73 results on '"Loessner, D."'
Search Results
2. KALLIKREIN-RELATED PEPTIDASE 4 IS A KEY REGULATOR OF THE PROSTATE CANCER MICROENVIRONMENT: P03-10
- Author
-
Fuhrman-Luck, R, Kleifeld, O, Ho, B, Hastie, M, Stoll, T, Stephens, C, Loessner, D, Stansfield, S, Gorman, J, and Clements, J A
- Published
- 2013
3. SpheroidSim—Preliminary evaluation of a new computational tool to predict the influence of cell cycle time and phase fraction on spheroid growth.
- Author
-
Little, J. P., Pettet, G. J., Hutmacher, D. W., and Loessner, D.
- Subjects
CELL cycle ,CELL proliferation ,CANCER cells ,GENE expression ,GROWTH factors - Abstract
Background: There is a relative paucity of research that integrates materials science and bioengineering with computational simulations to decipher the intricate processes promoting cancer progression. Therefore, a first‐generation computational model, SpheroidSim, was developed that includes a biological data set derived from a bioengineered spheroid model to obtain a quantitative description of cell kinetics. Results: SpheroidSim is a 3D agent‐based model simulating the growth of multicellular cancer spheroids. Cell cycle time and phases mathematically motivated the population growth. SpheroidSim simulated the growth dynamics of multiple spheroids by individually defining a collection of specific phenotypic traits and characteristics for each cell. Experimental data derived from a hydrogel‐based spheroid model were fit to the predictions providing insight into the influence of cell cycle time (CCT) and cell phase fraction (CPF) on the cell population. A comparison of the number of active cells predicted for each analysis showed that the value and method used to define CCT had a greater effect on the predicted cell population than CPF. The model predictions were similar to the experimental results for the number of cells, with the predicted total number of cells varying by 8% and 12%, respectively, compared to the experimental data. Conclusions: SpheroidSim is a first step in developing a biologically based predictive tool capable of revealing fundamental elements in cancer cell physiology. This computational model may be applied to study the effect of the microenvironment on spheroid growth and other cancer cell types that demonstrate a similar multicellular clustering behavior as the population develops. © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 34:1335–1343, 2018 [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
4. A humanised tissue‐engineered bone model allows species‐specific breast cancer‐related bone metastasis in vivo.
- Author
-
Quent, V. M. C., Taubenberger, A. V., Reichert, J. C., Martine, L. C., Clements, J. A., Hutmacher, D. W., and Loessner, D.
- Published
- 2018
- Full Text
- View/download PDF
5. Growth of confined cancer spheroids: a combined experimental and mathematical modelling approach.
- Author
-
Loessner, D., Flegg, J. A., Byrne, H. M., Clements, J. A., and Hutmacher, D. W.
- Published
- 2013
- Full Text
- View/download PDF
6. 251 Kallikrein-related Peptidase 4 and Prostate Cancer – an Extended Role in Tumour Progression
- Author
-
Fuhrman-Luck, R., Stansfield, S., Stephens, C., Loessner, D., and Clements, J.
- Published
- 2012
- Full Text
- View/download PDF
7. 113 Unravelling the Tumour-stroma Microenvironment of Late Stage Ovarian Cancer
- Author
-
Loessner, D., Baldwin, J., Rockstroh, A., Irving-Rodgers, H., Magdolen, V., Hutmacher, D.W., and Clements, J.A.
- Published
- 2012
- Full Text
- View/download PDF
8. 103: The tumour–stroma niche of ovarian cancer in 3D.
- Author
-
Loessner, D., Holzapfel, B.M., Baldwin, J., Rockstroh, A., Magdolen, V., Hutmacher, D.W., and Clements, J.A.
- Published
- 2014
- Full Text
- View/download PDF
9. The kallikrein-related peptidase family: Dysregulation and functions during cancer progression.
- Author
-
Kryza, T., Silva, M.L., Loessner, D., Heuzé-Vourc'h, N., and Clements, J.A.
- Subjects
- *
KALLIKREIN , *PEPTIDASE , *CANCER invasiveness , *PROSTATE cancer , *DESMOGLEINS - Abstract
Cancer is the second leading cause of death with 14 million new cases and 8.2 million cancer-related deaths worldwide in 2012. Despite the progress made in cancer therapies, neoplastic diseases are still a major therapeutic challenge notably because of intra- and inter-malignant tumour heterogeneity and adaptation/escape of malignant cells to/from treatment. New targeted therapies need to be developed to improve our medical arsenal and counter-act cancer progression. Human kallikrein-related peptidases (KLKs) are secreted serine peptidases which are aberrantly expressed in many cancers and have great potential in developing targeted therapies. The potential of KLKs as cancer biomarkers is well established since the demonstration of the association between KLK3/PSA (prostate specific antigen) levels and prostate cancer progression. In addition, a constantly increasing number of in vitro and in vivo studies demonstrate the functional involvement of KLKs in cancer-related processes. These peptidases are now considered key players in the regulation of cancer cell growth, migration, invasion, chemo-resistance, and importantly, in mediating interactions between cancer cells and other cell populations found in the tumour microenvironment to facilitate cancer progression. These functional roles of KLKs in a cancer context further highlight their potential in designing new anti-cancer approaches. In this review, we comprehensively review the biochemical features of KLKs, their functional roles in carcinogenesis, followed by the latest developments and the successful utility of KLK-based therapeutics in counteracting cancer progression. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
10. 263: Mimicking the tumour microenvironment (TME): Angiogenesis in tumour progression.
- Author
-
Bonda, U.H., Bray, L.J., Lister, N., Ellem, S., Risbridger, G., Freudenberg, U., Werner, C.C., Hutmacher, D.W., De-Juan-Pardo, E.M., and Loessner, D.
- Published
- 2014
- Full Text
- View/download PDF
11. 278: The kallikrein-related serine peptidase, KLK4, regulates the TGFb1 pathway in the tumour–stroma microenvironment in prostate cancer.
- Author
-
Clements, J., Fuhrman-Luck, R., Stansfield, S., Hastie, M., Stoll, T., Stephens, C., Loessner, D., Lehman, M., Nelson, C., and Gorman, J.
- Published
- 2014
- Full Text
- View/download PDF
12. Tailoring metabolic activity assays for tumour-engineered 3D models.
- Author
-
Clegg J, Curvello R, Gabrielyan A, Croagh D, Hauser S, and Loessner D
- Abstract
Monitoring cell behaviour in hydrogel-based 3D models is critical for assessing their growth and response to cytotoxic treatment. Resazurin-based PrestoBlue and AlamarBlue reagents are frequently used metabolic activity assays when determining cell responses. However, both assays are largely applied to cell monolayer cultures but yet to have a defined protocol for use in hydrogel-based 3D models. The assays' performance depends on the cell type, culture condition and measurement sensitivity. To better understand how both assays perform, we grew pancreatic cancer cells in gelatin methacryloyl and collagen hydrogels and evaluated their metabolic activity using different concentrations and incubation times of the PrestoBlue and AlamarBlue reagents. We tested reagent concentrations of 4 % and 10 % and incubation times of 45 min, 2 h and 4 h. In addition, we co-cultured cancer cells together with cancer-associated fibroblasts and peripheral blood mononuclear cells in gelatin methacryloyl hydrogels and subjected them to gemcitabine and nab-paclitaxel to evaluate how both assays perform when characterising cell responses upon drug treatment. CyQuant assays were conducted on the same samples and compared to data from the metabolic activity assays. In cancer monocultures, higher reagent concentration and incubation time increased fluorescent intensity. We found a reagent concentration of 10 % and an incubation time of 2 h suitable for all cell lines and both hydrogels. In multicellular 3D cultures, PrestoBlue and AlamarBlue assays detected similar cell responses upon drug treatment but overestimated cell growth. We recommend to assess cell viability and growth in conjunction with CyQuant assays that directly measure cell functions., Competing Interests: Declaration of competing interest The authors do not have any conflict of interest., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. Tumor-associated mesenchymal stromal cells modulate macrophage phagocytosis in stromal-rich colorectal cancer via PD-1 signaling.
- Author
-
Leonard NA, Corry SM, Reidy E, Egan H, O'Malley G, Thompson K, McDermott E, O'Neill A, Zakaria N, Egan LJ, Ritter T, Loessner D, Redmond K, Sheehan M, Canney A, Hogan AM, Hynes SO, Treacy O, Dunne PD, and Ryan AE
- Abstract
CMS4 colorectal cancer (CRC), based on the consensus molecular subtype (CMS), stratifies patients with the poorest disease-free survival rates. It is characterized by a strong mesenchymal stromal cell (MSC) signature, wound healing-like inflammation and therapy resistance. We utilized 2D and 3D in vitro, in vivo , and ex vivo models to assess the impact of inflammation and stromal cells on immunosuppression in CMS4 CRC. RNA sequencing data from untreated stage II/III CRC patients showed enriched TNF-α signatures in CMS1 and CMS4 tumors. Secretome from TNF-α treated cancer cells induced an immunomodulatory and chemotactic phenotype in MSC and cancer-associated fibroblasts (CAFs). Macrophages in CRC tumours migrate and preferentially localise in stromal compartment. Inflammatory CRC secretome enhances expression of PD-L1 and CD47 on both human and murine stromal cells. We demonstrate that TNF-α-induced inflammation in CRC suppresses macrophage phagocytosis via stromal cells. We show that stromal cell-mediated suppression of macrophage phagocytosis is mediated in part through PD-1 signaling. These data suggest that re-stratification of CRC by CMS may reveal patient subsets with microsatellite stable tumors, particularly CMS4-like tumors, that may respond to immunotherapies., Competing Interests: The authors declare no conflicts of interest., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
14. A Self-Assembled 3D Model Demonstrates How Stiffness Educates Tumor Cell Phenotypes and Therapy Resistance in Pancreatic Cancer.
- Author
-
Liu Y, Okesola BO, Osuna de la Peña D, Li W, Lin ML, Trabulo S, Tatari M, Lawlor RT, Scarpa A, Wang W, Knight M, Loessner D, Heeschen C, Mata A, and Pearce OMT
- Subjects
- Humans, Cell Line, Tumor, Carcinoma, Pancreatic Ductal pathology, Carcinoma, Pancreatic Ductal metabolism, Peptides chemistry, Peptides pharmacology, Phenotype, Receptors, CXCR4 metabolism, Pancreatic Neoplasms pathology, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms drug therapy, Hydrogels chemistry, Drug Resistance, Neoplasm drug effects, Epithelial-Mesenchymal Transition drug effects, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Extracellular Matrix metabolism, Tumor Microenvironment drug effects
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by a dense and stiff extracellular matrix (ECM) associated with tumor progression and therapy resistance. To further the understanding of how stiffening of the tumor microenvironment (TME) contributes to aggressiveness, a three-dimensional (3D) self-assembling hydrogel disease model is developed based on peptide amphiphiles (PAs, PA-E3Y) designed to tailor stiffness. The model displays nanofibrous architectures reminiscent of native TME and enables the study of the invasive behavior of PDAC cells. Enhanced tuneability of stiffness is demonstrated by interacting thermally annealed aqueous solutions of PA-E3Y (PA-E3Y
h ) with divalent cations to create hydrogels with mechanical properties and ultrastructure similar to native tumor ECM. It is shown that stiffening of PA-E3Yh hydrogels to levels found in PDAC induces ECM deposition, promotes epithelial-to-mesenchymal transition (EMT), enriches CD133+ /CXCR4+ cancer stem cells (CSCs), and subsequently enhances drug resistance. The findings reveal how a stiff 3D environment renders PDAC cells more aggressive and therefore more faithfully recapitulates in vivo tumors., (© 2024 The Authors. Advanced Healthcare Materials published by Wiley‐VCH GmbH.)- Published
- 2024
- Full Text
- View/download PDF
15. Recreating metabolic interactions of the tumour microenvironment.
- Author
-
Curvello R, Berndt N, Hauser S, and Loessner D
- Subjects
- Humans, Animals, Extracellular Matrix metabolism, Tumor Microenvironment physiology, Neoplasms metabolism, Neoplasms pathology
- Abstract
Tumours are heterogeneous tissues containing diverse populations of cells and an abundant extracellular matrix (ECM). This tumour microenvironment prompts cancer cells to adapt their metabolism to survive and grow. Besides epigenetic factors, the metabolism of cancer cells is shaped by crosstalk with stromal cells and extracellular components. To date, most experimental models neglect the complexity of the tumour microenvironment and its relevance in regulating the dynamics of the metabolism in cancer. We discuss emerging strategies to model cellular and extracellular aspects of cancer metabolism. We highlight cancer models based on bioengineering, animal, and mathematical approaches to recreate cell-cell and cell-matrix interactions and patient-specific metabolism. Combining these approaches will improve our understanding of cancer metabolism and support the development of metabolism-targeting therapies., Competing Interests: Declaration of interests No interests are declared., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
16. A Tumor Microenvironment Model of Pancreatic Cancer to Elucidate Responses toward Immunotherapy.
- Author
-
Kast V, Nadernezhad A, Pette D, Gabrielyan A, Fusenig M, Honselmann KC, Stange DE, Werner C, and Loessner D
- Subjects
- Humans, Immunotherapy, Immunomodulation, Tumor Microenvironment, Pancreatic Neoplasms drug therapy
- Abstract
Pancreatic cancer is a devastating malignancy with minimal treatment options. Standard-of-care therapy, including surgery and chemotherapy, is unsatisfactory, and therapies harnessing the immune system have been unsuccessful in clinical trials. Resistance to therapy and disease progression are mediated by the tumor microenvironment, which contains excessive amounts of extracellular matrix and stromal cells, acting as a barrier to drug delivery. There is a lack of preclinical pancreatic cancer models that reconstruct the extracellular, cellular, and biomechanical elements of tumor tissues to assess responses toward immunotherapy. To address this limitation and explore the effects of immunotherapy in combination with chemotherapy, a multicellular 3D cancer model using a star-shaped poly(ethylene glycol)-heparin hydrogel matrix is developed. Human pancreatic cancer cells, cancer-associated fibroblasts, and myeloid cells are grown encapsulated in hydrogels to mimic key components of tumor tissues, and cell responses toward treatment are assessed. Combining the CD11b agonist ADH-503 with anti-PD-1 immunotherapy and chemotherapy leads to a significant reduction in tumor cell viability, proliferation, metabolic activity, immunomodulation, and secretion of immunosuppressive and tumor growth-promoting cytokines., (© 2022 The Authors. Advanced Healthcare Materials published by Wiley-VCH GmbH.)
- Published
- 2023
- Full Text
- View/download PDF
17. Rab31-dependent regulation of transforming growth factor ß expression in breast cancer cells.
- Author
-
Soelch S, Beaufort N, Loessner D, Kotzsch M, Reuning U, Luther T, Kirchner T, and Magdolen V
- Subjects
- Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Line, Tumor, Female, Humans, Transforming Growth Factor beta1 metabolism, rab GTP-Binding Proteins metabolism, Breast Neoplasms genetics, Epithelial-Mesenchymal Transition genetics, Transforming Growth Factor beta1 genetics, rab GTP-Binding Proteins genetics
- Abstract
Background: The small GTP-binding protein Rab31 plays an important role in the modulation of tumor biological-relevant processes, including cell proliferation, adhesion, and invasion. As an underlying mechanism, Rab31 is presumed to act as a molecular switch between a more proliferative and an invasive phenotype. This prompted us to analyze whether Rab31 overexpression in breast cancer cells affects expression of genes involved in epithelial-to-mesenchymal transition (EMT)-like processes when compared to Rab31 low-expressing cells., Methods: Commercially available profiler PCR arrays were applied to search for differentially expressed genes in Rab31 high- and low-expressing CAMA-1 breast cancer cells. Differential expression of selected candidate genes in response to Rab31 overexpression in CAMA-1 cells was validated by independent qPCR and protein assays., Results: Gene expression profiling of key genes involved in EMT, or its reciprocal process MET, identified 9 genes being significantly up- or down-regulated in Rab31 overexpressing CAMA-1 cells, with the strongest effects seen for TGFB1, encoding TGF-ß1 (> 25-fold down-regulation in Rab31 overexpressing cells). Subsequent validation analyses by qPCR revealed a strong down-regulation of TGFB1 mRNA levels in response to increased Rab31 expression not only in CAMA-1 cells, but also in another breast cancer cell line, MDA-MB-231. Using ELISA and Western blot analysis, a considerable reduction of both intracellular and secreted TGF-ß1 antigen levels was determined in Rab31 overexpressing cells compared to vector control cells. Furthermore, reduced TGF-ß activity was observed upon Rab31 overexpression in CAMA-1 cells using a sensitive TGF-ß bioassay. Finally, the relationship between Rab31 expression and the TGF-ß axis was analyzed by another profiler PCR array focusing on genes involved in TGF-ß signaling. We found 12 out of 84 mRNAs significantly reduced and 7 mRNAs significantly increased upon Rab31 overexpression., Conclusions: Our results demonstrate that Rab31 is a potent modulator of the expression of TGF-ß and other components of the TGF-ß signaling pathway in breast cancer cells., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
18. Stromal Cells Promote Matrix Deposition, Remodelling and an Immunosuppressive Tumour Microenvironment in a 3D Model of Colon Cancer.
- Author
-
Leonard NA, Reidy E, Thompson K, McDermott E, Peerani E, Tomas Bort E, Balkwill FR, Loessner D, and Ryan AE
- Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related deaths worldwide. CRC develops in a complex tumour microenvironment (TME) with both mesenchymal stromal cells (MSCs) and immune infiltrate, shown to alter disease progression and treatment response. We hypothesised that an accessible, affordable model of CRC that combines multiple cell types will improve research translation to the clinic and enable the identification of novel therapeutic targets. A viable gelatine-methacrloyl-based hydrogel culture system that incorporates CRC cells with MSCs and a monocyte cell line was developed. Gels were analysed on day 10 by PCR, cytokine array, microscopy and flow cytometry. The addition of stromal cells increased transcription of matrix remodelling proteins FN1 and MMP9, induced release of tumour-promoting immune molecules MIF, Serpin E1, CXCL1, IL-8 and CXCL12 and altered cancer cell expression of immunotherapeutic targets EGFR, CD47 and PD-L1. Treatment with PD153035, an EGFR inhibitor, revealed altered CRC expression of PD-L1 but only in gels lacking MSCs. We established a viable 3D model of CRC that combined cancer cells, MSCs and monocytic cells that can be used to research the role the stroma plays in the TME, identify novel therapeutic targets and improve the transitional efficacy of therapies.
- Published
- 2021
- Full Text
- View/download PDF
19. Modeling the Tumor Microenvironment of Ovarian Cancer: The Application of Self-Assembling Biomaterials.
- Author
-
Mendoza-Martinez AK, Loessner D, Mata A, and Azevedo HS
- Abstract
Ovarian cancer (OvCa) is one of the leading causes of gynecologic malignancies. Despite treatment with surgery and chemotherapy, OvCa disseminates and recurs frequently, reducing the survival rate for patients. There is an urgent need to develop more effective treatment options for women diagnosed with OvCa. The tumor microenvironment (TME) is a key driver of disease progression, metastasis and resistance to treatment. For this reason, 3D models have been designed to represent this specific niche and allow more realistic cell behaviors compared to conventional 2D approaches. In particular, self-assembling peptides represent a promising biomaterial platform to study tumor biology. They form nanofiber networks that resemble the architecture of the extracellular matrix and can be designed to display mechanical properties and biochemical motifs representative of the TME. In this review, we highlight the properties and benefits of emerging 3D platforms used to model the ovarian TME. We also outline the challenges associated with using these 3D systems and provide suggestions for future studies and developments. We conclude that our understanding of OvCa and advances in materials science will progress the engineering of novel 3D approaches, which will enable the development of more effective therapies.
- Published
- 2021
- Full Text
- View/download PDF
20. TGFBI Production by Macrophages Contributes to an Immunosuppressive Microenvironment in Ovarian Cancer.
- Author
-
Lecker LSM, Berlato C, Maniati E, Delaine-Smith R, Pearce OMT, Heath O, Nichols SJ, Trevisan C, Novak M, McDermott J, Brenton JD, Cutillas PR, Rajeeve V, Hennino A, Drapkin R, Loessner D, and Balkwill FR
- Subjects
- Animals, Apoptosis, Cell Proliferation, Cystadenocarcinoma, Serous genetics, Cystadenocarcinoma, Serous immunology, Cystadenocarcinoma, Serous metabolism, Extracellular Matrix immunology, Extracellular Matrix metabolism, Female, Humans, Immunosuppressive Agents, Mice, Mice, Inbred C57BL, Ovarian Neoplasms genetics, Ovarian Neoplasms immunology, Ovarian Neoplasms metabolism, Peritoneal Neoplasms genetics, Peritoneal Neoplasms immunology, Peritoneal Neoplasms metabolism, Prognosis, Transforming Growth Factor beta1 genetics, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Cystadenocarcinoma, Serous pathology, Extracellular Matrix pathology, Macrophages immunology, Ovarian Neoplasms pathology, Peritoneal Neoplasms pathology, Transforming Growth Factor beta1 metabolism, Tumor Microenvironment
- Abstract
The tumor microenvironment evolves during malignant progression, with major changes in nonmalignant cells, cytokine networks, and the extracellular matrix (ECM). In this study, we aimed to understand how the ECM changes during neoplastic transformation of serous tubal intraepithelial carcinoma lesions (STIC) into high-grade serous ovarian cancers (HGSOC). Analysis of the mechanical properties of human fallopian tubes (FT) and ovaries revealed that normal FT and fimbria had a lower tissue modulus, a measure of stiffness, than normal or diseased ovaries. Proteomic analysis of the matrisome fraction between FT, fimbria, and ovaries showed significant differences in the ECM protein TGF beta induced (TGFBI, also known as βig-h3). STIC lesions in the fimbria expressed high levels of TGFBI, which was predominantly produced by CD163-positive macrophages proximal to STIC epithelial cells. In vitro stimulation of macrophages with TGFβ and IL4 induced secretion of TGFBI, whereas IFNγ/LPS downregulated macrophage TGFBI expression. Immortalized FT secretory epithelial cells carrying clinically relevant TP53 mutations stimulated macrophages to secrete TGFBI and upregulated integrin αvβ3, a putative TGFBI receptor. Transcriptomic HGSOC datasets showed a significant correlation between TGFBI expression and alternatively activated macrophage signatures. Fibroblasts in HGSOC metastases expressed TGFBI and stimulated macrophage TGFBI production in vitro . Treatment of orthotopic mouse HGSOC tumors with an anti-TGFBI antibody reduced peritoneal tumor size, increased tumor monocytes, and activated β3-expressing unconventional T cells. In conclusion, TGFBI may favor an immunosuppressive microenvironment in STICs that persists in advanced HGSOC. Furthermore, TGFBI may be an effector of the tumor-promoting actions of TGFβ and a potential therapeutic target. SIGNIFICANCE: Analysis of ECM changes during neoplastic transformation reveals a role for TGFBI secreted by macrophages in immunosuppression in early ovarian cancer., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
21. Bioengineered 3D models of human pancreatic cancer recapitulate in vivo tumour biology.
- Author
-
Osuna de la Peña D, Trabulo SMD, Collin E, Liu Y, Sharma S, Tatari M, Behrens D, Erkan M, Lawlor RT, Scarpa A, Heeschen C, Mata A, and Loessner D
- Subjects
- Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Cell Culture Techniques methods, Gene Expression Profiling methods, Gene Expression Regulation, Neoplastic, Humans, Neoplastic Stem Cells metabolism, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Reproducibility of Results, Stromal Cells metabolism, Tumor Cells, Cultured, Bioengineering methods, Carcinoma, Pancreatic Ductal pathology, Models, Biological, Neoplastic Stem Cells pathology, Pancreatic Neoplasms pathology, Stromal Cells pathology
- Abstract
Patient-derived in vivo models of human cancer have become a reality, yet their turnaround time is inadequate for clinical applications. Therefore, tailored ex vivo models that faithfully recapitulate in vivo tumour biology are urgently needed. These may especially benefit the management of pancreatic ductal adenocarcinoma (PDAC), where therapy failure has been ascribed to its high cancer stem cell (CSC) content and high density of stromal cells and extracellular matrix (ECM). To date, these features are only partially reproduced ex vivo using organoid and sphere cultures. We have now developed a more comprehensive and highly tuneable ex vivo model of PDAC based on the 3D co-assembly of peptide amphiphiles (PAs) with custom ECM components (PA-ECM). These cultures maintain patient-specific transcriptional profiles and exhibit CSC functionality, including strong in vivo tumourigenicity. User-defined modification of the system enables control over niche-dependent phenotypes such as epithelial-to-mesenchymal transition and matrix deposition. Indeed, proteomic analysis of these cultures reveals improved matrisome recapitulation compared to organoids. Most importantly, patient-specific in vivo drug responses are better reproduced in self-assembled cultures than in other models. These findings support the use of tuneable self-assembling platforms in cancer research and pave the way for future precision medicine approaches., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
22. A humanized orthotopic tumor microenvironment alters the bone metastatic tropism of prostate cancer cells.
- Author
-
McGovern JA, Bock N, Shafiee A, Martine LC, Wagner F, Baldwin JG, Landgraf M, Lahr CA, Meinert C, Williams ED, Pollock PM, Denham J, Russell PJ, Risbridger GP, Clements JA, Loessner D, Holzapfel BM, and Hutmacher DW
- Subjects
- Animals, Humans, Male, Mice, Mice, Inbred NOD, Mice, SCID, Neoplasm Metastasis, Bone Neoplasms secondary, Prostatic Neoplasms pathology, Tissue Engineering, Tumor Microenvironment
- Abstract
Prostate cancer (PCa) is the second most commonly diagnosed cancer in men, and bone is the most frequent site of metastasis. The tumor microenvironment (TME) impacts tumor growth and metastasis, yet the role of the TME in PCa metastasis to bone is not fully understood. We used a tissue-engineered xenograft approach in NOD-scid IL2Rγ
null (NSG) mice to incorporate two levels of humanization; the primary tumor and TME, and the secondary metastatic bone organ. Bioluminescent imaging, histology, and immunohistochemistry were used to study metastasis of human PC-3 and LNCaP PCa cells from the prostate to tissue-engineered bone. Here we show pre-seeding scaffolds with human osteoblasts increases the human cellular and extracellular matrix content of bone constructs, compared to unseeded scaffolds. The humanized prostate TME showed a trend to decrease metastasis of PC-3 PCa cells to the tissue-engineered bone, but did not affect the metastatic potential of PCa cells to the endogenous murine bones or organs. On the other hand, the humanized TME enhanced LNCaP tumor growth and metastasis to humanized and murine bone. Together this demonstrates the importance of the TME in PCa bone tropism, although further investigations are needed to delineate specific roles of the TME components in this context., (© 2021. Crown.)- Published
- 2021
- Full Text
- View/download PDF
23. Kallikrein-Related Peptidase 6 Is Associated with the Tumour Microenvironment of Pancreatic Ductal Adenocarcinoma.
- Author
-
Candido JB, Maiques O, Boxberg M, Kast V, Peerani E, Tomás-Bort E, Weichert W, Sananes A, Papo N, Magdolen V, Sanz-Moreno V, and Loessner D
- Abstract
As cancer-associated factors, kallikrein-related peptidases (KLKs) are components of the tumour microenvironment, which represents a rich substrate repertoire, and considered attractive targets for the development of novel treatments. Standard-of-care therapy of pancreatic cancer shows unsatisfactory results, indicating the need for alternative therapeutic approaches. We aimed to investigate the expression of KLKs in pancreatic cancer and to inhibit the function of KLK6 in pancreatic cancer cells. KLK6, KLK7, KLK8, KLK10 and KLK11 were coexpressed and upregulated in tissues from pancreatic cancer patients compared to normal pancreas. Their high expression levels correlated with each other and were linked to shorter survival compared to low KLK levels. We then validated KLK6 mRNA and protein expression in patient-derived tissues and pancreatic cancer cells. Coexpression of KLK6 with KRT19, αSMA or CD68 was independent of tumour stage, while KLK6 was coexpressed with KRT19 and CD68 in the invasive tumour area. High KLK6 levels in tumour and CD68+ cells were linked to shorter survival. KLK6 inhibition reduced KLK6 mRNA expression, cell metabolic activity and KLK6 secretion and increased the secretion of other serine and aspartic lysosomal proteases. The association of high KLK levels and poor prognosis suggests that inhibiting KLKs may be a therapeutic strategy for precision medicine.
- Published
- 2021
- Full Text
- View/download PDF
24. 3D Collagen-Nanocellulose Matrices Model the Tumour Microenvironment of Pancreatic Cancer.
- Author
-
Curvello R, Kast V, Abuwarwar MH, Fletcher AL, Garnier G, and Loessner D
- Abstract
Three-dimensional (3D) cancer models are invaluable tools designed to study tumour biology and new treatments. Pancreatic ductal adenocarcinoma (PDAC), one of the deadliest types of cancer, has been progressively explored with bioengineered 3D approaches by deconstructing elements of its tumour microenvironment. Here, we investigated the suitability of collagen-nanocellulose hydrogels to mimic the extracellular matrix of PDAC and to promote the formation of tumour spheroids and multicellular 3D cultures with stromal cells. Blending of type I collagen fibrils and cellulose nanofibres formed a matrix of controllable stiffness, which resembled the lower profile of pancreatic tumour tissues. Collagen-nanocellulose hydrogels supported the growth of tumour spheroids and multicellular 3D cultures, with increased metabolic activity and matrix stiffness. To validate our 3D cancer model, we tested the individual and combined effects of the anti-cancer compound triptolide and the chemotherapeutics gemcitabine and paclitaxel, resulting in differential cell responses. Our blended 3D matrices with tuneable mechanical properties consistently maintain the growth of PDAC cells and its cellular microenvironment and allow the screening of anti-cancer treatments., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Curvello, Kast, Abuwarwar, Fletcher, Garnier and Loessner.)
- Published
- 2021
- Full Text
- View/download PDF
25. Addressing the tumour microenvironment in early drug discovery: a strategy to overcome drug resistance and identify novel targets for cancer therapy.
- Author
-
Kaemmerer E, Loessner D, and Avery VM
- Subjects
- Animals, Drug Evaluation, Preclinical methods, Drug Resistance, Neoplasm, Extracellular Matrix metabolism, High-Throughput Screening Assays, Humans, Models, Biological, Molecular Targeted Therapy, Tumor Microenvironment, Antineoplastic Agents pharmacology, Drug Discovery methods, Neoplasms drug therapy
- Abstract
The tumour microenvironment (TME) comprises not only malignant and non-malignant cells, but also the extracellular matrix (ECM), secreted factors, and regulators of cellular functions. In addition to genetic alterations, changes of the biochemical/biophysical properties or cellular composition of the TME have been implicated in drug resistance. Here, we review the composition of the ECM and different elements of the TME contributing to drug resistance, including soluble factors, hypoxia, extracellular acidity, and cell adhesion properties. We discuss selected approaches for modelling the TME, current progress, and their use in low-and high-throughput assays for preclinical studies. Lastly, we summarise the status quo of advanced 3D cancer models compatible with high-throughput screening (HTS), the technical practicalities and challenges., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
26. Neuroblastoma Invasion Strategies Are Regulated by the Extracellular Matrix.
- Author
-
Gavin C, Geerts N, Cavanagh B, Haynes M, Reynolds CP, Loessner D, Ewald AJ, and Piskareva O
- Abstract
Neuroblastoma is a paediatric malignancy of the developing sympathetic nervous system. About half of the patients have metastatic disease at the time of diagnosis and a survival rate of less than 50%. Our understanding of the cellular processes promoting neuroblastoma metastases will be facilitated by the development of appropriate experimental models. In this study, we aimed to explore the invasion of neuroblastoma cells and organoids from patient-derived xenografts (PDXs) grown embedded in 3D extracellular matrix (ECM) hydrogels by time-lapse microscopy and quantitative image analysis. We found that the ECM composition influenced the growth, viability and local invasion of organoids. The ECM compositions induced distinct cell behaviours, with Matrigel being the preferred substratum for local organoid invasion. Organoid invasion was cell line- and PDX-dependent. We identified six distinct phenotypes in PDX-derived organoids. In contrast, NB cell lines were more phenotypically restricted in their invasion strategies, as organoids isolated from cell line-derived xenografts displayed a broader range of phenotypes compared to clonal cell line clusters. The addition of FBS and bFGF induced more aggressive cell behaviour and a broader range of phenotypes. In contrast, the repression of the prognostic neuroblastoma marker, MYCN , resulted in less aggressive cell behaviour. The combination of PDX organoids, real-time imaging and the novel 3D culture assays developed herein will enable rapid progress in elucidating the molecular mechanisms that control neuroblastoma invasion.
- Published
- 2021
- Full Text
- View/download PDF
27. 3D Models for Ovarian Cancer.
- Author
-
Kast V and Loessner D
- Subjects
- Cell Culture Techniques, Female, Humans, Organoids, Tumor Microenvironment, Ovarian Neoplasms drug therapy, Spheroids, Cellular
- Abstract
The main reasons for the slow progress in improving survival outcomes for ovarian cancer are the 'one-size-fits-all' therapy and lack of clinically relevant experimental models that represent the advanced stages of the human disease. The interaction of tumour cells with their surrounding niche, the tumour microenvironment, influences the spread of ovarian cancer cells within the peritoneum and their responses to therapeutics. Scientists are increasingly using 3D cell culture models to dissect the role of the tumour microenvironment in cancer development and progression and the treatment of this disease. In this chapter, we will briefly describe the tumour microenvironment of ovarian cancer. Then, we will review some of the clinically relevant experimental approaches, such as spheroid, organoid and organotypic models, that have been developed for the 3D culture of ovarian cancer cells using different tools, including hydrogels, scaffolds and cancer-on-a-chip devices, to mimic selected components of the tumour microenvironment., (© 2021. Springer Nature Switzerland AG.)
- Published
- 2021
- Full Text
- View/download PDF
28. Peptide-protein coassembling matrices as a biomimetic 3D model of ovarian cancer.
- Author
-
Hedegaard CL, Redondo-Gómez C, Tan BY, Ng KW, Loessner D, and Mata A
- Subjects
- Cell Line, Tumor, Extracellular Matrix metabolism, Humans, Peptides metabolism, Tumor Microenvironment, Biomimetics, Ovarian Neoplasms metabolism
- Abstract
Bioengineered three-dimensional (3D) matrices expand our experimental repertoire to study tumor growth and progression in a biologically relevant, yet controlled, manner. Here, we used peptide amphiphiles (PAs) to coassemble with and organize extracellular matrix (ECM) proteins producing tunable 3D models of the tumor microenvironment. The matrix was designed to mimic physical and biomolecular features of tumors present in patients. We included specific epitopes, PA nanofibers, and ECM macromolecules for the 3D culture of human ovarian cancer, endothelial, and mesenchymal stem cells. The multicellular constructs supported the formation of tumor spheroids with extensive F-actin networks surrounding the spheroids, enabling cell-cell communication, and comparative cell-matrix interactions and encapsulation response to those observed in Matrigel. We conducted a proof-of-concept study with clinically used chemotherapeutics to validate the functionality of the multicellular constructs. Our study demonstrates that peptide-protein coassembling matrices serve as a defined model of the multicellular tumor microenvironment of primary ovarian tumors., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)
- Published
- 2020
- Full Text
- View/download PDF
29. 3D approaches to model the tumor microenvironment of pancreatic cancer.
- Author
-
Tomás-Bort E, Kieler M, Sharma S, Candido JB, and Loessner D
- Subjects
- Animals, Cell Culture Techniques, Disease Models, Animal, Humans, Tumor Microenvironment, Pancreatic Neoplasms pathology, Spheroids, Cellular pathology, Tissue Engineering methods
- Abstract
In tumor engineering, 3D approaches are used to model components of the tumor microenvironment and to test new treatments. Pancreatic cancers are a cancer of substantial unmet need and survival rates are lower compared to any other cancer. Bioengineering techniques are increasingly applied to understand the unique biology of pancreatic tumors and to design patient-specific models. Here we summarize how extracellular and cellular elements of the pancreatic tumor microenvironment and their interactions have been studied in 3D cell cultures. We review selected clinical trials, assess the benefits of therapies interfering with the tumor microenvironment and address their limitations and future perspectives., Competing Interests: Competing Interests: The authors have declared that no competing interest exists., (© The author(s).)
- Published
- 2020
- Full Text
- View/download PDF
30. A humanized bone microenvironment uncovers HIF2 alpha as a latent marker for osteosarcoma.
- Author
-
Wagner F, Holzapfel BM, Martine LC, McGovern J, Lahr CA, Boxberg M, Prodinger PM, Grässel S, Loessner D, and Hutmacher DW
- Subjects
- Animals, Bone Morphogenetic Protein 7 pharmacology, Bone Neoplasms pathology, Heterografts, Humans, Mice, Mice, Inbred NOD, Mice, SCID, Neoplasm Transplantation, Osteosarcoma pathology, Basic Helix-Loop-Helix Transcription Factors metabolism, Biomarkers, Tumor metabolism, Bone Neoplasms metabolism, Neoplasm Proteins metabolism, Osteosarcoma metabolism, Tumor Microenvironment
- Abstract
The quest for predictive tumor markers for osteosarcoma (OS) has not well progressed over the last two decades due to a lack of preclinical models. The aim of this study was to investigate if microenvironmental modifications in an original humanized in vivo model alter the expression of OS tumor markers. Human bone micro-chips and bone marrow, harvested during hip arthroplasty, were implanted at the flanks of NOD/scid mice. We administered recombinant human bone morphogenetic protein 7 (rhBMP-7) in human bone micro-chips/bone marrow group I in order to modulate bone matrix and bone marrow humanization. Ten weeks post-implantation, human Luc-SAOS-2 OS cells were injected into the humanized tissue-engineered bone organs (hTEBOs). Tumors were harvested 5 weeks post-implantation to determine the expression of the previously described OS markers ezrin, periostin, VEGF, HIF1α and HIF2α. Representation of these proteins was analyzed in two different OS patient cohorts. Ezrin was downregulated in OS in hTEBOs with rhBMP-7, whereas HIF2α was significantly upregulated in comparison to hTEBOs without rhBMP-7. The expression of periostin, VEGF and HIF1α did not differ significantly between both groups. HIF2α was consistently present in OS patients and dependent on tumor site and clinical stage. OS patients post-chemotherapy had suppressed levels of HIF2α. In conclusion, we demonstrated the overall expression of OS-related factors in a preclinical model, which is based on a humanized bone organ. Our preclinical research results and analysis of two comprehensive patient cohorts imply that HIF2α is a potential prognostic marker and/or therapeutic target. STATEMENT OF SIGNIFICANCE: This study demonstrates the clinical relevance of the humanized organ bone microenvironment in osteosarcoma research and validates the expression of tumor markers, especially HIF2α. The convergence of clinically proven bone engineering concepts for the development of humanized mice models is a new starting point for investigations of OS-related marker expression. The validation and first data set in such a model let one conclude that further clinical studies on the role of HIF2α as a prognostic marker and its potential as therapeutic target is a condition sine qua non., (Copyright © 2019 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
31. A 3D tumor microenvironment regulates cell proliferation, peritoneal growth and expression patterns.
- Author
-
Loessner D, Rockstroh A, Shokoohmand A, Holzapfel BM, Wagner F, Baldwin J, Boxberg M, Schmalfeldt B, Lengyel E, Clements JA, and Hutmacher DW
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, Coculture Techniques, Epithelium pathology, Female, Gene Expression Regulation, Neoplastic, Humans, Mice, Inbred NOD, Mice, SCID, Ovarian Neoplasms genetics, Peritoneal Neoplasms genetics, Peritoneal Neoplasms pathology, Tissue Scaffolds chemistry, Transcriptome, Ovarian Neoplasms pathology, Peritoneal Neoplasms secondary, Peritoneum pathology, Tumor Microenvironment
- Abstract
Peritoneal invasion through the mesothelial cell layer is a hallmark of ovarian cancer metastasis. Using tissue engineering technologies, we recreated an ovarian tumor microenvironment replicating this aspect of disease progression. Ovarian cancer cell-laden hydrogels were combined with mesothelial cell-layered melt electrospun written scaffolds and characterized with proliferation and transcriptomic analyses and used as intraperitoneal xenografts. Here we show increased cancer cell proliferation in these 3D co-cultures, which we validated using patient-derived cells and linked to peritoneal tumor growth in vivo. Transcriptome-wide expression analysis identified IGFBP7, PTGS2, VEGFC and FGF2 as bidirectional factors deregulated in 3D co-cultures compared to 3D mono-cultures, which we confirmed by immunohistochemistry of xenograft and patient-derived tumor tissues and correlated with overall and progression-free survival. These factors were further increased upon expression of kallikrein-related proteases. This clinically predictive model allows us to mimic the complexity and processes of the metastatic disease that may lead to therapies that protect from peritoneal invasion or delay the development of metastasis., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
32. Cell Recovery of Hydrogel-Encapsulated Cells for Molecular Analysis.
- Author
-
Peerani E, Candido JB, and Loessner D
- Subjects
- Cell Line, Tumor, Cell Proliferation, Cell Survival, Culture Media chemistry, Flow Cytometry methods, Humans, Microscopy, Confocal, Microscopy, Fluorescence, Spheroids, Cellular, Cell Culture Techniques methods, Cell Encapsulation methods, Hydrogels
- Abstract
Tissue engineering technologies have produced controllable and reproducible three-dimensional (3D) models that mimic the architecture and complexity of native tissues. In particular cell biology-based research is driven by the development of cell culture platforms and techniques that allow the analysis of cells cultured in 3D. Here we summarize several easy-to-follow methods for the characterization of cells that have been encapsulated and grown in hydrogels to measure their cell viability, metabolic activity, and mechanical properties of cell-containing hydrogels. We also describe an enzymatic approach for the digestion of cell-containing hydrogels and cell recovery thereby maintaining high cell viability for subsequent analysis.
- Published
- 2019
- Full Text
- View/download PDF
33. Humanization of the Prostate Microenvironment Reduces Homing of PC3 Prostate Cancer Cells to Human Tissue-Engineered Bone.
- Author
-
McGovern JA, Shafiee A, Wagner F, Lahr CA, Landgraf M, Meinert C, Williams ED, Russell PJ, Clements JA, Loessner D, Holzapfel BM, Risbridger GP, and Hutmacher DW
- Abstract
The primary tumor microenvironment is inherently important in prostate cancer (PCa) initiation, growth and metastasis. However, most current PCa animal models are based on the injection of cancer cells into the blood circulation and bypass the first steps of the metastatic cascade, hence failing to investigate the influence of the primary tumor microenvironment on PCa metastasis. Here, we investigated the spontaneous metastasis of PC3 human PCa cells from humanized prostate tissue, containing cancer-associated fibroblasts (CAFs) and prostate lymphatic and blood vessel endothelial cells (BVEC), to humanized tissue-engineered bone constructs (hTEBC) in NOD-SCID IL2Rγ
null (NSG) mice. The hTEBC formed a physiologically mature organ bone which allowed homing of metastatic PCa cells. Humanization of prostate tissue had no significant effect on the tumor burden at the primary site over the 4 weeks following intraprostatic injection, yet reduced the incidence and burden of metastases in the hTEBC. Spontaneous PCa metastases were detected in the lungs and spleen with no significant differences between the humanized and non-humanized prostate groups. A significantly greater metastatic tumor burden was observed in the liver when metastasis occurred from the humanized prostate. Together, our data suggests that the presence of human-derived CAFs and BVECs in the primary PCa microenvironment influences selectively the metastatic and homing behavior of PC3 cells in this model. Our orthotopic and humanized prostate cancer model developed via convergence of cancer research and tissue engineering concepts provides an important platform to study species-specific PCa bone metastasis and to develop and test therapeutic strategies., Competing Interests: C.M. is a founder, shareholder and director of GELOMICS PTY Ltd, a start-up company developing and distributing hydrogels for 3D cell culture applications. D.W.H. is a founder and shareholder of GELOMICS PTY Ltd. All other authors declare no competing interests.- Published
- 2018
- Full Text
- View/download PDF
34. Kallikrein-related peptidases 4, 5, 6 and 7 regulate tumour-associated factors in serous ovarian cancer.
- Author
-
Wang P, Magdolen V, Seidl C, Dorn J, Drecoll E, Kotzsch M, Yang F, Schmitt M, Schilling O, Rockstroh A, Clements JA, and Loessner D
- Subjects
- Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Cell Line, Tumor, Cystadenocarcinoma, Serous genetics, Female, Gene Expression Regulation, Neoplastic, Humans, Ovarian Neoplasms genetics, Prognosis, Cystadenocarcinoma, Serous metabolism, Gene Expression Profiling methods, Gene Regulatory Networks, Kallikreins genetics, Ovarian Neoplasms metabolism, Proteomics methods
- Abstract
Background: Tissue kallikrein-related peptidases 4, 5, 6 and 7 (KLK4-7) strongly increase the malignancy of ovarian cancer cells. Deciphering their downstream effectors, we aimed at finding new potential prognostic biomarkers and treatment targets for ovarian cancer patients. KLK4-7-transfected (OV-KLK4-7) and vector-control OV-MZ-6 (OV-VC) ovarian cancer cells were established to select differentially regulated factors., Methods: With three independent approaches, PCR arrays, genome-wide microarray and proteome analyses, we identified 10 candidates (MSN, KRT19, COL5A2, COL1A2, BMP5, F10, KRT7, JUNB, BMP4, MMP1). To determine differential protein expression, we performed western blot analyses, immunofluorescence and immunohistochemistry for four candidates (MSN, KRT19, KRT7, JUNB) in cells, tumour xenograft and patient-derived tissues., Results: We demonstrated that KLK4-7 clearly regulates expression of MSN, KRT19, KRT7 and JUNB at the mRNA and protein levels in ovarian cancer cells and tissues. Protein expression of the top-upregulated effectors, MSN and KRT19, was investigated by immunohistochemistry in patients afflicted with serous ovarian cancer and related to KLK4-7 immunoexpression. Significant positive associations were found for KRT19/KLK4, KRT19/KLK5 and MSN/KLK7., Conclusion: These findings imply that KLK4-7 exert key modulatory effects on other cancer-related genes and proteins in ovarian cancer. These downstream effectors of KLK4-7, MSN and KRT19 may represent important therapeutic targets in serous ovarian cancer.
- Published
- 2018
- Full Text
- View/download PDF
35. Kallikrein-related peptidases represent attractive therapeutic targets for ovarian cancer.
- Author
-
Loessner D, Goettig P, Preis S, Felber J, Bronger H, Clements JA, Dorn J, and Magdolen V
- Subjects
- Animals, Biomarkers, Tumor metabolism, Drug Development methods, Female, Humans, Molecular Targeted Therapy, Ovarian Neoplasms pathology, Prognosis, Tumor Microenvironment, Antineoplastic Agents pharmacology, Kallikreins metabolism, Ovarian Neoplasms drug therapy
- Abstract
Introduction: Aberrant levels of kallikrein-related peptidases (KLK1-15) have been linked to cancer cell proliferation, invasion and metastasis. In ovarian cancer, the KLK proteolytic network has a crucial role in the tissue and tumor microenvironment. Publically available ovarian cancer genome and expression data from multiple patient cohorts show an upregulation of most KLKs. Areas covered: Here, we review the expression levels of all 15 members of this family in normal and ovarian cancer tissues, categorizing them into highly and moderately or weakly expressed KLKs, and their association with patient prognosis and survival. We summarize their tumor-biological functions determined in cell-based assays and xenograft models, further highlighting their suitability as cancer biomarkers and attractive candidates for drug development. Finally, we discuss some different pharmaceutical approaches, including peptide-based and small molecule inhibitors, cyclic peptides, depsipeptides, engineered natural inhibitors, antibodies, RNA/DNA-based aptamers, prodrugs, miRNA and siRNA. Expert opinion: In light of the results from clinical and tumor-biological studies, together with the available pharmaceutical tools, we suggest KLK4, KLK5, KLK6 and possibly KLK7 as preferred targets for inhibition in ovarian cancer.
- Published
- 2018
- Full Text
- View/download PDF
36. Humanization of bone and bone marrow in an orthotopic site reveals new potential therapeutic targets in osteosarcoma.
- Author
-
Wagner F, Holzapfel BM, McGovern JA, Shafiee A, Baldwin JG, Martine LC, Lahr CA, Wunner FM, Friis T, Bas O, Boxberg M, Prodinger PM, Shokoohmand A, Moi D, Mazzieri R, Loessner D, and Hutmacher DW
- Subjects
- Animals, Antigens, CD34 metabolism, Biomarkers, Tumor metabolism, Disease Models, Animal, Female, Human Umbilical Vein Endothelial Cells metabolism, Humans, Mesenchymal Stem Cells cytology, Mice, Minimally Invasive Surgical Procedures, Neovascularization, Physiologic, Regenerative Medicine, Tissue Engineering, Xenograft Model Antitumor Assays, Bone Marrow pathology, Bone and Bones pathology, Molecular Targeted Therapy, Osteosarcoma therapy
- Abstract
Background: Existing preclinical murine models often fail to predict effects of anti-cancer drugs. In order to minimize interspecies-differences between murine hosts and human bone tumors of in vivo xenograft platforms, we tissue-engineered a novel orthotopic humanized bone model., Methods: Orthotopic humanized tissue engineered bone constructs (ohTEBC) were fabricated by 3D printing of medical-grade polycaprolactone scaffolds, which were seeded with human osteoblasts and embedded within polyethylene glycol-based hydrogels containing human umbilical vein endothelial cells (HUVECs). Constructs were then implanted at the femur of NOD-scid and NSG mice. NSG mice were then bone marrow transplanted with human CD34
+ cells. Human osteosarcoma (OS) growth was induced within the ohTEBCs by direct injection of Luc-SAOS-2 cells. Tissues were harvested for bone matrix and marrow morphology analysis as well as tumor biology investigations. Tumor marker expression was analyzed in the humanized OS and correlated with the expression in 68 OS patients utilizing tissue micro arrays (TMA)., Results: After harvesting the femurs micro computed tomography and immunohistochemical staining showed an organ, which had all features of human bone. Around the original mouse femur new bone trabeculae have formed surrounded by a bone cortex. Staining for human specific (hs) collagen type-I (hs Col-I) showed human extracellular bone matrix production. The presence of nuclei staining positive for human nuclear mitotic apparatus protein 1 (hs NuMa) proved the osteocytes residing within the bone matrix were of human origin. Flow cytometry verified the presence of human hematopoietic cells. After injection of Luc-SAOS-2 cells a primary tumor and lung metastasis developed. After euthanization histological analysis showed pathognomic features of osteoblastic OS. Furthermore, the tumor utilized the previously implanted HUVECS for angiogenesis. Tumor marker expression was similar to human patients. Moreover, the recently discovered musculoskeletal gene C12orf29 was expressed in the most common subtypes of OS patient samples., Conclusion: OhTEBCs represent a suitable orthotopic microenvironment for humanized OS growth and offers a new translational direction, as the femur is the most common location of OS. The newly developed and validated preclinical model allows controlled and predictive marker studies of primary bone tumors and other bone malignancies., (Copyright © 2018 Elsevier Ltd. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
37. A Method for Prostate and Breast Cancer Cell Spheroid Cultures Using Gelatin Methacryloyl-Based Hydrogels.
- Author
-
Meinert C, Theodoropoulos C, Klein TJ, Hutmacher DW, and Loessner D
- Subjects
- Cell Culture Techniques, Cell Line, Tumor, Cell Proliferation, Cell Survival, Female, Humans, Male, Methacrylates chemistry, Tumor Cells, Cultured, Breast Neoplasms pathology, Gelatin chemistry, Hydrogels chemistry, Prostatic Neoplasms pathology, Spheroids, Cellular
- Abstract
Modern tissue engineering technologies have delivered tools to recreate a cell's naturally occurring niche in vitro and to investigate normal and pathological cell-cell and cell-niche interactions. Hydrogel biomaterials mimic crucial properties of native extracellular matrices, including mechanical support, cell adhesion sites and proteolytic degradability. As such, they are applied as 3D cell culture platforms to replicate tissue-like architectures observed in vivo, allowing physiologically relevant cell behaviors. Here we review bioengineered 3D approaches used for prostate and breast cancer. Furthermore, we describe the synthesis and use of gelatin methacryloyl-based hydrogels as in vitro 3D cancer model. This platform is used to engineer the microenvironments for prostate and breast cancer cells to study processes regulating spheroid formation, cell functions and responses to therapeutic compounds. Collectively, these bioengineered 3D approaches provide cell biologists with innovative pre-clinical tools that integrate the complexity of the disease seen in patients to advance our knowledge of cancer cell physiology and the contribution of a tumor's surrounding milieu.
- Published
- 2018
- Full Text
- View/download PDF
38. Lycopene reduces ovarian tumor growth and intraperitoneal metastatic load.
- Author
-
Holzapfel NP, Shokoohmand A, Wagner F, Landgraf M, Champ S, Holzapfel BM, Clements JA, Hutmacher DW, and Loessner D
- Abstract
Mutagens like oxidants cause lesions in the DNA of ovarian and fallopian tube epithelial cells, resulting in neoplastic transformation. Reduced exposure of surface epithelia to oxidative stress may prevent the onset or reduce the growth of ovarian cancer. Lycopene is well-known for its excellent antioxidant properties. In this study, the potential of lycopene in the prevention and treatment of ovarian cancer was investigated using an intraperitoneal animal model. Lycopene prevention significantly reduced the metastatic load of ovarian cancer-bearing mice, whereas treatment of already established ovarian tumors with lycopene significantly diminished the tumor burden. Lycopene treatment synergistically enhanced anti-tumorigenic effects of paclitaxel and carboplatin. Immunostaining of tumor and metastatic tissues for Ki67 revealed that lycopene reduced the number of proliferating cancer cells. Lycopene decreased the expression of the ovarian cancer biomarker, CA125. The anti-metastatic and anti-proliferative effects were accompanied by down-regulated expression of ITGA5, ITGB1, MMP9, FAK, ILK and EMT markers, decreased protein expression of integrin α5 and reduced activation of MAPK. These findings indicate that lycopene interferes with mechanisms involved in the development and progression of ovarian cancer and that its preventive and therapeutic use, combined with chemotherapeutics, reduces the tumor and metastatic burden of ovarian cancer in vivo ., Competing Interests: None.
- Published
- 2017
39. Endosteal-like extracellular matrix expression on melt electrospun written scaffolds.
- Author
-
Muerza-Cascante ML, Shokoohmand A, Khosrotehrani K, Haylock D, Dalton PD, Hutmacher DW, and Loessner D
- Subjects
- Cell Proliferation, Cell Survival, Cells, Cultured, Equipment Design, Female, Hematopoietic Stem Cells physiology, Hot Temperature, Humans, Male, Printing, Three-Dimensional, Tissue Engineering instrumentation, Tissue Engineering methods, Biomimetic Materials chemical synthesis, Bone Substitutes chemical synthesis, Electroplating methods, Extracellular Matrix chemistry, Hematopoietic Stem Cells cytology, Tissue Scaffolds
- Abstract
Tissue engineering technology platforms constitute a unique opportunity to integrate cells and extracellular matrix (ECM) proteins into scaffolds and matrices that mimic the natural microenvironment in vitro. The development of tissue-engineered 3D models that mimic the endosteal microenvironment enables researchers to discover the causes and improve treatments for blood and immune-related diseases. The aim of this study was to establish a physiologically relevant in vitro model using 3D printed scaffolds to assess the contribution of human cells to the formation of a construct that mimics human endosteum. Melt electrospun written scaffolds were used to compare the suitability of primary human osteoblasts (hOBs) and placenta-derived mesenchymal stem cells (plMSCs) in (non-)osteogenic conditions and with different surface treatments. Using osteogenic conditions, hOBs secreted a dense ECM with enhanced deposition of endosteal proteins, such as fibronectin and vitronectin, and osteogenic markers, such as osteopontin and alkaline phosphatase, compared to plMSCs. The expression patterns of these proteins were reproducibly identified in hOBs derived from three individual donors. Calcium phosphate-coated scaffolds induced the expression of osteocalcin by hOBs when maintained in osteogenic conditions. The tissue-engineered endosteal microenvironment supported the growth and migration of primary human haematopoietic stem cells (HSCs) when compared to HSCs maintained using tissue culture plastic. This 3D testing platform represents an endosteal bone-like tissue and warrants future investigation for the maintenance and expansion of human HSCs., Statement of Significance: This work is motivated by the recent interest in melt electrospinning writing, a 3D printing technique used to produce porous scaffolds for biomedical applications in regenerative medicine. Our team has been among the pioneers in building a new class of melt electrospinning devices for scaffold-based tissue engineering. These scaffolds allow structural support for various cell types to invade and deposit their own ECM, mimicking a characteristic 3D microenvironment for experimental studies. We used melt electrospun written polycaprolactone scaffolds to develop an endosteal bone-like tissue that promotes the growth of HSCs. We combine tissue engineering concepts with cell biology and stem cell research to design a physiologically relevant niche that is of prime interest to the scientific community., (Copyright © 2016 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
40. The distribution of the apparent diffusion coefficient as an indicator of the response to chemotherapeutics in ovarian tumour xenografts.
- Author
-
Tourell MC, Shokoohmand A, Landgraf M, Holzapfel NP, Poh PS, Loessner D, and Momot KI
- Subjects
- Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Carboplatin pharmacology, Carboplatin therapeutic use, Diffusion Magnetic Resonance Imaging, Drug Therapy, Combination, Elastic Modulus, Extracellular Matrix metabolism, Extracellular Matrix Proteins genetics, Extracellular Matrix Proteins metabolism, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Mice, Mice, Inbred NOD, Mice, SCID, Ovarian Neoplasms drug therapy, Ovarian Neoplasms metabolism, Paclitaxel pharmacology, Paclitaxel therapeutic use, Statistics, Nonparametric, Transplantation, Heterologous, Antineoplastic Agents metabolism, Carboplatin metabolism, Ovarian Neoplasms pathology, Paclitaxel metabolism
- Abstract
Diffusion-weighted magnetic resonance imaging (DW-MRI) was used to evaluate the effects of single-agent and combination treatment regimens in a spheroid-based animal model of ovarian cancer. Ovarian tumour xenografts grown in non-obese diabetic/severe-combined-immunodeficiency (NOD/SCID) mice were treated with carboplatin or paclitaxel, or combination carboplatin/paclitaxel chemotherapy regimens. After 4 weeks of treatment, tumours were extracted and underwent DW-MRI, mechanical testing, immunohistochemical and gene expression analyses. The distribution of the apparent diffusion coefficient (ADC) exhibited an upward shift as a result of each treatment regimen. The 99-th percentile of the ADC distribution ("maximum ADC") exhibited a strong correlation with the tumour size (r
2 = 0.90) and with the inverse of the elastic modulus (r2 = 0.96). Single-agent paclitaxel (n = 5) and combination carboplatin/paclitaxel (n = 2) treatment regimens were more effective in inducing changes in regions of higher cell density than single-agent carboplatin (n = 3) or the no-treatment control (n = 5). The maximum ADC was a good indicator of treatment-induced cell death and changes in the extracellular matrix (ECM). Comparative analysis of the tumours' ADC distribution, mechanical properties and ECM constituents provides insights into the molecular and cellular response of the ovarian tumour xenografts to chemotherapy. Increased sample sizes are recommended for future studies. We propose experimental approaches to evaluation of the timeline of the tumour's response to treatment.- Published
- 2017
- Full Text
- View/download PDF
41. Prostate Cancer-Associated Kallikrein-Related Peptidase 4 Activates Matrix Metalloproteinase-1 and Thrombospondin-1.
- Author
-
Fuhrman-Luck RA, Stansfield SH, Stephens CR, Loessner D, and Clements JA
- Subjects
- Bone Neoplasms chemistry, Bone Neoplasms metabolism, Bone Neoplasms secondary, Disease Progression, Humans, Male, Prostatic Neoplasms chemistry, Proteolysis, Kallikreins physiology, Matrix Metalloproteinase 1 metabolism, Prostatic Neoplasms pathology, Thrombospondin 1 metabolism
- Abstract
Prostate cancer metastasis to bone is terminal; thus, novel therapies are required to prevent end-stage disease. Kallikrein-related peptidase 4 (KLK4) is a serine protease that is overproduced in localized prostate cancer and is abundant in prostate cancer bone metastases. In vitro, KLK4 induces tumor-promoting phenotypes; however, the underlying proteolytic mechanism is undefined. The protein topography and migration analysis platform (PROTOMAP) was used for high-depth identification of KLK4 substrates secreted by prostate cancer bone metastasis-derived PC-3 cells to delineate the mechanism of KLK4 action in advanced prostate cancer. Thirty-six putative novel substrates were determined from the PROTOMAP analysis. In addition, KLK4 cleaved the established substrate, urokinase-type plasminogen activator, thus validating the approach. KLK4 activated matrix metalloproteinase-1 (MMP1), a protease that promotes prostate tumor growth and metastasis. MMP1 was produced in the tumor compartment of prostate cancer bone metastases, highlighting its accessibility to KLK4 at this site. KLK4 further liberated an N-terminal product, with purported angiogenic activity, from thrombospondin-1 (TSP1) and cleaved TSP1 in an osteoblast-derived matrix. This is the most comprehensive analysis of the proteolytic action of KLK4 in an advanced prostate cancer model to date, highlighting KLK4 as a potential multifunctional regulator of prostate cancer progression.
- Published
- 2016
- Full Text
- View/download PDF
42. A Validated Preclinical Animal Model for Primary Bone Tumor Research.
- Author
-
Wagner F, Holzapfel BM, Thibaudeau L, Straub M, Ling MT, Grifka J, Loessner D, Lévesque JP, and Hutmacher DW
- Subjects
- Animals, Bone Neoplasms metabolism, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Mice, Mice, Inbred NOD, Vascular Endothelial Growth Factor A metabolism, Bone Morphogenetic Protein 7 metabolism, Bone Neoplasms pathology, Disease Models, Animal
- Abstract
Background: Despite the introduction of 21st-century surgical and neoadjuvant treatment modalities, survival of patients with osteosarcoma (OS) has not improved in two decades. Advances will depend in part on the development of clinically relevant and reliable animal models. This report describes the engineering and validation of a humanized tissue-engineered bone organ (hTEBO) for preclinical research on primary bone tumors in order to minimize false-positive and false-negative results due to interspecies differences in current xenograft models., Methods: Pelvic bone and marrow fragments were harvested from patients during reaming of the acetabulum during hip arthroplasty. HTEBOs were engineered by embedding fragments in a fibrin matrix containing bone morphogenetic protein-7 (BMP-7) and implanted into NOD-scid mice. After 10 weeks of subcutaneous growth, one group of hTEBOs was harvested to analyze the degree of humanization. A second group was injected with human luciferase-labeled OS (Luc-SAOS-2) cells. Tumor growth was followed in vivo with bioluminescence imaging. After 5 weeks, the OS tumors were harvested and analyzed. They were also compared with tumors created via intratibial injection., Results: After 10 weeks of in vivo growth, a new bone organ containing human bone matrix as well as viable and functional human hematopoietic cells developed. Five weeks after injection of Luc-SAOS-2 cells into this humanized bone microenvironment, spontaneous metastatic spread to the lung was evident. Relevant prognostic markers such as vascular endothelial growth factor (VEGF) and periostin were found to be positive in OS tumors grown within the humanized microenvironment but not in tumors created in murine tibial bones. Hypoxia-inducible transcription factor-2α (HIF-2α) was detected only in the humanized OS., Conclusions: We report an in vivo model that contains human bone matrix and marrow components in one organ. BMP-7 made it possible to maintain viable mesenchymal and hematopoietic stem cells and created a bone microenvironment mimicking human physiology., Clinical Relevance: This novel platform enables preclinical research on primary bone tumors in order to test new treatment options., (Copyright © 2016 by The Journal of Bone and Joint Surgery, Incorporated.)
- Published
- 2016
- Full Text
- View/download PDF
43. Functionalization, preparation and use of cell-laden gelatin methacryloyl-based hydrogels as modular tissue culture platforms.
- Author
-
Loessner D, Meinert C, Kaemmerer E, Martine LC, Yue K, Levett PA, Klein TJ, Melchels FP, Khademhosseini A, and Hutmacher DW
- Subjects
- Animals, Humans, Tissue Engineering methods, Biopolymers, Gelatin, Hydrogels chemistry, Methacrylates, Tissue Culture Techniques methods, Tissue Scaffolds chemistry
- Abstract
Progress in advancing a system-level understanding of the complexity of human tissue development and regeneration is hampered by a lack of biological model systems that recapitulate key aspects of these processes in a physiological context. Hence, growing demand by cell biologists for organ-specific extracellular mimics has led to the development of a plethora of 3D cell culture assays based on natural and synthetic matrices. We developed a physiological microenvironment of semisynthetic origin, called gelatin methacryloyl (GelMA)-based hydrogels, which combine the biocompatibility of natural matrices with the reproducibility, stability and modularity of synthetic biomaterials. We describe here a step-by-step protocol for the preparation of the GelMA polymer, which takes 1-2 weeks to complete, and which can be used to prepare hydrogel-based 3D cell culture models for cancer and stem cell research, as well as for tissue engineering applications. We also describe quality control and validation procedures, including how to assess the degree of GelMA functionalization and mechanical properties, to ensure reproducibility in experimental and animal studies.
- Published
- 2016
- Full Text
- View/download PDF
44. Lycopene's Effects on Cancer Cell Functions within Monolayer and Spheroid Cultures.
- Author
-
Holzapfel NP, Holzapfel BM, Theodoropoulos C, Kaemmerer E, Rausch T, Feldthusen J, Champ S, Clements JA, Hutmacher DW, and Loessner D
- Subjects
- Breast Neoplasms drug therapy, Breast Neoplasms pathology, Cell Movement drug effects, Cell Proliferation drug effects, Cell Survival drug effects, Female, Humans, Lycopene, Male, Ovarian Neoplasms drug therapy, Ovarian Neoplasms pathology, Prostatic Neoplasms drug therapy, Prostatic Neoplasms pathology, Spheroids, Cellular, Tumor Cells, Cultured, Anticarcinogenic Agents pharmacology, Carotenoids pharmacology, Cell Culture Techniques methods, Neoplasms drug therapy, Neoplasms pathology
- Abstract
Lycopene, a compound that blocks the action of free radicals and oxygen molecules, is found in tomatoes and tomato-based products and linked to a reduced incidence of cancer. Increasing willingness of patients to maintain a healthy lifestyle by supplemental intake of nutrients and acceptance of alternative therapeutics has boosted research into nutraceuticals. The potential of lycopene to prevent or treat cancer has been investigated, but outcomes are inconsistent and its mode of action is still unknown. Further studies are needed to understand the role of lycopene in cancer prevention and treatment. The impact of lycopene on viability, proliferation, migration, and invasion of five different cancer cell lines was determined using monolayer and spheroid cultures. Cell viability was significantly reduced upon lycopene treatment at physiologically attainable concentrations. Cell proliferation, migration, and invasion did not change upon lycopene treatment. Ovarian cancer spheroids initially showed a decreased proliferation and after 14 days increased cell viability upon lycopene treatment, confirming the potential of lycopene to reduce cancer cell growth in short-term cultures and also indicate enhanced cell viability over prolonged exposure. This study cannot substantiate that lycopene inhibits cell functions associated with tumor growth, even in a 3D cancer model that mimics the natural tumor microenvironment.
- Published
- 2016
- Full Text
- View/download PDF
45. Assessment of kallikrein-related peptidase 5 (KLK5) protein expression in tumor tissue of advanced ovarian cancer patients by immunohistochemistry and ELISA: correlation with clinical outcome.
- Author
-
Dorn J, Yassouridis A, Walch A, Diamandis EP, Schmitt M, Kiechle M, Wang P, Drecoll E, Schmalfeldt B, Loessner D, Kotzsch M, and Magdolen V
- Abstract
Members of the human kallikrein-related peptidase (KLK) family, including KLK5, have been reported to play an important role in ovarian cancer progression. In the present study, we assessed KLK5 protein expression in ovarian cancer tissues by immunohistochemistry (IHC) and ELISA, and analyzed its association with clinicopathologic parameters and disease outcome in 95 patients with advanced ovarian cancer FIGO stage III/IV. KLK5 immunoexpression was evaluated in ovarian cancer tissue microarrays by IHC using a manual semiquantitative scoring system. KLK5 antigen levels were determined in ovarian cancer tumour tissue extracts by ELISA. KLK5 protein is expressed in ovarian cancer tissue by stromal and tumor cells. Mean KLK5 immunoscore values in tumor cells (KLK5-Tc; 5.7, range 0 to 12) were higher compared to stromal cells (KLK5-Sc; 1.2, range 0 to 9) but the correlation between KLK5-Tc and KLK5-Sc was rather low (rs = 0.34, P < 0.05). No significant associations of clinicopathological parameters with KLK5-Tc, KLK5-Sc, the combined overall score KLK5-Tc+Sc, or ELISA (KLK5-E) expression values were determined, except for KLK5-E protein expression with advanced age and high nuclear grade (G3). In univariate Cox regression analysis, elevated expression levels of KLK5-Sc are significantly linked with both prolonged overall survival (OS) (hazard ratio [HR] = 0.6, P = 0.046) and progression-free survival (PFS) (HR = 0.54, P = 0.032) of advanced ovarian cancer patients. KLK5-Tc and KLK5-Tc+Sc scores as well as the KLK5-E values were not associated with patients' outcome. In multivariable analysis, KLK5-Sc expression was found to be statistically significant for PFS. Patients with elevated KLK5-Sc had a two-fold lower risk of disease recurrence (HR = 0.53, P = 0.037) as compared to patients with low KLK5-Sc. For KLK5-Sc and OS, a trend towards statistical significance was observed (HR = 0.62, P = 0.077). These results indicate that KLK5 overexpression by stromal cells (KLK5-Sc) may be a positive modulator lowering aggressiveness of ovarian cancer.
- Published
- 2015
46. Convergence of regenerative medicine and synthetic biology to develop standardized and validated models of human diseases with clinical relevance.
- Author
-
Hutmacher DW, Holzapfel BM, De-Juan-Pardo EM, Pereira BA, Ellem SJ, Loessner D, and Risbridger GP
- Subjects
- Animals, Biomedical Research, Humans, Precision Medicine, Prostheses and Implants, Tissue Engineering methods, Regenerative Medicine methods, Synthetic Biology methods
- Abstract
In order to progress beyond currently available medical devices and implants, the concept of tissue engineering has moved into the centre of biomedical research worldwide. The aim of this approach is not to replace damaged tissue with an implant or device but rather to prompt the patient's own tissue to enact a regenerative response by using a tissue-engineered construct to assemble new functional and healthy tissue. More recently, it has been suggested that the combination of Synthetic Biology and translational tissue-engineering techniques could enhance the field of personalized medicine, not only from a regenerative medicine perspective, but also to provide frontier technologies for building and transforming the research landscape in the field of in vitro and in vivo disease models., (Crown Copyright © 2015. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
47. Tissue engineered humanized bone supports human hematopoiesis in vivo.
- Author
-
Holzapfel BM, Hutmacher DW, Nowlan B, Barbier V, Thibaudeau L, Theodoropoulos C, Hooper JD, Loessner D, Clements JA, Russell PJ, Pettit AR, Winkler IG, and Levesque JP
- Subjects
- Animals, Bone Substitutes chemical synthesis, Cell Differentiation physiology, Cells, Cultured, Equipment Design, Equipment Failure Analysis, Female, Humans, Mesenchymal Stem Cells physiology, Mice, Mice, Inbred C57BL, Osteoblasts physiology, Osteogenesis physiology, Stem Cell Niche physiology, Tissue Engineering instrumentation, Tissue Engineering methods, Bioartificial Organs, Bone Development physiology, Hematopoiesis physiology, Mesenchymal Stem Cells cytology, Osteoblasts cytology, Tissue Scaffolds
- Abstract
Advances in tissue-engineering have resulted in a versatile tool-box to specifically design a tailored microenvironment for hematopoietic stem cells (HSCs) in order to study diseases that develop within this setting. However, most current in vivo models fail to recapitulate the biological processes seen in humans. Here we describe a highly reproducible method to engineer humanized bone constructs that are able to recapitulate the morphological features and biological functions of the HSC niches. Ectopic implantation of biodegradable composite scaffolds cultured for 4 weeks with human mesenchymal progenitor cells and loaded with rhBMP-7 resulted in the development of a chimeric bone organ including a large number of human mesenchymal cells which were shown to be metabolically active and capable of establishing a humanized microenvironment supportive of the homing and maintenance of human HSCs. A syngeneic mouse-to-mouse transplantation assay was used to prove the functionality of the tissue-engineered ossicles. We predict that the ability to tissue engineer a morphologically intact and functional large-volume bone organ with a humanized bone marrow compartment will help to further elucidate physiological or pathological interactions between human HSCs and their native niches., (Crown Copyright © 2015. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
48. Biomaterial science meets computational biology.
- Author
-
Hutmacher DW, Little JP, Pettet GJ, and Loessner D
- Subjects
- Animals, Computer Simulation, Humans, Cell Membrane physiology, Computational Biology methods, Extracellular Matrix physiology, Mechanotransduction, Cellular physiology, Models, Biological, Tissue Engineering methods
- Abstract
There is a pressing need for a predictive tool capable of revealing a holistic understanding of fundamental elements in the normal and pathological cell physiology of organoids in order to decipher the mechanoresponse of cells. Therefore, the integration of a systems bioengineering approach into a validated mathematical model is necessary to develop a new simulation tool. This tool can only be innovative by combining biomaterials science with computational biology. Systems-level and multi-scale experimental data are incorporated into a single framework, thus representing both single cells and collective cell behaviour. Such a computational platform needs to be validated in order to discover key mechano-biological factors associated with cell-cell and cell-niche interactions.
- Published
- 2015
- Full Text
- View/download PDF
49. Engineered microenvironments provide new insights into ovarian and prostate cancer progression and drug responses.
- Author
-
Loessner D, Holzapfel BM, and Clements JA
- Subjects
- Animals, Antineoplastic Agents pharmacology, Biomimetics, Cell Culture Techniques, Disease Progression, Female, Humans, Male, Models, Biological, Ovarian Neoplasms drug therapy, Prostatic Neoplasms drug therapy, Tumor Microenvironment physiology, Ovarian Neoplasms pathology, Prostatic Neoplasms pathology, Tissue Engineering methods
- Abstract
Tissue engineering technologies, which have originally been designed to reconstitute damaged tissue structure and function, can mimic not only tissue regeneration processes but also cancer development and progression. Bioengineered approaches allow cell biologists to develop sophisticated experimentally and physiologically relevant cancer models to recapitulate the complexity of the disease seen in patients. Tissue engineering tools enable three-dimensionality based on the design of biomaterials and scaffolds that re-create the geometry, chemistry, function and signalling milieu of the native tumour microenvironment. Three-dimensional (3D) microenvironments, including cell-derived matrices, biomaterial-based cell culture models and integrated co-cultures with engineered stromal components, are powerful tools to study dynamic processes like proteolytic functions associated with cancer progression, metastasis and resistance to therapeutics. In this review, we discuss how biomimetic strategies can reproduce a humanised niche for human cancer cells, such as peritoneal or bone-like microenvironments, addressing specific aspects of ovarian and prostate cancer progression and therapy response., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
50. Kallikrein-Related Peptidases in Prostate Cancer: From Molecular Function to Clinical Application.
- Author
-
Fuhrman-Luck RA, Loessner D, and Clements JA
- Abstract
Prostate cancer is a leading contributor to male cancer-related deaths worldwide. Kallikrein-related peptidases (KLKs) are serine proteases that exhibit deregulated expression in prostate cancer, with KLK3, or prostate specific antigen (PSA), being the widely-employed clinical biomarker for prostate cancer. Other KLKs, such as KLK2, show promise as prostate cancer biomarkers and, additionally, their altered expression has been utilised for the design of KLK-targeted therapies. There is also a large body of in vitro and in vivo evidence supporting their role in cancer-related processes. Here, we review the literature on studies to date investigating the potential of other KLKs, in addition to PSA, as biomarkers and in therapeutic options, as well as their current known functional roles in cancer progression. Increased knowledge of these KLK-mediated functions, including degradation of the extracellular matrix, local invasion, cancer cell proliferation, interactions with fibroblasts, angiogenesis, migration, bone metastasis and tumour growth in vivo, may help define new roles as prognostic biomarkers and novel therapeutic targets for this cancer.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.