1
|
Bjerring JS, Khodour Y, Peterson EA, Sachs PC, Bruno RD. Intercellular mitochondrial transfer contributes to microenvironmental redirection of cancer cell fate. FEBS J 2025; 292:2306-2322. [PMID: 39934946 PMCID: PMC12062771 DOI: 10.1111/febs.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/04/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025]
Abstract
The mammary microenvironment has been shown to suppress tumor progression by redirecting cancer cells to adopt a normal mammary epithelial progenitor fate in vivo. However, the mechanism(s) by which this alteration occurs has yet to be defined. Here, we test the hypothesis that mitochondrial transfer from normal mammary epithelial cells to breast cancer cells plays a role in this redirection process. We evaluate mitochondrial transfer in 2D and 3D organoids using our unique 3D bioprinting system to produce chimeric organoids containing normal and cancer cells. We demonstrate that breast cancer tumoroid growth is hindered following interaction with mammary epithelial cells in both 2D and 3D environments. Furthermore, we show mitochondrial transfer occurs between donor mammary epithelial cells and recipient cancer cells primarily through tunneling nanotubes (TNTs) with minimal amounts seen from extracellular transfer of mitochondria, likely via extracellular vesicles (EVs). This organelle exchange results in various cellular and metabolic alterations within cancer cells, reducing their proliferative potential, and making them susceptible to microenvironmental control. Our results demonstrate that mitochondrial transfer contributes to microenvironmental redirection of cancer cells through alteration of metabolic and molecular functions of the recipient cancer cells. To the best of our knowledge, this is the first description of a 3D bioprinter-assisted organoid system for studying mitochondrial transfer. These studies are also the first mechanistic insights into the process of mammary microenvironmental redirection of cancer and provide a framework for new therapeutic strategies to control cancer.
Collapse
Affiliation(s)
- Julie Sofie Bjerring
- School of Medical Diagnostics and Translational Sciences, College of Health SciencesOld Dominion UniversityNorfolkVAUSA
| | - Yara Khodour
- School of Medical Diagnostics and Translational Sciences, College of Health SciencesOld Dominion UniversityNorfolkVAUSA
| | - Emilee Anne Peterson
- School of Medical Diagnostics and Translational Sciences, College of Health SciencesOld Dominion UniversityNorfolkVAUSA
| | - Patrick Christian Sachs
- School of Medical Diagnostics and Translational Sciences, College of Health SciencesOld Dominion UniversityNorfolkVAUSA
| | - Robert David Bruno
- School of Medical Diagnostics and Translational Sciences, College of Health SciencesOld Dominion UniversityNorfolkVAUSA
| |
Collapse
|
2
|
Kuracha MR, Radhakrishna U, Kuracha SV, Vegi N, Gurung JL, McVicker BL. New Horizons in Cancer Progression and Metastasis: Hippo Signaling Pathway. Biomedicines 2024; 12:2552. [PMID: 39595118 PMCID: PMC11591698 DOI: 10.3390/biomedicines12112552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/29/2024] [Accepted: 11/02/2024] [Indexed: 11/28/2024] Open
Abstract
The Hippo pathway is highly evolved to maintain tissue homeostasis in diverse species by regulating cell proliferation, differentiation, and apoptosis. In tumor biology, the Hippo pathway is a prime example of signaling molecules involved in cancer progression and metastasis. Hippo core elements LATS1, LATS2, MST1, YAP, and TAZ have critical roles in the maintenance of traditional tissue architecture and cell homeostasis. However, in cancer development, dysregulation of Hippo signaling results in tumor progression and the formation secondary cancers. Hippo components not only transmit biochemical signals but also act as mediators of mechanotransduction pathways during malignant neoplasm development and metastatic disease. This review confers knowledge of Hippo pathway core components and their role in cancer progression and metastasis and highlights the clinical role of Hippo pathway in cancer treatment. The Hippo signaling pathway and its unresolved mechanisms hold great promise as potential therapeutic targets in the emerging field of metastatic cancer research.
Collapse
Affiliation(s)
- Murali R. Kuracha
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Uppala Radhakrishna
- Department of Anesthesiology and Perioperative Medicine, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - Sreenaga V. Kuracha
- Comparative Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Navyasri Vegi
- Shri Vishnu College of Pharmacy, Andhra University, Bhimavaram 534202, Andhra Pradesh, India;
| | - Jhyama Lhamo Gurung
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Benita L. McVicker
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| |
Collapse
|
3
|
Ye Z, Nguyen TL, Dite GS, MacInnis RJ, Hopper JL, Li S. Mammographic Texture versus Conventional Cumulus Measure of Density in Breast Cancer Risk Prediction: A Literature Review. Cancer Epidemiol Biomarkers Prev 2024; 33:989-998. [PMID: 38787323 DOI: 10.1158/1055-9965.epi-23-1365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/01/2024] [Accepted: 05/22/2024] [Indexed: 05/25/2024] Open
Abstract
Mammographic textures show promise as breast cancer risk predictors, distinct from mammographic density. Yet, there is a lack of comprehensive evidence to determine the relative strengths as risk predictor of textures and density and the reliability of texture-based measures. We searched the PubMed database for research published up to November 2023, which assessed breast cancer risk associations [odds ratios (OR)] with texture-based measures and percent mammographic density (PMD), and their discrimination [area under the receiver operating characteristics curve (AUC)], using same datasets. Of 11 publications, for textures, six found stronger associations (P < 0.05) with 11% to 508% increases on the log scale by study, and four found weaker associations (P < 0.05) with 14% to 100% decreases, compared with PMD. Risk associations remained significant when fitting textures and PMD together. Eleven of 17 publications found greater AUCs for textures than PMD (P < 0.05); increases were 0.04 to 0.25 by study. Discrimination from PMD and these textures jointly was significantly higher than from PMD alone (P < 0.05). Therefore, different textures could capture distinct breast cancer risk information, partially independent of mammographic density, suggesting their joint role in breast cancer risk prediction. Some textures could outperform mammographic density for predicting breast cancer risk. However, obtaining reliable texture-based measures necessitates addressing various issues. Collaboration of researchers from diverse fields could be beneficial for advancing this complex field.
Collapse
Affiliation(s)
- Zhoufeng Ye
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
| | - Tuong L Nguyen
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
| | - Gillian S Dite
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
- Genetic Technologies Limited, Fitzroy, Australia
| | - Robert J MacInnis
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, East Melbourne, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
| | - Shuai Li
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Melbourne, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
4
|
Coelho LL, Vianna MM, da Silva DM, Gonzaga BMDS, Ferreira RR, Monteiro AC, Bonomo AC, Manso PPDA, de Carvalho MA, Vargas FR, Garzoni LR. Spheroid Model of Mammary Tumor Cells: Epithelial-Mesenchymal Transition and Doxorubicin Response. BIOLOGY 2024; 13:463. [PMID: 39056658 PMCID: PMC11273983 DOI: 10.3390/biology13070463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 07/28/2024]
Abstract
Breast cancer is the most prevalent cancer among women worldwide. Therapeutic strategies to control tumors and metastasis are still challenging. Three-dimensional (3D) spheroid-type systems more accurately replicate the features of tumors in vivo, working as a better platform for performing therapeutic response analysis. This work aimed to characterize the epithelial-mesenchymal transition and doxorubicin (dox) response in a mammary tumor spheroid (MTS) model. We evaluated the doxorubicin treatment effect on MCF-7 spheroid diameter, cell viability, death, migration and proteins involved in the epithelial-mesenchymal transition (EMT) process. Spheroids were also produced from tumors formed from 4T1 and 67NR cell lines. MTSs mimicked avascular tumor characteristics, exhibited adherens junction proteins and independently produced their own extracellular matrix. Our spheroid model supports the 3D culturing of cells isolated from mice mammary tumors. Through the migration assay, we verified a reduction in E-cadherin expression and an increase in vimentin expression as the cells became more distant from spheroids. Dox promoted cytotoxicity in MTSs and inhibited cell migration and the EMT process. These results suggest, for the first time, that this model reproduces aspects of the EMT process and describes the potential of dox in inhibiting the metastatic process, which can be further explored.
Collapse
Affiliation(s)
- Laura Lacerda Coelho
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| | - Matheus Menezes Vianna
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| | - Debora Moraes da Silva
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| | - Beatriz Matheus de Souza Gonzaga
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| | - Roberto Rodrigues Ferreira
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| | - Ana Carolina Monteiro
- Laboratory of Osteo and Tumor Immunology, Department of Immunobiology, Fluminense Federal University (UFF), Rio de Janeiro 24020-150, Brazil;
- Thymus Research Laboratory, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil;
| | - Adriana Cesar Bonomo
- Thymus Research Laboratory, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil;
| | - Pedro Paulo de Abreu Manso
- Laboratory of Pathology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil;
| | | | - Fernando Regla Vargas
- Laboratory of Epidemiology of Congenital Malformations, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil;
| | - Luciana Ribeiro Garzoni
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro 21040-900, Brazil; (L.L.C.); (M.M.V.); (D.M.d.S.); (B.M.d.S.G.); (R.R.F.)
| |
Collapse
|
5
|
Gu Y, Wang M, Gong Y, Li X, Wang Z, Wang Y, Jiang S, Zhang D, Li C. Unveiling breast cancer risk profiles: a survival clustering analysis empowered by an online web application. Future Oncol 2023; 19:2651-2667. [PMID: 38095059 DOI: 10.2217/fon-2023-0736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Aim: To develop a shiny app for doctors to investigate breast cancer treatments through a new approach by incorporating unsupervised clustering and survival information. Materials & methods: Analysis is based on the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) dataset, which contains 1726 subjects and 22 variables. Cox regression was used to identify survival risk factors for K-means clustering. Logrank tests and C-statistics were compared across different cluster numbers and Kaplan-Meier plots were presented. Results & conclusion: Our study fills an existing void by introducing a unique combination of unsupervised learning techniques and survival information on the clinician side, demonstrating the potential of survival clustering as a valuable tool in uncovering hidden structures based on distinct risk profiles.
Collapse
Affiliation(s)
- Yuan Gu
- Department of Statistics, The George Washington University, Washington, DC 20052, USA
| | - Mingyue Wang
- Department of Mathematics, Syracuse University, Syracuse, NY 13244, USA
| | - Yishu Gong
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, NY 02115, USA
| | - Xin Li
- Department of Statistics, The George Washington University, Washington, DC 20052, USA
| | - Ziyang Wang
- Department of Computer Science, University of Oxford, Oxford, OX1 3QD, UK
| | - Yuli Wang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Song Jiang
- Department of Biochemistry, Huzhou Institute of Biological Products Co., Ltd., 313017, China
| | - Dan Zhang
- Department of Information Science and Engineering, Shandong University, Shan Dong, China
| | - Chen Li
- Department of Biology, Chemistry and Pharmacy, Free University of Berlin, Berlin, 14195, Germany
| |
Collapse
|
6
|
Guo S, Huang X, Xu C, Yu M, Li Y, Wu Z, Zhou A, Xu P. Multiregional radiomic model for breast cancer diagnosis: value of ultrasound-based peritumoral and parenchymal radiomics. Quant Imaging Med Surg 2023; 13:3127-3139. [PMID: 37179905 PMCID: PMC10167447 DOI: 10.21037/qims-22-939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 03/10/2023] [Indexed: 05/15/2023]
Abstract
Background Breast cancer consists not only of neoplastic cells but also of significant changes in the surrounding and parenchymal stroma, which can be reflected in radiomics. This study aimed to perform breast lesion classification through an ultrasound-based multiregional (intratumoral, peritumoral, and parenchymal) radiomic model. Methods We retrospectively reviewed ultrasound images of breast lesions from institution #1 (n=485) and institution #2 (n=106). Radiomic features were extracted from different regions (intratumoral, peritumoral, and ipsilateral breast parenchymal) and selected to train the random forest classifier with the training cohort (n=339, a subset of the institution #1 dataset). Then, the intratumoral, peritumoral, and parenchymal, intratumoral & peritumoral (In&Peri), intratumoral & parenchymal (In&P), and intratumoral & peritumoral & parenchymal (In&Peri&P) models were developed and validated on the internal (n=146, another subset of institution 1) and external (n=106, institution #2 dataset) test cohorts. Discrimination was evaluated using the area under the curve (AUC). Calibration curve and Hosmer-Lemeshow test assessed calibration. Integrated discrimination improvement (IDI) was used to assess performance improvement. Results The performance of the In&Peri (AUC values 0.892 and 0.866), In&P (0.866 and 0.863), and In&Peri&P (0.929 and 0.911) models was significantly better than that of the intratumoral model (0.849 and 0.838) in the internal and external test cohorts (IDI test, all P<0.05). The intratumoral, In&Peri and In&Peri&P models showed good calibration (Hosmer-Lemeshow test, all P>0.05). The multiregional (In&Peri&P) model had the highest discrimination among the 6 radiomic models in the test cohorts, respectively. Conclusions The multiregional model combining radiomic information of intratumoral, peritumoral, and ipsilateral parenchymal regions yielded better performance than the intratumoral model in distinguishing malignant breast lesions from benign lesions.
Collapse
Affiliation(s)
- Suping Guo
- Department of Ultrasonography, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xingzhi Huang
- Department of Ultrasonography, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chao Xu
- Department of Ultrasonography, Jiangxi Tumor Hospital, Nanchang, China
| | - Meiqin Yu
- Department of Ultrasonography, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yaohui Li
- Department of Ultrasonography, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhenghua Wu
- Department of Ultrasonography, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Aiyun Zhou
- Department of Ultrasonography, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Pan Xu
- Department of Ultrasonography, First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
7
|
Suwannakot P, Nemec S, Peres NG, Du EY, Kilian KA, Gaus K, Kavallaris M, Gooding JJ. Electrostatic Assembly of Multiarm PEG-Based Hydrogels as Extracellular Matrix Mimics: Cell Response in the Presence and Absence of RGD Cell Adhesive Ligands. ACS Biomater Sci Eng 2023; 9:1362-1376. [PMID: 36826383 DOI: 10.1021/acsbiomaterials.2c01252] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Synthetic hydrogels have been used widely as extracellular matrix (ECM) mimics due to the ability to control and mimic physical and biochemical cues observed in natural ECM proteins such as collagen, laminin, and fibronectin. Most synthetic hydrogels are formed via covalent bonding resulting in slow gelation which is incompatible with drop-on-demand 3D bioprinting of cells and injectable hydrogels for therapeutic delivery. Herein, we developed an electrostatically crosslinked PEG-based hydrogel system for creating high-throughput 3D in vitro models using synthetic hydrogels to mimic the ECM cancer environment. A 3-arm PEG-based polymer backbone was first modified with either permanent cationic charged moieties (2-(methacryloyloxy)ethyl trimethylammonium) or permanent anionic charged moieties (3-sulfopropyl methacrylate potassium salt). The resulting charged polymers can be conjugated further with various amounts of cell adhesive RGD motifs (0, 25, 75, and 98%) to study the influences of RGD motifs on breast cancer (MCF-7) spheroid formation. Formation, stability, and mechanical properties of hydrogels were tested with, and without, RGD to evaluate the cellular response to material parameters in a 3D environment. The hydrogels can be degraded in the presence of salts at room temperature by breaking the interaction of oppositely charged polymer chains. MCF-7 cells could be released with high viability through brief exposure to NaCl solution. Flow cytometry characterization demonstrated that embedded MCF-7 cells proliferate better in a softer (60 Pa) 3D hydrogel environment compared to those that are stiffer (1160 Pa). As the stiffness increases, the RGD motif plays a role in promoting cell proliferation in the stiffer hydrogel. Flow cytometry characterization demonstrated that embedded MCF-7 cells proliferate better in a softer (60 Pa) 3D hydrogel environment compared to those that are stiffer (1160 Pa). As the stiffness increases, the RGD motif plays a role in promoting cell proliferation in the stiffer hydrogel. Additionally, cell viability was not impacted by the tested hydrogel stiffness range between 60 to 1160 Pa. Taken together, this PEG-based tuneable hydrogel system shows great promise as a 3D ECM mimic of cancer extracellular environments with controllable biophysical and biochemical properties. The ease of gelation and dissolution through salt concentration provides a way to quickly harvest cells for further analysis at any given time of interest without compromising cell viability.
Collapse
Affiliation(s)
- Panthipa Suwannakot
- School of Chemistry, UNSW, Sydney, New South Wales 2052, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, New South Wales 2052, Australia
| | - Stephanie Nemec
- School of Materials Science and Engineering, UNSW, Sydney, New South Wales 2052, Australia
| | - Newton Gil Peres
- School of Medical Sciences, EMBL Australia Node in Single Molecule Science, UNSW, Sydney, New South Wales 2052, Australia
| | - Eric Y Du
- School of Chemistry, UNSW, Sydney, New South Wales 2052, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, New South Wales 2052, Australia
| | - Kristopher A Kilian
- School of Chemistry, UNSW, Sydney, New South Wales 2052, Australia
- School of Materials Science and Engineering, UNSW, Sydney, New South Wales 2052, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, New South Wales 2052, Australia
| | - Katharina Gaus
- School of Medical Sciences, EMBL Australia Node in Single Molecule Science, UNSW, Sydney, New South Wales 2052, Australia
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW, Sydney, New South Wales 2052, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, New South Wales 2052, Australia
| | - J Justin Gooding
- School of Chemistry, UNSW, Sydney, New South Wales 2052, Australia
- Australian Centre for NanoMedicine, UNSW, Sydney, New South Wales 2052, Australia
| |
Collapse
|
8
|
Jung M, Skhinas JN, Du EY, Tolentino MAK, Utama RH, Engel M, Volkerling A, Sexton A, O'Mahony AP, Ribeiro JCC, Gooding JJ, Kavallaris M. A high-throughput 3D bioprinted cancer cell migration and invasion model with versatile and broad biological applicability. Biomater Sci 2022; 10:5876-5887. [PMID: 36149407 DOI: 10.1039/d2bm00651k] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Understanding the underlying mechanisms of migration and metastasis is a key focus of cancer research. There is an urgent need to develop in vitro 3D tumor models that can mimic physiological cell-cell and cell-extracellular matrix interactions, with high reproducibility and that are suitable for high throughput (HTP) drug screening. Here, we developed a HTP 3D bioprinted migration model using a bespoke drop-on-demand bioprinting platform. This HTP platform coupled with tunable hydrogel systems enables (i) the rapid encapsulation of cancer cells within in vivo tumor mimicking matrices, (ii) in situ and real-time measurement of cell movement, (iii) detailed molecular analysis for the study of mechanisms underlying cell migration and invasion, and (iv) the identification of novel therapeutic options. This work demonstrates that this HTP 3D bioprinted cell migration platform has broad applications across quantitative cell and cancer biology as well as drug screening.
Collapse
Affiliation(s)
- MoonSun Jung
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia. .,Australian Center for NanoMedicine, UNSW Sydney, Sydney, NSW, Australia.,School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| | - Joanna N Skhinas
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia. .,Australian Center for NanoMedicine, UNSW Sydney, Sydney, NSW, Australia
| | - Eric Y Du
- Australian Center for NanoMedicine, UNSW Sydney, Sydney, NSW, Australia.,School of Chemistry, UNSW Sydney, Sydney, NSW, Australia
| | - M A Kristine Tolentino
- Australian Center for NanoMedicine, UNSW Sydney, Sydney, NSW, Australia.,School of Chemistry, UNSW Sydney, Sydney, NSW, Australia
| | | | - Martin Engel
- Inventia Life Science Pty Ltd, Sydney, NSW, Australia
| | | | - Andrew Sexton
- Inventia Life Science Pty Ltd, Sydney, NSW, Australia
| | | | | | - J Justin Gooding
- Australian Center for NanoMedicine, UNSW Sydney, Sydney, NSW, Australia.,School of Chemistry, UNSW Sydney, Sydney, NSW, Australia
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia. .,Australian Center for NanoMedicine, UNSW Sydney, Sydney, NSW, Australia.,School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
9
|
Shi H, Rath EM, Lin RCY, Sarun KH, Clarke CJ, McCaughan BC, Ke H, Linton A, Lee K, Klebe S, Maitz J, Song K, Wang Y, Kao S, Cheng YY. 3-Dimensional mesothelioma spheroids provide closer to natural pathophysiological tumor microenvironment for drug response studies. Front Oncol 2022; 12:973576. [PMID: 36091141 PMCID: PMC9462830 DOI: 10.3389/fonc.2022.973576] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Traditional studies using cancer cell lines are often performed on a two-dimensional (2D) cell culture model with a low success rate of translating to Phase I or Phase II clinical studies. In comparison, with the advent of developments three-dimensional (3D) cell culture has been championed as the latest cellular model system that better mimics in vivo conditions and pathological conditions such as cancer. In comparison to biospecimens taken from in vivo tissue, the details of gene expression of 3D culture models are largely undefined, especially in mesothelioma – an aggressive cancer with very limited effective treatment options. In this study, we examined the veracity of the 3D mesothelioma cell culture model to study cell-to-cell interaction, gene expression and drug response from 3D cell culture, and compared them to 2D cell and tumor samples. We confirmed via SEM analysis that 3D cells grown using the spheroid methods expressed highly interconnected cell-to-cell junctions. The 3D spheroids were revealed to be an improved mini-tumor model as indicated by the TEM visualization of cell junctions and microvilli, features not seen in the 2D models. Growing 3D cell models using decellularized lung scaffold provided a platform for cell growth and infiltration for all cell types including primary cell lines. The most time-effective method was growing cells in spheroids using low-adhesive U-bottom plates. However, not every cell type grew into a 3D model using the the other methods of hanging drop or poly-HEMA. Cells grown in 3D showed more resistance to chemotherapeutic drugs, exhibiting reduced apoptosis. 3D cells stained with H&E showed cell-to-cell interactions and internal architecture that better represent that of in vivo patient tumors when compared to 2D cells. IHC staining revealed increased protein expression in 3D spheroids compared to 2D culture. Lastly, cells grown in 3D showed very different microRNA expression when compared to that of 2D counterparts. In conclusion, 3D cell models, regardless of which method is used. Showed a more realistic tumor microenvironment for architecture, gene expression and drug response, when compared to 2D cell models, and thus are superior preclinical cancer models.
Collapse
Affiliation(s)
- Huaikai Shi
- Asbestos Diseases Research Institute, Concord, Sydney, NSW, Australia
- *Correspondence: Huaikai Shi,
| | - Emma M. Rath
- Giannoulatou Laboratory, Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - Ruby C. Y. Lin
- Centre for Infectious Diseases and Microbiology, The Westmead Institute for Medical Research, Sydney, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Kadir Harun Sarun
- Asbestos Diseases Research Institute, Concord, Sydney, NSW, Australia
| | - Candice Julie Clarke
- Sydney Cardiothoracic Surgeons, Royal Prince Alfred Hospital (RPA) Medical Centre, Sydney, NSW, Australia
| | - Brian C. McCaughan
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
- Sydney Cardiothoracic Surgeons, Royal Prince Alfred Hospital (RPA) Medical Centre, Sydney, NSW, Australia
| | - Helen Ke
- Asbestos Diseases Research Institute, Concord, Sydney, NSW, Australia
- Concord Repatriation General Hospital, Sydney, NSW, Australia
| | - Anthony Linton
- Asbestos Diseases Research Institute, Concord, Sydney, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Concord Repatriation General Hospital, Sydney, NSW, Australia
| | - Kenneth Lee
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Concord Repatriation General Hospital, Sydney, NSW, Australia
| | - Sonja Klebe
- Asbestos Diseases Research Institute, Concord, Sydney, NSW, Australia
- Pathology, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia
| | - Joanneke Maitz
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Concord Repatriation General Hospital, Sydney, NSW, Australia
- The ANAZC Research Institute, Sydney, NSW, Australia
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian, China
| | - Yiwei Wang
- The ANAZC Research Institute, Sydney, NSW, Australia
- Jiangsu Provincial Engineering Research Centre of Traditional Chinese Medicine (TCM) External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, China
| | - Steven Kao
- Asbestos Diseases Research Institute, Concord, Sydney, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Yuen Yee Cheng
- Institute for Biomedical Materials & Devices, Faculty of Science, The University of Technology Sydney, NSW, Australia
| |
Collapse
|
10
|
Wilson GM, Dinh P, Pathmanathan N, Graham JD. Ductal Carcinoma in Situ: Molecular Changes Accompanying Disease Progression. J Mammary Gland Biol Neoplasia 2022; 27:101-131. [PMID: 35567670 PMCID: PMC9135892 DOI: 10.1007/s10911-022-09517-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/13/2022] [Indexed: 10/26/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) is a non-obligate precursor of invasive ductal carcinoma (IDC), whereby if left untreated, approximately 12% of patients develop invasive disease. The current standard of care is surgical removal of the lesion, to prevent potential progression, and radiotherapy to reduce risk of recurrence. There is substantial overtreatment of DCIS patients, considering not all DCIS lesions progress to invasive disease. Hence, there is a critical imperative to better predict which DCIS lesions are destined for poor outcome and which are not, allowing for tailored treatment. Active surveillance is currently being trialed as an alternative management practice, but this approach relies on accurately identifying cases that are at low risk of progression to invasive disease. Two DCIS-specific genomic profiling assays that attempt to distinguish low and high-risk patients have emerged, but imperfections in risk stratification coupled with a high price tag warrant the continued search for more robust and accessible prognostic biomarkers. This search has largely turned researchers toward the tumor microenvironment. Recent evidence suggests that a spectrum of cell types within the DCIS microenvironment are genetically and phenotypically altered compared to normal tissue and play critical roles in disease progression. Uncovering the molecular mechanisms contributing to DCIS progression has provided optimism for the search for well-validated prognostic biomarkers that can accurately predict the risk for a patient developing IDC. The discovery of such markers would modernize DCIS management and allow tailored treatment plans. This review will summarize the current literature regarding DCIS diagnosis, treatment, and pathology.
Collapse
Affiliation(s)
- Gemma M Wilson
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Phuong Dinh
- Westmead Breast Cancer Institute, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Nirmala Pathmanathan
- Westmead Breast Cancer Institute, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - J Dinny Graham
- Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia.
- Westmead Breast Cancer Institute, Westmead Hospital, Westmead, NSW, 2145, Australia.
| |
Collapse
|
11
|
Caballero D, Reis RL, Kundu SC. Current Trends in Microfluidics and Biosensors for Cancer Research Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:81-112. [DOI: 10.1007/978-3-031-04039-9_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
12
|
Kundu B, Caballero D, Abreu CM, Reis RL, Kundu SC. The Tumor Microenvironment: An Introduction to the Development of Microfluidic Devices. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:115-138. [DOI: 10.1007/978-3-031-04039-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
13
|
Structural Biology of the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1350:91-100. [PMID: 34888845 DOI: 10.1007/978-3-030-83282-7_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancers can be described as "rogue organs" (Balkwill FR, Capasso M, Hagemann T, J Cell Sci 125:5591-5596, 2012) because they are composed of multiple cell types and tissues. The transformed cells can recruit and alter healthy cells from surrounding tissues for their own benefit. It is these interactions that create the tumor microenvironment (TME). The TME describes the cells, factors, and extracellular matrix proteins that make up the tumor and the area around it; the biology of the TME influences tumor progression. Changes in the TME can lead to the growth and development of the tumor, the death of the tumor, or tumor metastasis. Metastasis is the process by which cancer spreads from its initial site to a different part of the body. Metastasis occurs when cancer cells enter the circulatory system or lymphatic system after they break away from a tumor. Once the cells leave, they can travel to a different part of the body and form new tumors. Therefore, understanding the TME is critical to fully understand cancer and find a way to successfully combat it. Knowledge of the TME can better inform researchers of the ability of potential therapies to reach tumor cells. It can also give researchers potential targets to kill the tumor. Instead of directly killing the cancer cells, therapies can target an aspect of the TME which could then halt tumor development or lead to tumor death. In other cases, targeting another aspect of the TME could make it easier for another therapy to kill the cancer cells, for example, using nanoparticles with collagenases to target the collagen in the surrounding environment to expose the cancer cells to drugs (Zinger A, et al, ACS Nano 13(10):11008-11021, 2019).The TME can be split simply into cells and the structural matrix. Within these groups are fibroblasts, structural proteins, immune cells, lymphocytes, bone marrow-derived inflammatory cells, blood vessels, and signaling molecules (Spill F, et al, Curr Opin Biotechnol 40:41-48, 2016; Del Prete A, et al, Curr Opin Pharmacol 35:40-47, 2017; Arneth B, Medicina (Kaunas) 56(1), 2019). From structure to providing nutrients for growth, each of these components plays a critical role in tumor maintenance. Together these components impact cancer growth, development, and resistance to therapies (Hanahan D, Coussens LM, Cancer Cell 21:309-322, 2012). In this chapter, we will describe the TME and express the importance of the cellular and structural elements of the TME.
Collapse
|
14
|
Modeling Tumor: Lymphatic Interactions in Lymphatic Metastasis of Triple Negative Breast Cancer. Cancers (Basel) 2021; 13:cancers13236044. [PMID: 34885152 PMCID: PMC8656640 DOI: 10.3390/cancers13236044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Lymphatic metastasis is a critical prognostic factor of breast cancer aggressiveness and patient survival. Since existing therapeutic approaches have shown limited efficacy, new strategies to identify effective therapeutic targets for reducing breast cancer lymphatic metastasis are needed. We have used novel culture chambers, designed and fabricated by our group, to develop 3D models in which we can study spat ial interactions between breast cancer cells and lymphatic cells as they occur in real-time. This approach provides information on the complex cell–cell interactions involved in lymphatic metastasis of breast cancers. Factors in the secretome of the lymphatic cells promote invasive outgrowths from 3D cultures of breast cancer cells, suggesting that targeting interactions between breast cancer cells and lymphatic cells could be a potential therapeutic approach for the prevention of lymphatic metastasis. Abstract Breast cancer frequently metastasizes to lymphatics and the presence of breast cancer cells in regional lymph nodes is an important prognostic factor. Delineating the mechanisms by which breast cancer cells disseminate and spatiotemporal aspects of interactions between breast cancer cells and lymphatics is needed to design new therapies to prevent lymphatic metastases. As triple-negative breast cancer (TNBC) has a high incidence of lymphatic metastasis, we used a three-dimensional (3D) coculture model of human TNBC cells and human microvascular lymphatic endothelial cells (LECs) to analyze TNBC:LEC interactions. Non-invasive analyses such as live-cell imaging in real-time and collection of conditioned media for secretomic analysis were facilitated by our novel microfluidic chambers. The volumes of 3D structures formed in TNBC:LEC cocultures are greater than that of 3D structures formed by either LEC or TNBC monocultures. Over 4 days of culture there is an increase in multicellular invasive outgrowths from TNBC spheroids and an association of TNBC spheroids with LEC networks. The increase in invasive phenotype also occurred when TNBC spheroids were cultured in LEC-conditioned media and in wells linked to ones containing LEC networks. Our results suggest that modeling spatiotemporal interactions between TNBC and LECs may reveal paracrine signaling that could be targeted to reduce lymphatic metastasis.
Collapse
|
15
|
Fabrication of 3D-Printed Interpenetrating Hydrogel Scaffolds for Promoting Chondrogenic Differentiation. Polymers (Basel) 2021; 13:polym13132146. [PMID: 34209853 DOI: 10.3390/polym13132146] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/13/2022] Open
Abstract
The limited self-healing ability of cartilage necessitates the application of alternative tissue engineering strategies for repairing the damaged tissue and restoring its normal function. Compared to conventional tissue engineering strategies, three-dimensional (3D) printing offers a greater potential for developing tissue-engineered scaffolds. Herein, we prepared a novel photocrosslinked printable cartilage ink comprising of polyethylene glycol diacrylate (PEGDA), gelatin methacryloyl (GelMA), and chondroitin sulfate methacrylate (CSMA). The PEGDA-GelMA-CSMA scaffolds possessed favorable compressive elastic modulus and degradation rate. In vitro experiments showed good adhesion, proliferation, and F-actin and chondrogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) on the scaffolds. When the CSMA concentration was increased, the compressive elastic modulus, GAG production, and expression of F-actin and cartilage-specific genes (COL2, ACAN, SOX9, PRG4) were significantly improved while the osteogenic marker genes of COL1 and ALP were decreased. The findings of the study indicate that the 3D-printed PEGDA-GelMA-CSMA scaffolds possessed not only adequate mechanical strength but also maintained a suitable 3D microenvironment for differentiation, proliferation, and extracellular matrix production of BMSCs, which suggested this customizable 3D-printed PEGDA-GelMA-CSMA scaffold may have great potential for cartilage repair and regeneration in vivo.
Collapse
|
16
|
Tissue Imaging and Quantification Relying on Endogenous Contrast. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 3233:257-288. [PMID: 34053031 DOI: 10.1007/978-981-15-7627-0_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Cell-matrix interactions play an important role in regulating a variety of essential processes in multicellular organisms, and are closely associated with numerous diseases. Modified interactions have major effects upon key features of both cells and extracellular matrix (ECM), and a thorough understanding of changes in these features can lead to critically important insights of diseases as well as the identification of effective therapeutic targets. Here, we summarize recent advances in quantitative, optical imaging of cellular metabolism and ECM spatial organization using endogenous sources of contrast. Specifically, we focus on the two-photon excited fluorescence (TPEF) imaging of autofluorescent cellular coenzymes, NAD(P)H and FAD, for the extraction of metabolic information described by optical biomarkers including cellular redox state, NAD(P)H fluorescence lifetime, and mitochondrial clustering. We show representative applications in assessing adipose tissue function and detecting malignant lesions in human skin, and further demonstrate that a combination of these optical metrics can provide complementary insights into the underlying biological mechanisms. In addition, we review the development of quantitative analysis methods to extract spatial orientation and organization metrics of collagen fibers, a major ECM component, and demonstrate applications of these approaches in two and three dimensions in several diseases, including would healing, osteoarthritis and cancer, as well as assessments of matrix remodeling in hormone-regulated engineered breast tissues. Finally, we summarize this chapter and discuss important research directions that we expect will evolve in the near future.
Collapse
|
17
|
Garry MG, Kren S, Wenger JB, Garry DJ. Biologically Derived, Three-Dimensional, Embryonic Scaffolds for Long-Term Cardiomyocyte Culture. Stem Cells Dev 2021; 30:697-704. [PMID: 33843275 DOI: 10.1089/scd.2021.0060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The ability to maintain viable cultures of mature, primary cardiomyocytes is challenging. The lack of viable cardiomyocyte cultures severely limits in vitro biochemical assays, toxicology assays, drug screening assays, and other analyses. Here, we describe a novel three-dimensional (3D) embryonic scaffold, which supports the culture of postnatal day 7 murine cardiomyocytes within the embryonic heart for, at least, 28 days. We have observed that these cardiomyocytes display normal differentiation, protein expression, and function after extended culture. This novel culture system will allow for prolonged treatment of cardiomyocytes in a natural 3D orientation and has the potential for providing a superior tool for the screening of therapeutic compounds.
Collapse
Affiliation(s)
- Mary G Garry
- Lillehei Heart Institute (LHI), University of Minnesota, Minneapolis, Minnesota, USA.,Institute for Engineering in Medicine (IEM), University of Minnesota, Minneapolis, Minnesota, USA.,Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Stefan Kren
- Lillehei Heart Institute (LHI), University of Minnesota, Minneapolis, Minnesota, USA.,Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Joseph B Wenger
- Lillehei Heart Institute (LHI), University of Minnesota, Minneapolis, Minnesota, USA.,Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Daniel J Garry
- Lillehei Heart Institute (LHI), University of Minnesota, Minneapolis, Minnesota, USA.,Institute for Engineering in Medicine (IEM), University of Minnesota, Minneapolis, Minnesota, USA.,Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
18
|
Xu L, Yang P, Hu K, Wu Y, Xu-Welliver M, Wan Y, Luo C, Wang J, Wang J, Qin J, Rong Y, Niu T. Prediction of neoadjuvant chemotherapy response in high-grade osteosarcoma: added value of non-tumorous bone radiomics using CT images. Quant Imaging Med Surg 2021; 11:1184-1195. [PMID: 33816159 DOI: 10.21037/qims-20-681] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background This study aimed to determine the impact of including radiomics analysis of non-tumorous bone region of interest in improving the performance of pathological response prediction to chemotherapy in high-grade osteosarcomas (HOS), compared to radiomics analysis of tumor region alone. Methods This retrospective study included 157 patients diagnosed with HOS between November 2013 and November 2017 (age range, 5-44 years; mean age, 16.99 ±7.42 years), in which 69 and 88 patients were diagnosed as pathological good response (pGR) and non-pGR, respectively. Radiomics features were extracted from tumor and non-tumorous bone regions based on diagnostic CT images. Pathological response classifiers were developed and validated via leave-one-out cross validation (LOOCV) and independent validation methods by using the area under the receiver operating characteristic curve (AUC) value as the figure of merit. Results Using the LOOCV, the classifiers combining features from tumor and non-tumorous regions showed better prediction performance than those from tumor region alone (AUC, 0.8207±0.0043 vs. 0.7799±0.0044). The combined classifier also showed better performance than the tumor feature-based classifier in both training and validation datasets [training dataset: 0.791, 95% confidence interval (CI), 0.706-0.860 vs. 0.766, 95% CI, 0.679-0.840; validation dataset: 0.816, 95% CI, 0.662-0.920 vs. 0.766, 95% CI, 0.606-0.885]. Conclusions Radiomics analysis of combined tumor and non-tumorous bone features showed improved performance of pathological response prediction to chemotherapy in HOS compared to that of tumor features alone. Moreover, the proposed classifier had the potential to predict pathological response to chemotherapy for HOS patients.
Collapse
Affiliation(s)
- Lei Xu
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Pengfei Yang
- College of Biomedical Engineering &Instrument Science, Zhejiang University, Hangzhou, China
| | - Kun Hu
- Nuclear & Radiological Engineering and Medical Physics Programs, Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Yan Wu
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Meng Xu-Welliver
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Yidong Wan
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Chen Luo
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Jing Wang
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Jinhua Wang
- Department of Radiology, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Jiale Qin
- Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Rong
- Department of Radiation Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Tianye Niu
- Nuclear & Radiological Engineering and Medical Physics Programs, Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
19
|
Banani MA, Rahmatullah M, Farhan N, Hancox Z, Yousaf S, Arabpour Z, Moghaddam ZS, Mozafari M, Sefat F. Adipose tissue-derived mesenchymal stem cells for breast tissue regeneration. Regen Med 2021; 16:47-70. [PMID: 33533667 DOI: 10.2217/rme-2020-0045] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 01/08/2021] [Indexed: 02/06/2023] Open
Abstract
With an escalating incidence of breast cancer cases all over the world and the deleterious psychological impact that mastectomy has on patients along with several limitations of the currently applied modalities, it's plausible to seek unconventional approaches to encounter such a burgeoning issue. Breast tissue engineering may allow that chance via providing more personalized solutions which are able to regenerate, mimicking natural tissues also facing the witnessed limitations. This review is dedicated to explore the utilization of adipose tissue-derived mesenchymal stem cells for breast tissue regeneration among postmastectomy cases focusing on biomaterials and cellular aspects in terms of harvesting, isolation, differentiation and new tissue formation as well as scaffolds types, properties, material-host interaction and an in vitro breast tissue modeling.
Collapse
Affiliation(s)
- Mohammed A Banani
- Division of Surgery & Interventional Science, University College London, London, NW3 2PS, UK
| | - Mohammed Rahmatullah
- Division of Surgery & Interventional Science, University College London, London, NW3 2PS, UK
| | - Nawras Farhan
- Division of Surgery & Interventional Science, University College London, London, NW3 2PS, UK
| | - Zoe Hancox
- Department of Biomedical & Electronics Engineering, School of Engineering, University of Bradford, Bradford, BD7 1DP, UK
| | - Safiyya Yousaf
- Department of Biomedical & Electronics Engineering, School of Engineering, University of Bradford, Bradford, BD7 1DP, UK
| | - Zohreh Arabpour
- Department of Biomedical & Electronics Engineering, School of Engineering, University of Bradford, Bradford, BD7 1DP, UK
| | - Zoha Salehi Moghaddam
- Department of Biomedical & Electronics Engineering, School of Engineering, University of Bradford, Bradford, BD7 1DP, UK
- Interdisciplinary Research Centre in Polymer Science & Technology (IRC Polymer), University of Bradford, Bradford, BD7 1DP, UK
| | - Masoud Mozafari
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, M5G 1X5, Canada
| | - Farshid Sefat
- Department of Biomedical & Electronics Engineering, School of Engineering, University of Bradford, Bradford, BD7 1DP, UK
- Interdisciplinary Research Centre in Polymer Science & Technology (IRC Polymer), University of Bradford, Bradford, BD7 1DP, UK
| |
Collapse
|
20
|
Moon HR, Ozcelikkale A, Yang Y, Elzey BD, Konieczny SF, Han B. An engineered pancreatic cancer model with intra-tumoral heterogeneity of driver mutations. LAB ON A CHIP 2020; 20:3720-3732. [PMID: 32909573 PMCID: PMC9178523 DOI: 10.1039/d0lc00707b] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a complex disease with significant intra-tumoral heterogeneity (ITH). Currently, no reliable PDAC tumor model is available that can present ITH profiles in a controlled manner. We develop an in vitro microfluidic tumor model mimicking the heterogeneous accumulation of key driver mutations of human PDAC using cancer cells derived from genetically engineered mouse models. These murine pancreatic cancer cell lines have KPC (Kras and Trp53 mutations) and KIC genotypes (Kras mutation and Cdkn2a deletion). Also, the KIC genotypes have two distinct phenotypes - mesenchymal or epithelial. The tumor model mimics the ITH of human PDAC to study the effects of ITH on the gemcitabine response. The results show gemcitabine resistance induced by ITH. Remarkably, it shows that cancer cell-cell interactions induce the gemcitabine resistance potentially through epithelial-mesenchymal-transition. The tumor model can provide a useful testbed to study interaction mechanisms between heterogeneous cancer cell subpopulations.
Collapse
Affiliation(s)
- Hye-Ran Moon
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Barros da Silva P, Coelho M, Bidarra SJ, Neves SC, Barrias CC. Reshaping in vitro Models of Breast Tissue: Integration of Stromal and Parenchymal Compartments in 3D Printed Hydrogels. Front Bioeng Biotechnol 2020; 8:494. [PMID: 32596217 PMCID: PMC7300215 DOI: 10.3389/fbioe.2020.00494] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 04/28/2020] [Indexed: 12/30/2022] Open
Abstract
Breast tissue consists of an epithelial parenchyma embedded in stroma, of heterogeneous and complex composition, undergoing several morphological and functional alterations throughout females' lifespan. Improved knowledge on the crosstalk between parenchymal and stromal mammary cells should provide important insights on breast tissue dynamics, both under healthy and diseased states. Here, we describe an advanced 3D in vitro model of breast tissue, combining multiple components, namely stromal cells and their extracellular matrix (ECM), as well as parenchymal epithelial cells, in a hybrid system. To build the model, porous scaffolds were produced by extrusion 3D printing of peptide-modified alginate hydrogels, and then populated with human mammary fibroblasts. Seeded fibroblasts were able to adhere, spread and produce endogenous ECM, providing adequate coverage of the scaffold surface, without obstructing the pores. On a second stage, a peptide-modified alginate pre-gel laden with mammary gland epithelial cells was used to fill the scaffold's pores, forming a hydrogel in situ by ionic crosslinking. Throughout time, epithelial cells formed prototypical mammary acini-like structures, in close proximity with fibroblasts and their ECM. This generated a heterotypic 3D model that partially recreates both stromal and parenchymal compartments of breast tissue, promoting cell-cell and cell-matrix crosstalk. Furthermore, the hybrid system could be easily dissolved for cell recovery and subsequent analysis by standard cellular/molecular assays. In particular, we show that retrieved cell populations could be discriminated by flow cytometry using cell-type specific markers. This integrative 3D model stands out as a promising in vitro platform for studying breast stroma-parenchyma interactions, both under physiological and pathological settings.
Collapse
Affiliation(s)
- Patrícia Barros da Silva
- i3S—Instituto de Inovação e Investigação em Saúde, Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- FEUP—Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| | - Mariana Coelho
- i3S—Instituto de Inovação e Investigação em Saúde, Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Sílvia Joana Bidarra
- i3S—Instituto de Inovação e Investigação em Saúde, Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Sara Carvalheira Neves
- i3S—Instituto de Inovação e Investigação em Saúde, Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- FEUP—Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| | - Cristina Carvalho Barrias
- i3S—Instituto de Inovação e Investigação em Saúde, Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
22
|
Tao S, Zhao Z, Zhang X, Guan X, Wei J, Yuan B, He S, Zhao D, Zhang J, Liu Q, Ding Y. The role of macrophages during breast cancer development and response to chemotherapy. Clin Transl Oncol 2020; 22:1938-1951. [PMID: 32279178 DOI: 10.1007/s12094-020-02348-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 03/21/2020] [Indexed: 12/14/2022]
Abstract
Macrophages play an important role in the immune system as a key host defense against pathogens. Non-polarized macrophages can differentiate into pro-inflammatory classical pathway-activated macrophages or anti-inflammatory alternative pathway-activated macrophages, both of which play central roles in breast cancer growth and progression in a process called polarization of macrophages. Classical pathway-activated and alternative pathway-activated macrophages can transform into each other and their transformational properties and orientation are determined by cytokines in the tumor microenvironment. Tumor-associated macrophages display many functions, such as tissue reforming, participating in inflammation and tumor growth in breast cancer progression. Some cytokines, such as interleukins and transcriptional activators, reside in the tumor microenvironment and influence tumor-associated macrophages. Chemotherapy is a common treatment for breast cancer and macrophages play an important role in mammary tumor cell migration, cancer invasion, and angiogenesis. This review summarizes the activities of tumor-associated macrophages in the mammary tumor, chemotherapeutic processes and some potential strategies for breast cancer therapy.
Collapse
Affiliation(s)
- S Tao
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Z Zhao
- The Second Clinical School of Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China.,The 2nd Clinical School of Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.,The 85th Hospital of CPLA, Shanghai, 200040, China.,Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, Zhuhai, 519015, China
| | - X Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - X Guan
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - J Wei
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - B Yuan
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - S He
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - D Zhao
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - J Zhang
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, 130062, China.
| | - Q Liu
- The Second Clinical School of Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510120, Guangdong, China. .,Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China. .,The 2nd Clinical School of Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China. .,Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, Zhuhai, 519015, China.
| | - Y Ding
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, 130062, China.
| |
Collapse
|
23
|
Revisiting Cancer Stem Cells as the Origin of Cancer-Associated Cells in the Tumor Microenvironment: A Hypothetical View from the Potential of iPSCs. Cancers (Basel) 2020; 12:cancers12040879. [PMID: 32260363 PMCID: PMC7226406 DOI: 10.3390/cancers12040879] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/17/2020] [Accepted: 03/30/2020] [Indexed: 12/18/2022] Open
Abstract
The tumor microenvironment (TME) has an essential role in tumor initiation and development. Tumor cells are considered to actively create their microenvironment during tumorigenesis and tumor development. The TME contains multiple types of stromal cells, cancer-associated fibroblasts (CAFs), Tumor endothelial cells (TECs), tumor-associated adipocytes (TAAs), tumor-associated macrophages (TAMs) and others. These cells work together and with the extracellular matrix (ECM) and many other factors to coordinately contribute to tumor growth and maintenance. Although the types and functions of TME cells are well understood, the origin of these cells is still obscure. Many scientists have tried to demonstrate the origin of these cells. Some researchers postulated that TME cells originated from surrounding normal tissues, and others demonstrated that the origin is cancer cells. Recent evidence demonstrates that cancer stem cells (CSCs) have differentiation abilities to generate the original lineage cells for promoting tumor growth and metastasis. The differentiation of CSCs into tumor stromal cells provides a new dimension that explains tumor heterogeneity. Using induced pluripotent stem cells (iPSCs), our group postulates that CSCs could be one of the key sources of CAFs, TECs, TAAs, and TAMs as well as the descendants, which support the self-renewal potential of the cells and exhibit heterogeneity. In this review, we summarize TME components, their interactions within the TME and their insight into cancer therapy. Especially, we focus on the TME cells and their possible origin and also discuss the multi-lineage differentiation potentials of CSCs exploiting iPSCs to create a society of cells in cancer tissues including TME.
Collapse
|
24
|
Bizzarri M, Giuliani A, Minini M, Monti N, Cucina A. Constraints Shape Cell Function and Morphology by Canalizing the Developmental Path along the Waddington's Landscape. Bioessays 2020; 42:e1900108. [DOI: 10.1002/bies.201900108] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 01/17/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Mariano Bizzarri
- Systems Biology Group Laboratory, Department of Experimental MedicineSapienza University 00161 Rome Italy
| | - Alessandro Giuliani
- Environment and Health DepartmentIstituto Superiore di Sanità 00161 Rome Italy
| | - Mirko Minini
- Systems Biology Group Laboratory, Department of Experimental MedicineSapienza University 00161 Rome Italy
- Department of Surgery “Pietro Valdoni,”Sapienza University of Rome 00161 Rome Italy
| | - Noemi Monti
- Systems Biology Group Laboratory, Department of Experimental MedicineSapienza University 00161 Rome Italy
- Department of Surgery “Pietro Valdoni,”Sapienza University of Rome 00161 Rome Italy
| | - Alessandra Cucina
- Department of Surgery “Pietro Valdoni,”Sapienza University of Rome 00161 Rome Italy
- Azienda Policlinico Umberto I 00161 Rome Italy
| |
Collapse
|
25
|
Bradney MJ, Venis SM, Yang Y, Konieczny SF, Han B. A Biomimetic Tumor Model of Heterogeneous Invasion in Pancreatic Ductal Adenocarcinoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1905500. [PMID: 31997571 PMCID: PMC7069790 DOI: 10.1002/smll.201905500] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/13/2019] [Indexed: 05/21/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a complex, heterogeneous, and genetically unstable disease. Its tumor microenvironment (TME) is complicated by heterogeneous cancer cell populations and strong desmoplastic stroma. This complex and heterogeneous environment makes it challenging to discover and validate unique therapeutic targets. Reliable and relevant in vitro PDAC tumor models can significantly advance the understanding of the PDAC TME and may enable the discovery and validation of novel drug targets. In this study, an engineered tumor model is developed to mimic the PDAC TME. This biomimetic model, named ductal tumor-microenvironment-on-chip (dT-MOC), permits analysis and experimentation on the epithelial-mesenchymal transition (EMT) and local invasion with intratumoral heterogeneity. This dT-MOC is a microfluidic platform where a duct of murine genetically engineered pancreatic cancer cells is embedded within a collagen matrix. The cancer cells used carry two of the three mutations of KRAS, CDKN2A, and TP53, which are key driver mutations of human PDAC. The intratumoral heterogeneity is mimicked by co-culturing these cancer cells. Using the dT-MOC model, heterogeneous invasion characteristics, and response to transforming growth factor-beta1 are studied. A mechanism of EMT and local invasion caused by the interaction between heterogeneous cancer cell populations is proposed.
Collapse
Affiliation(s)
- Michael J Bradney
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Stephanie M Venis
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Yi Yang
- Department of Biological Science, Purdue University, West Lafayette, IN, 47907, USA
| | - Stephen F Konieczny
- Department of Biological Science, Purdue University, West Lafayette, IN, 47907, USA
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| |
Collapse
|
26
|
Ding J, Yao Y, Huang G, Wang X, Yi J, Zhang N, Liu C, Wang K, Zhang Y, Wang M, Liu P, Ye M, Li M, Cheng H. Targeting the EphB4 receptor tyrosine kinase sensitizes HER2-positive breast cancer cells to Lapatinib. Cancer Lett 2020; 475:53-64. [PMID: 32006616 DOI: 10.1016/j.canlet.2020.01.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/23/2019] [Accepted: 01/24/2020] [Indexed: 02/07/2023]
Abstract
Clinical data analysis reveals that the expression of the EphB4 receptor tyrosine kinase is significantly elevated in HER2-positive breast cancer and high levels of EphB4 strongly correlate with poor disease prognosis. However, the impact of EphB4 activation on HER2-positive breast cancer cells and the potential of EphB4 as a therapeutic target remain to be explored. Here, we show that EphB4 overexpression confers gain-of-function activities to HER2-positive breast cancer cells, rendering resistance to a HER2/EGFR inhibitor Lapatinib. Furthermore, using integrated transcriptomic and tyrosine phosphoproteomic analyses, followed by biochemical confirmation, we establish that EphB4 activation engages the SHP2/GAB1-MEK signaling cascade and downstream c-MYC activation, and thereby limits the overall drug responses to Lapatinib. Finally, we demonstrate that, in HER2-positive breast tumors, inhibition of EphB4 combined with Lapatinib is more effective than either alone. These findings provide new insights into the signaling networks dictating therapeutic response to Lapatinib as well as a rationale for co-targeting EphB4 in HER2-positive breast cancer.
Collapse
Affiliation(s)
- Jinlei Ding
- Cancer Institute, Department of Oncology, Department of Thoracic Surgery, The Second Hospital of Dalian Medical University, Dalian Key Laboratory of Molecular Targeted Cancer Therapy, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yating Yao
- Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, China; University of Chinese Academy of Sciences, Beijing, China
| | - Gena Huang
- Cancer Institute, Department of Oncology, Department of Thoracic Surgery, The Second Hospital of Dalian Medical University, Dalian Key Laboratory of Molecular Targeted Cancer Therapy, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xiaonan Wang
- Cancer Institute, Department of Oncology, Department of Thoracic Surgery, The Second Hospital of Dalian Medical University, Dalian Key Laboratory of Molecular Targeted Cancer Therapy, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Jingyan Yi
- Cancer Institute, Department of Oncology, Department of Thoracic Surgery, The Second Hospital of Dalian Medical University, Dalian Key Laboratory of Molecular Targeted Cancer Therapy, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Nan Zhang
- Cancer Institute, Department of Oncology, Department of Thoracic Surgery, The Second Hospital of Dalian Medical University, Dalian Key Laboratory of Molecular Targeted Cancer Therapy, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Chongya Liu
- Cancer Institute, Department of Oncology, Department of Thoracic Surgery, The Second Hospital of Dalian Medical University, Dalian Key Laboratory of Molecular Targeted Cancer Therapy, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Kainan Wang
- Cancer Institute, Department of Oncology, Department of Thoracic Surgery, The Second Hospital of Dalian Medical University, Dalian Key Laboratory of Molecular Targeted Cancer Therapy, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yuan Zhang
- Cancer Institute, Department of Oncology, Department of Thoracic Surgery, The Second Hospital of Dalian Medical University, Dalian Key Laboratory of Molecular Targeted Cancer Therapy, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Min Wang
- Cancer Institute, Department of Oncology, Department of Thoracic Surgery, The Second Hospital of Dalian Medical University, Dalian Key Laboratory of Molecular Targeted Cancer Therapy, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Pixu Liu
- Cancer Institute, Department of Oncology, Department of Thoracic Surgery, The Second Hospital of Dalian Medical University, Dalian Key Laboratory of Molecular Targeted Cancer Therapy, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
| | - Mingliang Ye
- Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, China.
| | - Man Li
- Cancer Institute, Department of Oncology, Department of Thoracic Surgery, The Second Hospital of Dalian Medical University, Dalian Key Laboratory of Molecular Targeted Cancer Therapy, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
| | - Hailing Cheng
- Cancer Institute, Department of Oncology, Department of Thoracic Surgery, The Second Hospital of Dalian Medical University, Dalian Key Laboratory of Molecular Targeted Cancer Therapy, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
| |
Collapse
|
27
|
Li H, Mendel KR, Lan L, Sheth D, Giger ML. Digital Mammography in Breast Cancer: Additive Value of Radiomics of Breast Parenchyma. Radiology 2019; 291:15-20. [PMID: 30747591 PMCID: PMC6445042 DOI: 10.1148/radiol.2019181113] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 12/14/2018] [Accepted: 01/02/2019] [Indexed: 11/11/2022]
Abstract
Background Previous studies have suggested that breast parenchymal texture features may reflect the biologic risk factors associated with breast cancer development. Therefore, combining the characteristics of normal parenchyma from the contralateral breast with radiomic features of breast tumors may improve the accuracy of digital mammography in the diagnosis of breast cancer. Purpose To determine whether the addition of radiomic analysis of contralateral breast parenchyma to the characterization of breast lesions with digital mammography improves lesion classification over that with radiomic tumor features alone. Materials and Methods This HIPAA-compliant, retrospective study included 182 patients (age range, 25-90 years; mean age, 55.9 years ± 14.9) who underwent mammography between June 2002 and July 2009. There were 106 malignant and 76 benign lesions. Automatic lesion segmentation and radiomic analysis were performed for each breast lesion. Radiomic texture analysis was applied in the normal regions of interest in the contralateral breast parenchyma to assess the mammographic parenchymal patterns. The classification performance of both individual features and the output from a Bayesian artificial neural network classifier was evaluated with the leave-one-patient-out method by using the area under the receiver operating characteristic curve (AUC) as the figure of merit in the task of differentiating between malignant and benign lesions. Results The performance of the combined lesion and parenchyma classifier in the differentiation between malignant and benign mammographic lesions was better than that with the lesion features alone (AUC = 0.84 ± 0.03 vs 0.79 ± 0.03, respectively; P = .047). Overall, six radiomic features-spiculation, margin sharpness, size, circularity from the tumor feature set, and skewness and power law beta from the parenchymal feature set-were selected more than 50% of the time during the feature selection process on the combined feature set. Conclusion Combining quantitative radiomic data from tumors with contralateral parenchyma characterizations may improve diagnostic accuracy for breast cancer. © RSNA, 2019 Online supplemental material is available for this article. See also the editorial by Shaffer in this issue.
Collapse
Affiliation(s)
- Hui Li
- From the Department of Radiology, University of Chicago, 5841 S
Maryland Ave, Chicago, IL 60637
| | - Kayla R. Mendel
- From the Department of Radiology, University of Chicago, 5841 S
Maryland Ave, Chicago, IL 60637
| | - Li Lan
- From the Department of Radiology, University of Chicago, 5841 S
Maryland Ave, Chicago, IL 60637
| | - Deepa Sheth
- From the Department of Radiology, University of Chicago, 5841 S
Maryland Ave, Chicago, IL 60637
| | - Maryellen L. Giger
- From the Department of Radiology, University of Chicago, 5841 S
Maryland Ave, Chicago, IL 60637
| |
Collapse
|
28
|
Gryka MC, Comi TJ, Forsyth RA, Hadley PM, Deb S, Bhargava R. Controlled dissolution of freeform 3D printed carbohydrate glass scaffolds in hydrogels using a hydrophobic spray coating. ADDITIVE MANUFACTURING 2019; 26:193-201. [PMID: 30775269 PMCID: PMC6371974 DOI: 10.1016/j.addma.2018.12.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Freeform 3D printing combined with sacrificial molding promises to lead advances in production of highly complex tubular systems for biomedical applications. Here we leverage a purpose-built isomalt 3D printer to generate complex channel geometries in hydrogels which would be inaccessible with other techniques. To control the dissolution of the scaffold, we propose an enabling technology consisting of an automated nebulizer coating system which applies octadecane to isomalt scaffolds. Octadecane, a saturated hydrocarbon, protects the rigid mold from dissolution and provides ample time for gels to set around the sacrificial structure. With a simplified model of the nebulizer system, the robotic motion was optimized for uniform coating. Using a combination of stimulated Raman scattering (SRS) microscopy and X-ray computed tomography, the coating was characterized to assess surface roughness and consistency. Colorimetric measurements of dissolution rates allowed optimization of sprayer parameters, yielding a decrease in dissolution rates by at least 4 orders of magnitude. High fidelity channels are ensured by surfactant treatment of the coating, which prevents bubbles from clinging to the surface. Spontaneous Raman scattering microspectroscopy and white light microscopy indicate cleared channels are free of octadecane following gentle flushing. The capabilities of the workflow are highlighted with several complex channel architectures including helices, blind channels, and multiple independent channels within polyacrylamide hydrogels of varying stiffnesses.
Collapse
Affiliation(s)
- M C Gryka
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL
| | - T J Comi
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL
| | - R A Forsyth
- Department of Chemical Engineering, University of Illinois at Urbana-Champaign, Urbana, IL
| | - P M Hadley
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL
| | - S Deb
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL
| | - R Bhargava
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL
- Departments of Chemistry, Chemical and Biomolecular Engineering, Mechanical Science and Engineering, and Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL
| |
Collapse
|
29
|
Brock EJ, Ji K, Shah S, Mattingly RR, Sloane BF. In Vitro Models for Studying Invasive Transitions of Ductal Carcinoma In Situ. J Mammary Gland Biol Neoplasia 2019; 24:1-15. [PMID: 30056557 PMCID: PMC6641861 DOI: 10.1007/s10911-018-9405-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 07/13/2018] [Indexed: 12/11/2022] Open
Abstract
About one fourth of all newly identified cases of breast carcinoma are diagnoses of breast ductal carcinoma in situ (DCIS). Since we cannot yet distinguish DCIS cases that would remain indolent from those that may progress to life-threatening invasive ductal carcinoma (IDC), almost all women undergo aggressive treatment. In order to allow for more rational individualized treatment, we and others are developing in vitro models to identify and validate druggable pathways that mediate the transition of DCIS to IDC. These models range from conventional two-dimensional (2D) monolayer cultures on plastic to 3D cultures in natural or synthetic matrices. Some models consist solely of DCIS cells, either cell lines or primary cells. Others are co-cultures that include additional cell types present in the normal or cancerous human breast. The 3D co-culture models more accurately mimic structural and functional changes in breast architecture that accompany the transition of DCIS to IDC. Mechanistic studies of the dynamic and temporal changes associated with this transition are facilitated by adapting the in vitro models to engineered microfluidic platforms. Ultimately, the goal is to create in vitro models that can serve as a reproducible preclinical screen for testing therapeutic strategies that will reduce progression of DCIS to IDC. This review will discuss the in vitro models that are currently available, as well as the progress that has been made using them to understand DCIS pathobiology.
Collapse
MESH Headings
- Breast/pathology
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Intraductal, Noninfiltrating/drug therapy
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Cell Line, Tumor
- Coculture Techniques/methods
- Drug Screening Assays, Antitumor/methods
- Female
- Humans
- Neoplasm Invasiveness/pathology
- Neoplasm Invasiveness/prevention & control
- Primary Cell Culture/methods
Collapse
Affiliation(s)
- Ethan J Brock
- Program in Cancer Biology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Kyungmin Ji
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Seema Shah
- Program in Cancer Biology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Raymond R Mattingly
- Program in Cancer Biology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Bonnie F Sloane
- Program in Cancer Biology, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
- Department of Pharmacology, Wayne State University, 540 E. Canfield, Detroit, MI, 48201, USA.
| |
Collapse
|
30
|
Pallante P, Pisapia P, Bellevicine C, Malapelle U, Troncone G. Circulating Tumour Cells in Predictive Molecular Pathology: Focus on Drug-Sensitive Assays and 3D Culture. Acta Cytol 2019; 63:171-181. [PMID: 30759433 DOI: 10.1159/000496213] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/13/2018] [Indexed: 12/19/2022]
Abstract
Molecular cytopathology is a rapidly evolving field of cytopathology that provides biological information about the response to personalised therapy and about the prognosis of neoplasms diagnosed on cytological samples. Biomarkers such as circulating tumour cells and circulating tumour DNA are increasingly being evaluated in blood and in other body fluids. Such liquid biopsies are non-invasive, repeatable, and feasible also in patients with severe comorbidities. However, liquid biopsy may be challenging due to a low concentration of biomarkers. In such cases, biomarkers can be detected with highly sensitive molecular techniques, which in turn should be validated and integrated in a complex algorithm that includes tissue-based molecular assessments. The aim of this review is to provide the cytopathologist with practical information that is relevant to daily practice, particularly regarding the emerging role of circulating tumour cells in the field of predictive molecular pathology.
Collapse
Affiliation(s)
- Pierlorenzo Pallante
- Institute of Experimental Endocrinology and Oncology (IEOS) "G. Salvatore," National Research Council (CNR), Naples, Italy
| | - Pasquale Pisapia
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Claudio Bellevicine
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy,
| |
Collapse
|
31
|
Eglen RM, Reisine T. Human iPS Cell-Derived Patient Tissues and 3D Cell Culture Part 2: Spheroids, Organoids, and Disease Modeling. SLAS Technol 2019; 24:18-27. [DOI: 10.1177/2472630318803275] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Human induced pluripotent stem cells (HiPSCs) provide several advantages for drug discovery, but principally they provide a source of clinically relevant tissue. Furthermore, the use of HiPSCs cultured in three-dimensional (3D) systems, as opposed to traditional two-dimensional (2D) culture approaches, better represents the complex tissue architecture in vivo. The use of HiPSCs in 3D spheroid and organoid culture is now growing, but particularly when using myocardial, intestinal enteric nervous system, and retinal cell lines. However, organoid cell culture is perhaps making the most notable impact in research and drug discovery, in which 3D neuronal cell cultures allow direct modeling of cortical cell layering and neuronal circuit activity. Given the specific degeneration seen in discrete neuronal circuitry in Alzheimer’s disease (AD) and Parkinson’s disease (PD), HiPSC culture systems are proving to be a major advance. In the present review, the second part of a two-part review, we discuss novel methods in which 3D cell culture systems (principally organoids) are now being used to provide insights into disease mechanisms. (The use of HiPSCs in target identification was reviewed in detail in Part 1.)
Collapse
|
32
|
Abstract
This chapter elaborates on the state-of-the-art experimental procedures utilized in ex-vivo model systems of cancer-bone cell interactions under "static and dynamic" culture conditions and their potential use to understand cellular and molecular mechanisms as well as drug testing and discovery. An additional focus of this chapter is to provide details of how to incorporate varying oxygen tension, viz., hypoxic, normoxic, and hyperoxic, in such studies and regulate the bone biology toward dissociation of the bone remodeling stages to achieve only "bone resorption" or "bone formation" individually.
Collapse
Affiliation(s)
- Erdjan Salih
- Department of Periodontology, Henry M. Goldman School of Dental Medicine, Boston University Medical Center, Boston, MA, USA.
| |
Collapse
|
33
|
Livingston MK, Morgan MM, Daly WT, Murphy WL, Johnson BP, Beebe DJ, Virumbrales-Muñoz M. Evaluation of PEG-based hydrogel influence on estrogen receptor driven responses in MCF7 breast cancer cells. ACS Biomater Sci Eng 2019; 5:6089-6098. [PMID: 31942444 PMCID: PMC6961958 DOI: 10.1021/acsbiomaterials.9b00480] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Extracellular matrix (ECM) mimicking hydrogel scaffolds have greatly improved the physiological relevance of in vitro assays, but introduce another dimension that creates variability in cell related readouts when compared to traditional 2D cells-on-plastic assays. We have developed a synthetic poly(ethylene glycol) (PEG) based ECM mimicking hydrogel and tested it against two gold standard animal-based naturally derived hydrogel scaffolds in MCF7 cell response. We have used the percent coefficient of variation (CV) as a metric to evaluate the reproducibility of said responses. Results indicated that PEG hydrogels performed similarly to naturally derived gold standards, and variance was similar in basic characterization assays, such as viability and cell adherence. PEG based hydrogels had lower CV values in estrogen receptor driven responses to several doses of estrogen in both estrogen receptor transactivation and estrogen induced proliferation.
Collapse
Affiliation(s)
- Megan K. Livingston
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, WI 53706
- School of Medicine and Public Health, University of Wisconsin-Madison, 750 Highland Ave, Madison, WI 53726
| | - Molly M. Morgan
- School of Medicine and Public Health, University of Wisconsin-Madison, 750 Highland Ave, Madison, WI 53726
| | - William T. Daly
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 55705
| | - William L. Murphy
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 55705
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1550 Engineering Dr, Madison, WI 53706
- Department of Materials Science and Engineering, University of Wisconsin-Madison, 1509 University Ave, Madison, WI 53706
| | - Brian P. Johnson
- School of Medicine and Public Health, University of Wisconsin-Madison, 750 Highland Ave, Madison, WI 53726
| | - David J. Beebe
- School of Medicine and Public Health, University of Wisconsin-Madison, 750 Highland Ave, Madison, WI 53726
| | - Maria Virumbrales-Muñoz
- School of Medicine and Public Health, University of Wisconsin-Madison, 750 Highland Ave, Madison, WI 53726
| |
Collapse
|
34
|
Kwak BS, Choi W, Jeon JW, Won JI, Sung GY, Kim B, Sung JH. In vitro 3D skin model using gelatin methacrylate hydrogel. J IND ENG CHEM 2018. [DOI: 10.1016/j.jiec.2018.05.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
35
|
Saleem J, Wang L, Chen C. Carbon-Based Nanomaterials for Cancer Therapy via Targeting Tumor Microenvironment. Adv Healthc Mater 2018; 7:e1800525. [PMID: 30073803 DOI: 10.1002/adhm.201800525] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/04/2018] [Indexed: 12/12/2022]
Abstract
Cancer remains one of the major health problems all over the world and conventional therapeutic approaches have failed to attain an effective cure. Tumor microenvironments (TME) present a unique challenge in tumor therapy due to their complex structures and multiple components, which also serve as the soil for tumor growth, development, invasion, and migration. The complex TME includes immune cells, fibrous collagen structures, and tortuous blood vessels, in which conventional therapeutic approaches are rendered useless. State-of-the-art nanotechnologies have potential to cope with the threats of malignant tumors. With unique physiochemical properties, carbon nanomaterials (CNMs), including graphene, fullerenes, carbon nanotubes, and carbon quantum dots, offer opportunities to resolve the hurdles, by targeting not only cancer cells but also the TME. This review summarizes the progress about CNM-based cancer therapy strategies, which mainly focuses on both the treatment for cancer cells and TME-targeted modulation. In the last, the challenges for TME-based therapy via CNMs are discussed, which will be important in guiding current basic research to clinical translation in the future.
Collapse
Affiliation(s)
- Jabran Saleem
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience; National Center for Nanoscience and Technology of China; Beijing 100190 P. R. China
| | - Liming Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety; Institute of High Energy Physics; Chinese Academy of Sciences; Beijing 100049 P. R. China
| | - Chunying Chen
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience; National Center for Nanoscience and Technology of China; Beijing 100190 P. R. China
| |
Collapse
|
36
|
Luminal MCF-12A & myoepithelial-like Hs 578Bst cells form bilayered acini similar to human breast. Future Sci OA 2018; 4:FSO315. [PMID: 30112185 PMCID: PMC6088263 DOI: 10.4155/fsoa-2018-0010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/08/2018] [Indexed: 12/21/2022] Open
Abstract
The epithelium's functional unit is the bilayered acinus, made of a layer of luminal cells, surrounded by a layer of basal cells mainly composed of myoepithelial cells. Aim The aim of this study was to develop a reproducible and manipulable 3D co-culture model of the bilayered acinus in vitro to study the interactions between the two layers. Materials & methods Two different combinations of cell lines were co-cultured in Matrigel: SCp2 and SCg6 mice cells, or MCF-12A and Hs 578Bst human cell lines. Results Confocal microscopy analysis showed that only MCF-12A and Hs 578Bst cells could form some bilayered acini. This in vitro bilayered acini model will allow us to understand the role of interactions between luminal and myoepithelial cells in the normal breast development.
Collapse
|
37
|
Lim SH, Kathuria H, Tan JJY, Kang L. 3D printed drug delivery and testing systems - a passing fad or the future? Adv Drug Deliv Rev 2018; 132:139-168. [PMID: 29778901 DOI: 10.1016/j.addr.2018.05.006] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 04/12/2018] [Accepted: 05/12/2018] [Indexed: 12/23/2022]
Abstract
The US Food and Drug Administration approval of the first 3D printed tablet in 2015 has ignited growing interest in 3D printing, or additive manufacturing (AM), for drug delivery and testing systems. Beyond just a novel method for rapid prototyping, AM provides key advantages over traditional manufacturing of drug delivery and testing systems. These includes the ability to fabricate complex geometries to achieve variable drug release kinetics; ease of personalising pharmacotherapy for patient and lowering the cost for fabricating personalised dosages. Furthermore, AM allows fabrication of complex and micron-sized tissue scaffolds and models for drug testing systems that closely resemble in vivo conditions. However, there are several limitations such as regulatory concerns that may impede the progression to market. Here, we provide an overview of the advantages of AM drug delivery and testing, as compared to traditional manufacturing techniques. Also, we discuss the key challenges and future directions for AM enabled pharmaceutical applications.
Collapse
Affiliation(s)
- Seng Han Lim
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Block S4A, Level 3, 117543, Singapore
| | - Himanshu Kathuria
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Block S4A, Level 3, 117543, Singapore
| | - Justin Jia Yao Tan
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Block S4A, Level 3, 117543, Singapore
| | - Lifeng Kang
- School of Pharmacy, University of Sydney, Pharmacy and Bank Building A15, NSW 2006, Australia.
| |
Collapse
|
38
|
Liu Z, Speroni L, Quinn KP, Alonzo C, Pouli D, Zhang Y, Stuntz E, Sonnenschein C, Soto AM, Georgakoudi I. 3D organizational mapping of collagen fibers elucidates matrix remodeling in a hormone-sensitive 3D breast tissue model. Biomaterials 2018; 179:96-108. [PMID: 29980078 DOI: 10.1016/j.biomaterials.2018.06.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 06/08/2018] [Accepted: 06/22/2018] [Indexed: 12/11/2022]
Abstract
Hormones play an important role in normal and diseased breast tissue development. However, they can also disrupt cell-matrix interactions and their role in extracellular matrix reorganization during epithelial morphogenesis remains poorly understood, partly due to a lack of sensitive approaches for matrix characterization. Here, we assess the hormonal regulation of matrix reorganization in a three-dimensional (3D) breast tissue culture model using a novel metric, i.e., 3D directional variance, to characterize the 3D organization of collagen fibers visualized via high-resolution, second harmonic generation imaging. This metric enables resolving and quantifying patterns of spatial organization throughout the matrix surrounding epithelial structures treated with 17β-estradiol (E2) alone, and E2 in combination with either promegestone, a progestogen, or prolactin. Addition of promegestone results in the most disorganized fibers, while the E2 alone treatment leads to the most organized ones. Location-dependent organization mapping indicates that only the prolactin treatment leads to significant heterogeneities in the regional organization of collagen fibers, with higher levels of alignment observed at the end of the elongated epithelial structures. The observed collagen organization patterns for all groups persist for tens of micrometers. In addition, a comparison between 3D directional variance and typical 2D analysis approaches reveals an improved sensitivity of the 3D metric to identify organizational heterogeneities and differences among treatment groups. These results demonstrate that 3D directional variance is sensitive to subtle changes in the extracellular micro-environment and has the potential to elucidate reciprocal cell-matrix interactions in the context of numerous applications involving the study of normal and diseased tissue morphogenesis.
Collapse
Affiliation(s)
- Zhiyi Liu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA; Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Lucia Speroni
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Kyle P Quinn
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA; Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, USA
| | - Carlo Alonzo
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Dimitra Pouli
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Yang Zhang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Emily Stuntz
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Carlos Sonnenschein
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Ana M Soto
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
39
|
Zhu W, Cui H, Boualam B, Masood F, Flynn E, Rao RD, Zhang ZY, Zhang LG. 3D bioprinting mesenchymal stem cell-laden construct with core-shell nanospheres for cartilage tissue engineering. NANOTECHNOLOGY 2018; 29:185101. [PMID: 29446757 DOI: 10.1088/1361-6528/aaafa1] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Cartilage tissue is prone to degradation and has little capacity for self-healing due to its avascularity. Tissue engineering, which provides artificial scaffolds to repair injured tissues, is a novel and promising strategy for cartilage repair. 3D bioprinting offers even greater potential for repairing degenerative tissue by simultaneously integrating living cells, biomaterials, and biological cues to provide a customized scaffold. With regard to cell selection, mesenchymal stem cells (MSCs) hold great capacity for differentiating into a variety of cell types, including chondrocytes, and could therefore be utilized as a cartilage cell source in 3D bioprinting. In the present study, we utilize a tabletop stereolithography-based 3D bioprinter for a novel cell-laden cartilage tissue construct fabrication. Printable resin is composed of 10% gelatin methacrylate (GelMA) base, various concentrations of polyethylene glycol diacrylate (PEGDA), biocompatible photoinitiator, and transforming growth factor beta 1 (TGF-β1) embedded nanospheres fabricated via a core-shell electrospraying technique. We find that the addition of PEGDA into GelMA hydrogel greatly improves the printing resolution. Compressive testing shows that modulus of the bioprinted scaffolds proportionally increases with the concentrations of PEGDA, while swelling ratio decreases with the increase of PEGDA concentration. Confocal microscopy images illustrate that the cells and nanospheres are evenly distributed throughout the entire bioprinted construct. Cells grown on 5%/10% (PEGDA/GelMA) hydrogel present the highest cell viability and proliferation rate. The TGF-β1 embedded in nanospheres can keep a sustained release up to 21 d and improve chondrogenic differentiation of encapsulated MSCs. The cell-laden bioprinted cartilage constructs with TGF-β1-containing nanospheres is a promising strategy for cartilage regeneration.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington DC 20052, United States of America
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Contribution of three-dimensional architecture and tumor-associated fibroblasts to hepcidin regulation in breast cancer. Oncogene 2018; 37:4013-4032. [PMID: 29695834 PMCID: PMC6054540 DOI: 10.1038/s41388-018-0243-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 02/02/2018] [Accepted: 03/06/2018] [Indexed: 01/08/2023]
Abstract
Hepcidin is a peptide hormone that negatively regulates iron efflux and plays an important role in controlling the growth of breast tumors. In patients with breast cancer, the combined expression of hepcidin and its membrane target, ferroportin, predict disease outcome. However, mechanisms that control hepcidin expression in breast cancer cells remain largely unknown. Here we use three-dimensional breast cancer spheroids derived from cell lines and breast cancer patients to probe mechanisms of hepcidin regulation in breast cancer. We observe that the extent of hepcidin induction and pathways of its regulation are markedly changed in breast cancer cells grown in three dimensions. In monolayer culture, BMPs, particularly BMP6, regulate hepcidin transcription. When breast cancer cells are grown as spheroids, there is a >10 fold induction in hepcidin transcripts. Microarray analysis combined with knockdown experiments reveal that GDF-15 is the primary mediator of this change. The increase in hepcidin as breast cells develop a three-dimensional architecture increases intracellular iron, as indicated by an increase in the iron storage protein ferritin. Immunohistochemical staining of human breast tumors confirms that both GDF-15 and hepcidin are expressed in breast cancer specimens. Further, levels of GDF-15 are significantly correlated with levels of hepcidin at both the mRNA and protein level in patient samples, consistent with a role for GDF-15 in control of hepcidin in human breast tumors. Inclusion of tumor-associated fibroblasts in breast cancer spheroids further induces hepcidin. This induction is mediated by fibroblast-dependent secretion of IL-6. Breast cancer cells grown as spheroids are uniquely receptive to IL-6-dependent induction of hepcidin by tumor-associated fibroblasts, since IL-6 does not induce hepcidin in cells grown as monolayers. Collectively, our results suggest a new paradigm for tumor-mediated control of iron through the control of hepcidin by tumor architecture and the breast tumor microenvironment.
Collapse
|
41
|
Karimpoor M, Yebra-Fernandez E, Parhizkar M, Orlu M, Craig D, Khorashad JS, Edirisinghe M. Alginate foam-based three-dimensional culture to investigate drug sensitivity in primary leukaemia cells. J R Soc Interface 2018; 15:20170928. [PMID: 29695605 PMCID: PMC5938583 DOI: 10.1098/rsif.2017.0928] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 03/28/2018] [Indexed: 01/14/2023] Open
Abstract
The development of assays for evaluating the sensitivity of leukaemia cells to anti-cancer agents is becoming an important aspect of personalized medicine. Conventional cell cultures lack the three-dimensional (3D) structure of the bone marrow (BM), the extracellular matrix and stromal components which are crucial for the growth and survival of leukaemia stem cells. To accurately predict the sensitivity of the leukaemia cells in an in vitro assay a culturing system containing the essential components of BM is required. In this study, we developed a porous calcium alginate foam-based scaffold to be used for 3D culture. The new 3D culture was shown to be cell compatible as it supported the proliferation of both normal haematopoietic and leukaemia cells. Our cell differential assay for myeloid markers showed that the porous foam-based 3D culture enhanced myeloid differentiation in both leukaemia and normal haematopoietic cells compared to two-dimensional culture. The foam-based scaffold reduced the sensitivity of the leukaemia cells to the tested antileukaemia agents in K562 and HL60 leukaemia cell line model and also primary myeloid leukaemia cells. This observation supports the application of calcium alginate foams as scaffold components of the 3D cultures for investigation of sensitivity to antileukaemia agents in primary myeloid cells.
Collapse
Affiliation(s)
- Mahroo Karimpoor
- Department of Mechanical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
- Centre for Haematology, Department of Medicine, Imperial College, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Eva Yebra-Fernandez
- Molecular Pathology, North West London Pathology, Hammersmith Hospital, London, W12 0HS, UK
| | - Maryam Parhizkar
- Department of Mechanical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
| | - Mine Orlu
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Duncan Craig
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Jamshid S Khorashad
- Centre for Haematology, Department of Medicine, Imperial College, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
- Molecular Pathology, North West London Pathology, Hammersmith Hospital, London, W12 0HS, UK
| | - Mohan Edirisinghe
- Department of Mechanical Engineering, University College London, Torrington Place, London WC1E 7JE, UK
| |
Collapse
|
42
|
Molla A, Couvet M, Coll JL. Unsuccessful mitosis in multicellular tumour spheroids. Oncotarget 2018; 8:28769-28784. [PMID: 28430635 PMCID: PMC5438690 DOI: 10.18632/oncotarget.15673] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 02/07/2017] [Indexed: 11/25/2022] Open
Abstract
Multicellular spheroids are very attractive models in oncology because they mimic the 3D organization of the tumour cells with their microenvironment. We show here using 3 different cell types (mammary TSA/pc, embryonic kidney Hek293 and cervical cancer HeLa), that when the cells are growing as spheroids the frequency of binucleated cells is augmented as occurs in some human tumours. We therefore describe mitosis in multicellular spheroids by following mitotic markers and by time-lapse experiments. Chromosomes alignment appears to be correct on the metaphasic plate and the passenger complex is well localized on centromere. Moreover aurora kinases are fully active and histone H3 is phosphorylated on Ser 10. Consequently, the mitotic spindle checkpoint is satisfied and, anaphase proceeds as illustrated by the transfer of survivin on the spindle and by the segregation of the two lots of chromosomes. However, the segregation plane is not well defined and oscillations of the dividing cells are observed. Finally, cytokinesis fails and the absence of separation of the two daughter cells gives rise to binucleated cells. Division orientation is specified during interphase and persists throughout mitosis. Our data indicate that the cancer cells, in multicellular spheroids, lose their ability to regulate their orientation, a feature commonly encountered in tumours. Moreover, multicellular spheroid expansion is still sensitive to mitotic drugs as pactlitaxel and aurora kinase inhibitors. The spheroids thus represent a highly relevant model for studying drug efficiency in tumours.
Collapse
Affiliation(s)
- Annie Molla
- Institute for Advance Biosciences, Centre de recherche UGA, INSERM U1209, CNRS UMR 5309, 38700 La Tronche, France
| | - Morgane Couvet
- Institute for Advance Biosciences, Centre de recherche UGA, INSERM U1209, CNRS UMR 5309, 38700 La Tronche, France
| | - Jean-Luc Coll
- Institute for Advance Biosciences, Centre de recherche UGA, INSERM U1209, CNRS UMR 5309, 38700 La Tronche, France
| |
Collapse
|
43
|
Watson SS, Dane M, Chin K, Tatarova Z, Liu M, Liby T, Thompson W, Smith R, Nederlof M, Bucher E, Kilburn D, Whitman M, Sudar D, Mills GB, Heiser LM, Jonas O, Gray JW, Korkola JE. Microenvironment-Mediated Mechanisms of Resistance to HER2 Inhibitors Differ between HER2+ Breast Cancer Subtypes. Cell Syst 2018; 6:329-342.e6. [PMID: 29550255 PMCID: PMC5927625 DOI: 10.1016/j.cels.2018.02.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 08/16/2017] [Accepted: 02/02/2018] [Indexed: 01/19/2023]
Abstract
Extrinsic signals are implicated in breast cancer resistance to HER2-targeted tyrosine kinase inhibitors (TKIs). To examine how microenvironmental signals influence resistance, we monitored TKI-treated breast cancer cell lines grown on microenvironment microarrays composed of printed extracellular matrix proteins supplemented with soluble proteins. We tested ~2,500 combinations of 56 soluble and 46 matrix microenvironmental proteins on basal-like HER2+ (HER2E) or luminal-like HER2+ (L-HER2+) cells treated with the TKIs lapatinib or neratinib. In HER2E cells, hepatocyte growth factor, a ligand for MET, induced resistance that could be reversed with crizotinib, an inhibitor of MET. In L-HER2+ cells, neuregulin1-β1 (NRG1β), a ligand for HER3, induced resistance that could be reversed with pertuzumab, an inhibitor of HER2-HER3 heterodimerization. The subtype-specific responses were also observed in 3D cultures and murine xenografts. These results, along with bioinformatic pathway analysis and siRNA knockdown experiments, suggest different mechanisms of resistance specific to each HER2+ subtype: MET signaling for HER2E and HER2-HER3 heterodimerization for L-HER2+ cells.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Breast Neoplasms/drug therapy
- Cell Line, Tumor
- Databases, Genetic
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Enzyme Inhibitors/pharmacology
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Genes, erbB-2/drug effects
- Genes, erbB-2/genetics
- Genes, erbB-2/physiology
- High-Throughput Screening Assays/methods
- Humans
- Lapatinib/pharmacology
- MCF-7 Cells
- Mice
- Protein Kinase Inhibitors/pharmacology
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Proto-Oncogene Proteins c-met/antagonists & inhibitors
- Quinazolines/pharmacology
- Quinolines/pharmacology
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-3/antagonists & inhibitors
- Signal Transduction/drug effects
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- Tumor Microenvironment/physiology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Spencer S Watson
- Department of Biomedical Engineering, Knight Cancer Institute, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Mark Dane
- Department of Biomedical Engineering, Knight Cancer Institute, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Koei Chin
- Department of Biomedical Engineering, Knight Cancer Institute, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Zuzana Tatarova
- Department of Biomedical Engineering, Knight Cancer Institute, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Moqing Liu
- Department of Biomedical Engineering, Knight Cancer Institute, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Tiera Liby
- Department of Biomedical Engineering, Knight Cancer Institute, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Wallace Thompson
- Department of Biomedical Engineering, Knight Cancer Institute, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Rebecca Smith
- Department of Biomedical Engineering, Knight Cancer Institute, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Michel Nederlof
- Department of Biomedical Engineering, Knight Cancer Institute, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Quantitative Imaging Systems LLC, 1410 NW Kearney Street, #1114, Portland, OR 97209, USA
| | - Elmar Bucher
- Department of Biomedical Engineering, Knight Cancer Institute, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - David Kilburn
- Department of Biomedical Engineering, Knight Cancer Institute, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Matthew Whitman
- Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115, USA
| | - Damir Sudar
- Department of Biomedical Engineering, Knight Cancer Institute, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA; Quantitative Imaging Systems LLC, 1410 NW Kearney Street, #1114, Portland, OR 97209, USA
| | - Gordon B Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Laura M Heiser
- Department of Biomedical Engineering, Knight Cancer Institute, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Oliver Jonas
- Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115, USA
| | - Joe W Gray
- Department of Biomedical Engineering, Knight Cancer Institute, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| | - James E Korkola
- Department of Biomedical Engineering, Knight Cancer Institute, OHSU Center for Spatial Systems Biomedicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
44
|
Murekatete B, Shokoohmand A, McGovern J, Mohanty L, Meinert C, Hollier BG, Zippelius A, Upton Z, Kashyap AS. Targeting Insulin-Like Growth Factor-I and Extracellular Matrix Interactions in Melanoma Progression. Sci Rep 2018; 8:583. [PMID: 29330502 PMCID: PMC5766529 DOI: 10.1038/s41598-017-19073-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 12/20/2017] [Indexed: 01/21/2023] Open
Abstract
Insulin-like growth factor (IGF)-I binds to the ECM protein vitronectin (VN) through IGF binding proteins (IGFBPs) to enhance proliferation and migration of skin keratinocytes and fibroblasts. Although evidence exists for the role of individual components of the complex (IGF-I, IGFBP-3 and VN), the cellular functions stimulated by these proteins together as a complex remains un-investigated in melanoma cells. We report here that the IGF-I:IGFBP-3:VN trimeric complex stimulates a dose-dependent increase in the proliferation and migration of WM35 and Sk-MEL28 melanoma cells. In 3D Matrigel™ and hydrogel cultures, both cell lines formed primary tumor-like spheroids, which increased in size in a dose-dependent manner in response to the trimeric complex. Furthermore, we reveal IGFBP-3:VN protein complexes in malignant melanoma and squamous cell carcinoma patient tissues, where the IGFBP-3:VN complex was seen to be predominantly tumor cell-associated. Peptide antagonists designed to target the binding of IGF-I:IGFBP-3 to VN were demonstrated to inhibit IGF-I:IGFBP-3:VN-stimulated cell migration, invasion and 3D tumor cell growth of melanoma cells. Overall, this study provides new data on IGF:ECM interactions in skin malignancies and demonstrates the potential usefulness of a growth factor:ECM-disrupting strategy for abrogating tumor progression.
Collapse
Affiliation(s)
- Berline Murekatete
- Institute of Health and Biomedical Innovation, School of Biomedical Science, Queensland University of Technology, Brisbane, QLD, Australia
| | - Ali Shokoohmand
- Institute of Health and Biomedical Innovation, School of Biomedical Science, Queensland University of Technology, Brisbane, QLD, Australia.,Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Brisbane, Queensland, Australia
| | - Jacqui McGovern
- Institute of Health and Biomedical Innovation, School of Biomedical Science, Queensland University of Technology, Brisbane, QLD, Australia
| | - Lipsa Mohanty
- Institute of Health and Biomedical Innovation, School of Biomedical Science, Queensland University of Technology, Brisbane, QLD, Australia
| | - Christoph Meinert
- Institute of Health and Biomedical Innovation, School of Biomedical Science, Queensland University of Technology, Brisbane, QLD, Australia
| | - Brett G Hollier
- Australian Prostate Cancer Research Centre - Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Brisbane, Queensland, Australia
| | - Alfred Zippelius
- Cancer Immunology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Zee Upton
- Institute of Health and Biomedical Innovation, School of Biomedical Science, Queensland University of Technology, Brisbane, QLD, Australia.,Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Abhishek S Kashyap
- Institute of Health and Biomedical Innovation, School of Biomedical Science, Queensland University of Technology, Brisbane, QLD, Australia. .,Cancer Immunology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland.
| |
Collapse
|
45
|
Ionizing radiation-mediated premature senescence and paracrine interactions with cancer cells enhance the expression of syndecan 1 in human breast stromal fibroblasts: the role of TGF-β. Aging (Albany NY) 2017; 8:1650-69. [PMID: 27434331 PMCID: PMC5032688 DOI: 10.18632/aging.100989] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/26/2016] [Indexed: 12/12/2022]
Abstract
The cell surface proteoglycan syndecan 1 (SDC1) is overexpressed in the malignant breast stromal fibroblasts, creating a favorable milieu for tumor cell growth. In the present study, we found that ionizing radiation, a well-established treatment in human breast cancer, provokes premature senescence of human breast stromal fibroblasts in vitro, as well as in the breast tissue in vivo. These senescent cells were found to overexpress SDC1 both in vitro and in vivo. By using a series of specific inhibitors and siRNA approaches, we showed that this SDC1 overexpression in senescent cells is the result of an autocrine action of Transforming Growth Factor-β (TGF-β) through the Smad pathway and the transcription factor Sp1, while the classical senescence pathways of p53 or p38 MAPK - NF-kB are not involved. In addition, the highly invasive human breast cancer cells MDA-MB-231 (in contrast to the low-invasive MCF-7) can also enhance SDC1 expression, both in early-passage and senescent fibroblasts via a paracrine action of TGF-β. The above suggest that radiation-mediated premature senescence and invasive tumor cells, alone or in combination, enhance SDC1 expression in breast stromal fibroblasts, a poor prognostic factor for cancer growth, and that TGF-β plays a crucial role in this process.
Collapse
|
46
|
Eritja N, Yeramian A, Chen BJ, Llobet-Navas D, Ortega E, Colas E, Abal M, Dolcet X, Reventos J, Matias-Guiu X. Endometrial Carcinoma: Specific Targeted Pathways. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 943:149-207. [PMID: 27910068 DOI: 10.1007/978-3-319-43139-0_6] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Endometrial cancer (EC) is the most common gynecologic malignancy in the western world with more than 280,000 cases per year worldwide. Prognosis for EC at early stages, when primary surgical resection is the most common initial treatment, is excellent. Five-year survival rate is around 70 %.Several molecular alterations have been described in the different types of EC. They occur in genes involved in important signaling pathways. In this chapter, we will review the most relevant altered pathways in EC, including PI3K/AKT/mTOR, RAS-RAF-MEK-ERK, Tyrosine kinase, WNT/β-Catenin, cell cycle, and TGF-β signaling pathways. At the end of the chapter, the most significant clinical trials will be briefly discussed.This information is important to identify specific targets for therapy.
Collapse
Affiliation(s)
- Nuria Eritja
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
- GEICEN Research Group, Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
| | - Andree Yeramian
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
- GEICEN Research Group, Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
| | - Bo-Juen Chen
- New York Genome Center, New York, NY, 10013, USA
| | - David Llobet-Navas
- Institute of Genetic Medicine, Newcastle University, Newcastle-Upon-Tyne, NE1 3BZ, UK
| | - Eugenia Ortega
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
| | - Eva Colas
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
- GEICEN Research Group, Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
- Research Unit in Biomedicine and Translational and Pediatric Oncology, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Miguel Abal
- GEICEN Research Group, Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
- Translational Medical Oncology, Health Research Institute of Santiago (IDIS), Santiago de Compostela, Spain
| | - Xavier Dolcet
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
- GEICEN Research Group, Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
| | - Jaume Reventos
- GEICEN Research Group, Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain
- Research Unit in Biomedicine and Translational and Pediatric Oncology, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Xavier Matias-Guiu
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain.
- GEICEN Research Group, Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Av Rovira Roure, 80, 25198, Lleida, Spain.
| |
Collapse
|
47
|
Tumor cells and their crosstalk with endothelial cells in 3D spheroids. Sci Rep 2017; 7:10428. [PMID: 28874803 PMCID: PMC5585367 DOI: 10.1038/s41598-017-10699-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 08/14/2017] [Indexed: 12/18/2022] Open
Abstract
Recapitulating the tumor microenvironment is a central challenge in the development of experimental model for cancer. To provide a reliable tool for drug development and for personalized cancer therapy, it is critical to maintain key features that exist in the original tumor. Along with this effort, 3-dimentional (3D) cellular models are being extensively studied. Spheroids are self-assembled cell aggregates that possess many important components of the physiological spatial growth and cell-cell interactions. In this study we aimed to investigate the interconnection between tumor and endothelial cells (EC) in hybrid spheroids containing either tumor cell (TC) lines or patient derived cancer cells. Preparation protocols of hybrid spheroids were optimized and their morphology and tissue-like features were analyzed. Our finding show that capillary-like structures are formed upon assembly and growth of TC:EC spheroids and that spheroids’ shape and surface texture may be an indication of spatial invasiveness of cells in the extra-cellular matrix (ECM). Establishing a model of hybrid tumor/stroma spheroids has a crucial importance in the experimental approach for personalized medicine, and may offer a reliable and low-cost method for the goal of predicting drug effects.
Collapse
|
48
|
Muraro MG, Muenst S, Mele V, Quagliata L, Iezzi G, Tzankov A, Weber WP, Spagnoli GC, Soysal SD. Ex-vivo assessment of drug response on breast cancer primary tissue with preserved microenvironments. Oncoimmunology 2017; 6:e1331798. [PMID: 28811974 DOI: 10.1080/2162402x.2017.1331798] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 12/31/2022] Open
Abstract
Interaction between cancerous, non-transformed cells, and non-cellular components within the tumor microenvironment plays a key role in response to treatment. However, short-term culture or xenotransplantation of cancer specimens in immunodeficient animals results in dramatic modifications of the tumor microenvironment, thus preventing reliable assessment of compounds or biologicals of potential therapeutic relevance. We used a perfusion-based bioreactor developed for tissue engineering purposes to successfully maintain the tumor microenvironment of freshly excised breast cancer tissue obtained from 27 breast cancer patients and used this platform to test the therapeutic effect of antiestrogens as well as checkpoint-inhibitors on the cancer cells. Viability and functions of tumor and immune cells could be maintained for over 2 weeks in perfused bioreactors. Next generation sequencing authenticated cultured tissue specimens as closely matching the original clinical samples. Anti-estrogen treatment of cultured estrogen receptor positive breast cancer tissue as well as administration of pertuzumab to a Her2 positive breast cancer both had an anti-proliferative effect. Treatment with anti-programmed-death-Ligand (PD-L)-1 and anti-cytotoxic T lymphocyte-associated protein (CTLA)-4 antibodies lead to immune activation, evidenced by increased lymphocyte proliferation, increased expression of IFNγ, and decreased expression of IL10, accompanied by a massive cancer cell death in ex vivo triple negative breast cancer specimens. In the era of personalized medicine, the ex vivo culture of breast cancer tissue represents a promising approach for the pre-clinical evaluation of conventional and immune-mediated treatments and provides a platform for testing of innovative treatments.
Collapse
Affiliation(s)
- Manuele G Muraro
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Simone Muenst
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Valentina Mele
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Luca Quagliata
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Giandomenica Iezzi
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Alexandar Tzankov
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Walter P Weber
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Giulio C Spagnoli
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Savas D Soysal
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland.,Department of Surgery, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
49
|
Inhibition of extracellular matrix mediated TGF-β signalling suppresses endometrial cancer metastasis. Oncotarget 2017; 8:71400-71417. [PMID: 29069715 PMCID: PMC5641058 DOI: 10.18632/oncotarget.18069] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 05/07/2017] [Indexed: 01/06/2023] Open
Abstract
Although aggressive invasion and distant metastases are an important cause of morbidity and mortality in patients with endometrial cancer (EC), the requisite events determining this propensity are currently unknown. Using organotypic three-dimensional culture of endometrial cancer cell lines, we demonstrated anti-correlated TGF-β signalling gene expression patterns that arise among extracellular matrix (ECM)-attached cells. TGF-β pathway seemed to be active in EC cells forming non-glandular colonies in 3D-matrix but weaker in glandular colonies. Functionally we found that out of several ECM proteins, fibronectin relatively promotes Smad phosphorylation suggesting a potential role in regulating TGF-β signalling in non-glandular colonies. Importantly, alteration of TGF-β pathway induced EMT and MET in both type of colonies through slug protein. The results exemplify a crucial role of TGF-β pathway during EC metastasis in human patients and inhibition of the pathway in a murine model impaired tumour cell invasion and metastasis depicting an attractive target for therapeutic intervention of malignant tumour progression. These findings provide key insights into the role of ECM-derived TGF-β signalling to promote endometrial cancer metastasis and offer an avenue for therapeutic targeting of microenvironment derived signals along with tumour cells.
Collapse
|
50
|
Wang L, Yu X, Wang C, Pan S, Liang B, Zhang Y, Chong X, Meng Y, Dong J, Zhao Y, Yang Y, Wang H, Gao J, Wei H, Zhao J, Wang H, Hu C, Xiao W, Li B. The anti-ErbB2 antibody H2-18 and the pan-PI3K inhibitor GDC-0941 effectively inhibit trastuzumab-resistant ErbB2-overexpressing breast cancer. Oncotarget 2017; 8:52877-52888. [PMID: 28881779 PMCID: PMC5581078 DOI: 10.18632/oncotarget.17907] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 05/05/2017] [Indexed: 11/25/2022] Open
Abstract
Trastuzumab, an anti-ErbB2 humanized antibody, brings benefit to patients with ErbB2-amplified metastatic breast cancers. However, the resistance to trastuzumab is common. Our previously reported H2-18, an anti-ErbB2 antibody, potently induced programmed cell death in trastuzumab-resistant breast cancer cells. Here, we aim to investigate the antitumor efficacy of H2-18 in combination with the pan-PI3K inhibitor GDC-0941 in trastuzumab-resistant breast cancer cell lines. The results showed that H2-18 and GDC-0941 synergistically inhibited the in vitro proliferation of BT-474, SKBR-3, HCC-1954 and HCC-1419 breast cancer cells. H2-18 plus GDC-0941 showed significantly enhanced programmed cell death-inducing activity compared with each drug used alone. The combination of H2-18 and GDC-0941 did not increase the effect of single agent on ROS production, cell cycle and ErbB2 signaling. Importantly, the in vivo antitumor efficacy of H2-18 plus GDC-0941 was superior to that of single agent. Thus, the enhanced in vivo antitumor efficacy of H2-18 plus GDC-0941 may mainly be attributable to its increased programmed cell death-inducing activity. Collectively, H2-18 plus GDC-0941 could effectively inhibit tumor growth, suggesting the potential to be translated into clinic as an efficient strategy for ErbB2-overexpressing breast cancers.
Collapse
Affiliation(s)
- Lingfei Wang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.,International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Xiaojie Yu
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Chao Wang
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Shujun Pan
- Hangzhou Sanatorium of People's Liberation Army, Hangzhou 310007, China
| | - Beibei Liang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Yajun Zhang
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Xiaodan Chong
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Yanchun Meng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Jian Dong
- Department of Vascular Surgery, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Yirong Zhao
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Yang Yang
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Huajing Wang
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Jie Gao
- Department of Pharmaceutical Sciences, Second Military Medical University, Shanghai 200433, China
| | - Huafeng Wei
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.,International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Jian Zhao
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.,International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Hao Wang
- International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| | - Chaohua Hu
- Department of General Surgery, Xiaogan Central Hospital Affiliated to Wuhan University of Science and Technology, Wuhan 432000, China
| | - Wenze Xiao
- Department of Rheumatology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Bohua Li
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.,International Joint Cancer Institute, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|