David E. Gyorki
Peter MacCallum Cancer Centre
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by David E. Gyorki.
Nature Medicine | 2009
Elgene Lim; François Vaillant; Di Wu; Natasha C. Forrest; Bhupinder Pal; Adam H. Hart; Marie-Liesse Asselin-Labat; David E. Gyorki; Teresa Ward; Audrey Partanen; Frank Feleppa; Lily I. Huschtscha; Heather Thorne; Stephen B. Fox; Max Yan; Juliet D. French; Melissa A. Brown; Gordon K. Smyth; Jane E. Visvader; Geoffrey J. Lindeman
Basal-like breast cancers arising in women carrying mutations in the BRCA1 gene, encoding the tumor suppressor protein BRCA1, are thought to develop from the mammary stem cell. To explore early cellular changes that occur in BRCA1 mutation carriers, we have prospectively isolated distinct epithelial subpopulations from normal mammary tissue and preneoplastic specimens from individuals heterozygous for a BRCA1 mutation. We describe three epithelial subsets including basal stem/progenitor, luminal progenitor and mature luminal cells. Unexpectedly, we found that breast tissue from BRCA1 mutation carriers harbors an expanded luminal progenitor population that shows factor-independent growth in vitro. Moreover, gene expression profiling revealed that breast tissue heterozygous for a BRCA1 mutation and basal breast tumors were more similar to normal luminal progenitor cells than any other subset, including the stem cell–enriched population. The c-KIT tyrosine kinase receptor (encoded by KIT) emerged as a key marker of luminal progenitor cells and was more highly expressed in BRCA1-associated preneoplastic tissue and tumors. Our findings suggest that an aberrant luminal progenitor population is a target for transformation in BRCA1-associated basal tumors .
Molecular and Cellular Biology | 2011
Marie-Liesse Asselin-Labat; Kate D. Sutherland; François Vaillant; David E. Gyorki; Di Wu; Sheridan L Holroyd; Kelsey Breslin; Teresa Ward; Wei Shi; Mary L. Bath; Siddhartha Deb; Stephen B. Fox; Gordon K. Smyth; Geoffrey J. Lindeman; Jane E. Visvader
ABSTRACT The transcription factor Gata-3 is a definitive marker of luminal breast cancers and a key regulator of mammary morphogenesis. Here we have explored a role for Gata-3 in tumor initiation and the underlying cellular mechanisms using a mouse model of “luminal-like” cancer. Loss of a single Gata-3 allele markedly accelerated tumor progression in mice carrying the mouse mammary tumor virus promoter-driven polyomavirus middle T antigen (MMTV-PyMT mice), while overexpression of Gata-3 curtailed tumorigenesis. Through the identification of two distinct luminal progenitor cells in the mammary gland, we demonstrate that Gata-3 haplo-insufficiency increases the tumor-initiating capacity of these progenitors but not the stem cell-enriched population. Overexpression of a conditional Gata-3 transgene in the PyMT model promoted cellular differentiation and led to reduced tumor-initiating capacity as well as diminished angiogenesis. Transcript profiling studies identified caspase-14 as a novel downstream target of Gata-3, in keeping with its roles in differentiation and tumorigenesis. A strong association was evident between GATA-3 and caspase-14 expression in preinvasive ductal carcinoma in situ samples, where GATA-3 also displayed prognostic significance. Overall, these studies identify GATA-3 as an important regulator of tumor initiation through its ability to promote the differentiation of committed luminal progenitor cells.
CA | 2017
Jeffrey E. Gershenwald; Richard A. Scolyer; Kenneth R. Hess; Vernon K. Sondak; Merrick I. Ross; Alexander J. Lazar; Mark B. Faries; John M. Kirkwood; Grant A. McArthur; Lauren E. Haydu; Alexander M.M. Eggermont; Keith T. Flaherty; Charles M. Balch; John F. Thompson; Michael B. Atkins; Raymond L. Barnhill; Karl Y. Bilimoria; Antonio C. Buzaid; David R. Byrd; Alistair J. Cochran; David E. Elder; Claus Garbe; Julie M. Gardner; Phyllis A. Gimotty; Allan C. Halpern; Timothy M. Johnson; Anne W. M. Lee; Martin C. Mihm; Victor G. Prieto; Arthur J. Sober
Answer questions and earn CME/CNE
Breast Cancer Research | 2009
David E. Gyorki; Marie-Liesse Asselin-Labat; Nico van Rooijen; Geoffrey J. Lindeman; Jane E. Visvader
IntroductionMacrophages in the mammary gland are essential for morphogenesis of the ductal epithelial tree and have been implicated in promoting breast tumor metastasis. Although it is well established that macrophages influence normal mammopoiesis, the mammary cell types that these accessory cells influence have not been determined. Here we have explored a role for macrophages in regulating mammary stem cell (MaSC) activity, by assessing the ability of MaSCs to reconstitute a mammary gland in a macrophage-depleted fat pad.MethodsTwo different in vivo models were used to deplete macrophages from the mouse mammary fat pad, allowing us to examine the effect of macrophage deficiency on the mammary repopulating activity of MaSCs. Both the Csf1op/opmice and clodronate liposome-mediated ablation models entailed transplantation studies using the MaSC-enriched population.ResultsWe show that mammary repopulating ability is severely compromised when the wild-type MaSC-enriched subpopulation is transplanted into Csf1op/opfat pads. In reciprocal experiments, the MaSC-enriched subpopulation from Csf1op/opglands had reduced regenerative capacity in a wild-type environment. Utilizing an alternative strategy for selective depletion of macrophages from the mammary gland, we demonstrate that co-implantation of the MaSC-enriched subpopulation with clodronate-liposomes leads to a marked decrease in repopulating frequency and outgrowth potential.ConclusionsOur data reveal a key role for mammary gland macrophages in supporting stem/progenitor cell function and suggest that MaSCs require macrophage-derived factors to be fully functional. Macrophages may therefore constitute part of the mammary stem cell niche.
Journal of Clinical Investigation | 2017
Paul A. Beavis; Melissa A. Henderson; Lauren Giuffrida; Jane Mills; Kevin Sek; Ryan S. Cross; Alexander J. Davenport; Liza B. John; Sherly Mardiana; Clare Y. Slaney; Ricky W. Johnstone; Joseph A. Trapani; John Stagg; Sherene Loi; Lev Kats; David E. Gyorki; Michael H. Kershaw; Phillip K. Darcy
Chimeric antigen receptor (CAR) T cells have been highly successful in treating hematological malignancies, including acute and chronic lymphoblastic leukemia. However, treatment of solid tumors using CAR T cells has been largely unsuccessful to date, partly because of tumor-induced immunosuppressive mechanisms, including adenosine production. Previous studies have shown that adenosine generated by tumor cells potently inhibits endogenous antitumor T cell responses through activation of adenosine 2A receptors (A2ARs). Herein, we have observed that CAR activation resulted in increased A2AR expression and suppression of both murine and human CAR T cells. This was reversible using either A2AR antagonists or genetic targeting of A2AR using shRNA. In 2 syngeneic HER2+ self-antigen tumor models, we found that either genetic or pharmacological targeting of the A2AR profoundly increased CAR T cell efficacy, particularly when combined with PD-1 blockade. Mechanistically, this was associated with increased cytokine production of CD8+ CAR T cells and increased activation of both CD8+ and CD4+ CAR T cells. Given the known clinical relevance of the CD73/adenosine pathway in several solid tumor types, and the initiation of phase I trials for A2AR antagonists in oncology, this approach has high translational potential to enhance CAR T cell efficacy in several cancer types.
Seminars in Immunology | 2016
Paul A. Beavis; Clare Y. Slaney; Michael H. Kershaw; David E. Gyorki; Paul Neeson; Phillip K. Darcy
The frontiers of cancer immunotherapy are extending in terms of both the range of cancer types that can potentially be targeted and the types of therapeutics that are in clinical development. The use of adoptive cellular therapy (ACT) and its derivative, chimeric antigen receptor (CAR) T cells, is currently limited to hematological malignancies and immunogenic cancers such as melanoma and renal cell carcinoma. Although ACT utilizing ex vivo expanded tumor-infiltrating lymphocytes (TIL) or engineered CAR/TCR T cells have undergone clinical trials for other solid cancers, their efficacy to date has been limited. This may be due, in part, to the immunosuppressive nature of the tumor microenvironment. The development of novel combination approaches which target the immunosuppressive network engineered by tumors has raised the possibility of using ACT for a broader range of cancers. This review summarizes the potential of such strategies and outlines the clinical relevance of these observations.
Nature Biotechnology | 2016
Kathryn Alsop; Heather Thorne; Shahneen Sandhu; Anne Hamilton; Christopher P. Mintoff; Elizabeth L. Christie; Odette Spruyt; Scott Williams; Orla McNally; Linda Mileshkin; Sumitra Ananda; Julene Hallo; Sherene Loi; Clare L. Scott; Peter Savas; Lisa Devereux; Patricia C. M. O'Brien; Sameera Gunawardena; Clare Hampson; Kate Strachan; Rufaro Diana Jaravaza; Victoria Francis; Gregory Young; David Ranson; Ravindra Samaranayake; David B. Stevens; Samantha E. Boyle; Clare G Fedele; Monique Topp; Gwo Ho
To the Editor: Systematic genomic studies, including the Cancer Genome Atlas (TCGA)1 and the International Cancer Genome Consortium (ICGC)2, have provided an unprecedented catalog of driver mutations in human cancer. However, these studies use mainly primary, pre-treatment tumor material obtained at surgery with curative intent. There is an urgent need to identify and characterize resistance mechanisms to understand how cancers can evade even the best medical efforts and kill patients; therefore, access to end-stage disease is important. Solid cancers show considerable spatial3, temporal4,5 and genomic heterogeneity at diagnosis. Selective pressure and mutagenic impact of treatment6 drives intra-patient evolution of cancer cell populations4,7. Understanding acquired resistance requires access to paired preand post-treatment samples4,7; however, curative surgery is typically confined to patients with locoregional disease, and opportunities for tumor sampling in advanced disseminated disease are limited. Here, we describe Cancer Tissue Collection After Death (CASCADE), an autopsy program that overcomes logistical challenges to enable collection of samples at end stage for research in melanoma and breast, ovarian and prostate cancers. For the CASCADE study, we aimed to recruit cancer patients close to the end of life, including those outside the minority of patients who die in hospitals. To preserve tissue integrity, autopsies must commence within a few hours of death, requiring access to around-the-clock services. Intervention in the emotionally charged end-of-life environment must be managed in an ethical manner and to a high standard. Finally, we aimed for the study to be highly cost-effective. We believe our approach to meeting these challenges is applicable to researchers in other large urban centers. Here we summarize the main steps in CASCADE’s operating protocol and our experiences from the initial 3 years and 30 autopsies performed (Fig. 1). Information about institutional review board approvals (including a detailed patient informationand-consent form), the autopsy procedure and certain laboratory processes is given in Supplementary Methods and Supplementary Figure 1. Recruitment of participants was led by the clinicians. Such discussions require careful consideration, in timing and in language, and were initiated only if there was a perception that tissue donation would be acceptable to the patients and their families. Factors suggesting acceptability include the emotional stability of the participant and family members and their clarity about and acceptance of the terminal nature of the disease. On occasion, participants prompted discussion by asking about organ or body donation. Consent discussions typically involved oncologists and/or palliative care physicians employed at recruiting hospitals who had established a care relationship with the participant and their family during the patient’s cancer journey. Frequently, the study was introduced at one meeting and discussed over several subsequent clinic visits, allowing patients and their families time to consider participation. We view the involvement of family members in the consent process as essential to support the participant and facilitate decisionmaking. Involvement of family members also ensures that they are fully aware of the autopsy process and helps to clarify funeral arrangements for the study team. After obtaining consent, study investigators collated clinical information, including that related to past and current treatment and diagnostic procedures such as imaging, on an ongoing basis. Between September 2012 and August 2015, 40 patients were approached, and 37 (92.5%) expressed interest in participating. Of those 32 patients (80%) consented; the other 5 had rapid clinical deterioration precluding
Anz Journal of Surgery | 2010
David E. Gyorki; C. E. Brooks; Rohan M. Gett; Rodney Woods; Michael J. Johnston; J. Keck; John Mackay; Alexander G. Heriot
Background: Enterocutaneous fistulae (ECFs) present a difficult management problem and can cause significant morbidity. The aim of the study was to assess the outcome of these patients.
Nature Medicine | 2018
Peter Savas; Balaji Virassamy; Chengzhong Ye; Agus Salim; Christopher P. Mintoff; Franco Caramia; Roberto Salgado; David J. Byrne; Zhi L. Teo; Sathana Dushyanthen; Ann Byrne; Lironne Wein; Stephen J. Luen; Catherine Poliness; Sophie S. Nightingale; Anita S. Skandarajah; David E. Gyorki; Chantel M. Thornton; Paul A. Beavis; Stephen B. Fox; Phillip K. Darcy; Terence P. Speed; Laura K. Mackay; Paul Neeson; Sherene Loi
The quantity of tumor-infiltrating lymphocytes (TILs) in breast cancer (BC) is a robust prognostic factor for improved patient survival, particularly in triple-negative and HER2-overexpressing BC subtypes1. Although T cells are the predominant TIL population2, the relationship between quantitative and qualitative differences in T cell subpopulations and patient prognosis remains unknown. We performed single-cell RNA sequencing (scRNA-seq) of 6,311 T cells isolated from human BCs and show that significant heterogeneity exists in the infiltrating T cell population. We demonstrate that BCs with a high number of TILs contained CD8+ T cells with features of tissue-resident memory T (TRM) cell differentiation and that these CD8+ TRM cells expressed high levels of immune checkpoint molecules and effector proteins. A CD8+ TRM gene signature developed from the scRNA-seq data was significantly associated with improved patient survival in early-stage triple-negative breast cancer (TNBC) and provided better prognostication than CD8 expression alone. Our data suggest that CD8+ TRM cells contribute to BC immunosurveillance and are the key targets of modulation by immune checkpoint inhibition. Further understanding of the development, maintenance and regulation of TRM cells will be crucial for successful immunotherapeutic development in BC.Extensive, high-dimensional characterization of T cells in breast cancer reveals activated TRM population and a gene signature associated with improved prognosis.
Journal of Oncology Practice | 2016
Patricia Banks; Shahneen Sandhu; David E. Gyorki; Meredith L. Johnston; Danny Rischin
Merkel cell carcinoma (MCC) is a rare and highly aggressive neuroendocrine malignancy with a propensity for recurrence and a poor prognosis. Incidence of MCC is on the rise and is known to increase with advanced age, immunosuppression, and UV exposure. Merkel cell polyomavirus is implicated in the pathogenesis of virus-positive MCC and accounts for 80% of MCCs in the northern hemisphere and 25% in southern latitudes. In contrast, tumorigenesis of virus-negative MCC is linked to UV-induced DNA damage. Interplay between ubiquitous Merkel cell polyomavirus skin infections that commonly occur in healthy skin and other established risk factors, such as immunosuppression and UV exposure, remains poorly understood. Surgery and radiotherapy achieves excellent locoregional control; however, invariably, a significant proportion of patients develop disseminated disease that is incurable. Chemotherapy offers a high response rate for metastatic disease, but responses are short-lived and the impact on survival is not established. Recent advances in our understanding of the genetic landscape and immunobiology of MCC has led to investigation of novel treatments, including immune checkpoint inhibitors, which are likely to rapidly transform the way we manage these patients. We review epidemiologic, clinical, and histopathologic features of MCC; describe recent insights in MCC biology; and discuss novel therapeutic approaches.