Network


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

Hotspot


Dive into the research topics where Steve E. Kalloger is active.

Publication


Featured researches published by Steve E. Kalloger.


The New England Journal of Medicine | 2010

ARID1A Mutations in Endometriosis-Associated Ovarian Carcinomas

Kimberly C. Wiegand; Sohrab P. Shah; Osama M. Al-Agha; Yongjun Zhao; Kane Tse; Thomas Zeng; Janine Senz; Melissa K. McConechy; Michael S. Anglesio; Steve E. Kalloger; Winnie Yang; Alireza Heravi-Moussavi; Ryan Giuliany; Christine Chow; John Fee; Abdalnasser Zayed; Leah M Prentice; Nataliya Melnyk; Gulisa Turashvili; Allen Delaney; Jason Madore; Stephen Yip; Andrew McPherson; Gavin Ha; Lynda Bell; Sian Fereday; Angela Tam; Laura Galletta; Patricia N. Tonin; Diane Provencher

BACKGROUND Ovarian clear-cell and endometrioid carcinomas may arise from endometriosis, but the molecular events involved in this transformation have not been described. METHODS We sequenced the whole transcriptomes of 18 ovarian clear-cell carcinomas and 1 ovarian clear-cell carcinoma cell line and found somatic mutations in ARID1A (the AT-rich interactive domain 1A [SWI-like] gene) in 6 of the samples. ARID1A encodes BAF250a, a key component of the SWI–SNF chromatin remodeling complex. We sequenced ARID1A in an additional 210 ovarian carcinomas and a second ovarian clear-cell carcinoma cell line and measured BAF250a expression by means of immunohistochemical analysis in an additional 455 ovarian carcinomas. RESULTS ARID1A mutations were seen in 55 of 119 ovarian clear-cell carcinomas (46%), 10 of 33 endometrioid carcinomas (30%), and none of the 76 high-grade serous ovarian carcinomas. Seventeen carcinomas had two somatic mutations each. Loss of the BAF250a protein correlated strongly with the ovarian clear-cell carcinoma and endometrioid carcinoma subtypes and the presence of ARID1A mutations. In two patients, ARID1A mutations and loss of BAF250a expression were evident in the tumor and contiguous atypical endometriosis but not in distant endometriotic lesions. CONCLUSIONS These data implicate ARID1A as a tumor-suppressor gene frequently disrupted in ovarian clear-cell and endometrioid carcinomas. Since ARID1A mutation and loss of BAF250a can be seen in the preneoplastic lesions, we speculate that this is an early event in the transformation of endometriosis into cancer. (Funded by the British Columbia Cancer Foundation and the Vancouver General Hospital–University of British Columbia Hospital Foundation.).


PLOS Medicine | 2008

Ovarian Carcinoma Subtypes Are Different Diseases: Implications for Biomarker Studies

Martin Köbel; Steve E. Kalloger; Niki Boyd; Steven McKinney; Erika Mehl; Chana Palmer; Samuel Leung; Nathan J. Bowen; Diana N. Ionescu; Ashish Rajput; Leah M Prentice; Dianne Miller; Jennifer F. De Los Santos; Kenneth D. Swenerton; C. Blake Gilks; David Huntsman

Background Although it has long been appreciated that ovarian carcinoma subtypes (serous, clear cell, endometrioid, and mucinous) are associated with different natural histories, most ovarian carcinoma biomarker studies and current treatment protocols for women with this disease are not subtype specific. With the emergence of high-throughput molecular techniques, distinct pathogenetic pathways have been identified in these subtypes. We examined variation in biomarker expression rates between subtypes, and how this influences correlations between biomarker expression and stage at diagnosis or prognosis. Methods and Findings In this retrospective study we assessed the protein expression of 21 candidate tissue-based biomarkers (CA125, CRABP-II, EpCam, ER, F-Spondin, HE4, IGF2, K-Cadherin, Ki-67, KISS1, Matriptase, Mesothelin, MIF, MMP7, p21, p53, PAX8, PR, SLPI, TROP2, WT1) in a population-based cohort of 500 ovarian carcinomas that was collected over the period from 1984 to 2000. The expression of 20 of the 21 biomarkers differs significantly between subtypes, but does not vary across stage within each subtype. Survival analyses show that nine of the 21 biomarkers are prognostic indicators in the entire cohort but when analyzed by subtype only three remain prognostic indicators in the high-grade serous and none in the clear cell subtype. For example, tumor proliferation, as assessed by Ki-67 staining, varies markedly between different subtypes and is an unfavourable prognostic marker in the entire cohort (risk ratio [RR] 1.7, 95% confidence interval [CI] 1.2%–2.4%) but is not of prognostic significance within any subtype. Prognostic associations can even show an inverse correlation within the entire cohort, when compared to a specific subtype. For example, WT1 is more frequently expressed in high-grade serous carcinomas, an aggressive subtype, and is an unfavourable prognostic marker within the entire cohort of ovarian carcinomas (RR 1.7, 95% CI 1.2%–2.3%), but is a favourable prognostic marker within the high-grade serous subtype (RR 0.5, 95% CI 0.3%–0.8%). Conclusions The association of biomarker expression with survival varies substantially between subtypes, and can easily be overlooked in whole cohort analyses. To avoid this effect, each subtype within a cohort should be analyzed discretely. Ovarian carcinoma subtypes are different diseases, and these differences should be reflected in clinical research study design and ultimately in the management of ovarian carcinoma.


The New England Journal of Medicine | 2009

Mutation of FOXL2 in granulosa-cell tumors of the ovary

Sohrab P. Shah; Martin Köbel; Janine Senz; Ryan D. Morin; Blaise Clarke; Kimberly C. Wiegand; Gillian Leung; Abdalnasser Zayed; Erika Mehl; Steve E. Kalloger; Mark Sun; Ryan Giuliany; Erika Yorida; Steven J.M. Jones; Richard Varhol; Kenneth D. Swenerton; Dianne Miller; Philip B. Clement; Colleen Crane; Jason Madore; Diane Provencher; Peter C. K. Leung; Anna deFazio; Jaswinder Khattra; Gulisa Turashvili; Yongjun Zhao; Thomas Zeng; J.N. Mark Glover; Barbara C. Vanderhyden; Chengquan Zhao

BACKGROUND Granulosa-cell tumors (GCTs) are the most common type of malignant ovarian sex cord-stromal tumor (SCST). The pathogenesis of these tumors is unknown. Moreover, their histopathological diagnosis can be challenging, and there is no curative treatment beyond surgery. METHODS We analyzed four adult-type GCTs using whole-transcriptome paired-end RNA sequencing. We identified putative GCT-specific mutations that were present in at least three of these samples but were absent from the transcriptomes of 11 epithelial ovarian tumors, published human genomes, and databases of single-nucleotide polymorphisms. We confirmed these variants by direct sequencing of complementary DNA and genomic DNA. We then analyzed additional tumors and matched normal genomic DNA, using a combination of direct sequencing, analyses of restriction-fragment-length polymorphisms, and TaqMan assays. RESULTS All four index GCTs had a missense point mutation, 402C-->G (C134W), in FOXL2, a gene encoding a transcription factor known to be critical for granulosa-cell development. The FOXL2 mutation was present in 86 of 89 additional adult-type GCTs (97%), in 3 of 14 thecomas (21%), and in 1 of 10 juvenile-type GCTs (10%). The mutation was absent in 49 SCSTs of other types and in 329 unrelated ovarian or breast tumors. CONCLUSIONS Whole-transcriptome sequencing of four GCTs identified a single, recurrent somatic mutation (402C-->G) in FOXL2 that was present in almost all morphologically identified adult-type GCTs. Mutant FOXL2 is a potential driver in the pathogenesis of adult-type GCTs.


The New England Journal of Medicine | 2012

Recurrent Somatic DICER1 Mutations in Nonepithelial Ovarian Cancers

Alireza Heravi-Moussavi; Michael S. Anglesio; S.-W. Grace Cheng; Janine Senz; Winnie Yang; Leah M Prentice; Anthony P. Fejes; Christine Chow; Alicia A. Tone; Steve E. Kalloger; Nancy Hamel; Andrew Roth; Gavin Ha; Adrian Wan; Sarah Maines-Bandiera; Clara Salamanca; Barbara Pasini; Blaise Clarke; Anna F. Lee; Cheng-Han Lee; Chengquan Zhao; Robert H. Young; Samuel Aparicio; Poul H. Sorensen; Michelle Woo; Niki Boyd; Steven J.M. Jones; Martin Hirst; Marco A. Marra; Blake Gilks

BACKGROUND Germline truncating mutations in DICER1, an endoribonuclease in the RNase III family that is essential for processing microRNAs, have been observed in families with the pleuropulmonary blastoma-family tumor and dysplasia syndrome. Mutation carriers are at risk for nonepithelial ovarian tumors, notably sex cord-stromal tumors. METHODS We sequenced the whole transcriptomes or exomes of 14 nonepithelial ovarian tumors and noted closely clustered mutations in the region of DICER1 encoding the RNase IIIb domain of DICER1 in four samples. We then sequenced this region of DICER1 in additional ovarian tumors and in certain other tumors and queried the effect of the mutations on the enzymatic activity of DICER1 using in vitro RNA cleavage assays. RESULTS DICER1 mutations in the RNase IIIb domain were found in 30 of 102 nonepithelial ovarian tumors (29%), predominantly in Sertoli-Leydig cell tumors (26 of 43, or 60%), including 4 tumors with additional germline DICER1 mutations. These mutations were restricted to codons encoding metal-binding sites within the RNase IIIb catalytic centers, which are critical for microRNA interaction and cleavage, and were somatic in all 16 samples in which germline DNA was available for testing. We also detected mutations in 1 of 14 nonseminomatous testicular germ-cell tumors, in 2 of 5 embryonal rhabdomyosarcomas, and in 1 of 266 epithelial ovarian and endometrial carcinomas. The mutant DICER1 proteins had reduced RNase IIIb activity but retained RNase IIIa activity. CONCLUSIONS Somatic missense mutations affecting the RNase IIIb domain of DICER1 are common in nonepithelial ovarian tumors. These mutations do not obliterate DICER1 function but alter it in specific cell types, a novel mechanism through which perturbation of microRNA processing may be oncogenic. (Funded by the Terry Fox Research Institute and others.).


BMC Cancer | 2008

Ovarian carcinomas with genetic and epigenetic BRCA1 loss have distinct molecular abnormalities

Joshua Z. Press; Alessandro De Luca; Niki Boyd; Sean Young; Armelle Troussard; Yolanda Ridge; Pardeep Kaurah; Steve E. Kalloger; Katherine A Blood; Margaret Smith; Paul T. Spellman; Yuker Wang; Dianne Miller; Doug Horsman; Malek Faham; C. Blake Gilks; Joe W. Gray; David Huntsman

BackgroundSubclassification of ovarian carcinomas can be used to guide treatment and determine prognosis. Germline and somatic mutations, loss of heterozygosity (LOH), and epigenetic events such as promoter hypermethylation can lead to decreased expression of BRCA1/2 in ovarian cancers. The mechanism of BRCA1/2 loss is a potential method of subclassifying high grade serous carcinomas.MethodsA consecutive series of 49 ovarian cancers was assessed for mutations status of BRCA1 and BRCA2, LOH at the BRCA1 and BRCA2 loci, methylation of the BRCA1 promoter, BRCA1, BRCA2, PTEN, and PIK3CA transcript levels, PIK3CA gene copy number, and BRCA1, p21, p53, and WT-1 immunohistochemistry.ResultsEighteen (37%) of the ovarian carcinomas had germline or somatic BRCA1 mutations, or epigenetic loss of BRCA1. All of these tumours were high-grade serous or undifferentiated type. None of the endometrioid (n = 5), clear cell (n = 4), or low grade serous (n = 2) carcinomas showed loss of BRCA1, whereas 47% of the 38 high-grade serous or undifferentiated carcinomas had loss of BRCA1. It was possible to distinguish high grade serous carcinomas with BRCA1 mutations from those with epigenetic BRCA1 loss: tumours with BRCA1 mutations typically had decreased PTEN mRNA levels while those with epigenetic loss of BRCA1 had copy number gain of PIK3CA. Overexpression of p53 with loss of p21 expression occurred significantly more frequently in high grade serous carcinomas with epigenetic loss of BRCA1, compared to high grade serous tumors without loss of BRCA1.ConclusionHigh grade serous carcinomas can be subclassified into three groups: BRCA1 loss (genetic), BRCA1 loss (epigenetic), and no BRCA1 loss. Tumors in these groups show distinct molecular alterations involving the PI3K/AKT and p53 pathways.


Clinical Cancer Research | 2007

Amplification of PVT1 Contributes to the Pathophysiology of Ovarian and Breast Cancer

Yinghui Guan; Wen Lin Kuo; Jackie L. Stilwell; Hirokuni Takano; Anna Lapuk; Jane Fridlyand; Jian-Hua Mao; Mamie Yu; Melinda A. Miller; Jennifer F. De Los Santos; Steve E. Kalloger; Joseph W. Carlson; David G. Ginzinger; Susan E. Celniker; Gordon B. Mills; David Huntsman; Joe W. Gray

Purpose: This study was designed to elucidate the role of amplification at 8q24 in the pathophysiology of ovarian and breast cancer because increased copy number at this locus is one of the most frequent genomic abnormalities in these cancers. Experimental Design: To accomplish this, we assessed the association of amplification at 8q24 with outcome in ovarian cancers using fluorescence in situ hybridization to tissue microarrays and measured responses of ovarian and breast cancer cell lines to specific small interfering RNAs against the oncogene MYC and a putative noncoding RNA, PVT1, both of which map to 8q24. Results: Amplification of 8q24 was associated with significantly reduced survival duration. In addition, small interfering RNA–mediated reduction in either PVT1 or MYC expression inhibited proliferation in breast and ovarian cancer cell lines in which they were both amplified and overexpressed but not in lines in which they were not amplified/overexpressed. Inhibition of PVT1 expression also induced a strong apoptotic response in cell lines in which it was overexpressed but not in lines in which it was not amplified/overexpressed. Inhibition of MYC, on the other hand, did not induce an apoptotic response in cell lines in which MYC was amplified and overexpressed. Conclusions: These results suggest that MYC and PVT1 contribute independently to ovarian and breast pathogenesis when overexpressed because of genomic abnormalities. They also suggest that PVT1-mediated inhibition of apoptosis may explain why amplification of 8q24 is associated with reduced survival duration in patients treated with agents that act through apoptotic mechanisms.


The Journal of Pathology | 2013

Distinct evolutionary trajectories of primary high-grade serous ovarian cancers revealed through spatial mutational profiling

Ali Bashashati; Gavin Ha; Alicia A. Tone; Jiarui Ding; Leah M Prentice; Andrew Roth; Jamie Rosner; Karey Shumansky; Steve E. Kalloger; Janine Senz; Winnie Yang; Melissa K. McConechy; Nataliya Melnyk; Michael S. Anglesio; Margaret Luk; Kane Tse; Thomas Zeng; Richard G. Moore; Yongjun Zhao; Marco A. Marra; Blake Gilks; Stephen Yip; David Huntsman; Jessica N. McAlpine; Sohrab P. Shah

High‐grade serous ovarian cancer (HGSC) is characterized by poor outcome, often attributed to the emergence of treatment‐resistant subclones. We sought to measure the degree of genomic diversity within primary, untreated HGSCs to examine the natural state of tumour evolution prior to therapy. We performed exome sequencing, copy number analysis, targeted amplicon deep sequencing and gene expression profiling on 31 spatially and temporally separated HGSC tumour specimens (six patients), including ovarian masses, distant metastases and fallopian tube lesions. We found widespread intratumoural variation in mutation, copy number and gene expression profiles, with key driver alterations in genes present in only a subset of samples (eg PIK3CA, CTNNB1, NF1). On average, only 51.5% of mutations were present in every sample of a given case (range 10.2–91.4%), with TP53 as the only somatic mutation consistently present in all samples. Complex segmental aneuploidies, such as whole‐genome doubling, were present in a subset of samples from the same individual, with divergent copy number changes segregating independently of point mutation acquisition. Reconstruction of evolutionary histories showed one patient with mixed HGSC and endometrioid histology, with common aetiologic origin in the fallopian tube and subsequent selection of different driver mutations in the histologically distinct samples. In this patient, we observed mixed cell populations in the early fallopian tube lesion, indicating that diversity arises at early stages of tumourigenesis. Our results revealed that HGSCs exhibit highly individual evolutionary trajectories and diverse genomic tapestries prior to therapy, exposing an essential biological characteristic to inform future design of personalized therapeutic solutions and investigation of drug‐resistance mechanisms.


International Journal of Gynecological Pathology | 2010

Differences in Tumor Type in Low-stage Versus High-stage Ovarian Carcinomas

Martin Köbel; Steve E. Kalloger; David Huntsman; Jennifer F. De Los Santos; Kenneth D. Swenerton; Jeffrey D. Seidman; C. Blake Gilks

Although there are recognized differences in the type of ovarian carcinomas between those tumors diagnosed at low versus high stage, there is a lack of data on stage distribution of ovarian carcinomas diagnosed according to the current histopathologic criteria from large population-based cohorts. We reviewed full slide sets of 1009 cases of 2555 patients diagnosed with ovarian carcinoma that were referred to the British Columbia Cancer Agency over a 16-year period (1984 to 2000). On the basis of the reviewed cases we extrapolated the distribution of tumor type in low-stage (I/II) and high-stage (III/IV) tumors. We then compared the frequencies with those seen in a large hospital practice. The overall frequency of tumor types was as follows: high-grade serous—68.1%, clear-cell—12.2%, endometrioid—11.3%, mucinous—3.4%, low-grade serous—3.4%, rare types—1.6%. High-grade serous carcinomas accounted for 35.5% of stage I/II tumors and 87.7% of stage III/IV tumors. In contrast, clear-cell (26.2% vs. 4.5%), endometrioid (26.6% vs. 2.5%), and mucinous (7.5% vs. 1.2%) carcinomas were relatively more common among the low-stage versus high-stage tumors. This distribution was found to be very similar in 410 consecutive cases from the Washington Hospital Center. The distribution of ovarian carcinoma types differs significantly in patients with low-stage versus high-stage ovarian carcinoma when contemporary diagnostic criteria are used, with consistent results seen in 2 independent case series. These findings reflect important biological differences in the behavior of the major tumor types, with important clinical implications.


Human Pathology | 2008

Tumor cell type can be reproducibly diagnosed and is of independent prognostic significance in patients with maximally debulked ovarian carcinoma

C. Blake Gilks; Diana N. Ionescu; Steve E. Kalloger; Martin Köbel; Julie Irving; Blaise Clarke; Jennifer F. De Los Santos; Nhu D. Le; Veronika Moravan; Kenneth D. Swenerton

Ovarian surface epithelial carcinomas are routinely subclassified by pathologists based on tumor cell type and grade. It is controversial whether cell type or grade is superior in predicting patient response to treatment or survival, in patients stratified by stage of disease. The aim of this study was to uniformly apply updated criteria for cell-type and grade assignment to a series of 575 cases of ovarian surface epithelial carcinoma. All patients were optimally surgically debulked, with no macroscopic residual disease after primary surgery. Slides from these cases were reviewed by a single pathologist, who was blinded to patient outcomes. In 50 cases, 2 additional pathologists reviewed the slides independently to determine interobserver variation in assessment of cell type and grade. The distribution of tumor stage was as follows: stage I--233 cases, stage II--246 cases, stage III--96 cases. The most common cell type encountered was serous carcinoma (229/575, 40%), followed by clear cell (149/575, 26%), endometrioid (139/575, 24%), and mucinous (36/575, 6%). Serous carcinomas were significantly more likely to present with advanced stage disease (76/229 [33.2%] were stage III, and 82% of all stage III tumors were serous), whereas all nonserous cell types were stage I or II at diagnosis in greater than 90% of cases. Both FIGO grade and Silverberg grade stratified patients into groups with significantly different risks of relapse and survival, but the Silverberg grading system was a more powerful prognosticator. In multivariate analysis, stage was the most powerful prognostic indicator (P < .0001), followed by tumor cell type (P = .015), but grade was not of independent significance. Interobserver variation in assignment of cell type was very good (kappa = 0.77) with moderate reproducibility in assignment of Silverberg grade (kappa = 0.40) and minimal reproducibility in assignment of FIGO grade (kappa = 0.27). Thus, in this series of cases of ovarian surface epithelial carcinomas with no macroscopic residual disease after primary debulking surgery, assignment of tumor cell type was both more reproducible and provided superior prognostic information compared with assignment of tumor grade. As tumor cell type also correlates with underlying molecular abnormalities and may predict response to chemotherapy, this suggests that tumor cell type could be used to guide treatment decisions for patients with ovarian surface epithelial carcinoma.


Modern Pathology | 2009

Intraepithelial T cells and prognosis in ovarian carcinoma: novel associations with stage, tumor type, and BRCA1 loss.

Blaise Clarke; Anna V. Tinker; Cheng Han Lee; Subbaya Subramanian; Matt van de Rijn; Dmitry Turbin; Steve E. Kalloger; Guangming Han; Kathy Ceballos; Mark G. Cadungog; David Huntsman; George Coukos; C. Blake Gilks

Intraepithelial tumor-infiltrating T cells have been correlated with improved outcomes in ovarian carcinoma, however, it is not known whether there is an association with disease stage, histological subtype, or BRCA mutation/expression. Two case series of ovarian carcinomas were included in the study; a retrospective series of 500 patients, and 40 prospectively collected cases fully characterized for BRCA1 mutation status and expression. Intraepithelial immune cells were assessed as present or absent by immunohistochemical staining of tissue microarrays. In the retrospective case series, the presence of intraepithelial CD8+ T-cells correlated with improved disease-specific survival (P=0.027), whereas intraepithelial CD3+ T cells did not (P=0.49). For serous ovarian carcinomas, the presence of intraepithelial CD3+ and CD8+ T-cells correlated with improved disease-specific survival (P=0.0016 and P≤0.0001, respectively). The presence of intraepithelial CD8+ T cells was not associated with improved survival in endometrioid or clear cell carcinomas. On multivariate analysis, disease stage and CD8+ T cells were found to be independently predictive of improved disease-specific survival, whereas grade, age at surgery, and type of adjuvant treatment were not. In the prospective patient cohort, intraepithelial CD8+ T-cells correlated with the presence of mutation or loss of expression of BRCA1 through promoter methylation (P=0.019). Intraepithelial CD8+ tumor-infiltrating T-cells correlate with improved clinical outcomes for all stages of ovarian cancer; this association is restricted to the serous ovarian cancer subtype, and is an independent prognostic factor on multivariate analysis. The presence of intraepithelial CD8+ T cells also significantly correlates with loss of BRCA1.

Collaboration


Dive into the Steve E. Kalloger's collaboration.

Top Co-Authors

Avatar

David Huntsman

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

C. Blake Gilks

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael S. Anglesio

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

David F. Schaeffer

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Janine Senz

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Blaise Clarke

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Blake Gilks

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Leah M Prentice

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Jessica N. McAlpine

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge