Network


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

Hotspot


Dive into the research topics where Kathleen A. Burke is active.

Publication


Featured researches published by Kathleen A. Burke.


Cancer Research | 2006

The expression of CD30 in anaplastic large cell lymphoma is regulated by nucleophosmin-anaplastic lymphoma kinase-mediated JunB level in a cell type-specific manner.

Faye Yuan Yi Hsu; Patrick B. Johnston; Kathleen A. Burke; Yi Zhao

Chromosomal translocation t(2;5) and the resulting fusion protein nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) are detected in 50% to 70% of anaplastic large cell lymphoma (ALCL), which is a T/null cell non-Hodgkins lymphoma showing anaplastic morphology with cell surface expression of CD30. Because aberrant CD30 expression was also observed in the T-cell lymphoma derived from lineage-specific NPM-ALK transgenic mice, we tested the hypothesis that there might be a functional relationship between the two neoplastic-related proteins: NPM-ALK and CD30. In this study, we used the RNA interference method to modulate NPM-ALK protein expression in ALCL-derived, t(2;5)-positive Karpas 299 cells. We observed decreased CD30 expression when NPM-ALK was repressed. Further analysis suggested that JunB functioned as the mediator of NPM-ALK-derived CD30 transcriptional regulation. The NPM-ALK-repressed cells, which had low CD30 expression, were characterized with lower cell proliferation compared with cells in the control group, suggesting that altered CD30 expression may correlate to NPM-ALK-mediated tumor cell growth inhibition. Combination of NPM-ALK repression and CD30 ligand leads to significantly increased tumor cell growth inhibition compared with one method alone, suggesting its potential application for ALCL-specific cancer treatment.


The Journal of Pathology | 2016

Massively parallel sequencing of phyllodes tumours of the breast reveals actionable mutations, and TERT promoter hotspot mutations and TERT gene amplification as likely drivers of progression

Salvatore Piscuoglio; Charlotte K.Y. Ng; Melissa P. Murray; Kathleen A. Burke; Marcia Edelweiss; Felipe C. Geyer; Gabriel S. Macedo; Akiko Inagaki; Anastasios D. Papanastasiou; Luciano G. Martelotto; Caterina Marchiò; Raymond S. Lim; Rafael A. Ioris; Pooja K. Nahar; Ino de Bruijn; Lillian Mary Smyth; Muzaffar Akram; Dara S. Ross; John H.J. Petrini; Larry Norton; David B. Solit; José Baselga; Edi Brogi; Marc Ladanyi; Britta Weigelt; Jorge S. Reis-Filho

Phyllodes tumours (PTs) are breast fibroepithelial lesions that are graded based on histological criteria as benign, borderline or malignant. PTs may recur locally. Borderline PTs and malignant PTs may metastasize to distant sites. Breast fibroepithelial lesions, including PTs and fibroadenomas, are characterized by recurrent MED12 exon 2 somatic mutations. We sought to define the repertoire of somatic genetic alterations in PTs and whether these may assist in the differential diagnosis of these lesions. We collected 100 fibroadenomas, 40 benign PTs, 14 borderline PTs and 22 malignant PTs; six, six and 13 benign, borderline and malignant PTs, respectively, and their matched normal tissue, were subjected to targeted massively parallel sequencing (MPS) using the MSK‐IMPACT sequencing assay. Recurrent MED12 mutations were found in 56% of PTs; in addition, mutations affecting cancer genes (eg TP53, RB1, SETD2 and EGFR) were exclusively detected in borderline and malignant PTs. We found a novel recurrent clonal hotspot mutation in the TERT promoter (−124 C>T) in 52% and TERT gene amplification in 4% of PTs. Laser capture microdissection revealed that these mutations were restricted to the mesenchymal component of PTs. Sequencing analysis of the entire cohort revealed that the frequency of TERT alterations increased from benign (18%) to borderline (57%) and to malignant PTs (68%; p < 0.01), and TERT alterations were associated with increased levels of TERT mRNA (p < 0.001). No TERT alterations were observed in fibroadenomas. An analysis of TERT promoter sequencing and gene amplification distinguished PTs from fibroadenomas with a sensitivity and a positive predictive value of 100% (CI 95.38–100%) and 100% (CI 85.86–100%), respectively, and a sensitivity and a negative predictive value of 39% (CI 28.65–51.36%) and 68% (CI 60.21–75.78%), respectively. Our results suggest that TERT alterations may drive the progression of PTs, and may assist in the differential diagnosis between PTs and fibroadenomas. Copyright


The Journal of Pathology | 2016

Uterine adenosarcomas are mesenchymal neoplasms

Salvatore Piscuoglio; Kathleen A. Burke; Charlotte K.Y. Ng; Anastasios D. Papanastasiou; Felipe C. Geyer; Gabriel S. Macedo; Luciano G. Martelotto; Ino de Bruijn; Maria Rosaria De Filippo; Anne M. Schultheis; Rafael A. Ioris; Douglas A. Levine; Robert A. Soslow; Brian P. Rubin; Jorge S. Reis-Filho; Britta Weigelt

Uterine adenosarcomas (UAs) are biphasic lesions composed of a malignant mesenchymal (ie stromal) component and an epithelial component. UAs are generally low‐grade and have a favourable prognosis, but may display sarcomatous overgrowth (SO), which is associated with a worse outcome. We hypothesized that, akin to breast fibroepithelial lesions, UAs are mesenchymal neoplasms in which clonal somatic genetic alterations are restricted to the mesenchymal component. To characterize the somatic genetic alterations in UAs and to test this hypothesis, we subjected 20 UAs to a combination of whole‐exome (n = 6), targeted capture (n = 13) massively parallel sequencing (MPS) and/or RNA sequencing (n = 6). Only three genes, FGFR2, KMT2C and DICER1, were recurrently mutated, all in 2/19 cases; however, 26% (5/19) and 21% (4/19) of UAs harboured MDM2/CDK4/HMGA2 and TERT gene amplification, respectively, and two cases harboured fusion genes involving NCOA family members. Using a combination of laser‐capture microdissection and in situ techniques, we demonstrated that the somatic genetic alterations detected by MPS were restricted to the mesenchymal component. Furthermore, mitochondrial DNA sequencing of microdissected samples revealed that epithelial and mesenchymal components of UAs were clonally unrelated. In conclusion, here we provide evidence that UAs are genetically heterogeneous lesions and mesenchymal neoplasms. Copyright


npj Breast Cancer | 2016

Triple-negative breast cancer: the importance of molecular and histologic subtyping, and recognition of low-grade variants

Fresia Pareja; Felipe C. Geyer; Caterina Marchiò; Kathleen A. Burke; Britta Weigelt; Js Reis-Filho

Triple-negative breast cancers (TNBCs), defined by lack of expression of estrogen receptor, progesterone receptor and HER2, account for 12–17% of breast cancers and are clinically perceived as a discrete breast cancer subgroup. Nonetheless, TNBC has been shown to constitute a vastly heterogeneous disease encompassing a wide spectrum of entities with marked genetic, transcriptional, histological and clinical differences. Although most TNBCs are high-grade tumors, there are well-characterized low-grade TNBCs that have an indolent clinical course, whose natural history, molecular features and optimal therapy vastly differ from those of high-grade TNBCs. Secretory and adenoid cystic carcinomas are two histologic types of TNBCs underpinned by specific fusion genes; these tumors have an indolent clinical behavior and lack all of the cardinal molecular features of high-grade triple-negative disease. Recent studies of rare entities, including lesions once believed to constitute mere benign breast disease (e.g., microglandular adenosis), have resulted in the identification of potential precursors of TNBC and suggested the existence of a family of low-grade triple-negative lesions that, despite having low-grade morphology and indolent clinical behavior, have been shown to harbor the complex genomic landscape of common forms of TNBC, and may progress to high-grade disease. In this review, we describe the heterogeneity of TNBC and focus on the histologic and molecular features of low-grade forms of TNBC. Germane to addressing the challenges posed by the so-called triple-negative disease is the realization that TNBC is merely a descriptive term, and that low-grade types of TNBC may be driven by distinct sets of genetic alterations.


Clinical Cancer Research | 2017

The Landscape of Somatic Genetic Alterations in Metaplastic Breast Carcinomas

Charlotte K.Y. Ng; Salvatore Piscuoglio; Felipe C. Geyer; Kathleen A. Burke; Fresia Pareja; Carey A. Eberle; Raymond S. Lim; Rachael Natrajan; Nadeem Riaz; Odette Mariani; Larry Norton; Anne Vincent-Salomon; Y Hannah Wen; Britta Weigelt; Jorge S. Reis-Filho

Purpose: Metaplastic breast carcinoma (MBC) is a rare and aggressive histologic type of breast cancer, predominantly of triple-negative phenotype, and characterized by the presence of malignant cells showing squamous and/or mesenchymal differentiation. We sought to define the repertoire of somatic genetic alterations and the mutational signatures of MBCs. Experimental Design: Whole-exome sequencing was performed in 35 MBCs, with 16, 10, and 9 classified as harboring chondroid, spindle, and squamous metaplasia as the predominant metaplastic component. The genomic landscape of MBCs was compared with that of triple-negative invasive ductal carcinomas of no special type (IDC-NST) from The Cancer Genome Atlas. Wnt and PI3K/AKT/mTOR pathway activity was assessed using a qPCR assay. Results: MBCs harbored complex genomes with frequent TP53 (69%) mutations. In contrast to triple-negative IDC-NSTs, MBCs more frequently harbored mutations in PIK3CA (29%), PIK3R1 (11%), ARID1A (11%), FAT1 (11%), and PTEN (11%). PIK3CA mutations were not found in MBCs with chondroid metaplasia. Compared with triple-negative IDC-NSTs, MBCs significantly more frequently harbored mutations in PI3K/AKT/mTOR pathway–related (57% vs. 22%) and canonical Wnt pathway–related (51% vs. 28%) genes. MBCs with somatic mutations in PI3K/AKT/mTOR or Wnt pathway–related genes displayed increased activity of the respective pathway. Conclusions: MBCs are genetically complex and heterogeneous, and are driven by a repertoire of somatic mutations distinct from that of triple-negative IDC-NSTs. Our study highlights the genetic basis and the importance of PI3K/AKT/mTOR and Wnt pathway dysregulation in MBCs and provides a rationale for the metaplastic phenotype and the reported responses to PI3K/AKT/mTOR inhibitors in these tumors. Clin Cancer Res; 23(14); 3859–70. ©2017 AACR.


The Journal of Pathology | 2017

The genetic landscape of endometrial clear cell carcinomas

Deborah DeLair; Kathleen A. Burke; Pier Selenica; Raymond S. Lim; Sasinya N. Scott; Sumit Middha; Abhinita Mohanty; Donavan T. Cheng; Michael F. Berger; Robert A. Soslow; Britta Weigelt

Clear cell carcinoma of the endometrium is a rare type of endometrial cancer that is generally associated with an aggressive clinical behaviour. Here, we sought to define the repertoire of somatic genetic alterations in endometrial clear cell carcinomas (ECCs), and whether ECCs could be classified into the molecular subtypes described for endometrial endometrioid and serous carcinomas. We performed a rigorous histopathological review, immunohistochemical analysis and massively parallel sequencing targeting 300 cancer‐related genes of 32 pure ECCs. Eleven (34%), seven (22%) and six (19%) ECCs showed abnormal expression patterns for p53, ARID1A, and at least one DNA mismatch repair (MMR) protein, respectively. Targeted sequencing data were obtained from 30 of the 32 ECCs included in this study, and these revealed that two ECCs (7%) were ultramutated and harboured mutations affecting the exonuclease domain of POLE. In POLE wild‐type ECCs, TP53 (46%), PIK3CA (36%), PPP2R1A (36%), FBXW7 (25%), ARID1A (21%), PIK3R1 (18%) and SPOP (18%) were the genes most commonly affected by mutations; 18% and 11% harboured CCNE1 and ERBB2 amplifications, respectively, and 11% showed DAXX homozygous deletions. ECCs less frequently harboured mutations affecting CTNNB1 and PTEN but more frequently harboured PPP2R1A and TP53 mutations than non‐POLE endometrioid carcinomas from The Cancer Genome Atlas (TCGA). Compared to endometrial serous carcinomas (TCGA), ECCs less frequently harboured TP53 mutations. When a surrogate model for the molecular‐based TCGA classification was used, all molecular subtypes previously identified in endometrial endometrioid and serous carcinomas were present in the ECCs studied, including POLE, MMR‐deficient, copy‐number high (serous‐like)/p53 abnormal, and copy‐number low (endometrioid)/p53 wild‐type, which were significantly associated with disease‐free survival in univariate analysis. These findings demonstrate that ECCs constitute a histologically and genetically heterogeneous group of tumours with varying outcomes. Furthermore, our data suggest that the classification of ECCs as being generally ‘high‐grade’ or ‘type II’ tumours may not be warranted. Copyright


Nature Medicine | 2017

Whole-genome single-cell copy number profiling from formalin-fixed paraffin-embedded samples

Luciano G. Martelotto; Timour Baslan; Jude Kendall; Felipe C. Geyer; Kathleen A. Burke; Lee Spraggon; Salvatore Piscuoglio; Kalyani Chadalavada; Gouri Nanjangud; Charlotte K.Y. Ng; Pamela Moody; Sean D'Italia; Linda Rodgers; Hilary Cox; Arnaud Da Cruz Paula; Asya Stepansky; Michail Schizas; Hannah Y. Wen; Tari A. King; Larry Norton; Britta Weigelt; James Hicks; Jorge S. Reis-Filho

A substantial proportion of tumors consist of genotypically distinct subpopulations of cancer cells. This intratumor genetic heterogeneity poses a substantial challenge for the implementation of precision medicine. Single-cell genomics constitutes a powerful approach to resolve complex mixtures of cancer cells by tracing cell lineages and discovering cryptic genetic variations that would otherwise be obscured in tumor bulk analyses. Because of the chemical alterations that result from formalin fixation, single-cell genomic approaches have largely remained limited to fresh or rapidly frozen specimens. Here we describe the development and validation of a robust and accurate methodology to perform whole-genome copy-number profiling of single nuclei obtained from formalin-fixed paraffin-embedded clinical tumor samples. We applied the single-cell sequencing approach described here to study the progression from in situ to invasive breast cancer, which revealed that ductal carcinomas in situ show intratumor genetic heterogeneity at diagnosis and that these lesions may progress to invasive breast cancer through a variety of evolutionary processes.


Behavioural Brain Research | 2009

Growth deficits in a postnatal day 3 rat model of hypoxic-ischemic brain injury.

Wei Cheng Tai; Kathleen A. Burke; Jesus F. Dominguez; Lalitha Gundamraj; Jack E. Turman

The postnatal day (P) 3 rat model of hypoxic-ischemic (HI) brain injury provides valuable information regarding the cellular response to HI injury in a very immature brain. Our present study is the first to examine growth, metabolic, and behavioral outcomes following a P3 HI brain injury. Rats were injured by cauterizing the right common carotid, and exposure to 8% oxygen for 1.5h. Control rats received sham surgery and exposure to 1.5h of room air. One cohort of rats was examined for growth patterns through P33, evaluated using a battery of tests focused on early postnatal feeding behaviors, and studied using the open field paradigm during the early postnatal and postweaning periods. Another cohort of rats was used to examine metabolic parameters using indirect calorimetry. Significant growth deficits emerged in injured rats during the second postnatal week. No significant differences between groups were noted in the expression of feeding-related behaviors or in metabolic parameters between groups. However, we did observe significant associations between feeding-related behaviors and P14 growth parameters in injured rats. In the open field assessment, HI rats showed increased circling and supination behaviors only during the early postnatal period. Our data reveal that P3 HI brain injury results in generalized growth deficits that persist through postweaning. Analyses suggest that alterations in feeding-related behaviors contribute to growth deficits following a P3 HI brain injury.


Cancer Research | 2016

IDH2 Mutations Define a Unique Subtype of Breast Cancer with Altered Nuclear Polarity.

Sarah Chiang; Britta Weigelt; Huei Chi Wen; Fresia Pareja; Ashwini Raghavendra; Luciano G. Martelotto; Kathleen A. Burke; Thais Basili; Anqi Li; Felipe C. Geyer; Salvatore Piscuoglio; Charlotte K.Y. Ng; Achim A. Jungbluth; Jörg Balss; Stefan Pusch; Gabrielle Baker; Kimberly S. Cole; Andreas von Deimling; Julie M. Batten; Jonathan D. Marotti; Hwei Choo Soh; Benjamin L. McCalip; Jonathan Serrano; Raymond S. Lim; Kalliopi P. Siziopikou; Song Lu; Xiaolong Liu; Tarek Hammour; Edi Brogi; Matija Snuderl

Solid papillary carcinoma with reverse polarity (SPCRP) is a rare breast cancer subtype with an obscure etiology. In this study, we sought to describe its unique histopathologic features and to identify the genetic alterations that underpin SPCRP using massively parallel whole-exome and targeted sequencing. The morphologic and immunohistochemical features of SPCRP support the invasive nature of this subtype. Ten of 13 (77%) SPCRPs harbored hotspot mutations at R172 of the isocitrate dehydrogenase IDH2, of which 8 of 10 displayed concurrent pathogenic mutations affecting PIK3CA or PIK3R1 One of the IDH2 wild-type SPCRPs harbored a TET2 Q548* truncating mutation coupled with a PIK3CA H1047R hotspot mutation. Functional studies demonstrated that IDH2 and PIK3CA hotspot mutations are likely drivers of SPCRP, resulting in its reversed nuclear polarization phenotype. Our results offer a molecular definition of SPCRP as a distinct breast cancer subtype. Concurrent IDH2 and PIK3CA mutations may help diagnose SPCRP and possibly direct effective treatment. Cancer Res; 76(24); 7118-29. ©2016 AACR.


Oncogene | 2017

Compromised BRCA1|[ndash]|PALB2 interaction is associated with breast cancer risk

Tzeh Keong Foo; M Tischkowitz; Srilatha Simhadri; Talia Boshari; N Zayed; Kathleen A. Burke; Samuel H. Berman; P Blecua; N Riaz; Yanying Huo; Yuan Chun Ding; Susan L. Neuhausen; Britta Weigelt; Jorge S. Reis-Filho; W D Foulkes; Bing Xia

The major breast cancer suppressor proteins BRCA1 and BRCA2 play essential roles in homologous recombination (HR)-mediated DNA repair, which is thought to be critical for tumor suppression. The two BRCA proteins are linked by a third tumor suppressor, PALB2, in the HR pathway. While truncating mutations in these genes are generally pathogenic, interpretation of missense variants remains a challenge. To date, patient-derived missense variants that disrupt PALB2 binding have been identified in BRCA1 and BRCA2; however, there has not been sufficient evidence to prove their pathogenicity in humans, and no variants in PALB2 that disrupt either its BRCA1 or BRCA2 binding have been reported. Here we report on the identification of a novel PALB2 variant, c.104T>C (p.L35P), that segregates in a family with a strong history of breast cancer. Functional analyses showed that L35P abrogates the PALB2–BRCA1 interaction and completely disables its abilities to promote HR and confer resistance to platinum salts and PARP inhibitors. Whole-exome sequencing of a breast cancer from a c.104T>C carrier revealed a second, somatic, truncating mutation affecting PALB2, and the tumor displays hallmark genomic features of tumors with BRCA mutations and HR defects, cementing the pathogenicity of L35P. Parallel analyses of other germline variants in the PALB2 N-terminal BRCA1-binding domain identified multiple variants that affect HR function to varying degrees, suggesting their possible contribution to cancer development. Our findings establish L35P as the first pathogenic missense mutation in PALB2 and directly demonstrate the requirement of the PALB2-BRCA1 interaction for breast cancer suppression.

Collaboration


Dive into the Kathleen A. Burke's collaboration.

Top Co-Authors

Avatar

Britta Weigelt

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Felipe C. Geyer

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jorge S. Reis-Filho

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Salvatore Piscuoglio

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fresia Pareja

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Larry Norton

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Edi Brogi

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Luciano G. Martelotto

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Raymond S. Lim

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge