1
|
Sama S, Rosqvist S, Savage T, Lomo L, Sibbald K, Straubhar A, Werner TL. Durable response to BRAF inhibitor monotherapy in recurrent metastatic low grade serous ovarian cancer. Gynecol Oncol Rep 2024; 53:101412. [PMID: 38779189 PMCID: PMC11109349 DOI: 10.1016/j.gore.2024.101412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/05/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
Low grade serous ovarian cancers (LGSOC) in an advanced setting have limited systemic treatment options. In this paper we report a case of metastatic LGSOC harboring a BRAF mutation, treated with dabrafenib. We discuss the clinical, pathologic and molecular characteristics as well as surgical considerations and ongoing investigations in LGSOC.
Collapse
Affiliation(s)
- Shashank Sama
- Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT, USA
| | | | - Talicia Savage
- Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT, USA
| | - Lesley Lomo
- Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT, USA
| | | | | | - Theresa L. Werner
- Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
2
|
Au-Yeung G, MacArthur E, Chan J, Ilenkovan N, Frumovitz M, Fader AN, Gourley C. Innovations in Rare Gynecologic Cancer: Melanoma, Neuroendocrine, and Low-Grade Serous Ovarian. Am Soc Clin Oncol Educ Book 2024; 44:e431818. [PMID: 39177646 DOI: 10.1200/edbk_431818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
In the field of gynecologic cancer, low-grade serous ovarian cancer (LGSOC) has been poorly understood and underinvestigated until recently. Similarly, understanding of the distinct properties and therapeutic sensitivities of gynecologic melanoma and cervical neuroendocrine tumors has recently accelerated. For each of these rare cancers, we explore the epidemiology and natural history, discuss the prognosis, diagnostic testing, and contemporary molecular classification, and then deliberate existing and emerging therapeutic strategies. In LGSOC, we focus on the clinical relevance of recent molecular studies that shed light on the importance of mitogen-activated protein kinase (MAPK) pathway gene mutation and chromosome 1 copy-number change on prognosis and MEK inhibitor sensitivity. We also discuss the relative chemoresistance of this disease and the fact that attention is shifting to combinations of molecular therapies such as endocrine agents plus cyclin-dependent kinase 4/6 inhibitors or MEK inhibitors plus FAK inhibitors. Gynecologic tract melanomas harbor a lower frequency of canonical BRAF mutations, and have lower tumor mutational burden and immune cell infiltration than cutaneous melanomas (CMs). As a result, patients with this disease are less likely to respond to BRAF/MEK or immune checkpoint inhibition than patients with CM. Emerging strategies include the combination of antiangiogenic agents with immune checkpoint inhibitors and the use of adoptive cellular therapies. In cervical neuroendocrine cancer, we discuss the use of surgery in early-stage disease, and the uncertainties regarding the role of radiotherapy. We also explore the evidence for chemotherapy and emerging investigational strategies including the use of poly (ADP-ribose) polymerase inhibitors. For all situations, we explore the shared decision-making process with the patient.
Collapse
Affiliation(s)
- George Au-Yeung
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Emily MacArthur
- Kelly Gynecologic Oncology Service, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, MD
| | - Joanna Chan
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Narthana Ilenkovan
- Cancer Research UK Scotland Institute, University of Glasgow, Glasgow, United Kingdom
| | - Michael Frumovitz
- Department of Gynecologic Oncology and Reproductive Medicine, MD Anderson Cancer Center, Houston, TX
| | - Amanda N Fader
- Kelly Gynecologic Oncology Service, Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, MD
| | - Charlie Gourley
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
3
|
Lliberos C, Richardson G, Papa A. Oncogenic Pathways and Targeted Therapies in Ovarian Cancer. Biomolecules 2024; 14:585. [PMID: 38785992 PMCID: PMC11118117 DOI: 10.3390/biom14050585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/06/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
Epithelial ovarian cancer (EOC) is one of the most aggressive forms of gynaecological malignancies. Survival rates for women diagnosed with OC remain poor as most patients are diagnosed with advanced disease. Debulking surgery and platinum-based therapies are the current mainstay for OC treatment. However, and despite achieving initial remission, a significant portion of patients will relapse because of innate and acquired resistance, at which point the disease is considered incurable. In view of this, novel detection strategies and therapeutic approaches are needed to improve outcomes and survival of OC patients. In this review, we summarize our current knowledge of the genetic landscape and molecular pathways underpinning OC and its many subtypes. By examining therapeutic strategies explored in preclinical and clinical settings, we highlight the importance of decoding how single and convergent genetic alterations co-exist and drive OC progression and resistance to current treatments. We also propose that core signalling pathways such as the PI3K and MAPK pathways play critical roles in the origin of diverse OC subtypes and can become new targets in combination with known DNA damage repair pathways for the development of tailored and more effective anti-cancer treatments.
Collapse
Affiliation(s)
- Carolina Lliberos
- Cancer Program, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia;
- Neil Beauglehall Department of Medical Oncology Research, Cabrini Health, Malvern, VIC 3144, Australia
| | - Gary Richardson
- Neil Beauglehall Department of Medical Oncology Research, Cabrini Health, Malvern, VIC 3144, Australia
| | - Antonella Papa
- Cancer Program, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia;
| |
Collapse
|
4
|
Ottenbourgs T, Van Nieuwenhuysen E. Novel Endocrine Therapeutic Opportunities for Estrogen Receptor-Positive Ovarian Cancer-What Can We Learn from Breast Cancer? Cancers (Basel) 2024; 16:1862. [PMID: 38791941 PMCID: PMC11119209 DOI: 10.3390/cancers16101862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Low-grade serous ovarian cancer (LGSOC) is a rare ovarian malignancy primarily affecting younger women and is characterized by an indolent growth pattern. It exhibits indolent growth and high estrogen/progesterone receptor expression, suggesting potential responsiveness to endocrine therapy. However, treatment efficacy remains limited due to the development of endocrine resistance. The mechanisms of resistance, whether primary or acquired, are still largely unknown and present a significant hurdle in achieving favorable treatment outcomes with endocrine therapy in these patients. In estrogen receptor-positive breast cancer, mechanisms of endocrine resistance have been largely explored and novel treatment strategies to overcome resistance have emerged. Considering the shared estrogen receptor positivity in LGSOC and breast cancer, we wanted to explore whether there are any parallel mechanisms of resistance and whether we can extend endocrine breast cancer treatments to LGSOC. This review aims to highlight the underlying molecular mechanisms possibly driving endocrine resistance in ovarian cancer, while also exploring the available therapeutic opportunities to overcome this resistance. By unraveling the potential pathways involved and examining emerging strategies, this review explores valuable insights for advancing treatment options and improving patient outcomes in LGSOC, which has limited therapeutic options available.
Collapse
Affiliation(s)
- Tine Ottenbourgs
- Gynaecological Oncology Laboratory, KU Leuven, Leuven Cancer Institute, 3000 Leuven, Belgium;
| | - Els Van Nieuwenhuysen
- Department of Gynaecology and Obstetrics, University Hospitals Leuven, BGOG and Leuven Cancer Institute, 3000 Leuven, Belgium
| |
Collapse
|
5
|
Zhou I, Plana D, Palmer AC. Tumor-Specific Activity of Precision Medicines in the NCI-MATCH Trial. Clin Cancer Res 2024; 30:786-792. [PMID: 38109210 PMCID: PMC10922532 DOI: 10.1158/1078-0432.ccr-23-0983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 09/07/2023] [Accepted: 12/13/2023] [Indexed: 12/20/2023]
Abstract
PURPOSE National Cancer Institute Molecular Analysis for Therapy Choice (NCI-MATCH) is a precision medicine basket trial designed to test the effectiveness of treating cancers based on specific genetic changes in patients' tumors, regardless of cancer type. Multiple subprotocols have each tested different targeted therapies matched to specific genetic aberrations. Most subprotocols exhibited low rates of tumor shrinkage as evaluated across all tumor types enrolled. We hypothesized that these results may arise because these precision cancer therapies have tumor type-specific efficacy, as is common among other cancer therapies. EXPERIMENTAL DESIGN To test the hypothesis that certain tumor types are more sensitive to specific therapies than other tumor types, we applied permutation testing to tumor volume change and progression-free survival data from 10 published NCI-MATCH subprotocols (together n = 435 patients). FDR was controlled by the Benjamini-Hochberg procedure. RESULTS Six of ten subprotocols exhibited statistically significant evidence of tumor-specific drug sensitivity, four of which were previously considered negative based on response rate across all tumors. This signal-finding analysis highlights potential uses of FGFR tyrosine kinase inhibition in urothelial carcinomas with actionable FGFR aberrations and MEK inhibition in lung cancers with BRAF non-V600E mutations. In addition, it identifies low-grade serious ovarian carcinoma with BRAF v600E mutation as especially sensitive to BRAF and MEK co-inhibition (dabrafenib plus trametinib), a treatment that received accelerated FDA approval for advanced solid tumors with BRAF v600E mutation. CONCLUSIONS These findings support the value of basket trials because even when precision medicines do not have tumor-agnostic activity, basket trials can identify tumor-specific activity for future study.
Collapse
Affiliation(s)
- Ivvone Zhou
- Department of Pharmacology, Computational Medicine Program, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| | - Deborah Plana
- Laboratory of Systems Pharmacology, and the Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts, 02139, USA
| | - Adam C. Palmer
- Department of Pharmacology, Computational Medicine Program, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, 27599, USA
| |
Collapse
|
6
|
Antunes Meireles P, Mira B, Vaz F. Complete Response With Trametinib in Advanced Low-Grade Serous Ovarian Carcinoma: A Case Report. Cureus 2024; 16:e53600. [PMID: 38449951 PMCID: PMC10915706 DOI: 10.7759/cureus.53600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2024] [Indexed: 03/08/2024] Open
Abstract
Low-grade serous ovarian carcinoma (LGSOC) is an uncommon subtype of ovarian cancer, and it is usually associated with reduced sensitivity to chemotherapy and worse outcomes. We present a case involving a 45-year-old female patient diagnosed with stage III-C low-grade serous ovarian carcinoma (LGSOC) in 2013. She achieved a complete response for 29 months after undergoing platinum-based chemotherapy and interval cytoreduction. However, in 2016, both local and distant relapses were observed. As there was no benefit from hormonal therapy and the patient refused chemotherapy, bevacizumab was initiated, resulting in disease stabilization for 30 months. At disease progression, trametinib was proposed, and the patient experienced an ongoing sustained complete response for over 36 months. To the best of our knowledge, this is the first report, outside of a clinical trial, regarding a complete response with single agent MEK inhibitor therapy in a patient with recurrent LGSOC, with unknown BRAF V600E mutation. We present the following case in accordance with the CAse REports (CARE) checklist.
Collapse
Affiliation(s)
- Pedro Antunes Meireles
- Department of Medical Oncology, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, PRT
| | - Beatriz Mira
- Department of Medical Oncology, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, PRT
| | - Fátima Vaz
- Department of Medical Oncology, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, PRT
| |
Collapse
|
7
|
Zwimpfer TA, Tal O, Geissler F, Coelho R, Rimmer N, Jacob F, Heinzelmann-Schwarz V. Low grade serous ovarian cancer - A rare disease with increasing therapeutic options. Cancer Treat Rev 2023; 112:102497. [PMID: 36525716 DOI: 10.1016/j.ctrv.2022.102497] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
High-grade serous ovarian cancers (HGSOCs) most commonly arise from the fimbrial end of the fallopian tube and harbor TP53 gene mutations. In contrast, low-grade serous ovarian cancers (LGSOCs) appear to have different pathological, epidemiological, and clinical features and should be seen as a distinct serous epithelial ovarian cancer subtype. Our current understanding of LGSOC is limited, and treatment has generally been derived from the more common HGSOCs due to a lack of separate trial data. LGSOCs are characterized by slow tumor growth and are assumed to develop from serous borderline ovarian tumors as precursors. These cancers are often estrogen-receptor positive and show an activated mitogen-activated protein kinase pathway together with KRAS and BRAF mutations and, rarely, TP53 mutations. These characteristics are now commonly used to guide therapeutical decision making and, consequently, a substantial part of treatment consists of maintenance with endocrine treatment, thus balancing disease stabilization and mild toxicity. Additionally, new trials are ongoing that examine the role of targeted therapies such as MEK inhibitors in combination with endocrine treatments. The purpose of this work is to summarize current knowledge and present ongoing trial efforts for LGSOCs.
Collapse
Affiliation(s)
- Tibor A Zwimpfer
- Peter MacCallum Cancer Center, East Melbourne, Victoria 3002, Australia; Department of Gynecological Oncology, University Hospital Basel, 4031 Basel, Switzerland.
| | - Ori Tal
- Department of Obstetrics and Gynecology, Edith Wolfson Medical Center, Holon, Israel
| | - Franziska Geissler
- Department of Gynecological Oncology, University Hospital Basel, 4031 Basel, Switzerland
| | - Ricardo Coelho
- Ovarian Cancer Research, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Natalie Rimmer
- Ovarian Cancer Research, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Francis Jacob
- Ovarian Cancer Research, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Viola Heinzelmann-Schwarz
- Peter MacCallum Cancer Center, East Melbourne, Victoria 3002, Australia; Ovarian Cancer Research, Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| |
Collapse
|
8
|
Llaurado Fernandez M, Hijmans EM, Gennissen AM, Wong NK, Li S, Wisman GBA, Hamilton A, Hoenisch J, Dawson A, Lee CH, Bittner M, Kim H, DiMattia GE, Lok CA, Lieftink C, Beijersbergen RL, de Jong S, Carey MS, Bernards R, Berns K. NOTCH Signaling Limits the Response of Low-Grade Serous Ovarian Cancers to MEK Inhibition. Mol Cancer Ther 2022; 21:1862-1874. [PMID: 36198031 PMCID: PMC9716250 DOI: 10.1158/1535-7163.mct-22-0004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 05/30/2022] [Accepted: 10/03/2022] [Indexed: 01/12/2023]
Abstract
Low-grade serous ovarian cancer (LGSOC) is a rare subtype of epithelial ovarian cancer with high fatality rates in advanced stages due to its chemoresistant properties. LGSOC is characterized by activation of MAPK signaling, and recent clinical trials indicate that the MEK inhibitor (MEKi) trametinib may be a good treatment option for a subset of patients. Understanding MEKi-resistance mechanisms and subsequent identification of rational drug combinations to suppress resistance may greatly improve LGSOC treatment strategies. Both gain-of-function and loss-of-function CRISPR-Cas9 genome-wide libraries were used to screen LGSOC cell lines to identify genes that modulate the response to MEKi. Overexpression of MAML2 and loss of MAP3K1 were identified, both leading to overexpression of the NOTCH target HES1, which has a causal role in this process as its knockdown reversed MEKi resistance. Interestingly, increased HES1 expression was also observed in selected spontaneous trametinib-resistant clones, next to activating MAP2K1 (MEK1) mutations. Subsequent trametinib synthetic lethality screens identified SHOC2 downregulation as being synthetic lethal with MEKis. Targeting SHOC2 with pan-RAF inhibitors (pan-RAFis) in combination with MEKi was effective in parental LGSOC cell lines, in MEKi-resistant derivatives, in primary ascites cultures from patients with LGSOC, and in LGSOC (cell line-derived and patient-derived) xenograft mouse models. We found that the combination of pan-RAFi with MEKi downregulated HES1 levels in trametinib-resistant cells, providing an explanation for the synergy that was observed. Combining MEKis with pan-RAFis may provide a promising treatment strategy for patients with LGSOC, which warrants further clinical validation.
Collapse
Affiliation(s)
- Marta Llaurado Fernandez
- Department of Obstetrics and Gynaecology, University of British Columbia Vancouver, British Columbia, Canada
| | - E. Marielle Hijmans
- Division of Molecular Carcinogenesis, Oncode Institute, Cancer Genomics Center Netherlands, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annemiek M.C. Gennissen
- Division of Molecular Carcinogenesis, Oncode Institute, Cancer Genomics Center Netherlands, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Nelson K.Y. Wong
- Department of Obstetrics and Gynaecology, University of British Columbia Vancouver, British Columbia, Canada.,Department of Experimental Therapeutics, BC Cancer, Vancouver, British Columbia, Canada
| | - Shang Li
- Department of Medical Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - G. Bea A. Wisman
- Department of Gynecologic Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Aleksandra Hamilton
- Department of Obstetrics and Gynaecology, University of British Columbia Vancouver, British Columbia, Canada
| | - Joshua Hoenisch
- Department of Obstetrics and Gynaecology, University of British Columbia Vancouver, British Columbia, Canada
| | - Amy Dawson
- Department of Obstetrics and Gynaecology, University of British Columbia Vancouver, British Columbia, Canada
| | - Cheng-Han Lee
- Department of Obstetrics and Gynaecology, University of British Columbia Vancouver, British Columbia, Canada
| | - Madison Bittner
- Department of Obstetrics and Gynaecology, University of British Columbia Vancouver, British Columbia, Canada
| | - Hannah Kim
- Department of Obstetrics and Gynaecology, University of British Columbia Vancouver, British Columbia, Canada
| | - Gabriel E. DiMattia
- Mary and John Knight Translational Ovarian Cancer Research Unit, London Health Sciences Center
| | - Christianne A.R. Lok
- Center for Gynecologic Oncology Amsterdam, Antoni van Leeuwenhoek/The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Cor Lieftink
- Division of Molecular Carcinogenesis, Oncode Institute, Cancer Genomics Center Netherlands, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Roderick L. Beijersbergen
- Division of Molecular Carcinogenesis, Oncode Institute, Cancer Genomics Center Netherlands, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Steven de Jong
- Department of Medical Oncology, Cancer Research Center Groningen, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mark S. Carey
- Department of Obstetrics and Gynaecology, University of British Columbia Vancouver, British Columbia, Canada.,Corresponding Authors: Katrien Berns, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands. Phone: 31-20-5121955. E-mail: ; and Mark S. Carey, Vancouver, British Columbia V6Z 2K8, Canada. Phone: 160-4875-4268; E-mail: ; René Bernards, Plesmanlaan 121,1066 CX Amsterdam, the Netherlands. Phone: 31-20-5121952; E-mail:
| | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, Cancer Genomics Center Netherlands, the Netherlands Cancer Institute, Amsterdam, the Netherlands.,Corresponding Authors: Katrien Berns, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands. Phone: 31-20-5121955. E-mail: ; and Mark S. Carey, Vancouver, British Columbia V6Z 2K8, Canada. Phone: 160-4875-4268; E-mail: ; René Bernards, Plesmanlaan 121,1066 CX Amsterdam, the Netherlands. Phone: 31-20-5121952; E-mail:
| | - Katrien Berns
- Division of Molecular Carcinogenesis, Oncode Institute, Cancer Genomics Center Netherlands, the Netherlands Cancer Institute, Amsterdam, the Netherlands.,Corresponding Authors: Katrien Berns, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands. Phone: 31-20-5121955. E-mail: ; and Mark S. Carey, Vancouver, British Columbia V6Z 2K8, Canada. Phone: 160-4875-4268; E-mail: ; René Bernards, Plesmanlaan 121,1066 CX Amsterdam, the Netherlands. Phone: 31-20-5121952; E-mail:
| |
Collapse
|
9
|
Gilks CB, Selinger CI, Davidson B, Köbel M, Ledermann JA, Lim D, Malpica A, Mikami Y, Singh N, Srinivasan R, Vang R, Lax SF, McCluggage WG. Data Set for the Reporting of Ovarian, Fallopian Tube and Primary Peritoneal Carcinoma: Recommendations From the International Collaboration on Cancer Reporting (ICCR). Int J Gynecol Pathol 2022; 41:S119-S142. [PMID: 36305537 DOI: 10.1097/pgp.0000000000000908] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The move toward consistent and comprehensive surgical pathology reports for cancer resection specimens has been a key development in supporting evidence-based patient management and consistent cancer staging. The International Collaboration on Cancer Reporting (ICCR) previously developed a data set for reporting of the ovarian, fallopian tube and primary peritoneal carcinomas which was published in 2015. In this paper, we provide an update on this data set, as a second edition, that reflects changes in the 2020 World Health Organization (WHO) Classification of Female Genital Tumours as well as some other minor modifications. The data set has been developed by a panel of internationally recognized expert pathologists and a clinician and consists of "core" and "noncore" elements to be included in surgical pathology reports, with detailed commentary to guide users, including references. This data set replaces the widely used first edition, and will facilitate consistent and accurate case reporting, data collection for quality assurance and research, and allow for comparison of epidemiological and pathologic parameters between different populations.
Collapse
|
10
|
Genomic and TCR profiling data reveal the distinct molecular traits in epithelial ovarian cancer histotypes. Oncogene 2022; 41:3093-3103. [PMID: 35468938 DOI: 10.1038/s41388-022-02277-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 01/24/2022] [Accepted: 03/11/2022] [Indexed: 11/08/2022]
Abstract
Epithelial ovarian cancer (EOC) is classified into five major histotypes: high-grade serous (HGSOC), low-grade serous (LGSOC), clear cell (CCOC), endometrioid (ENOC), and mucinous (MOC). However, the landscape of molecular and immunological alterations in these histotypes, especially LGSOC, CCOC, ENOC, and MOC, is largely uncharacterized. We collected 101 treatment-naive EOC patients. The resected tumor tissues and paired preoperative peripheral blood samples were collected and subjected to target sequencing of 1021 cancer-associated genes and T cell repertoire sequencing. Distinct characteristics of mutations were identified among the five histotypes. Furthermore, tumor mutation burden (TMB) was found to be higher in CCOC and ENOC, but lower in LGSOC and HGSOC. Alterations associated with DNA damage repair (DDR) pathways and homologous recombination deficiencies (HRD) were prevalent in five histotypes. CCOC demonstrated increased level of T cell clonality compared with HSGOC. Interestingly, the proportion of the 100 most common T cell clones was associated with TMB and tumor neoantigen burden in CCOC, highlighting more sensitive anti-tumor responses in this histotype, which was also evidenced by the enhanced convergent recombination of T cell clones. These findings shed light on the molecular traits of genomic alteration and T cell repertoire in the five major EOC histotypes and may help optimize clinical management of EOC with different histotypes.
Collapse
|
11
|
Impressive and durable clinical responses obtained with dabrafenib and trametinib in low-grade serous ovarian cancer harbouring a BRAF V600E mutation. Gynecol Oncol Rep 2022; 40:100942. [PMID: 35242981 PMCID: PMC8873542 DOI: 10.1016/j.gore.2022.100942] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 11/30/2022] Open
Abstract
LGSOC is characterized scarce therapeutic options in the advanced setting. BRAF mutations are frequent on these types of tumours. Dabrafenib/trametinib yield impressive responses in patients with BRAF mutations.
Low-grade serous ovarian cancer (LGSOC) is now considered a different entity from high-grade serous ovarian cancer. The chemoresistance inherent to this type of ovarian cancer narrows the therapeutic options, especially in the recurrent setting. It is thought that the mitogen-activated protein kinase (MAPK) pathway plays a significant role in the pathogenesis of these tumours, and about 2 to 20% of LGSOC harbour a BRAF mutation. Here we present a case report of two patients with a BRAF V600E mutation that achieved sustained clinical responses with combination treatment with dabrafenib (BRAF inhibitor) and trametinib (MEK inhibitor).
Collapse
|