Network


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

Hotspot


Dive into the research topics where Adrian Ishkanian is active.

Publication


Featured researches published by Adrian Ishkanian.


Nature Genetics | 2004

A tiling resolution DNA microarray with complete coverage of the human genome.

Adrian Ishkanian; Chad A. Malloff; Spencer Watson; Ronald J. deLeeuw; Bryan Chi; Bradley P. Coe; Antoine M. Snijders; Donna G. Albertson; Daniel Pinkel; Marco A. Marra; Victor Ling; Calum MacAulay; Wan L. Lam

We constructed a tiling resolution array consisting of 32,433 overlapping BAC clones covering the entire human genome. This increases our ability to identify genetic alterations and their boundaries throughout the genome in a single comparative genomic hybridization (CGH) experiment. At this tiling resolution, we identified minute DNA alterations not previously reported. These alterations include microamplifications and deletions containing oncogenes, tumor-suppressor genes and new genes that may be associated with multiple tumor types. Our findings show the need to move beyond conventional marker-based genome comparison approaches, that rely on inference of continuity between interval markers. Our submegabase resolution tiling set for array CGH (SMRT array) allows comprehensive assessment of genomic integrity and thereby the identification of new genes associated with disease.


Lancet Oncology | 2014

Tumour genomic and microenvironmental heterogeneity for integrated prediction of 5-year biochemical recurrence of prostate cancer: a retrospective cohort study

Emilie Lalonde; Adrian Ishkanian; Jenna Sykes; Michael Fraser; Helen Ross-Adams; Nicholas Erho; Mark J. Dunning; Silvia Halim; Alastair D. Lamb; Nathalie C Moon; Gaetano Zafarana; Anne Warren; Xianyue Meng; John Thoms; Michal R Grzadkowski; Alejandro Berlin; Cherry Have; Varune Rohan Ramnarine; Cindy Q. Yao; Chad A. Malloff; Lucia L. Lam; Honglei Xie; Nicholas J. Harding; Denise Y. F. Mak; Kenneth C. Chu; Lauren C. Chong; Dorota H Sendorek; Christine P'ng; Colin Collins; Jeremy A. Squire

BACKGROUND Clinical prognostic groupings for localised prostate cancers are imprecise, with 30-50% of patients recurring after image-guided radiotherapy or radical prostatectomy. We aimed to test combined genomic and microenvironmental indices in prostate cancer to improve risk stratification and complement clinical prognostic factors. METHODS We used DNA-based indices alone or in combination with intra-prostatic hypoxia measurements to develop four prognostic indices in 126 low-risk to intermediate-risk patients (Toronto cohort) who will receive image-guided radiotherapy. We validated these indices in two independent cohorts of 154 (Memorial Sloan Kettering Cancer Center cohort [MSKCC] cohort) and 117 (Cambridge cohort) radical prostatectomy specimens from low-risk to high-risk patients. We applied unsupervised and supervised machine learning techniques to the copy-number profiles of 126 pre-image-guided radiotherapy diagnostic biopsies to develop prognostic signatures. Our primary endpoint was the development of a set of prognostic measures capable of stratifying patients for risk of biochemical relapse 5 years after primary treatment. FINDINGS Biochemical relapse was associated with indices of tumour hypoxia, genomic instability, and genomic subtypes based on multivariate analyses. We identified four genomic subtypes for prostate cancer, which had different 5-year biochemical relapse-free survival. Genomic instability is prognostic for relapse in both image-guided radiotherapy (multivariate analysis hazard ratio [HR] 4·5 [95% CI 2·1-9·8]; p=0·00013; area under the receiver operator curve [AUC] 0·70 [95% CI 0·65-0·76]) and radical prostatectomy (4·0 [1·6-9·7]; p=0·0024; AUC 0·57 [0·52-0·61]) patients with prostate cancer, and its effect is magnified by intratumoral hypoxia (3·8 [1·2-12]; p=0·019; AUC 0·67 [0·61-0·73]). A novel 100-loci DNA signature accurately classified treatment outcome in the MSKCC low-risk to intermediate-risk cohort (multivariate analysis HR 6·1 [95% CI 2·0-19]; p=0·0015; AUC 0·74 [95% CI 0·65-0·83]). In the independent MSKCC and Cambridge cohorts, this signature identified low-risk to high-risk patients who were most likely to fail treatment within 18 months (combined cohorts multivariate analysis HR 2·9 [95% CI 1·4-6·0]; p=0·0039; AUC 0·68 [95% CI 0·63-0·73]), and was better at predicting biochemical relapse than 23 previously published RNA signatures. INTERPRETATION This is the first study of cancer outcome to integrate DNA-based and microenvironment-based failure indices to predict patient outcome. Patients exhibiting these aggressive features after biopsy should be entered into treatment intensification trials. FUNDING Movember Foundation, Prostate Cancer Canada, Ontario Institute for Cancer Research, Canadian Institute for Health Research, NIHR Cambridge Biomedical Research Centre, The University of Cambridge, Cancer Research UK, Cambridge Cancer Charity, Prostate Cancer UK, Hutchison Whampoa Limited, Terry Fox Research Institute, Princess Margaret Cancer Centre Foundation, PMH-Radiation Medicine Program Academic Enrichment Fund, Motorcycle Ride for Dad (Durham), Canadian Cancer Society.


Clinical Cancer Research | 2012

Tumor hypoxia predicts biochemical failure following radiotherapy for clinically localized prostate cancer

Michael Milosevic; Padraig Warde; Cynthia Ménard; Peter Chung; Ants Toi; Adrian Ishkanian; M. McLean; Melania Pintilie; Jenna Sykes; Mary Gospodarowicz; Charles Catton; Richard P. Hill; Robert E. Bristow

Purpose: Tumor hypoxia is an important determinant of outcome in many human malignancies and is associated with treatment resistance and metastases. The aim of this study was to determine the effect of hypoxia in patients with prostate cancer treated with radiotherapy. Experimental Design: Tumor hypoxia was measured in 247 patients with clinically localized prostate cancer before radiotherapy, with or without hormonal therapy. The median pO2 was 6.8 mm Hg and the median hypoxic percentage less than 10 mm Hg (HP10) was 63%. The median follow-up was 6.6 years. Results: The 5-year biochemical relapse-free rate (bRFR) was 78%. Prostrate-specific antigen and Gleason score were both associated with biochemical relapse and formed a baseline clinical model. The effect of hypoxia was found to vary with the duration of patient follow-up. HP10, when added to the clinical model, was an independent predictor of early bRFR (P = 0.019). The relationship between hypoxia and early bRFR was more pronounced when the analysis was restricted to 142 patients with bulk tumor at the site of the oxygen measurements (P = 0.004). Hypoxia was the only factor predictive of local recurrence in 70 patients who had biopsies conducted during follow-up (P = 0.043), again with the effect being greatest early after completing treatment. Conclusions: This is the largest clinical study of prostate cancer hypoxia with direct measurement of tumor oxygen levels. It shows that hypoxia is associated with early biochemical relapse after radiotherapy and also with local recurrence in the prostate gland. Clin Cancer Res; 18(7); 2108–14. ©2012 AACR.


Cancer | 2012

Copy number alterations of c-MYC and PTEN are prognostic factors for relapse after prostate cancer radiotherapy

Gaetano Zafarana; Adrian Ishkanian; Chad A. Malloff; Jennifer A. Locke; Jenna Sykes; John Thoms; Wan L. Lam; Jeremy A. Squire; Maisa Yoshimoto; Varune Rohan Ramnarine; Alice Meng; Igor Jurisca; Michael Milosevic; Melania Pintilie; Theo H. van der Kwast; Robert G. Bristow

Despite the use of PSA, Gleason score, and T‐category as prognosticators in intermediate‐risk prostate cancer, 20–40% of patients will fail local therapy. In order to optimize treatment approaches for intermediate‐risk patients, additional genetic prognosticators are needed. Previous reports using array comparative genomic hybridization (aCGH) in radical prostatectomy cohorts suggested a combination of allelic loss of the PTEN gene on 10q and allelic gain of the c‐MYC gene on 8q were associated with metastatic disease. We tested whether copy number alterations (CNAs) in PTEN (allelic loss) and c‐MYC (allelic gain) were associated with biochemical relapse following modern‐era, image‐guided radiotherapy (mean dose 76.4 Gy). We used aCGH analyses validated by fluorescence in‐situ hybridization (FISH) of DNA was derived from frozen, pre‐treatment biopsies in 126 intermediate‐risk prostate cancer patients. Patients whose tumors had CNAs in both PTEN and c‐MYC had significantly increased genetic instability (percent genome alteration; PGA) compared to tumors with normal PTEN and c‐MYC status (p < 0.0001). We demonstrate that c‐MYC gain alone, or combined c‐MYC gain and PTEN loss, were increasingly prognostic for relapse on multivariable analyses (hazard ratios (HR) of 2.58/p = 0.005 and 3.21/p = 0.0004; respectively). Triaging patients by the use of CNAs within pre‐treatment biopsies may allow for better use of systemic therapies to target sub‐clinical metastases or locally recurrent disease and improve clinical outcomes. Cancer 2012.


The Prostate | 2009

High-resolution array CGH identifies novel regions of genomic alteration in intermediate-risk prostate cancer

Adrian Ishkanian; Chad Mallof; J. Ho; Alice Meng; Monique Albert; Amena Syed; Theodorus van der Kwast; M. Milosevic; Maisa Yoshimoto; Jeremy A. Squire; Wan L. Lam; Robert G. Bristow

Approximately one‐third of prostate cancer patients present with intermediate risk disease. Interestingly, while this risk group is clinically well defined, it demonstrates the most significant heterogeneity in PSA‐based biochemical outcome. Further, the majority of candidate genes associated with prostate cancer progression have been identified using cell lines, xenograft models, and high‐risk androgen‐independent or metastatic patient samples. We used a global high‐resolution array comparative genomic hybridization (CGH) assay to characterize copy number alterations (CNAs) in intermediate risk prostate cancer. Herein, we show this risk group contains a number of alterations previously associated with high‐risk disease: (1) deletions at 21q22.2 (TMPRSS2:ERG), 16q22–24 (containing CDH1), 13q14.2 (RB1), 10q23.31 (PTEN), 8p21 (NKX3.1); and, (2) amplification at 8q21.3–24.3 (containing c‐MYC). In addition, we identified six novel microdeletions at high frequency: 1q42.12–q42.3 (33.3%), 5q12.3–13.3 (21%), 20q13.32–13.33 (29.2%), 22q11.21 (25%), 22q12.1 (29.2%), and 22q13.31 (33.3%). Further, we show there is little concordance between CNAs from these clinical samples and those found in commonly used prostate cancer cell models. These unexpected findings suggest that the intermediate‐risk category is a crucial cohort warranting further study to determine if a unique molecular fingerprint can predict aggressive versus indolent phenotypes. Prostate 69:1091–1100, 2009.


Genes, Chromosomes and Cancer | 2004

Novel Regions of Amplification on 8q Distinct from the MYC Locus and Frequently Altered in Oral Dysplasia and Cancer

Cathie Garnis; Bradley P. Coe; Adrian Ishkanian; Lewei Zhang; Miriam P. Rosin; Wan L. Lam

Genetic studies aimed at identifying key alterations in oral cancers have focused on analysis of tumors, with few such studies using early oral premalignant lesions (OPLs) because of limitations in both sample availability and size. In this study, we used a randomly amplified polymorphic DNA (RAPD)‐PCR approach to fingerprint DNA from microdissected normal and dysplastic cells and identified two recurrent genetic alterations on the long arm of chromosome 8 in OPLs, one mapping to 8q22 and the other to 8q24 near the MYC locus. We constructed a high‐resolution bacterial artificial chromosome (BAC) comparative genomic hybridization array consisting of 166 overlapping BAC clones that spans about 52 Mbp, from 8q21 to 8q24. Hybridization of DNA from microdissected oral tumors to the array revealed alteration at 8q24, with amplification of the BAC containing MYC. Strikingly, at least two other novel regions of amplification at 8q22 were identified. Microsatellite analysis of 93 oral dysplasias and tumors confirmed the presence of one of the alterations at 8q22. Loss of heterozygosity (LOH) at D8S1830, mapping within one of the regions of amplification, was observed in high frequency in both OPLs and tumors. Of the 37 cases with LOH at D8S1830, 23 (62%) showed retention at D8S1793, which maps 1.6 Mbp centromeric to MYC. This is further support for the alteration at 8q22 being distinct from MYC. These data raise the possibility of additional oncogenes on 8q near the MYC locus that are potentially involved in OPL disease progression.


Modern Pathology | 2002

An Efficient Method for the Assessment of DNA Quality of Archival Microdissected Specimens

Arek Siwoski; Adrian Ishkanian; Cathie Garnis; Lewei Zhang; Miriam P. Rosin; Wan L. Lam

There will be an increasing need of methods for assessing the suitability of specimens for genetic-based assays as DNA markers become an integral part of molecular diagnosis. The targeting of specimens for specific analyses will require the ability to rapidly screen for DNA quality. Conventional methods such as Southern analysis and gene specific-polymerase chain reaction (PCR) often require quantities of material that represent a significant portion of the specimen, especially in microdissected samples. Here we describe a novel application of a commonly used PCR-based DNA-fingerprinting technology that requires minimal quantities of DNA to simultaneously assess multiple regions throughout the genome for DNA quality. Randomly amplified polymorphic DNA (RAPD) PCR generates DNA fragments of a broad size range with the product size reflecting the degree of sample fragmentation. Fourteen DNA samples extracted from cells microdissected from seven formalin-fixed, paraffin-embedded oral cancer biopsies were assessed for DNA quality using gene-specific PCR and RAPD-PCR. Although the more conventional assay required 2-ng DNA (or 300-cell equivalents) to examine DNA quality at a single locus, RAPD-PCR provided a more informative profile of DNA quality from the same microdissected archival specimens.


Acta Oncologica | 2010

Array CGH as a potential predictor of radiocurability in intermediate risk prostate cancer

Adrian Ishkanian; Gaetano Zafarana; John Thoms; Robert G. Bristow

Abstract Prostate cancer is the most common male cancer and up to one fifth of diagnosed patients will die of their disease. Current prognostic variables including T-category (of the TNM staging), the absolute or kinetics of prostatic specific antigen (PSA) and the pathologic Gleason score (GS) are utilized to place men in low, intermediate and high-risk prostate cancer risk groupings. There is great heterogeneity within the non-indolent intermediate risk group with respect to clinical response. It is therefore imperative that further genetic and other prognostic factors be identified to better individualize treatment. Somatic alterations in prostate cancer. Herein, we review the potential for somatic alterations in tumor-associated genes (based on comparative genomic hybridization (CGH) in prostate cancers to be novel prognostic, and possibly predictive, factors for prostate cancer radiotherapy response. Intermediate risk prostate cancers show alterations in a number of genes thought to be involved in radiosensitivity, DNA repair, cell death and stem cell renewal. These include deletions at 21q (TMPRSS2: ERG), 13q (RB1), 10q (PTEN), 8p (NKX3.1), additions at 8q21 (containing c-Myc)) and haplo-insufficiency for p53, PARP1, ATM and DNA-PKcs. Conclusions. The use of high-resolution CGH for fine-mapping of deletions and amplifications in pre-radiotherapy prostate cancer biopsies is feasible. Genetic alterations may delineate localized prostate cancer from systemic disease and be used as a predictive factor in that patients would be individually triaged to local (surgery versus radiotherapy) and/or adjuvant (adjuvant androgen ablation or post-operative radiotherapy) therapies in a prospective fashion to improve outcome. The knowledge of abnormal DNA repair pathways within in a given patient could allow for the judicious use of targeted agents (PARP/ATM inhibitors) as personalized medicine.


Clinical Cancer Research | 2012

NKX3.1 Haploinsufficiency Is Prognostic for Prostate Cancer Relapse following Surgery or Image-Guided Radiotherapy

Jennifer A. Locke; Gaetano Zafarana; Adrian Ishkanian; Michael Milosevic; John Thoms; Cherry Have; Chad A. Malloff; Wan L. Lam; Jeremy A. Squire; Melania Pintilie; Jenna Sykes; Varune Rohan Ramnarine; Alice Meng; Omer Ahmed; Igor Jurisica; Theo H. van der Kwast; Robert G. Bristow

Background: Despite the use of prostate specific antigen (PSA), Gleason-score, and T-category as prognostic factors, up to 40% of patients with intermediate-risk prostate cancer will fail radical prostatectomy or precision image-guided radiotherapy (IGRT). Additional genetic prognosticators are needed to triage these patients toward intensified combination therapy with novel targeted therapeutics. We tested the role of the NKX3.1 gene as a determinant of treatment outcome given its reported roles in tumor initiating cell (TIC) renewal, the DNA damage response, and cooperation with c-MYC during prostate cancer progression. Methods: Using high-resolution array comparative genomic hybridization (aCGH), we profiled the copy number alterations in TIC genes using tumor DNA from frozen needle biopsies derived from 126 intermediate-risk patients who underwent IGRT. These data were correlated to biochemical relapse-free rate (bRFR) by the Kaplan–Meier method and Cox proportional hazards models. Results: A screen of the aCGH-IGRT data for TIC genes showed frequent copy number alterations for NKX3.1, PSCA, and c-MYC. NKX3.1 haploinsufficiency was associated with increased genomic instability independent of PSA, T-category, and Gleason-score. After adjusting for clinical factors in a multivariate model, NKX3.1 haploinsufficiency was associated with bRFR when tested alone (HR = 3.05, 95% CI: 1.46–6.39, P = 0.0030) or when combined with c-MYC gain (HR = 3.88, 95% CI: 1.78–8.49, P = 0.00067). A similar association was observed for patients following radical prostatectomy with a public aCGH database. NKX3.1 status was associated with positive biopsies post-IGRT and increased clonogen radioresistance in vitro. Conclusions: Our results support the use of genomic predictors, such as NKX3.1 status, in needle biopsies for personalized approaches to prostate cancer management. Clin Cancer Res; 18(1); 308–16. ©2011 AACR.


Oncotarget | 2016

Association of multiparametric MRI quantitative imaging features with prostate cancer gene expression in MRI-targeted prostate biopsies

Radka Stoyanova; Alan Pollack; Mandeep Takhar; Charles M. Lynne; Nestor A. Parra; Lucia L.C. Lam; Mohammed Alshalalfa; Christine Buerki; Rosa Castillo; Merce Jorda; Hussam Al-Deen Ashab; Oleksandr N. Kryvenko; Sanoj Punnen; Dipen J. Parekh; M.C. Abramowitz; Robert J. Gillies; Elai Davicioni; Nicholas Erho; Adrian Ishkanian

Standard clinicopathological variables are inadequate for optimal management of prostate cancer patients. While genomic classifiers have improved patient risk classification, the multifocality and heterogeneity of prostate cancer can confound pre-treatment assessment. The objective was to investigate the association of multiparametric (mp)MRI quantitative features with prostate cancer risk gene expression profiles in mpMRI-guided biopsies tissues. Global gene expression profiles were generated from 17 mpMRI-directed diagnostic prostate biopsies using an Affimetrix platform. Spatially distinct imaging areas (‘habitats’) were identified on MRI/3D-Ultrasound fusion. Radiomic features were extracted from biopsy regions and normal appearing tissues. We correlated 49 radiomic features with three clinically available gene signatures associated with adverse outcome. The signatures contain genes that are over-expressed in aggressive prostate cancers and genes that are under-expressed in aggressive prostate cancers. There were significant correlations between these genes and quantitative imaging features, indicating the presence of prostate cancer prognostic signal in the radiomic features. Strong associations were also found between the radiomic features and significantly expressed genes. Gene ontology analysis identified specific radiomic features associated with immune/inflammatory response, metabolism, cell and biological adhesion. To our knowledge, this is the first study to correlate radiogenomic parameters with prostate cancer in men with MRI-guided biopsy.

Collaboration


Dive into the Adrian Ishkanian's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chad A. Malloff

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wan L. Lam

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jenna Sykes

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

John Thoms

University Health Network

View shared research outputs
Top Co-Authors

Avatar

M. Milosevic

University Health Network

View shared research outputs
Researchain Logo
Decentralizing Knowledge