Network


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

Hotspot


Dive into the research topics where Carl O. Postenka is active.

Publication


Featured researches published by Carl O. Postenka.


Journal of Cellular and Molecular Medicine | 2009

High aldehyde dehydrogenase and expression of cancer stem cell markers selects for breast cancer cells with enhanced malignant and metastatic ability.

Alysha K. Croker; David Goodale; Jenny E. Chu; Carl O. Postenka; Benjamin D. Hedley; David A. Hess; Alison L. Allan

Cancer stem cells (CSCs) have recently been identified in leukaemia and solid tumours; however, the role of CSCs in metastasis remains poorly understood. This dearth of knowledge about CSCs and metastasis is due largely to technical challenges associated with the use of primary human cancer cells in pre‐clinical models of metastasis. Therefore, the objective of this study was to develop suitable pre‐clinical model systems for studying stem‐like cells in breast cancer metastasis, and to test the hypothesis that stem‐like cells play a key role in metastatic behaviour. We assessed four different human breast cancer cell lines (MDA‐MB‐435, MDA‐MB‐231, MDA‐MB‐468, MCF‐7) for expression of prospective CSC markers CD44/CD24 and CD133, and for functional activity of aldehyde dehydrogenase (ALDH), an enzyme involved in stem cell self‐protection. We then used fluorescence‐activated cell sorting and functional assays to characterize differences in malignant/metastatic behaviour in vitro (proliferation, colony‐forming ability, adhesion, migration, invasion) and in vivo (tumorigenicity and metastasis). Sub‐populations of cells demonstrating stem‐cell‐like characteristics (high expression of CSC markers and/or high ALDH) were identified in all cell lines except MCF‐7. When isolated and compared to ALDHlowCD44low/– cells, ALDHhiCD44+CD24− (MDA‐MB‐231) and ALDHhiCD44+CD133+ (MDA‐MB‐468) cells demonstrated increased growth (P < 0.05), colony formation (P < 0.05), adhesion (P < 0.001), migration (P < 0.001) and invasion (P < 0.001). Furthermore, following tail vein or mammary fat pad injection of NOD/SCID/IL2γ receptor null mice, ALDHhiCD44+CD24− and ALDHhiCD44+CD133+ cells showed enhanced tumorigenicity and metastasis relative to ALDHlowCD44low/– cells (P < 0.05). These novel results suggest that stem‐like ALDHhiCD44+CD24− and ALDHhiCD44+CD133+ cells may be important mediators of breast cancer metastasis.


Cell Reports | 2014

Invadopodia are required for cancer cell extravasation and are a therapeutic target for metastasis.

Hon S. Leong; Amy E. Robertson; Konstantin Stoletov; Sean J. Leith; Curtis A. Chin; Andrew E. Chien; M. Nicole Hague; Amber Ablack; Katia Carmine-Simmen; Victor A. McPherson; Carl O. Postenka; Eva A. Turley; Sara A. Courtneidge; Ann F. Chambers; John D. Lewis

Tumor cell extravasation is a key step during cancer metastasis, yet the precise mechanisms that regulate this dynamic process are unclear. We utilized a high-resolution time-lapse intravital imaging approach to visualize the dynamics of cancer cell extravasation in vivo. During intravascular migration, cancer cells form protrusive structures identified as invadopodia by their enrichment of MT1-MMP, cortactin, Tks4, and importantly Tks5, which localizes exclusively to invadopodia. Cancer cells extend invadopodia through the endothelium into the extravascular stroma prior to their extravasation at endothelial junctions. Genetic or pharmacological inhibition of invadopodia initiation (cortactin), maturation (Tks5), or function (Tks4) resulted in an abrogation of cancer cell extravasation and metastatic colony formation in an experimental mouse lung metastasis model. This provides direct evidence of a functional role for invadopodia during cancer cell extravasation and distant metastasis and reveals an opportunity for therapeutic intervention in this clinically important process.


Cancer Research | 2005

Three-dimensional High-Frequency Ultrasound Imaging for Longitudinal Evaluation of Liver Metastases in Preclinical Models

Kevin C. Graham; Lauren A. Wirtzfeld; Lisa T. MacKenzie; Carl O. Postenka; Alan C. Groom; Ian C. MacDonald; Aaron Fenster; James C. Lacefield; Ann F. Chambers

Liver metastasis is a clinically significant contributor to the mortality associated with melanoma, colon, and breast cancer. Preclinical mouse models are essential to the study of liver metastasis, yet their utility has been limited by the inability to study this dynamic process in a noninvasive and longitudinal manner. This study shows that three-dimensional high-frequency ultrasound can be used to noninvasively track the growth of liver metastases and evaluate potential chemotherapeutics in experimental liver metastasis models. Liver metastases produced by mesenteric vein injection of B16F1 (murine melanoma), PAP2 (murine H-ras-transformed fibroblast), HT-29 (human colon carcinoma), and MDA-MB-435/HAL (human breast carcinoma) cells were identified and tracked longitudinally. Tumor size and location were verified by histologic evaluation. Tumor volumes were calculated from the three-dimensional volumetric data, with individual liver metastases showing exponential growth. The importance of volumetric imaging to reduce uncertainty in tumor volume measurement was shown by comparing three-dimensional segmented volumes with volumes estimated from diameter measurements and the assumption of an ellipsoid shape. The utility of high-frequency ultrasound imaging in the evaluation of therapeutic interventions was established with a doxorubicin treatment trial. These results show that three-dimensional high-frequency ultrasound imaging may be particularly well suited for the quantitative assessment of metastatic progression and the evaluation of chemotherapeutics in preclinical liver metastasis models.


Cancer Research | 2005

Dietary Genistein Reduces Metastasis in a Postsurgical Orthotopic Breast Cancer Model

Sharon A. Vantyghem; Sylvia M. Wilson; Carl O. Postenka; Waleed Al-Katib; Alan B. Tuck; Ann F. Chambers

Metastatic spread, not primary tumor burden, is the leading cause of breast cancer deaths. For patient prognosis to improve, new systemic adjuvant therapies that are capable of effectively inhibiting the outgrowth of seeded tumor cells after surgical treatment of the primary breast tumor are needed. To facilitate the preclinical development of such therapies, relevant animal models of breast cancer metastasis that can mimic the postsurgical adjuvant setting are required. Here we developed a preclinical xenograft model of breast cancer metastasis where the primary tumor was removed by surgical resection before systemic adjuvant treatment. We used this model to assess the antimetastatic effect of postsurgical dietary intervention with the soy isoflavone genistein. The anticancer activity of genistein has been established in vitro and in vivo, however, few studies have tested the potential of genistein as an antimetastatic therapy. Using our model, we tested the efficacy of adjuvant treatment with genistein to inhibit the outgrowth of metastases postsurgery. To establish primary tumors, human breast carcinoma cells, MDA-MB-435/HAL, were implanted into the mammary fat pad of female nude mice. Primary tumors were left to grow for 5 weeks before being surgically removed. Mice were then randomized into two diet groups: control soy-free diet versus genistein-supplemented diet. Five weeks later, metastatic burden was assessed. Genistein reduced the percent metastatic burden in the lungs by 10-fold. These results indicate that dietary intervention following cancer surgery can affect the outgrowth of seeded tumor cells. The availability of well-characterized, clinically relevant animal models for studying factors that regulate metastatic outgrowth postsurgery will provide an important tool for developing new systemic adjuvant therapies.


Clinical & Experimental Metastasis | 2005

A new model for lymphatic metastasis : Development of a variant of the MDA-MB-468 human breast cancer cell line that aggressively metastasizes to lymph nodes

Sharon A. Vantyghem; Alison L. Allan; Carl O. Postenka; Waleed Al-Katib; Michael Keeney; Alan B. Tuck; Ann F. Chambers

Breast cancer often spreads from the primary tumor to regional lymph nodes. Lymph node status provides clinically important information for making treatment decisions. Spread via lymphatics is also important for the biology of breast cancer, as tumor cells in lymph nodes may provide a reservoir of cells leading to distant, lethal metastases. Improved understanding of the biology of lymphatic spread thus is important for improved breast cancer survival. Advances towards understanding the interactions between tumors cells and lymphatic vessels have in part been limited by the lack of suitable cell lines and experimental models. We have addressed this need by developing a new model of lymphatic metastasis. Here we describe the establishment of 468LN cells, a variant of the MDA-MB-468 human breast adenocarcinoma cell line, which produces extensive lymph node metastasis following orthotopic injection of nude mice. 468LN cells are also more aggressive in vitro, produce more osteopontin and express different surface integrins compared to the parent line. The dramatic in vitro and in vivo phenotypic and molecular differences of 468LN and parental 468GFP cells make this pair of cell lines a unique model for the specific study of lymph node metastasis of breast cancer.


Laboratory Investigation | 2011

Nuclear localization of maspin is essential for its inhibition of tumor growth and metastasis

Brigitte Goulet; Wendy Kennette; Amber Ablack; Carl O. Postenka; M. Nicole Hague; Joe S. Mymryk; Alan B. Tuck; Vincent Giguère; Ann F. Chambers; John D. Lewis

Maspin (mammary serine protease inhibitor or SerpinB5) acts as a tumor suppressor when overexpressed in aggressive cancer cell lines. However, its role in human cancer is controversial. Maspin expression has been associated with a poor prognosis in some studies, whereas in others, with favorable outcome. The clinical data suggest, however, that nuclear-localized maspin is associated with improved survival. We hypothesized that the tumor suppressor activity of maspin may require nuclear localization, and that the discordance between clinical and experimental reports is a consequence of the variable subcellular distribution of maspin. Furthermore, we surmized that nuclear maspin could function as a tumor suppressor through the regulation of genes involved in tumor growth and invasion. Maspin or maspin fused to a nuclear export signal were expressed in metastatic human breast and epidermoid carcinoma cell lines. We found that pan-cellular localized maspin inhibited in vivo tumor growth and metastasis when assessed in xenograft chicken embryo and murine mammary fat pad injection models. However, when maspin was excluded from the nucleus via a nuclear exclusion signal, it no longer functioned as a metastasis suppressor. Using chromatin immunoprecipitation, we show that nuclear maspin was enriched at the promoter of colony-stimulating factor-1 (CSF-1) and associated with diminished levels of CSF-1 mRNA. Our findings demonstrate that the nuclear localization of maspin is required for its tumor and metastasis suppressor functions in vivo, and suggest that its mechanism of action involves, in part, direct association of maspin with target genes.


Investigative Radiology | 2008

Noninvasive Quantification of Tumor Volume in Preclinical Liver Metastasis Models Using Contrast-enhanced X-ray Computed Tomography

Kevin C. Graham; Nancy L. Ford; Lisa T. MacKenzie; Carl O. Postenka; Alan C. Groom; Ian C. MacDonald; David W. Holdsworth; Maria Drangova; Ann F. Chambers

Objectives:To determine a timepoint after contrast injection that yields equal liver parenchymal and vascular enhancement in micro-computed tomography images. To evaluate the utility of images acquired during this time period for the noninvasive measurement of liver-tumor volume. Materials and Methods:The imaging timepoint was determined by quantifying the enhancement kinetics of Fenestra VC (0.015 mL/g) in NIH III mice. In respiratory-gated images of tumor bearing mice, the ability to measure tumor volume was evaluated with a measurement variability study, and by comparing in vivo and histologically measured tumor volume. Results:Eight hours after contrast injection the liver parenchyma and vasculature were equally enhanced allowing for clear delineation of the unenhanced tumors. The smallest tumor detected in this study was 1.1 mm in diameter. The coefficient of variation for tumor-volume measurement ranged from 3.6% to 12.9% and from 6.3% to 25.8% for intra and interobserver variability, respectively. In vivo and histologic tumor-volume measurements were closely correlated (r = 0.98, P < 0.0001). Conclusions:Imaging at a time period of equal liver parenchyma and vascular enhancement after contrast injection allows for clear delineation of liver-tumor borders, thereby enabling quantitative tumor-volume monitoring.


Cancer Biology & Therapy | 2005

Expression of osteopontin and HGF/Met in adult soft tissue tumors

Vivien Bramwell; Alan B. Tuck; Sylvia M. Wilson; Larry Stitt; A. Cherian; Stewart C. Rorke; Waleed Al-Katib; Carl O. Postenka; Ann F. Chambers

There is a need for molecular markers that predict biological behavior of adult soft tissue tumors. Elevated levels of osteopontin (OPN) a transformation-linked protein, have been associated with poor survival in many cancers. OPN induces cell migration in cancer cells, in part through activation of the hepatocyte growth factor (HGF) receptor (Met) and its signaling pathway. Met expression has been associated with a poor prognosis in some sarcomas. In a series of 15 patients with adult soft tissue tumors, we found that mRNA levels of OPN (p=0.015), Met (p=0.03) and HGF (p


Oncotarget | 2016

Epithelial-to-mesenchymal transition leads to disease-stage differences in circulating tumor cell detection and metastasis in pre-clinical models of prostate cancer

Lori E. Lowes; David Goodale; Ying Xia; Carl O. Postenka; Matthew M. Piaseczny; Freeman Paczkowski; Alison L. Allan

Metastasis is the cause of most prostate cancer (PCa) deaths and has been associated with circulating tumor cells (CTCs). The presence of ≥5 CTCs/7.5mL of blood is a poor prognosis indicator in metastatic PCa when assessed by the CellSearch® system, the “gold standard” clinical platform. However, ~35% of metastatic PCa patients assessed by CellSearch® have undetectable CTCs. We hypothesize that this is due to epithelial-to-mesenchymal transition (EMT) and subsequent loss of necessary CTC detection markers, with important implications for PCa metastasis. Two pre-clinical assays were developed to assess human CTCs in xenograft models; one comparable to CellSearch® (EpCAM-based) and one detecting CTCs semi-independent of EMT status via combined staining with EpCAM/HLA (human leukocyte antigen). In vivo differences in CTC generation, kinetics, metastasis and EMT status were determined using 4 PCa models with progressive epithelial (LNCaP, LNCaP-C42B) to mesenchymal (PC-3, PC-3M) phenotypes. Assay validation demonstrated that the CellSearch®-based assay failed to detect a significant number (~40-50%) of mesenchymal CTCs. In vivo, PCa with an increasingly mesenchymal phenotype shed greater numbers of CTCs more quickly and with greater metastatic capacity than PCa with an epithelial phenotype. Notably, the CellSearch®-based assay captured the majority of CTCs shed during early-stage disease in vivo, and only after establishment of metastases were a significant number of undetectable CTCs present. This study provides important insight into the influence of EMT on CTC generation and subsequent metastasis, and highlights that novel technologies aimed at capturing mesenchymal CTCs may only be useful in the setting of advanced metastatic disease.


Cancer Research | 2014

Abstract 4055: The transcriptional regulator TBX3 promotes progression of cells representing early premalignant breast cancer

Connor D. MacMillan; Hon S. Leong; Allen G. Clifford; Milica Krstic; Siddika Pardhan; David W. Dales; Carl O. Postenka; Ann F. Chambers; Alan B. Tuck

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CAnnThe transcriptional regulator TBX3 has been shown to be elevated in metastatic breast cancer and is thought to be involved in promoting malignancy of breast and other carcinomas. In a series of cell lines derived from the same patient with breast cancer, we previously showed that metastatic 21MT-1 cells express elevated levels of TBX3, while low levels are expressed by the ductal carcinoma in situ-like 21NT cells. Here we hypothesized that upregulation of TBX3 in 21NT cells would promote their progression to a more malignant phenotype. We found that 21NT cells engineered to overexpress either isoform of TBX3 (iso1, iso2) have an altered growth in 3D Matrigel, with increased colony-forming ability, higher number of cells per colony and less spherical, more irregular colonies, consistent with increased invasiveness. Transwell assays through Matrigel showed increased invasiveness of 21NT cells overexpressing either TBX3 isoform and in vivo assay in the chick embryo similarly showed increased propensity of TBX3 over-expressing 21NT cells for extravasation. These TBX3-induced changes were associated with increased vimentin expression, but minimal change in e-cadherin expression. Overall, these results indicate that TBX3 (iso1 or iso2) expression can promote progression in a model of early breast cancer, by altering cell properties involved in cell survival/colony formation and invasiveness.nnCitation Format: Connor D. MacMillan, Hon S. Leong, Allen G. Clifford, Milica Krstic, Siddika Pardhan, David W. Dales, Carl O. Postenka, Ann F. Chambers, Alan B. Tuck. The transcriptional regulator TBX3 promotes progression of cells representing early premalignant breast cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4055. doi:10.1158/1538-7445.AM2014-4055

Collaboration


Dive into the Carl O. Postenka's collaboration.

Top Co-Authors

Avatar

Ann F. Chambers

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Alan B. Tuck

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Waleed Al-Katib

London Health Sciences Centre

View shared research outputs
Top Co-Authors

Avatar

Alison L. Allan

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Sharon A. Vantyghem

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Sylvia M. Wilson

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Alan C. Groom

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Amber Ablack

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

David Goodale

London Health Sciences Centre

View shared research outputs
Top Co-Authors

Avatar

Hon S. Leong

London Health Sciences Centre

View shared research outputs
Researchain Logo
Decentralizing Knowledge