1
|
Szafron LA, Sobiczewski P, Dansonka-Mieszkowska A, Kupryjanczyk J, Szafron LM. An Analysis of Genetic Polymorphisms in 76 Genes Related to the Development of Ovarian Tumors of Different Aggressiveness. Int J Mol Sci 2024; 25:10876. [PMID: 39456660 PMCID: PMC11507582 DOI: 10.3390/ijms252010876] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/12/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Borderline ovarian tumors (BOTS) are rare neoplasms of intermediate aggressiveness between cystadenomas and low-grade ovarian cancers (lgOvCa), which they share some molecular resemblances with. In contrast to the most frequent and well-described high-grade ovarian carcinomas (hgOvCa), the molecular background of BOTS and lgOvCa is less thoroughly characterized. Here, we aimed to analyze genetic variants in crucial tumor suppressors and oncogenes in BOTS (with or without the BRAF V600E mutation), lgOvCa, and hgOvCa in two gene panels using next-generation sequencing. Then, we verified the existence of selected polymorphisms by Sanger sequencing. Finally, Western blot analyses were carried out to check the impact of the selected polymorphisms on the expression of the corresponding proteins. Our study contributes to the molecular characterization of ovarian neoplasms, demonstrating divergent polymorphic patterns pointing to distinct signaling pathways engaged in their development. Certain mutations seem to play an important role in BOTS without the BRAF V600E variant (KRAS) and in lgOvCa (KRAS and NRAS), but not in hgOvCa. Additionally, based on multivariable regression analyses, potential biomarkers in BOTS (PARP1) and hgOvCa (FANCI, BRCA2, TSC2, FANCF) were identified. Noteworthy, for some of the analyzed genes, such as FANCI, FANCD2, and FANCI, FANCF, TSC2, the status of BRCA1/2 and TP53, respectively, turned out to be crucial. Our results shed new light on the similarities and differences in the polymorphic patterns between ovarian tumors of diverse aggressiveness. Furthermore, the biomarkers identified herein are of potential use as predictors of the prognosis and/or response to therapy.
Collapse
Affiliation(s)
- Laura A. Szafron
- Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| | - Piotr Sobiczewski
- Department of Gynecological Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Agnieszka Dansonka-Mieszkowska
- Cancer Molecular and Genetic Diagnostics Department, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Jolanta Kupryjanczyk
- Department of Cancer Pathomorphology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Lukasz M. Szafron
- Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| |
Collapse
|
2
|
Badlaeva A, Tregubova A, Palicelli A, Asaturova A. Eosinophilic Cells in Ovarian Borderline Serous Tumors as a Predictor of BRAF Mutation. Cancers (Basel) 2024; 16:2322. [PMID: 39001384 PMCID: PMC11240704 DOI: 10.3390/cancers16132322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
According to recent reports, ovarian serous borderline tumor (SBT) harboring the BRAF V600E mutation is associated with a lower risk of progression to low-grade serous carcinoma. Preliminary observations suggest that there may be an association between eosinophilic cells (ECs) and the above-mentioned mutation, so this study aimed to evaluate interobserver reproducibility for assessing ECs. Forty-two samples of SBTs were analyzed for ECs with abundant eosinophilic cytoplasm. Immunohistochemical staining and genetic pro-filing were performed in all cases to verify the BRAF V600E mutation. A BRAF V600E mutation was found in 19 of 42 (45%) cases. Inter-observer reproducibility in the assessment of ECs was substantial (κ = 0.7). The sensitivity and specificity for predicting the mutation were 79% and 91%, respectively. Patients with BRAF-mutated SBTs were significantly younger than those without mutation (p = 0.005). SBTs with BRAF mutation were less likely to be accompanied by non-invasive implants than wild-type SBT: 12% (2/17) versus 33% (6/18). Seven cases were excluded due to incomplete cytoreductive surgery. Nevertheless, Fisher's exact test showed no significant differences between the two groups (p = 0.228). Overall, this study strengthens the idea that ECs in ovarian SBTs may represent a mutation with prognostic significance, which can serve as a primary screening test for BRAF V600E mutation in this pathologic entity.
Collapse
Affiliation(s)
- Alina Badlaeva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Health of Russia, Bldg. 4, Oparina Street, 117513 Moscow, Russia; (A.B.); (A.T.)
| | - Anna Tregubova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Health of Russia, Bldg. 4, Oparina Street, 117513 Moscow, Russia; (A.B.); (A.T.)
| | - Andrea Palicelli
- Pathology Unit, Azienda Unità Sanitaria Locale—IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Aleksandra Asaturova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of the Ministry of Health of Russia, Bldg. 4, Oparina Street, 117513 Moscow, Russia; (A.B.); (A.T.)
| |
Collapse
|
3
|
Grisham RN, Slomovitz BM, Andrews N, Banerjee S, Brown J, Carey MS, Chui H, Coleman RL, Fader AN, Gaillard S, Gourley C, Sood AK, Monk BJ, Moore KN, Ray-Coquard I, Shih IM, Westin SN, Wong KK, Gershenson DM. Low-grade serous ovarian cancer: expert consensus report on the state of the science. Int J Gynecol Cancer 2023; 33:1331-1344. [PMID: 37591609 PMCID: PMC10511962 DOI: 10.1136/ijgc-2023-004610] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2023] [Indexed: 08/19/2023] Open
Abstract
Compared with high-grade serous carcinoma, low-grade serous carcinoma of the ovary or peritoneum is a less frequent epithelial ovarian cancer type that is poorly sensitive to chemotherapy and affects younger women, many of whom endure years of ineffective treatments and poor quality of life. The pathogenesis of this disease and its management remain incompletely understood. However, recent advances in the molecular characterization of the disease and identification of novel targeted therapies with activity in low-grade serous carcinoma offer the promise of improved outcomes. To update clinicians regarding recent scientific and clinical trial advancements and discuss unanswered questions related to low-grade serous carcinoma diagnosis and treatment, a panel of experts convened for a workshop in October 2022 to develop a consensus document addressing pathology, translational research, epidemiology and risk, clinical management, and ongoing research. In addition, the patient perspective was discussed. The recommendations developed by this expert panel-presented in this consensus document-will guide practitioners in all settings regarding the clinical management of women with low-grade serous carcinoma and discuss future opportunities to improve research and patient care.
Collapse
Affiliation(s)
- Rachel N Grisham
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| | - Brian M Slomovitz
- Department of Gynecologic Oncology, Mount Sinai Medical Center, Miami Beach, Florida, USA
- Florida International University, Miami, Florida, USA
| | - Nicole Andrews
- STAAR Ovarian Cancer Foundation, Western Springs, Illinois, USA
| | | | - Jubilee Brown
- Department of Gynecologic Oncology, Levine Cancer Institute at Atrium Health, Wake Forest University, Charlotte, North Carolina, USA
| | - Mark S Carey
- Division of Gynecologic Oncology, Vancouver Coastal Health, Vancouver, British Columbia, Canada
| | - Herman Chui
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Robert L Coleman
- Sarah Cannon Research Institute (SCRI), Nashville, Tennessee, USA
| | - Amanda N Fader
- Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Stephanie Gaillard
- Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Charlie Gourley
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh, UK
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bradley J Monk
- Division of Gynecologic Oncology, Honor Health, University of Arizona, Creighton University, Phoenix, Arizona, USA
| | - Kathleen N Moore
- Department of Gynecologic Oncology, Stephenson Cancer Center at the University of Oklahoma Health Sciences, Oklahoma City, Oklahoma, USA
| | - Isabelle Ray-Coquard
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
- Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Ie-Ming Shih
- Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kwong-Kwok Wong
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David M Gershenson
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
4
|
McHenry A, Rottmann DA, Buza N, Hui P. KRAS mutation in primary ovarian serous borderline tumors correlates with tumor recurrence. Virchows Arch 2023:10.1007/s00428-023-03564-z. [PMID: 37219599 DOI: 10.1007/s00428-023-03564-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/05/2023] [Accepted: 05/16/2023] [Indexed: 05/24/2023]
Abstract
Oncogenic activation of the mitogen-activated protein kinase (MAPK) pathway due to KRAS or BRAF gain-of-function mutation is frequently found in ovarian serous borderline tumor (SBT) and their extraovarian implants. We investigated mutational status of KRAS and BRAF of the primary ovarian SBTs that had a high stage presentation in correlation with clinical outcome. Among 39 consecutive primary SBTs with either invasive implants (20 cases) or non-invasive implants (19 cases), KRAS and BRAF mutational analysis was informative in 34 cases. Sixteen cases (47%) harbored a KRAS mutation, while 5 cases (15%) had a BRAF V600E mutation. High-stage disease (IIIC) was seen in 31% (5/16) of patients with a KRAS mutation and 39% (7/18) of patients without a KRAS mutation (p = 0.64). KRAS mutations were present in 9/16 (56%) tumors with invasive implants/LGSC versus 7/18 (39%) tumors with non-invasive implants (p = 0.31). BRAF mutation was seen in 5 cases with non-invasive implants. Tumor recurrence was seen in 31% (5/16) of patients with a KRAS mutation, compared to 6% (1/18) of patients without a KRAS mutation (p = 0.04). A KRAS mutation predicted an adverse disease-free survival (31% survival at 160 months) compared to those with wild-type KRAS (94% at 160 months; log-rank test, p = 0.037; HR 4.47). In conclusion, KRAS mutation in primary ovarian SBTs is significantly associated with a worse disease-free survival, independent of the high tumor stage or histological subtypes of extraovarian implant. KRAS mutation testing of primary ovarian SBT may servce as a useful biomarker for tumor recurrence.
Collapse
Affiliation(s)
- Austin McHenry
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Natalia Buza
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Pei Hui
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
5
|
Nilforoushan N, Liu L, Finkelman BS, Andersen J, Liu Y, James J, Hung CF, Wu TC, Vang R, Xing D. Ovarian Combined Serous Borderline Tumor/Low-grade Serous Carcinoma and Mesonephric-like Lesion: Report of 2 Cases With New Observations. Int J Gynecol Pathol 2023; 42:182-191. [PMID: 35348533 PMCID: PMC9675888 DOI: 10.1097/pgp.0000000000000868] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Ovarian combined serous borderline tumor/low-grade serous carcinomas (SBT/LGSC) and mesonephric-like adenocarcinomas (MLA) have been previously reported and the presence of identical oncogenic somatic mutations in both components supports the concept that at least some of MLAs arise from a Müllerian origin. We report 2 cases of ovarian combined SBT/LGSC and mesonephric-like lesion. Case 1 was a 70-yr-old woman presented with a liver lesion and omental carcinomatosis. Histologic examination revealed biphasic tumors in bilateral ovaries consisting of conventional SBT and invasive MLA with extraovarian spread. The right ovary also had a component of cribriform variant of SBT/noninvasive LGSC. The SBT/LGSC component was diffusely positive for Pax8, WT-1, and ER, focally positive for PR, and negative for GATA3, while the MLA component was diffusely positive for GATA3 but negative for WT-1, ER, and PR. Molecular analysis revealed a KRAS G12V mutation in both the SBT/LGSC and MLA components, indicating their clonal origin. Case 2 was a 58-yr-old woman who presented with conventional type SBT in both ovaries. In addition, the left ovarian tumor demonstrated a few areas (each <5 mm) of mesonephric-like differentiation/hyperplasia in close proximity to the serous-type epithelium, with an immunophenotype of focal GATA3 expression, luminal pattern of CD10 staining and negative WT-1, ER, and PR staining. This phenomenon has been reported in endometrioid borderline tumor but not in any serous type lesions. The findings in case 1 provide further evidence to demonstrate the clonal relationship between these morphologically and immunophenotypically distinct components. It also supports the theory that, unlike cervical mesonephric carcinomas originating from mesonephric remnants, MLAs are derived from a Müllerian-type lesion with differentiation into mesonephric lineage. The presence of a hyperplastic mesonephric-like lesion/differentiation in case 2 indicates that a precursor lesion in the same lineage with the potential to develop into MLA exists in the ovary.
Collapse
|
6
|
Xing D, Liang SX, Gao FF, Epstein JI. Mesonephric Adenocarcinoma and Mesonephric-like Adenocarcinoma of the Urinary Tract. Mod Pathol 2023; 36:100031. [PMID: 36788068 DOI: 10.1016/j.modpat.2022.100031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/19/2022] [Accepted: 09/21/2022] [Indexed: 01/19/2023]
Abstract
Given the association of mesonephric adenocarcinoma (MA) of the uterine cervix with florid mesonephric hyperplasia, one would expect MAs to rarely arise in other anatomical locations that harbor mesonephric remnants. In contrast, mesonephric-like adenocarcinoma (MLA) is thought to arise from Müllerian origin without an association with mesonephric remnants. The current case series characterizes 4 cases of MA arising in the urinary bladder (1 woman and 3 men), 1 case of MA in the perirenal region (woman), and 1 case of MLA in the ureter (woman). All cases displayed morphologic features similar to MA of the uterine cervix and MLA of the ovary and endometrium, characterized by predominant tubular and focal glandular/ductal architecture. Mesonephric remnants in the bladder wall were closely associated with adjacent MA in cases 1 and 4. MLA in case 6 was associated with mesonephric-like proliferations and endometriosis. All cases (6/6) were diffusely positive for Pax8, and all displayed a luminal pattern of CD10 staining, except case 4 for which CD10 immunostain was not available for review. Gata3 was either focally positive (cases 1, 2, and 6), negative (case 3), or diffusely positive (case 5). TTF-1 was diffusely expressed in cases 1 and 3 and negative in cases 2, 5, and 6. Although a KRAS G12C somatic mutation was detected in case 6, hotspot mutations in KRAS, NRAS, and PIK3CA were not present in other tested cases. Our study demonstrates that MAs and MLAs of the urinary tract share similar histopathogenesis, morphology, and immunophenotype to their counterparts in the female genital tract. We propose that, in the urinary tract, MA might be classified as a distinctive tumor that arises from mesonephric remnants or presumed Wolffian origin if they are not related to Müllerian-type precursors. The tumor displaying similar morphology and immunoprofile to MA but associated with Müllerian-type precursors should be classified as MLA.
Collapse
Affiliation(s)
- Deyin Xing
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, Maryland; Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, Maryland; Department of Gynecology and Obstetrics, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Sharon X Liang
- Department of Pathology, Allegheny Health Network/West Penn Hospital, Pittsburgh, Pennsylvania
| | - Faye F Gao
- Department of Pathology, MedStar Washington Hospital Center, Washington, District of Columbia
| | - Jonathan I Epstein
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, Maryland; Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, Maryland; Department of Urology, The Johns Hopkins Medical Institutions, Baltimore, Maryland.
| |
Collapse
|
7
|
Colic E, Patel PU, Kent OA. Aberrant MAPK Signaling Offers Therapeutic Potential for Treatment of Ovarian Carcinoma. Onco Targets Ther 2022; 15:1331-1346. [PMID: 36388156 PMCID: PMC9645123 DOI: 10.2147/ott.s361512] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 11/01/2022] [Indexed: 08/22/2023] Open
Abstract
Ovarian cancer remains the most lethal gynecological malignancy worldwide due to lack of effective screening, vague early symptoms, poor description of biomarkers, and absence of effective treatment regimes. Epithelial ovarian carcinoma (EOC) is categorized into five distinct disease subtypes which collectively account for ~90% of ovarian carcinomas. Most women present at advanced stages contributing to a poor overall 5-year survival rate. Standard treatment for EOC is cytoreductive surgery and platinum-based chemotherapy; however, most patients suffer from recurrence and platinum-resistant disease, which highlights an urgent need for targeted therapy. The high frequency of molecular alterations affecting gain-of-function signaling through the RAS mitogen-activated protein kinase (MAPK) pathway in EOC has prompted pre-clinical and clinical efforts toward research into the effectiveness of MAPK pathway inhibition as a second-line treatment. The RAS/MAPK pathway is a highly conserved signal transduction cascade, often disrupted in cancer, that regulates tumorigenic phenotypes including cellular proliferation, survival, migration, apoptosis, and differentiation. Herein, the role of the MAPK pathway in EOC with emphasis on targetability of the pathway is described. Pre-clinical and clinical efforts to target MAPK signaling in EOC have identified several MAPK pathway inhibitors that offer efficacious potential for monotherapy and in combination with other compounds. Thus, inhibition of the RAS/MAPK pathway is emerging as a tractable strategy for treatment of ovarian cancer that may permit development of personalized therapy and improved prognosis for women challenged by this disease.
Collapse
Affiliation(s)
- Eva Colic
- Department of Pharmacology, adMare BioInnovations, Montreal, Quebec, Canada
| | - Preya U Patel
- Department of Pharmacology, adMare BioInnovations, Montreal, Quebec, Canada
| | - Oliver A Kent
- Department of Pharmacology, adMare BioInnovations, Montreal, Quebec, Canada
| |
Collapse
|
8
|
Abstract
The RAS family of proteins is among the most frequently mutated genes in human malignancies. In ovarian cancer (OC), the most lethal gynecological malignancy, RAS, especially KRAS mutational status at codons 12, 13, and 61, ranges from 6-65% spanning different histo-types. Normally RAS regulates several signaling pathways involved in a myriad of cellular signaling cascades mediating numerous cellular processes like cell proliferation, differentiation, invasion, and death. Aberrant activation of RAS leads to uncontrolled induction of several downstream signaling pathways such as RAF-1/MAPK (mitogen-activated protein kinase), PI3K phosphoinositide-3 kinase (PI3K)/AKT, RalGEFs, Rac/Rho, BRAF (v-Raf murine sarcoma viral oncogene homolog B), MEK1 (mitogen-activated protein kinase kinase 1), ERK (extracellular signal-regulated kinase), PKB (protein kinase B) and PKC (protein kinase C) involved in cell proliferation as well as maintenance pathways thereby driving tumorigenesis and cancer cell propagation. KRAS mutation is also known to be a biomarker for poor outcome and chemoresistance in OC. As a malignancy with several histotypes showing varying histopathological characteristics, we focus on reviewing recent literature showcasing the involvement of oncogenic RAS in mediating carcinogenesis and chemoresistance in OC and its subtypes.
Collapse
Affiliation(s)
- Lubna Therachiyil
- Hamad Medical Corporation, Doha, Qatar, 3050, Qatar
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, 2713, Qatar
| | - Anjana Anand
- Hamad Medical Corporation, Doha, Qatar, 3050, Qatar
| | | | | | - Hesham M. Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, 2713, Qatar
| | - Shahab Uddin
- Hamad Medical Corporation, Doha, Qatar, 3050, Qatar
| |
Collapse
|
9
|
MicroRNAs miR-451a and Let-7i-5p Profiles in Circulating Exosomes Vary among Individuals with Different Sickle Hemoglobin Genotypes and Malaria. J Clin Med 2022; 11:jcm11030500. [PMID: 35159951 PMCID: PMC8837188 DOI: 10.3390/jcm11030500] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/10/2022] [Accepted: 01/13/2022] [Indexed: 01/22/2023] Open
Abstract
Sickle cell disease (SCD) occurs when two alleles of mutated hemoglobin (HbS or HbC) are inherited (HbSS and HbSC) rather than one (HbAS or HbAC), which indicates a person carries the sickle cell trait. The high prevalence of these two alleles in Africa have been associated with reduced malaria susceptibility. Recent in vitro research has been shown that microRNAs (miRNAs) miR-451a and let-7i-5p are differentially expressed in HbSS erythrocytes compared to healthy controls (HbAA) and are overexpressed in Plasmodium-infected malaria erythrocytes. However, these miRNAs have not been fully examined in the plasma of people with different sickle hemoglobin genotypes. Plasma circulating miRNAs are commonly encapsulated in extracellular vesicles, such as exosomes, and are thought to play a role in disease development. Circulating exosomal miR-451a and let-7i-5p were quantified from individuals with various hemoglobin genotypes (HbAA, HbAS, HbAC, HbSS, HbSC, and HbCC) with (+) and without (-) malaria. The results showed a higher level of exosomal let-7i-5p and miR-451a in HbSS-. Exosomal let-7i-5p and miR-451a levels were lower in HbSS+ compared to other genotypes. Based on the area under the curve (AUC) of the Receiver Operating Characteristics (ROCs), both exosomal miRNAs may be useful disease biomarkers for SCD with malaria. Finally, miR-451a and let-7i-5p modulate genes involved in inflammation, making them potential biomarkers of pathogenesis for both diseases.
Collapse
|
10
|
Mutational spectrum in clinically aggressive low-grade serous carcinoma/serous borderline tumors of the ovary-Clinical significance of BRCA2 gene variants in genomically stable tumors. Gynecol Oncol 2021; 161:762-768. [PMID: 33773808 DOI: 10.1016/j.ygyno.2021.03.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/15/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVE The mutational spectra of low-grade serous carcinomas (LGSCs) and serous borderline tumors (SBTs) of the ovary are poorly characterized. We present 17 cases of advanced or recurrent LGSC/SBT patients who underwent molecular profiling. METHODS Thirteen LGSCs and four SBTs underwent targeted gene panel testing by massively parallel sequencing. Microsatellite stability and tumor mutation burdens (TMBs) were determined based on panel sequencing data. RESULTS The mean TMB was 5.2 mutations/megabase (range 3-10) in 14 cases. Twelve of twelve (12/12) cases were microsatellite stable. Clear driver mutations were identified in 11 cases, namely KRAS (5/17), BRAF (2/17), NRAS (2/17) and ERBB2 (2/17). Five cases harbored BRCA2 alterations (allele fractions: 44-51%), including two classified as likely benign/benign variants, and three classified as variants of uncertain significance (VUSs), with two variants being confirmed to be germline. The three BRCA2 VUSs were missense variants that were assessed to be of unlikely clinical significance, based on family cancer history and expected impact on protein function. Two patients received PARP inhibitors during their disease course, with neither of the patients demonstrating appreciable response. CONCLUSIONS The mutational spectra in 17 clinically aggressive SBT/LGSC cases demonstrate genomically stable tumors, frequently driven by the RTK/RAS/MAPK pathway. While BRCA2 variants were identified, our data demonstrate BRCA2 gene variants are at most VUSs and of dubious clinical significance, in contrast to disease-associated BRCA1/2 variants that may be identified in high-grade serous carcinoma. Germline testing and PARP inhibitors are thus expected to provide limited benefit to patients with LGSC/SBTs.
Collapse
|
11
|
Shrestha R, Llaurado Fernandez M, Dawson A, Hoenisch J, Volik S, Lin YY, Anderson S, Kim H, Haegert AM, Colborne S, Wong NKY, McConeghy B, Bell RH, Brahmbhatt S, Lee CH, DiMattia GE, Le Bihan S, Morin GB, Collins CC, Carey MS. Multiomics Characterization of Low-Grade Serous Ovarian Carcinoma Identifies Potential Biomarkers of MEK Inhibitor Sensitivity and Therapeutic Vulnerability. Cancer Res 2021; 81:1681-1694. [PMID: 33441310 DOI: 10.1158/0008-5472.can-20-2222] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/12/2020] [Accepted: 01/11/2021] [Indexed: 11/16/2022]
Abstract
Low-grade serous ovarian carcinoma (LGSOC) is a rare tumor subtype with high case fatality rates in patients with metastatic disease. There is a pressing need to develop effective treatments using newly available preclinical models for therapeutic discovery and drug evaluation. Here, we use multiomics integration of whole-exome sequencing, RNA sequencing, and mass spectrometry-based proteomics on 14 LGSOC cell lines to elucidate novel biomarkers and therapeutic vulnerabilities. Comparison of LGSOC cell line data with LGSOC tumor data enabled predictive biomarker identification of MEK inhibitor (MEKi) efficacy, with KRAS mutations found exclusively in MEKi-sensitive cell lines and NRAS mutations found mostly in MEKi-resistant cell lines. Distinct patterns of Catalogue of Somatic Mutations in Cancer mutational signatures were identified in MEKi-sensitive and MEKi-resistant cell lines. Deletions of CDKN2A/B and MTAP genes were more frequent in cell lines than tumor samples and possibly represent key driver events in the absence of KRAS/NRAS/BRAF mutations. These LGSOC cell lines were representative models of the molecular aberrations found in LGSOC tumors. For prediction of in vitro MEKi efficacy, proteomic data provided better discrimination than gene expression data. Condensin, minichromosome maintenance, and replication factor C protein complexes were identified as potential treatment targets in MEKi-resistant cell lines. This study suggests that CDKN2A/B or MTAP deficiency may be exploited using synthetically lethal treatment strategies, highlighting the importance of using proteomic data as a tool for molecular drug prediction. Multiomics approaches are crucial to improving our understanding of the molecular underpinnings of LGSOC and applying this information to develop new therapies. SIGNIFICANCE: These findings highlight the utility of global multiomics to characterize LGSOC cell lines as research models, to determine biomarkers of MEKi resistance, and to identify potential novel therapeutic targets.
Collapse
Affiliation(s)
- Raunak Shrestha
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Marta Llaurado Fernandez
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amy Dawson
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joshua Hoenisch
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stanislav Volik
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Yen-Yi Lin
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shawn Anderson
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Hannah Kim
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anne M Haegert
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Shane Colborne
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Nelson K Y Wong
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Pathology & Laboratory Medicine, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Brian McConeghy
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Robert H Bell
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | | | - Cheng-Han Lee
- Department of Pathology & Laboratory Medicine, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Gabriel E DiMattia
- Translational Ovarian Cancer Research Program, London Health Science Centre, London, Ontario, Canada
| | | | - Gregg B Morin
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Colin C Collins
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada. .,Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mark S Carey
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
12
|
Van Nieuwenhuysen E, Busschaert P, Laenen A, Moerman P, Han SN, Neven P, Lambrechts D, Vergote I. Loss of 1p36.33 Frequent in Low-Grade Serous Ovarian Cancer. Neoplasia 2019; 21:582-590. [PMID: 31054497 PMCID: PMC6500912 DOI: 10.1016/j.neo.2019.03.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/29/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND: Low-grade serous ovarian cancer (LGSOC) is a rare subtype of epithelial ovarian carcinoma. Limited data regarding the molecular-genetic background exist beyond mutations in the RAS signaling pathway. There is a growing need to better characterize these tumors due to chemoresistance and limited therapeutic options in advanced or recurrent disease. METHODS: We performed genome-wide copy number aberration (CNA) profiles and mutation hotspot screening (KRAS, BRAF, NRAS, ERBB2, PIK3CA, TP53) in 38 LGSOC tumor samples. RESULTS: We detected mutations in the RAS-signaling pathway in 36.8% of cases, including seven KRAS, four BRAF, and three NRAS mutations. We identified two mutations in PIK3CA and one mutation in MAP3K1, EGFR, and TP53. CNAs were detected in 86.5% of cases. None of the focal aberrations was correlated with specific clinical characteristics. The most frequently detected CNA was loss of 1p36.33 in 54.1% of cases, with a trend towards lower progression-free survival and overall survival in patients with 1p36.33 loss. CONCLUSIONS: Activating RAS mutations were dominant in our series, with supplementary detection of two PIK3CA mutations which may lead to therapeutic options. Furthermore, we detected 1p36.33 deletions in half of the cases, indicating a role in tumorigenesis, and these deletions may serve as a prognostic marker.
Collapse
Affiliation(s)
- Els Van Nieuwenhuysen
- Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium; Division of Gynaecological Oncology, Leuven Cancer Institute, Kuleuven, Leuven, Belgium.
| | - Pieter Busschaert
- Laboratory for Translational Genetics Department of Oncology, KU, Leuven, Belgium; Center for Cancer Biology, VIB, Leuven, Belgium
| | - Annouschka Laenen
- Leuven Biostatistics and Statistical Bioinformatics Centre, KULeuven, Leuven, Belgium
| | - Philippe Moerman
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Sileny N Han
- Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium
| | - Patrick Neven
- Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics Department of Oncology, KU, Leuven, Belgium; Center for Cancer Biology, VIB, Leuven, Belgium
| | - Ignace Vergote
- Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium; Division of Gynaecological Oncology, Leuven Cancer Institute, Kuleuven, Leuven, Belgium
| |
Collapse
|
13
|
Khan AQ, Kuttikrishnan S, Siveen KS, Prabhu KS, Shanmugakonar M, Al-Naemi HA, Haris M, Dermime S, Uddin S. RAS-mediated oncogenic signaling pathways in human malignancies. Semin Cancer Biol 2019; 54:1-13. [PMID: 29524560 DOI: 10.1016/j.semcancer.2018.03.001] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 03/06/2018] [Indexed: 02/07/2023]
Abstract
Abnormally activated RAS proteins are the main oncogenic driver that governs the functioning of major signaling pathways involved in the initiation and development of human malignancies. Mutations in RAS genes and or its regulators, most frequent in human cancers, are the main force for incessant RAS activation and associated pathological conditions including cancer. In general, RAS is the main upstream regulator of the highly conserved signaling mechanisms associated with a plethora of important cellular activities vital for normal homeostasis. Mutated or the oncogenic RAS aberrantly activates a web of interconnected signaling pathways including RAF-MEK (mitogen-activated protein kinase kinase)-ERK (extracellular signal-regulated kinase), phosphoinositide-3 kinase (PI3K)/AKT (protein kinase B), protein kinase C (PKC) and ral guanine nucleotide dissociation stimulator (RALGDS), etc., leading to uncontrolled transcriptional expression and reprogramming in the functioning of a range of nuclear and cytosolic effectors critically associated with the hallmarks of carcinogenesis. This review highlights the recent literature on how oncogenic RAS negatively use its signaling web in deregulating the expression and functioning of various effector molecules in the pathogenesis of human malignancies.
Collapse
Affiliation(s)
- Abdul Q Khan
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Shilpa Kuttikrishnan
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Kodappully S Siveen
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Kirti S Prabhu
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | | | - Hamda A Al-Naemi
- Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Mohammad Haris
- Translational Medicine Research Branch, Sidra Medical and Research Center, Doha, Qatar
| | - Said Dermime
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
14
|
Fernandez ML, Dawson A, Hoenisch J, Kim H, Bamford S, Salamanca C, DiMattia G, Shepherd T, Cremona M, Hennessy B, Anderson S, Volik S, Collins CC, Huntsman DG, Carey MS. Markers of MEK inhibitor resistance in low-grade serous ovarian cancer: EGFR is a potential therapeutic target. Cancer Cell Int 2019. [PMID: 30636931 DOI: 10.1186/s12935-019-0725-1]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Although low-grade serous ovarian cancer (LGSC) is rare, case-fatality rates are high as most patients present with advanced disease and current cytotoxic therapies are not overly effective. Recognizing that these cancers may be driven by MAPK pathway activation, MEK inhibitors (MEKi) are being tested in clinical trials. LGSC respond to MEKi only in a subgroup of patients, so predictive biomarkers and better therapies will be needed. METHODS We evaluated a number of patient-derived LGSC cell lines, previously classified according to their MEKi sensitivity. Two cell lines were genomically compared against their matching tumors samples. MEKi-sensitive and MEKi-resistant lines were compared using whole exome sequencing and reverse phase protein array. Two treatment combinations targeting MEKi resistance markers were also evaluated using cell proliferation, cell viability, cell signaling, and drug synergism assays. RESULTS Low-grade serous ovarian cancer cell lines recapitulated the genomic aberrations from their matching tumor samples. We identified three potential predictive biomarkers that distinguish MEKi sensitive and resistant lines: KRAS mutation status, and EGFR and PKC-alpha protein expression. The biomarkers were validated in three newly developed LGSC cell lines. Sub-lethal combination of MEK and EGFR inhibition showed drug synergy and caused complete cell death in two of four MEKi-resistant cell lines tested. CONCLUSIONS KRAS mutations and the protein expression of EGFR and PKC-alpha should be evaluated as predictive biomarkers in patients with LGSC treated with MEKi. Combination therapy using a MEKi with EGFR inhibition may represent a promising new therapy for patients with MEKi-resistant LGSC.
Collapse
Affiliation(s)
| | - Amy Dawson
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Joshua Hoenisch
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Hannah Kim
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Sylvia Bamford
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Clara Salamanca
- 2Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Gabriel DiMattia
- 3Translational Ovarian Cancer Research Program, London Health Science Centre, London, ON Canada.,4Oncology, University of Western Ontario, London, ON Canada
| | - Trevor Shepherd
- 3Translational Ovarian Cancer Research Program, London Health Science Centre, London, ON Canada.,4Oncology, University of Western Ontario, London, ON Canada
| | - Mattia Cremona
- Medical Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Bryan Hennessy
- Medical Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Shawn Anderson
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - Stanislav Volik
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - Colin C Collins
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - David G Huntsman
- 2Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada.,7Molecular Oncology, British Columbia Cancer Agency, Vancouver, BC Canada
| | - Mark S Carey
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada.,Division of Gynecologic Oncology, Diamond Health Centre, 2775 Laurel St., 6th Floor, Vancouver, BC V5Z 1M9 Canada
| |
Collapse
|
15
|
Fernandez ML, Dawson A, Hoenisch J, Kim H, Bamford S, Salamanca C, DiMattia G, Shepherd T, Cremona M, Hennessy B, Anderson S, Volik S, Collins CC, Huntsman DG, Carey MS. Markers of MEK inhibitor resistance in low-grade serous ovarian cancer: EGFR is a potential therapeutic target. Cancer Cell Int 2019. [PMID: 30636931 DOI: 10.1186/s12935-019-0725-1] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Background Although low-grade serous ovarian cancer (LGSC) is rare, case-fatality rates are high as most patients present with advanced disease and current cytotoxic therapies are not overly effective. Recognizing that these cancers may be driven by MAPK pathway activation, MEK inhibitors (MEKi) are being tested in clinical trials. LGSC respond to MEKi only in a subgroup of patients, so predictive biomarkers and better therapies will be needed. Methods We evaluated a number of patient-derived LGSC cell lines, previously classified according to their MEKi sensitivity. Two cell lines were genomically compared against their matching tumors samples. MEKi-sensitive and MEKi-resistant lines were compared using whole exome sequencing and reverse phase protein array. Two treatment combinations targeting MEKi resistance markers were also evaluated using cell proliferation, cell viability, cell signaling, and drug synergism assays. Results Low-grade serous ovarian cancer cell lines recapitulated the genomic aberrations from their matching tumor samples. We identified three potential predictive biomarkers that distinguish MEKi sensitive and resistant lines: KRAS mutation status, and EGFR and PKC-alpha protein expression. The biomarkers were validated in three newly developed LGSC cell lines. Sub-lethal combination of MEK and EGFR inhibition showed drug synergy and caused complete cell death in two of four MEKi-resistant cell lines tested. Conclusions KRAS mutations and the protein expression of EGFR and PKC-alpha should be evaluated as predictive biomarkers in patients with LGSC treated with MEKi. Combination therapy using a MEKi with EGFR inhibition may represent a promising new therapy for patients with MEKi-resistant LGSC.
Collapse
Affiliation(s)
| | - Amy Dawson
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Joshua Hoenisch
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Hannah Kim
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Sylvia Bamford
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Clara Salamanca
- 2Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Gabriel DiMattia
- 3Translational Ovarian Cancer Research Program, London Health Science Centre, London, ON Canada.,4Oncology, University of Western Ontario, London, ON Canada
| | - Trevor Shepherd
- 3Translational Ovarian Cancer Research Program, London Health Science Centre, London, ON Canada.,4Oncology, University of Western Ontario, London, ON Canada
| | - Mattia Cremona
- Medical Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Bryan Hennessy
- Medical Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Shawn Anderson
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - Stanislav Volik
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - Colin C Collins
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - David G Huntsman
- 2Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada.,7Molecular Oncology, British Columbia Cancer Agency, Vancouver, BC Canada
| | - Mark S Carey
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada.,Division of Gynecologic Oncology, Diamond Health Centre, 2775 Laurel St., 6th Floor, Vancouver, BC V5Z 1M9 Canada
| |
Collapse
|
16
|
Fernandez ML, Dawson A, Hoenisch J, Kim H, Bamford S, Salamanca C, DiMattia G, Shepherd T, Cremona M, Hennessy B, Anderson S, Volik S, Collins CC, Huntsman DG, Carey MS. Markers of MEK inhibitor resistance in low-grade serous ovarian cancer: EGFR is a potential therapeutic target. Cancer Cell Int 2019; 19:10. [PMID: 30636931 PMCID: PMC6325847 DOI: 10.1186/s12935-019-0725-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/02/2019] [Indexed: 02/06/2023] Open
Abstract
Background Although low-grade serous ovarian cancer (LGSC) is rare, case-fatality rates are high as most patients present with advanced disease and current cytotoxic therapies are not overly effective. Recognizing that these cancers may be driven by MAPK pathway activation, MEK inhibitors (MEKi) are being tested in clinical trials. LGSC respond to MEKi only in a subgroup of patients, so predictive biomarkers and better therapies will be needed. Methods We evaluated a number of patient-derived LGSC cell lines, previously classified according to their MEKi sensitivity. Two cell lines were genomically compared against their matching tumors samples. MEKi-sensitive and MEKi-resistant lines were compared using whole exome sequencing and reverse phase protein array. Two treatment combinations targeting MEKi resistance markers were also evaluated using cell proliferation, cell viability, cell signaling, and drug synergism assays. Results Low-grade serous ovarian cancer cell lines recapitulated the genomic aberrations from their matching tumor samples. We identified three potential predictive biomarkers that distinguish MEKi sensitive and resistant lines: KRAS mutation status, and EGFR and PKC-alpha protein expression. The biomarkers were validated in three newly developed LGSC cell lines. Sub-lethal combination of MEK and EGFR inhibition showed drug synergy and caused complete cell death in two of four MEKi-resistant cell lines tested. Conclusions KRAS mutations and the protein expression of EGFR and PKC-alpha should be evaluated as predictive biomarkers in patients with LGSC treated with MEKi. Combination therapy using a MEKi with EGFR inhibition may represent a promising new therapy for patients with MEKi-resistant LGSC. Electronic supplementary material The online version of this article (10.1186/s12935-019-0725-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Amy Dawson
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Joshua Hoenisch
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Hannah Kim
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Sylvia Bamford
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada
| | - Clara Salamanca
- 2Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada
| | - Gabriel DiMattia
- 3Translational Ovarian Cancer Research Program, London Health Science Centre, London, ON Canada.,4Oncology, University of Western Ontario, London, ON Canada
| | - Trevor Shepherd
- 3Translational Ovarian Cancer Research Program, London Health Science Centre, London, ON Canada.,4Oncology, University of Western Ontario, London, ON Canada
| | - Mattia Cremona
- Medical Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Bryan Hennessy
- Medical Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Shawn Anderson
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - Stanislav Volik
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - Colin C Collins
- 6Laboratory for Advanced Genome Analysis, Vancouver Prostate Centre, Vancouver, BC Canada
| | - David G Huntsman
- 2Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC Canada.,7Molecular Oncology, British Columbia Cancer Agency, Vancouver, BC Canada
| | - Mark S Carey
- 1Obstetrics and Gynecology, University of British Columbia, Vancouver, BC Canada.,Division of Gynecologic Oncology, Diamond Health Centre, 2775 Laurel St., 6th Floor, Vancouver, BC V5Z 1M9 Canada
| |
Collapse
|
17
|
Ilenkovan N, Gourley C. Pathogenesis, Genetics, and Genomics of Non-High Grade Serous Ovarian Cancers. Hematol Oncol Clin North Am 2018; 32:929-942. [PMID: 30390766 DOI: 10.1016/j.hoc.2018.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The 5 main non-high grade serous epithelial ovarian cancers (clear cell, low grade endometrioid, low grade serous, mucinous, and carcinosarcoma) are discrete in terms of their pathogenesis, molecular biology, and treatment sensitivity. This article reviews the current understanding of their pathogenesis and molecular biology, highlighting areas of uncertainty where future research efforts should be focused.
Collapse
Affiliation(s)
- Narthana Ilenkovan
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK
| | - Charlie Gourley
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK.
| |
Collapse
|
18
|
Moujaber T, Etemadmoghadam D, Kennedy CJ, Chiew YE, Balleine RL, Saunders C, Wain GV, Gao B, Hogg R, Srirangan S, Kan C, Fereday S, Traficante N, Patch AM, Pearson JV, Waddell N, Grimmond SM, Dobrovic A, Bowtell DD, Harnett PR, deFazio A. BRAF Mutations in Low-Grade Serous Ovarian Cancer and Response to BRAF Inhibition. JCO Precis Oncol 2018; 2:1-14. [DOI: 10.1200/po.17.00221] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Low-grade serous ovarian carcinoma (LGSC) responds poorly to chemotherapy and is characterized by activating mutations in the Ras sarcoma–mitogen-activated protein kinase (RAS-MAPK) pathway, including oncogenic BRAF. However, response to BRAF inhibitors is tumor-type specific. Significant improvement in survival is seen in patients with BRAF-mutant melanoma, but other cancer types, such as colorectal cancers, are generally less sensitive. We examined the frequency and characteristics of BRAF-mutated LGSC and described the response to treatment with BRAF inhibitors. Patients and Methods Mutations were assessed in LGSC (N = 65) by using targeted, exome, and whole-genome sequencing. Patient characteristics, treatment, and clinical outcome were assessed, and the median follow-up time was more than 5 years. BRAF inhibitors were trialed in two patients with a somatic BRAF V600E mutation: one patient received dabrafenib monotherapy and was monitored clinically, biochemically (cancer antigen [CA]-125 levels), and with positron emission tomography (PET) imaging. Expression of the BRAF V600E protein in this patient was assessed by immunohistochemistry. Results Among patients with LGSC, nine (13.8%) of 65 had a somatic BRAF mutation. Of the nine patients with BRAF mutation–positive LGSC, four experienced progressive disease that did not respond to conventional chemotherapy. Two of the patients experienced progression quickly and died as a result of disease progression, and two received targeted treatment. Two patients with BRAF V600E mutation received BRAF inhibitors at relapse and both achieved durable responses. Conclusion BRAF mutations are not uncommon in patients with LGSC and should be routinely tested, because BRAF inhibitors can be an effective treatment for these patients. The results highlight the need for targeted treatment in this rare tumor type, and a prospective study is needed to formally assess the response rate and clinical benefit.
Collapse
Affiliation(s)
- Tania Moujaber
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Dariush Etemadmoghadam
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Catherine J. Kennedy
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Yoke-Eng Chiew
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Rosemary L. Balleine
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Catherine Saunders
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Gerard V. Wain
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Bo Gao
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Russell Hogg
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Sivatharsny Srirangan
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Casina Kan
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Sian Fereday
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Nadia Traficante
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Ann-Marie Patch
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - John V. Pearson
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Nicola Waddell
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Sean M. Grimmond
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Alexander Dobrovic
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - David D.L. Bowtell
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Paul R. Harnett
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | - Anna deFazio
- Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Bo Gao, Sivatharsny Srirangan, Casina Kan, Paul R. Harnett, and Anna deFazio, Westmead Institute for Medical Research; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Russell Hogg, Paul R. Harnett, and Anna DeFazio, University of Sydney; Tania Moujaber, Catherine J. Kennedy, Yoke-Eng Chiew, Rosemary L. Balleine, Catherine Saunders, Gerard V. Wain, Bo Gao, Paul R. Harnett, and Anna
| | | |
Collapse
|
19
|
Testing for NRAS Mutations in Serous Borderline Ovarian Tumors and Low-Grade Serous Ovarian Carcinomas. DISEASE MARKERS 2018; 2018:1497879. [PMID: 29682098 PMCID: PMC5845515 DOI: 10.1155/2018/1497879] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/31/2017] [Accepted: 01/30/2018] [Indexed: 12/24/2022]
Abstract
The Idylla NRAS Mutation Test, performed on the Biocartis Idylla system, is an in vitro diagnostic tool for the qualitative assessment of 18 NRAS mutations in codons 12, 13, 59, 61, 117, and 146. Low-grade serous ovarian cancer (LGSC) represents less than 10% of all serous ovarian carcinomas. LGSCs are believed to arise from preexisting cystadenomas or serous borderline tumors (SBOTs) that eventually progress to an invasive carcinoma. The molecular analysis of cancer-causing mutations and the development of targeted biological therapies constitute a milestone in the diagnosis and therapy of ovarian malignancies. According to some authors, NRAS may be an important oncogene for the progression of SBOT to a frankly invasive disease. The primary aim of this study was to verify if a fully integrated, real-time PCR-based Idylla system can be used for the rapid determination of the NRAS mutation status in patients with serous borderline ovarian tumors and low-grade serous ovarian carcinomas. The study included tissue specimens from 12 patients with histopathologically verified ovarian masses, operated on at the Department of Obstetrics and Gynecology, Nicolaus Copernicus University, Collegium Medicum in Bydgoszcz (Poland), between January 2009 and June 2012. The mean age of the study patients was 52.5 years (range 27-80 years). NRAS mutation in codon 13 (G13D, p.Gly13Asp; nucleotide: c.38G>A) was found in one patient, a woman with low-grade serous ovarian carcinoma. To the best of our knowledge, our experiment was the first published study using the novel Idylla NRAS Mutation Test for the evaluation of ovarian tumors in a clinical setting. The Idylla platform is an interesting ancillary first-line rapid and fully automated instrument to detect NRAS mutations in SBOTs and LGSCs. However, the clinical usefulness of this method still needs to be verified in larger groups of cancer patients.
Collapse
|