Network


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

Hotspot


Dive into the research topics where Mannan Nouri is active.

Publication


Featured researches published by Mannan Nouri.


Frontiers in Oncology | 2014

Androgen-targeted therapy-induced epithelial mesenchymal plasticity and neuroendocrine transdifferentiation in prostate cancer: an opportunity for intervention

Mannan Nouri; Ellca Ratther; Nataly Stylianou; Colleen C. Nelson; Brett G. Hollier; Elizabeth D. Williams

Androgens regulate biological pathways to promote proliferation, differentiation, and survival of benign and malignant prostate tissue. Androgen receptor (AR) targeted therapies exploit this dependence and are used in advanced prostate cancer to control disease progression. Contemporary treatment regimens involve sequential use of inhibitors of androgen synthesis or AR function. Although targeting the androgen axis has clear therapeutic benefit, its effectiveness is temporary, as prostate tumor cells adapt to survive and grow. The removal of androgens (androgen deprivation) has been shown to activate both epithelial-to-mesenchymal transition (EMT) and neuroendocrine transdifferentiation (NEtD) programs. EMT has established roles in promoting biological phenotypes associated with tumor progression (migration/invasion, tumor cell survival, cancer stem cell-like properties, resistance to radiation and chemotherapy) in multiple human cancer types. NEtD in prostate cancer is associated with resistance to therapy, visceral metastasis, and aggressive disease. Thus, activation of these programs via inhibition of the androgen axis provides a mechanism by which tumor cells can adapt to promote disease recurrence and progression. Brachyury, Axl, MEK, and Aurora kinase A are molecular drivers of these programs, and inhibitors are currently in clinical trials to determine therapeutic applications. Understanding tumor cell plasticity will be important in further defining the rational use of androgen-targeted therapies clinically and provides an opportunity for intervention to prolong survival of men with metastatic 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.


Oncogene | 2017

Neuropilin-1 is upregulated in the adaptive response of prostate tumors to androgen-targeted therapies and is prognostic of metastatic progression and patient mortality

Brian Wan-Chi Tse; Marianna Volpert; Ellca Ratther; Nataly Stylianou; Mannan Nouri; K McGowan; Melanie Lehman; Stephen McPherson; Mani Roshan-Moniri; M S Butler; C.Y. Gregory-Evans; Jacqui A. McGovern; Rajdeep Das; Mandeep Takhar; Nicholas Erho; Mohammed Alshalafa; Elai Davicioni; Edward M. Schaeffer; Robert B. Jenkins; Ashley E. Ross; R.J. Karnes; Robert B. Den; Ladan Fazli; Philip A. Gregory; Martin Gleave; Elizabeth D. Williams; Paul S. Rennie; Ralph Buttyan; Jennifer H. Gunter; Luke A. Selth

Recent evidence has implicated the transmembrane co-receptor neuropilin-1 (NRP1) in cancer progression. Primarily known as a regulator of neuronal guidance and angiogenesis, NRP1 is also expressed in multiple human malignancies, where it promotes tumor angiogenesis. However, non-angiogenic roles of NRP1 in tumor progression remain poorly characterized. In this study, we define NRP1 as an androgen-repressed gene whose expression is elevated during the adaptation of prostate tumors to androgen-targeted therapies (ATTs), and subsequent progression to metastatic castration-resistant prostate cancer (mCRPC). Using short hairpin RNA (shRNA)-mediated suppression of NRP1, we demonstrate that NRP1 regulates the mesenchymal phenotype of mCRPC cell models and the invasive and metastatic dissemination of tumor cells in vivo. In patients, immunohistochemical staining of tissue microarrays and mRNA expression analyses revealed a positive association between NRP1 expression and increasing Gleason grade, pathological T score, positive lymph node status and primary therapy failure. Furthermore, multivariate analysis of several large clinical prostate cancer (PCa) cohorts identified NRP1 expression at radical prostatectomy as an independent prognostic biomarker of biochemical recurrence after radiation therapy, metastasis and cancer-specific mortality. This study identifies NRP1 for the first time as a novel androgen-suppressed gene upregulated during the adaptive response of prostate tumors to ATTs and a prognostic biomarker of clinical metastasis and lethal PCa.


Oncotarget | 2017

Discovery and characterization of small molecules targeting the DNA-binding ETS domain of ERG in prostate cancer

Miriam S. Butler; Mani Roshan-Moniri; Michael Hsing; Desmond Lau; Ari Kim; Paul Yen; Marta Mroczek; Mannan Nouri; Scott Lien; Peter Axerio-Cilies; Kush Dalal; Clement Yau; Fariba Ghaidi; Yubin Guo; Takeshi Yamazaki; Sam Lawn; Martin Gleave; Cheryl Y. Gregory-Evans; Lawrence P. McIntosh; Michael E. Cox; Paul S. Rennie; Artem Cherkasov

Genomic alterations involving translocations of the ETS-related gene ERG occur in approximately half of prostate cancer cases. These alterations result in aberrant, androgen-regulated production of ERG protein variants that directly contribute to disease development and progression. This study describes the discovery and characterization of a new class of small molecule ERG antagonists identified through rational in silico methods. These antagonists are designed to sterically block DNA binding by the ETS domain of ERG and thereby disrupt transcriptional activity. We confirmed the direct binding of a lead compound, VPC-18005, with the ERG-ETS domain using biophysical approaches. We then demonstrated VPC-18005 reduced migration and invasion rates of ERG expressing prostate cancer cells, and reduced metastasis in a zebrafish xenograft model. These results demonstrate proof-of-principal that small molecule targeting of the ERG-ETS domain can suppress transcriptional activity and reverse transformed characteristics of prostate cancers aberrantly expressing ERG. Clinical advancement of the developed small molecule inhibitors may provide new therapeutic agents for use as alternatives to, or in combination with, current therapies for men with ERG-expressing metastatic castration-resistant prostate cancer.


Oncogene | 2018

Non-canonical activation of hedgehog in prostate cancer cells mediated by the interaction of transcriptionally active androgen receptor proteins with Gli3

Na Li; Sarah Truong; Mannan Nouri; Jackson Moore; Nader Al Nakouzi; Amy A. Lubik; Ralph Buttyan

Hedgehog (Hh) is an oncogenic signaling pathway that regulates the activity of Gli transcription factors. Canonical Hh is a Smoothened- (Smo-) driven process that alters the post-translational processing of Gli2/Gli3 proteins. Though evidence supports a role for Gli action in prostate cancer (PCa) cell growth and progression, there is little indication that Smo is involved. Here we describe a non-canonical means for activation of Gli transcription in PCa cells mediated by the binding of transcriptionally-active androgen receptors (ARs) to Gli3. Androgens stimulated reporter expression from a Gli-dependent promoter in a variety of AR + PCa cells and this activity was suppressed by an anti-androgen, Enz, or by AR knockdown. Androgens also upregulated expression of endogenous Gli-dependent genes. This activity was associated with increased intranuclear binding of Gli3 to AR that was antagonized by Enz. Fine mapping of the AR binding domain on Gli2 showed that AR recognizes the Gli protein processing domain (PPD) in the C-terminus. Mutations in the arginine-/serine repeat elements of the Gli2 PPD involved in phosphorylation and ubiquitinylation blocked the binding to AR. β-TrCP, a ubiquitin ligase that recognizes the Gli PPD, competed with AR for binding to this site. AR binding to Gli3 suppressed its proteolytic processing to the Gli3 repressor form (Gli3R) whereas AR knockdown increased Gli3R. Both full-length and truncated ARs were able to activate Gli transcription. Finally, we found that an ARbinding decoy polypeptide derived from the Gli2 C-terminus can compete with Gli3 for binding to AR. Exogenous overexpression of this decoy suppressed Gli transcriptional activity in PCa cells. Collectively, this work identifies a novel pathway for non-canonical activation of Hh signaling in PCa cells and identifies a means for interference that may have clinical relevance for PCa patients.


GigaScience | 2018

The long noncoding RNA landscape of neuroendocrine prostate cancer and its clinical implications

Varune Rohan Ramnarine; Mohammed Alshalalfa; Fan Mo; Noushin Nabavi; Nicholas Erho; Mandeep Takhar; Robert Shukin; Sonal Brahmbhatt; Alexander Gawronski; Maxim Kobelev; Mannan Nouri; Dong Lin; Harrison Tsai; Tamara L. Lotan; R Jefferey Karnes; Mark A. Rubin; Amina Zoubeidi; Martin Gleave; Cenk Sahinalp; Alexander W. Wyatt; Stanislav Volik; Himisha Beltran; Elai Davicioni; Yuzhuo Wang; Colin Collins

Abstract Background Treatment-induced neuroendocrine prostate cancer (tNEPC) is an aggressive variant of late-stage metastatic castrate-resistant prostate cancer that commonly arises through neuroendocrine transdifferentiation (NEtD). Treatment options are limited, ineffective, and, for most patients, result in death in less than a year. We previously developed a first-in-field patient-derived xenograft (PDX) model of NEtD. Longitudinal deep transcriptome profiling of this model enabled monitoring of dynamic transcriptional changes during NEtD and in the context of androgen deprivation. Long non-coding RNA (lncRNA) are implicated in cancer where they can control gene regulation. Until now, the expression of lncRNAs during NEtD and their clinical associations were unexplored. Results We implemented a next-generation sequence analysis pipeline that can detect transcripts at low expression levels and built a genome-wide catalogue (n = 37,749) of lncRNAs. We applied this pipeline to 927 clinical samples and our high-fidelity NEtD model LTL331 and identified 821 lncRNAs in NEPC. Among these are 122 lncRNAs that robustly distinguish NEPC from prostate adenocarcinoma (AD) patient tumours. The highest expressed lncRNAs within this signature are H19, LINC00617, and SSTR5-AS1. Another 742 are associated with the NEtD process and fall into four distinct patterns of expression (NEtD lncRNA Class I, II, III, and IV) in our PDX model and clinical samples. Each class has significant (z-scores >2) and unique enrichment for transcription factor binding site (TFBS) motifs in their sequences. Enriched TFBS include (1) TP53 and BRN1 in Class I, (2) ELF5, SPIC, and HOXD1 in Class II, (3) SPDEF in Class III, (4) HSF1 and FOXA1 in Class IV, and (5) TWIST1 when merging Class III with IV. Common TFBS in all NEtD lncRNA were also identified and include E2F, REST, PAX5, PAX9, and STAF. Interrogation of the top deregulated candidates (n = 100) in radical prostatectomy adenocarcinoma samples with long-term follow-up (median 18 years) revealed significant clinicopathological associations. Specifically, we identified 25 that are associated with rapid metastasis following androgen deprivation therapy (ADT). Two of these lncRNAs (SSTR5-AS1 and LINC00514) stratified patients undergoing ADT based on patient outcome. Discussion To date, a comprehensive characterization of the dynamic landscape of lncRNAs during the NEtD process has not been performed. A temporal analysis of the PDX-based NEtD model has for the first time provided this dynamic landscape. TFBS analysis identified NEPC-related TF motifs present within the NEtD lncRNA sequences, suggesting functional roles for these lncRNAs in NEPC pathogenesis. Furthermore, select NEtD lncRNAs appear to be associated with metastasis and patients receiving ADT. Treatment-related metastasis is a clinical consequence of NEPC tumours. Top candidate lncRNAs FENDRR, H19, LINC00514, LINC00617, and SSTR5-AS1 identified in this study are implicated in the development of NEPC. We present here for the first time a genome-wide catalogue of NEtD lncRNAs that characterize the transdifferentiation process and a robust NEPC lncRNA patient expression signature. To accomplish this, we carried out the largest integrative study that applied a PDX NEtD model to clinical samples. These NEtD and NEPC lncRNAs are strong candidates for clinical biomarkers and therapeutic targets and warrant further investigation.


International Journal of Cancer | 2017

Paracrine sonic hedgehog signaling contributes significantly to acquired steroidogenesis in the prostate tumor microenvironment.

Amy A. Lubik; Mannan Nouri; Sarah Truong; Mazyar Ghaffari; Hans Adomat; Eva Corey; Michael E. Cox; Na Li; Emma S. Guns; Parvin Yenki; Steven Pham; Ralph Buttyan

Despite the substantial benefit of androgen deprivation therapy (ADT) for metastatic prostate cancer, patients often progress to castration‐resistant disease (CRPC) that is more difficult to treat. CRPC is associated with renewed androgen receptor activity in tumor cells and restoration of tumor androgen levels through acquired intratumoral steroidogenesis (AIS). Although prostate cancer (PCa) cells have been shown to have steroidogenic capability in vitro, we previously found that benign prostate stromal cells (PrSCs) can also synthesize testosterone (T) from an adrenal precursor, DHEA, when stimulated with a hedgehog (Hh) pathway agonist, SAG. Here, we show exposure of PrSCs to a different Smoothened (Smo) agonist, Ag1.5, or to conditioned medium from sonic hedgehog overexpressing LNCaP cells induces steroidogenic enzyme expression in PrSCs and significantly increases production of T and its precursor steroids in a Smo‐dependent manner from 22‐OH‐cholesterol substrate. Hh agonist‐/ligand‐treated PrSCs produced androgens at a rate similar to or greater than that of PCa cell lines. Likewise, primary bone marrow stromal cells became more steroidogenic and produced T under the influence of Smo agonist. Treatment of mice bearing LNCaP xenografts with a Smo antagonist, TAK‐441, delayed the onset of CRPC after castration and substantially reduced androgen levels in residual tumors. These outcomes support the idea that stromal cells in ADT‐treated primary or metastatic prostate tumors can contribute to AIS as a consequence of a paracrine Hh signaling microenvironment. As such, Smo antagonists may be useful for targeting prostate tumor stromal cell‐derived AIS and delaying the onset of CRPC after ADT.


The Journal of Urology | 2017

PD33-01 NON-CANONICAL ACTIVATION OF HEDGEHOG SIGNALING IN PROSTATE CANCER CELLS MEDIATED BY THE BINDING OF TRANSCRIPTIONALLY ACTIVE ANDROGEN RECEPTORS TO GLI TRANSCRIPTION FACTORS

Na Li; Sarah Truong; Mannan Nouri; Amy A. Lubik; Ralph Buttyan

hospitals with excess readmission rates for certain targeted conditions. We aim to evaluate the effect of this program on targeted and nontargeted surgical conditions. METHODS: We used a 20% Medicare sample to identify readmissions following targeted (total hip arthroplasty, total knee arthroplasty) and non-targeted (cystectomy, abdominal aortic aneurysm repair, colectomy, lung resection) procedures from 2006 to 2014. Multivariable logistic regression was used to calculate adjusted readmission rates. Changes in hospital level readmission rates were analyzed for three distinct time periods (Pre, Measurement, Penalty) corresponding to the HRRP implementation timeline, using an interrupted time series approach. RESULTS: We identified 538,293 targeted and 165,432 nontargeted procedures performed at 2,779 hospitals. There was a significant decrease in the odds of readmission for all procedures, except cystectomy (Table) which also had the highest readmission rate at 27%. Prior to the policy, the readmission rate for non-targeted procedures was decreasing faster than that of targeted procedures (Figure). However, this trend reversed during the Measurement period (difference in slope for targeted to non-targeted -0.10 [95% Confidence Interval -0.16 to -0.044]). Neither group had a significant change during the Penalty period. CONCLUSIONS: While the HRRP effectively decreased readmissions for targeted surgery, there was no spillover benefit for nontargeted procedures. To decrease the burden of readmission after cystectomy, future efforts should focus on identifying the interventions that resulted in readmission reduction for targeted procedures and evaluating their effectiveness in this population.


Cancer Research | 2017

Abstract 4492: Androgen receptor affects GLI activity by promoting transcriptionally active states of GLI upon androgen stimulation

Sarah Truong; Na Li; Mannan Nouri; Ralph Buttyan

Introduction: While “castration” therapies benefit metastatic prostate cancer (PCa) patients, their effects are transient and progression to castration resistant disease (CRPC) remains a vexing clinical problem. Despite low levels of circulating androgens in treated patients, CRPC typically remains dependent on androgen/androgen receptor (AR) signaling for further growth. Previously we showed that Gli protein binding to AR co-activates AR transcriptional activity and supports androgen-independent growth. Here we show evidence that the interaction of AR with Gli proteins, particularly Gli3, allows for a “non-canonical” activation of Hedgehog signaling that contributes to PCa cell growth and progression to CRPC. Methods: Androgen growth-dependent (LNCaP) or -independent (LNCaP-AI, LN95) PCa cells were transfected with a Gli-luciferase (Gli-luc) reporter and were treated with R1881, enzalutamide (Enz), or a combination. Luciferase activity was measured. R1-D567 (harboring a genomic deletion of AR) served as a negative control. Knockdown of AR-FL with siRNA or overexpression of a small decoy peptide from the Gli2 AR-binding domain (Gli-DP) was also performed in above cells. Western blot and real-time PCR confirmed both AR and Gli target gene/protein expressions in LNCaP and LNCaP-AI cells (-/+ R1881). Sonic hedgehog (Shh) ligand-responsive 3T3-E1 cells were transiently expressed with AR-FL to confirm the involvement of canonical Shh-Gli activation pathway in AR co-activation of Gli transcription. Co-immunoprecipitation (co-IP) was performed to verify the blocking efficiency of Gli-DP in AR-Gli3 interaction in LN95 cells and AR-overexpressing 293FT. Results: Androgen (R1881) treatment strongly induced Gli transcription activity in AR+ PCa cells while Enz reversed this effect; AR-FL-specific knockdown or exogenous Gli-DP expression suppressed androgen-induced Gli reporter expression as well as expression of endogenous Gli-responsive genes. R1881 treatment promoted Gli2/3 protein processing to transcriptionally active states (Gli3-FullLength and Gli2-Active) and enhanced the expression of Gli-target genes, Gli1/Ptch1, at protein and/or mRNA levels. Co-IP revealed that exogenous Gli-DP blocked the interaction between AR and transcriptionally active Gli3-FL in PCa cells. Overexpression of AR in 3T3-E1 cells confirmed that AR interaction with Gli stabilizes the active Gli3-FL form by suppressing degradation of Gli3 in a manner independent of the canonical Shh-Gli activation pathway. Conclusions: Our findings further identify the importance of AR-Gli interaction in progression of PCa to CRPC and shows that cooperative binding of AR to Gli proteins enhances both AR and Hedgehog signaling in PCa cells. Interference with the AR-Gli interaction using a decoy peptide provides a means to suppress AR activation of Hedgehog in PCa cells. Citation Format: Sarah Truong, Na Li, Mannan Nouri, Ralph Buttyan. Androgen receptor affects GLI activity by promoting transcriptionally active states of GLI upon androgen stimulation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4492. doi:10.1158/1538-7445.AM2017-4492


Cancer Research | 2017

Abstract 4908: Neuropilin-1 is up-regulated in the adaptive response of prostate tumors to androgen targeted therapies and is prognostic of metastatic progression and patient mortality

Marianna Volpert; Brian Wan-Chi Tse; Ellca Ratther; Nataly Stylianou; Mannan Nouri; Melanie Lehman; Stephen McPherson; Mani Roshan-Moniri; Mandeep Takhar; Nicholas Erho; Mohamed Alshalafa; Elai Davicioni; Robert B. Jenkins; Ashley E. Ross; Jeffrey Karnes; Robert B. Den; Ladan Fazli; Martin Gleave; Elizabeth D. Williams; Paul S. Rennie; Ralph Buttyan; Pamela J. Russell; Colleen C. Nelson; Brett G. Hollier

Aims: Androgen-targeted therapies (ATTs) are the mainstay treatment for metastatic prostate cancer (PCa). However, ATTs promote adaptation of tumour cells and lead to castration resistant disease (CRPC). We have recently identified the cell surface receptor, Neuropilin-1 (NRP1) as increased during EMT and in CRPC. However, the role of NRP1 in the prostate epithelium is poorly understood. This study aims to determine whether the inhibition of NRP1 will be a feasible therapeutic strategy for blocking PCa metastasis and therapy resistance. Methods: qPCR and western blotting were used to assess NRP1 expression in PCa cell lines. NRP1 expression in CRPC was assessed using a murine LNCaP xenograft model of castration. NRP1 was knocked down with shRNA sequences from the pLKO.1 lentiviral construct. For metastasis assays, PC3 cells were microinjected into the zebrafish yolk sac and metastatic dissemination imaged 5 days later. NRP1 expression in radical prostatectomy (RP) samples from Mayo Clinic (545 patients) and Johns Hopkins Medical Institutions (JHMI; 188 patients) cohorts was quantified by Affymetrix exon arrays and multivariable analysis performed. Wound scratch migration and invasion assays were performed with the WoundMaker™ tool and IncuCyte™ FLR imaging systems. Results: NRP1 levels were elevated in humanCRPC xenografts, metastatic and castrate resistant clinical PCa samples (p Conclusion: These results will provide the preclinical data necessary to rationalise the use of anti-NRP1 directed adjuvant therapies for clinical use in PCa patients receiving ATTs, and will pave the way for larger scale preclinical and clinical trials in the PCa setting. Citation Format: Marianna Volpert, Brian Tse, Ellca Ratther, Nataly Stylianou, Mannan Nouri, Melanie Lehman, Stephen McPherson, Mani Roshan-Moniri, Mandeep Takhar, Nicholas Erho, Mohamed Alshalafa, Elai Davicioni, Robert Jenkins, Ashley Ross, Jeffrey Karnes, Robert Den, Ladan Fazli, Martin Gleave, Elizabeth Williams, Paul Rennie, Ralph Buttyan, Pamela Russell, Colleen Nelson, Brett Hollier. Neuropilin-1 is up-regulated in the adaptive response of prostate tumors to androgen targeted therapies and is prognostic of metastatic progression and patient mortality [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4908. doi:10.1158/1538-7445.AM2017-4908

Collaboration


Dive into the Mannan Nouri's collaboration.

Top Co-Authors

Avatar

Ralph Buttyan

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Sarah Truong

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Martin Gleave

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Amy A. Lubik

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Mani Roshan-Moniri

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Brett G. Hollier

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Paul S. Rennie

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Colleen C. Nelson

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Elizabeth D. Williams

Queensland University of Technology

View shared research outputs
Top Co-Authors

Avatar

Ellca Ratther

Queensland University of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge