Network


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

Hotspot


Dive into the research topics where Amy A. Lubik is active.

Publication


Featured researches published by Amy A. Lubik.


Cancer Research | 2008

Androgen levels increase by intratumoral de novo steroidogenesis during progression of castration-resistant prostate cancer.

Jennifer A. Locke; Emma S. Guns; Amy A. Lubik; Hans Adomat; Stephen C. Hendy; Catherine Wood; Susan Ettinger; Martin Gleave; Colleen C. Nelson

Although systemic androgen deprivation prolongs life in advanced prostate cancer, remissions are temporary because patients almost uniformly progress to a state of a castration-resistant prostate cancer (CRPC) as indicated by recurring PSA. This complex process of progression does not seem to be stochastic as the timing and phenotype are highly predictable, including the observation that most androgen-regulated genes are reactivated despite castrate levels of serum androgens. Recent evidence indicates that intraprostatic levels of androgens remain moderately high following systemic androgen deprivation therapy, whereas the androgen receptor (AR) remains functional, and silencing the AR expression following castration suppresses tumor growth and blocks the expression of genes known to be regulated by androgens. From these observations, we hypothesized that CRPC progression is not independent of androgen-driven activity and that androgens may be synthesized de novo in CRPC tumors leading to AR activation. Using the LNCaP xenograft model, we showed that tumor androgens increase during CRPC progression in correlation to PSA up-regulation. We show here that all enzymes necessary for androgen synthesis are expressed in prostate cancer tumors and some seem to be up-regulated during CRPC progression. Using an ex vivo radiotracing assays coupled to high-performance liquid chromatography-radiometric/mass spectrometry detection, we show that tumor explants isolated from CRPC progression are capable of de novo conversion of [(14)C]acetic acid to dihydrotestosterone and uptake of [(3)H]progesterone allows detection of the production of six other steroids upstream of dihydrotestosterone. This evidence suggests that de novo androgen synthesis may be a driving mechanism leading to CRPC progression following castration.


Cancer Research | 2011

Insulin Increases De Novo Steroidogenesis in Prostate Cancer Cells

Amy A. Lubik; Jennifer H. Gunter; Stephen C. Hendy; Jennifer A. Locke; Hans Adomat; Vanessa C. Thompson; Adrian C. Herington; Martin Gleave; Michael Pollak; Colleen C. Nelson

Androgen-dependent pathways regulate maintenance and growth of normal and malignant prostate tissues. Androgen deprivation therapy (ADT) exploits this dependence and is used to treat metastatic prostate cancer; however, regression initially seen with ADT gives way to development of incurable castration-resistant prostate cancer (CRPC). Although ADT generates a therapeutic response, it is also associated with a pattern of metabolic alterations consistent with metabolic syndrome including elevated circulating insulin. Because CRPC cells are capable of synthesizing androgens de novo, we hypothesized that insulin may also influence steroidogenesis in CRPC. In this study, we examined this hypothesis by evaluating the effect of insulin on steroid synthesis in prostate cancer cell lines. Treatment with 10 nmol/L insulin increased mRNA and protein expression of steroidogenesis enzymes and upregulated the insulin receptor substrate insulin receptor substrate 2 (IRS-2). Similarly, insulin treatment upregulated intracellular testosterone levels and secreted androgens, with the concentrations of steroids observed similar to the levels reported in prostate cancer patients. With similar potency to dihydrotestosterone, insulin treatment resulted in increased mRNA expression of prostate-specific antigen. CRPC progression also correlated with increased expression of IRS-2 and insulin receptor in vivo. Taken together, our findings support the hypothesis that the elevated insulin levels associated with therapeutic castration may exacerbate progression of prostate cancer to incurable CRPC in part by enhancing steroidogenesis.


Organogenesis | 2010

Interactions between human osteoblasts and prostate cancer cells in a novel 3D in vitro model

Shirly Sieh; Amy A. Lubik; Judith A. Clements; Colleen C. Nelson; Dietmar W. Hutmacher

Cell-cell and cell-matrix interactions play a major role in tumor morphogenesis and cancer metastasis. Therefore, it is crucial to create a model with a biomimetic microenvironment that allows such interactions to fully represent the pathophysiology of a disease for an in vitro study. This is achievable by using three-dimensional (3D) models instead of conventional two-dimensional (2D) cultures with the aid of tissue engineering technology. We are now able to better address the complex intercellular interactions underlying prostate cancer (CaP) bone metastasis through such models. In this study, we assessed the interaction of CaP cells and human osteoblasts (hOBs) within a tissue engineered bone (TEB) construct. Consistent with other in vivo studies, our findings show that intercellular and CaP cell-bone matrix interactions lead to elevated levels of matrix metalloproteinases, steroidogenic enzymes and the CaP biomarker, prostate specific antigen (PSA); all associated with CaP metastasis. Hence, it highlights the physiological relevance of this model. We believe that this model will provide new insights for understanding of the previously poorly understood molecular mechanisms of bone metastasis, which will foster further translational studies, and ultimately offer a potential tool for drug screening.


The Prostate | 2009

Arachidonic acid activation of intratumoral steroid synthesis during prostate cancer progression to castration resistance.

Jennifer A. Locke; Emma S. Guns; Melanie Lehman; Susan Ettinger; Amina Zoubeidi; Amy A. Lubik; Katia Margiotti; Ladan Fazli; Hans Adomat; Kishor M. Wasan; Martin Gleave; Colleen C. Nelson

De novo androgen synthesis and subsequent androgen receptor (AR) activation has recently been shown to contribute to castration‐resistant prostate cancer (CRPC) progression. Herein we provide evidence that fatty acids (FA) can trigger androgen synthesis within steroid starved prostate cancer (CaP) tumor cells.


The Prostate | 2009

A novel communication role for CYP17A1 in the progression of castration‐resistant prostate cancer

Jennifer A. Locke; Ladan Fazli; Hans Adomat; Jil Smyl; Kristin Weins; Amy A. Lubik; Dale B. Hales; Colleen C. Nelson; Martin Gleave; Emma S. Guns

CYP17A1 is currently a target for total androgen blockade in advanced prostate cancer (CaP) patients. After castration, or removal of testicular androgens, CYP17A1 can act as a rate‐limiting enzyme in androgen synthesis from cholesterol or other adrenal precursors within the tumor microenvironment ultimately contributing to disease progression. Herein we provide evidence that CYP17A1 could also be a mediator of cell‐to‐cell communication within the CaP tumor microenvironment.


Endocrine-related Cancer | 2013

IGF2 increases de novo steroidogenesis in prostate cancer cells

Amy A. Lubik; Jennifer H. Gunter; Brett G. Hollier; Susan Ettinger; Ladan Fazli; Nataly Stylianou; Stephen C. Hendy; Hans Adomat; Martin Gleave; Michael Pollak; Adrian C. Herington; Colleen C. Nelson

IGF2 is a mitogenic foetal growth factor commonly over-expressed in cancers, including prostate cancer (PC). We recently demonstrated that insulin can activate de novo steroidogenesis in PC cells, a major pathway for reactivation of androgen pathways and PC progression. IGF2 can activate the IGF1 receptor (IGF1R) or insulin receptor (INSR) or hybrids of these two receptors. We therefore hypothesized that IGF2 may contribute to PC progression via de novo steroidogenesis. IGF2 mRNA but not IGF2 receptor mRNA expression was increased in patient samples during progression to castrate-resistant PC as was immunoreactivity to INSR and IGF1R antibodies. Treatment of androgen receptor (AR)-positive PC cell lines LNCaP and 22RV1 with IGF2 for 48 h resulted in increased expression of steroidogenic enzyme mRNA and protein, including steroid acute regulatory protein (StAR), cytochrome p450 family member (CYP)17A1, aldo-keto reductase family member (AKR)1C3 and hydroxysteroid dehydrogenase (HSD)17B3. IGF2 treatment resulted in increased steady state steroid levels and increased de novo steroidogenesis resulting in AR activation as demonstrated by PSA mRNA induction. Inhibition of the IGF1R/INSR signalling axis attenuated the effects of IGF2 on steroid hormone synthesis. We present a potential mechanism for prostatic IGF2 contributing to PC progression by inducing steroidogenesis and that IGF2 signalling and related pathways present attractive targets for PC therapy.


Journal of Molecular Endocrinology | 2013

Cloning of a novel insulin-regulated ghrelin transcript in prostate cancer.

Inge Seim; Amy A. Lubik; Melanie Lehman; Nadine Tomlinson; Eliza Whiteside; Adrian C. Herington; Colleen C. Nelson; Lisa K. Chopin

Ghrelin is a multifunctional hormone, with roles in stimulating appetite and regulating energy balance, insulin secretion and glucose homoeostasis. The ghrelin gene locus (GHRL) is highly complex and gives rise to a range of novel transcripts derived from alternative first exons and internally spliced exons. The wild-type transcript encodes a 117 amino acid preprohormone that is processed to yield the 28 amino acid peptide ghrelin. Here, we identified insulin-responsive transcription corresponding to cryptic exons in intron 2 of the human ghrelin gene. A transcript, termed in2c-ghrelin (intron 2-cryptic), was cloned from the testis and the LNCaP prostate cancer cell line. This transcript may encode an 83 amino acid preproghrelin isoform that codes for ghrelin, but not obestatin. It is expressed in a limited number of normal tissues and in tumours of the prostate, testis, breast and ovary. Finally, we confirmed that in2c-ghrelin transcript expression, as well as the recently described in1-ghrelin transcript, is significantly upregulated by insulin in cultured prostate cancer cells. Metabolic syndrome and hyperinsulinaemia have been associated with prostate cancer risk and progression. This may be particularly significant after androgen deprivation therapy for prostate cancer, which induces hyperinsulinaemia, and this could contribute to castrate-resistant prostate cancer growth. We have previously demonstrated that ghrelin stimulates prostate cancer cell line proliferation in vitro. This study is the first description of insulin regulation of a ghrelin transcript in cancer and should provide further impetus for studies into the expression, regulation and function of ghrelin gene products.


International Journal of Cell Biology | 2013

New players for advanced prostate cancer and the rationalisation of insulin-sensitising medication.

Jennifer H. Gunter; Phoebe Sarkar; Amy A. Lubik; Colleen C. Nelson

Obesity and type 2 diabetes are recognised risk factors for the development of some cancers and, increasingly, predict more aggressive disease, treatment failure, and cancer-specific mortality. Many factors may contribute to this clinical observation. Hyperinsulinaemia, dyslipidaemia, hypoxia, ER stress, and inflammation associated with expanded adipose tissue are thought to be among the main culprits driving malignant growth and cancer advancement. This observation has led to the proposal of the potential utility of “old players” for the treatment of type 2 diabetes and metabolic syndrome as new cancer adjuvant therapeutics. Androgen-regulated pathways drive proliferation, differentiation, and survival of benign and malignant prostate tissue. Androgen deprivation therapy (ADT) exploits this dependence to systemically treat advanced prostate cancer resulting in anticancer response and improvement of cancer symptoms. However, the initial therapeutic response from ADT eventually progresses to castrate resistant prostate cancer (CRPC) which is currently incurable. ADT rapidly induces hyperinsulinaemia which is associated with more rapid treatment failure. We discuss current observations of cancer in the context of obesity, diabetes, and insulin-lowering medication. We provide an update on current treatments for advanced prostate cancer and discuss whether metabolic dysfunction, developed during ADT, provides a unique therapeutic window for rapid translation of insulin-sensitising medication as combination therapy with antiandrogen targeting agents for the management of advanced prostate cancer.


Advances in Urology | 2012

The interactions between insulin and androgens in progression to castrate resistant prostate cancer

Jennifer H. Gunter; Amy A. Lubik; Ian McKenzie; Michael Pollak; Colleen C. Nelson

An association between the metabolic syndrome and reduced testosterone levels has been identified, and a specific inverse relationship between insulin and testosterone levels suggests that an important metabolic crosstalk exists between these two hormonal axes; however, the mechanisms by which insulin and androgens may be reciprocally regulated are not well described. Androgen-dependant gene pathways regulate the growth and maintenance of both normal and malignant prostate tissue, and androgen-deprivation therapy (ADT) in patients exploits this dependence when used to treat recurrent and metastatic prostate cancer resulting in tumour regression. A major systemic side effect of ADT includes induction of key features of the metabolic syndrome and the consistent feature of hyperinsulinaemia. Recent studies have specifically identified a correlation between elevated insulin and high-grade PCa and more rapid progression to castrate resistant disease. This paper examines the relationship between insulin and androgens in the context of prostate cancer progression. Prostate cancer patients present a promising cohort for the exploration of insulin stabilising agents as adjunct treatments for hormone deprivation or enhancers of chemosensitivity for treatment of advanced prostate cancer.


Oncotarget | 2017

Therapy-induced developmental reprogramming of prostate cancer cells and acquired therapy resistance

Mannan Nouri; Amy A. Lubik; Na Li; Brett G. Hollier; Mandeep Takhar; Manuel Altimirano-Dimas; Mengqian Chen; Mani Roshan-Moniri; Miriam S. Butler; Melanie Lehman; Jennifer L. Bishop; Sarah Truong; Shih Chieh Huang; Dawn R. Cochrane; Michael E. Cox; Colin Collins; Martin Gleave; Nicholas Erho; Mohamed Alshalafa; Elai Davicioni; Colleen C. Nelson; Sheryl Gregory-Evans; R. Jeffrey Karnes; Robert B. Jenkins; Eric A. Klein; Ralph Buttyan

Treatment-induced neuroendocrine transdifferentiation (NEtD) complicates therapies for metastatic prostate cancer (PCa). Based on evidence that PCa cells can transdifferentiate to other neuroectodermally-derived cell lineages in vitro, we proposed that NEtD requires first an intermediary reprogramming to metastable cancer stem-like cells (CSCs) of a neural class and we demonstrate that several different AR+/PSA+ PCa cell lines were efficiently reprogrammed to, maintained and propagated as CSCs by growth in androgen-free neural/neural crest (N/NC) stem medium. Such reprogrammed cells lost features of prostate differentiation; gained features of N/NC stem cells and tumor-initiating potential; were resistant to androgen signaling inhibition; and acquired an invasive phenotype in vitro and in vivo. When placed back into serum-containing mediums, reprogrammed cells could be re-differentiated to N-/NC-derived cell lineages or return back to an AR+ prostate-like state. Once returned, the AR+ cells were resistant to androgen signaling inhibition. Acute androgen deprivation or anti-androgen treatment in serum-containing medium led to the transient appearance of a sub-population of cells with similar characteristics. Finally, a 132 gene signature derived from reprogrammed PCa cell lines distinguished tumors from PCa patients with adverse outcomes. This model may explain neural manifestations of PCa associated with lethal disease. The metastable nature of the reprogrammed stem-like PCa cells suggests that cycles of PCa cell reprogramming followed by re-differentiation may support disease progression and therapeutic resistance. The ability of a gene signature from reprogrammed PCa cells to identify tumors from patients with metastasis or PCa-specific mortality implies that developmental reprogramming is linked to aggressive tumor behaviors.

Collaboration


Dive into the Amy A. Lubik's collaboration.

Top Co-Authors

Avatar

Colleen C. Nelson

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Hans Adomat

Vancouver General Hospital

View shared research outputs
Top Co-Authors

Avatar

Martin Gleave

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Emma S. Guns

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Ralph Buttyan

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Jennifer A. Locke

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Jennifer H. Gunter

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Ladan Fazli

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Mannan Nouri

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Sarah Truong

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge