Network


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

Hotspot


Dive into the research topics where Gillian Vandekerkhove is active.

Publication


Featured researches published by Gillian Vandekerkhove.


European Urology | 2017

Treatment Outcomes and Tumor Loss of Heterozygosity in Germline DNA Repair–deficient Prostate Cancer

Matti Annala; Werner J. Struss; Evan W. Warner; Kevin Beja; Gillian Vandekerkhove; Amanda Wong; Daniel Khalaf; Irma-Liisa Seppälä; Alan So; Gregory Lo; Rahul Aggarwal; Eric J. Small; Matti Nykter; Martin Gleave; Kim N. Chi; Alexander W. Wyatt

BACKGROUND Germline mutations in DNA repair genes were recently reported in 8-12% of patients with metastatic castration-resistant prostate cancer (mCRPC). It is unknown whether these mutations associate with differential response to androgen receptor (AR)-directed therapy. OBJECTIVE To determine the clinical response of mCRPC patients with germline DNA repair defects to AR-directed therapies and to establish whether biallelic DNA repair gene loss is detectable in matched circulating tumor DNA (ctDNA). DESIGN, SETTING, AND PARTICIPANTS We recruited 319 mCRPC patients and performed targeted germline sequencing of 22 DNA repair genes. In patients with deleterious germline mutations, plasma cell-free DNA was also sequenced. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Prostate-specific antigen response and progression were assessed in relation to initial androgen deprivation therapy (ADT) and subsequent therapy for mCRPC using Kaplan-Meier analysis. RESULTS AND LIMITATIONS Of the 319 patients, 24 (7.5%) had deleterious germline mutations, with BRCA2 (n=16) being the most frequent. Patients (n=22) with mutations in genes linked to homologous recombination were heterogeneous at initial presentation but, after starting ADT, progressed to mCRPC with a median time of 11.8 mo (95% confidence interval [CI] 5.1-18.4). The median time to prostate-specific antigen progression on first-line AR-targeted therapy in the mCRPC setting was 3.3 mo (95% CI 2.7-3.9). Ten out of 11 evaluable patients with germline BRCA2 mutations had somatic deletion of the intact allele in ctDNA. A limitation of this study is absence of a formal control cohort for comparison of clinical outcomes. CONCLUSIONS Patients with mCRPC who have germline DNA repair defects exhibit attenuated responses to AR-targeted therapy. Biallelic gene loss was robustly detected in ctDNA, suggesting that this patient subset could be prioritized for therapies exploiting defective DNA repair using a liquid biopsy. PATIENT SUMMARY Patients with metastatic prostate cancer and germline DNA repair defects exhibit a poor response to standard hormonal therapies, but may be prioritized for potentially more effective therapies using a blood test.


Clinical Cancer Research | 2017

Circulating Tumor DNA Reveals Clinically Actionable Somatic Genome of Metastatic Bladder Cancer

Gillian Vandekerkhove; Tilman Todenhöfer; Matti Annala; Werner J. Struss; Amanda Wong; Kevin Beja; Elie Ritch; Sonal Brahmbhatt; Stanislav Volik; Jörg Hennenlotter; Matti Nykter; Kim N. Chi; Scott North; A. Stenzl; Colin Collins; Bernhard J. Eigl; Peter C. Black; Alexander W. Wyatt

Purpose: Targeted agents and immunotherapies promise to transform the treatment of metastatic bladder cancer, but therapy selection will depend on practical tumor molecular stratification. Circulating tumor DNA (ctDNA) is established in several solid malignancies as a minimally invasive tool to profile the tumor genome in real-time, but is critically underexplored in bladder cancer. Experimental Design: We applied a combination of whole-exome sequencing and targeted sequencing across 50 bladder cancer driver genes to plasma cell-free DNA (cfDNA) from 51 patients with aggressive bladder cancer, including 37 with metastatic disease. Results: The majority of patients with metastasis, but only 14% of patients with localized disease, had ctDNA proportions above 2% of total cfDNA (median 16.5%, range 3.9%–72.6%). Twelve percent of estimable samples had evidence of genome hypermutation. We reveal an aggressive mutational landscape in metastatic bladder cancer with 95% of patients harboring deleterious alterations to TP53, RB1, or MDM2, and 70% harboring a mutation or disrupting rearrangement affecting chromatin modifiers such as ARID1A. Targetable alterations in MAPK/ERK or PI3K/AKT/mTOR pathways were robustly detected, including amplification of ERBB2 (20% of patients) and activating hotspot mutations in PIK3CA (20%), with the latter mutually exclusive to truncating mutations in TSC1. A novel FGFR3 gene fusion was identified in consecutive samples from one patient. Conclusions: Our study demonstrates that ctDNA provides a practical and cost-effective snapshot of driver gene status in metastatic bladder cancer. The identification of a wide spectrum of clinically informative somatic alterations nominates ctDNA as a tool to dissect disease pathogenesis and guide therapy selection in patients with metastatic bladder cancer. Clin Cancer Res; 23(21); 6487–97. ©2017 AACR.


Cancer Discovery | 2018

Circulating tumor DNA genomics correlate with resistance to abiraterone and enzalutamide in prostate cancer

Matti Annala; Gillian Vandekerkhove; Daniel Khalaf; Sinja Taavitsainen; Kevin Beja; Evan W. Warner; Katherine Sunderland; Christian Kollmannsberger; Bernhard J. Eigl; Daygen L. Finch; Conrad D. Oja; Joanna Vergidis; Muhammad Zulfiqar; Arun Azad; Matti Nykter; Martin Gleave; Alexander W. Wyatt; Kim N. Chi

Primary resistance to androgen receptor (AR)-directed therapies in metastatic castration-resistant prostate cancer (mCRPC) is poorly understood. We randomized 202 patients with treatment-naïve mCRPC to abiraterone or enzalutamide and performed whole-exome and deep targeted 72-gene sequencing of plasma cell-free DNA prior to therapy. For these agents, which have never been directly compared, time to progression was similar. Defects in BRCA2 and ATM were strongly associated with poor clinical outcomes independently of clinical prognostic factors and circulating tumor DNA abundance. Somatic alterations in TP53, previously linked to reduced tumor dependency on AR signaling, were also independently associated with rapid resistance. Although detection of AR amplifications did not outperform standard prognostic biomarkers, AR gene structural rearrangements truncating the ligand binding domain were identified in several patients with primary resistance. These findings establish genomic drivers of resistance to first-line AR-directed therapy in mCRPC and identify potential minimally invasive biomarkers.Significance: Leveraging plasma specimens collected in a large randomized phase II trial, we report the relative impact of common circulating tumor DNA alterations on patient response to the most widely used therapies for advanced prostate cancer. Our findings suggest that liquid biopsy analysis can guide the use of AR-targeted therapy in general practice. Cancer Discov; 8(4); 444-57. ©2018 AACR.See related commentary by Jayaram et al., p. 392This article is highlighted in the In This Issue feature, p. 371.


The Journal of Pathology | 2018

Biallelic tumour suppressor loss and DNA repair defects in de novo small-cell prostate carcinoma: Molecular dissection of de novo small-cell prostate carcinoma

Edmund Cp Chedgy; Gillian Vandekerkhove; Cameron Herberts; Matti Annala; Adam Donoghue; Michael Sigouros; Elie Ritch; Werner J. Struss; Saki Konomura; Janet Liew; Sunil Parimi; Joanna Vergidis; Antonio Hurtado-Coll; Andrea Sboner; Ladan Fazli; Himisha Beltran; Kim N. Chi; Alexander W. Wyatt

Small‐cell prostate carcinoma (SCPC) is an aggressive malignancy that is managed similarly to small‐cell lung cancer. SCPC can evolve from prostate adenocarcinoma in response to androgen deprivation therapy, but, in rare cases, is present at initial cancer diagnosis. The molecular aetiology of de novo SCPC is incompletely understood, owing to the scarcity of tumour tissue and the short life‐expectancy of patients. Through a retrospective search of our regional oncology pharmacy database, we identified 18 patients diagnosed with de novo SCPC between 2004 and 2017. Ten patients had pure SCPC pathology, and the remainder had some admixed adenocarcinoma foci, but all were treated with first‐line platinum‐based chemotherapy. The median overall survival was 28 months. We performed targeted DNA sequencing, whole exome sequencing and mRNA profiling on formalin‐fixed paraffin‐embedded archival tumour tissue. We observed frequent biallelic deletion and/or mutation of the tumour suppressor genes TP53, RB1, and PTEN, similarly to what was found in treatment‐related SCPC. Indeed, at the RNA level, pure de novo SCPC closely resembled treatment‐related SCPC. However, five patients had biallelic loss of DNA repair genes, including BRCA1, BRCA2, ATM, and MSH2/6, potentially underlying the high genomic instability of this rare disease variant. Two patients with pure de novo SCPC harboured ETS gene rearrangements involving androgen‐driven promoters, consistent with the evolution of de novo SCPC from an androgen‐driven ancestor. Overall, our results reveal a highly aggressive molecular landscape that underlies this unusual pathological variant, and suggest opportunities for targeted therapy strategies in a disease with few treatment options. Copyright


Communications Biology | 2018

Frequent mutation of the FOXA1 untranslated region in prostate cancer

Matti Annala; Sinja Taavitsainen; Gillian Vandekerkhove; Jack V. W. Bacon; Kevin Beja; Kim N. Chi; Matti Nykter; Alexander W. Wyatt

Prostate cancer has a low somatic mutation rate but non-coding regions remain underexplored. We sequenced the untranslated regions (UTRs) of 72 established driver genes in 428 patients with metastatic prostate cancer and identified FOXA1 3′-UTR mutations in 12% of patients. The mutations were predominantly insertions or deletions, covered the entire UTR without motif enrichment, and were not detected in other cancers. FOXA1 lies in head-on orientation with the androgen-regulated non-coding gene AL121790.1, resulting in strong prostate lineage-specific bidirectional transcription across the FOXA1 3′-UTR. This suggests transcriptional activity as a cause for the localized hypermutation. The indel-dominant pattern of somatic mutation extends into the FOXA1 coding region, where it is shaped by clonal selection to yield a cluster of non-frameshift indels inside the forkhead domain. Somatic FOXA1 3′-UTR mutations may prove useful for diagnostic and screening approaches, given their high frequency and lineage specificity.Matti Annala et al. report the recurrence of new FOXA1 mutations in prostate cancer. These FOXA1 mutations in 3’ untranslated region may prove useful as diagnostic markers for prostate cancer.


Journal of Clinical Oncology | 2017

A randomized phase II cross-over study of abiraterone + prednisone (ABI) vs enzalutamide (ENZ) for patients (pts) with metastatic, castration-resistant prostate cancer (mCRPC).

Kim N. Chi; Matti Annala; Katherine Sunderland; Daniel Khalaf; Daygen L. Finch; Conrad D. Oja; Joanna Vergidis; Muhammad Zulfiqar; Kevin Beja; Gillian Vandekerkhove; Martin Gleave; Alexander W. Wyatt


Cancer Genetics and Cytogenetics | 2017

Clinical utility of emerging liquid biomarkers in advanced prostate cancer

Gillian Vandekerkhove; Kim N. Chi; Alexander W. Wyatt


Journal of Clinical Oncology | 2017

Circulating tumor DNA (ctDNA) and correlations with clinical prognostic factors in patients with metastatic castration-resistant prostate cancer (mCRPC).

Daniel Khalaf; Matti Annala; Kevin Beja; Gillian Vandekerkhove; Muhammad Zulfiqar; Daygen L. Finch; Conrad D. Oja; Joanna Vergidis; Martin Gleave; Alexander W. Wyatt; Kim N. Chi


Journal of Clinical Oncology | 2018

Outcomes in patients (Pts) with advanced prostate cancer and inactivating germline mutations in BRCA2 or ATM.

Steven Yip; Daniel Khalaf; Werner J. Struss; Katherine Sunderland; Gillian Vandekerkhove; Evan W. Warner; Matti Annala; Alexander W. Wyatt; Kim N. Chi


Journal of Clinical Oncology | 2017

Germline DNA repair mutations in metastatic castration-resistant prostate cancer: Therapy response and applicability of circulating tumor DNA.

Werner J. Struss; Matti Annala; Evan W. Warner; Kevin Beja; Gillian Vandekerkhove; Amanda Wong; Martin Gleave; Kim N. Chi; Alexander W. Wyatt

Collaboration


Dive into the Gillian Vandekerkhove's collaboration.

Top Co-Authors

Avatar

Alexander W. Wyatt

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kevin Beja

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martin Gleave

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Werner J. Struss

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Evan W. Warner

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Joanna Vergidis

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge