Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Laure Thibaudeau is active.

Publication


Featured researches published by Laure Thibaudeau.


Biointerphases | 2012

Design and fabrication of tubular scaffolds via direct writing in a melt electrospinning mode.

Toby D. Brown; Anna Slotosch; Laure Thibaudeau; Anna Taubenberger; Daniela Loessner; Cedryck Vaquette; Paul D. Dalton; Dietmar W. Hutmacher

Flexible tubular structures fabricated from solution electrospun fibers are finding increasing use in tissue engineering applications. However it is difficult to control the deposition of fibers due to the chaotic nature of the solution electrospinning jet. By using non-conductive polymer melts instead of polymer solutions the path and collection of the fiber becomes predictable. In this work we demonstrate the melt electrospinning of polycaprolactone in a direct writing mode onto a rotating cylinder. This allows the design and fabrication of tubes using 20 μm diameter fibers with controllable micropatterns and mechanical properties. A key design parameter is the fiber winding angle, where it allows control over scaffold pore morphology (e.g. size, shape, number and porosity). Furthermore, the establishment of a finite element model as a predictive design tool is validated against mechanical testing results of melt electrospun tubes to show that a lesser winding angle provides improved mechanical response to uniaxial tension and compression. In addition, we show that melt electrospun tubes support the growth of three different cell types in vitro and are therefore promising scaffolds for tissue engineering applications. Electronic supplementary material The online version of this article (doi:10.1007/s13758-011-0013-7) contains supplementary material, which is available to authorized users.


Biomaterials | 2014

Species-specific homing mechanisms of human prostate cancer metastasis in tissue engineered bone ☆

Boris Michael Holzapfel; Ferdinand Wagner; Daniela Loessner; Nina Pauline Holzapfel; Laure Thibaudeau; Ross Crawford; Ming-Tat Ling; Judith A. Clements; Pamela J. Russell; Dietmar W. Hutmacher

The development of effective therapeutic strategies against prostate cancer bone metastases has been impeded by the lack of adequate animal models that are able to recapitulate the biology of the disease in humans. Bioengineered approaches allow researchers to create sophisticated experimentally and physiologically relevant in vivo models to study interactions between cancer cells and their microenvironment under reproducible conditions. The aim of this study was to engineer a morphologically and functionally intact humanized organ bone which can serve as a homing site for human prostate cancer cells. Transplantation of biodegradable tubular composite scaffolds seeded with human mesenchymal progenitor cells and loaded with rhBMP-7 resulted in the development of a chimeric bone construct including a large number of human mesenchymal cells which were shown to be metabolically active and capable of producing extracellular matrix components. Micro-CT analysis demonstrated that the newly formed ossicle recapitulated the morphological features of a physiological organ bone with a trabecular network surrounded by a cortex-like outer structure. This microenvironment was supportive of the lodgement and maintenance of murine haematopoietic cell clusters, thus mimicking a functional organ bone. Bioluminescence imaging demonstrated that luciferase-transduced human PC3 cells reproducibly homed to the humanized tissue engineered bone constructs, proliferated, and developed macro-metastases. This model allows the analysis of interactions between human prostate cancer cells and a functional humanized bone organ within an immuno-incompetent murine host. The system can serve as a reproducible platform to study effects of therapeutics against prostate cancer bone metastases within a humanized microenvironment.


Stem Cells | 2015

Concise review: Humanized models of tumor immunology in the 21st century: Convergence of cancer research and tissue engineering

Boris Michael Holzapfel; Ferdinand Wagner; Laure Thibaudeau; Jean-Pierre Levesque; Dietmar W. Hutmacher

Despite positive testing in animal studies, more than 80% of novel drug candidates fail to proof their efficacy when tested in humans. This is primarily due to the use of preclinical models that are not able to recapitulate the physiological or pathological processes in humans. Hence, one of the key challenges in the field of translational medicine is to “make the model organism mouse more human.” To get answers to questions that would be prognostic of outcomes in human medicine, the mouses genome can be altered in order to create a more permissive host that allows the engraftment of human cell systems. It has been shown in the past that these strategies can improve our understanding of tumor immunology. However, the translational benefits of these platforms have still to be proven. In the 21st century, several research groups and consortia around the world take up the challenge to improve our understanding of how to humanize the animals genetic code, its cells and, based on tissue engineering principles, its extracellular microenvironment, its tissues, or entire organs with the ultimate goal to foster the translation of new therapeutic strategies from bench to bedside. This article provides an overview of the state of the art of humanized models of tumor immunology and highlights future developments in the field such as the application of tissue engineering and regenerative medicine strategies to further enhance humanized murine model systems. Stem Cells 2015;33:1696–1704


Cancer and Metastasis Reviews | 2013

Humanised xenograft models of bone metastasis revisited: novel insights into species-specific mechanisms of cancer cell osteotropism

Boris Michael Holzapfel; Laure Thibaudeau; Parisa Hesami; Anna Taubenberger; Nina Pauline Holzapfel; Susanne Mayer-Wagner; Carl A. Power; Judith A. Clements; Pamela J. Russell; Dietmar W. Hutmacher

The determinants and key mechanisms of cancer cell osteotropism have not been identified, mainly due to the lack of reproducible animal models representing the biological, genetic and clinical features seen in humans. An ideal model should be capable of recapitulating as many steps of the metastatic cascade as possible, thus facilitating the development of prognostic markers and novel therapeutic strategies. Most animal models of bone metastasis still have to be derived experimentally as most syngeneic and transgeneic approaches do not provide a robust skeletal phenotype and do not recapitulate the biological processes seen in humans. The xenotransplantation of human cancer cells or tumour tissue into immunocompromised murine hosts provides the possibility to simulate early and late stages of the human disease. Human bone or tissue-engineered human bone constructs can be implanted into the animal to recapitulate more subtle, species-specific aspects of the mutual interaction between human cancer cells and the human bone microenvironment. Moreover, the replication of the entire “organ” bone makes it possible to analyse the interaction between cancer cells and the haematopoietic niche and to confer at least a partial human immunity to the murine host. This process of humanisation is facilitated by novel immunocompromised mouse strains that allow a high engraftment rate of human cells or tissue. These humanised xenograft models provide an important research tool to study human biological processes of bone metastasis.


Journal of Bone and Mineral Research | 2013

Delineating breast cancer cell interactions with engineered bone microenvironments.

Anna Taubenberger; Verena M.C. Quent; Laure Thibaudeau; Judith A. Clements; Dietmar W. Hutmacher

The mechanisms leading to colonization of metastatic breast cancer cells (BCa) in the skeleton are still not fully understood. Here, we demonstrate that mineralized extracellular matrices secreted by primary human osteoblasts (hOBM) modulate cellular processes associated with BCa colonization of bone. A panel of four BCa cell lines of different bone‐metastatic potential (T47D, SUM1315, MDA‐MB‐231, and the bone‐seeking subline MDA‐MB‐231BO) was cultured on hOBM. After 3 days, the metastatic BCa cells had undergone morphological changes on hOBM and were aligned along the hOBMs collagen type I fibrils that were decorated with bone‐specific proteins. In contrast, nonmetastatic BCa cells showed a random orientation on hOBM. Atomic force microscopy‐based single‐cell force spectroscopy revealed that the metastatic cell lines adhered more strongly to hOBM compared with nonmetastatic cells. Function‐blocking experiments indicated that β1‐integrins mediated cell adhesion to hOBM. In addition, metastatic BCa cells migrated directionally and invaded hOBM, which was accompanied by enhanced MMP‐2 and ‐9 secretion. Furthermore, we observed gene expression changes associated with osteomimickry in BCa cultured on hOBM. As such, osteopontin mRNA levels were significantly increased in SUM1315 and MDA‐MB‐231BO cells in a β1‐integrin–dependent manner after growing for 3 days on hOBM compared with tissue culture plastic. In conclusion, our results show that extracellular matrices derived from human osteoblasts represent a powerful experimental platform to dissect mechanisms underlying critical steps in the development of bone metastases.


Journal of Bone and Joint Surgery, American Volume | 2016

A Validated Preclinical Animal Model for Primary Bone Tumor Research

Ferdinand Wagner; Boris Michael Holzapfel; Laure Thibaudeau; Melanie Straub; Ming-Tat Ling; Joachim Grifka; Daniela Loessner; Jean-Pierre Levesque; Dietmar W. Hutmacher

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.


Faculty of Health; Institute of Health and Biomedical Innovation; Science & Engineering Faculty | 2014

Species-specific homing mechanisms of human prostate cancer metastasis in tissue engineered bone

Boris Michael Holzapfel; Ferdinand Wagner; Daniela Loessner; Nina Pauline Holzapfel; Laure Thibaudeau; Ross Crawford; Ming-Tat Ling; Judith A. Clements; Pamela J. Russell; Dietmar W. Hutmacher


Biomaterials | 2015

Tissue engineered humanized bone supports human hematopoiesis in vivo

Boris Michael Holzapfel; Dietmar W. Hutmacher; Bianca Nowlan; Valerie Barbier; Laure Thibaudeau; Christina Theodoropoulos; John D. Hooper; Daniela Loessner; Judith A. Clements; Pamela J. Russell; Allison R. Pettit; Ingrid G. Winkler; Jean-Pierre Levesque


Cancer and Metastasis Reviews | 2014

Mimicking breast cancer-induced bone metastasis in vivo: current transplantation models and advanced humanized strategies

Laure Thibaudeau; Verena M.C. Quent; Boris Michael Holzapfel; Anna Taubenberger; Melanie Straub; Dietmar W. Hutmacher


Clinical & Experimental Metastasis | 2014

A humanized tissue-engineered in vivo model to dissect interactions between human prostate cancer cells and human bone

Parisa Hesami; Boris Michael Holzapfel; Anna Taubenberger; Martine Roudier; Ladan Fazli; Shirly Sieh; Laure Thibaudeau; Laura S. Gregory; Dietmar W. Hutmacher; Judith A. Clements

Collaboration


Dive into the Laure Thibaudeau's collaboration.

Top Co-Authors

Avatar

Dietmar W. Hutmacher

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Boris Michael Holzapfel

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Anna Taubenberger

Dresden University of Technology

View shared research outputs
Top Co-Authors

Avatar

Judith A. Clements

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Daniela Loessner

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Pamela J. Russell

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Verena M.C. Quent

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ming-Tat Ling

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Nina Pauline Holzapfel

Queensland University of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge