Network


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

Hotspot


Dive into the research topics where Alan Dal Pra is active.

Publication


Featured researches published by Alan Dal Pra.


Nature Genetics | 2015

Spatial genomic heterogeneity within localized, multifocal prostate cancer

Paul C. Boutros; Michael Fraser; Nicholas J. Harding; Richard de Borja; Dominique Trudel; Emilie Lalonde; Alice Meng; Pablo H. Hennings-Yeomans; Andrew McPherson; Veronica Y. Sabelnykova; Amin Zia; Natalie S. Fox; Julie Livingstone; Yu Jia Shiah; Jianxin Wang; Timothy Beck; Cherry Have; Taryne Chong; Michelle Sam; Jeremy Johns; Lee Timms; Nicholas Buchner; Ada Wong; John D. Watson; Trent T. Simmons; Christine P'ng; Gaetano Zafarana; Francis Nguyen; Xuemei Luo; Kenneth C. Chu

Herein we provide a detailed molecular analysis of the spatial heterogeneity of clinically localized, multifocal prostate cancer to delineate new oncogenes or tumor suppressors. We initially determined the copy number aberration (CNA) profiles of 74 patients with index tumors of Gleason score 7. Of these, 5 patients were subjected to whole-genome sequencing using DNA quantities achievable in diagnostic biopsies, with detailed spatial sampling of 23 distinct tumor regions to assess intraprostatic heterogeneity in focal genomics. Multifocal tumors are highly heterogeneous for single-nucleotide variants (SNVs), CNAs and genomic rearrangements. We identified and validated a new recurrent amplification of MYCL, which is associated with TP53 deletion and unique profiles of DNA damage and transcriptional dysregulation. Moreover, we demonstrate divergent tumor evolution in multifocal cancer and, in some cases, tumors of independent clonal origin. These data represent the first systematic relation of intraprostatic genomic heterogeneity to predicted clinical outcome and inform the development of novel biomarkers that reflect individual prognosis.


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

BACKGROUNDnClinical 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.nnnMETHODSnWe 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.nnnFINDINGSnBiochemical 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.nnnINTERPRETATIONnThis 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.nnnFUNDINGnMovember 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.


Nature | 2017

Genomic hallmarks of localized, non-indolent prostate cancer

Michael Fraser; Veronica Y. Sabelnykova; Takafumi N. Yamaguchi; Lawrence E. Heisler; Julie Livingstone; Vincent Huang; Yu Jia Shiah; Fouad Yousif; Xihui Lin; Andre P. Masella; Natalie S. Fox; Michael Xie; Stephenie D. Prokopec; Alejandro Berlin; Emilie Lalonde; Musaddeque Ahmed; Dominique Trudel; Xuemei Luo; Timothy Beck; Alice Meng; Junyan Zhang; Alister D'Costa; Robert E. Denroche; Haiying Kong; Shadrielle Melijah G. Espiritu; Melvin Lee Kiang Chua; Ada Wong; Taryne Chong; Michelle Sam; Jeremy Johns

Prostate tumours are highly variable in their response to therapies, but clinically available prognostic factors can explain only a fraction of this heterogeneity. Here we analysed 200 whole-genome sequences and 277 additional whole-exome sequences from localized, non-indolent prostate tumours with similar clinical risk profiles, and carried out RNA and methylation analyses in a subset. These tumours had a paucity of clinically actionable single nucleotide variants, unlike those seen in metastatic disease. Rather, a significant proportion of tumours harboured recurrent non-coding aberrations, large-scale genomic rearrangements, and alterations in which an inversion repressed transcription within its boundaries. Local hypermutation events were frequent, and correlated with specific genomic profiles. Numerous molecular aberrations were prognostic for disease recurrence, including several DNA methylation events, and a signature comprised of these aberrations outperformed well-described prognostic biomarkers. We suggest that intensified treatment of genomically aggressive localized prostate cancer may improve cure rates.


Clinical Cancer Research | 2013

Reprogramming Metabolism with Metformin Improves Tumor Oxygenation and Radiotherapy Response

Vanessa E. Zannella; Alan Dal Pra; Hala Muaddi; Trevor D. McKee; Shawn Stapleton; Jenna Sykes; Rachel Glicksman; Selim Chaib; Paul Zamiara; Michael Milosevic; Bradly G. Wouters; Robert G. Bristow; Marianne Koritzinsky

Purpose: Tumor hypoxia is a negative prognostic factor in multiple cancers, due in part to its role in causing resistance to radiotherapy. Hypoxia arises in tumor regions distal to blood vessels as oxygen is consumed by more proximal tumor cells. Reducing the rate of oxygen consumption is therefore a potential strategy to reduce tumor hypoxia. We hypothesized that the anti-diabetic drug metformin, which reduces oxygen consumption through inhibition of mitochondrial complex I, would improve radiation response by increasing tumor oxygenation. Experimental Design: Tumor hypoxia was measured in xenografts before and after metformin treatment using 2-nitroimidazole hypoxia markers quantified by immunohistochemistry (IHC), flow cytometry, and positron emission tomography (PET) imaging. Radiation response was determined by tumor growth delay and clonogenic survival in xenografts with and without administration of metformin. The impact of metformin use on outcome was assessed in 504 patients with localized prostate cancer treated with curative-intent, image-guided radiotherapy (IGRT) from 1996 to 2012. Three-year biochemical relapse-free rates were assessed using the Kaplan–Meier method. Results: Metformin treatment significantly improved tumor oxygenation in two xenograft models as measured by IHC, flow cytometry, and PET imaging. Metformin also led to improved radiotherapy responses when mice were administered metformin immediately before irradiation. Clinically, metformin use was associated with an independent and significant decrease in early biochemical relapse rates (P = 0.0106). Conclusion: Our data demonstrate that metformin can improve tumor oxygenation and response to radiotherapy. Our study suggests that metformin may represent an effective and inexpensive means to improve radiotherapy outcome with an optimal therapeutic ratio. Clin Cancer Res; 19(24); 6741–50. ©2013 AACR.


Clinical Cancer Research | 2013

TMPRSS2-ERG Status Is Not Prognostic Following Prostate Cancer Radiotherapy: Implications for Fusion Status and DSB Repair

Alan Dal Pra; Emilie Lalonde; Jenna Sykes; Fiona Warde; Adrian Ishkanian; Alice Meng; Chad Maloff; John R. Srigley; Anthony M. Joshua; Gyorgy Petrovics; Theodorus H. van der Kwast; Andrew Evans; Michael Milosevic; Fred Saad; Colin Collins; Jeremy A. Squire; Wan L. Lam; Tarek A. Bismar; Paul C. Boutros; Robert G. Bristow

Background: Preclinical data suggest that TMPRSS2-ERG gene fusions, present in about 50% of prostate cancers, may be a surrogate for DNA repair status and therefore a biomarker for DNA-damaging agents. To test this hypothesis, we examined whether TMPRSS2-ERG status was associated with biochemical failure after clinical induction of DNA damage following image-guided radiotherapy (IGRT). Methods: Pretreatment biopsies from two cohorts of patients with intermediate-risk prostate cancer [T1/T2, Gleason score (GS) < 8, prostate-specific antigen (PSA) < 20 ng/mL; >7 years follow-up] were analyzed: (i) 126 patients [comparative genomic hybridization (CGH) cohort] with DNA samples assayed by array CGH (aCGH) for the TMPRSS2-ERG fusion; and (ii) 118 patients [immunohistochemical (IHC) cohort] whose biopsy samples were scored within a defined tissue microarray (TMA) immunostained for ERG overexpression (known surrogate for TMPRSS2-ERG fusion). Patients were treated with IGRT with a median dose of 76 Gy. The potential role of TMPRSS2-ERG status as a prognostic factor for biochemical relapse-free rate (bRFR; nadir + 2 ng/mL) was evaluated in the context of clinical prognostic factors in multivariate analyses using a Cox proportional hazards model. Results: TMPRSS2-ERG fusion by aCGH was identified in 27 (21%) of the cases in the CGH cohort, and ERG overexpression was found in 59 (50%) patients in the IHC cohort. In both cohorts, TMPRSS2-ERG status was not associated with bRFR on univariate or multivariate analysis. Conclusions: In two similarly treated IGRT cohorts, TMPRSS2-ERG status was not prognostic for bRFR, in disagreement with the hypothesis that these prostate cancers have DNA repair defects that render them clinically more radiosensitive. TMPRSS2-ERG is therefore unlikely to be a predictive factor for IGRT response. Clin Cancer Res; 19(18); 5202–9. ©2013 AACR.


Advances in Experimental Medicine and Biology | 2014

Intratumoral hypoxia as the genesis of genetic instability and clinical prognosis in prostate cancer

Daria Taiakina; Alan Dal Pra; Robert G. Bristow

Intratumoral hypoxia is prevalent in many solid tumors and is a marker of poor clinical prognosis in prostate cancer. The presence of hypoxia is associated with increased chromosomal instability, gene amplification, downregulation of DNA damage repair pathways, and altered sensitivity to agents that damage DNA. These genomic changes could also lead to oncogene activation or tumor suppressor gene inactivation during prostate cancer progression. We review here the concept of repair-deficient hypoxic tumor cells that can adapt to low oxygen levels and acquire an aggressive unstable mutator phenotype. We speculate that hypoxia-induced genomic instability may also be a consequence of aberrant mitotic function in hypoxic cells, which leads to increased chromosomal instability and aneuploidy. Because both hypoxia and aneuploidy are prognostic factors in prostate cancer, a greater understanding of these biological states in prostate cancer may lead to novel prognostic and predictive tests and drive new therapeutic strategies in the context of personalized cancer medicine.


Nature Reviews Urology | 2015

Synergistic action of image-guided radiotherapy and androgen deprivation therapy

Jennifer A. Locke; Alan Dal Pra; S. Supiot; Padraig Warde; Robert G. Bristow

The combined use of androgen deprivation therapy (ADT) and image-guided radiotherapy (IGRT) can improve overall survival in aggressive, localized prostate cancer. However, owing to the adverse effects of prolonged ADT, it is imperative to identify the patients who would benefit from this combined-modality therapy relative to the use of IGRT alone. Opportunities exist for more personalized approaches in treating aggressive, locally advanced prostate cancer. Biomarkers—such as disseminated tumour cells, circulating tumour cells, genomic signatures and molecular imaging techniques—could identify the patients who are at greatest risk for systemic metastases and who would benefit from the addition of systemic ADT. By contrast, when biomarkers of systemic disease are not present, treatment could proceed using local IGRT alone. The choice of drug, treatment duration and timing of ADT relative to IGRT could be predicated on these personalized approaches to prostate cancer medicine. These novel treatment intensification and reduction strategies could result in improved prostate-cancer-specific survival and overall survival, without incurring the added expense of metabolic syndrome and other adverse effects of ADT in all patients.


European Urology | 2017

A Prostate Cancer “Nimbosus”: Genomic Instability and SChLAP1 Dysregulation Underpin Aggression of Intraductal and Cribriform Subpathologies

Melvin Lee Kiang Chua; Winnie Lo; Melania Pintilie; Jure Murgic; Emilie Lalonde; Vinayak Bhandari; Osman Mahamud; Anuradha Gopalan; Charlotte F. Kweldam; Geert J.L.H. van Leenders; Esther I. Verhoef; Agnes Marije Hoogland; Julie Livingstone; Alejandro Berlin; Alan Dal Pra; Alice Meng; Junyan Zhang; Michèle Orain; Valerie Picard; Hélène Hovington; Alain Bergeron; Louis Lacombe; Yves Fradet; Bernard Têtu; Victor E. Reuter; Neil Fleshner; Michael Fraser; Paul C. Boutros; Theodorus H. van der Kwast; Robert G. Bristow

BACKGROUNDnIntraductal carcinoma (IDC) and cribriform architecture (CA) represent unfavorable subpathologies in localized prostate cancer. We recently showed that IDC shares a clonal ancestry with the adjacent glandular adenocarcinoma.nnnOBJECTIVEnWe investigated for the co-occurrence of aggression factors, genomic instability and hypoxia, and performed gene expression profiling of these tumors.nnnDESIGN, SETTING, AND PARTICIPANTSnA total of 1325 men were treated for localized prostate cancer from four academic institutions (University Health Network, CHU de Québec-Université Laval, Memorial Sloan Kettering Cancer Center [MSKCC], and Erasmus Medical Center). Pathological specimens were centrally reviewed. Gene copy number and expression, and intraprostatic oxygenation were assessed.nnnOUTCOME MEASUREMENTS AND STATISTICAL ANALYSISnIDC/CA was separately assessed for biochemical relapse risk in the Canadian and MSKCC cohorts. Both cohorts were pooled for analyses on metastasis.nnnRESULTS AND LIMITATIONnPresence of IDC/CA independently predicted for increased risks of biochemical relapse (HRCanadian 2.17, p<0.001; HRMSKCC 2.32, p=0.0035) and metastasis (HRpooled 3.31, p<0.001). IDC/CA+ cancers were associated with an increased percentage of genome alteration (PGA [median] 7.2 vs 3.0, p<0.001), and hypoxia (64.0% vs 45.5%, p=0.17). Combinatorial genomic-pathological indices offered the strongest discrimination for metastasis (C-index 0.805 [clinical+IDC/CA+PGA] vs 0.786 [clinical+IDC/CA] vs 0.761 [clinical]). Profiling of mRNA abundance revealed that long noncoding RNA, SChLAP1, was the only gene expressed at >3-fold higher (p<0.0001) in IDC/CA+ than in IDC/CA- tumors, independently corroborated by increased SChLAP1 RNA in situ hybridization signal. Optimal treatment intensification for IDC/CA+ prostate cancer requires prospective testing.nnnCONCLUSIONSnThe poor outcome associated with IDC and CA subpathologies is associated with a constellation of genomic instability, SChLAP1 expression, and hypoxia. We posit a novel concept in IDC/CA+ prostate cancer, nimbosus (gathering of stormy clouds, Latin), which manifests as increased metastatic capacity and lethality.nnnPATIENT SUMMARYnA constellation of unfavorable molecular characteristics co-occur with intraductal and cribriform subpathologies in prostate cancer. Modern imaging for surveillance and treatment intensification trials should be considered in this adverse subgroup.


Frontiers in Oncology | 2016

Mechanistic Insights into Molecular Targeting and Combined Modality Therapy for Aggressive, Localized Prostate Cancer

Alan Dal Pra; Jennifer A. Locke; Gerben R. Borst; S. Supiot; Robert G. Bristow

Radiation therapy (RT) is one of the mainstay treatments for prostate cancer (PCa). The potentially curative approaches can provide satisfactory results for many patients with non-metastatic PCa; however, a considerable number of individuals may present disease recurrence and die from the disease. Exploiting the rich molecular biology of PCa will provide insights into how the most resistant tumor cells can be eradicated to improve treatment outcomes. Important for this biology-driven individualized treatment is a robust selection procedure. The development of predictive biomarkers for RT efficacy is therefore of utmost importance for a clinically exploitable strategy to achieve tumor-specific radiosensitization. This review highlights the current status and possible opportunities in the modulation of four key processes to enhance radiation response in PCa by targeting the: (1) androgen signaling pathway; (2) hypoxic tumor cells and regions; (3) DNA damage response (DDR) pathway; and (4) abnormal extra-/intracell signaling pathways. In addition, we discuss how and which patients should be selected for biomarker-based clinical trials exploiting and validating these targeted treatment strategies with precision RT to improve cure rates in non-indolent, localized PCa.


European Urology | 2016

Re: Ronald C. Chen. Postprostatectomy Radiotherapy: Whether and How Long to Give Concurrent Androgen Deprivation Therapy. Eur Urol. In press. http://dx.doi.org/10.1016/j.eururo.2015.05.036: Which patients need treatment intensification?

Alan Dal Pra; Martin Spahn

The study by Jackson et al in this month’s issue of European Urology [1] addresses a clinically relevant question of whether adding androgen deprivation therapy (ADT) to postprostatectomy radiotherapy (RT) improves cancer control and survival. Furthermore, the study examines whether duration of ADT is associated with these outcomes. This question is commonly faced by practicing urologists, radiation oncologists, and medical oncologists but has not been well studied. Consequently, current practice is guided partly by studies that examined the use of RT with ADT in the primary treatment setting. Primary RT with concurrent ADT for patients with aggressive prostate cancer is, by far, the best-studied treatment regimenfor this disease, with multiple randomized trials demonstrating that ADT added to RT improves overall survival compared with RT alone [2]. In addition, long-term ADT (2–3 yr) added to RT is associated with improved overall survival compared with short-term ADT (4–6 mo) with RT [3,4]. Why does ADT improve survival? A possibility is that ADT potentiates the local tumoricidal effect of radiation. Indeed, a common criticism of the abovementioned randomized trials is that the radiation doses used (commonly 65–70 Gy) were lower than standard doses used today ( 75.6 Gy). However, higher doses of radiation compared with lower doses have never been able to demonstrate the survival benefit that ADT has demonstrated [2]. Consequently, the survival benefit from ADT when added to RT is at least partly due to its effect on distant micrometastatic disease. If this were true, then the survival benefit from ADT that has been repeatedly demonstrated in the primary treatment setting should also be demonstrable in the postprostatectomy

Collaboration


Dive into the Alan Dal Pra's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alejandro Berlin

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Alice Meng

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Emilie Lalonde

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Jenna Sykes

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Julie Livingstone

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Michael Fraser

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Michael Milosevic

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Paul C. Boutros

Ontario Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Gaetano Zafarana

Princess Margaret Cancer Centre

View shared research outputs
Researchain Logo
Decentralizing Knowledge