Network


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

Hotspot


Dive into the research topics where S.B. Lele is active.

Publication


Featured researches published by S.B. Lele.


Journal of the National Cancer Institute | 2015

Germline Mutations in the BRIP1, BARD1, PALB2, and NBN Genes in Women With Ovarian Cancer

Susan J. Ramus; Honglin Song; Ed Dicks; Jonathan Tyrer; Adam N. Rosenthal; Maria P. Intermaggio; Lindsay Fraser; Aleksandra Gentry-Maharaj; Jane Hayward; Susan Philpott; Christopher E. Anderson; Christopher K. Edlund; David V. Conti; Patricia Harrington; Daniel Barrowdale; David Bowtell; Kathryn Alsop; Gillian Mitchell; Mine S. Cicek; Julie M. Cunningham; Brooke L. Fridley; Jennifer Alsop; Mercedes Jimenez-Linan; Samantha Poblete; S.B. Lele; Lara E. Sucheston-Campbell; Kirsten B. Moysich; Weiva Sieh; Valerie McGuire; Jenny Lester

BACKGROUND Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy, responsible for 13 000 deaths per year in the United States. Risk prediction based on identifying germline mutations in ovarian cancer susceptibility genes could have a clinically significant impact on reducing disease mortality. METHODS Next generation sequencing was used to identify germline mutations in the coding regions of four candidate susceptibility genes-BRIP1, BARD1, PALB2 and NBN-in 3236 invasive EOC case patients and 3431 control patients of European origin, and in 2000 unaffected high-risk women from a clinical screening trial of ovarian cancer (UKFOCSS). For each gene, we estimated the prevalence and EOC risks and evaluated associations between germline variant status and clinical and epidemiological risk factor information. All statistical tests were two-sided. RESULTS We found an increased frequency of deleterious mutations in BRIP1 in case patients (0.9%) and in the UKFOCSS participants (0.6%) compared with control patients (0.09%) (P = 1 x 10(-4) and 8 x 10(-4), respectively), but no differences for BARD1 (P = .39), NBN1 ( P = .61), or PALB2 (P = .08). There was also a difference in the frequency of rare missense variants in BRIP1 between case patients and control patients (P = 5.5 x 10(-4)). The relative risks associated with BRIP1 mutations were 11.22 for invasive EOC (95% confidence interval [CI] = 3.22 to 34.10, P = 1 x 10(-4)) and 14.09 for high-grade serous disease (95% CI = 4.04 to 45.02, P = 2 x 10(-5)). Segregation analysis in families estimated the average relative risks in BRIP1 mutation carriers compared with the general population to be 3.41 (95% CI = 2.12 to 5.54, P = 7×10(-7)). CONCLUSIONS Deleterious germline mutations in BRIP1 are associated with a moderate increase in EOC risk. These data have clinical implications for risk prediction and prevention approaches for ovarian cancer and emphasize the critical need for risk estimates based on very large sample sizes before genes of moderate penetrance have clinical utility in cancer prevention.


Journal of Clinical Oncology | 2015

Contribution of Germline Mutations in the RAD51B, RAD51C, and RAD51D Genes to Ovarian Cancer in the Population

Honglin Song; Ed Dicks; Susan J. Ramus; Jonathan Tyrer; Maria P. Intermaggio; Jane Hayward; Christopher K. Edlund; David V. Conti; Patricia Harrington; Lindsay Fraser; Susan Philpott; Christopher N. G. Anderson; Adam Rosenthal; Aleksandra Gentry-Maharaj; David Bowtell; Kathryn Alsop; Mine S. Cicek; Julie M. Cunningham; Brooke L. Fridley; Jennifer Alsop; Mercedes Jimenez-Linan; Estrid Høgdall; C Hogdall; Allan Jensen; Susanne Kriiger Kjaer; Jan Lubinski; Tomasz Huzarski; Anna Jakubowska; Jacek Gronwald; Samantha Poblete

PURPOSE The aim of this study was to estimate the contribution of deleterious mutations in the RAD51B, RAD51C, and RAD51D genes to invasive epithelial ovarian cancer (EOC) in the population and in a screening trial of individuals at high risk of ovarian cancer. PATIENTS AND METHODS The coding sequence and splice site boundaries of the three RAD51 genes were sequenced and analyzed in germline DNA from a case-control study of 3,429 patients with invasive EOC and 2,772 controls as well as in 2,000 unaffected women who were BRCA1/BRCA2 negative from the United Kingdom Familial Ovarian Cancer Screening Study (UK_FOCSS) after quality-control analysis. RESULTS In the case-control study, we identified predicted deleterious mutations in 28 EOC cases (0.82%) compared with three controls (0.11%; P < .001). Mutations in EOC cases were more frequent in RAD51C (14 occurrences, 0.41%) and RAD51D (12 occurrences, 0.35%) than in RAD51B (two occurrences, 0.06%). RAD51C mutations were associated with an odds ratio of 5.2 (95% CI, 1.1 to 24; P = .035), and RAD51D mutations conferred an odds ratio of 12 (95% CI, 1.5 to 90; P = .019). We identified 13 RAD51 mutations (0.65%) in unaffected UK_FOCSS participants (RAD51C, n = 7; RAD51D, n = 5; and RAD51B, n = 1), which was a significantly greater rate than in controls (P < .001); furthermore, RAD51 mutation carriers were more likely than noncarriers to have a family history of ovarian cancer (P < .001). CONCLUSION These results confirm that RAD51C and RAD51D are moderate ovarian cancer susceptibility genes and suggest that they confer levels of risk of EOC that may warrant their use alongside BRCA1 and BRCA2 in routine clinical genetic testing.


British Journal of Cancer | 2014

Evidence for a time-dependent association between FOLR1 expression and survival from ovarian carcinoma: implications for clinical testing. An Ovarian Tumour Tissue Analysis consortium study

Martin Köbel; J Madore; Susan J. Ramus; Blaise Clarke; Paul Pharoah; Suha Deen; David Bowtell; Kunle Odunsi; Usha Menon; Carl Morrison; S.B. Lele; Wiam Bshara; Lara Sucheston; Matthias W. Beckmann; Alexander Hein; Falk C. Thiel; Arndt Hartmann; David L. Wachter; Michael S. Anglesio; Estrid Høgdall; Allan Jensen; Claus Høgdall; Kimberly R. Kalli; Brooke L. Fridley; Gary L. Keeney; Zachary C. Fogarty; Robert A. Vierkant; Suzanne Liu; S Cho; Gregg Nelson

Background:Folate receptor 1 (FOLR1) is expressed in the majority of ovarian carcinomas (OvCa), making it an attractive target for therapy. However, clinical trials testing anti-FOLR1 therapies in OvCa show mixed results and require better understanding of the prognostic relevance of FOLR1 expression. We conducted a large study evaluating FOLR1 expression with survival in different histological types of OvCa.Methods:Tissue microarrays composed of tumour samples from 2801 patients in the Ovarian Tumour Tissue Analysis (OTTA) consortium were assessed for FOLR1 expression by centralised immunohistochemistry. We estimated associations for overall (OS) and progression-free (PFS) survival using adjusted Cox regression models. High-grade serous ovarian carcinomas (HGSC) from The Cancer Genome Atlas (TCGA) were evaluated independently for association between FOLR1 mRNA upregulation and survival.Results:FOLR1 expression ranged from 76% in HGSC to 11% in mucinous carcinomas in OTTA. For HGSC, the association between FOLR1 expression and OS changed significantly during the years following diagnosis in OTTA (Pinteraction=0.01, N=1422) and TCGA (Pinteraction=0.01, N=485). In OTTA, particularly for FIGO stage I/II tumours, patients with FOLR1-positive HGSC showed increased OS during the first 2 years only (hazard ratio=0.44, 95% confidence interval=0.20–0.96) and patients with FOLR1-positive clear cell carcinomas (CCC) showed decreased PFS independent of follow-up time (HR=1.89, 95% CI=1.10–3.25, N=259). In TCGA, FOLR1 mRNA upregulation in HGSC was also associated with increased OS during the first 2 years following diagnosis irrespective of tumour stage (HR: 0.48, 95% CI: 0.25–0.94).Conclusions:FOLR1-positive HGSC tumours were associated with an increased OS in the first 2 years following diagnosis. Patients with FOLR1-negative, poor prognosis HGSC would be unlikely to benefit from anti-FOLR1 therapies. In contrast, a decreased PFS interval was observed for FOLR1-positive CCC. The clinical efficacy of FOLR1-targeted interventions should therefore be evaluated according to histology, stage and time following diagnosis.


Journal of Ovarian Research | 2011

Decreased levels of serum glutathione peroxidase 3 are associated with papillary serous ovarian cancer and disease progression

Deep Agnani; Olga Camacho-Vanegas; Catalina Camacho; S.B. Lele; Kunle Odunsi; Samantha Cohen; Peter Dottino; John A. Martignetti

BackgroundGlutathione peroxidase 3 (GPX3) is a selenocysteine-containing antioxidant enzyme that reacts with hydrogen peroxide and soluble fatty acid hydroperoxides, thereby helping to maintain redox balance within cells. Serum levels of GPX3 have been found to be reduced in various cancers including prostrate, thyroid, colorectal, breast and gastric cancers. Intriguingly, GPX3 has been reported to be upregulated in clear cell ovarian cancer tissues and thus may have implications in chemotherapeutic resistance. Since clear cell and serous subtypes of ovarian cancer represent two distinct disease entities, the aim of this study was to determine GPX3 levels in serous ovarian cancer patients and establish its potential as a biomarker for detection and/or surveillance of papillary serous ovarian cancer, the most frequent form of ovarian tumors in women.Patients and MethodsSerum was obtained from 66 patients (median age: 62 years, range: 22-89) prior to surgery and 65 controls with a comparable age-range (median age: 53 years, range: 25-83). ELISA was used to determine the levels of serum GPX3. The Mann Whitney U test was performed to determine statistical significance between the levels of serum GPX3 in patients and controls.ResultsSerum levels of GPX3 were found to be significantly lower in patients than controls (p = 1 × 10-2). Furthermore, this was found to be dependent on the stage of disease. While levels in early stage (I/II) patients showed no significant difference when compared to controls, there was a significant reduction in late stage (III/IV, p = 9 × 10-4) and recurrent (p = 1 × 10-2) patients. There was a statistically significant reduction in levels of GPX3 between early and late stage (p = 5 × 10-4) as well as early and recurrent (p = 1 × 10-2) patients. Comparison of women and controls stratified to include only women at or above 50 years of age shows that the same trends were maintained and the differences became more statistically significant.ConclusionsSerum GPX3 levels are decreased in women with papillary serous ovarian cancer in a stage-dependent manner and also decreased in women with disease recurrence. Whether this decrease represents a general feature in response to the disease or a link to the progression of the cancer is unknown. Understanding this relationship may have clinical and therapeutic consequences for women with papillary serous adenocarcinoma.


Journal of the National Cancer Institute | 2016

PPM1D Mosaic Truncating Variants in Ovarian Cancer Cases May Be Treatment-Related Somatic Mutations

Paul Pharoah; Honglin Song; Ed Dicks; Maria P. Intermaggio; Patricia Harrington; Caroline Baynes; Kathryn Alsop; Natalia Bogdanova; Mine S. Cicek; Julie M. Cunningham; Brooke L. Fridley; Aleksandra Gentry-Maharaj; Peter Hillemanns; S.B. Lele; Jenny Lester; Valerie McGuire; Kirsten B. Moysich; Samantha Poblete; Weiva Sieh; Lara E. Sucheston-Campbell; Martin Widschwendter; Alice S. Whittemore; Thilo Dörk; Usha Menon; Kunle Odunsi; Ellen L. Goode; Beth Y. Karlan; David Bowtell; Simon A. Gayther; Susan J. Ramus

Mosaic truncating mutations in the protein phosphatase, Mg(2+)/Mn(2+)-dependent, 1D (PPM1D) gene have recently been reported with a statistically significantly greater frequency in lymphocyte DNA from ovarian cancer case patients compared with unaffected control patients. Using massively parallel sequencing (MPS) we identified truncating PPM1D mutations in 12 of 3236 epithelial ovarian cancer (EOC) case patients (0.37%) but in only one of 3431 unaffected control patients (0.03%) (P = .001). All statistical tests were two-sided. A combination of Sanger sequencing, pyrosequencing, and MPS data suggested that 12 of the 13 mutations were mosaic. All mutations were identified in post-chemotherapy treatment blood samples from case patients (n = 1827) (average 1234 days post-treatment in carriers) rather than from cases collected pretreatment (less than 14 days after diagnosis, n = 1384) (P = .002). These data suggest that PPM1D variants in EOC cases are primarily somatic mosaic mutations caused by treatment and are not associated with germline predisposition to EOC.


Oncotarget | 2017

Germline whole exome sequencing and large-scale replication identifies FANCM as a likely high grade serous ovarian cancer susceptibility gene

Ed Dicks; Honglin Song; Susan J. Ramus; Elke Van Oudenhove; Jonathan Tyrer; Maria P. Intermaggio; Siddhartha Kar; Patricia Harrington; David Bowtell; Mine S. Cicek; Julie M. Cunningham; Brooke L. Fridley; Jennifer Alsop; Mercedes Jimenez-Linan; Anna Piskorz; Teodora Goranova; Emma Kent; Nadeem Siddiqui; James Paul; Robin Crawford; Samantha Poblete; S.B. Lele; Lara E. Sucheston-Campbell; Kirsten B. Moysich; Weiva Sieh; Valerie McGuire; Jenny Lester; Kunle Odunsi; Alice S. Whittemore; Natalia Bogdanova

We analyzed whole exome sequencing data in germline DNA from 412 high grade serous ovarian cancer (HGSOC) cases from The Cancer Genome Atlas Project and identified 5,517 genes harboring a predicted deleterious germline coding mutation in at least one HGSOC case. Gene-set enrichment analysis showed enrichment for genes involved in DNA repair (p = 1.8×10-3). Twelve DNA repair genes - APEX1, APLF, ATX, EME1, FANCL, FANCM, MAD2L2, PARP2, PARP3, POLN, RAD54L and SMUG1 – were prioritized for targeted sequencing in up to 3,107 HGSOC cases, 1,491 cases of other epithelial ovarian cancer (EOC) subtypes and 3,368 unaffected controls of European origin. We estimated mutation prevalence for each gene and tested for associations with disease risk. Mutations were identified in both cases and controls in all genes except MAD2L2, where we found no evidence of mutations in controls. In FANCM we observed a higher mutation frequency in HGSOC cases compared to controls (29/3,107 cases, 0.96 percent; 13/3,368 controls, 0.38 percent; P=0.008) with little evidence for association with other subtypes (6/1,491, 0.40 percent; P=0.82). The relative risk of HGSOC associated with deleterious FANCM mutations was estimated to be 2.5 (95% CI 1.3 – 5.0; P=0.006). In summary, whole exome sequencing of EOC cases with large-scale replication in case-control studies has identified FANCM as a likely novel susceptibility gene for HGSOC, with mutations associated with a moderate increase in risk. These data may have clinical implications for risk prediction and prevention approaches for high-grade serous ovarian cancer in the future and a significant impact on reducing disease mortality.We analyzed whole exome sequencing data in germline DNA from 412 high grade serous ovarian cancer (HGSOC) cases from The Cancer Genome Atlas Project and identified 5,517 genes harboring a predicted deleterious germline coding mutation in at least one HGSOC case. Gene-set enrichment analysis showed enrichment for genes involved in DNA repair (p = 1.8x10-3). Twelve DNA repair genes - APEX1, APLF, ATX, EME1, FANCL, FANCM, MAD2L2, PARP2, PARP3, POLN, RAD54L and SMUG1 - were prioritized for targeted sequencing in up to 3,107 HGSOC cases, 1,491 cases of other epithelial ovarian cancer (EOC) subtypes and 3,368 unaffected controls of European origin. We estimated mutation prevalence for each gene and tested for associations with disease risk. Mutations were identified in both cases and controls in all genes except MAD2L2, where we found no evidence of mutations in controls. In FANCM we observed a higher mutation frequency in HGSOC cases compared to controls (29/3,107 cases, 0.96 percent; 13/3,368 controls, 0.38 percent; P=0.008) with little evidence for association with other subtypes (6/1,491, 0.40 percent; P=0.82). The relative risk of HGSOC associated with deleterious FANCM mutations was estimated to be 2.5 (95% CI 1.3 - 5.0; P=0.006). In summary, whole exome sequencing of EOC cases with large-scale replication in case-control studies has identified FANCM as a likely novel susceptibility gene for HGSOC, with mutations associated with a moderate increase in risk. These data may have clinical implications for risk prediction and prevention approaches for high-grade serous ovarian cancer in the future and a significant impact on reducing disease mortality.


British Journal of Cancer | 2012

Pair-Box (PAX8) protein-positive expression is associated with poor disease outcome in women with endometrial cancer

Paulette Mhawech-Fauceglia; Dan Wang; D Samrao; Heidi Godoy; Tanja Pejovic; Song Liu; S.B. Lele

Background:The Pax8 transcription factor genes have a role in cell differentiation and cell growth, and silencing of Pax8 in cell cultures results in cell death. The aims of this study were to determine the expression and correlation of Pax8 protein with several clinicopathological variables in patients with endometrial cancer.Methods:The following clinical parameters from 229 patients were used for correlation with Pax8 expression; age, histological subtype, myometrial depth of invasion, lymphovascular invasion (LVI), the International Federation of Gynecology and Obstetrics grade, lymph nodes status, and disease status.Results:A positive association of Pax8+ expression was found with high tumour grade (P=0.002), LVI+(P=0.0186), and type II tumour subtype (P<0.0001) in univariate analysis. Survival analysis showed an association of Pax8 and 5-year overall survival probability (P=0.01486), 80.04% for patients with Pax8− and 55.59% for patients with Pax8+. There was also an association of Pax8 and 5-year disease-free survival probability (P=0.02028), 72.12% for patients with Pax8− vs 49.88% for patients with Pax8+. Finally, an association of Pax8+ and shorter recurrence-free survival was also found (P=0.00203), with 74.36% for Pax8− and 52.11% for Pax8+.Conclusion:Overexpression of Pax8 protein by endometrial cancer is associated with poor disease outcomes. Inhibition of Pax8 may be a very attractive targeted therapy for selective patients.


International Journal of Gynecological Pathology | 2006

Ovarian primary primitive neurectodermal tumor coexisting with endometrioid adenocarcinoma: a case report.

Gabor Fischer; Kunle Odunsi; S.B. Lele; Paulette Mhawech


Archives of Pathology & Laboratory Medicine | 2004

Pathologic Quiz Case: An Asymptomatic 79-Year-Old Woman With an Abnormal Papanicolaou Test

Thaer Khoury; S.B. Lele; Dongfeng Tan


Seminars in Oncology | 2015

A Young Female With Metastatic Nongestational Choriocarcinoma

Budhi Singh Yadav; Bhavana Rai; Vanita Suri; Kanchan K. Mukherjee; Amanjit Bal; Robert J. Morgan; Nicole Shonka; S.B. Lele; Gloria J. Morris

Collaboration


Dive into the S.B. Lele's collaboration.

Top Co-Authors

Avatar

Kunle Odunsi

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

P.J. Frederick

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

J.B. Szender

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Emese Zsiros

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

S.N. Akers

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Kevin H. Eng

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

K.S. Grzankowski

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Kirsten B. Moysich

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

P.C. Mayor

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

K. Morrell

Roswell Park Cancer Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge