tumorigenesis. Recently, however, studies have shown that the tumor microenvironment, especially for cancer-associated fibroblasts (CAFs) and the infiltration of platelets into tumors, plays an essential role in the progression of human malignant disease. METHODS In this study, we investigated the presence and prognostic role of podoplanin-expressing CAFs (CAF+), the infiltration of platelets into tumors (CD61+) and platelet count before surgery in a large sample of patients with breast cancer. Podoplanin expression and platelet infiltration were evaluated by immunohistochemistry in 164 patients with breast cancer. RESULTS Seventy-two patients (44%) showed CAF+, while fifty-seven patients (35%) showed CD61+. Several strong positive correlations were found, including CD61+ with blood platelet count before surgery (P=0.004), and CAF+ with CD61+ (P=0.048). Patients with CAF+, CD61+ or platelet count >280×109/L before surgery had a significantly shorter disease-free survival according to univariate analysis. Multivariable analysis showed that CAF+ was an independent prognostic factor (Hazard ratio=3.928; p=0.005). CONCLUSIONS CAF+ and CD61+ were found to be good negative prognosis factors for invasive breast cancer patients. CD61+ also had strong positive correlation with blood platelet count before surgery. These targets may be used as strategies for the treatment of breast cancer in the future.
Collapse
Affiliation(s)
- Dongyan Cai
- Department of Oncology, The Fourth People's Hospital of Wuxi, No. 200, Huihe road, Wuxi 214062, China
| | - Xiaohong Wu
- Department of Oncology, The Fourth People's Hospital of Wuxi, No. 200, Huihe road, Wuxi 214062, China
| | - Tingting Hong
- Department of Oncology, The Fourth People's Hospital of Wuxi, No. 200, Huihe road, Wuxi 214062, China
| | - Yong Mao
- Department of Oncology, The Fourth People's Hospital of Wuxi, No. 200, Huihe road, Wuxi 214062, China
| | - Xiaosong Ge
- Department of Oncology, The Fourth People's Hospital of Wuxi, No. 200, Huihe road, Wuxi 214062, China
| | - Dong Hua
- Department of Oncology, The Fourth People's Hospital of Wuxi, No. 200, Huihe road, Wuxi 214062, China.
| |
Collapse
|
26
|
Grinde MT, Vik J, Camilio KA, Martinez-Zubiaurre I, Hellevik T. Ionizing radiation abrogates the pro-tumorigenic capacity of cancer-associated fibroblasts co-implanted in xenografts. Sci Rep 2017; 7:46714. [PMID: 28440285 PMCID: PMC5404232 DOI: 10.1038/srep46714] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/28/2017] [Indexed: 01/06/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are abundantly present in solid tumors and affect tumorigenesis and therapeutic responses. In the context of clinical radiotherapy, the impact of irradiated CAFs to treatment outcomes is largely unexplored. Aiming at improving radiotherapy efficacy, we have here explored the effect of radiation on the inherent pro-tumorigenic capacity of CAFs in animals. Ionizing radiation was delivered to cultured CAFs as single-high or fractionated doses. Tumor development was compared in mice receiving A549 lung tumor cells admixed with irradiated or control CAFs. Biological mechanisms behind tumor growth regulation were investigated by quantitative histology and immunohistochemistry. Viability assessments confirmed that irradiated CAFs are fully functional prior to implantation. However, the enhanced tumorigenic effect observed in tumors co-implanted with control CAFs was abrogated in tumors established with irradiated CAFs. Experiments to ascertain fate of implanted fibroblasts showed that exogenously administered CAFs reside at the implantation site for few days, suggesting that tumor growth regulation from admixed CAFs take place during initial tumor formation. Our work demonstrate that irradiated CAFs lose their pro-tumorigenic potential in vivo, affecting angiogenesis and tumor engraftment. This finding propose a previously unknown advantageous effect induced by radiotherapy, adding to the direct cytotoxic effects on transformed epithelial cells.
Collapse
Affiliation(s)
- Maria Tunset Grinde
- Department of Radiology, University Hospital of Northern-Norway, Tromsø, Norway.,Molecular &Clinical Inflammation Research Group, Department of Clinical Medicine, University of Tromsø, Norway
| | - Jørg Vik
- Molecular &Clinical Inflammation Research Group, Department of Clinical Medicine, University of Tromsø, Norway
| | | | - Inigo Martinez-Zubiaurre
- Molecular &Clinical Inflammation Research Group, Department of Clinical Medicine, University of Tromsø, Norway
| | - Turid Hellevik
- Molecular &Clinical Inflammation Research Group, Department of Clinical Medicine, University of Tromsø, Norway.,Department of Radiation Oncology, University Hospital of Northern-Norway, Tromsø, Norway
| |
Collapse
|
27
|
Amornsupak K, Jamjuntra P, Warnnissorn M, O-Charoenrat P, Sa-Nguanraksa D, Thuwajit P, Eccles SA, Thuwajit C. High ASMA + Fibroblasts and Low Cytoplasmic HMGB1 + Breast Cancer Cells Predict Poor Prognosis. Clin Breast Cancer 2017; 17:441-452.e2. [PMID: 28533055 DOI: 10.1016/j.clbc.2017.04.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 04/04/2017] [Accepted: 04/06/2017] [Indexed: 10/19/2022]
Abstract
INTRODUCTION The influence of cancer-associated fibroblasts (CAFs) and high mobility group box 1 (HMGB1) has been recognized in several cancers, although their roles in breast cancer are unclear. The present study aimed to determine the levels and prognostic significance of α-smooth muscle actin-positive (ASMA+) CAFs, plus HMGB1 and receptor for advanced glycation end products (RAGE) in cancer cells. MATERIALS AND METHODS A total of 127 breast samples, including 96 malignant and 31 benign, were examined for ASMA, HMGB1, and RAGE by immunohistochemistry. The χ2 test and Fisher's exact test were used to test the association of each protein with clinicopathologic parameters. The Kaplan-Meier method or log-rank test and Cox regression were used for survival analysis. RESULTS ASMA+ fibroblast infiltration was significantly increased in the tumor stroma compared with that in benign breast tissue. The levels of cytoplasmic HMGB1 and RAGE were significantly greater in the breast cancer tissue than in the benign breast tissues. High ASMA expression correlated significantly with large tumor size, clinical stage III-IV, and angiolymphatic and perinodal invasion. In contrast, increased cytoplasmic HMGB1 correlated significantly with small tumor size, pT stage, early clinical stage, luminal subtype (but not triple-negative subtype), and estrogen receptor and progesterone receptor expression. The levels of ASMA (hazard ratio, 14.162; P = .010) and tumor cytoplasmic HMGB1 (hazard ratio, 0.221; P = .005) could serve as independent prognostic markers for metastatic relapse in breast cancer patients. The ASMA-high/HMGB1-low profile provided the most reliable prediction of metastatic relapse. CONCLUSION We present for the first time, to the best of our knowledge, the potential clinical implications of the combined assessment of ASMA+ fibroblasts and cytoplasmic HMGB1 in breast cancer.
Collapse
Affiliation(s)
- Kamolporn Amornsupak
- Department of Immunology, Graduate Program in Immunology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pranisa Jamjuntra
- Department of Immunology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Malee Warnnissorn
- Department of Pathology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pornchai O-Charoenrat
- Division of Head, Neck and Breast Surgery, Department of Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Doonyapat Sa-Nguanraksa
- Division of Head, Neck and Breast Surgery, Department of Surgery, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Peti Thuwajit
- Department of Immunology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Suzanne A Eccles
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, Sutton, United Kingdom
| | - Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
28
|
Aponte PM, Caicedo A. Stemness in Cancer: Stem Cells, Cancer Stem Cells, and Their Microenvironment. Stem Cells Int 2017; 2017:5619472. [PMID: 28473858 PMCID: PMC5394399 DOI: 10.1155/2017/5619472] [Citation(s) in RCA: 250] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 01/31/2017] [Accepted: 02/19/2017] [Indexed: 02/06/2023] Open
Abstract
Stemness combines the ability of a cell to perpetuate its lineage, to give rise to differentiated cells, and to interact with its environment to maintain a balance between quiescence, proliferation, and regeneration. While adult Stem Cells display these properties when participating in tissue homeostasis, Cancer Stem Cells (CSCs) behave as their malignant equivalents. CSCs display stemness in various circumstances, including the sustaining of cancer progression, and the interaction with their environment in search for key survival factors. As a result, CSCs can recurrently persist after therapy. In order to understand how the concept of stemness applies to cancer, this review will explore properties shared between normal and malignant Stem Cells. First, we provide an overview of properties of normal adult Stem Cells. We thereafter elaborate on how these features operate in CSCs. We then review the organization of microenvironment components, which enables CSCs hosting. We subsequently discuss Mesenchymal Stem/Stromal Cells (MSCs), which, although their stemness properties are limited, represent essential components of the Stem Cell niche and tumor microenvironment. We next provide insights of the therapeutic strategies targeting Stem Cell properties in tumors and the use of state-of-the-art techniques in future research. Increasing our knowledge of the CSCs microenvironment is key to identifying new therapeutic solutions.
Collapse
Affiliation(s)
- Pedro M. Aponte
- Colegio de Ciencias Biológicas y Ambientales, Universidad San Francisco de Quito (USFQ), 170901 Quito, Ecuador
- Colegio de Ciencias de la Salud, Escuela de Medicina Veterinaria, Universidad San Francisco de Quito (USFQ), 170901 Quito, Ecuador
- Mito-Act Research Consortium, Quito, Ecuador
| | - Andrés Caicedo
- Mito-Act Research Consortium, Quito, Ecuador
- Colegio de Ciencias de la Salud, Escuela de Medicina, Universidad San Francisco de Quito (USFQ), 170901 Quito, Ecuador
- Colegio de Ciencias Biológicas y Ambientales, Instituto de Microbiología, Universidad San Francisco de Quito (USFQ), 170901 Quito, Ecuador
| |
Collapse
|
29
|
Pasquereau S, Al Moussawi F, Karam W, Diab Assaf M, Kumar A, Herbein G. Cytomegalovirus, Macrophages and Breast Cancer. Open Virol J 2017; 11:15-27. [PMID: 28567162 PMCID: PMC5420183 DOI: 10.2174/1874357901711010015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/05/2017] [Accepted: 01/17/2017] [Indexed: 12/14/2022] Open
Abstract
The human cytomegalovirus (HCMV) is a betaherpesvirus that is highly host specific, infects among others epithelial cells and macrophages, and has been recently mentioned as having oncomodulatory properties. HCMV is detected in the breast tumor tissue where macrophages, especially tumor associated macrophages, are associated with a poor prognosis. In this review, we will discuss the potential implication of HCMV in breast cancer with emphasis on the role played by macrophages.
Collapse
Affiliation(s)
- S Pasquereau
- Pathogens & Inflammation/EPILAB Laboratory, Department of Virology, University of Franche-Comté, COMUE Bourgogne Franche-Comté University, UPRES EA4266, SFR FED 4234, CHRU Besançon, Besançon, France
| | - F Al Moussawi
- Pathogens & Inflammation/EPILAB Laboratory, Department of Virology, University of Franche-Comté, COMUE Bourgogne Franche-Comté University, UPRES EA4266, SFR FED 4234, CHRU Besançon, Besançon, France
| | - W Karam
- Université Libanaise, Beyrouth, Lebanon
| | | | - A Kumar
- Pathogens & Inflammation/EPILAB Laboratory, Department of Virology, University of Franche-Comté, COMUE Bourgogne Franche-Comté University, UPRES EA4266, SFR FED 4234, CHRU Besançon, Besançon, France
| | - G Herbein
- Pathogens & Inflammation/EPILAB Laboratory, Department of Virology, University of Franche-Comté, COMUE Bourgogne Franche-Comté University, UPRES EA4266, SFR FED 4234, CHRU Besançon, Besançon, France
| |
Collapse
|
30
|
Verdonck M, Denayer A, Delvaux B, Garaud S, De Wind R, Desmedt C, Sotiriou C, Willard-Gallo K, Goormaghtigh E. Characterization of human breast cancer tissues by infrared imaging. Analyst 2017; 141:606-19. [PMID: 26535413 DOI: 10.1039/c5an01512j] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Fourier Transform InfraRed (FTIR) spectroscopy coupled to microscopy (IR imaging) has shown unique advantages in detecting morphological and molecular pathologic alterations in biological tissues. The aim of this study was to evaluate the potential of IR imaging as a diagnostic tool to identify characteristics of breast epithelial cells and the stroma. In this study a total of 19 breast tissue samples were obtained from 13 patients. For 6 of the patients, we also obtained Non-Adjacent Non-Tumor tissue samples. Infrared images were recorded on the main cell/tissue types identified in all breast tissue samples. Unsupervised Principal Component Analyses and supervised Partial Least Square Discriminant Analyses (PLS-DA) were used to discriminate spectra. Leave-one-out cross-validation was used to evaluate the performance of PLS-DA models. Our results show that IR imaging coupled with PLS-DA can efficiently identify the main cell types present in FFPE breast tissue sections, i.e. epithelial cells, lymphocytes, connective tissue, vascular tissue and erythrocytes. A second PLS-DA model could distinguish normal and tumor breast epithelial cells in the breast tissue sections. A patient-specific model reached particularly high sensitivity, specificity and MCC rates. Finally, we showed that the stroma located close or at distance from the tumor exhibits distinct spectral characteristics. In conclusion FTIR imaging combined with computational algorithms could be an accurate, rapid and objective tool to identify/quantify breast epithelial cells and differentiate tumor from normal breast tissue as well as normal from tumor-associated stroma, paving the way to the establishment of a potential complementary tool to ensure safe tumor margins.
Collapse
Affiliation(s)
- M Verdonck
- Laboratory of Structure and Function of Biological Membranes, Center of Structural Biology and Bioinformatics, Université Libre de Bruxelles, Brussels, Belgium.
| | - A Denayer
- Laboratory of Structure and Function of Biological Membranes, Center of Structural Biology and Bioinformatics, Université Libre de Bruxelles, Brussels, Belgium.
| | - B Delvaux
- Laboratory of Structure and Function of Biological Membranes, Center of Structural Biology and Bioinformatics, Université Libre de Bruxelles, Brussels, Belgium.
| | - S Garaud
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - R De Wind
- Pathological Anatomy Department, Institut Jules Bordet, Brussels, Belgium
| | - C Desmedt
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Brussels, Belgium
| | - C Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Brussels, Belgium
| | - K Willard-Gallo
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - E Goormaghtigh
- Laboratory of Structure and Function of Biological Membranes, Center of Structural Biology and Bioinformatics, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
31
|
Pradhan S, Hassani I, Clary JM, Lipke EA. Polymeric Biomaterials for In Vitro Cancer Tissue Engineering and Drug Testing Applications. TISSUE ENGINEERING PART B-REVIEWS 2016; 22:470-484. [PMID: 27302080 DOI: 10.1089/ten.teb.2015.0567] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Biomimetic polymers and materials have been widely used in tissue engineering for regeneration and replication of diverse types of both normal and diseased tissues. Cancer, being a prevalent disease throughout the world, has initiated substantial interest in the creation of tissue-engineered models for anticancer drug testing. The development of these in vitro three-dimensional (3D) culture models using novel biomaterials has facilitated the investigation of tumorigenic and associated biological phenomena with a higher degree of complexity and physiological context than that provided by established two-dimensional culture models. In this review, an overview of a wide range of natural, synthetic, and hybrid biomaterials used for 3D cancer cell culture and investigation of cancer cell behavior is presented. The role of these materials in modulating cell-matrix interactions and replicating specific tumorigenic characteristics is evaluated. In addition, recent advances in biomaterial design, synthesis, and fabrication are also assessed. Finally, the advantages of incorporating polymeric biomaterials in 3D cancer models for obtaining efficacy data in anticancer drug testing applications are highlighted.
Collapse
Affiliation(s)
- Shantanu Pradhan
- Department of Chemical Engineering, Auburn University , Auburn, Alabama
| | - Iman Hassani
- Department of Chemical Engineering, Auburn University , Auburn, Alabama
| | - Jacob M Clary
- Department of Chemical Engineering, Auburn University , Auburn, Alabama
| | - Elizabeth A Lipke
- Department of Chemical Engineering, Auburn University , Auburn, Alabama
| |
Collapse
|
32
|
Roberts GC, Morris PG, Moss MA, Maltby SL, Palmer CA, Nash CE, Smart E, Holliday DL, Speirs V. An Evaluation of Matrix-Containing and Humanised Matrix-Free 3-Dimensional Cell Culture Systems for Studying Breast Cancer. PLoS One 2016; 11:e0157004. [PMID: 27300768 PMCID: PMC4907459 DOI: 10.1371/journal.pone.0157004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 05/23/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND 3D cell cultures are emerging as more physiologically meaningful alternatives to monolayer cultures for many biological applications. They are attractive because they more closely mimic in vivo morphology, especially when co-cultured with stromal fibroblasts. METHODOLOGY/PRINCIPAL FINDINGS We compared the efficacy of 3 different 3D cell culture systems; collagen I, low attachment culture vessels and a modification of Fibrolife®, a specialised humanised cell culture medium devoid of animal-derived components, using breast cancer cell lines representative of the different molecular subtypes of breast cancer, cultured alone or with human mammary fibroblasts with a view to developing matrix-free humanised systems. 3D collagen I culture supported the growth of a range of breast cancer cell lines. By modifying the composition of Fibrolife® to epiFL, matrix-free cell culture was possible. During sequential transfer to epiFL breast cancer cells gradually detached from the flask, growing progressively as spheroids. Phenotype was stable and reversible with cells remaining actively proliferating and easily accessible throughout culture. They could also be revived from frozen stocks. To achieve co-culture with fibroblasts in epiFL required use of low attachment culture vessels instead of standard plastic as fibroblasts remained adherent in epiFL. Here, cancer cell spheroids were allowed to form before adding fibroblasts. Immunohistochemical examination showed fibroblasts scattered throughout the epithelial spheroid, not dissimilar to the relationship of tumour stroma in human breast cancer. CONCLUSIONS Because of its ease of handling, matrix-free 3D cell culture may be a useful model to study the influence of fibroblasts on breast cancer epithelial cells with use of epiFL culture medium taking this a step further towards a fully humanised 3D model. This methodology could be applied to other types of cancer cell lines, making this a versatile technique for cancer researchers wishing to use in vitro systems that better reflect cancer in vivo.
Collapse
Affiliation(s)
- Grace C. Roberts
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Paul G. Morris
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Marcus A. Moss
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Sarah L. Maltby
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Chelsea A. Palmer
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Claire E. Nash
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Emily Smart
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Deborah L. Holliday
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Valerie Speirs
- Leeds Institute of Cancer and Pathology, Wellcome Trust Brenner Building, University of Leeds, Leeds, LS9 7TF, United Kingdom
| |
Collapse
|
33
|
Nash CE, Mavria G, Baxter EW, Holliday DL, Tomlinson DC, Treanor D, Novitskaya V, Berditchevski F, Hanby AM, Speirs V. Development and characterisation of a 3D multi-cellular in vitro model of normal human breast: a tool for cancer initiation studies. Oncotarget 2016; 6:13731-41. [PMID: 25915532 PMCID: PMC4537045 DOI: 10.18632/oncotarget.3803] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/18/2015] [Indexed: 11/25/2022] Open
Abstract
Multicellular 3-dimensional (3D) in vitro models of normal human breast tissue to study cancer initiation are required. We present a model incorporating three of the major functional cell types of breast, detail the phenotype and document our breast cancer initiation studies. Myoepithelial cells and fibroblasts were isolated and immortalised from breast reduction mammoplasty samples. Tri-cultures containing non-tumorigenic luminal epithelial cells HB2, or HB2 overexpressing different HER proteins, together with myoepithelial cells and fibroblasts were established in collagen I. Phenotype was assessed morphologically and immunohistochemically and compared to normal breast tissue. When all three cell types were present, polarised epithelial structures with lumens and basement membrane production were observed, akin to normal human breast tissue. Overexpression of HER2 or HER2/3 caused a significant increase in size, while HER2 overexpression resulted in development of a DCIS-like phenotype. In summary, we have developed a 3D tri-cellular model of normal human breast, amenable to comparative analysis after genetic manipulation and with potential to dissect the mechanisms behind the early stages of breast cancer initiation.
Collapse
Affiliation(s)
- Claire E Nash
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK.,Current address: The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Georgia Mavria
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Euan W Baxter
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | | | - Darren C Tomlinson
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Leeds, UK
| | - Darren Treanor
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK.,Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Vera Novitskaya
- School of Cancer Sciences, University of Birmingham, Birmingham, UK
| | | | - Andrew M Hanby
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Valerie Speirs
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| |
Collapse
|
34
|
Rijal G, Li W. 3D scaffolds in breast cancer research. Biomaterials 2016; 81:135-156. [DOI: 10.1016/j.biomaterials.2015.12.016] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/12/2015] [Accepted: 12/15/2015] [Indexed: 12/15/2022]
|
35
|
Booth ME, Nash CE, Roberts NP, Magee DR, Treanor D, Hanby AM, Speirs V. 3-D Tissue Modelling and Virtual Pathology as New Approaches to Study Ductal Carcinoma In Situ. Altern Lab Anim 2015; 43:377-83. [DOI: 10.1177/026119291504300605] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Widespread screening mammography programmes mean that ductal carcinoma in situ (DCIS), a pre-invasive breast lesion, is now more frequently diagnosed. However, not all diagnosed DCIS lesions progress to invasive breast cancer, which presents a dilemma for clinicians. As such, there is much interest in studying DCIS in the laboratory, in order to help understand more about its biology and determine the characteristics of those that progress to invasion. Greater knowledge would lead to targeted and better DCIS treatment. Here, we outline some of the models available to study DCIS, with a particular focus on animal-free systems.
Collapse
Affiliation(s)
- Mary E. Booth
- Leeds Institute of Cancer and Pathology, Leeds, UK
- Joint first authors
| | - Claire E. Nash
- Leeds Institute of Cancer and Pathology, Leeds, UK
- Joint first authors
- Current address: The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Montreal, Quebec H4A 3J1, Canada
| | | | | | - Darren Treanor
- Leeds Institute of Cancer and Pathology, Leeds, UK
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | | | | |
Collapse
|
36
|
Maris P, Blomme A, Palacios AP, Costanza B, Bellahcène A, Bianchi E, Gofflot S, Drion P, Trombino GE, Di Valentin E, Cusumano PG, Maweja S, Jerusalem G, Delvenne P, Lifrange E, Castronovo V, Turtoi A. Asporin Is a Fibroblast-Derived TGF-β1 Inhibitor and a Tumor Suppressor Associated with Good Prognosis in Breast Cancer. PLoS Med 2015; 12:e1001871. [PMID: 26327350 PMCID: PMC4556693 DOI: 10.1371/journal.pmed.1001871] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/21/2015] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Breast cancer is a leading malignancy affecting the female population worldwide. Most morbidity is caused by metastases that remain incurable to date. TGF-β1 has been identified as a key driving force behind metastatic breast cancer, with promising therapeutic implications. METHODS AND FINDINGS Employing immunohistochemistry (IHC) analysis, we report, to our knowledge for the first time, that asporin is overexpressed in the stroma of most human breast cancers and is not expressed in normal breast tissue. In vitro, asporin is secreted by breast fibroblasts upon exposure to conditioned medium from some but not all human breast cancer cells. While hormone receptor (HR) positive cells cause strong asporin expression, triple-negative breast cancer (TNBC) cells suppress it. Further, our findings show that soluble IL-1β, secreted by TNBC cells, is responsible for inhibiting asporin in normal and cancer-associated fibroblasts. Using recombinant protein, as well as a synthetic peptide fragment, we demonstrate the ability of asporin to inhibit TGF-β1-mediated SMAD2 phosphorylation, epithelial to mesenchymal transition, and stemness in breast cancer cells. In two in vivo murine models of TNBC, we observed that tumors expressing asporin exhibit significantly reduced growth (2-fold; p = 0.01) and metastatic properties (3-fold; p = 0.045). A retrospective IHC study performed on human breast carcinoma (n = 180) demonstrates that asporin expression is lowest in TNBC and HER2+ tumors, while HR+ tumors have significantly higher asporin expression (4-fold; p = 0.001). Assessment of asporin expression and patient outcome (n = 60; 10-y follow-up) shows that low protein levels in the primary breast lesion significantly delineate patients with bad outcome regardless of the tumor HR status (area under the curve = 0.87; 95% CI 0.78-0.96; p = 0.0001). Survival analysis, based on gene expression (n = 375; 25-y follow-up), confirmed that low asporin levels are associated with a reduced likelihood of survival (hazard ratio = 0.58; 95% CI 0.37-0.91; p = 0.017). Although these data highlight the potential of asporin to serve as a prognostic marker, confirmation of the clinical value would require a prospective study on a much larger patient cohort. CONCLUSIONS Our data show that asporin is a stroma-derived inhibitor of TGF-β1 and a tumor suppressor in breast cancer. High asporin expression is significantly associated with less aggressive tumors, stratifying patients according to the clinical outcome. Future pre-clinical studies should consider options for increasing asporin expression in TNBC as a promising strategy for targeted therapy.
Collapse
Affiliation(s)
- Pamela Maris
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
| | - Arnaud Blomme
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
| | - Ana Perez Palacios
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
| | - Brunella Costanza
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
| | - Akeila Bellahcène
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
| | - Elettra Bianchi
- Department of Pathology, University Hospital Liège, University of Liège, Liège, Belgium
| | | | - Pierre Drion
- Animal Facility, GIGA–Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - Giovanna Elvi Trombino
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | | | - Pino G. Cusumano
- Department of Senology, University Hospital Liège, University of Liège, Liège, Belgium
| | - Sylvie Maweja
- Department of Abdominal Surgery, University of Liège, Liège, Belgium
| | - Guy Jerusalem
- Department of Medical Oncology, University Hospital Liège, University of Liège, Liège, Belgium
| | - Philippe Delvenne
- Department of Pathology, University Hospital Liège, University of Liège, Liège, Belgium
| | - Eric Lifrange
- Department of Senology, University Hospital Liège, University of Liège, Liège, Belgium
| | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
- * E-mail: (VC); (AT)
| | - Andrei Turtoi
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
- * E-mail: (VC); (AT)
| |
Collapse
|
37
|
Thakur S, Nabbi A, Klimowicz A, Riabowol K. Stromal ING1 expression induces a secretory phenotype and correlates with breast cancer patient survival. Mol Cancer 2015; 14:164. [PMID: 26306560 PMCID: PMC4549945 DOI: 10.1186/s12943-015-0434-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/13/2015] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Previous studies have established that levels of the Inhibitor of Growth 1(ING1) tumor suppressor are reduced in a significant proportion of different cancer types. Here we analyzed levels of ING1 in breast cancer patients to determine its prognostic significance as a biomarker for breast cancer prognosis. METHODS We used automated quantitative analysis (AQUA) to determine the levels of ING1 in the tumor associated stromal cells of 462 breast cancer samples. To better understand how high ING1 levels affect nearby epithelium, we measured the levels of cytokines and secreted matrix metalloproteases (MMPs), using an ELISA based assay in mammary fibroblasts overexpressing ING1. These cells were also used in a 3-dimensional co-culture with MCF7 cells to determine the effect of released MMPs and other cytokines on growing colonies. RESULTS We find that high levels of ING1 in stroma are associated with tumor grade (p = 0.001) and size (p = 0.02), and inversely associated with patient survival (p = 0.0001) in luminal, but not in non-luminal cancers, suggesting that high stromal ING1 promotes cancer development. In this group of patients ING1 could also predict patient survival and act as a biomarker (HR = 2.125). While ING1 increased or decreased the expression of different cytokines, ING1 also increased the levels of MMP1, MMP3 and MMP10 by 5-8 fold, and concomitantly decreased levels of the tissue inhibitors of metalloproteases TIMP2, TIMP3 and TIMP4 by 1.5-3.3 fold, resulting in significant increases in MMP activity as determined by zymography. Co-culturing of MCF7 cells with stromal cells expressing ING1 in 3-dimensional organoid cultures suggested that MCF7 colonies were less well defined, suggesting that secreted MMPs might promote migration. CONCLUSION These data indicate that stromal ING1 expression can predict the survival of patients with luminal breast cancer. High levels of ING1 in stromal cells can promote the development of breast cancer through increased expression and release of MMPs and down regulation of TIMPs, which may be an underlying mechanism of reduced patient survival.
Collapse
Affiliation(s)
- Satbir Thakur
- Department of Biochemistry and Molecular Biology, University of Calgary, 3330 Hospital Drive, Calgary, T2N 4N1, AB, Canada
| | - Arash Nabbi
- Department of Medical Science, University of Calgary, 3330 Hospital Drive, Calgary, T2N 4N1, AB, Canada
| | - Alexander Klimowicz
- Functional Tissue Imaging Unit, Translational Research Laboratory, Tom Baker Cancer Center, 1331- 29 street NW, Calgary, T2N 4N2, AB, Canada.,Present Address: Boehringer Ingelheim Pharmaceuticals Inc, 900 Ridgebury Road, Ridgefield, CT, 06877, USA
| | - Karl Riabowol
- Department of Biochemistry and Molecular Biology, University of Calgary, 3330 Hospital Drive, Calgary, T2N 4N1, AB, Canada. .,Department of Oncology, University of Calgary, 311 HMRB, 3330 Hospital Drive, NW, Calgary, T2N 4N1, AB, Canada.
| |
Collapse
|
38
|
Establishment of a heterotypic 3D culture system to evaluate the interaction of TREG lymphocytes and NK cells with breast cancer. J Immunol Methods 2015. [PMID: 26215372 DOI: 10.1016/j.jim.2015.07.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Three-dimensional (3D) culture approaches to investigate breast tumour progression are yielding information more reminiscent of the in vivo microenvironment. We have established a 3D Matrigel system to determine the interactions of luminal phenotype MCF-7 cells and basal phenotype MDA-MB-231 cells with regulatory T lymphocytes and Natural Killer cells. Immune cells were isolated from peripheral blood using magnetic cell sorting and their phenotype validated using flow cytometry both before and after activation with IL-2 and phytohaemagglutinin. Following the establishment of the heterotypic culture system, tumour cells displayed morphologies and cell-cell associations distinct to that observed in 2D monolayer cultures, and associated with tissue remodelling and invasion processes. We found that the level of CCL4 secretion was influenced by breast cancer phenotype and immune stimulation. We further established that for RNA extraction, the use of proteinase K in conjunction with the Qiagen RNeasy Mini Kit and only off-column DNA digestion gave the best RNA yield, purity and integrity. We also investigated the efficacy of the culture system for immunolocalisation of the biomarkers oestrogen receptor-α and the glycoprotein mucin 1 in luminal phenotype breast cancer cells; and epidermal growth factor receptor in basal phenotype breast cancer cells, in formalin-fixed, paraffin-wax embedded cultures. The expression of these markers was shown to vary under immune mediation. We thus demonstrate the feasibility of using this co-culture system for downstream applications including cytokine analysis, immunolocalisation of tumour biomarkers on serial sections and RNA extraction in accordance with MIQE guidelines.
Collapse
|
39
|
Ham SL, Atefi E, Fyffe D, Tavana H. Robotic production of cancer cell spheroids with an aqueous two-phase system for drug testing. J Vis Exp 2015:e52754. [PMID: 25939084 PMCID: PMC4541593 DOI: 10.3791/52754] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cancer cell spheroids present a relevant in vitro model of avascular tumors for anti-cancer drug testing applications. A detailed protocol for producing both mono-culture and co-culture spheroids in a high throughput 96-well plate format is described in this work. This approach utilizes an aqueous two-phase system to confine cells into a drop of the denser aqueous phase immersed within the second aqueous phase. The drop rests on the well surface and keeps cells in close proximity to form a single spheroid. This technology has been adapted to a robotic liquid handler to produce size-controlled spheroids and expedite the process of spheroid production for compound screening applications. Spheroids treated with a clinically-used drug show reduced cell viability with increase in the drug dose. The use of a standard micro-well plate for spheroid generation makes it straightforward to analyze viability of cancer cells of drug-treated spheroids with a micro-plate reader. This technology is straightforward to implement both robotically and with other liquid handling tools such as manual pipettes.
Collapse
Affiliation(s)
| | - Ehsan Atefi
- Department of Biomedical Engineering, The University of Akron
| | - Darcy Fyffe
- Department of Biomedical Engineering, The University of Akron
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron;
| |
Collapse
|
40
|
Abstract
For many decades, fundamental cancer research has relied on two-dimensional in vitro cell culture models. However, these provide a poor representation of the complex three-dimensional (3D) architecture of living tissues. The more recent 3D culture systems, which range from ridged scaffolds to semiliquid gels, resemble their natural counterparts more closely. The arrangement of the cells in 3D systems allows better cell-cell interaction and facilitates extracellular matrix secretion, with concomitant effects on gene and protein expression and cellular behavior. Many studies have reported differences between 3D and 2D systems as regards responses to therapeutic agents and pivotal cellular processes such as cell differentiation, morphology, and signaling pathways, demonstrating the importance of 3D culturing for various cancer cell lines.
Collapse
|
41
|
Benton G, DeGray G, Kleinman HK, George J, Arnaoutova I. In vitro microtumors provide a physiologically predictive tool for breast cancer therapeutic screening. PLoS One 2015; 10:e0123312. [PMID: 25856378 PMCID: PMC4391795 DOI: 10.1371/journal.pone.0123312] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 03/03/2015] [Indexed: 12/14/2022] Open
Abstract
Many anti-cancer drugs fail in human trials despite showing efficacy in preclinical models. It is clear that the in vitro assays involving 2D monoculture do not reflect the complex extracellular matrix, chemical, and cellular microenvironment of the tumor tissue, and this may explain the failure of 2D models to predict clinical efficacy. We first optimized an in vitro microtumor model using a tumor-aligned ECM, a tumor-aligned medium, MCF-7 and MDA-MB-231 breast cancer spheroids, human umbilical vein endothelial cells, and human stromal cells to recapitulate the tissue architecture, chemical environment, and cellular organization of a growing and invading tumor. We assayed the microtumor for cell proliferation and invasion in a tumor-aligned extracellular matrix, exhibiting collagen deposition, acidity, glucose deprivation, and hypoxia. We found maximal proliferation and invasion when the multicellular spheroids were cultured in a tumor-aligned medium, having low pH and low glucose, with 10% fetal bovine serum under hypoxic conditions. In a 7-day assay, varying doses of fluorouracil or paclitaxel had differential effects on proliferation for MCF-7 and MDA-MB-231 tumor spheroids in microtumor compared to 2D and 3D monoculture. The microtumors exhibited a tumor morphology and drug response similar to published xenograft data, thus demonstrating a more physiologically predictive in vitro model.
Collapse
Affiliation(s)
- Gabriel Benton
- Trevigen Inc., Gaithersburg, MD, United States of America
| | - Gerald DeGray
- Trevigen Inc., Gaithersburg, MD, United States of America
| | | | - Jay George
- Trevigen Inc., Gaithersburg, MD, United States of America
| | | |
Collapse
|
42
|
Nash C, Hanby AM, Speirs V. Modelling the Molecular Pathology of Breast Cancer Initiation. MOLECULAR PATHOLOGY LIBRARY 2015. [DOI: 10.1007/978-1-4939-2886-6_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
43
|
Engineered microenvironments provide new insights into ovarian and prostate cancer progression and drug responses. Adv Drug Deliv Rev 2014; 79-80:193-213. [PMID: 24969478 DOI: 10.1016/j.addr.2014.06.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 05/30/2014] [Accepted: 06/16/2014] [Indexed: 02/06/2023]
Abstract
Tissue engineering technologies, which have originally been designed to reconstitute damaged tissue structure and function, can mimic not only tissue regeneration processes but also cancer development and progression. Bioengineered approaches allow cell biologists to develop sophisticated experimentally and physiologically relevant cancer models to recapitulate the complexity of the disease seen in patients. Tissue engineering tools enable three-dimensionality based on the design of biomaterials and scaffolds that re-create the geometry, chemistry, function and signalling milieu of the native tumour microenvironment. Three-dimensional (3D) microenvironments, including cell-derived matrices, biomaterial-based cell culture models and integrated co-cultures with engineered stromal components, are powerful tools to study dynamic processes like proteolytic functions associated with cancer progression, metastasis and resistance to therapeutics. In this review, we discuss how biomimetic strategies can reproduce a humanised niche for human cancer cells, such as peritoneal or bone-like microenvironments, addressing specific aspects of ovarian and prostate cancer progression and therapy response.
Collapse
|
44
|
Benton G, Arnaoutova I, George J, Kleinman HK, Koblinski J. Matrigel: from discovery and ECM mimicry to assays and models for cancer research. Adv Drug Deliv Rev 2014; 79-80:3-18. [PMID: 24997339 DOI: 10.1016/j.addr.2014.06.005] [Citation(s) in RCA: 308] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 06/19/2014] [Accepted: 06/24/2014] [Indexed: 01/06/2023]
Abstract
The basement membrane is an important extracellular matrix that is found in all epithelial and endothelial tissues. It maintains tissue integrity, serves as a barrier to cells and to molecules, separates different tissue types, transduces mechanical signals, and has many biological functions that help to maintain tissue specificity. A well-defined soluble basement membrane extract, termed BME/Matrigel, prepared from an epithelial tumor is similar in content to authentic basement membrane, and forms a hydrogel at 24-37°C. It is used in vitro as a substrate for 3D cell culture, in suspension for spheroid culture, and for various assays, such as angiogenesis, invasion, and dormancy. In vivo, BME/Matrigel is used for angiogenesis assays and to promote xenograft and patient-derived biopsy take and growth. Studies have shown that both the stiffness of the BME/Matrigel and its components (i.e. chemical signals) are responsible for its activity with so many different cell types. BME/Matrigel has widespread use in assays and in models that improve our understanding of tumor biology and help define therapeutic approaches.
Collapse
|
45
|
Kumar S, Shabi TS, Goormaghtigh E. A FTIR imaging characterization of fibroblasts stimulated by various breast cancer cell lines. PLoS One 2014; 9:e111137. [PMID: 25390361 PMCID: PMC4229076 DOI: 10.1371/journal.pone.0111137] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 09/29/2014] [Indexed: 12/21/2022] Open
Abstract
It is well known that the microenvironment plays a major role in breast cancer progression. Yet, the mechanism explaining the transition from normal fibroblasts to cancer-stimulated fibroblasts remains to be elucidated. Here we report a FTIR imaging study of the effects of three different breast cancer cell lines on normal fibroblasts in culture. Fibroblast activation process was monitored by FTIR imaging and spectra compared by multivariate statistical analyses. Principal component analysis evidenced that the fibroblasts stimulated by these cancer cell lines grouped together and remained distinctly separated from normal fibroblasts indicating a modified different chemical composition in the cancer-stimulated fibroblasts. Similar changes in fibroblasts were induced by the various breast cancer cell lines belonging to different sub-types. Most significant changes were observed in the region of 2950 and 1230 cm−1, possibly related to changes in lipids and in the 1230 cm−1 area assigned to phosphate vibrations (nucleotides). Interestingly, the cancer-cell induced changes in the fibroblasts also occurred when there was no possible direct contact between the two cell lines in the co-culture. When contact was possible, the spectral changes were similar, suggesting that soluble factors but not direct cell-cell interactions were responsible for fibroblast activation. Overall, the results indicate that IR imaging could be used in the future for analyzing the microenvironment of breast tumors.
Collapse
Affiliation(s)
- Saroj Kumar
- Laboratory for the Structure and Function of Biological Membranes, Center for Structural Biology and Bioinformatics, Université Libre de Bruxelles, Brussels, Belgium
- * E-mail: (SK); (EG)
| | - Thankaraj Salammal Shabi
- Organic Semiconductor Lab, Department of Polymer Science and Engineering, Zhejiang University, P. R. China
| | - Erik Goormaghtigh
- Laboratory for the Structure and Function of Biological Membranes, Center for Structural Biology and Bioinformatics, Université Libre de Bruxelles, Brussels, Belgium
- * E-mail: (SK); (EG)
| |
Collapse
|
46
|
Silva EJ, Argyris PP, Zou X, Ross KF, Herzberg MC. S100A8/A9 regulates MMP-2 expression and invasion and migration by carcinoma cells. Int J Biochem Cell Biol 2014; 55:279-87. [PMID: 25236491 DOI: 10.1016/j.biocel.2014.09.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 08/19/2014] [Accepted: 09/05/2014] [Indexed: 12/12/2022]
Abstract
Intracellular calprotectin (S100A8/A9) functions in the control of the cell cycle checkpoint at G2/M. Dysregulation of S100A8/A9 appears to cause loss of the checkpoint, which frequently characterizes head and neck squamous cell carcinoma (HNSCC). In the present study, we analyzed carcinoma cells for other S100A8/A9-directed changes in malignant phenotype. Using a S100A8/A9-negative human carcinoma cell line (KB), transfection to express S100A8 and S100A9 caused selective down-regulation of MMP-2 and inhibited in vitro invasion and migration. Conversely, silencing of endogenous S100A8 and S100A9 expression in TR146 cells, a well-differentiated HNSCC cell line, increased MMP-2 activity and in vitro invasion and migration. When MMP-2 expression was silenced, cells appeared to assume a less malignant phenotype. To more closely model the architecture of cell growth in vivo, cells were grown in a 3D collagen substrate, which was compared to 2D. Growth on 3D substrates caused greater MMP-2 expression. Whereas hypermethylation of CpG islands occurs frequently in HNSCC, S100A8/A9-dependent regulation of MMP-2 could not be explained by modification of the upstream promoters of MMP2 or TIMP2. Collectively, these results suggest that intracellular S100A8/A9 contributes to the cancer cell phenotype by modulating MMP-2 expression and activity to regulate cell migration and mobility.
Collapse
Affiliation(s)
- Emmanuel J Silva
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Prokopios P Argyris
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Xianqiong Zou
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Karen F Ross
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; Mucosal and Vaccine Research Center, Minneapolis Veterans Affairs Medical Center, Minneapolis, MN 55417, USA
| | - Mark C Herzberg
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; Mucosal and Vaccine Research Center, Minneapolis Veterans Affairs Medical Center, Minneapolis, MN 55417, USA.
| |
Collapse
|
47
|
Mimicking breast cancer-induced bone metastasis in vivo: current transplantation models and advanced humanized strategies. Cancer Metastasis Rev 2014; 33:721-35. [DOI: 10.1007/s10555-014-9499-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
48
|
Collazo J, Zhu B, Larkin S, Martin SK, Pu H, Horbinski C, Koochekpour S, Kyprianou N. Cofilin drives cell-invasive and metastatic responses to TGF-β in prostate cancer. Cancer Res 2014; 74:2362-73. [PMID: 24509905 PMCID: PMC4488067 DOI: 10.1158/0008-5472.can-13-3058] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cofilin (CFL) is an F-actin-severing protein required for the cytoskeleton reorganization and filopodia formation, which drives cell migration. CFL binding and severing of F-actin is controlled by Ser3 phosphorylation, but the contributions of this step to cell migration during invasion and metastasis of cancer cells are unclear. In this study, we addressed the question in prostate cancer cells, including the response to TGF-β, a critical regulator of migration. In cells expressing wild-type CFL, TGF-β treatment increased LIMK-2 activity and cofilin phosphorylation, decreasing filopodia formation. Conversely, constitutively active CFL (SerAla) promoted filipodia formation and cell migration mediated by TGF-β. Notably, in cocultures of prostate cancer epithelial cells and cancer-associated fibroblasts, active CFL promoted invasive migration in response to TGF-β in the microenvironment. Further, constitutively active CFL elevated the metastatic ability of prostate cancer cells in vivo. We found that levels of active CFL correlated with metastasis in a mouse model of prostate tumor and that in human prostate cancer, CFL expression was increased significantly in metastatic tumors. Our findings show that the actin-severing protein CFL coordinates responses to TGF-β that are needed for invasive cancer migration and metastasis.
Collapse
Affiliation(s)
- Joanne Collazo
- Department of Toxicology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Beibei Zhu
- Department of Urology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Spencer Larkin
- Department of Urology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Sarah K. Martin
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Hong Pu
- Department of Urology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Craig Horbinski
- Department of Pathology, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Shahriar Koochekpour
- Departments of Cancer Genetics and Urology, Roswell Park Cancer Institute, Buffalo, New York
| | - Natasha Kyprianou
- Department of Toxicology, University of Kentucky College of Medicine, Lexington, Kentucky
- Department of Urology, University of Kentucky College of Medicine, Lexington, Kentucky
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky
- Department of Pathology, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
49
|
Wang C, Tang Z, Zhao Y, Yao R, Li L, Sun W. Three-dimensional
in vitro
cancer models: a short review. Biofabrication 2014; 6:022001. [DOI: 10.1088/1758-5082/6/2/022001] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
50
|
Kan S, Konishi E, Arita T, Ikemoto C, Takenaka H, Yanagisawa A, Katoh N, Asai J. Podoplanin expression in cancer-associated fibroblasts predicts aggressive behavior in melanoma. J Cutan Pathol 2014; 41:561-7. [PMID: 24588302 DOI: 10.1111/cup.12322] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 02/24/2014] [Accepted: 02/25/2014] [Indexed: 11/27/2022]
Abstract
BACKGROUND Recent studies have showed podoplanin expression in several tumors, which has been associated with lymph node metastasis and poor prognosis. Podoplanin expression in cancer-associated fibroblasts also correlates with tumor progression. However, the association of podoplanin expression with melanomas remains unclear. METHODS To clarify the prognostic significance of podoplanin in melanoma, podoplanin expression in tumor cells and cancer-associated fibroblasts was examined by immunohistochemistry in tissue samples collected from 55 melanoma patients. RESULTS Podoplanin expression in tumor cells was identified in 38 patients (69.1%) but did not show correlation with characteristics of tumor progression such as tumor thickness (p = 0.52) and sentinel lymph node (SLN) metastasis (p = 0.79). Podoplanin expression in cancer-associated fibroblasts was observed in 25 patients (45.5%), 11 of whom (44.0%) had SLN metastasis. In contrast, only 4 of 30 patients (13.3%) with podoplanin-negative cancer-associated fibroblasts exhibited SLN metastasis. Podoplanin-positive cancer-associated fibroblasts were associated with increased tumor thickness and SLN metastasis. Furthermore, patients with podoplanin-positive cancer-associated fibroblasts had poorer survival than those with podoplanin-negative cancer-associated fibroblasts (p = 0.0148). CONCLUSION The presence of podoplanin expression in cancer-associated fibroblasts correlates with aggressive behavior in melanoma and might therefore serve as a useful prognostic factor for patients with melanoma.
Collapse
Affiliation(s)
- Saori Kan
- Department of Dermatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|